• Duke University »
  • Pratt School of Engineering »
  • Degree Program
  • Enrollment and Graduation Rates
  • Career Outcomes
  • Campus Tours
  • How to Apply
  • Double Majors
  • BME Design Fellows
  • For Current Students
  • 4+1: BSE+Master's Degree
  • Degree Programs
  • Concentrations
  • Certificates
  • PhD Program
  • Meet Our Students
  • Certificates & Training Programs
  • Major Research Programs
  • Centers & Initiatives
  • Research News
  • Faculty Profiles
  • Awards & Recognition
  • About the Partnership
  • Proposal Process
  • Oversight Committee
  • Welcome from the Chair
  • Vision & Mission
  • Facts & Stats
  • Serving Society
  • Our History
  • Community Resources
  • DEIC Across Duke BME
  • Report a Concern
  • Email Newsletter
  • Media Coverage
  • Driving Directions

microscope image of muscle tissue from Bursac lab

Tissue Engineering and Regenerative Medicine

Research in tissue engineering and regenerative medicine seeks to replace or regenerate diseased or damaged tissues, organs, and cells – a challenging endeavor, but one that has tremendous potential for the practice of medicine.

Technologies under investigation range from biomaterial/cell constructs for repairing various tissues and organs, to stem cell therapies, to immune therapies. Our work in this area is highly multidisciplinary, combining materials science, cell biology, clinical science, immunology, stem cell biology, genome science, and others.

Accordingly, researchers in this area within Duke BME are broadly interactive with departments throughout the university including Duke University Medical Center clinical departments, the Duke University School of Medicine departments of Cell Biology and Immunology, the Duke Department of Chemistry, and others. This community is also supported by centers and programs such as Regeneration Next and the Center for Biomolecular and Tissue Engineering (CBTE) .

Primary Faculty

phd tissue engineering and regenerative medicine

Nenad Bursac

Professor of Biomedical Engineering

Research Interests: Embryonic and adult stem cell therapies for heart and muscle disease; cardiac and skeletal muscle tissue engineering; cardiac electrophysiology and arrhythmias; genetic modifications of stem and somatic cells; micropatterning of proteins and hydrogels.

phd tissue engineering and regenerative medicine

Pranam D. Chatterjee

Assistant Professor of Biomedical Engineering

Research Interests: Integration of computational and experimental methodologies to design novel proteins for applications in genome editing, targeted protein modulation, and reproductive bioengineering

phd tissue engineering and regenerative medicine

Joel Collier

Theodore Kennedy Professor of Biomedical Engineering

Research Interests: The design of biomaterials for a range of biomedical applications, with a focus on understanding and controlling adaptive immune responses. Most materials investigated are created from molecular assemblies- proteins, peptides or bioconjugates that self-organize into useful…

phd tissue engineering and regenerative medicine

Sharon Gerecht

Paul M. Gross Distinguished Professor of Biomedical Engineering

Research Interests: stem cells, biomaterials, hypoxia, blood vessels, physics of cancer, regenerative medicine

phd tissue engineering and regenerative medicine

Charles Gersbach

John W. Strohbehn Distinguished Professor of Biomedical Engineering

Research Interests: Gene therapy, genomics and epigenomics, biomolecular and cellular engineering, regenerative medicine, and synthetic biology.

phd tissue engineering and regenerative medicine

John Wirthlin Hickey

phd tissue engineering and regenerative medicine

Samira Musah

Assistant Professor in the Department of Biomedical Engineering

Research Interests: Induced pluripotent stem cells (iPS cells), disease mechanisms, regenerative medicine, molecular and cellular basis of human kidney development and disease, organ engineering, patient-specific disease models, biomarkers, therapeutic discovery, tissue and organ transplantation,…

phd tissue engineering and regenerative medicine

Tatiana Segura

Research Interests: The design of biomaterials to promote endogenous repair and reducing inflammation through the design of the geometry of the material, and delivering genes, proteins and drugs.

phd tissue engineering and regenerative medicine

George A. Truskey

R. Eugene and Susie E. Goodson Distinguished Professor of Biomedical Engineering

Research Interests: Cardiovascular tissue engineering, mechanisms of atherogenesis, cell adhesion, and cell biomechanics.

phd tissue engineering and regenerative medicine

Shyni Varghese

Professor of Biomedical Engineering, Mechanical Engineering & Materials Science and Orthopaedics

Research Interests: Musculoskeletal tissue repair, disease biophysics and organ-on-a-chip technology

Secondary Faculty

phd tissue engineering and regenerative medicine

Geoffrey Steven Ginsburg

Adjunct Professor in the Department of Medicine

phd tissue engineering and regenerative medicine

Cynthia Ann Toth

Joseph A.C. Wadsworth Distinguished Professor of Ophthalmology

phd tissue engineering and regenerative medicine

Stefan Zauscher

Professor in the Thomas Lord Department of Mechanical Engineering and Materials Science

Research Interests: Nano-mechanical and nano-tribological characterization (elasticity, friction, adhesion) of materials including organic thin films; self-assembled monolayers, polymeric gels, and cellulosics; Fabrication of polymeric nanostructures by scanning probe lithography; Colloidal probe…

Adjunct Faculty

phd tissue engineering and regenerative medicine

Jennifer L West

Adjunct Professor of Biomedical Engineering

Research Interests: Biomaterials, nanotechnology and tissue engineering that involves the synthesis, development, and application of novel biofunctional materials, and the use of biomaterials and engineering approaches to study biological problems.

Faculty Emeritus

phd tissue engineering and regenerative medicine

William M. Reichert

Professor Emeritus of Biomedical Engineering

Research Interests: Biosensors, protein mediated cell adhesion, and wound healing.

Updates on COVID-19 for Grad Students and Postdocs

Graduate program in stem cell biology & regenerative medicine, stanford is a world leader in stem cell research and regenerative medicine. central discoveries in stem cell biology – tissue stem cells and their use for regenerative therapies, transdifferentiation into mature cell-types, isolation of cancerous stem cells, and stem cell signaling pathways – were made by stanford faculty and students. our mission is to train the next generation of stem cell scientists..

Lab_handsW350

About the SCBRM Graduate Program

Majeti_Lab

Unique Opportunities for Medical Students

2019 class photo

Stem Cell PhD Program

weissman talk

Faculty and Their Research Interests

lokey building

Application Process

ludwig beachy3

Curriculum and Handbooks

Learn about the many ways to support the institute for Stem Cell Biology and Regenerative Medicine

  • Student/Faculty Portal
  • Learning Hub (Brightspace)
  • Continuous Professional Development

Regenerative Sciences

Regenerative sciences track.

faculty spanning multidisciplinary departments

education in discovery, clinical translation, and application of regenerative solutions

Guaranteed 5-year internal fellowship

includes full tuition, stipend and benefits

Seeking to spur development of innovative medical breakthroughs, Mayo Clinic Graduate School of Biomedical Sciences, in partnership with the Center for Regenerative Biotherapeutics , started one of the nation's first doctoral research training programs in regenerative sciences.

Regenerative medicine is transforming clinical practice with the development of new therapies, treatments and surgeries for patients with chronic conditions, debilitating injuries and degenerative diseases. Advances in developmental and cell biology, immunology, and other fields unlock new opportunities for innovative breakthroughs for the next generation of regenerative diagnostic and therapeutic medical solutions.

The Regenerative Sciences (REGS) Ph.D. track at Mayo Clinic is a transdisciplinary Ph.D. Program designed to prepare the next generation of scientists to accelerate the discovery, translation, and application of cutting-edge regenerative diagnostics and therapeutics. The REGS Ph.D. track builds on the existing Mayo Clinic Regenerative Sciences Training Program (RSTP) to now offer in-depth curriculum and advanced training opportunities. 

The Regenerative Sciences Track places a significant emphasis on laboratory-based research training. Laboratory research is complemented with both core and track-specific courses, as well as advanced courses on current topics in regenerative science and medicine.

The regenerative sciences curriculum encompasses the full spectrum of regenerative science topics, including molecular and cell biology, stem cell biology, developmental biology, tissue engineering, biomaterials and nanomedicine, genome editing and gene therapies, regulatory and translational science, product development, biomanufacturing, entrepreneurship and more.

Students in Regenerative Sciences join a close-knit community of learners, are provided unique hands-on- experiences and collaborate with some of the brightest minds in the field.

See the full Regenerative Sciences Track curriculum (PDF)

Graduates of the Regenerative Sciences Ph.D. track will be integral to forming the multidisciplinary workforce needed to drive the future of health care at Mayo Clinic and across the world.

Learn more:  What is Regenerative Medicine - Mayo Clinic Radio

Focus areas

  • Molecular and epigenetic mechanisms of stem and progenitor cell proliferation and differentiation, as well as tissue degeneration and regeneration
  • Immune responses to viral insult and tissue healing
  • Gene editing for cell therapy applications and to alter disease progression
  • Extracellular vesicles in disease progression and for tissue regeneration
  • Tissue engineering and bioengineering of novel therapies, including 3-D printing, electrospinning, and advanced biomanufacturing 

Profile photo of Shan Gao, a Ph.D. student at Mayo Clinic. She is looking at the camera and smiling.

Mayo Clinic is an incredible place for doctoral training in regenerative science. The interdisciplinary strategy here allows research and courses to be tailored according to each student’s interests and ability. Moreover, Mayo Clinic provides a wealth resource to develop collaborations within the institution, which will offer students more ways to communicate and promote students to achieve their personal goals.

Shan Gao Ph.D. student, Regenerative Sciences Track

Armin Garmany

Mayo Clinic provides unparalleled access to world-renowned clinicians and researchers all focused on clinically relevant research. Mayo Clinic’s Center for Regenerative Medicine permeates throughout the institution. Thus, the REGS program gives students the necessary experience and knowledge to drive future research in restoring form and function in any field of medicine.

Armin Garmany M.D.-Ph.D. student, Regenerative Sciences Track

Profile photo of Samuel Buchl, a Ph.D. student at Mayo Clinic.

The study of Regenerative Sciences (REGS) at Mayo Clinic is unparalleled. Students are funded to study cutting-edge biomedical science in their domain of interest with plentiful opportunities to translate benchside discoveries to the patient bedside and beyond. I chose Mayo Clinic's REGS program to join its community of researchers, practitioners, and entrepreneurs who everyday advance the science and practice of regenerative medicine and bring new regenerative solutions to the world.

Samuel Buchl Ph.D. student, Regenerative Sciences Track

Profile photo of Delaney Liskey, a Ph.D. student at Mayo Clinic

The Regenerative Sciences Ph.D. track at Mayo Clinic thoroughly equips students to be leaders in biomedical research through an unmatched curriculum of multidisciplinary science and world-class research training. REGS is a collaborative and supportive program in a promising field of medicine that provides the foundational skills to pipeline research to patient care.

Delaney Liskey Ph.D. student, Regenerative Sciences Track

Thesis topics

Current students thesis topics.

  • "Targeted Regenerative Therapies for Heart Failure Susceptibility," Armin Garmany (Mentor: Andre Terzic, M.D., Ph.D.)
  • "Novel Look Into the Crude Stromal Vascular Fraction (SVF) from Human Adipose-Derived Tissue and Its Role in Regulating the Self-Renewing Capacity of Brain Tumor-Initiating Cells," Rawan Alkharboosh (Mentor: Alfredo Quinones-Hinojosa, M.D.)
  • "Tissue Quality in Existing and Emerging Treatments for Osteoarthritis," Katherine Arnold (Mentor: Jennifer Westendorf, Ph.D.)
  • "Harnessing the Mesenchymal Stem Cell Secretome to Target Alpha-Synuclein-Associated Dysfunction in Parkinson's Disease," Jeremy Burgess (Mentor: Pamela McLean, Ph.D.)
  • "Retinal Neuroprotection Properties of an ATP-Sensitive Potassium Channel Opener," Catherine Knier (Mentor: Michael Fautsch, Ph.D.)
  • "Towards a Subcutaneous Combination Biodevice for the Treatment of Type 1 Diabetes," Ethan Law (Mentor: Quinn Peterson, Ph.D.)
  • "Modulation of CART Cell Activation to Enhance Antitumor Response via CRISPR-mediated Gene Editing and Combined Immunotherapy," Claudia Manriquez Roman (Mentor: Saad Kenderian, M.B., Ch.B.)
  • "Systems Biology for Engineering Regenerative Immunotherapies in Precision Neuro-oncology," Dileep Monie (Mentors: Hu Li, Ph.D. and Richard Vile, Ph.D.
  • "APOE2 Effects on Central and Peripheral Vasculature," Francis Shue (Mentor: Guojun Bu, Ph.D.)
  • "Engineering of Antiviral Extracellular Vesicles," Amanda Terlap (Mentor: Atta Behfar, M.D., Ph.D.) 
  • "Glycome of Breast Cancer-Derived Extracellular Vesicles in Metastasis," Sierra Walker (Mentor: Joy Wolfram, Ph.D.)
  • "Bidirectional Interactions Between Stem Cell Populations of the Subventricular Zone and Glioblastoma," Emily Norton (Mentor: Hugo Guerrero Cazares, M.D., Ph.D.)
  • "Measles Virus Vector for Gene Editing and Reprogramming of Human Fibroblasts," Ramya Rallabandi (Mentor: Patricia Devaux, Ph.D.)
  • "Precise Genetic Engineering of Human Primary Cells for Cell Therapy-Based Applications," (Mentor: Stephen Ekker, Ph.D.)

Recent graduates thesis topics

  • "Epigenetic Control of the Architectural and Trophic Functions of Mesenchymal Stem Cells in Musculoskeletal Tissue Regeneration Therapies," (Mentor: Andre van Wijnen, Ph.D.)
  • "Metabolic Regulation of Muscle Stem Cells," (Mentor: Jason Doles, Ph.D.)
  • "Purified Exosome Product Enhances Neovascularization in Peripheral Arterial Disease," (Mentors: Atta Behfar, M.D., Ph.D. and Andre Terzic, M.D., Ph.D.)
  • "Antigen Presentation by CNS-Resident Microglia and Macrophages is Required for Antigen-Specific CD8 T Cell Responses in the Brain Following Viral Challenge," (Mentor: Aaron Johnson, Ph.D.)

Meet the director

Isobel Scarisbrick, Ph.D.

Training opportunities extend from fundamental science principles through laboratory skills and hands-on experiences. Students will also have the opportunity to develop an understanding of national and international regulatory agencies, and business requirements and procedures needed to implement the discovery, translation, application pipeline for new regenerative technologies.

We are excited to provide a program of training that will serve as an incubator to develop the next generation of leaders in regenerative science and medicine.

Isobel Scarisbrick, Ph.D. Regenerative Sciences Track Director Professor of Physical Medicine & Rehabilitation Phone: 507-284-0124 Email: [email protected] See research interests

Browse a list of Regenerative Sciences Track faculty members

Shield

KEY APPLICATION AREA

Tissue Engineering & Regenerative Medicine

Research in tissue engineering and regenerative medicine encompasses all aspects of the research and development continuum from mechanistic studies to translational approaches. Collaborative efforts with colleagues at Rice and the Texas Medical Center address unmet clinical needs for a plethora of tissues ranging from bone to cartilage to heart valve to inner ear.

Specific areas of interest include structure and function relationships in living tissues, synthesis and fabrication of biomimetic materials and extracellular matrix constructs, combinations of biomaterials with cell populations for modulating cell function and guiding tissue growth, stem cell programming, drug and gene delivery systems for tissue induction and regeneration, 3D printing and bioprinting, and bioreactor designs for cell culture and disease modeling.

Rice BIOE researchers working in this key application area:

Caleb bashor, phd, faculty profile | laboratory website, jane grande-allen, phd, isaac hilton, phd, kevin mchugh, phd, antonios mikos, phd, jordan miller, phd, robert raphael, phd, omid veiseh, phd.

U.S. flag

An official website of the United States government

Here’s how you know

Official websites use .gov A .gov website belongs to an official government organization in the United States.

Secure .gov websites use HTTPS A lock ( Lock A locked padlock ) or https:// means you’ve safely connected to the .gov website. Share sensitive information only on official, secure websites.

National Institute of Dental and Craniofacial Research

  • Grants & Funding
  • Grant Programs

Tissue Engineering & Regenerative Medicine Research Program

Staff contacts.

Scott Verbridge, Ph.D.

Regenerative Medicine

18 Dec 2023 Turning the Tap Back On

07 Jan 2021 Stem Cell Treatment Corrects Skull Shape and Restores Brain Function in Mouse Model of Childhood Disorder

28 Jan 2021 Surgical Adhesive Inspired by Slug Slime

30 Sep 2019 How Does a Mouse’s Tooth Grow?

19 Nov 2018 The Quest to Understand Dental Stem Cells

15 Oct 2018 Human Skeletal Stem Cell Identified

Grantee News

26 Apr 2023 Do Spiny Mice Hold the Key to Regenerative Healing? UK Study Explores

17 Apr 2023 Healing the Unhealable: New Approach Helps Bones Mend Themselves

22 Mar 2023 Researchers Find Key to Healing Muscle Injuries in Elderly

09 Jan 2022 A Crowning Achievement in Understanding Head Development

18 Nov 2021 A Stunning 3D Map of Blood Vessels and Cells in a Mouse Skull Could Help Scientists Make New Bones

07 Sep 2021 Nerve Repair, With Help From Stem Cells

06 Oct 2021 Massage Doesn’t Just Make Muscles Feel Better, it Makes Them Heal Faster and Stronger

26 Apr 2021 Skin and Bones Repaired by Bioprinting during Surgery

14 May 2018 First Description of mEAK-7 Gene Could Suggest Path toward Therapies for Cancer, Other Diseases

12 Jul 2017 Diabetes Causes Shift in Oral Microbiome that Fosters Periodontitis, Penn Study Finds

Funding Opportunities & Notices

Previously funded grants.

NIH RePORTER - Research Portfolio Online Reporting Tool (RePORT)

  • U.S. Department of Health & Human Services
  • National Institutes of Health

NIBIB Logo

En Español | Site Map | Staff Directory | Contact Us

  • Science Education
  • Science Topics

Tissue Engineering and Regenerative Medicine

What are tissue engineering and regenerative medicine, how do tissue engineering and regenerative medicine work, how do tissue engineering and regenerative medicine fit in with current medical practices, what are nih-funded researchers developing in the areas of tissue engineering and regenerative medicine.

An image of a small human tissue engineered liver on the tip of a gloved hand

Tissue engineering evolved from the field of biomaterials development and refers to the practice of combining scaffolds, cells, and biologically active molecules into functional tissues. The goal of tissue engineering is to assemble functional constructs that restore, maintain, or improve damaged tissues or whole organs. Artificial skin and cartilage are examples of engineered tissues that have been approved by the FDA; however, currently they have limited use in human patients.  

Regenerative medicine is a broad field that includes tissue engineering but also incorporates research on self-healing – where the body uses its own systems, sometimes with help foreign biological material to recreate cells and rebuild tissues and organs.  The terms “tissue engineering” and “regenerative medicine” have become largely interchangeable, as the field hopes to focus on cures instead of treatments for complex, often chronic, diseases.

This field continues to evolve. In addition to medical applications, non-therapeutic applications include using tissues as biosensors to detect biological or chemical threat agents, and tissue chips that can be used to test the toxicity of an experimental medication.

A baboon occyte surrounded by granulosa (cumulus) is cultured in a 3D alginate matrix

Cells are the building blocks of tissue, and tissues are the basic unit of function in the body. Generally, groups of cells make and secrete their own support structures, called extra-cellular matrix. This matrix, or scaffold, does more than just support the cells; it also acts as a relay station for various signaling molecules. Thus, cells receive messages from many sources that become available from the local environment. Each signal can start a chain of responses that determine what happens to the cell. By understanding how individual cells respond to signals, interact with their environment, and organize into tissues and organisms, researchers have been able to manipulate these processes to mend damaged tissues or even create new ones.

The process often begins with building a scaffold from a wide set of possible sources, from proteins to plastics. Once scaffolds are created, cells with or without a “cocktail” of growth factors can be introduced. If the environment is right, a tissue develops.  In some cases, the cells, scaffolds, and growth factors are all mixed together at once, allowing the tissue to “self-assemble.”  

Another method to create new tissue uses an existing scaffold. The cells of a donor organ are stripped and the remaining collagen scaffold is used to grow new tissue. This process has been used to bioengineer heart, liver, lung, and kidney tissue. This approach holds great promise for using scaffolding from human tissue discarded during surgery and combining it with a patient’s own cells to make customized organs that would not be rejected by the immune system.

An image of two gloved hands stretching a blueish material with white lines all over it

Currently, tissue engineering plays a relatively small role in patient treatment. Supplemental bladders, small arteries, skin grafts, cartilage, and even a full trachea have been implanted in patients, but the procedures are still experimental and very costly. While more complex organ tissues like heart, lung, and liver tissue have been successfully recreated in the lab, they are a long way from being fully reproducible and ready to implant into a patient. These tissues, however, can be quite useful in research, especially in drug development. Using functioning human tissue to help screen medication candidates could speed up development and provide key tools for facilitating personalized medicine while saving money and reducing the number of animals used for research.

Research supported by NIBIB includes development of new scaffold materials and new tools to fabricate, image, monitor, and preserve engineered tissues. Some examples of research in this area are described below.

  • Controlling stem cells through their environment: For many years, scientists have searched for ways to control how stems cells develop into other cell types, in the hopes of creating new therapies. Two NIBIB researchers have grown pluripotent cells—stem cells that have the ability to turn into any kind of cell—in different types of defined spaces and found that this confinement triggered very specific gene networks that determined the ultimate fate for the cells. Most other medical research on pluripotent stem cells has focused on modifying the combination of growth solutions in which the cells are placed. The discovery that there is a biomechanical element to controlling how stem cells transform into other cell types is an important piece of the puzzle as scientists try to harness stems cells for medical uses.

Thumbnail

  • Implanting human livers in mice: NIBIB-funded researchers have engineered human liver tissue that can be implanted in a mouse. The mouse retains its own liver as well, and therefore its normal function-but the added piece of engineered human liver can metabolize drugs in the same way humans do. This allows researchers to test susceptibility to toxicity and to demonstrate species-specific responses that typically do not show up until clinical trials. Using engineered human tissue in this way could cut down on the time and cost of producing new drugs, as well as allow for critical examinations of drug-drug interactions within a human-like system.  
  • Engineering mature bone stem cells : Researchers funded by NIBIB completed the first published study that has been able to take stem cells all the way from their pluripotent state to mature bone grafts that could potentially be transplanted into a patient. Previously, investigators could only differentiate the cells to a primitive version of the tissue which was not fully functional. Additionally, the study found that when the bone was implanted in immunodeficient mice there were no abnormal growths afterwards—a problem that often occurs after implanting stem cells or bone scaffolds alone.    
  • Using lattices to help engineered tissue survive: Currently, engineered tissues that are larger than 200 microns (about twice the width of a human hair) in any dimension cannot survive because they do not have vascular networks (veins or arteries). Tissues need a good “plumbing system”—a way to bring nutrients to the cells and carry away the waste—and without a blood supply or similar mechanism, the cells quickly die. Ideally, scientists would like to be able to create engineered tissue with this plumbing system already built in.  One NIBIB funded researcher is working on a very simple and easily reproducible system to solve this problem: a modified ink-jet printer that lays down a lattice made of a sugar solution. This solution hardens and the engineered tissue (in a gel form) surrounds the lattice. Later, blood is added which easily dissolves the sugar lattice, leaving pre-formed channels to act as blood vessels.

3 MRI images of a knee before a season of NBA basketball, 3 post-season

  • New hope for the bum knee: Until now, cartilage has been very difficult, if not impossible, to repair due to the fact that cartilage lacks a blood supply to promote regeneration. There has been a 50% long-term success rate using microfracture surgery in young adults suffering from sports injuries, and little to no success in patients with widespread cartilage degeneration such as osteoarthritis. An NIBIB-funded tissue engineer has developed a biological gel that can be injected into a cartilage defect following microfracture surgery to create an environment that facilitates regeneration. However, in order for this gel to stay in place within the knee, researchers also developed a new biological adhesive that is able to bond to both the gel as well as the damaged cartilage in the knee, keeping the newly regrown cartilage in place. The gel/adhesive combo was successful in regenerating cartilage tissue following surgery in a recent clinical trial of fifteen patients, all of whom reported decreased pain at six months post-surgery. In contrast, the majority of microfracture patients, after an initial decrease in pain, returned to their original pain level within six months. This researcher worked in collaboration with another NIBIB grantee to image the patients who had undergone surgery enabling scientists to combine new, non-invasive methods to see the evolving results in real-time.  
  • Regenerating a new kidney:   The ability to regenerate a new kidney from a patient’s own cells would provide major relief for the hundreds of thousands of patients suffering from kidney disease. Experimenting on rat, pig and human kidney cells, NIDDK supported researchers broke new ground on this front by first stripping cells from a donor organ and using the remaining collagen scaffold to help guide the growth of new tissue. To regenerate viable kidney tissue, researchers seeded the kidney scaffolds with epithelial and endothelial cells. The resulting organ tissue was able to clear metabolites, reabsorb nutrients, and produce urine both in vitro and in vivo in rats. This process was previously used to bioengineer heart, liver, and lung tissue. The creation of transplantable tissue to permanently replace kidney function is a leap forward in overcoming the problems of donor organ shortages and the morbidity associated with immunosuppression in organ transplants.

download icon

Explore More

Heath Topics

  • Heart Disease
  • Kidney Disease
  • Osteoarthritis

Research Topics

  • Tissue engineering
  • Tissue Regeneration

Scientific Program Areas

  • Division of Applied Science & Technology (Bioimaging)
  • Division of Discovery Science & Technology (Bioengineering)
  • Division of Health Informatics Technologies (Informatics)
  • Division of Interdisciplinary Training (DIDT)

Inside NIBIB

  • Director's Corner
  • Funding Policies
  • NIBIB Fact Sheets
  • Press Releases

website

Institute for Stem Cell & Regenerative Medicine

Tissue engineering.

These are the faculty members that are specialized in tissue engineering.

Nancy Allbritton, MD, PhD (Bioengineering) Research in my laboratory focuses on the development of novel methods and technologies to answer fundamental questions in biology & medicine.  Much of biology & medicine is technology limited in that leaps in knowledge follow closely on the heels of new discoveries and inventions in the physical and engineering sciences; consequently, interdisciplinary groups which bridge these different disciplines are playing increasingly important roles in biomedical research.  Our lab has developed partnerships with other investigators in the areas of biology, medicine, chemistry, physics, and engineering to design, fabricate, test, and utilize new tools for biomedical and clinical research.  Collaborative projects include novel strategies to measure enzyme activity in single cells using microelectrophoresis innovations, to build organ-on-a-chips particularly intestine-on-chip, array-based methods for cell screening and sorting.  An additional focus area is the development of software and instrumentation to support these applications areas. The ultimate goal is to design and build novel technologies and then translate these technologies into the marketplace to insure their availability to the biomedical research and clinical communities to enable humans to lead healthier and more productive lives.

Cole A. DeForest, PhD  (Chemical Engineering) While the potential for biomaterial-based strategies to improve and extend the quality of human health through tissue regeneration and the treatment of disease continues to grow, the majority of current strategies rely on outdated technology initially developed and optimized for starkly different applications. Therefore, the DeForest Group seeks to integrate the governing principles of rational design with fundamental concepts from material science, synthetic chemistry, and stem cell biology to conceptualize, create, and exploit next-generation materials to address a variety of health-related problems. We are currently interested in the development of new classes of user-programmable hydrogels whose biochemical and biophysical properties can be tuned in time and space over a variety of scales. Our work relies heavily on the utilization of cytocompatible bioorthogonal chemistries, several of which can be initiated with light and thereby confined to specific sub-volumes of a sample. By recapitulating the dynamic nature of the native tissue through 4D control of the material properties, these synthetic environments are utilized to probe and better understand basic cell function as well as to engineer complex heterogeneous tissue.

David A. Dichek, MD (Medicine/Cardiology) Our work focuses on defining the molecular mechanisms that drive aortic aneurysm formation and that precipitate atherosclerotic plaque rupture (the proximal cause of most heart attacks). We are also developing a gene therapy—delivered to the blood vessel wall—that prevents and reverses atherosclerosis. Experiments are performed in a mouse model of heritable thoracic aortic aneurysms, a mouse model of atherosclerotic plaque rupture, and with advanced human plaque tissue. Our gene therapy research uses helper-dependent adenoviral vectors to test therapies in rabbit models of carotid artery and vein graft atherosclerosis.  We anticipate that insights from our work will lead to therapies that prevent or stabilize aortic aneurysms and that prevent and reverse atherosclerosis.

Benjamin Freedman, PhD  (Medicine/Nephrology) Our laboratory has developed techniques to efficiently differentiate hPSCs into kidney organoids in a reproducible, multi-well format – a prototype ‘kidney-in-a-dish’. In addition, we have generated hPSC lines carrying naturally occurring or engineered mutations relevant to human kidney diseases, such as polycystic kidney disease and nephrotic syndrome. The goal of our research is to use these new tools to model human kidney disease and identify therapeutic approaches, including kidney regeneration.

Cecilia Giachelli, PhD  (Bioengineering) My lab is interested in applying stem cell and regenerative medicine strategies to the areas of ectopic calcification, tissue engineering, biomaterials development and biocompatibility.

Ray Monnat, PhD  (Pathology and Genome Sciences) Our research focuses on human RecQ helicase deficiency syndromes such as Werner syndrome; high resolution analyses of DNA replication dynamics; and the engineering of homing endonucleases for targeted gene modification or repair in human and other animal cells.

Tracy E. Popowics, PhD (Oral Health Sciences) Our team focusses on regeneration of the periodontal ligament (PDL) that maintains tooth position and provides support during chewing. Our approach is to engineer three-dimensional (3D) periodontal constructs that mimic the native tissue structure and function. Our 3D PDL constructs include cells that are suspended in collagen matrix and recreate the living PDL tissue. Periodontal tissue loss not only includes loss of the ligament, but also the alveolar bone and cementum that anchor the periodontal ligament and hold the tooth in place. This tissue loss may occur to different degrees during an individual’s lifespan due to changes in oral care, periodontal disease, systemic disease or other health problems. This is particularly true for the aged population in which diminished oral care can contribute to persistent and recurring periodontal inflammation and tissue breakdown. Regenerating these three layers is essential to restore the structural and functional integrity of PDL and to prevent tooth loss.

Feini (Sylvia) Qu, VMD, PhD (Orthopaedics & Sports Medicine, Mechanical Engineering) The long-term goal of our research is to understand the cellular and molecular mechanisms of musculoskeletal tissue regeneration, especially with respect to the bones and connective tissues of limbs and joints, and then leverage this knowledge to regenerate lost or diseased structures using stem cells, gene editing, and biomaterials. Our lab uses the mouse digit tip, one of the few mammalian systems that exhibits true regeneration, to identify pathways that regulate tissue patterning and outgrowth after amputation. Armed with a better understanding of the cues that direct complex tissue formation in adulthood, we will develop therapeutic strategies that enhance the regeneration of limbs and joints after injury and degenerative disease in patients.

Buddy Ratner, PhD  (Bioengineering) Stem cells proliferate and differentiate in response to micromechanical cues, surface biological signals, orientational directives and chemical gradients. To control stem cell proliferation and differentiation, the Ratner lab brings 30 years experience in surface control of biology, polymer scaffold fabrication and controlled release of bioactive agents to address the challenges of directing stem cell differentiation and subsequent tissue formation.

Michael Regnier, PhD  (Bioengineering) The Regnier lab works in a highly collaborative environment to develop both cell replacement and gene therapies approaches to treat diseased and failing hearts and skeletal muscle. Cell replacement strategies include development and testing of tissue engineered constructs. Gene therapies are target and improve myofilament contractile protein function.

Jenny Robinson, PhD (Orthopaedics & Sports Medicine and Mechanical Engineering) Our primary goal is to understand what cues are needed to promote connective tissue (ligament, cartilage, fibrocartilage) regeneration after knee injuries and reduce the onset of osteoarthritis. We have a particular interest on how these cues may differ in male and female athletes. We engineer biomaterial-based environments that mimic native tissue biochemical and mechanical properties to pinpoint specific cues that are required for regeneration of the connective tissues in the knee. We aim to use this knowledge to inform the treatment options for patients with knee injuries to ensure they can get back to performance with reduced or minimal chance for the development of osteoarthritis.

Shelly Sakiyama-Elbert, PhD (Bioengineering) Our lab works on developing novel approaches to treat peripheral nerve and spinal cord injury.  We use stem cell derived neurons and glia for transplantation following injury to replace cells that are lost as well as model systems to test potential drugs to promote regeneration.  Our ultimate goal is to provide patients with new therapies that will improve functional outcomes after injury.

Mehmet Sarikaya, PhD  (Materials Science and Engineering) Our research focuses on Molecular Biomimetics in which we use combinatorial mutagenesis to select peptides with specific affinity to desired materials, use bioinformatics-based pathways to in-silico design peptides, tailor their structure and function using genetic engineering protocols, couple them with synthetic self-assembled molecular hybrids, and use them as molecular tools in practical medicine and materials technologies. Our focus at the biology/materials interface incorporates molecular biology and nanotechnology, computational biology and bioinformatics, molecular assemblers, bio-enabled nanophotonics (quantum-dot and surface-enhanced probes), and peptide-based matrices for neural, dental and soft tissue regeneration.

Drew L. Sellers, PhD  (Bioengineering) Despite possessing a resident pool of neural stem cells, the mammalian brain and spinal cord shows a limited ability to regenerate damaged tissue after traumatic injury.  Instead, injury initiates a cascade of events that direct reactive gliosis to wall off an injury with a glial scar to mitigate damage and preserve function. My current research interests explore approaches to re-engineer the stem cell niche, to utilize gene-therapy and genome editing approaches to reprogram and engineer stem cells directly, and to enhance drug delivery into the central nervous system (CNS) to drive regenerative strategies that augment functional recovery in the diseased or traumatically injured CNS.

Alec Smith, PhD (Physiology & Biophysics) My lab’s research is focused on understanding the mechanistic pathways that underpin muscle and nervous tissue development in health and disease. To achieve this, we are developing human stem cell-derived models of neuromuscular diseases, such as amyotrophic lateral sclerosis (ALS). By analyzing the behavior of these cells, we aim to better define how the causal mutation leads to the development and progression of neurodegenerative disease. Ultimately, identification of pathways critical to disease progression will provide new targets for therapeutic intervention, leading to the development of new treatments for patients suffering from these debilitating and life-threatening conditions.

Nathan Sniadecki, PhD   (Mechanical Engineering) Our mission is to understand how mechanics affects human biology and disease at the cellular level. If we can formulate how cells are guided by mechanics, then we can direct cellular response in order to engineer cells and tissue for medical applications. We specialize in the design and development of micro- and nano-tools, which allows us to probe the role of cell mechanics at a length scale appropriate to the size of cells and their proteins.

Kelly R. Stevens, PhD  (Bioengineering and Pathology) Our research is focused on developing new technologies to assemble synthetic human tissues from stem cells, and to remotely control these tissues after implantation in a patient. To do this, we use diverse tools from stem cell biology, tissue engineering, synthetic biology, microfabrication, and bioprinting. We seek to translate our work into new regenerative therapies for patients with heart and liver disease.

Thomas N. Wight, PhD  (Benaroya Research Institute) This investigator leads a research program focused on the role that the extracellular matrix molecules, proteoglycans and hyaluronan, play in regulating vascular cell type and the regulation of extracellular matrix assembly. These pathways are fundamental to understanding the growth of new blood vessels in different tissues of the body, and have potential for direct tissue regeneration applications through the use of proteoglycan genes to bioengineer vascular tissue.

Ying Zheng, PhD  (Bioengineering) Dr. Zheng’s research focuses on understanding and engineering the fundamental structure and functions in living tissue and organ systems from nanometer, micrometer to centimeter scale.

Bioengineering

Tissue engineering & regenerative medicine.

The promise of regenerative medicine is truly remarkable. Over the last two decades, significant breakthroughs in understanding within the regenerative medicine and tissue engineering fields have yielded a more intimate understanding of the functioning of human tissue. In the future, new technologies may deliver islet cells for diabetes, neural regeneration for spinal cord injuries and more substantial heart repair. In addition, as biology, bioengineering and medicine continue to converge, the regenerative medicine field may succeed in building three-dimensional organs like hearts, kidneys or livers.

Traditionally, researchers in the BioE program focus was on replacement of tissues or growing cell-based substitutes outside the body for implantation into the body. However, as the field has evolved over the last decade, researchers have broadened their approach from a focus on tissue engineering to one that includes repair and regeneration.

Projects range from creating better techniques for wound repair to peripheral nerve regeneration. In addition, BioE researchers are using advanced bioengineering methods to develop technologies that will facilitate the transfer of research in musculoskeletal biology and regenerative medicine for treatment of wounded soldiers.

phd tissue engineering and regenerative medicine

Julia Babensee

Associate professor.

Host responses to combination products, biomaterial interactions with dendritic cells, tissue engineering for rheumatoid arthritis, targeted DNA vaccine delivery, and biomaterial-applied immunology.

phd tissue engineering and regenerative medicine

Stephen Balakirsky

Principal research scientist.

Collaborative robotics

phd tissue engineering and regenerative medicine

Edward Botchwey

Tissue engineering and biomaterials, microvascular growth and remodeling, stem cell engineering The Botchwey Laboratory takes a multidisciplinary approach for improvement of tissue engineering therapies through study of microvascular remodeling, inflammation resolution and host stem cells. Our goal is development of effective new strategies to repair, replace, preserve or enhance tissue or organ function. 

phd tissue engineering and regenerative medicine

Julie Champion

Developing therapeutic protein materials, where the protein is both the drug and the delivery system Engineering proteins to control and understand protein particle self-assembly Repurposing and engineering pathogenic proteins for human therapeutics Creating materials that mimic cell-cell interactions to modulate immunological functions for various applications, including inflammation, cancer, autoimmune disease, and vaccination

phd tissue engineering and regenerative medicine

Ahmet Coskun

Assistant professor.

phd tissue engineering and regenerative medicine

Bone Tissue Engineering, Computational Design and Fabrication of Resorbable Scaffolds, Composite Scaffolds, Direct Digital Manufacturing Technologies, Large-Area Micro- and Nanoscale Photopolymerization, Patterning, and Interference Lithography Technologies, Laser Materials Processing.

phd tissue engineering and regenerative medicine

Prasad Dasi

phd tissue engineering and regenerative medicine

Michael Davis

Our laboratory focuses on various aspects of cardiac regeneration and preservation using molecular-based and biomaterials-based approaches to restoring function after cardiac injury

phd tissue engineering and regenerative medicine

Brandon Dixon

Bioengineering: lymphatics, lipid metabolism, biomechanics, biomedical optics, image processing, and tissue engineering.

phd tissue engineering and regenerative medicine

Erik Dreaden

Assistant professor of biomedical engineering, assistant professor of pediatrics.

The Dreaden Lab uses molecular engineering to impart augmented, amplified, or non-natural function to tumor therapies and immunotherapies. The overall goal of our research is to engineer molecular and nanoscale tools that can (i) improve our understanding of fundamental tumor biology and (ii) simultaneously serve as cancer therapies that are more tissue-exclusive and patient-personalized. The lab is housed on the Emory SOM campus and currently focuses on three main application areas: optically-triggered immunotherapies, combination therapies for pediatric cancers, and nanoscale cancer vaccines. Our work aims to translate these technologies into the clinic and beyond.

phd tissue engineering and regenerative medicine

Ross Ethier

My research area is biomechanics and mechanobiology with focus on: Glaucoma, including studies of aqueous humour drainage, optic nerve head biomechanics, and stem cell therapies in glaucoma; and Mechanobiology of osteoarthritis.

phd tissue engineering and regenerative medicine

Andrés García

Executive director, parker h. petit institute for bioengineering and bioscience, regents' professor, george w. woodruff school of mechanical engineering.

Biomolecular, cellular, and tissue engineering strategies to direct cell function for biomaterial and regenerative medicine applications.

phd tissue engineering and regenerative medicine

Rudy Gleason

Associate professor - joint appointment in the school of biomedical engineering.

Cardiovascular mechanics, soft tissue growth and remodeling, and tissue engineering.

phd tissue engineering and regenerative medicine

Scott Hollister

Professor and patsy and alan dorris chair in pediatric technology.

My research interests focus on image-based computational design and 3D biomaterial printing for patient specific devices and regenerative medicine, with specific interests in pediatric applications. Clinical application interests include airway reconstruction and tissue engineering, structural heart defects, craniofacial and facial plastics, orthopaedics, and gastrointestinal reconstruction. We specifically utilize patient image data as a foundation to for multiscale design of devices, reconstructive implants and regenerative medicine porous scaffolds. We are also interested in multiscale computational simulation of how devices and implants mechanically interact with patient designs, combining these simulations with experimental measures of tissue mechanics. We then transfer these designs to both laser sintering and nozzle based platforms to build devices from a wide range of biomaterials.  Subsequently, we are interested in combining these 3D printed biomaterial platforms with biologics for patient specific regenerative medicine solutions to tissue reconstruction.

phd tissue engineering and regenerative medicine

Stem cell, tissue engineering, mitochondrial engineering and bioenergetics, aging, muscular dystrophy, and neuromuscular disease.

phd tissue engineering and regenerative medicine

Michelle LaPlaca

Traumatic brain and spinal cord injury, Neural tissue engineering, Injury biomechanics, Neural interfacing, and Cognitive impairment associated with brain injury and aging.

phd tissue engineering and regenerative medicine

Joe Le Doux

I am a co-founder of STELAR: the Studio for Transforming Engineering Learning and Research. STELAR is a one-of-its-kind learning sciences research group embedded within the Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory University. Our goal is to contribute to changing the engineering workforce so that it is diverse and inclusive and fully leverages all of our national talent to effectively address the complex problems of the 21st century. We aspire to graduate students who have agency as inclusive engineers who understand the need for a diverse and inclusive engineering workforce and have the knowledge, skills, and dispositions to create, and thrive within, inclusive study and work environments.

phd tissue engineering and regenerative medicine

Shuming Nie

The wallace h. coulter distinguished faculty chair in biomedical engineering & professor.

Our research is in the areas of biomolecular engineering and nanotechnology, with a focus on bioconjugated nanoparticles for cancer molecular imaging, molecular profiling, pharmacogenomics, and targeted therapy.

phd tissue engineering and regenerative medicine

John Oshinski

Dr. Oshinski is well known for his collaborative efforts between Emory and Georgia Tech's Department of Biomedical Engineering, along with his dedication to advancing the technologies of MR imaging. One area of concentration is the development of Cardiovascular MRI for clinical and basic science applications. Dr. Oshinski has worked on development of the contrast-enhanced MRA and phase-contrast MR for rapid assessment of the aorta and the peripheral runoff vessels. He also Implemented SSFP cine imaging for rapid breath-hold assessment of cardiac function, IR recovery sequences for myocardial perfusion imaging, and creating a protocol for using MR coronary angiography to diagnose the proximal course of the coronary arteries.

phd tissue engineering and regenerative medicine

Muralidhar Padala

Associate professor of surgery & bme.

phd tissue engineering and regenerative medicine

Sung-JIn Park

phd tissue engineering and regenerative medicine

Felipe Quiroz

phd tissue engineering and regenerative medicine

Philip Santangelo

RNA regulation, single molecule imaging, and RNA virus pathogenesis

phd tissue engineering and regenerative medicine

Gregory Sawicki

phd tissue engineering and regenerative medicine

Todd Streelman

Professor and director.

Evolution and development of functional systems.

phd tissue engineering and regenerative medicine

Todd Sulchek

Bioengineering and Microelectromechanical Systems: Atomic force microscopy, pathogen adhesion and endocytosis, cell biomechanics, single molecule biophysics, drug delivery and targeting, cell membrane mimetics, and biosensors.

phd tissue engineering and regenerative medicine

Shuichi Takayama

The MNM Biotech Lab uses engineering expertise to assist life scientists in the study, diagnosis, and treatment of human disease. By developing better models of the body, we help advance drug discovery, increase understanding of the mechanisms of disease, and develop clinical treatments.

phd tissue engineering and regenerative medicine

Vascular biology with an emphasis on the role of vascular inflammation in the pathogenesis of hypertension, diabetes and atherosclerosis.

phd tissue engineering and regenerative medicine

Johnna Temenoff

Carol ann and david d. flanagan professor.

Development of novel polymeric biomaterials, regeneration of tendon/ligament, and protein delivery for orthopaedic tissue engineering.

phd tissue engineering and regenerative medicine

Randy Trumbower

Director of research & assistant professor.

Spinal cord injury, stroke, intermittent hypoxia, rehabilitation, motor control, and whole limb mechanics.

phd tissue engineering and regenerative medicine

Our research focuses on applying systems analysis approaches and engineering tools to identify novel clinical therapeutic targets for complex diseases. It is challenging to develop new treatments for these diseases, such as Alzheimer's disease (AD) and Traumatic Brain Injury (TBI), because they do not have a single genetic cause and they simultaneously present broad physiologic changes. By combining novel engineered in vitro platforms, mouse models, and multivariate computational systems analysis, we will be able to 1) capture a holistic systems-level understanding of complex diseases, and 2) isolate specific mechanisms driving disease. The ultimate goal of our laboratory is to use these tools to identify new mechanisms driving disease onset and progression that will translate to effective therapeutic strategies.

phd tissue engineering and regenerative medicine

Disruptive technologies enabled by nanoscale materials and devices will define our future in the same way that microtechnology has done over the past several decades. Our current research centers on the design and synthesis of novel nanomaterials for a broad range of applications, including nanomedicine, regenerative medicine, cancer theranostics, tissue engineering, controlled release, catalysis, and fuel cell technology. We are design and synthesize/fabricate novel nanomaterials that could serve as: 1) theranostic agents for cancer and other diseases; 2) multifucntional probes for cellular tracking; 3) smart capsules for site-specific, on-demand delivery; and 4) scaffolds for the repair or regeneration of tissues.

phd tissue engineering and regenerative medicine

Shawn Hochman

Professor and interim chair,.

The spinal cord is the gateway for information transfer between body and brain but is not simply a conduit. Within its central gray matter lie millions of neurons that integrate and coordinate complex sensory, motor and autonomic events. Spinal cord injuries can permanently sever descending command pathways and produce paralysis. After some time the injured cord often become hyper-responsive and may result in spasticity (hyperreflexia), autonomic dysfunction and devastating chronic pain syndromes. Brain modulatory systems regulate spinal cord excitability and it may be disruption of their actions that are primarily responsible for the hyperactivity seen after cord injury or in diseases states. A major goal of our lab is to understand how the major brain monoamine modulatory transmitters (serotonin, dopamine, and nor-adrenaline) regulate cord function. These transmitters have been linked to activation of the spinal cord circuitry generating locomotion, control of autonomic NS function, as well as the potent inhibition of spinal cord pain systems. Dysfunction is spinal dopamine is also strongly implicated in the emergence of Restless Legs Syndrome (RLS). We are also very interested in additional mechanisms that limit body sensations.

Current research in the lab focuses on:
 1. Non-classical control of body sensations. 
2. Restless Legs Syndrome (RLS) and spinal cord dopamine.
 3. Plasticity of spinal cord function after injury.
 4. Regulation of spinal cord locomotor activity.
 5. Unusual modulatory mechanisms controlling spinal cord function.

phd tissue engineering and regenerative medicine

Song Ih Ahn

Neuroengineering, Tissue Engineering, Organ-on-chips, Microfluidics, Drug Delivery, Cell Mechanics

phd tissue engineering and regenerative medicine

Alexander Beach

Retinal research, tissue regeneration

phd tissue engineering and regenerative medicine

Nicholas Beskid

phd tissue engineering and regenerative medicine

Fredrick Bulondo

Ms. in bioengineering.

ImmunoEngineering, Biomaterials, Bioinstrumentation

phd tissue engineering and regenerative medicine

Thomas Burkholder

Associate professor,.

Exploration of the coordination of skeletal muscle structure and function. This work has two thrusts: understanding the mechanism by which mechanical signals alter muscle structure and understanding the functional demands on muscle

phd tissue engineering and regenerative medicine

Olivia Burnsed

Olivia is interested in the effects of both the structural and biochemical cues provided by the extracellular matrix on modulating cell phenotype and inflammation. She is currently working to create microcarriers to expand chondrocytes while maintaining their phenotype and direct the differentiation of stem cells. Olivia is also investigating the use of MSCs in combination with human amniotic membrane for osteoarthritis (OA) treatments in the context of modulating inflammation and OA progression.

phd tissue engineering and regenerative medicine

Seleipiri Charles

phd tissue engineering and regenerative medicine

Albert Cheng

phd tissue engineering and regenerative medicine

Ricardo Cruz-Acuña

Biomaterials and Regenerative Medicine

phd tissue engineering and regenerative medicine

Gilad Doron

phd tissue engineering and regenerative medicine

Aaron Enten

Phd, mba, managing innovation and technology commercialization.

Medical Device Innovation / Clinically Translational Technology

phd tissue engineering and regenerative medicine

Shaun Eshraghi

phd tissue engineering and regenerative medicine

Jose Garcia

Biomaterials, Tissue Engineering, Therapies for Increased Vascularization, Stem Cell Therapies

phd tissue engineering and regenerative medicine

Michael Griffin

Microfluidics, Arterial Thrombosis, Platelets, vWF

phd tissue engineering and regenerative medicine

Robert Guldberg

Adjunct professor.

Guldberg’s research interests focus on musculoskeletal growth and development, functional regeneration following traumatic injury, and degenerative diseases, including skeletal fragility and osteoarthritis. His research is supported by the NIH, NSF, DoD, and several biotechnology companies and has resulted in over 150 book chapters and publications. Guldberg is a Fellow of the American Institute for Medical and Biological Engineering (AIMBE) and holds several national leadership positions.

phd tissue engineering and regenerative medicine

Elijah Holland

Bioengineering.

phd tissue engineering and regenerative medicine

Michael Hunckler

phd tissue engineering and regenerative medicine

Kelly Hyland

phd tissue engineering and regenerative medicine

Timothy Kassis

Lymphatic biology, lipid uptake and metabolism, obesity, lymphatic imaging, in situ imaging, image processing, vessel biomechanics, microfluidics, machine learning.

phd tissue engineering and regenerative medicine

Myeongsoo Kim

phd tissue engineering and regenerative medicine

Johnathon Long

phd tissue engineering and regenerative medicine

Lina Maria Mancipe Castro

Drug delivery, Tissue Engineering & Regenerative Medicine, Biomaterials    

phd tissue engineering and regenerative medicine

Karen Martin

phd tissue engineering and regenerative medicine

Larry McIntire

Department chair emeritus.

Bioengineering aspects of vascular biology, thrombosis, inflammatory response, and infectious disease Cellular engineering

phd tissue engineering and regenerative medicine

Alyssa Montalbine

phd tissue engineering and regenerative medicine

Leandro Moretti

phd tissue engineering and regenerative medicine

Adriana Mulero-Russe

My research is focused on the growth, expansion, and differentiation of human pluripotent stem cells (hPSC) into human intestinal organoids (HIOs) using an engineered synthetic polyethylene glycol (PEG) hydrogel platform.

phd tissue engineering and regenerative medicine

Andre Norfleet

Systems Biology Computational Modeling, Personalized Medicine, Immunotherapeutics

phd tissue engineering and regenerative medicine

Michelle Quizon

phd tissue engineering and regenerative medicine

Mohammad Razavi

phd tissue engineering and regenerative medicine

Rachel Ringquist

Immunology, organ-on-chip, immunotherapies

phd tissue engineering and regenerative medicine

Stephen Robinson

In-vitro models of disease

Apoorva-Salimath (Kalasuramath)

Apoorva Salimath (Kalasuramath)

phd tissue engineering and regenerative medicine

Angel Santiago-Lopez

phd tissue engineering and regenerative medicine

Rebecca Schneider

phd tissue engineering and regenerative medicine

Farbod Sedaghati

Cardiovascular Mechanics Soft tissue growth & remodeling Tissue engineering

phd tissue engineering and regenerative medicine

Generation and regeneration of cells into hepatopancreatic lineages, Bone Morphogenetic Protein (BMP) 2b signaling, Zebrafish genetics, Morphogenesis and organogenesis

phd tissue engineering and regenerative medicine

Sangeetha Srinivasan

Biomaterials, Immunology, Tissue Engineering, Polymers

phd tissue engineering and regenerative medicine

Brennan Torstrick

Graduate student.

   

phd tissue engineering and regenerative medicine

Vishwa Vasani

Microfluidics, Organoids, Microphysiological Systems

phd tissue engineering and regenerative medicine

Hannah Viola

phd tissue engineering and regenerative medicine

Muhammad Minhal Yusufali

Ph.d. student + r&d engineer.

Neural Engineering + Biomaterials

phd tissue engineering and regenerative medicine

Nicholas Zhang

phd tissue engineering and regenerative medicine

Daniel Zhang

Regenerative medicine, aging, longevity, stem cell engineering, tissue engineering, neuroengineering

phd tissue engineering and regenerative medicine

Luiza DaMotta

phd tissue engineering and regenerative medicine

Justin Damon

phd tissue engineering and regenerative medicine

Laura Hansen

Assistant professor of medicine.

phd tissue engineering and regenerative medicine

Anant Paravastu

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • View all journals
  • Explore content
  • About the journal
  • Publish with us
  • Sign up for alerts
  • Review Article
  • Published: 19 December 2023

Single-cell transcriptomics in tissue engineering and regenerative medicine

  • Anna Ruta 1   na1 ,
  • Kavita Krishnan   ORCID: orcid.org/0000-0003-1345-0249 1   na1 &
  • Jennifer H. Elisseeff   ORCID: orcid.org/0000-0002-5066-1996 1  

Nature Reviews Bioengineering volume  2 ,  pages 101–119 ( 2024 ) Cite this article

1288 Accesses

25 Altmetric

Metrics details

  • Computational biology and bioinformatics
  • Regenerative medicine
  • Tissue engineering

Regenerative medicine and tissue engineering aim to promote functional rebuilding of damaged tissue. Comprehensively profiling cell identity, function and interaction in healthy tissues, as well as understanding how these change upon tissue disruption, such as that caused by injury, ageing or infection, is foundational to advancing tissue engineering and regenerative therapeutics. Tissue injury response is a highly dynamic process driven by complex interactions between immune and stromal cell populations, with dysregulation leading to deleterious fibrosis and chronic inflammation. Advances in single-cell RNA sequencing now allow in-depth mapping of the complex cellular response to injury and biomaterial implantation. In this Review, we first describe the fundamentals of sequencing and computational methods for the generation and analysis of high-dimensional single-cell RNA sequencing data sets. We then highlight how these methods can be applied to study tissue injury responses and guide the rational design of biomaterials and regenerative therapeutics.

Single-cell RNA sequencing (scRNA-seq) affords unprecedented resolution in profiling cellular transcriptomics by simultaneously detecting the expression of thousands of genes on an individual cell basis.

Tissue engineers can leverage scRNA-seq to comprehensively map healthy and perturbed (such as injured or diseased) tissue environments and explore cellular heterogeneity, gene expression shifts, differentiation trajectories and interaction networks.

Insights gained by scRNA-seq profiling of biological systems can be leveraged to guide the rational design of new biomaterials and regenerative therapeutics.

scRNA-seq can be used to characterize the host response to implanted engineered constructs or regenerative therapeutics and discern mechanisms of action (regenerative or fibrotic).

Sharing of data sets in public repositories, development of large-scale atlases and formation of dedicated consortiums promote low-cost accessibility, increase diversity and maximize exploration of generated scRNA-seq data sets.

Interdisciplinary teams of basic scientists, bioinformaticians, tissue engineers and clinicians should work together to connect computational approaches to outstanding biological questions, driving innovation of new regenerative therapeutics.

This is a preview of subscription content, access via your institution

Access options

Subscribe to this journal

Receive 12 digital issues and online access to articles

92,52 € per year

only 7,71 € per issue

Rent or buy this article

Prices vary by article type

Prices may be subject to local taxes which are calculated during checkout

phd tissue engineering and regenerative medicine

Similar content being viewed by others

phd tissue engineering and regenerative medicine

Computational reconstruction of the signalling networks surrounding implanted biomaterials from single-cell transcriptomics

Christopher Cherry, David R. Maestas, … Jennifer H. Elisseeff

phd tissue engineering and regenerative medicine

Profiling joint tissues at single-cell resolution: advances and insights

Akshay Pandey & Nidhi Bhutani

phd tissue engineering and regenerative medicine

Large-scale integration of single-cell transcriptomic data captures transitional progenitor states in mouse skeletal muscle regeneration

David W. McKellar, Lauren D. Walter, … Benjamin D. Cosgrove

Slyper, M. et al. A single-cell and single-nucleus RNA-Seq toolbox for fresh and frozen human tumors. Nat. Med. 26 , 792–802 (2020).

Article   CAS   PubMed   PubMed Central   Google Scholar  

Wu, H., Kirita, Y., Donnelly, E. L. & Humphreys, B. D. Advantages of single-nucleus over single-cell RNA sequencing of adult kidney: rare cell types and novel cell states revealed in fibrosis. J. Am. Soc. Nephrol. 30 , 23–32 (2019).

Article   CAS   PubMed   Google Scholar  

Grindberg, R. V. et al. RNA-sequencing from single nuclei. Proc. Natl Acad. Sci. USA 110 , 19802–19807 (2013).

Article   CAS   PubMed   PubMed Central   ADS   Google Scholar  

Autengruber, A., Gereke, M., Hansen, G., Hennig, C. & Bruder, D. Impact of enzymatic tissue disintegration on the level of surface molecule expression and immune cell function. Eur. J. Microbiol. Immunol. 2 , 112–120 (2012).

Article   CAS   Google Scholar  

Reichard, A. & Asosingh, K. Best practices for preparing a single cell suspension from solid tissues for flow cytometry. Cytometry A 95 , 219–226 (2019).

van den Brink, S. C. et al. Single-cell sequencing reveals dissociation-induced gene expression in tissue subpopulations. Nat. Methods 14 , 935–936 (2017).

Article   PubMed   Google Scholar  

Denisenko, E. et al. Systematic assessment of tissue dissociation and storage biases in single-cell and single-nucleus RNA-seq workflows. Genome Biol. 21 , 130 (2020). This study compares gene expression and cellular composition of single-cell and single-nucleus suspensions generated implementing different dissociation protocols and different storage methods to identify potential artefacts and biases.

Sutermaster, B. A. & Darling, E. M. Considerations for high-yield, high-throughput cell enrichment: fluorescence versus magnetic sorting. Sci. Rep. 9 , 227 (2019).

Article   PubMed   PubMed Central   ADS   Google Scholar  

Stoeckius, M. et al. Cell Hashing with barcoded antibodies enables multiplexing and doublet detection for single cell genomics. Genome Biol. 19 , 224 (2018).

Gehring, J., Hwee Park, J., Chen, S., Thomson, M. & Pachter, L. Highly multiplexed single-cell RNA-seq by DNA oligonucleotide tagging of cellular proteins. Nat. Biotechnol. 38 , 35–38 (2020).

Srivatsan, S. R. et al. Massively multiplex chemical transcriptomics at single-cell resolution. Science 367 , 45–51 (2020).

Article   CAS   PubMed   ADS   Google Scholar  

Ding, J. et al. Systematic comparison of single-cell and single-nucleus RNA-sequencing methods. Nat. Biotechnol. 38 , 737–746 (2020).

Mereu, E. et al. Benchmarking single-cell RNA-sequencing protocols for cell atlas projects. Nat. Biotechnol. 38 , 747–755 (2020).

Zhao, S. & Zhang, B. A comprehensive evaluation of ensembl, RefSeq, and UCSC annotations in the context of RNA-seq read mapping and gene quantification. BMC Genomics 16 , 97 (2015).

Article   PubMed   PubMed Central   Google Scholar  

Cunningham, F. et al. Ensembl 2022. Nucleic Acids Res. 50 , D988–D995 (2022).

O’Leary, N. A. et al. Reference sequence (RefSeq) database at NCBI: current status, taxonomic expansion, and functional annotation. Nucleic Acids Res. 44 , D733–D745 (2016).

Nassar, L. R. et al. The UCSC Genome Browser database: 2023 update. Nucleic Acids Res. 51 , D1188–D1195 (2023).

Brüning, R. S., Tombor, L., Schulz, M. H., Dimmeler, S. & John, D. Comparative analysis of common alignment tools for single-cell RNA sequencing. Gigascience 11 , giac001 (2022).

10x Genomics. Cell Ranger. 10x Genomics https://support.10xgenomics.com/single-cell-vdj/software/pipelines/latest/what-is-cell-ranger (2020).

Kaminow, B., Yunusov, D. & Dobin, A. STARsolo: accurate, fast and versatile mapping/quantification of single-cell and single-nucleus RNA-seq data. Preprint at bioRxiv https://doi.org/10.1101/2021.05.05.442755 (2021).

Slovin, S. et al. Single-cell RNA sequencing analysis: a step-by-step overview. Methods Mol. Biol. 2284 , 343–365 (2021). This review covers the main considerations on the laboratory and computational sides of scRNA-seq data generation and analysis with pipelines for data processing.

McGinnis, C. S., Murrow, L. M. & Gartner, Z. J. DoubletFinder: doublet detection in single-cell RNA sequencing data using artificial nearest neighbors. Cell Syst. 8 , 329–337.e4 (2019).

Xi, N. M. & Li, J. J. Benchmarking computational doublet-detection methods for single-cell RNA sequencing data. Cell Syst. 12 , 176–194.e6 (2021).

Young, M. D. & Behjati, S. SoupX removes ambient RNA contamination from droplet-based single-cell RNA sequencing data. Gigascience 9 , giaa151 (2020).

Lytal, N., Ran, D. & An, L. Normalization methods on single-cell RNA-seq data: an empirical survey. Front. Genet. 11 , 41 (2020).

Chen, W. et al. A comparison of methods accounting for batch effects in differential expression analysis of UMI count based single cell RNA sequencing. Comput. Struct. Biotechnol. J. 18 , 861–873 (2020).

Tran, H. T. N. et al. A benchmark of batch-effect correction methods for single-cell RNA sequencing data. Genome Biol. 21 , 12 (2020).

Luecken, M. D. et al. Benchmarking atlas-level data integration in single-cell genomics. Nat. Methods 19 , 41–50 (2022).

Kobak, D. & Berens, P. The art of using t-SNE for single-cell transcriptomics. Nat. Commun. 10 , 5416 (2019).

Becht, E. et al. Dimensionality reduction for visualizing single-cell data using UMAP. Nat. Biotechnol. 37 , 38–44 (2019).

Moon, K. R. et al. Visualizing structure and transitions in high-dimensional biological data. Nat. Biotechnol. 37 , 1482–1492 (2019).

Xiang, R. et al. A comparison for dimensionality reduction methods of single-cell RNA-seq data. Front. Genet. 12 , 646936 (2021).

Duò, A., Robinson, M. D. & Soneson, C. A systematic performance evaluation of clustering methods for single-cell RNA-seq data. F1000Research 7 , 1141 (2018).

Kiselev, V. Y., Andrews, T. S. & Hemberg, M. Challenges in unsupervised clustering of single-cell RNA-seq data. Nat. Rev. Genet. 20 , 273–282 (2019).

Pasquini, G., Rojo Arias, J. E., Schäfer, P. & Busskamp, V. Automated methods for cell type annotation on scRNA-seq data. Comput. Struct. Biotechnol. J. 19 , 961–969 (2021).

Huang, Q., Liu, Y., Du, Y. & Garmire, L. X. Evaluation of cell type annotation R packages on single-cell RNA-seq data. Genomics Proteomics Bioinformatics 19 , 267–281 (2021).

Yi, H., Plotkin, A. & Stanley, N. Benchmarking differential abundance methods for finding condition-specific prototypical cells in multi-sample single-cell datasets. Preprint at bioRxiv https://doi.org/10.1101/2023.02.24.529894 (2023).

Robinson, M. D., McCarthy, D. J. & Smyth, G. K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26 , 139–140 (2010).

Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15 , 550 (2014).

Wang, T., Li, B., Nelson, C. E. & Nabavi, S. Comparative analysis of differential gene expression analysis tools for single-cell RNA sequencing data. BMC Bioinformatics   20 , 40 (2019).

Liberzon, A. et al. Molecular signatures database (MSigDB) 3.0. Bioinformatics 27 , 1739–1740 (2011).

Culhane, A. C. et al. GeneSigDB: a manually curated database and resource for analysis of gene expression signatures. Nucleic Acids Res. 40 , D1060–D1066 (2012).

Cao, J. et al. The single-cell transcriptional landscape of mammalian organogenesis. Nature 566 , 496–502 (2019).

Deconinck, L., Cannoodt, R., Saelens, W., Deplancke, B. & Saeys, Y. Recent advances in trajectory inference from single-cell omics data. Curr. Opin. Syst. Biol. 27 , 100344 (2021).

Bergen, V., Lange, M., Peidli, S., Wolf, F. A. & Theis, F. J. Generalizing RNA velocity to transient cell states through dynamical modeling. Nat. Biotechnol. 38 , 1408–1414 (2020).

Gorin, G., Fang, M., Chari, T. & Pachter, L. RNA velocity unraveled. PLoS Comput. Biol. 18 , e1010492 (2022).

Weiler, P., Van den Berge, K., Street, K. & Tiberi, S. in Single Cell Transcriptomics Methods and Protocols (eds Calogero, R. A. & Benes, V.) 269–292 (Springer, 2022).

Alemany, A., Florescu, M., Baron, C. S., Peterson-Maduro, J. & Van Oudenaarden, A. Whole-organism clone tracing using single-cell sequencing. Nature 556 , 108–112 (2018).

Shlyakhtina, Y., Bloechl, B. & Portal, M. M. BdLT-Seq as a barcode decay-based method to unravel lineage-linked transcriptome plasticity. Nat. Commun. 14 , 1085 (2023).

Pratapa, A., Jalihal, A. P., Law, J. N., Bharadwaj, A. & Murali, T. M. Benchmarking algorithms for gene regulatory network inference from single-cell transcriptomic data. Nat. Methods 17 , 147–154 (2020).

Chen, S. & Mar, J. C. Evaluating methods of inferring gene regulatory networks highlights their lack of performance for single cell gene expression data. BMC Bioinformatics   19 , 232 (2018).

Efremova, M., Vento-Tormo, M., Teichmann, S. A. & Vento-Tormo, R. CellPhoneDB: inferring cell–cell communication from combined expression of multi-subunit ligand–receptor complexes. Nat. Protoc. 15 , 1484–1506 (2020).

Browaeys, R., Saelens, W. & Saeys, Y. NicheNet: modeling intercellular communication by linking ligands to target genes. Nat. Methods 17 , 159–162 (2020).

Shao, X., Lu, X., Liao, J., Chen, H. & Fan, X. New avenues for systematically inferring cell-cell communication: through single-cell transcriptomics data. Protein Cell 11 , 866–880 (2020).

Almet, A. A., Cang, Z., Jin, S. & Nie, Q. The landscape of cell–cell communication through single-cell transcriptomics. Curr. Opin. Syst. Biol. 26 , 12–23 (2021).

Fischer, D. S., Schaar, A. C. & Theis, F. J. Modeling intercellular communication in tissues using spatial graphs of cells. Nat. Biotechnol. 41 , 332–336 (2023).

Jerby-Arnon, L. & Regev, A. DIALOGUE maps multicellular programs in tissue from single-cell or spatial transcriptomics data. Nat. Biotechnol. 40 , 1467–1477 (2022).

Clark, I. C. et al. Barcoded viral tracing of single-cell interactions in central nervous system inflammation. Science 372 , eabf1230 (2021).

Giladi, A. et al. Dissecting cellular crosstalk by sequencing physically interacting cells. Nat. Biotechnol. 38 , 629–637 (2020).

Stickels, R. R. et al. Highly sensitive spatial transcriptomics at near-cellular resolution with Slide-seqV2. Nat. Biotechnol. 39 , 313–319 (2021).

Wei, X. et al. Single-cell Stereo-seq reveals induced progenitor cells involved in axolotl brain regeneration. Science 377 , eabp9444 (2022). This article reports the development of a high-resolution, single-cell spatial transcriptomics approach Stereo-seq to profile developmental and post-injury regenerative neurogenesis in axolotl telencephalon.

Moffitt, J. R. et al. High-throughput single-cell gene-expression profiling with multiplexed error-robust fluorescence in situ hybridization. Proc. Natl Acad. Sci. USA 113 , 11046–11051 (2016).

Alon, S. et al. Expansion sequencing: spatially precise in situ transcriptomics in intact biological systems. Science 371 , eaax2656 (2021).

Lee, J., Yoo, M. & Choi, J. Recent advances in spatially resolved transcriptomics: challenges and opportunities. BMB Rep. 55 , 113–124 (2022).

Williams, C. G., Lee, H. J., Asatsuma, T., Vento-Tormo, R. & Haque, A. An introduction to spatial transcriptomics for biomedical research. Genome Med. 14 , 68 (2022).

Thomas, S. M., Ackert-Bicknell, C. L., Zuscik, M. J. & Payne, K. A. Understanding the Transcriptomic Landscape to Drive New Innovations in Musculoskeletal Regenerative Medicine. Curr. Osteoporos. Rep. 20 , 141–152 (2022).

Rai, M. F. et al. Single cell omics for musculoskeletal research. Curr. Osteoporos. Rep. 19 , 131–140 (2021).

Sarmiento, P. & Little, D. Tendon and multiomics: advantages, advances, and opportunities. NPJ Regen. Med. 6 , 61 (2021).

Baldwin, M. J., Cribbs, A. P., Guilak, F. & Snelling, S. J. B. Mapping the musculoskeletal system one cell at a time. Nat. Rev. Rheumatol. 17 , 247–248 (2021).

Paik, D. T., Cho, S., Tian, L., Chang, H. Y. & Wu, J. C. Single-cell RNA sequencing in cardiovascular development, disease and medicine. Nat. Rev. Cardiol. 17 , 457–473 (2020).

Chaudhry, F. et al. Single-cell RNA sequencing of the cardiovascular system: new looks for old diseases. Front. Cardiovasc. Med. 6 , 173 (2019).

Schreibing, F. & Kramann, R. Mapping the human kidney using single-cell genomics. Nat. Rev. Nephrol. 18 , 347–360 (2022).

Clark, A. R. & Greka, A. The power of one: advances in single-cell genomics in the kidney. Nat. Rev. Nephrol. 16 , 73–74 (2020).

Alexander, M. J., Budinger, G. R. S. & Reyfman, P. A. Breathing fresh air into respiratory research with single-cell RNA sequencing. Eur. Resp. Rev. 29 , 200060 (2020).

Article   Google Scholar  

Theocharidis, G., Tekkela, S., Veves, A., McGrath, J. A. & Onoufriadis, A. Single‐cell transcriptomics in human skin research: available technologies, technical considerations and disease applications. Exp. Dermatol. 31 , 655–673 (2022).

Dubois, A., Gopee, N., Olabi, B. & Haniffa, M. Defining the skin cellular community using single-cell genomics to advance precision medicine. J. Invest. Dermatol. 141 , 255–264 (2021).

Colonna, M. & Brioschi, S. Neuroinflammation and neurodegeneration in human brain at single-cell resolution. Nat. Rev. Immunol. 20 , 81–82 (2020).

Cao, Y., Zhu, S., Yu, B. & Yao, C. Single‐cell RNA sequencing for traumatic spinal cord injury. FASEB J. 36 , e22656 (2022).

Guerrero-Juarez, C. F. et al. Single-cell analysis reveals fibroblast heterogeneity and myeloid-derived adipocyte progenitors in murine skin wounds. Nat. Commun. 10 , 650 (2019).

Oprescu, S. N., Yue, F., Qiu, J., Brito, L. F. & Kuang, S. Temporal dynamics and heterogeneity of cell populations during skeletal muscle regeneration. iScience 23 , 100993 (2020). This study reports the use of scRNA-seq and cell lineage tracing to profile the kinetics and transcriptional dynamics of skeletal muscle regeneration, considering both the stromal and immune cell compartments in various tissue injury phases (uninjured to 21 days post-injury).

Farbehi, N. et al. Single-cell expression profiling reveals dynamic flux of cardiac stromal, vascular and immune cells in health and injury. eLife 8 , e43882 (2019).

Dick, S. A. et al. Self-renewing resident cardiac macrophages limit adverse remodeling following myocardial infarction. Nat. Immunol. 20 , 29–39 (2019).

Vafadarnejad, E. et al. Dynamics of cardiac neutrophil diversity in murine myocardial infarction. Circ. Res. 127 , e232–e249 (2020).

Ruiz-Villalba, A. et al. Single-cell RNA sequencing analysis reveals a crucial role for CTHRC1 (collagen triple helix repeat containing 1) cardiac fibroblasts after myocardial infarction. Circulation 142 , 1831–1847 (2020).

Kirita, Y., Wu, H., Uchimura, K., Wilson, P. C. & Humphreys, B. D. Cell profiling of mouse acute kidney injury reveals conserved cellular responses to injury. Proc. Natl Acad. Sci. USA 117 , 15874–15883 (2020).

Abbasi, S. et al. Distinct regulatory programs control the latent regenerative potential of dermal fibroblasts during wound healing. Cell Stem Cell 27 , 396–412.e6 (2020).

Lin, Y. et al. Single-cell RNA-seq of UVB-radiated skin reveals landscape of photoaging-related inflammation and protection by vitamin D. Gene 831 , 146563 (2022).

Foster, D. S. et al. Integrated spatial multiomics reveals fibroblast fate during tissue repair. Proc. Natl Acad. Sci. USA 118 , e2110025118 (2021). This article reports the use of multi-modal integration (scRNA-seq, scATAC-seq and spatial transcriptomics) to map the kinetics of splinted excisional skin injury to compare cell populations at various wound locations (inner or outer) over the wound-healing time course (uninjured to 14 days post-injury).

Croft, A. P. et al. Distinct fibroblast subsets drive inflammation and damage in arthritis. Nature 570 , 246–251 (2019).

Zhang, F. et al. Defining inflammatory cell states in rheumatoid arthritis joint synovial tissues by integrating single-cell transcriptomics and mass cytometry. Nat. Immunol. 20 , 928–942 (2019).

Alivernini, S. et al. Distinct synovial tissue macrophage subsets regulate inflammation and remission in rheumatoid arthritis. Nat. Med. 26 , 1295–1306 (2020).

Wei, K. et al. Notch signalling drives synovial fibroblast identity and arthritis pathology. Nature 582 , 259–264 (2020).

Knights, A. J. et al. Synovial fibroblasts assume distinct functional identities and secrete R-spondin 2 in osteoarthritis. Ann. Rheum. Dis. 82 , 272–282 (2023).

Habermann, A. C. et al. Single-cell RNA sequencing reveals profibrotic roles of distinct epithelial and mesenchymal lineages in pulmonary fibrosis. Sci. Adv. 6 , eaba1972 (2020).

Aran, D. et al. Reference-based analysis of lung single-cell sequencing reveals a transitional profibrotic macrophage. Nat. Immunol. 20 , 163–172 (2019).

Zhao, C. Q. et al. Heterogeneity of T cells and macrophages in chlorine-induced acute lung injury in mice using single-cell RNA sequencing. Inhal. Toxicol. 34 , 399–411 (2022).

Peyser, R. et al. Defining the activated fibroblast population in lung fibrosis using single-cell sequencing. Am. J. Respir. Cell Mol. Biol. 61 , 74–85 (2019).

Milich, L. M. et al. Single-cell analysis of the cellular heterogeneity and interactions in the injured mouse spinal cord. J. Exp. Med. 218 , e20210040 (2021).

Hammond, T. R. et al. Single-cell RNA sequencing of microglia throughout the mouse lifespan and in the injured brain reveals complex cell-state changes. Immunity 50 , 253–271.e6 (2019).

Jordão, M. J. C. et al. Single-cell profiling identifies myeloid cell subsets with distinct fates during neuroinflammation. Science 363 , eaat7554 (2019).

Dell’Orso, S. et al. Single cell analysis of adult mouse skeletal muscle stem cells in homeostatic and regenerative conditions. Development 146 , dev174177 (2019).

Reyes, N. S. et al. Sentinel p16 INK4a+ cells in the basement membrane form a reparative niche in the lung. Science 378 , 192–201 (2022).

Leigh, N. D. et al. Transcriptomic landscape of the blastema niche in regenerating adult axolotl limbs at single-cell resolution. Nat. Commun. 9 , 5153 (2018).

Gerber, T. et al. Single-cell analysis uncovers convergence of cell identities during axolotl limb regeneration. Science 362 , eaaq0681 (2018).

Benhar, I. et al. Temporal single-cell atlas of non-neuronal retinal cells reveals dynamic, coordinated multicellular responses to central nervous system injury. Nat. Immunol. 24 , 700–713 (2023).

Lust, K. et al. Single-cell analyses of axolotl telencephalon organization, neurogenesis, and regeneration. Science 377 , eabp9262 (2022).

Armingol, E., Officer, A., Harismendy, O. & Lewis, N. E. Deciphering cell–cell interactions and communication from gene expression. Nat. Rev. Genet. 22 , 71–88 (2021).

De Micheli, A. J. et al. Single-cell analysis of the muscle stem cell hierarchy identifies heterotypic communication signals involved in skeletal muscle regeneration. Cell Rep. 30 , 3583–3595.e5 (2020).

Linnerbauer, M. et al. Intranasal delivery of a small-molecule ErbB inhibitor promotes recovery from acute and late-stage CNS inflammation. JCI Insight 7 , e154824 (2022).

Theocharidis, G. et al. Single cell transcriptomic landscape of diabetic foot ulcers. Nat. Commun. 13 , 181 (2022). This study reports the use of scRNA-seq to profile the cellular landscape of human DFU injuries (local tissue biopsies and peripheral blood) and identify unique populations enriched in patients with effective wound healing.

Mascharak, S. et al. Multi-omic analysis reveals divergent molecular events in scarring and regenerative wound healing. Cell Stem Cell 29 , 315–327.e6 (2022).

Phan, Q. M., Sinha, S., Biernaskie, J. & Driskell, R. R. Single‐cell transcriptomic analysis of small and large wounds reveals the distinct spatial organization of regenerative fibroblasts. Exp. Dermatol. 30 , 92–101 (2021).

Cui, M. et al. Dynamic transcriptional responses to injury of regenerative and non-regenerative cardiomyocytes revealed by single-nucleus RNA sequencing. Dev. Cell 53 , 102–116.e8 (2020).

Wang, Z. et al. Cell-type-specific gene regulatory networks underlying murine neonatal heart regeneration at single-cell resolution. Cell Rep. 33 , 108472 (2020).

Aztekin, C. et al. Identification of a regeneration-organizing cell in the Xenopus tail. Science 364 , 653–658 (2019).

Londono, R. et al. Single cell sequencing analysis of lizard phagocytic cell populations and their role in tail regeneration. J. Immunol. Regen. Med. 8 , 100029 (2020).

PubMed   PubMed Central   Google Scholar  

Qin, T. et al. A population of stem cells with strong regenerative potential discovered in deer antlers. Science 379 , 840–847 (2023).

Chen, T. et al. A road map from single-cell transcriptome to patient classification for the immune response to trauma. JCI Insight 6 , e145108 (2021).

Gaudilliere, B. et al. Coordinated surgical immune signatures contain correlates of clinical recovery. Sci. Transl Med. 6 , 255ra131 (2014).

Pummerer, C. L. et al. Identification of cardiac myosin peptides capable of inducing autoimmune myocarditis in BALB/c mice. J. Clin. Invest. 97 , 2057–2062 (1996).

Rieckmann, M. et al. Myocardial infarction triggers cardioprotective antigen-specific T helper cell responses. J. Clin. Invest. 129 , 4922–4936 (2019).

Xia, N. et al. A unique population of regulatory T cells in heart potentiates cardiac protection from myocardial infarction. Circulation 142 , 1956–1973 (2020).

Delgobo, M. et al. Myocardial milieu favors local differentiation of regulatory T cells. Circ. Res. 132 , 565–582 (2023).

Guo, F. et al. Distinct injury responsive regulatory T cells identified by multi-dimensional phenotyping. Front. Immunol. 13 , 833100 (2022).

Hanna, B. S. et al. The gut microbiota promotes distal tissue regeneration via RORγ + regulatory T cell emissaries. Immunity 56 , 829–846.e8 (2023).

Boland, B. S. et al. Heterogeneity and clonal relationships of adaptive immune cells in ulcerative colitis revealed by single-cell analyses. Sci. Immunol. 5 , eabb4432 (2020).

Koda, Y. et al. CD8+ tissue-resident memory T cells promote liver fibrosis resolution by inducing apoptosis of hepatic stellate cells. Nat. Commun. 12 , 4474 (2021).

Melo Ferreira, R. et al. Integration of spatial and single-cell transcriptomics localizes epithelial cell–immune cross-talk in kidney injury. JCI Insight 6 , e147703 (2021).

McKellar, D. W. et al. Large-scale integration of single-cell transcriptomic data captures transitional progenitor states in mouse skeletal muscle regeneration. Commun. Biol. 4 , 1280 (2021). This article reports the integration of new and publicly available scRNA-seq and snRNA-seq data sets to create a large-scale atlas of murine skeletal muscle injury for in-depth exploration of rare MuSC differentiation states, and it serves as cell cluster annotation reference for muscle-injury spatial transcriptomics.

Konieczny, P. et al. Interleukin-17 governs hypoxic adaptation of injured epithelium. Science 377 , eabg9302 (2022).

Kim, H. K., Ha, T. W. & Lee, M. R. Single-cell transcriptome analysis as a promising tool to study pluripotent stem cell reprogramming. Int. J. Mol. Sci. 22 , 5988 (2021).

Camp, J. G., Wollny, D. & Treutlein, B. Single-cell genomics to guide human stem cell and tissue engineering. Nat. Methods 15 , 661–667 (2018).

Chen, K. et al. Disrupting mechanotransduction decreases fibrosis and contracture in split-thickness skin grafting. Sci. Transl Med. 14 , eabj9152 (2022).

Henn, D. et al. Xenogeneic skin transplantation promotes angiogenesis and tissue regeneration through activated Trem2 + macrophages. Sci. Adv. 7 , eabi4528 (2021). This study reports the use of scRNA-seq to investigate the innate immune response to xenogeneic skin transplants, identify unique TREM2 + regenerative macrophages and develop a new cell-laden hydrogel construct to mitigate fibrosis and improve healing of complex skin wounds.

Wang, H. et al. Decoding the annulus fibrosus cell atlas by scRNA-seq to develop an inducible composite hydrogel: a novel strategy for disc reconstruction. Bioact. Mater. 14 , 350–363 (2022).

Zhang, X. et al. Msx1+ stem cells recruited by bioactive tissue engineering graft for bone regeneration. Nat. Commun. 13 , 5211 (2022).

Xiao, W. et al. Recombinant DTβ4-inspired porous 3D vascular graft enhanced antithrombogenicity and recruited circulating CD93 + /CD34 + cells for endothelialization. Sci. Adv. 8 , eabn1958 (2022).

Jiang, Y. et al. Wireless, closed-loop, smart bandage with integrated sensors and stimulators for advanced wound care and accelerated healing. Nat. Biotechnol. 41 , 652–662 (2022).

Hu, C. et al. Dissecting the microenvironment around biosynthetic scaffolds in murine skin wound healing. Sci. Adv. 7 , eabf0787 (2021).

Liang, R. et al. Silk gel recruits specific cell populations for scarless skin regeneration. Appl. Mater. Today 23 , 101004 (2021).

Huang, J. et al. Single-cell RNA-seq reveals functionally distinct biomaterial degradation-related macrophage populations. Biomaterials 277 , 121116 (2021).

Sadtler, K. et al. Developing a pro-regenerative biomaterial scaffold microenvironment requires T helper 2 cells. Science 352 , 366–370 (2016).

Heredia, J. E. et al. Type 2 innate signals stimulate fibro/adipogenic progenitors to facilitate muscle regeneration. Cell 153 , 376–388 (2013).

Brown, B. N. et al. Macrophage phenotype as a predictor of constructive remodeling following the implantation of biologically derived surgical mesh materials. Acta Biomater. 8 , 978–987 (2012).

Chung, L. et al. Interleukin 17 and senescent cells regulate the foreign body response to synthetic material implants in mice and humans. Sci. Transl Med. 12 , eaax3799 (2020).

Sadtler, K. et al. Divergent immune responses to synthetic and biological scaffolds. Biomaterials 192 , 405–415 (2019).

Sommerfeld, S. D. et al. Interleukin-36γ–producing macrophages drive IL-17–mediated fibrosis. Sci. Immunol. 4 , eaax4783 (2019). This article reports the use of scRNA-seq to profile macrophages from various muscle injury and biomaterial (pro-regenerative ECM scaffold and pro-fibrotic synthetic scaffold) environments to identify unique phenotypes and mechanistic drivers of divergent wound-healing outcomes.

Wang, J. et al. Break monopoly of polarization: CD301b + macrophages play positive roles in osteoinduction of calcium phosphate ceramics. Appl. Mater. Today 24 , 101111 (2021).

Wang, J. et al. CD301b+ macrophages mediate angiogenesis of calcium phosphate bioceramics by CaN/NFATc1/VEGF axis. Bioact. Mater. 15 , 446–455 (2022).

CAS   PubMed   PubMed Central   Google Scholar  

Anderson, J. M. Inflammatory response to implants. ASAIO Trans. 34 , 101–107 (1988).

Henderson, N. C., Rieder, F. & Wynn, T. A. Fibrosis: from mechanisms to medicines. Nature 587 , 555–566 (2020).

Doloff, J. C. et al. The surface topography of silicone breast implants mediates the foreign body response in mice, rabbits and humans. Nat. Biomed. Eng. 5 , 1115–1130 (2021).

Padmanabhan, J., Chen, K., Sivaraj, D. et al. Allometrically scaling tissue forces drive pathological foreign-body responses to implants via Rac2 -activated myeloid cells. Nat. Biomed. Eng . 7 , 1419–1436 (2023).

Sivaraj, D. et al. IQGAP1‐mediated mechanical signaling promotes the foreign body response to biomedical implants. FASEB J. 36 , e22007 (2022).

Cherry, C. et al. Transfer learning in a biomaterial fibrosis model identifies in vivo senescence heterogeneity and contributions to vascularization and matrix production across species and diverse pathologies. Geroscience 45 , 2559–2587 (2023).

Cherry, C. et al. Computational reconstruction of the signalling networks surrounding implanted biomaterials from single-cell transcriptomics. Nat. Biomed. Eng. 5 , 1228–1238 (2021).

Jones, R. C. et al. The Tabula Sapiens: a multiple-organ, single-cell transcriptomic atlas of humans. Science 376 , eabl4896 (2022).

Schaum, N. et al. Single-cell transcriptomics of 20 mouse organs creates a Tabula Muris . Nature 562 , 367–372 (2018).

Article   PubMed Central   ADS   Google Scholar  

Almanzar, N. et al. A single-cell transcriptomic atlas characterizes ageing tissues in the mouse. Nature 583 , 590–595 (2020).

Buechler, M. B. et al. Cross-tissue organization of the fibroblast lineage. Nature 593 , 575–579 (2021).

Cao, J. et al. A human cell atlas of fetal gene expression. Science 370 , eaba7721 (2020).

Domcke, S. et al. A human cell atlas of fetal chromatin accessibility. Science 370 , eaba7612 (2020).

Sanchez-Vega, F. et al. Oncogenic signaling pathways in the cancer genome atlas. Cell 173 , 321–337.e10 (2018).

Prieto, C., Barrios, D. & Villaverde, A. SingleCAnalyzer: interactive analysis of single cell RNA-Seq data on the cloud. Front. Bioinform. 2 , 793309 (2022).

Megill, C. et al. Cellxgene: a performant, scalable exploration platform for high dimensional sparse matrices. Preprint at bioRxiv https://doi.org/10.1101/2021.04.05.438318 (2021).

Speir, M. L. et al. UCSC cell browser: visualize your single-cell data. Bioinformatics 37 , 4578–4580 (2021).

Reich, M. et al. GenePattern 2.0. Nat. Genet. 38 , 500–501 (2006).

Luecken, M. D. & Theis, F. J. Current best practices in single‐cell RNA‐seq analysis: a tutorial. Mol. Syst. Biol. 15 , e8746 (2019). This review describes best practices and commonly used tools for scRNA-seq analysis and applies them to a publicly available data set as a guide; it also provides recommendations and points out potential pitfalls at each step of the process.

Cirulli, E. T. et al. A missense variant in PTPN22 is a risk factor for drug-induced liver injury. Gastroenterology 156 , 1707–1716.e2 (2019).

Delacher, M. et al. Single-cell chromatin accessibility landscape identifies tissue repair program in human regulatory T cells. Immunity 54 , 702–720.e17 (2021).

Llorens-Bobadilla, E. et al. A latent lineage potential in resident neural stem cells enables spinal cord repair. Science 370 , eabb8795 (2020).

Wang, L. et al. Serum proteomics identifies biomarkers associated with the pathogenesis of idiopathic pulmonary fibrosis. Mol. Cell. Proteom. 22 , 100524 (2023).

Ogbeide, S., Giannese, F., Mincarelli, L. & Macaulay, I. C. Into the multiverse: advances in single-cell multiomic profiling. Trends Genet. 38 , 831–843 (2022).

Rodriguez-Meira, A. et al. Unravelling intratumoral heterogeneity through high-sensitivity single-cell mutational analysis and parallel RNA sequencing. Mol. Cell 73 , 1292–1305.e8 (2019).

Zhu, C. et al. An ultra high-throughput method for single-cell joint analysis of open chromatin and transcriptome. Nat. Struct. Mol. Biol. 26 , 1063–1070 (2019).

Bock, C. et al. High-content CRISPR screening. Nat. Rev. Methods Primers 2 , 9 (2022).

Tian, F. et al. Core transcription programs controlling injury-induced neurodegeneration of retinal ganglion cells. Neuron 110 , 2607–2624.e8 (2022).

Zheng, G. X. Y. et al. Massively parallel digital transcriptional profiling of single cells. Nat. Commun. 8 , 14049 (2017).

Macosko, E. Z. et al. Highly parallel genome-wide expression profiling of individual cells using nanoliter droplets. Cell 161 , 1202–1214 (2015).

Picelli, S. et al. Full-length RNA-seq from single cells using Smart-seq2. Nat. Protoc. 9 , 171–181 (2014).

Hashimshony, T. et al. CEL-Seq2: sensitive highly-multiplexed single-cell RNA-Seq. Genome Biol. 17 , 77 (2016).

Bray, N. L., Pimentel, H., Melsted, P. & Pachter, L. Near-optimal probabilistic RNA-seq quantification. Nat. Biotechnol. 34 , 525–527 (2016).

Grandi, F. et al. popsicleR: a R package for pre-processing and quality control analysis of single cell RNA-seq data. J. Mol. Biol. 434 , 167560 (2022).

Hong, R. et al. Comprehensive generation, visualization, and reporting of quality control metrics for single-cell RNA sequencing data. Nat. Commun. 13 , 1688 (2022).

Hippen, A. A. et al. miQC: an adaptive probabilistic framework for quality control of single-cell RNA-sequencing data. PLoS Comput. Biol. 17 , e1009290 (2021).

Wolock, S. L., Lopez, R. & Klein, A. M. Scrublet: computational identification of cell doublets in single-cell transcriptomic data. Cell Syst. 8 , 281–291.e9 (2019).

Bernstein, N. J. et al. Solo: doublet identification in single-cell RNA-seq via semi-supervised deep learning. Cell Syst. 11 , 95–101.e5 (2020).

Yang, S. et al. Decontamination of ambient RNA in single-cell RNA-seq with DecontX. Genome Biol. 21 , 57 (2020).

Berg, M. et al. FastCAR: fast Correction for Ambient RNA to facilitate differential gene expression analysis in single-cell RNA-sequencing datasets. Preprint at bioRxiv https://doi.org/10.1101/2022.07.19.500594 (2022).

Lun, A. T. L., McCarthy, D. J. & Marioni, J. C. A step-by-step workflow for low-level analysis of single-cell RNA-seq data with Bioconductor. F1000Research 5 , 2122 (2016).

Bacher, R. et al. SCnorm: robust normalization of single-cell RNA-seq data. Nat. Methods 14 , 584–586 (2017).

Yip, S. H., Wang, P., Kocher, J.-P. A., Sham, P. C. & Wang, J. Linnorm: improved statistical analysis for single cell RNA-seq expression data. Nucleic Acids Res. 45 , e179 (2017).

Korsunsky, I. et al. Fast, sensitive and accurate integration of single-cell data with Harmony. Nat. Methods 16 , 1289–1296 (2019).

Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177 , 1888–1902.e21 (2019).

Hie, B., Bryson, B. & Berger, B. Efficient integration of heterogeneous single-cell transcriptomes using Scanorama. Nat. Biotechnol. 37 , 685–691 (2019).

van Dijk, D. et al. Recovering gene interactions from single-cell data using data diffusion. Cell 174 , 716–729.e27 (2018).

Wagner, F., Yan, Y. & Yanai, I. K-nearest neighbor smoothing for high-throughput single-cell RNA-Seq data. Preprint at bioRxiv https://doi.org/10.1101/217737 (2018).

Huang, M. et al. SAVER: gene expression recovery for single-cell RNA sequencing. Nat. Methods 15 , 539–542 (2018).

van der Maaten, L. & Hinton, G. Visualizing data using t-SNE. J. Mach. Learn. Res. 9 , 2579–2605 (2008).

Google Scholar  

McInnes, L., Healy, J., Saul, N. & Großberger, L. UMAP: uniform manifold approximation and projection. J. Open Source Softw. 3 , 861 (2018).

Kiselev, V. Y. et al. SC3: consensus clustering of single-cell RNA-seq data. Nat. Methods 14 , 483–486 (2017).

Žurauskienė, J. & Yau, C. pcaReduce: hierarchical clustering of single cell transcriptional profiles. BMC Bioinformatics   17 , 140 (2016).

Levine, J. H. et al. Data-driven phenotypic dissection of AML reveals progenitor-like cells that correlate with prognosis. Cell 162 , 184–197 (2015).

Hou, R., Denisenko, E. & Forrest, A. R. R. scMatch: a single-cell gene expression profile annotation tool using reference datasets. Bioinformatics 35 , 4688–4695 (2019).

Fu, R. et al. clustifyr: an R package for automated single-cell RNA sequencing cluster classification. F1000Research 9 , 223 (2020).

Tan, Y. & Cahan, P. SingleCellNet: a computational tool to classify single cell RNA-seq data across platforms and across species. Cell Syst. 9 , 207–213.e2 (2019).

Kharchenko, P. V., Silberstein, L. & Scadden, D. T. Bayesian approach to single-cell differential expression analysis. Nat. Methods 11 , 740–742 (2014).

Finak, G. et al. MAST: a flexible statistical framework for assessing transcriptional changes and characterizing heterogeneity in single-cell RNA sequencing data. Genome Biol. 16 , 278 (2015).

Korthauer, K. D. et al. A statistical approach for identifying differential distributions in single-cell RNA-seq experiments. Genome Biol. 17 , 222 (2016).

Ji, Z. & Ji, H. TSCAN: pseudo-time reconstruction and evaluation in single-cell RNA-seq analysis. Nucleic Acids Res. 44 , e117 (2016).

Street, K. et al. Slingshot: cell lineage and pseudotime inference for single-cell transcriptomics. BMC Genomics 19 , 477 (2018).

La Manno, G. et al. RNA velocity of single cells. Nature 560 , 494–498 (2018).

Aibar, S. et al. SCENIC: single-cell regulatory network inference and clustering. Nat. Methods 14 , 1083–1086 (2017).

Jin, S. et al. Inference and analysis of cell-cell communication using CellChat. Nat. Commun. 12 , 1088 (2021).

Chou, C.-H. et al. Synovial cell cross-talk with cartilage plays a major role in the pathogenesis of osteoarthritis. Sci. Rep. 10 , 10868 (2020).

do Valle Duraes, F. et al. Immune cell landscaping reveals a protective role for regulatory T cells during kidney injury and fibrosis. JCI Insight 5 , e130651 (2020).

Rudman-Melnick, V. et al. Single-cell profiling of AKI in a murine model reveals novel transcriptional signatures, profibrotic phenotype, and epithelial-to-stromal crosstalk. J. Am. Soc. Nephrol. 31 , 2793–2814 (2020).

Misra, A. et al. Characterizing neonatal heart maturation, regeneration, and scar resolution using spatial transcriptomics. J. Cardiovasc. Dev. Dis. 9 , 1 (2022).

CAS   Google Scholar  

Conlon, T. M. et al. Inhibition of LTβR signalling activates WNT-induced regeneration in lung. Nature 588 , 151–156 (2020).

Tran, N. M. et al. Single-cell profiles of retinal ganglion cells differing in resilience to injury reveal neuroprotective genes. Neuron 104 , 1039–1055.e12 (2019).

Wheeler, M. A. et al. MAFG-driven astrocytes promote CNS inflammation. Nature 578 , 593–599 (2020).

Schirmer, L. et al. Neuronal vulnerability and multilineage diversity in multiple sclerosis. Nature 573 , 75–82 (2019).

Henn, D. et al. Cas9-mediated knockout of Ndrg2 enhances the regenerative potential of dendritic cells for wound healing. Nat. Commun. 14 , 4729 (2023).

Jin, R. M., Warunek, J. & Wohlfert, E. A. Chronic infection stunts macrophage heterogeneity and disrupts immune-mediated myogenesis. JCI Insight 3 , e121459 (2018).

Vu, R. et al. Wound healing in aged skin exhibits systems-level alterations in cellular composition and cell-cell communication. Cell Rep. 40 , 111155 (2022).

Han, J. et al. Age-associated senescent - T cell signaling promotes type 3 immunity that inhibits regenerative response. Preprint at bioRxiv https://doi.org/10.1101/2021.08.17.456641 (2022).

Zhang, C. et al. Age‐related decline of interferon‐gamma responses in macrophage impairs satellite cell proliferation and regeneration. J. Cachexia Sarcopenia Muscle 11 , 1291–1305 (2020).

Download references

Acknowledgements

This work was funded in part by the National Institutes of Health (NIH) Pioneer Award DP1AR076959 (to J.H.E.), Bloomberg~Kimmel Institute and Morton Goldberg Professorship (to J.H.E.). A.R. is funded through NSF GRFP DGE-1746891.

Author information

These authors contributed equally: Anna Ruta, Kavita Krishnan.

Authors and Affiliations

Translational Tissue Engineering Center and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA

Anna Ruta, Kavita Krishnan & Jennifer H. Elisseeff

You can also search for this author in PubMed   Google Scholar

Contributions

All authors contributed equally to the preparation of this manuscript.

Corresponding author

Correspondence to Jennifer H. Elisseeff .

Ethics declarations

Competing interests.

J.H.E. holds equity in Unity Biotechnology and Aegeria Soft Tissue, and is an advisor for Tessera Therapeutics, HapInScience and Font Bio. The remaining authors declare no competing interests.

Peer review

Peer review information.

Nature Reviews Bioengineering thanks Omer Bayraktar and the other, anonymous, reviewers for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Cite this article.

Ruta, A., Krishnan, K. & Elisseeff, J.H. Single-cell transcriptomics in tissue engineering and regenerative medicine. Nat Rev Bioeng 2 , 101–119 (2024). https://doi.org/10.1038/s44222-023-00132-7

Download citation

Accepted : 11 October 2023

Published : 19 December 2023

Issue Date : February 2024

DOI : https://doi.org/10.1038/s44222-023-00132-7

Share this article

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

Quick links

  • Explore articles by subject
  • Guide to authors
  • Editorial policies

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

phd tissue engineering and regenerative medicine

  • Sponsored Article

Brush Up: Tissue Engineering and Regenerative Medicine

A new frontier in repairing organ damage.

Jennifer Zieba, PhD headshot

Jen has a PhD in human genetics from the University of California, Los Angeles where she is currently a project scientist. She enjoys teaching and communicating complex scientific concepts to a wide audience.

View full profile.

Learn about our editorial policies.

What Is Regenerative Medicine?   Regenerative medicine replaces tissue or organs that are damaged by trauma, disease, or congenital disorders. This is different from more traditional therapies that treat the symptoms of tissue damage. There are three main concentrations within the field of regenerative medicine: tissue engineering, cellular therapy, and artificial organs. The use of tissue engineering in regenerative medicine, known as TERM, is an active area of research that involves creating functional tissue through the combination of cells, scaffolds, and growth factors to restore normal biological function. 1  Clinicians treat millions of patients with tissue engineered regenerative devices. So far, the most successful tissue regeneration therapies occur in soft tissues such as skin, cartilage, and corneal tissues.

Using Tissue Engineering to Regenerate Damaged Tissue

How does tissue engineering work?

During healthy tissue development, cells build and surround themselves with an extracellular matrix. This matrix, or scaffold, contains structural proteins and acts as a reservoir for signaling molecules that cells use to communicate and organize themselves into functional complexes or tissues. 

The overall goal of tissue engineering in the context of regenerative medicine is to establish a 3D cell or biomaterial complex that functions similarly to the in vivo tissue extracellular matrix. In general, tissue engineering involves the design and implantation of a scaffold that is biologically compatible with the area to be regenerated. New cells are then either attracted to or grown directly onto the scaffold. 2 The FDA has approved engineered artificial cartilage and skin therapies, and researchers are developing many other therapies for different tissues and disorders (see table below).

Scaffolds in tissue engineering Scientists seed scaffolds with their desired cell type during or following implantation. Alternatively, they may add growth factors to the scaffold and wait until the structure is populated by the surrounding tissue.

Choosing a scaffold type and source for tissue engineering is imperative for regenerating functional tissue. Pore size and overall architecture are important variables to consider when designing a scaffold. Pores play a crucial role in tissue regeneration because they allow for the exchange of nutrition and oxygen with surrounding tissue as well as expulsion of waste products and vascularization. The overall architecture of the tissue is important for exposing surfaces for cell attachment as well as mechanical cell stimuli.

Scaffolds can be natural or synthetic. Natural scaffolds are derived from donor tissues where the cells are chemically removed, leaving only the extracellular matrix. Natural scaffolds can either come from a patient or a healthy donor, and they have the advantage of retaining the unique structural and functional architecture of complex tissues. Researchers can also create natural scaffolds in vitro, such as those made from collagen and Matrigel, which are comprised of basement membrane proteins. 3,4

Scientists can develop synthetic scaffolds from various polymers, including polyglycolic acid (PGA), polylactic acid (PLA), and poly(lactide-co-glycolide) acid (PLGA). Scaffolds made with these polymers are flexible and porous, making them ideal structures for cellular integration. Synthetic scaffolds are also biodegradable, with different polymers degrading at different times, allowing damaged tissue to regenerate without the use of permanent prosthetic implants. Synthetic scaffolds also have consistent structures between replicates as they are generated in a laboratory; however, they can cause inflammation in the recipient more readily than natural scaffolds. 5

3D printing in tissue engineering With recent progress in 3D printing methods, researchers create complex synthetic scaffold structures with more consistent architecture and pore sizes. Hydrogel materials, such as alginate hydrogel and gelatin, are typically used in 3D printing due to their effective crosslinking and biocompatible properties. 6

Stem cells in tissue engineering and regenerative therapy Mesenchymal, embryonic, and induced pluripotent stem cells effectively promote damaged tissue regeneration. However, in many tissues, transplanted stem cells have poor survival and differentiation capabilities. The development of stem cell technology in combination with tissue engineering techniques, such as scaffolds and the addition of growth factors, has allowed researchers to improve the viability and proliferation of stem cells in regenerative medicine. 7

Flowchart depicting the stages of development including fertilization, blastocyst formation, the three germ layers, and specialized cells that arise from each layer. The flow chart also shows corresponding developmental potency at each stage, including totipotent, pluripotent, multipotent, and unipotent.Pluripotent stem cell lines are derived from an embryo or scientists artificially generate induced pluripotent stem cells (iPSCs) by reprogramming somatic cells with forced expression of embryonic transcription factors. Pluripotent stem cells and iPSCs function similarly, and the discovery of iPSCs helped researchers understand how transcription factors affect cell differentiation during reprogramming and normal development.1,3,4

2. F. Han et al., “Tissue engineering and regenerative medicine: Achievements, future, and sustainability in Asia,” Front Bioeng Biotechnol , 8:83, 2020.

3. S. Sundaram et al., “Tissue engineering and regenerative medicine” in   Rossi's Principles of Transfusion Medicine . Fifth edition. T.L. Simon, ed., New York, N.Y.: John Wiley & Sons Inc., 2016, pp. 488-504.

4. C. Motta et al., “Tissue engineering and regenerative medicine” in Rossi's Principles of Transfusion Medicine . Sixth edition. T.L. Simon, ed., New York, N.Y.: John Wiley & Sons Inc., 2022, pp. 648-660.

5. Y. Li et al., “The effect of mechanical loads on the degradation of aliphatic biodegradable polyesters,” Regen Biomater , 4:179-190, 2017.

6. Z. Yazdanpanah et al., “3D bioprinted scaffolds for bone tissue engineering: State-of-the-art and emerging technologies,” Front Bioeng Biotechnol , 10:824156, 2022.

7. S.G. Kwon et al., “Recent advances in stem cell therapeutics and tissue engineering strategies,” Biomater Res , 22:36, 2018.

rr logo

Related cell biology Research Resources

<em >The Scientist</em>&rsquo;s Journal Club: Neuroscience and Cell Biology

The Scientist ’s Journal Club: Neuroscience and Cell Biology

Microscopic image of a live amoeba.

Illuminating Specimens Through Live Cell Imaging

Exploring Stem Cell Strategies for Spinal Cord Repair

Exploring Stem Cell Strategies for Spinal Cord Repair

Bio-Techne logo

The University of Edinburgh home

  • Schools & departments

Postgraduate study

Regenerative Medicine PhD

Awards: PhD

Study modes: Full-time, Part-time

Funding opportunities

Programme website: Regenerative Medicine

Discovery Day

Join us online on 18th April to learn more about postgraduate study at Edinburgh

View sessions and register

Research profile

Research excellence.

The Centre for Regenerative Medicine (CRM) is a world leading research centre based at the University of Edinburgh’s Institute for Regeneration and Repair.

Our scientists and clinicians study stem cells, disease and tissue repair to advance human health. By better understanding how stem cells are controlled and how diseases develop in a lab environment, we hope to find new ways to treat patients.

Our research is aimed at developing new treatments for major diseases including cancer, heart disease, diabetes, degenerative diseases such as multiple sclerosis and Parkinson's disease, and liver failure.

The Centre houses 25 research groups and has a staff of more than 270 scientists, graduate students, support and ancillary staff.

Research themes

Our work is currently organised into five themes. To promote collaboration within the Centre, we adopt a flexible approach to these themes, with each Principal Investigator (PI) having one or more secondary affiliations.

Two themes focus on fundamental research:

  • pluripotency and iPS
  • lineage and cell specification

The other three themes aim to translate fundamental research discoveries into clinical programmes relevant to brain, blood and liver diseases and to tissue repair.

The Centre has strong collaborative links to other centres within the University, such as the Euan MacDonald Centre for MND Research, the MS Centre and the Roslin Institute.

We also invest in technological development in all areas.

Training and support

Training within the Centre is provided through a structured series of seminars and literature reviews, in addition to the laboratory and scientific research skills training provided to you by your supervisors.

Many of our PhD students are involved in collaborative projects that provide cross-disciplinary experience and/or promote translation into the biotechnology or clinical fields.

How will I learn?

Our programme includes short courses taught by basic and clinical stem cell scientists, providing a state-of-the-art theoretical background in a variety of areas relating to regenerative medicine including:

  • developmental biology
  • pluripotent and tissue stem cell biology
  • degeneration and regeneration of adult tissues
  • genetic engineering
  • bioinformatics

We provide specialist lectures and short practical modules covering key technologies, including:

  • DNA analysis and genetic engineering
  • flow cytometry

In Year 1, you will participate in a weekly Centre for Regenerative Medicine ( CRM ) Postgraduate Discussion Group led by CRM group leaders. These discussion groups aim to widen your knowledge of stem cell and regenerative medicine research and to enhance your ability to critically review the literature in this field.

In addition to the taught components and research project, you will participate in a number of activities, including:

  • regular lab meetings of your research group
  • an internal seminar series
  • seminars by visiting national and international speakers
  • Journal Club
  • poster presentations
  • Three Minute Thesis presentation session.

Generic and transferable skills training is provided through the University's Institute for Academic Development (IAD).

  • Institute of Academic Development

Since 2011, the Centre has been housed in a new, specially designed building that provides high quality research facilities, including:

  • state of the art centralised cell culture facility for isolation and culture of primary and established cell lines including embryonic and induced pluripotent stem cells
  • clinical-grade GMP cell culture facility
  • specific pathogen free animal facility
  • transgenic service covering derivation and provision of mouse embryonic stem cells, blastocyst injection, morula aggregation and production of defined genetic alterations
  • ultrasound micro-injection equipment
  • flow cytometry service consisting of a suite of cell sorters and analysers operated by facility staff that can be operated by users following comprehensive training
  • a recently established single cell genomic analysis service using a 10x Genomics Chromium Controller
  • quantitative real-time polymerase chain reaction equipment
  • Fluidigm Biomark and CellPrep for single cell transcriptomics

Imaging facilities

We also have imaging facilities, including:

  • standard compound microscopy
  • widefield, confocal, and lightsheet microscopes
  • high-content and timelapse imaging

The facility has dedicated imaging managers and offers two high-end workstations for bio-image processing and analysis.

Take a virtual tour of our facilities at the Centre for Regenerative Medicine:

  • Virtual tour

Entry requirements

These entry requirements are for the 2024/25 academic year and requirements for future academic years may differ. Entry requirements for the 2025/26 academic year will be published on 1 Oct 2024.

A UK 2:1 honours degree or its international equivalent.

International qualifications

Check whether your international qualifications meet our general entry requirements:

  • Entry requirements by country
  • English language requirements

Regardless of your nationality or country of residence, you must demonstrate a level of English language competency at a level that will enable you to succeed in your studies.

English language tests

We accept the following English language qualifications at the grades specified:

  • IELTS Academic: total 6.5 with at least 6.0 in each component. We do not accept IELTS One Skill Retake to meet our English language requirements.
  • TOEFL-iBT (including Home Edition): total 92 with at least 20 in each component. We do not accept TOEFL MyBest Score to meet our English language requirements.
  • C1 Advanced ( CAE ) / C2 Proficiency ( CPE ): total 176 with at least 169 in each component.
  • Trinity ISE : ISE II with distinctions in all four components.
  • PTE Academic: total 62 with at least 59 in each component.

Your English language qualification must be no more than three and a half years old from the start date of the programme you are applying to study, unless you are using IELTS , TOEFL, Trinity ISE or PTE , in which case it must be no more than two years old.

Degrees taught and assessed in English

We also accept an undergraduate or postgraduate degree that has been taught and assessed in English in a majority English speaking country, as defined by UK Visas and Immigration:

  • UKVI list of majority English speaking countries

We also accept a degree that has been taught and assessed in English from a university on our list of approved universities in non-majority English speaking countries (non-MESC).

  • Approved universities in non-MESC

If you are not a national of a majority English speaking country, then your degree must be no more than five years old* at the beginning of your programme of study. (*Revised 05 March 2024 to extend degree validity to five years.)

Find out more about our language requirements:

Fees and costs

Additional programme costs.

Most laboratories require a bench fee of up to £5,000 per year. This cost can be covered in Research Council studentships.

Living costs

You will be responsible for covering living costs for the duration of your studies.

Tuition fees

Scholarships and funding, featured funding.

  • College of Medicine & Veterinary Medicine funding opportunities

UK government postgraduate loans

If you live in the UK, you may be able to apply for a postgraduate loan from one of the UK’s governments.

The type and amount of financial support you are eligible for will depend on your programme, the duration of your studies, and your residency status.

Programmes studied on a part-time intermittent basis are not eligible.

  • UK government and other external funding

Other funding opportunities

Search for scholarships and funding opportunities:

  • Search for funding

Further information

  • Postgraduate Administrator, Kelly Douglas
  • Phone: +44 (0)131 651 9500
  • Contact: [email protected]
  • Centre for Regenerative Medicine
  • Institute for Regeneration and Repair
  • The University of Edinburgh
  • Little France
  • Programme: Regenerative Medicine
  • School: Edinburgh Medical School: Clinical Sciences
  • College: Medicine & Veterinary Medicine

Select your programme and preferred start date to begin your application.

PhD Regenerative Medicine - 3 Years (Full-time)

Phd regenerative medicine - 6 years (part-time), application deadlines.

We encourage you to apply at least one month prior to entry so that we have enough time to process your application. If you are also applying for funding or will require a visa then we strongly recommend you apply as early as possible.

  • How to apply

You must submit two references with your application.

Before making your application, you must make contact with a potential supervisor to discuss your research proposal. Further information on making a research degree application can be found on the College website:

  • How to apply for a research degree

Find out more about the general application process for postgraduate programmes:

Van Cleve Cardiac Regenerative Medicine Program

Researcher reviewing lab specimens

Unprecedented hope

Cardiovascular disease, the leading cause of premature death around the world, is on the rise. Traditional medical therapies are not able to fully address the burden of heart disease, and the shortage of organs available for transplantation remains a key barrier for offering solutions to millions of patients across the globe.

Through the visionary leadership and generous philanthropy of Russell and Kathy Van Cleve, unprecedented hope is beginning to translate into future healing options for those with heart disease.

By harnessing the body's natural ability to heal, regenerative medicine is addressing the unmet needs of patients with chronic or complex conditions who often have no other viable treatment options.

Broad impact

The goal of the Mayo Clinic Van Cleve Cardiac Regenerative Medicine Program is to advance stem cell therapies, cell-free regeneration and tissue engineering. To do this, the program accelerates the discovery, translation and application of innovative regenerative products for heart disease.

Importantly, results from this work will not only make an impact in cardiovascular health but also advance novel regenerative solutions across medical and surgical specialties to address vast unmet patient needs.

Strategic focus

Through a shared vision with Russell and Kathy Van Cleve, the Mayo Clinic Van Cleve Cardiac Regenerative Medicine Program has the goal of creating novel, affordable and accessible cardiac regenerative therapies to restore the health of people with heart disease.

After meeting the highest standards of quality control and best manufacturing practices, products undergo rigorous testing in clinical trials to ensure their safe translation to clinical practice. Finally, research findings are applied to patient care in areas such as surgery, radiology, laboratory medicine and more, fostering Mayo Clinic's regenerative care model of not only reversing chronic disease but also restoring health to individuals and their communities.

Research pillars

Research in the Mayo Clinic Van Cleve Cardiac Regenerative Medicine Program is based on three pillars of investigative focus:

  • Cellular therapies
  • Acellular therapies
  • Tissue engineering

Contemporary regenerative medicine is underpinned by the recognition that full regeneration ultimately restores the person as a whole. Fostering the Mayo Clinic regenerative care model, the Mayo Clinic Van Cleve Cardiac Regenerative Medicine Program is advancing not only the restoration of organ health but also the rebuilding of individuals in their communities.

The mission of the Mayo Clinic Van Cleve Cardiac Regenerative Medicine Program is to position Mayo Clinic as the premier regenerative medicine destination: leading in the discovery of novel cell-therapy platforms that address cardiac pathologies, advancing off-the-shelf cell-free therapies from the bench toward clinical cardiovascular practice and establishing cardiac tissue engineering — thereby transforming the reach of modern regenerative care.

Investing in Humanity

The Mayo Clinic Van Cleve Cardiac Regenerative Medicine Program is funded with the generous support of Russ and Kathy Van Cleve.

More about research at Mayo Clinic

  • Research Faculty
  • Laboratories
  • Core Facilities
  • Centers & Programs
  • Departments & Divisions
  • Clinical Trials
  • Institutional Review Board
  • Postdoctoral Fellowships
  • Training Grant Programs
  • Publications

Mayo Clinic Footer

  • Request Appointment
  • About Mayo Clinic
  • About This Site

Legal Conditions and Terms

  • Terms and Conditions
  • Privacy Policy
  • Notice of Privacy Practices
  • Notice of Nondiscrimination
  • Manage Cookies

Advertising

Mayo Clinic is a nonprofit organization and proceeds from Web advertising help support our mission. Mayo Clinic does not endorse any of the third party products and services advertised.

  • Advertising and sponsorship policy
  • Advertising and sponsorship opportunities

Reprint Permissions

A single copy of these materials may be reprinted for noncommercial personal use only. "Mayo," "Mayo Clinic," "MayoClinic.org," "Mayo Clinic Healthy Living," and the triple-shield Mayo Clinic logo are trademarks of Mayo Foundation for Medical Education and Research.

Open Access is an initiative that aims to make scientific research freely available to all. To date our community has made over 100 million downloads. It’s based on principles of collaboration, unobstructed discovery, and, most importantly, scientific progression. As PhD students, we found it difficult to access the research we needed, so we decided to create a new Open Access publisher that levels the playing field for scientists across the world. How? By making research easy to access, and puts the academic needs of the researchers before the business interests of publishers.

We are a community of more than 103,000 authors and editors from 3,291 institutions spanning 160 countries, including Nobel Prize winners and some of the world’s most-cited researchers. Publishing on IntechOpen allows authors to earn citations and find new collaborators, meaning more people see your work not only from your own field of study, but from other related fields too.

Brief introduction to this section that descibes Open Access especially from an IntechOpen perspective

Want to get in touch? Contact our London head office or media team here

Our team is growing all the time, so we’re always on the lookout for smart people who want to help us reshape the world of scientific publishing.

Home > Books > Tissue Engineering and Regenerative Medicine

Regenerative Medicine and Tissue Engineering

Regenerative Medicine and Tissue Engineering

Book metrics overview

109,142 Chapter Downloads

Impact of this book and its chapters

Total Chapter Downloads on intechopen.com

IntechOpen

Total Chapter Views on intechopen.com

Overall attention for this book and its chapters

Book Citations

Total Chapter Citations

Academic Editor

University of Malaga , Spain

Published 22 May 2013

Doi 10.5772/46192

ISBN 978-953-51-1108-5

eBook (PDF) ISBN 978-953-51-4248-5

Copyright year 2013

Number of pages 868

Few events in science have captured the same level of sustained interest and imagination of the nonscientific community as Stem Cells, Tissue Engineering, and Regenerative Medicine. The fundamental concept of Tissue Engineering and Regenerative Medicine is appealing to scientists, physicians, and lay people alike: to heal tissue or organ defects that the current medical practice deems difficult or...

Few events in science have captured the same level of sustained interest and imagination of the nonscientific community as Stem Cells, Tissue Engineering, and Regenerative Medicine. The fundamental concept of Tissue Engineering and Regenerative Medicine is appealing to scientists, physicians, and lay people alike: to heal tissue or organ defects that the current medical practice deems difficult or impossible to cure. Tissue engineering combines cells, engineering, and materials methods with suitable biochemical and physiochemical factors to improve or replace biologic functions. Regenerative medicine is a new branch of medicine that attempts to change the course of chronic disease, in many instances regenerating failing organ systems lost due to age, disease, damage, or congenital defects. The area is rapidly becoming one of the most promising treatment options for patients suffering from tissue failure. This book of Regenerative Medicine and Tissue Engineering fairly reflects the state of the art of these two disciplines at this time as well as their therapeutic application. It covers numerous topics, such as stem cells, cell culture, polymer synthesis, novel biomaterials, drug delivery, therapeutics, and the creation of tissues and organs. The goal is to have this book serve as a reference for graduate students, post-docs, teachers, scientists and physicians, and as an explanatory analysis for executives in biotech and pharmaceutical companies.

By submitting the form you agree to IntechOpen using your personal information in order to fulfil your library recommendation. In line with our privacy policy we won’t share your details with any third parties and will discard any personal information provided immediately after the recommended institution details are received. For further information on how we protect and process your personal information, please refer to our privacy policy .

Indexed in the Book Citation Index (BKCI) in Web of Science Core Collection™

Cite this book

There are two ways to cite this book:

Edited Volume and chapters are indexed in

Table of contents.

By Annalucia Carbone, Stefano Castellani, Valentina Paracchini, Sante Di Gioia, Carla Colombo and Massimo Conese

By Hideki Agata

By Patricia Zuk

By Razieh Karamzadeh and Mohamadreza Baghaban Eslaminejad

By Fengming Yue, Sakiko Shirasawa, Hinako Ichikawa, Susumu Yoshie, Akimi Mogi, Shoko Masuda, Mika Nagai, Tadayuki Yokohama, Tomotsune Daihachiro and Katsunori Sasaki

By Morikuni Tobita and Hiroshi Mizuno

By José M. López-Puerta, Plácido Zamora-Navas, Silvia Claros, Gustavo A. Rico-Llanos, Inés Avedillo, José A. Andrades and José Becerra

By Vincenzo Vindigni, Giorgio Giatsidis, Francesco Reho , Erica Dalla Venezia , Marco Mammana and Bassetto Franco

By Bridget M. Deasy, Jordan E. Anderson and Shannon Zelina

By Aleksandar Evangelatov and Roumen Pankov

By Tran Le Bao Ha, To Minh Quan, Doan Nguyen Vu and Do Minh Si

By M. Arnal-Pastor, J. C. Chachques, M. Monleón Pradas and A. Vallés- Lluch

By Dragica Smrke, Primož Rožman, Matjaž Veselko and Borut Gubina

By Norbert W. Guldner, Peter Klapproth, Hangörg Zimmermann and Hans- H. Sievers

By Zaira Y. García-Carvajal, David Garciadiego-Cázares, Carmen Parra- Cid, Rocío Aguilar-Gaytán, Cristina Velasquillo , Clemente Ibarra and Javier S. Castro Carmona

By Qiong Li, Lu Zhang, Guangdong Zhou, Wei Liu and Yilin Cao

By Tetsuya Imamura, Osamu Ishizuka and Osamu Nishizawa

By Tatsuya Mimura, Seiichi Yokoo and Satoru Yamagami

By Susanne Jung and Johannes Kleinheinz

By Jan O. Gordeladze, Janne E. Reseland, Tommy A. Karlsen, Rune B. Jakobsen, Astrid K. Stunes, Unni Syversen, Lars Engebretsen, Ståle P. Lyngstadaas and Christian Jorgensen

By Ali Mobasheri and Mark Lewis

By Shigeru Kobayashi

By Sara Bouhout, Alexandre Rousseau, Stéphane Chabaud, Amélie Morissette and Stéphane Bolduc

By Chao Le Meng Bao, Erin Y. Teo, Mark S.K. Chong, Yuchun Liu, Mahesh Choolani and Jerry K.Y. Chan

By José A. Andrades, Lucía Narváez-Ledesma, Luna Cerón-Torres, Anyith P. Cruz-Amaya, Daniel López-Guillén, M. Laura Mesa- Almagro and José A. Moreno-Moreno

By Michele Mario Ciulla, Gianluca Lorenzo Perrucci and Fabio Magrini

By Fahd Azzabi Zouraq, Meline Stölting and Daniel Eberli

By Stefano Sivolella, Marleen De Biagi, Giulia Brunello, Sara Ricci, Drazen Tadic, Christiane Marinc, Diego Lops, Letizia Ferroni, Chiara Gardin, Eriberto Bressan and Barbara Zavan

By Masahiro Kameda

By Isabelle Gendreau, Laetitia Angers, Jessica Jean and Roxane Pouliot

By Yingai Shi, YuLin Li, JinYu Liu and Yuanyuan Zhang

By Norbert W. Guldner, Peter Klapproth and Hans-H. Sievers

By Raffaele Girlanda

IMPACT OF THIS BOOK AND ITS CHAPTERS

109,142 Total Chapter Downloads

15,973 Total Chapter Views

115 Crossref Citations

210 Web of Science Citations

306 Dimensions Citations

3 Altmetric Score

Order a print copy of this book

Available on

Amazon

Delivered by

£169 (ex. VAT)*

Hardcover | Printed Full Colour

FREE SHIPPING WORLDWIDE

* Residents of European Union countries need to add a Book Value-Added Tax Rate based on their country of residence. Institutions and companies, registered as VAT taxable entities in their own EU member state, will not pay VAT by providing IntechOpen with their VAT registration number. This is made possible by the EU reverse charge method.

As an IntechOpen contributor, you can buy this book for an Exclusive Author price with discounts from 30% to 50% on retail price.

Log in to your Author Panel to purchase a book at the discounted price.

For any assistance during ordering process, contact us at [email protected]

Related books

Edited by Daniel Eberli

Tissue Engineering

Advances in regenerative medicine.

Edited by Sabine Wislet

Bone Regeneration

Edited by Haim Tal

Tissue Engineering for Tissue and Organ Regeneration

Muscle cell and tissue.

Edited by Kunihiro Sakuma

Tissue Regeneration

Edited by Jamie Davies

Edited by Hussein Abdelhay Essayed Kaoud

Current Basic and Pathological Approaches to the Function of Muscle Cells and Tissues

Edited by Haruo Sugi

Call for authors

Submit your work to intechopen.

phd tissue engineering and regenerative medicine

Biomedical Engineering

Marco C. Bottino, D.D.S., MSc., Ph.D.

[email protected]

School of Dentistry, Room 2303 1011 N University Ave Ann Arbor, MI 48109

(734) 763-2206

Primary Website

Bottino Lab

Regenerative Dentistry Graduate Program

Research Interests

My lab focuses on identifying and translating regenerative materials and technologies to reestablish dental, oral, and craniofacial (DOC) tissue health. We use both in vitro and in vivo pre-clinical animal models to gain further insight into the potential clinical safety and efficacy of the developed biomaterials and overall regenerative strategies.

Research Interests: Biofabrication, 3D Bioprinting

Research Areas:

Biomaterials, Drug Delivery and Therapeutics, Tissue Engineering and Biomaterials, Tissue Engineering and Regenerative Medicine

The University of Manchester

Alternatively, use our A–Z index

Attend an open day

Download our course brochure

Discover more about Medicine at Manchester

MSc Tissue Engineering for Regenerative Medicine / Overview

Year of entry: 2024

  • View full page

We require an honours degree (minimum Upper Second) or overseas equivalent in:

  • biological sciences
  • medical sciences
  • biomedical materials
  • veterinary medicine
  • pharmacology

Full entry requirements

Please apply via our online application form. See the application and selection section for details of the supporting documents we require.

We recommend that you apply as early as possible. We reserve the right to close applications if the course is full.

Course options

Course overview.

  • Learn how to research strategies to repair, replace and ultimately regenerate various tissues and organs to solve major clinical problems.
  • Gain a comprehensive insight into topical issues such as stem cells, design and characterisation of biomaterials and nanomaterials (including graphene), biofabrication (including 3D bioprinting), cell and gene therapies, commercialisation and clinical translation of regenerative therapies.
  • Prepare for PhD study, specialist clinical training or a career in related industries, including pharmaceutical, biotechnology and regenerative medicine sectors.
  • Study at a university ranked 8th in the UK and among the top 30 in the world for Medicine (QS Rankings 2023).

For entry in the academic year beginning September 2024, the tuition fees are as follows:

  • MSc (full-time) UK students (per annum): £14,500 International, including EU, students (per annum): £34,500

Further information for EU students can be found on our dedicated EU page.

The fees quoted above will be fully inclusive for the course tuition, administration and computational costs during your studies.

All fees for entry will be subject to yearly review and incremental rises per annum are also likely over the duration of courses lasting more than a year for UK students (fees are typically fixed for international students for the course duration at the year of entry).

For general fees information please visit postgraduate fees. Always contact the department if you are unsure which fee applies to your qualification award and method of attendance.

Policy on additional costs

All students should normally be able to complete their programme of study without incurring additional study costs over and above the tuition fee for that programme. Any unavoidable additional compulsory costs totalling more than 1% of the annual home undergraduate fee per annum, regardless of whether the programme in question is undergraduate or postgraduate taught, will be made clear to you at the point of application. Further information can be found in the University's Policy on additional costs incurred by students on undergraduate and postgraduate taught programmes (PDF document, 91KB).

Scholarships/sponsorships

Contact details, courses in related subject areas.

Use the links below to view lists of courses in related subject areas.

  • Anatomical Science
  • Biological Sciences
  • Biomedical Sciences
  • Biochemistry
  • Molecular Biology
  • Nanoscience

Regulated by the Office for Students

The University of Manchester is regulated by the Office for Students (OfS). The OfS aims to help students succeed in Higher Education by ensuring they receive excellent information and guidance, get high quality education that prepares them for the future and by protecting their interests. More information can be found at the OfS website .

You can find regulations and policies relating to student life at The University of Manchester, including our Degree Regulations and Complaints Procedure, on our regulations website .

phd tissue engineering and regenerative medicine

Human Nasal Inferior Turbinate-Derived Neural Stem Cells Improve the Niche of Substantia Nigra Par Compacta in a Parkinson’s Disease Model by Modulating Hippo Signaling

  • Original Article
  • Open access
  • Published: 10 April 2024

Cite this article

You have full access to this open access article

  • Junwon Choi 1 , 2 ,
  • Sun Wha Park 1 , 2 ,
  • Hyunji Lee 1 , 2 ,
  • Do Hyun Kim 1 &
  • Sung Won Kim   ORCID: orcid.org/0000-0002-8981-2536 1 , 2  

Explore all metrics

Background:

Parkinson’s disease (PD) is one of the most prevalent neurodegenerative diseases, following Alzheimer’s disease. The onset of PD is characterized by the loss of dopaminergic neurons in the substantia nigra. Stem cell therapy has great potential for the treatment of neurodegenerative diseases, and human nasal turbinate-derived stem cells (hNTSCs) have been found to share some characteristics with mesenchymal stem cells. Although the Hippo signaling pathway was originally thought to regulate cell size in organs, recent studies have shown that it can also control inflammation in neural cells.

Dopaminergic neuron-like cells were differentiated from SH-SY5Y cells (DA-Like cells) and treated with 1-Methyl-4-phenylpyridinium iodide to stimulate Reactive oxidative species (ROS) production. A transwell assay was conducted to validate the effect of hNTSCs on the Hippo pathway. We generated an MPTP-induced PD mouse model and transplanted hNTSCs into the substantia nigra of PD mice via stereotaxic surgery. After five weeks of behavioral testing, the brain samples were validated by immunoblotting and immunostaining to confirm the niche control of hNTSCs.

In-vitro experiments showed that hNTSCs significantly increased cell survival and exerted anti-inflammatory effects by controlling ROS-mediated ER stress and hippocampal signaling pathway factors . Similarly, the in-vivo experiments demonstrated an increase in anti-inflammatory effects and cell survival rate. After transplantation of hNTSCs, the PD mouse model showed improved mobility and relief from PD symptoms.

Conclusion:

hNTSCs improved the survival rate of dopaminergic neurons by manipulating the hippocampal pathway through Yes-associated protein (YAP)/transcriptional coactivator with a PDZ-binding motif (TAZ) by reducing inflammatory cytokines. In this study, we found that controlling the niche of hNTSCs had a therapeutic effect on PD lesions.

Avoid common mistakes on your manuscript.

1 Introduction

Parkinson’s disease is the second most prevalent neurodegenerative disorder and affects motor function, causes resting tremors, and affects the autonomic nervous system [ 1 , 2 ]. The etiology of the disease is not clear, but alpha-synuclein (αSyn) deposition and the Lewy-body (LB) formation have been observed to affect the loss of dopaminergic neurons in the substantia nigra par compacta through the post mortem analyses [ 3 , 4 ]. Syn is a small abnormal protein that is found in various cell types, mostly in the presynaptic terminals of the central nervous system [ 5 ]. The domains of αSyn play a role membrane attachment and fibril aggregation. The N-terminal domain of αSyn is composed of an amphipathic α-helix structure which has high affinity to the mitochondrial membrane. The C-terminal domain contains numerous negatively charged amino acids including Ser129. This promotes αSyn aggregation by strengthening the interaction between metal ions and proteins [ 6 , 7 , 8 ]. In contrast, ROS are by-products of cellular metabolism in the mitochondria. ROS is necessary for maintaining mitochondrial homeostasis through the mitophagy quality control system [ 9 , 10 , 11 ]; however, aggregated αSyn bounds to the mitochondria membrane and disrupts electron transfer system (ETS) complex I, which leads to increase ROS levels in mitochondria [ 10 , 12 ]. Excessive ROS levels alter mitochondrial membrane permeability and lead to mitochondrial dysfunction and cell death [ 13 , 14 ]. Thus, αSyn deposition in the central nerve system produces ROS, which disrupts mitochondrial homeostasis [ 15 , 16 , 17 , 18 ].

The Hippo signaling pathway controls organ size, cancer development, and tissue regeneration [ 19 , 20 ]. Recent studies have shown that the Hippo-signaling pathway also plays a critical role in controlling neuroinflammation, neuronal cell differentiation, and neuronal cell death [ 21 , 22 ]. The canonical Hippo-signaling pathway is mainly composed of mammalian Ste20-like kinases 1/2 (MST 1/2), large tumor suppressor 1/2 (LATS 1/2), YAP/TAZ. MST 1/2 phosphorylates LAT 1/2, which leads to the phosphorylation of the downstream YAP/TAZ under oxidative stress. Phosphorylation of YAP/TAZ prevents their translocation into the nucleus and interaction with cytosolic protein 14-3-3, resulting in cell death [ 23 ]. In contrast, Unphosphorylated YAP/TAZ translocates to the nucleus and binds to the TEAD family of transcription factors, promoting the expression of genes related to cell proliferation, differentiation, and survival [ 24 , 25 ].

Mesenchymal stem cells (MSCs) are promising for cell therapy because of their immunomodulatory and therapeutic functions in lesions. A sufficient number of cells are needed for clinical trials. MSCs have various sources, such as bone marrow, umbilical cord blood, and adipocytes, and are relatively easy to obtain. Several studies have investigated the therapeutic effects of MSCs [ 26 , 27 ], and MSCs have been observed to secrete anti-inflammatory cytokines with immune suppression effects [ 28 , 29 ]. Human nasal turbinate-derived neural crest stem cells (hNTSCs) represent a new source of light for the treatment of neurodegenerative diseases. hNTSCs show characteristics similar to MSCs, are relatively functional, and differentiate into neurons and astrocytes. As hNTSCs are derived from the neural crest and have neural lineage features, hNTSCs have better compatibility than MSCs [ 30 , 31 , 32 ]. Restoring the nigrostriatal pathway through dopaminergic neuron regeneration is a promising method for the treatment of Parkinson’s [ 33 , 34 ]. Furthermore, improving the survival rate of the transplanted cells is important for successful therapeutic effects [ 35 , 36 ]. In this study, we generated a Parkinson’s disease model using SH-SY5Y cells that differentiated into dopaminergic neurons and treated them with 1-methyl-4-phenyl pyridinium (MPP + ) and utilized a MPTP-mediated mice model. The substantia nigra par compacta was relieved by niche control of hNTSCs in the lesion.

2 Materials and methods

2.1 hntscs generation and cell culture.

The study using hNTSCs followed the guidelines of the Institutional Review Board of Seoul St. Mary’s Hospital (KC08TISS0341), Catholic University of Korea, including informed consent regulations and the Declaration of Helsinki. Before surgery, the participants provided written informed consent to participate in the study. The tissue was donated by patient who conducted hypertrophied nasal inferior turbinate volume reduction surgery. hNTSCs were isolated from the discarded tissue following partial turbinectomy using previously described methods [ 30 ]. First, the tissues were washed with 0.9% saline and phosphate-buffered saline (Thermo Fisher Scientific). The specimens were cut into 1 mm pieces and placed in a culture dish. Finally, a sterilized glass cover slide was placed on top. The tissue was placed in a humidified incubator and maintained at a temperature of 37 °C with 5% CO 2 . The specimen was cultured in α-minimum essential medium (α-MEM; Thermo Fisher Scientific) which was supplemented with 1% penicillin/streptomycin (Invitrogen, CA, USA), and 10% fetal bovine serum (FBS; Thermo Fisher Scientific). The culture medium was changed every two days for three weeks. The cells were harvested from the tissue by removing the glass cover slide and using 0.25% trypsin and 1 mM EDTA solution. hNTSCs were cultured and expanded for use in experiments.

2.2 hBM-MSC generation and cell culture

Healthy donors provided human bone marrow aspirates from the iliac crest following approval from the Institutional Review Board of Seoul St. Mary’s Hospital (KC10CSSE0651). Bone marrow aspirates were sent to the good manufacturing practice-compliant facility of the Catholic Institute of Cell Therapy (Seoul, Korea, http://www.cic.re.kr ) for hBM-MSCs isolation, expansion, and quality control, with written consent from the participants [ 26 ]. The hBM-MSCs were cultured in Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 1% penicillin/streptomycin (Invitrogen) and 20% FBS (Thermo Fisher Scientific). The cells were incubated at 37 °C in a humidified atmosphere containing 5% of CO 2 .

2.3 SH-SY5Y cell culture and differentiation

SH-SY5Y cells were purchased from the Korean Cell Line Bank (Seoul, Korea). The cells were cultured in the humidified incubator, which maintained 37 °C and contained 5% CO 2 , in α-MEM supplemented with 1% penicillin/streptomycin (Invitrogen), and 10% FBS (Thermo Fisher Scientific). SH-SY5Y cells were seeded at a density of 3 × 10 4 cells in a 6-well-plate, and the differentiation medium was changed according to a previously described protocol [ 37 ].

2.4 Trans-well assay

DA-like cells (1 × 10 6 cells of DA-like cells were cultured in the bottom chamber, treated with 500 mM MPP + in culture medium overnight, and the following day, the medium was replaced with fresh medium. One day later, 5 × 10 5 hNTSCs and hBC-MSCs were cultured in the upper chamber, after being placed at the chamber bottom, and incubated overnight.

2.5 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mouse model generation

Seven-week-old C57BL/6N mice were obtained from Orient Bio (Gyeonggi-do, South Korea). Ten mice were randomly assigned to one of four groups: control, MPTP-saline, MPTP_hNTSCs, and MPTP_hBC-MSCs. MPTP was administered at a dose of 25 mg/kg for seven consecutive days. The motor dysfunction of the PD mouse model was validated using the rotarod and open field tests.

2.6 Stereotaxic surgery

hNTSCs and hBM-MSCs were incubated for 5 min with trypsin–EDTA (Thermo Fisher Scientific), centrifuged, and then, the supernatant was removed. Mice were anesthetized, and their heads were fixed in a stereotaxic apparatus. After cleaning the surface of the skull, according to the Allen Brain Atlas, the distance between the two points of the lambda and bregma was identified. The hNTSCs and hBM-MSCs [1 × 10 5 cells/10 µl N-Acetyl-L-cysteine(NAC)] were injected into the substantia nigra par compacta. The coordinates were as follows: AP = − 3.2, ML = 0.71 mm and DV = 4.6 mm.

2.7 Behavior test

One week after MPTP IP injection, the motor function of the mice was validated by rotarod and open field tests, using a rotarod machine with falling sensors (MED-Associates Inc., FL, USA). In the rotarod test, ten mice for each group were placed side by side in a rotarod machine. The mice were allowed to remain in the machine for 5 min. Each session was performed after habituation. The acceleration of the rotation and rotation speed were set to 0–15 m/min and 0–50 rpm, respectively. Each rail on the machine was turned off as soon as the mouse fell off the platform. The rotarod test was conducted consecutively for four days. The open field test was conducted using SMART software (version 3.0) and an open field box (Panlab, MA, USA). The open field box was a 42 × 42 × 42 cm polyvinyl chloride box, which was monitored and recorded using a camera connected to the SMART 3.0 software. The camera measured the movements of the mice in the peripheral and central zones. Trajectory tracing, total travel distance, and time spent in the zone were measured to analyze the motor function of the animal.

2.8 Flow cytometry

hNTSCs (1 × 10 5 cells) were collected in a round bottom tube and resuspended to 100 μL of 5% FBS (Thermo Fisher Scientific) diluted in Dulbecco’s phosphate-buffered saline (DPBS; Thermo Fisher Scientific). Cells were stained with human CD90-PE conjugated (BD Bioscience, NJ, USA) and CD34 (FITC) at RT for 2 h. The cells were then evaluated using a FACSAria III (BD Biosciences) machine.

2.9 Immunofluorescence staining

DA-Like cells were washed three times with DPBS (Thermo Fisher Scientific) and then fixed in cold methanol on ice for 15 min. After fixation, normal goat serum was used as a blocking buffer, for an hour at RT. Primary antibodies were diluted to 1:100 in blocking buffer and added to the cells. The cells were incubated overnight at 4 °C and then washed with DPBS three times. Secondary antibodies were diluted 1:500 in DPBS and incubated in the buffer for an hour. DAPI (Vector Laboratories) was diluted 1:5000 and incubated with the samples for 5 min at RT. The brain of MPTP-induced PD mouse model was fixed in 4% paraformaldehyde for 2 h and incubated in 30% of sucrose until it is crysectioned. The brain was sectioned 7 μm from the bregma to the end. The brain section slide was selected by referring to mouse brain atlas map and the slide was blocked with blocking buffer for an hour at RT. Primary antibody was diluted to 1:100 in blocking buffer and added to the slide. The slide was incubated overnight at 4 °C and washed with DPBS three times. Secondary antibody was diluted 1:500 in DPBS and incubated with the slide for an hour. DAPI was diluted 1:5000 and incubated for 5 min at RT. The fluorescence intensity of the cells and the slide were measured using a Zeiss LSM 800 Confocal Laser Scanning Microscope (Zeiss, Jena, Germany).

2.10 Protein isolation

Brain proteins were extracted using RIPA buffer (LPS Solution, Daejeon, Korea) with EDTA-free Complete Ultra tablets and an EASYpack Protease Inhibitor Cocktail (Roche, Basel, Switzerland). The tissues were homogenized using a tissue grinder with lysis buffer containing proteinase and phosphatase inhibitors. Lysates were then sonicated for 10 secs in an ice bath sonication machine, followed by centrifuging at 14,000 × g for 20 min at 4 °C. The protein concentrations were measured using a bicinchoninic acid protein assay kit (Thermo Fisher Scientific).

2.11 Immunoblotting

The proteins (15 μg) used for immunoblotting were mixed with 5 μL of NuPAGE™ LDS Sample Buffer (4x; Thermo Fisher Scientific), 2 μL of dithiothreitol (10x; Sigma-Aldrich, MO, USA), and distilled water, added to a final volume of 20 μL. The protein mixture was heated up to 75 °C for 10 min to denature the proteins. The protein mixture then underwent electrophoresis in a 10% SDS-PAGE gel for 40 min, in Invitrogen™ NuPAGE™ MOPS SDS Running Buffer (20x; Invitrogen). Proteins were separated by SDS-PAGE using the Trans-Blot Turbo Transfer System (Bio-Rad, Hercules, CA, USA). The PVDF membranes were blocked with 5% skim milk for 2 h at RT. The membranes were then washed three times with 0.1% tween-20 diluted in tris-buffered saline (0.1% T-TBS) for 5 min, three times, at RT. The transferred PVDF was then incubated with the primary antibody, either eukaryotic translation initiation factor 2 alpha subunit (EIF2α; Cell signaling technology, MA, USA), phosphorylated EIF2α activating transcription factor 4 (ATF4, Cell signaling technology), NEDD4L (Cell signaling), Yes-associated protein/transcription activator with PDZ binding motif (YAP/TAZ; Cell signaling), phosphorylated YAP/TAZ (Cell signaling), Large neutral amino acids transporter small subunit 1(LAT1; Cell signaling), or phosphorylated LAT1(Cell signaling). All the antibodies were diluted to 1:1000 with 0.1% sodium azide (Sigma-Aldrich) and incubated with the membranes at 4 °C overnight. The PVDF membranes were then washed with T-TBS for 10 min, three times, and then incubated with a goat anti-rabbit IgG antibody labeled with peroxidase (Vector Laboratory, Inc., CA, USA), diluted 1:5000, for 10 min at RT. The PVDF membranes were then analyzed using an LAS-3000 (FUJI PHOTO FILM CO., LTD, Tokyo, Japan).

2.12 RNA isolation and cDNA synthesis

Midbrain tissue was homogenized using a tissue grinder and 300 μL of TRIzol ® reagent (Invitrogen). Tissue lysates were added to 60 μL of chloroform (DAEJUNG CHEMICALS & METALS Co., Ltd, Gyeonggi-do, Korea) and vortexed for 3 secs, to mix. The lysates were then centrifuged at 12,000 × g for 15 min at 4 °C. Supernatants were transferred to new 1.7 mL tubes, incubated in 0.5 mL of isopropanol (Sigma-Aldrich), and centrifuged at 12,000 × g for 15 min at 4 °C. The resulting RNA pellet was washed with 70% cold ethanol and centrifuged at 12,000 × g for 10 min at 4 °C. The RNA pellet was then dissolved in 30–50 μL of diethyl pyrocarbonate-treated water, according to the size of the pellet. The RNA was then incubated for 10 min at 65 °C. cDNA was synthesized from 1 µg of total RNA using the iScript™ cDNA Synthesis Kit (Bio-RAD).

2.13 Poly-chain reaction (PCR) assay

The mRNA was isolated from the midbrain of each group using a manual protocol. cDNA was synthesized using the iScript™ cDNA Synthesis Kit (Bio-RAD). Then, 1 µg of cDNA was used for gene expression level analysis. CFX386 touch (Bio-RAD) was used to conduct qRT-PCR. The reaction efficiency and number of cycles were determined using innate software.

2.14 Statistics

The Kruskal–Wallis and Mann–Whitney U post-hoc tests were used to compare the results across groups. SPSS version 22 (IBM Corporation, NY, USA) was used to determine statistically significant differences between groups. The results are expressed as mean ± standard deviation (SD). Differences were considered significant at * p  < 0.05 versus Control; ** p  < 0.01 versus Control; *** p  < 0.001 versus control; # p  < 0.05 versus MPTP-saline and MPP + -saline; ## p  < 0.01 versus MPTP-saline and MPP + -saline; ### p  < 0.001 versus MPTP-saline and MPP + -saline.

3.1 hNTSCs generation and characteristics analysis

Nasal turbinate-derived normal tissues were obtained from each patient. The tissues were washed several times with DPBS (−/−) and reduced to a size of 1.5 cm. The tissue was chopped into a 1 mm size sample, placed in a cell culture dish under an autoclaved cover glass, and incubated in growth media for three weeks (Fig.  1 A). hNTSCs expressed neuronal stem cell bio markers TUJ1, Nestin, P75, and NGFR [ 31 ]. To confirm the characteristics that are similar to those of hBM-MSCs, CD34 and CD90 that are considered stem cell surface markers. Notably, the hNTSCs showed similar characteristics to hBM-MSCs in the flow cytometry analysis (Fig.  1 B, C).

figure 1

hNTSCs generated from nasal turbinate tissue. Nasal turbinate normal tissue donated from patients was chopped into 1 mm size and placed in a culture dish under the autoclaved cover slide. The dishes were then filled with culture media, and hNTSCs were grown from the tissue ( A ). Images show CD 90 (PE), CD 34 (FITC), stem cell marker flow cytometry data of hBM-MSCs ( B ). Images show the CD 90 (PE), CD 34 (FITC), and stem cell marker flow cytometry data of hNTSCs ( C ). bar = 50 μm

3.2 hNTSCs foster an MPP + -induced dopaminergic neuron-like cell niche

The SH-SY5Y human neuroblastoma cell line relies on gradual serum deprivation and retinoic acid supplementation, which reduce the period of time for differentiation into DA-Like cells. SH-SY5Y cells were cultured to differentiate into DA-Like cells in the bottom chamber of a trans-well chamber for three weeks (Fig.  2 A). Three types of differentiation media composed of differentiation-related small molecules and chemicals were used (Table  1 ). The cells were completely differentiated by day 18 (Supplementary 1 , 2 ). The fully differentiated DA-like cells were treated with a solution of 500 mM of MPP + overnight. hNTSCs and hBM-MSCs were cultured in the upper chamber of a transwell culture dish. The relative TH/TUJ1 immunofluorescent staining intensity of DA-Like cells showed that cells treated with MPP + saline had a significantly decreased intensity compared to that of the control group, MPP + _hNTSCs, and MPP + _hBM_MSCs. MPP + hNTSCs and MPP + hBM_MSCs showed increased relative intensity compared to MPP + saline-treated cells. Notably, MPP + hNTSCs showed no significant differences from MPP + hBM_MSCs.

figure 2

Differentiating SH-SY5Y cells to dopaminergic neuron-like cells. The diagram shows the morphological changes of SH-SY5Y cells during differentiation ( A ). The images show the TH and Tuj1 immunofluorescence staining of DA-Like cells after MPP + treatment ( B ). The graph indicates the intensity of TH/Tuj1 of DA-Like cells ( C ). The graph indicates the impact of a ROS concentration gradient on SH-SY5Y derived DA-Like cells ( D ). * p  < 0.05 versus control; *** p  < 0.001 versus control; ### p  < 0.001 versus MPP + -saline. bar = 50 μm

3.3 hNTSCs modulate hippo-pathway signaling factors of MPP + induced dopaminergic neuron-like cells

MPP + -induced PD DA-like cells are damaged by ROS-mediated ER stress in the cytoplasm, and the Hippo signaling pathway is involved in cell death. The protein expression levels of phosphorylated EIF2α and ATF4 of the MPP + saline group were significantly higher than those of control, MPP + _hNTSCs, and MPP + _hBM_MSCs cohorts. In contrast, the protein expression level of NEDD4.2, which inhibits the phosphorylation of LAT1, was lower than that of the control, MPP + _hNTSCs, and MPP + _hBM_MSCs groups (Fig.  3 A). Furthermore, the protein expression levels of phosphorylated LAT1 and YAP/TAZ in the MPP + _saline group were significantly higher than those in the control, MPP + _hNTSCs, and MPP + _hBM_MSCs groups (Fig.  3 B). The relative gene expression levels of inflammatory factors, TNF-α, IL-6, and INF-γ, were significantly decreased in MPP + _hNTSCs and MPP + _hBM-MSCs groups, compared to the MPP + _saline group (Fig.  3 C–E). The sequence is informed in Table  2 .

figure 3

Images show the western blot results of ER-Stress factors, EIF2α, p-EIF2α, ATF4, NEDD4.2, and β-actin. The immunoblotting was performed on MPP + -treated DA-like cells ( A ). Images show the results for hippo-signaling pathway factors, p-MST1/2, LAT1/2, p-LAT1/2, YAP/TAZ, p-YAP/TAZ, upon immunoblotting of MPP + -treated DA-like cells ( B ). The graph indicates the relative gene expression levels of TNF-α, IL-6, and interferon-γ in DA-like cells ( C – E ). ** p  < 0.01 versus control; *** p  < 0.001 versus control; # p  < 0.05 versus MPP + -saline; ## p  < 0.01 versus MPP + -saline; ### p  < 0.001 versus MPP + -saline

3.4 MPTP-induced Parkinson’s disease mouse model generation and validation

Seven-week old male mice were used as a PD mimic animal model, and each group consisted of 10 mice. The mice were separated randomly and intraperitoneally injected with MPTP (25 mg/kg IP for a week, consecutively). The animals were validated by PD modeling using the rotarod and open field tests. Most mice injected with MPTP showed motor dysfunction, which was validated by the rotarod and open field tests. Latency, drop speed, and distance were significantly decreased in MPTP-injected mice (Fig.  4 B). Furthermore, in the open field test, the MPTP-injected mice showed a significant decrease in motor function (Fig.  4 C). The PD mouse model was then subjected to stereotaxic surgery two days after the behavioral test. The hNTSCs and hBM-MSCs were transplanted into the substantia nigra pars compacta via stereotaxic surgery (Fig.  4 A).

figure 4

MPTP-mediated PD animal model generation and validation. The diagram indicates the MPTP-induced PD animal preparation schematic and a list of behavior tests ( A ). The graph shows the results of the first week of the Rotarod test in the MPTP-induced PD animal model ( B ). The graph shows the first week of the open field test of the MPTP-induced PD animal model ( C ). *** p  < 0.001 versus control

3.5 hNTSCs regulate hippo-pathway signaling factors of the MPTP-induced PD mouse model

The mice were sacrificed four weeks after cell transplantation. The midbrains were isolated and homogenized for immunoblotting and PCR analyses. eIF2α, p-eIF2α, ATF4, and NEDD4.2 primary antibodies were used to confirm the presence of ROS-mediated ER-stress. The protein expression level of p-eIF2α and ATF4 were downregulated in MPTP-hNTSCs and MPTP-hBM-MSCs, compared to those of the MPTP-saline group, and NEDD4.2 levels were upregulated (Fig.  5 A). The protein levels of p-MST 1/2, p-LAT 1/2, and p-YAP were downregulated in MPTP-hNTSCs and MPTP-hBM-MSCs, compared to the respective levels in the MPTP-saline group (Fig.  5 B). The inflammatory gene expression levels of TNF-α, IL-6, and INF-γ were significantly downregulated as well (Fig.  5 C–E and Table  3 ).

figure 5

The images show the immunoblotting results for ER-Stress factors, EIF2α, p-EIF2α, ATF4, NEDD4.2, and β-actin from midbrain samples of MPTP-induced Parkinson’s model mice ( A ). Images indicate the immunoblotting results of hippo-signaling pathway factors, p-MST1/2, LAT1/2, p-LAT1/2, YAP/TAZ, p-YAP/TAZ, from mid-brain samples of MPTP-induced Parkinson’s model mice ( B ). The graphs indicate TNF-α, IL-6 and INF-γ gene expression levels ( C – E ). ** p  < 0.01 versus control; *** p  < 0.001 versus control; # p  < 0.05 versus MPTP-saline; ## p  < 0.01 versus MPTP-saline; ### p  < 0.001 versus MPTP-saline

3.6 hNTSCs restore the dopaminergic neuron of MPTP-induced PD mouse model

The brains of the MPTP-induced PD mouse model were cut 7 μm cryo-section and stained to TH antibody to validate the influence of hNTSCs on dopaminergic neuron survival (Fig.  5 A). The MPTP-hNTSCs and MPTP-hBM-MSCs group showed higher number of TH positive cells than those of MPTP-saline group statistically. However, MPTP-hNTSCs and MPTP-hBM-MSCs group showed less number of TH positive cells than those of control group. On the other hands, MPTP-hNTSCs group showed higher number of TH positive cells than those of MPTP-hBM-MSCs group statistically (Fig.  5 B).

3.7 The MPTP-induced PD mouse model presented recovered and improved motor function

The mice showed valid behavioral differences during the four weeks after cell transplantation. The rotarod test showed a gradual restoration of latency, drop speed, and travel distance of hNTSCs and hBM-MSCs, compared with those of the MPTP-saline group (Fig.  6 A). In the open field trajectory test, the trace data also showed increased mobility of MPTP-hNTSCs and MPTP-hBM-MSCs, compared to that of the MPTP-saline group (Fig.  6 B). In addition, the travel distance and time in the zone also reflected increased mobility (Figs. 6 C and 7 ).

figure 6

Presenting images indicate tyrosine hydroxylase (TH) positive immunofluorescence staining, DAPI (Blue), TH (Green) ( A ). The graph indicates the number of TH cell/DAPI ( B ). *** p  < 0.001 versus control; ## p  < 0.01 versus MPTP-saline. Scale bar = 200 μm

figure 7

MPTP-induced PD animal model validation after cell transplantation. The graph shows the results of the Rotarod test over four weeks ( A ). The graph shows the result of the Rotarod test on the fourth week The images indicate the trajectory tracking of animal models in the open field test on the fourth week ( B ). The graph indicates the travel distance, zone distance and time in zone data over four weeks ( C ) * p  < 0.05 versus control; ** p  < 0.01 versus control; *** p  < 0.001 versus control; # p  < 0.05 versus MPTP-Saline; ## p  < 0.01 versus MPTP-Saline; ### p  < 0.001 versus MPTP-Saline

4 Discussion

Stem cell therapy has been highlighted as a regenerative medicine for neurodegenerative diseases such as Alzheimer’s disease and Parkinson’s disease. Despite considerable efforts to elucidate the etiology of Parkinson’s disease, Parkinson’s disease modeling is still having difficulty implementing an accurate model. The human neuroblastoma SH-SY5Y cell line is an alternative source of human Parkinson’s disease modelling, and it is relatively easy to differentiate dopaminergic neuron-like cell phenotypes within three weeks upon lowering FBS to 1% and adding 10 μM retinoic acid. SH-SY5Y-derived dopaminergic neuron-like cells not only express DJ-1 protein and TH, which are extensively related to the early onset of PD, but also the dopamine transporter (DAT), which regulates dopamine homeostasis, incorporates MPP + , and can be utilized in MPP + -induced neurotoxicity [ 38 , 39 ]. MPP + induces ROS production, which disrupts the electron transportation system of the mitochondria and leads to DNA laddering. Also, alpha-synuclein (α-syn) protein aggregation commences the ROS stress in mitochondria and the α-syn overexpression and aggregation are affected by numeral genes such as apolipoprotein E (APOE). In Parkinson’s disease, APOE-ε4, one of the phenotypes of APOE, is well known to promote neurodegenerative disease. APOE-ε4 is highly expressed in PD patients and the overexpressed APOE-ε4 accelerates aggregating α-syn in the lesion, leading to ROS stress in the lesion [ 40 ]. In this study, we mimicked ros-mediated neurotoxicity by using the MPP + . In the transwell assay, MPP + -treated dopaminergic neuron-like cells showed shortened dendrites and neuronal cell death. In contrast, the Hippo signaling pathway is well known for its role in organ size, cancer development, and tissue regeneration. There are many hurdles to overcome in the perception of transplanting cells into lesions. First, it is important to improve the viability of the transplanted cells to regenerate their functions. The lesion of substantia nigra in Parkinson’s disease is upregulated and produced higher levels of inflammatory factors, TNF-α, Interleukin-6, and interferon gamma. The upregulation of these inflammatory factors and cytokines induces glial cell migration and exacerbates inflammation, which affects transplanted cell viability and leads to a lower engraftment rate. hNTSCs show characteristics similar to those of mesenchymal stem cells (MSCs), and the anti-inflammatory effects of MSCs are well known, though many studies before [ 27 , 28 , 29 ]. The trans-well in vitro assay showed that MPP + saline-treated dopaminergic neuron-like cells upregulated inflammatory genes, TNF-α, and IL-6. However, after the hNTSCs were placed in the upper chamber, the expression of the inflammatory genes was downregulated, while anti-inflammatory gene expression levels were upregulated. The MPTP-induced in vivo test showed results similar to those of the in vitro test. Inflammatory gene expression levels in the mid-brain were downregulated in MPTP-hNTSCs and MPTP-hBC-MSCs groups, whereas anti-inflammatory gene expression levels were upregulated. In the live/dead assay with brain slides, the substantia nigra showed better cell viability in the lesions. Moreover, because hNTSCs are derived from the neural crest, they express neural lineage markers such as nestin, neural growth factor receptor p75, and sox2, which are more compatible with the central nervous system when engrafted. Although the same number of cells was transplanted into the MPTP-induced PD model, hNTSCs showed better cell viability than hBC-MSCs. Both stem cell types exhibit anti-inflammatory effects and relieve niches.

Data availability statement

The datasets generated during and/or analysed during the current study are available from the corresponding author on reasonable request.

Bloem BR, Okun MS, Klein C. Parkinson’s disease. The Lancet. 2021;397:2284–303.

Article   CAS   Google Scholar  

Sveinbjornsdottir S. The clinical symptoms of Parkinson’s disease. J Neurochem. 2016;139:318–24.

Article   CAS   PubMed   Google Scholar  

Wakabayashi K, Tanji K, Odagiri S, Miki Y, Mori F, Takahashi H. The Lewy body in Parkinson’s disease and related neurodegenerative disorders. Mol Neurobiol. 2013;47:495–508.

Spillantini MG, Goedert M. The α-synucleinopathies: Parkinson’s disease, dementia with lewy bodies, and multiple system atrophy. Ann N Y Acad Sci. 2000;920:16–27.

Bendor JT, Logan TP, Edwards RH. The function of α-synuclein. Neuron. 2013;79:1044–66.

Thorne NJ, Tumbarello DA. The relationship of alpha-synuclein to mitochondrial dynamics and quality control. Front Mol Neurosci. 2022;15:947191.

Article   CAS   PubMed   PubMed Central   Google Scholar  

Park JH, Burgess JD, Faroqi AH, DeMeo NN, Fiesel FC, Springer W, et al. Alpha-synuclein-induced mitochondrial dysfunction is mediated via a sirtuin 3-dependent pathway. Mol Neurodegener. 2020;15:5.

Risiglione P, Zinghirino F, Di Rosa MC, Magrì A, Messina A. Alpha-synuclein and mitochondrial dysfunction in Parkinson’s disease: the emerging role of VDAC. Biomolecules. 2021;11:718.

Lin KJ, Lin KL, Chen SD, Liou CW, Chuang YC, Lin HY, et al. The overcrowded crossroads: mitochondria, alpha-synuclein, and the endo-lysosomal system interaction in Parkinson’s disease. Int J Mol Sci. 2019;20:5312.

Ganjam GK, Bolte K, Matschke LA, Neitemeier S, Dolga AM, Höllerhage M, et al. Mitochondrial damage by α-synuclein causes cell death in human dopaminergic neurons. Cell Death Dis. 2019;10:865.

Puspita L, Chung SY, Shim JW. Oxidative stress and cellular pathologies in Parkinson’s disease. Mol Brain. 2017;10:53.

Article   PubMed   PubMed Central   Google Scholar  

Tripathi T, Chattopadhyay K. Interaction of α-synuclein with ATP synthase: switching role from physiological to pathological. ACS Chem Neurosci. 2019;10:16–7.

Fernández-Checa JC, Fernández A, Morales A, Marí M, García-Ruiz C, Colell A. Oxidative stress and altered mitochondrial function in neurodegenerative diseases: lessons from mouse models. CNS Neurol Disord Drug Targets. 2010;9:439–54.

Article   PubMed   Google Scholar  

Wang J, Fröhlich H, Torres FB, Silva RL, Poschet G, Agarwal A, et al. Mitochondrial dysfunction and oxidative stress contribute to cognitive and motor impairment in FOXP1 syndrome. Proc Natl Acad Sci U S A. 2022;119:e2112852119.

Pozo Devoto VM, Falzone TL. Mitochondrial dynamics in Parkinson’s disease: a role for α-synuclein? Dis Model Mech. 2017;10:1075–87.

Angelova PR, Abramov AY. Role of mitochondrial ROS in the brain: from physiology to neurodegeneration. FEBS Lett. 2018;592:692–702.

Liu XL, Wang YD, Yu XM, Li DW, Li GR. Mitochondria-mediated damage to dopaminergic neurons in Parkinson’s disease. Int J Mol Med. 2018;41:615–23.

CAS   PubMed   Google Scholar  

Devi L, Raghavendran V, Prabhu BM, Avadhani NG, Anandatheerthavarada HK. Mitochondrial import and accumulation of alpha-synuclein impair complex I in human dopaminergic neuronal cultures and Parkinson disease brain. J Biol Chem. 2008;283:9089–100.

Wei X, Huang G, Liu J, Ge J, Zhang W, Mei Z. An update on the role of Hippo signaling pathway in ischemia-associated central nervous system diseases. Biomed Pharmacother. 2023;162:114619.

Sahu MR, Mondal AC. The emerging role of Hippo signaling in neurodegeneration. J Neurosci Res. 2020;98:796–814.

Sahu MR, Mondal AC. Neuronal Hippo signaling: from development to diseases. Dev Neurobiol. 2021;81:92–109.

Li X, Li K, Chen Y, Fang F. The role of Hippo signaling pathway in the development of the nervous system. Dev Neurosci. 2021;43:63–70.

Hindley CJ, Condurat AL, Menon V, Thomas R, Azmitia LM, Davis JA, et al. The Hippo pathway member YAP enhances human neural crest cell fate and migration. Sci Rep. 2016;6:23208.

Cheng J, Wang S, Dong Y, Yuan Z. The role and regulatory mechanism of Hippo signaling components in the neuronal system. Front Immunol. 2020;11:281.

Ding R, Weynans K, Bossing T, Barros CS, Berger C. The Hippo signalling pathway maintains quiescence in Drosophila neural stem cells. Nat Commun. 2016;7:10510.

Wei X, Yang X, Han ZP, Qu FF, Shao L, Shi YF. Mesenchymal stem cells: a new trend for cell therapy. Acta Pharmacol Sin. 2013;34:747–54.

Brown C, McKee C, Bakshi S, Walker K, Hakman E, Halassy S, et al. Mesenchymal stem cells: cell therapy and regeneration potential. J Tissue Eng Regen Med. 2019;13:1738–55.

Kim Y-J, Park H-J, Lee G, Bang OY, Ahn YH, Joe E, et al. Neuroprotective effects of human mesenchymal stem cells on dopaminergic neurons through anti-inflammatory action. Glia. 2009;57:13–23.

Miao Z, Sun H, Xue Y. Isolation and characterization of human chorionic membranes mesenchymal stem cells and their neural differentiation. Tissue Eng Regen Med. 2017;14:143–51.

Zhang R, Liu Y, Yan K, Chen L, Chen XR, Li P, et al. Anti-inflammatory and immunomodulatory mechanisms of mesenchymal stem cell transplantation in experimental traumatic brain injury. J Neuroinflamm. 2013;10:871.

Article   Google Scholar  

Lim JY, In Park S, Park SA, Jeon JH, Jung HY, Yon JM, et al. Potential application of human neural crest-derived nasal turbinate stem cells for the treatment of neuropathology and impaired cognition in models of Alzheimer’s disease. Stem Cell Res Ther. 2021;12:402.

Lim JY, Lee JE, Park SA, Park SI, Yon JM, Park JA, et al. Protective effect of human-neural-crest-derived nasal turbinate stem cells against amyloid-β; neurotoxicity through inhibition of osteopontin in a human cerebral organoid model of Alzheimer’s disease. Cells. 2022;11:1029.

Nivet E, Vignes M, Girard SD, Pierrisnard C, Baril N, Devèze A, et al. Engraftment of human nasal olfactory stem cells restores neuroplasticity in mice with hippocampal lesions. J Clin Invest. 2011;121:2808–20.

Hodaie M, Neimat JS, Lozano AM. The dopaminergic nigrostriatal system and Parkinson’s disease: molecular events in development, disease, and cell death, and new therapeutic strategies. Neurosurgery. 2007;60:17–30.

Struzyna LA, Browne KD, Brodnik ZD, Burrell JC, Harris JP, Chen HI, et al. Tissue engineered nigrostriatal pathway for treatment of Parkinson’s disease. J Tissue Eng Regen Med. 2018;12:1702–16.

Osborn TM, Hallett PJ, Schumacher JM, Isacson O. Advantages and recent developments of autologous cell therapy for Parkinson’s disease patients. Front Cell Neurosci. 2020;14:58.

Wenker SD, Leal MC, Farías MI, Zeng X, Pitossi FJ. Cell therapy for Parkinson’s disease: functional role of the host immune response on survival and differentiation of dopaminergic neuroblasts. Brain Res. 2016;1638:15–29.

Lopes FM, Schröder R, da Frota Jr ML, Zanotto-Filho A, Müller CB, Pires AS, et al. Comparison between proliferative and neuron-like SH-SY5Y cells as an in vitro model for Parkinson disease studies. Brain Res. 2010;1337:85–94.

Yun JW, Ahn JB, Kwon E, Ahn JH, Park HW, Heo H, et al. Behavior, PET and histology in novel regimen of MPTP marmoset model of Parkinson’s disease for long-term stem cell therapy. Tissue Eng Regen Med. 2016;13:100–9.

Pang S, Li J, Zhang Y, Chen J. Meta-analysis of the relationship between the APOE gene and the onset of Parkinson’s disease dementia. Parkinsons Dis. 2018;2018:9497147.

PubMed   PubMed Central   Google Scholar  

Download references

Acknowledgements

This research was supported by a National Research Foundation of Korea (NRF) grant funded by the Ministry of Science and ICT (Grant Number 2021M3F7A1083232) and Korean Fund for Regenerative Medicine (KFRM) grant funded by the Korean Government (23C0121L1).

Author information

Authors and affiliations.

Department of Otolaryngology-Head and Neck Surgery, College of Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, Republic of Korea

Junwon Choi, Sun Wha Park, Hyunji Lee, Do Hyun Kim & Sung Won Kim

Postech-Catholic Biomedical Engineering Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea

Junwon Choi, Sun Wha Park, Hyunji Lee & Sung Won Kim

You can also search for this author in PubMed   Google Scholar

Corresponding author

Correspondence to Sung Won Kim .

Ethics declarations

Conflict of interest.

The authors declare that there is no conflict of interest.

Ethical statement

The study was conducted in compliance with the Institutional Review Board of Seoul St. Mary’s Hospital, Catholic University of Korea (IRB no. KC10CSSE0651), informed consent regulations, and the Declaration of Helsinki. Before surgery, the patients provided written informed consent to participate in the study.

Additional information

Publisher's note.

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Below is the link to the electronic supplementary material.

Supplementary file1 (DOCX 1217 kb)

Rights and permissions.

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/ .

Reprints and permissions

About this article

Choi, J., Park, S.W., Lee, H. et al. Human Nasal Inferior Turbinate-Derived Neural Stem Cells Improve the Niche of Substantia Nigra Par Compacta in a Parkinson’s Disease Model by Modulating Hippo Signaling. Tissue Eng Regen Med (2024). https://doi.org/10.1007/s13770-024-00635-3

Download citation

Received : 03 November 2023

Revised : 30 January 2024

Accepted : 15 February 2024

Published : 10 April 2024

DOI : https://doi.org/10.1007/s13770-024-00635-3

Share this article

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

  • Cell-based therapy
  • Regenerative medicine
  • Parkinson’s disease
  • Human nasal turbinate derived stem cells
  • Niche control
  • Find a journal
  • Publish with us
  • Track your research

Regenerative medicine and tissue engineering: Six companies leading the way

cropped Bio.Picture.Jules scaled 1

The field of tissue engineering blends principles from medtech and biotech. In the modern era, tissue engineering has seen rapid development, particularly with the advent of microfabrication and three-dimensional bioprinting technologies , with many companies being created in the field.

These advancements have enabled the creation of tissue constructs with greater precision and customization, improving outcomes in terms of biocompatibility, immune response reduction, and the integration and longevity of the engineered tissues. Big names in the biotech industry such as Abbvie have stakes in the market, and in this article, we take a closer look at six tissue engineering companies.

Table of contents

Founded in 1997 under the name TissueTech, the Miami-based company rebranded as BioTissue in November 2022. This biotech is focused on the field of regenerative medicine and is particularly known for its use of human birth tissue in creating products for ocular surface disease, surgical, chronic wound, and musculoskeletal applications. The company utilizes its proprietary CryoTek cryopreservation technology to maintain the structural and functional integrity of tissues in its products like Prokera, AmnioGraft, AmnioGuard for ocular application, Neox, and Clarix for surgical applications.

The company’s technology relies on human birth tissues, specifically the heavy chain hyaluronic acid/pentraxin3 (HC-HA/PTX3) complex that plays a role in the healing process in the fetal environment. Their tissue processing technology, CryoTek allows the preservation of the structural integrity of the placental tissues and HC-HA/PTX3 in the graft. Through its “Sharing Miracles” program, BioTissue partners with organizations like Dakota Lions Sight & Health to promote birth tissue donation, promoting a circular model.

BioTissue has conducted studies demonstrating the effectiveness of its CryoTek-preserved amniotic membrane in surgeries for pterygium , cataracts , and conjunctivochalasis, leading to improved patient outcomes. It has also shown that its amniotic allografts are effective in treating complex foot ulcers and traumatic wounds. In another study , the use of Prokera has led to improvement in moderate to severe dry eye disease.

Cellular Logistics

Cellular Logistics, based in Madison, Wisconsin, has developed CFX (Cardiac Fibroblast matriX), a therapeutic biomaterial produced using induced pluripotent stem cell technology. CFX is designed for local administration to prevent the pathological changes that lead to heart failure following myocardial infarction (heart attack). Moreover, CFX can also be used in conjunction with therapeutic stem cells to help remuscularize and restore function in scarred myocardium, particularly in cases of chronic heart failure.

The CFX technology presents several advantages. It is a cell-free product that is immune-privileged, allowing it to be administered to patients without the need for immunosuppressive drugs. It can modulate immune cells to promote healing properties and allows for the production of injectable particulates or patches, with the ability to adjust sizes according to clinical needs. These formulations enhance the retention of injected stem cells, preventing their rapid dispersal in the heart.

Earlier this year, Cellular Logistics and Allele Biotechnology entered into a partnership to develop regenerative medicine products for treating ischemic heart disease. The partnership leverages Allele’s cell reprogramming technology and aims to advance Solus to clinical-grade production for treating heart disease.

Japan Tissue Engineering

Japan Tissue Engineering (J-TEC), a company based in Gamagori, Aichi, Japan, is a pioneer in the field of regenerative medicine. Established in 1999, J-TEC has focused on developing, manufacturing, and selling tissue-engineered medical products. 

J-TEC’s business is divided into three main segments: Regenerative medicine, custom development and manufacturing, and R&D support. Its flagship products include autologous cultured epidermis JACE, and autologous cultured cartilage JACK.

J-TEC has developed autologous cultured epidermis products for treating severe burns, congenital giant pigmented nevus and epidermolysis bullosa. It is also working on treatments for vitiligo and second-degree burns using cultured epidermis.

Its autologous cultured cartilage product, approved for traumatic cartilage defects or osteochondritis dissecans, is one of Japan’s first regenerative medical products. Ongoing clinical trials aim to expand its use to secondary knee osteoarthritis .

In the field of ophthalmology, J-TEC has introduced autologous cultured corneal and oral mucosal epithelium products for treating corneal epithelial stem cell deficiency.

Moreover, J-TEC’s involvement in the broader landscape of regenerative medicine in Japan is significant, where the regulatory environment, such as the Regenerative Medicine Promotion Act of 2014, has catalyzed the development and approval of cell therapies. Japan’s strategic focus on regenerative medicine, supported by institutions like the Agency for Medical Research and Development (AMED), positions J-TEC in a thriving ecosystem.

Lattice Medical

Lattice Medical, established in 2017, is a French company specializing in the development of innovative implants for reconstructive surgery using tissue engineering, biomaterials, and 3D printing technologies. The company’s flagship products include Mattisse, a 3D-printed resorbable breast implant, and rodin, designed for hypodermal reconstruction. 

Mattisse stands out for being a 3D-printed, bioresorbable tissue engineering chamber designed for women who have undergone mastectomy due to breast cancer. This implant is made from a medical-grade resorbable biomaterial that naturally degrades over a few months post-implantation and is currently tested in the TIDE clinical trial in France. The design of Mattisse includes a porous base for fixing the autologous tissue to be regenerated and a dome that helps achieve the desired volume and shape.

Tissue Regenix

Tissue Regenix is a medical technology company headquartered in Leeds, England, specializing in regenerative medicine. The tissue engineering company is focused on the development and commercialization of regenerative products utilizing its proprietary technologies, dCELL and BioRinse.

The dCELL technology is a decellularization process that removes DNA and cellular material from animal and human tissue, leaving an acellular scaffold that can be used for the repair of damaged body parts without being rejected by the patient’s body. This technology has applications in sports injuries, foot and ankle surgery, and wound care. 

BioRinse is a technology used to sterilize bone allografts, minimizing tissue damage and preserving the quality of the grafts used primarily in sports medicine for tendon and ligament reconstruction.

Tissue Regenix has shown notable business growth, with a reported turnover of $29.5 million in 2023, indicating a substantial increase from the previous year. Additionally, Tissue Regenix has successfully expanded its market reach with new distributor agreements in Turkey and Ireland.

Vericel Corporation

Vericel Corporation, based in Cambridge, Massachusetts, specializes in advanced therapies for the sports medicine and severe burn care markets. Founded in 1989 and initially known as Aastrom Biosciences, Vericel underwent significant transformation after acquiring Sanofi’s cell therapy and regenerative medicine business in 2014, which led to its name change.

Their key products include Maci, Epicel, and NexoBrid, each utilizing different biotechnological approaches.

Maci (autologous cultured chondrocytes on porcine collagen membrane) is a product used for knee cartilage repair. It involves cultivating a patient’s own cells, which are then expanded and placed onto a collagen membrane. This membrane is then implanted into the damaged cartilage area, where it integrates with the surrounding tissue to repair the defect. MACI is specifically tailored to the size and shape of the cartilage damage, ensuring complete coverage and optimal healing​​.

Epicel (cultured epidermal autografts) is a skin graft technology used for treating patients with severe burns. It involves growing skin grafts from a small sample of the patient’s own skin, which can then be expanded to cover extensive burn areas​.

NexoBrid is a topical treatment used for the removal of eschar in patients with deep partial-thickness or full-thickness thermal burns. It contains proteolytic enzymes derived from pineapples that selectively remove nonviable burn tissue without harming surrounding healthy tissue. This process is crucial for the subsequent healing and treatment of severe burn injuries​.

The tissue engineering company is currently exploring other applications for its products as FDA approval for arthroscopic delivery of MACI is expected later this year and is on track to initiate a clinical trial for the treatment of ankle cartilage defects next year. Vericel also expects a FDA decision on NexoBrid pediatric usage later this year. 

Tissue engineering: an evolving industry

The current state of the tissue engineering industry is marked by a growing enthusiasm, driven by increasing clinical studies in regenerative medicine and tissue engineering. The market faces challenges such as high development costs and ethical issues surrounding stem cell research . Orthopedics, musculoskeletal, and spine applications dominate the market due to the rising prevalence of related disorders. The industry is also experiencing significant advancements in cardiology and vascular segments, fueled by the development of stem cell therapies and regenerative treatments for heart tissue .

Regionally, North America leads the market, supported by strong investment in research and development. The Asia Pacific region is also emerging as a significant player, with countries like Japan at the forefront of technological advances in the field.

Partnering 2030: The Biotech Perspective 2023

inpart logo

U.S. flag

An official website of the United States government

The .gov means it’s official. Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

The site is secure. The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

  • Publications
  • Account settings

Preview improvements coming to the PMC website in October 2024. Learn More or Try it out now .

  • Advanced Search
  • Journal List
  • Skin Appendage Disord
  • v.4(4); 2018 Oct

Logo of sad

Trichologist, Dermatotrichologist, or Trichiatrist? A Global Perspective on a Strictly Medical Discipline

Ralph michel trüeb.

a Center for Dermatology and Hair Diseases Professor Trüeb and University of Zurich, Zurich, Switzerland

Sergio Vañó-Galván

b Ramon y Cajal Hospital, University of Alcala, Madrid, Spain

Daisy Kopera

c Center of Aesthetic Medicine, Department of Dermatology, Medical University Graz, Graz, Austria

Vicky M.L. Jolliffe

d The Royal London Hospital, London, United Kingdom

Demetrios Ioannides

e 1st Department of Dermatology-Venereology, Hospital of Skin and Venereal Diseases, Aristotle University Medical School, Thessaloniki, Greece

Maria Fernanda Reis Gavazzoni Dias

f Department of Dermatology, Centro de Ciências Médicas, Hospital Universitário Antonia Pedro, Universidade Federal Fluminense, Niterói, Brazil

Melanie Macpherson

g Department of Dermatology and Venereology, San Gabriel Clinic, Lima, Peru

Javier Ruíz Ávila

h Dermedica Clinic, Polanco, Mexico City, Mexico

Aida Gadzhigoroeva

i Moscow Scientific and Practical Center of Dermatology and Cosmetology, Moscow City Health Department, Moscow, Russian Federation

Julya Ovcharenko

j General and Clinical Immunology and Allergology Department, School of Medicine, V.N. Karazin Kharkiv National University, Kharkiv, Ukraine

Won-Soo Lee

k Department of Dermatology, Wonju College of Medicine, Yonsei University, Gangwon-do, Republic of Korea

Sundaram Murugusundram

l Chennai Skin Foundation and Yesudian Research Institute, Chennai, India

Sotaro Kurata

m Beppu Garden Hill Clinic and Kurata Clinic, Beppu, Japan

n Prince of Wales Hospital and the Chinese University of Hong Kong, Hong Kong, China

Chuchai Tanglertsampan

o Department of Dermatology, Bumrungrad International Hospital, Mae Fah Luang University Hospital, Bangkok, Thailand

Few dermatologic problems carry as much emotional overtones as the complaint of hair loss. The best way to alleviate the distress related to hair loss is to effectively treat it. In fact, one of the oldest medical professions is the Egyptian physician who specialized on diseases of the head. And yet, from ancient Egypt down to modern times, human hair has been the object of superstition and mystery. Remarkably and despite the genuine advances in effective medical treatments, hair cosmetics, and surgical procedures, phony hair loss solutions continue to be marketed with an amazing success. In 1860, a quasi-scientific interest in hair loss and hair care originated in a London barbershop and became known as trichology, with the Institute of Trichologists being founded. Other corporations successively followed internationally, but it was only in 2010 that the term dermatotrichologist was proposed for board-certified dermatologists dealing with the scientific study of the hair and scalp, in contrast to the trichologist who is rather associated with laity and cosmetics than with medical professionalism, or – worse – offers opportunities to possible imposters with a primary commercial interest. The new term “trichiatrist” is proposed, literally meaning the “medical treatment of the hair,” to designate the strictly medical professional dealing with the hair and scalp in health and disease. Trichiatrists differ from trichologists by virtue of being physicians. The quality and stringency of their graduate medical training is identical to that of other physicians.

The medical practice is divided among them (the Egyptians) as follows: each physician is for one kind of sickness, and no more, and all places are crowded with physicians: for there are physicians for the eyes, physicians for the head , physicians for the teeth, physicians for the stomach, and for internal disease.

Herodotus, Histories 2,84

Few dermatologic problems carry as much emotional overtones as the complaint of hair loss, and the best way of alleviating the distress related to hair loss is to treat it effectively. In fact, one of the oldest medical professions documented in Greek historian and traveler Herodotus' “Histories” is the Egyptian physician who specialized on diseases of the head. Herodotus (484–425 BC) was the first historian known to have broken from the Homeric tradition to treat historical subjects as a method of investigation by collecting his materials systematically and critically and arranging them into a historiographic narrative. Thereby, Herodotus provides us with much information about the nature of the world and the status of science during his lifetime [ 1 ].

The most remote representations of hair loss that can be traced back around 30′000 years to wall engravings in a prehistorical cave are somewhat reminiscent of today's categorization of pattern hair loss, and the story is still ongoing in our modern society [ 2 ]. The first written document on the importance of making a distinction between inflammatory alopecia (tzaraat, Hebrew תערצ, breaking out on the head) from common baldness dates back to the Old Testament. The term tzaraat describes disfigurative conditions of the skin, hair of the beard and head. All variations are mainly referred to in chapters 13–14 of Leviticus (written ca. 538–332 BC) [ 3 ].

And yet, from the 4′000-year-old medical papyri of the ancient Egyptians down to modern times, human hair growth and color have been the object of superstition and mystery, besides arousing cosmetic and medical interest [ 4 ]. For the prevention or treatment of hair loss, countless herbal solutions, oils, lotions, magic pills, and even spiritual invocations have been advocated. What is remarkable about the history of hair loss cures is that despite the more recent genuine advances in effective medical treatments, hair cosmetics, and surgical procedures, phony hair loss solutions continue to be marketed today with an amazing success. Despite their outrageous claims, most lack scientifically measurable efficacy in preventing hair loss or promoting hair growth. As much today as in biblical times, people are so desperately concerned about their hair loss that they want to believe some miracle cure or some charismatic healer will help them.

The first genuine scientific studies on hair probably began when the English natural philosopher and polymath Robert Hooke (1635–1703) studied the hair shaft under the microscope [ 5 ] and the Italian biologist and physician Marcello Malphighi (1628–1694) described the anatomy of the hair follicle in his treatise “De pilis observationes” [ 6 ]. However, the biology of hair growth was not understood at this time. In search of information before engaging in the development of a new hair growth-promoting agent, makers of cosmetics turned to the medical faculty and received only very vague indications. When questioned, doctors remained evasive, and the hair was thus abated to malpractices of all sorts. The charlatan chemists of this age were as ineffectual but significantly more risky than their physician colleagues. With certain lotions in which toxic ingredients playing a role, accidents must have occurred frequently, and the French chemist Antoine Laurent de Lavoisier (1743–1794) urged for regulatory control of ingredients, which was never realized. France held the monopoly for miraculous elixirs and exported to America its “Eau de Ninon de L'enclos,” named after a beautiful courtesan whom had preserved her hair to the age of 85 years [ 7 ]. The French physician Auguste Caron, who published an “Encyclopedia of Beauty” [ 8 ] in 1806, warned the women of fashion of his time against the potential toxicity of products of mysterious origin, as a wretched woman had been driven to madness after using a tonic called “Eau de Chine.” The products remained just as dubious, whilst acquiring sentimental names as befitting the Romantic period. With the advance of medical technologies, ultraviolet light-emitting lamps, electrical scalp stimulators, and vacuum-cap machines have joined the repertory of treatments alleged to help stimulating the follicles to grow hair.

Probably the first sound scientific attempt toward hair restoration was the demonstration that it was possible to transplant hair from a hair-bearing to a non-hair bearing area in 1822 by the German medical student Johann Dieffenbach (1792–1847) [ 9 ]. However, it was not until 1939 that the era of hair transplantation began in earnest with the efforts of the Japanese physician Shoji Okuda (1886–1962) and later the New York dermatologist Norman Orentreich. Today, we hardly know anything about the medical background of Shoji Okuda, and his seminal work on hair transplantation remained virtually unknown outside Japan because of the outbreak of World War II. Moreover, the papers were written using old kanji (Japanese pictographs) and are consequently unintelligible even to modern Japanese medical readers. It was only in 2004 that the “Okuda Papers” were translated into English by Yoshihiro Imagawa, a retired gynecologist trained in the USA [ 10 ]. Okuda developed a circular scalpel for the purpose of transplanting hair into areas of alopecia [ 11 ]. In 1959, Norman Orentreich established the pivotal theory of donor dominance of the hair transplant. The basis of the theory was that plugs of hair follicles taken from the occipital scalp would continue growing when moved to the balding frontal scalp because those hair follicles were genetically programmed to do so. The concept became the foundation for the entire ensuing field of hair restoration surgery, and for the following decades, dedicated surgeons all over the globe have worked on refining the method with today close to natural results [ 12 ].

In 1860, a quasi-scientific interest in hair loss and hair care originated in a London barbershop under a self-styled Professor Wheeler. By1902, this interest in hair disorders became known as trichology, and the first Institute of Trichologists was founded. The International Association of Trichologists (IAT) was established in California in 1974 and offers a course by home-study for the training of students internationally who desire more knowledge about hair. Registered members can use the letters IAT after their name. Other corporations that have evolved globally are the Australian Institute of Trichology, the US Trichology Institute, the Argentine Association of Trichology (AATRI), and the World Trichology Society. Trichologists themselves are not normally medically qualified, although members of the medical profession can undertake courses and/or careers within trichology. Trichologists are not medically qualified but are taught the practice of care and treatment of the human hair and scalp in health and disease within their restricted but specialized role [ 13 ]. Nevertheless, there has been criticism regarding the ability of the public to reliably differentiate nonmedical trichologists from unqualified charlatans who monopolize publicity and proliferate in the high street, and concerns have been voiced about how to educate the public in choosing appropriate practitioners [ 14 , 15 ].

The dawn of the modern age of pharmacological therapy of hair loss can be traced back to the original clinical studies performed with topical minoxidil in the 1980s [ 16 ] and with oral finasteride in the 1990s [ 17 ]. For the first time in 4′000 years of history, pharmacological agents have been scientifically proven to stop hair loss and to promote hair growth. It is the introduction of these drugs into the treatment of hair loss that has heralded the emancipation of the treatment of hair loss from its age-old tradition of quackery. With respect to the study design and criteria for efficacy and safety, the respective clinical studies have set the standards for any agent with the claim of promoting hair growth.

Although testing medical interventions for efficacy had existed since the time of Avicenna's (980–1037) “The Canon of Medicine” in the 11th century [ 18 ], it was only in the 20th century that this effort evolved to impact almost all fields of health care and policy. In 1967, the American physician and mathematician Alvan R. Feinstein (1925–2001) published his seminal work “Clinical Judgment” [ 19 ], which together with Archie Cochrane's (1909–1988) celebrated book “Effectiveness and Efficiency” [ 20 ] led to an increasing acceptance of clinical epidemiology and controlled studies during the 1970s and 1980s and prepared the way for the institutional development of evidence-based medicine (EBM) in the 1990s. Ultimately, EBM aims for the ideal that healthcare professionals should make conscientious, explicit, and judicious use of the best available evidence gained from the scientific method to clinical decision making. It seeks to assess the strength of the evidence of risks and benefits of diagnostic tests and treatments, using techniques from science, engineering, and statistics, such as the systematic review of medical literature, meta-analysis, risk-benefit analysis, and randomized controlled trials.

Nonetheless, the limited success rate of evidence based therapies points to a more important complexity of hair loss and its management. Ultimately, EBM guidelines do not remove the problem of extrapolation to different populations. Even if several top-quality studies are available, questions remain as to how far and to which populations their results may be generalized. Certain groups have been historically under-researched, such as special age groups, ethnic minorities, and people with comorbid conditions. EBM applies to groups of people, but this should not preclude clinicians from using their personal experience in deciding how to treat the individual patient at hand.

For centuries, physicians propagated the viability of a complex approach in the diagnosis and treatment of disease, while modern medicine, which boasts a wide range of diagnostic methods and variety of therapeutic procedures, stresses specification. This raises the question: How does one wholly evaluate the state of a patient who suffers from a number of diseases simultaneously, where to start from and which disease(s) require(s) primary and subsequent treatment? This crucial question remained unanswered until 1970 when Alvan R. Feinstein coined the term “comorbidity” [ 21 ], which has been defined as “presence of one or more additional diseases co-occurring with a primary disease; or the effect of such additional diseases, whereby the additional disorder may also be a behavioural or mental disorder.” The effect of comorbid pathologies on clinical implications, diagnosis, prognosis, and therapy of trichologic conditions is polyhedral and patient specific. Therefore, presence of comorbidity must be taken into account when selecting the algorithm of the diagnosis and treatment plan for any given condition, including trichologic disease [ 22 ].

Ultimately, the dermatologist participates with the other medical disciplines in the diagnosis and treatment of all types of hair problems as they may relate to systemic disease [ 23 ].

On the other hand, hair loss is an important cause of discomfort and disability. The general physician is not always aware of the significance of hair loss and therefore may fail to refer patients with hair disorders to the dermatologist for appropriate management. Too often a delay in correct diagnosis and the resultant delay in ini­tiation of appropriate therapy results in potentially irreversible loss of hair, prolonged discomfort, and possible disfigurement.

As with any medical problem, the patient complaining of hair loss requires a comprehensive medical and drug history, physical examination of the hair and scalp, and appropriate laboratory evaluation to identify the cause. Dermatologic conditions are satisfying to diagnose as most conditions are visibly present at the time of consultation. Just looking would seem to be the simplest of diagnostic skills, and yet its very simplicity can result in its being overlooked. To reach the level of true artistry, looking must be a skillful active undertaking. The skill comes in interpreting the visual signs and having made a diagnosis hunting for the cause. The diagnostic process may be one of instantaneous recognition. The informed look is the one most practiced by the knowledgeable dermatologist, and is a combination of understanding, experience, and visual memory. If a visual diagnosis is not possible, then diagnostic tests are needed in the forms of specific dermatological examination techniques such as dermoscopy and laboratory evaluation (trichogram, biochemical investigations, microbiological studies, or scalp biopsy) as needed.

A prerequisite for delivering appropriate patient care is an understanding of the underlying pathologic dynamics of hair loss and its potentially multitudinous causes. By approaching the hair loss patient in a methodical way, commencing with objects the simplest and easiest to recognize, and ascending step by step to understanding the more complex aspects, an individualized treatment plan can be designed. Once the diagnosis is established, appropriate treatment is likely to be successful.

Alongside progress in clinical diagnosis and care, advances in the understanding of hair growth biology and its pathologic conditions is being made at a high pace thanks to the impetus of a generation of both astute clinicians and basic scientists interested in the hair follicle, the sophistication of molecular biology, and new technologies. Across the globe, Hair Research Societies have evolved such as the Australasian Hair and Wool Research Society; the European Hair Research Society; the North American Hair Research Society; the Society of Hair Research, Japan; the Korean Hair Research Society; the Hair Research Society of India; the Association of Professional Society of Trichologists, Moscow, Russia; and the Ukrainian Hair Research Society. These communities of interest and of practice regularly meet to bring together enthusiastic hair biologists and dermatologists for the exchange and discussion of the advances in hair research and clinical practice in the fields of genetics, molecular biology, immunology, aging, neurobiology, psychosomatics, diagnostic techniques and technologies, pharmacology, hair transplantation surgery, stem cells, and tissue-engineering research [ 24 ].

It was with the backdrop that in 2010 Dr. Patrick Yesudian, dermatologist practicing in India and founder of the Hair Research Society of India, proposed the term “dermatotrichologist” for board-certified dermatologists dealing with the scientific study of the hair and scalp in health and disease to distinguish them from the trichologist, who is not medically qualified and more involved with the cosmetic aspects of hair, or – worse – could offer opportunities to imposters with a primary commercial interest [ 25 ]. At the 2012 meeting of the Hair Research Society of India in UNESCO world heritage site Mamallapuram, the theme of the meeting was “To Abolish Quackery in Trichology,” and the consensus was that good medical practice in clinical trichology aims at (from [ 26 ]):

  • Understanding the hair patient on an emotional level and the medical problem on a technical level
  • Delivering sound patient education and effective trichologic therapy
  • Representing trichology as a discipline based on evidence from science, engineering, and statistics
  • Setting standards of good medical practice in trichology
  • Supporting progress in trichology through continuous medical education
  • Abolishing quackery in trichology

The ultimate question that arises, however, is whether the term “trichiatrist” for board-certified health care professionals (MDs) dealing with hair may be the yet more appropriate designation than trichologist or dermatotrichologist, in analogy to the term psychiatrist versus psychologist.

The term psychiatrist was originally coined by the German physician Johann Christian Reil in 1808 [ 27 ] and literally means the “medical treatment of the soul” (psych – “soul,” and –iatry – “medical treatment” from ancient Greek). Psychiatrists differ from psychologists in that they are physicians and have postgraduate training called residency in psychiatry (usually 4–5 years). The quality and stringency of their graduate medical training is identical to that of all other medical disciplines. Psychiatrists can therefore counsel patients, prescribe medication, conduct physical examinations, and order laboratory tests.

Parallels may easily be drawn with the care of hair disorders, when the same concept applies to the trichiatrist versus the trichologist, literally meaning the “medical treatment of the hair” (trich – “hair” from ancient Greek) to designate the strictly medical professional dealing with the hair and scalp in health and disease, with the capacity to counsel patients, prescribe medication, conduct physical examinations, and order pertinent laboratory tests as needed.

Furthermore, psychiatrists, more than other physicians, specialize in the doctor–patient relationship and are trained in therapeutic communication techniques [ 28 , 29 ].

In very much the same manner, prerequisites for a successful management of hair loss are twofold: on the technical and on the psychological level. On the technical level, prerequisites are a specific diagnosis, a profound understanding of the underlying pathophysiology, and the best available evidence gained from the scientific method for clinical decision making. On the psychological level, one must be sure that the patient's key concerns have been directly and specifically solicited and addressed: acknowledge the patient's perspective on the hair loss problem, explore patient's expectations from treatment, and educate patients into the basics of the hair cycle, and why patience is required for effective cosmetic recovery. Physicians should recognize that alopecia goes well beyond the simple physical aspects of hair loss and acknowledge the psychological impact of hair loss.

Ultimately, successful communication is the main reason for patient satisfaction and treatment success, while failed communication is the main reason for patient dissatisfaction, irrespective of treatment success [ 30 ].

In summary, as a trichiatrist, communication skills and treatment success require a genuine interest in recognizing and treating hair loss with knowledgeability on the scientific level and a genuine interest in supporting the patient complaining of hair loss with compassion on the emotional and psychological level. Ideally, credentials should include:

  • Medical degree and residency in a medical discipline relevant to the management of the hair and scalp in health and disease (usually dermatology)
  • Certification of traineeship or fellowship in the scientific study of the hair and scalp in health and disease with a syllabus expressing accountability and commitment
  • Membership in one of the Hair Research Societies or Societies of Hair Restoration Surgery with regular participation at respective scientific meetings
  • Accredited CME in the respective professional activity

Let's welcome the trichiatrist to the list of tricky “trichs” in dermatology [ 31 ]!

Statement of Ethics

The authors have no ethical conflicts to disclose.

Disclosure Statement

The authors have no conflicts of interest to disclose.

  • Search UNH.edu
  • Search Inquiry Journal

Commonly Searched Items:

  • Academic Calendar
  • Feature Article
  • Research Articles
  • Commentaries
  • Research Briefs
  • Mentor Highlights
  • Editorial Staff

Hydrogels and Injectable Therapeutics: How “Jell-O” May Become a Tissue Engineering Marvel

Yes, I know what you are thinking: Jell-O? Why would Jell-O be used in a materials lab for growing cells? Well, it isn’t the taste of this childhood snack that makes it perfect for tissue engineering; it is its composition. The most abundant protein in our body is a compound called collagen, which makes up most of the extracellular matrix that cells live in. If you take collagen and break it up into smaller pieces, you get gelatin, the material that makes up Jell-O. Tissue engineers, who work to maintain, improve, and/or restore biological tissues in our bodies, can use gelatin to simulate an in vivo environment, meaning the natural environment of a living organism. This not only opens applications in tissue culture but also has innumerable applications for injectable therapeutics, which are injectable solutions that can regrow or enhance existing tissue. While the field of tissue engineering is in its infancy, promising therapeutics with tissue culture using scaffolds like gelatin are already being applied to synthetic skin and organs for injured patients.

Injuries and degenerative diseases, such as Alzheimer’s and multiple sclerosis, can lead to long recoveries and/or irreversible damage. To alleviate these effects, one could inject specific stem cells into the damaged area t o grow themselves, trigger new growth within your body, and limit inflammation. However, direct injection of cells into the body results in low viability and significant dispersion of the cells throughout the body, moving them away from where they are needed most. Here is where our “scientific Jell-O” comes in. Gelatin, which is a hydrated complex of polymers, can be used to surround encapsulated cells, thereby increasing viability and stabilizing cell location following their injection. My research in Dr. Kyung Jae Jeong’s lab at the University of New Hampshire, which was funded by the Research Experience and Apprenticeship Program (REAP) through the Hamel Center for Undergraduate Research, looked at ways to alter hydrogels made of gelatin to make them more efficient when used in injectable therapeutics.

Bubble-looking circles of various sizes overlap on a gray and white background.

Figure 1. A microscope image of Jeong lab's novel 10% gelatin microgels. 

Microporous Hydrogel Methodology and Characterization

Conventional hydrogels are a complex of polymer chains that hold large amounts of water in comparison to their mass. However, these hydrogels lack porosity, mitigating cell growth and penetration of host biology. To address this, my faculty mentor, Dr. Jeong, and my graduate mentor, Dr. Seth Edwards, use novel injectable microporous hydrogels made of gelatin to promote cell spreading and proliferation. It may be easier to think of conventional hydrogels as one solid block of Jell-O with cells stuck inside, while a microgel structure is composed of many small beads of Jell-O with cells growing in the space bet ween. My summer research focused on the alteration of microgel diameter. Controlling microgel diameter can influence hydrogel properties such as nutrient transfer and available surface area, and cellular responses such as cell morphology, spreading, and differentiation. These variables help characterize cell behavior for predictability and safety of therapeutics in vivo.  

small bubble-looking circles are spread throughout the slide on a gray-blue background.

Figure 2. A microscope image of 5% gelatin microgels with a TWEEN 20 emulsifier developed by the author. 

I formed the microgels by dissolving gelatin in water and dropping the solution into an oil bath, creating an emulsion. The gelatin solution breaks up into small droplets in the oil that can be collected and cured together. The small spheres of gelatin are connected by microbial transglutaminase (mTG), a bacterial enzyme that connects the glutamine and lysine substituents of gelatin. After curing, a bulk hydrogel is left with space for cells to grow in between the microspheres. (Figure 1)

I tried lower concentrations of gelatin during the emulsion process, varying the mixing speeds and microgel curing times. Each variable had its own effect, some greater than others. I also integrated an emulsifier (TWEEN 20) of varying concentrations to reduce polarity on the hydrated gelatin’s surface. By integrating an emulsifier into the hydrogel emulsion protocol and decreasing gelatin concentration to 5% (from the original 10%), the diameter of the microgels were reduced to an eighth of their original size (around 40 µm). (Figure 2)

With this new manipulation of microgels I further researched cytotoxicity (toxicity of the environment to the cells). To do this, I cultured 3T3 cells (a fast-growing cell line of mouse fibroblasts) and formed microgels with small diameters and the control diameters. After three days of culture, I compared the control of pure gelatin microgels with the TWEEN 20 with a lowered gelatin concentration.

Right square is a black background with swirling green circles almost covering the slide. The left square is mostly black with green circles showing as the negative space.

Figure 3. A confocal microscopy stack image (left) and slice image (right) of living cells (green) and dead cells (red) growing within a microporous hydrogel structure.   

I measured viability by performing a live/dead assay with confocal imaging. This entails using a high-definition microscope and adding dye to stain living cells green and dead cells red. I detected no difference in cell viability between the two hydrogels. (Figure 3)

For further analysis I conducted a rheology test on the bulk hydrogel I had created with smaller microgels to measure and characterize its elasticity. This showed that the smaller hydrogels formed a stiffer bulk hydrogel than the one formed with larger control microgels at all points in the curing process. Rheology was performed only once, making the results less reliable. However, the stiffness is most likely caused by a larger surface area, which makes more interactions through mTG cross-links/bonds.

Results and Avenues for Application within Tissue Engineering

In the end I was able to decrease the diameter of the gelatin microgels, but I have yet to show their relation to cell growth. In theory, the differing concentrations of gelatin and emulsifier used in the microgels I created should induce different cell proliferation and differentiation. It has been shown through conventional nonporous hydrogels that stiffness does change cell differentiation trends. However, it is unclear if the cells are affected by the stiffness change in the porous environment. While the bulk hydrogel may have more mTG cross-linking, and thus be stiffer, there is still a large interior network that could limit the cells from experiencing overall stiffness. If that is the case, then a variation in hydrogel stiffness is important for different applications within the body. Depending on the biological environment this technology is applied to, less stiff hydrogel that degrades more quickly may be required, while others require the opposite. For example, neurons might like a less rigid structure with a smaller microgel for increased surface area, while bone cells may like a stiffer gel with larger microgels for better penetration of host biology.

Having control over all variables in the hydrogel leads to greater specificity and customization, depending on the intended use. As of now, conventional hydrogels are nonporous, and therefore restrict cell growth, penetration of host biology, nutrient transfer, and cell viability. The novel microporous hydrogels fix these problems but are not fully understood. Every cell location and cell type will need different conditions for efficient use in injectable therapies. These conditions can theoretically be accommodated with this technology, but these possibilities must be researched. If my research could be extrapolated, a full understanding of microgel cross-linking and diameter could lead to a new field of injectable therapeutics. Personally, I hope to continue my work in the Jeong lab by applying this technology to the culture of red blood cells. And to think, all of this possibility was sitting right in front of our faces in Jell-O’s composition.

I would like to acknowledge my faculty research mentor Dr. Kyung Jae Jeong for not only taking me on as a first-year researcher but accelerating my learning and cultivating an environment of opportunity. Further, I am grateful to Dr. Seth Edwards, my graduate mentor, for being alongside me every step of the way and for always being in my corner—truly the guiding voice in my process. None of this would be possible without the amazing staff and donors  (Mr. Dana Hamel, Dr. George Wildman, Mr. Nicholas Bencivenga)  at the Hamel Center for Undergraduate Research at UNH; without the Research, Experience, and Apprenticeship Program, I would not be writing today. I owe my future to the work and charity of these people, and I am forever grateful to the numerous efforts that have been made to benefit me.

Author and Mentor Bios

Jack Reynolds

Originally from Concord, New Hampshire, Jack Fenway Reynolds will graduate in May 2026 with a bachelor of science degree in bioengineering. He conducted his research on microgels through a Research Experience and Apprenticeship Program (REAP) grant funded by the Hamel Center for Undergraduate Research. Jack was looking for a project involving neurons and therapeutics, specifically in neurodegenerative diseases due to his mother's diagnosis with multiple sclerosis. He is very interested in the limited regenerative capacity of neurons and methods of circumnavigating this limitation. In talking with Dr. Jeong about possible topics relating to Jack’s interests, Dr. Joeng proposed the microgels his lab had been working on. Jack was pleasantly surprised at how accessible research was. As a new researcher, Jack describes having a steep learning curve, but he was mostly independent and was involved in scientific conversation with Ph.D. students and professors within a few weeks. He decided to submit to Inquiry because he is passionate about the public understanding of science. While using gelatin in this project, he wanted to convey the simplicity of this technology to excite people about the future, highlight the possibilities, and maybe inspire someone else. Jack would like to pursue a Ph.D. in a related scientific field and work in pharmaceutical/therapeutic development. While the research was very enlightening for the content and methodology, writing for Inquiry has shown him where science meets the public. Jack now wants to be on the cutting edge of science and educate the public about scientific developments.  

Dr. Kyung Jae Jeong is an associate professor of chemical engineering and bioengineering at the University of New Hampshire, beginning in 2013. His research interests revolve around biomaterials, drug delivery systems, medical devices, and tissue engineering. He mentored author, Jack Reynolds, for a Research Experience Apprenticeship Program (REAP) during the summer of 2023. Dr. Jeong has been interested in creating functional injectable hydrogels for medicine. However, the injectable hydrogels were nonporous and not optimal for cell delivery. His lab worked to develop the approach of assembling gelatin microgels using an enzymatic reaction in 2018 and applied the approach to neural stem cell encapsulation last year. Dr. Jeong has mentored Inquiry author, Ryan Boudreau previously. He describes Jack as “pleasant to work with” as well as being “highly motivated to learn new things, intellectually bright, and hard-working.”

Contact the author 

Copyright 2024, Jack Reynolds

Inquiry Journal

Spring 2024 issue.

University of New Hampshire Shield Logo

  • Sustainability
  • Embrace New Hampshire
  • University News
  • The Future of UNH
  • Campus Locations
  • Calendars & Events
  • Directories
  • Facts & Figures
  • Academic Advising
  • Colleges & Schools
  • Degrees & Programs
  • Undeclared Students
  • Course Search
  • Study Abroad
  • Career Services
  • How to Apply
  • Visit Campus
  • Undergraduate Admissions
  • Costs & Financial Aid
  • Net Price Calculator
  • Graduate Admissions
  • UNH Franklin Pierce School of Law
  • Housing & Residential Life
  • Clubs & Organizations
  • New Student Programs
  • Student Support
  • Fitness & Recreation
  • Student Union
  • Health & Wellness
  • Student Life Leadership
  • Sport Clubs
  • UNH Wildcats
  • Intramural Sports
  • Campus Recreation
  • Centers & Institutes
  • Undergraduate Research
  • Research Office
  • Graduate Research
  • FindScholars@UNH
  • Business Partnerships with UNH
  • Professional Development & Continuing Education
  • Research and Technology at UNH
  • Request Information
  • Current Students
  • Faculty & Staff
  • Alumni & Friends

Exosome-loaded decellularized tissue: Opening a new window for regenerative medicine

Affiliations.

  • 1 Cancer Research Center, Shahrekord University of Medical Sciences, Shahrekord, Iran.
  • 2 Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; Department of Tissue Engineering, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran.
  • 3 Department of Tissue Engineering, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran.
  • 4 Faculty of Medicine, Graduate School 'Molecular Medicine, University of Ulm, 89081, Ulm, Germany.
  • 5 Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; Department of Tissue Engineering, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran. Electronic address: [email protected].
  • PMID: 38594147
  • DOI: 10.1016/j.jtv.2024.04.005

Mesenchymal stem cell-derived exosomes (MSCs-EXO) have received a lot of interest recently as a potential therapeutic tool in regenerative medicine. Extracellular vesicles (EVs) known as exosomes (EXOs) are crucial for cell-cell communication throughout a variety of activities including stress response, aging, angiogenesis, and cell differentiation. Exploration of the potential use of EXOs as essential therapeutic effectors of MSCs to encourage tissue regeneration was motivated by success in the field of regenerative medicine. EXOs have been administered to target tissues using a variety of methods, including direct, intravenous, intraperitoneal injection, oral delivery, and hydrogel-based encapsulation, in various disease models. Despite the significant advances in EXO therapy, various methods are still being researched to optimize the therapeutic applications of these nanoparticles, and it is not completely clear which approach to EXO administration will have the greatest effects. Here, we will review emerging developments in the applications of EXOs loaded into decellularized tissues as therapeutic agents for use in regenerative medicine in various tissues.

Keywords: Decellularized tissue; Exosome; Regenerative medicine; Tissue engineering.

Copyright © 2024 Tissue Viability Society / Society of Tissue Viability. Published by Elsevier Ltd. All rights reserved.

Publication types

IMAGES

  1. Phd in tissue engineering and regenerative medicine

    phd tissue engineering and regenerative medicine

  2. Tissue Engineering and Regenerative Medicine (MSc)

    phd tissue engineering and regenerative medicine

  3. (PDF) Tissue Engineering and Regenerative Medicine

    phd tissue engineering and regenerative medicine

  4. Key concepts of the tissue engineering and regenerative medicine

    phd tissue engineering and regenerative medicine

  5. Regenerative Medicine, Cellular Therapy and Tissue Engineering

    phd tissue engineering and regenerative medicine

  6. The Future of Medicine: Tissue Engineering and Regenerative Medicine

    phd tissue engineering and regenerative medicine

VIDEO

  1. 8th Engineering in Medicine Symposium- Tissue Engineering

  2. Haute Culture: Tailoring Stem Cells to Make Us Well

  3. Vascular Tissue Engineering

  4. 👆 Full video

  5. Tissue Engineering and Regenerative Medicine

  6. MyDerm & Ops Luka

COMMENTS

  1. Tissue Engineering and Regenerative Medicine

    Research in tissue engineering and regenerative medicine seeks to replace or regenerate diseased or damaged tissues, organs, and cells - a challenging endeavor, but one that has tremendous potential for the practice of medicine. Technologies under investigation range from biomaterial/cell constructs for repairing various tissues and organs ...

  2. Stem Cell PhD Program

    Stanford is a world leader in stem cell research and regenerative medicine. Central discoveries in stem cell biology - tissue stem cells and their use for regenerative therapies, transdifferentiation into mature cell-types, isolation of cancerous stem cells, and stem cell signaling pathways - were made by Stanford faculty and students.

  3. Regenerative Sciences

    Seeking to spur development of innovative medical breakthroughs, Mayo Clinic Graduate School of Biomedical Sciences, in partnership with the Center for Regenerative Biotherapeutics, started one of the nation's first doctoral research training programs in regenerative sciences.. Regenerative medicine is transforming clinical practice with the development of new therapies, treatments and ...

  4. Tissue Engineering & Regenerative Medicine

    Tissue Engineering &. Regenerative Medicine. Research in tissue engineering and regenerative medicine encompasses all aspects of the research and development continuum from mechanistic studies to translational approaches. Collaborative efforts with colleagues at Rice and the Texas Medical Center address unmet clinical needs for a plethora of ...

  5. Tissue Engineering & Regenerative Medicine Research Program

    The Tissue Engineering and Regenerative Medicine Program supports basic and translational research on employing bioengineering- and stem cell biology- based approaches for the reconstruction, repair, and regeneration of dental, oral, and craniofacial (DOC) tissues damaged because of disease or injury.

  6. Tissue Engineering and Regenerative Medicine

    The goal of tissue engineering is to assemble functional constructs that restore, maintain, or improve damaged tissues or whole organs. Artificial skin and cartilage are examples of engineered tissues that have been approved by the FDA; however, currently they have limited use in human patients. Regenerative medicine is a broad field that ...

  7. Tissue Engineering

    Ying Zheng, PhD (Bioengineering) Dr. Zheng's research focuses on understanding and engineering the fundamental structure and functions in living tissue and organ systems from nanometer, micrometer to centimeter scale. These are the faculty members that are specialized in tissue engineering at the Institute for Stem Cell & Regenerative Medicine.

  8. Tissue Engineering & Regenerative Medicine

    The promise of regenerative medicine is truly remarkable. Over the last two decades, significant breakthroughs in understanding within the regenerative medicine and tissue engineering fields have yielded a more intimate understanding of the functioning of human tissue. In the future, new technologies may deliver islet cells for diabetes, neural ...

  9. Home

    Tissue Engineering and Regenerative Medicine is a platform for the advancement and dissemination of research and technologies related to its field. Indexed and included in PubMed/MEDLINE with all articles from 2019 searchable. Promotes sharing and dissemination of knowledge among members and provides insights into new research trends.

  10. Tissue engineering and regenerative medicine

    Tissue engineering is a branch of regenerative medicine, itself a branch of biomedical engineering. Tissue engineering and regenerative medicine are concerned with the replacement or regeneration ...

  11. Single-cell transcriptomics in tissue engineering and regenerative medicine

    Regenerative medicine and tissue engineering aim to promote functional rebuilding of damaged tissue. Comprehensively profiling cell identity, function and interaction in healthy tissues, as well ...

  12. Brush Up: Tissue Engineering and Regenerative Medicine

    The use of tissue engineering in regenerative medicine, known as TERM, is an active area of research that involves creating functional tissue through the combination of cells, scaffolds, and growth factors to restore normal biological function. 1 Clinicians treat millions of patients with tissue engineered regenerative devices.

  13. Regenerative Medicine PhD

    The Centre for Regenerative Medicine (CRM) is a world leading research centre based at the University of Edinburgh's Institute for Regeneration and Repair. Our scientists and clinicians study stem cells, disease and tissue repair to advance human health. By better understanding how stem cells are controlled and how diseases develop in a lab ...

  14. Tissue Engineering and Regenerative Medicine: Recent Innovations and

    Methods of Review. The first challenge in conducting this review was the sheer number of recent publications in the TERM field. The origins of the terms "tissue engineering" and "regenerative medicine" have been previously discussed in this journal, 1 with the former coming into common parlance in the mid-to-late 1980s and the latter gaining momentum around the turn of the 21st century.

  15. Tissue Engineering for Musculoskeletal Regeneration and Disease

    Regenerative medicine, as the term indicates, aims to enable and enhance the body's natural repair mechanisms to restore the function of otherwise irreparable tissues or organs in situ. The tissue engineering triad consists of three key elements: scaffolds, cells, and signaling molecules (O'Brien 2011). In the past few decades, considerable ...

  16. Van Cleve Cardiac Regenerative Medicine Program

    The goal of the Mayo Clinic Van Cleve Cardiac Regenerative Medicine Program is to advance stem cell therapies, cell-free regeneration and tissue engineering. To do this, the program accelerates the discovery, translation and application of innovative regenerative products for heart disease. Importantly, results from this work will not only make ...

  17. Regenerative Medicine and Tissue Engineering

    Few events in science have captured the same level of sustained interest and imagination of the nonscientific community as Stem Cells, Tissue Engineering, and Regenerative Medicine. The fundamental concept of Tissue Engineering and Regenerative Medicine is appealing to scientists, physicians, and lay people alike: to heal tissue or organ defects that the current medical practice deems ...

  18. regenerative medicine PhD Projects, Programmes & Scholarships

    Dr. Mahetab Amer is seeking motivated students with backgrounds in tissue engineering, cell biology, materials science, or related fields to join her dynamic, multidisciplinary research group at the Division of Cell Matrix Biology and Regenerative Medicine, University of Manchester.

  19. tissue engineering PhD Projects, Programmes & Scholarships

    Fully Funded PhD Scholarship in Biomaterials Synthesis and Cartilage Tissue Engineering. University of Galway School of Medicine. Application (s) are invited from suitably qualified candidates for full-time funded PhD scholarship starting in September, 2024 affiliated to the School of Medicine at the University of Galway. Read more.

  20. Marco C. Bottino, D.D.S., MSc., Ph.D.

    My lab focuses on identifying and translating regenerative materials and technologies to reestablish dental, oral, and craniofacial (DOC) tissue health. We use both in vitro and in vivo pre-clinical animal models to gain further insight into the potential clinical safety and efficacy of the developed biomaterials and overall regenerative ...

  21. The Main Mechanisms of Mesenchymal Stem Cell-Based ...

    Tissue Engineering and Regenerative Medicine - Coronavirus disease 2019 (COVID-19) has a clinical manifestation of hypoxic respiratory failure and acute respiratory distress syndrome. ... Ruoss M, Farzaneh Z, et al. Tissue engineering in liver regenerative medicine: insights into novel translational technologies. Cells. 2020;9:304. Article CAS ...

  22. MSc Tissue Engineering for Regenerative Medicine / Overview

    Our MSc Tissue Engineering for Regenerative Medicine looks at regenerating and ... commercialisation and clinical translation of regenerative therapies. Prepare for PhD study, specialist clinical training or a career in related industries, including pharmaceutical, biotechnology and regenerative medicine sectors. Study at a university ranked ...

  23. How a new drug prototype regenerates lung tissue

    "My approach toward regenerative medicine has been figuring out how to promote regenerative, proliferative repair of organs using druglike molecules that act on endogenous stem cell populations," says co-senior author, Michael J. Bollong, PhD, an associate professor and the Early Career Endowed Roon Chair for Cardiovascular Research in the ...

  24. Human Nasal Inferior Turbinate-Derived Neural Stem Cells ...

    Background: Parkinson's disease (PD) is one of the most prevalent neurodegenerative diseases, following Alzheimer's disease. The onset of PD is characterized by the loss of dopaminergic neurons in the substantia nigra. Stem cell therapy has great potential for the treatment of neurodegenerative diseases, and human nasal turbinate-derived stem cells (hNTSCs) have been found to share some ...

  25. Tissue engineering: Six key companies to put on your radar

    Japan Tissue Engineering (J-TEC), a company based in Gamagori, Aichi, Japan, is a pioneer in the field of regenerative medicine. Established in 1999, J-TEC has focused on developing, manufacturing, and selling tissue-engineered medical products. J-TEC's business is divided into three main segments: Regenerative medicine, custom development ...

  26. Trichologist, Dermatotrichologist, or Trichiatrist? A Global

    The quality and stringency of their graduate medical training is identical to that of other physicians. Keywords: Hair restoration, Evidence-based ... testing medical interventions for efficacy had existed since the time of Avicenna's (980-1037) "The Canon of Medicine" in the 11th ... stem cells, and tissue-engineering ...

  27. Hydrogels and Injectable Therapeutics: How "Jell-O" May Become a Tissue

    His research interests revolve around biomaterials, drug delivery systems, medical devices, and tissue engineering. He mentored author, Jack Reynolds, for a Research Experience Apprenticeship Program (REAP) during the summer of 2023. Dr. Jeong has been interested in creating functional injectable hydrogels for medicine.

  28. Exosome-loaded decellularized tissue: Opening a new window for ...

    Abstract. Mesenchymal stem cell-derived exosomes (MSCs-EXO) have received a lot of interest recently as a potential therapeutic tool in regenerative medicine. Extracellular vesicles (EVs) known as exosomes (EXOs) are crucial for cell-cell communication throughout a variety of activities including stress response, aging, angiogenesis, and cell ...

  29. Platelets: A Potential Factor that Offers Strategies for Promoting Bone

    Bone defects represent a prevalent category of clinical injuries, causing significant pain and escalating health care burdens. Effectively addressing bone defects is thus of paramount importance. Platelets, formed from megakaryocyte lysis, have emerged as pivotal players in bone tissue repair, inflammatory responses, and angiogenesis. Their intracellular storage of various growth factors ...

  30. Call for Special Issue Papers: Special Issue for Prof. James

    Authors: Guest Editors: Laura De Laporte, PhD, and Jeroen van den Beucken, PhD Authors Info & Affiliations Publication : Tissue Engineering Part A Volume 29 , Issue Number 23-24