Neurology and Neurosurgery

  • Study finds stem cell therapy is safe and may benefit people with spinal cord injuries

May 23, 2024

research article on stem cell therapy

Mayo Clinic researchers have demonstrated the safety and potential benefit of stem cell regenerative medicine therapy for patients with subacute and chronic spinal cord injury.

The results of the phase 1 Clinical Trial of Autologous Adipose-Derived Mesenchymal Stem Cells in the Treatment of Paralysis Due to Traumatic Spinal Cord Injury, known as CELLTOP, were published in Nature Communications.

Treatment process

Illustration shows the process of fat harvest via biopsy, adipose-derived mesenchymal stem cells (AD-MSC) preparation and administration of treatment.

All trial participants had experienced traumatic spinal injury classified as grade A or B on the American Spinal Injury Association Impairment Scale (AIS). Stem cell treatment was initiated on average 11 months after injury. Participants were evaluated over a two-year period.

Key findings:

  • Stem cells were successfully manufactured, and products were delivered to all 10 enrolled participants.
  • No serious adverse effects occurred among any participants. The most commonly reported side effects were headache and musculoskeletal pain, which resolved with over-the-counter treatment.
  • Seven participants demonstrated improvement, with each moving up at least one AIS grade.

As reported earlier in Mayo Clinic Proceedings, the first participant in the phase 1 trial was a superresponder who, after stem cell therapy, saw significant improvements in the function of his upper and lower extremities.

"Future research may show whether stem cells in combination with other therapies could be part of a new paradigm of treatment to improve outcomes for patients," says Mohamad Bydon, M.D. , a neurosurgeon at Mayo Clinic in Rochester, Minnesota, and the first author of both studies. "Not every patient who receives stem cell treatment is going to be a superresponder. One objective in our future studies is to delineate the optimal treatment protocols and understand why patients respond differently."

Dr. Bydon notes that stem cells' mechanism of action isn't fully understood. The researchers are analyzing changes in participants' MRI and cerebrospinal fluid to identify avenues for potential regeneration. Work is also underway on a larger, controlled trial of stem cell regenerative therapy.

"For years, treatment of spinal cord injury has been limited to stabilization surgery and physical therapy," Dr. Bydon says. "Many historical textbooks state that this condition does not improve. We have seen findings in recent years that challenge prior assumptions. This research is a step forward toward the ultimate goal of improving treatments for patients."

For more information

Bydon M, et al. Intrathecal delivery of adipose-derived mesenchymal stem cells in traumatic spinal cord injury: Phase I trial . Nature Communications. 2024;15:2201.

Bydon M, et al. CELLTOP clinical trial: First report from a phase I trial of autologous adipose tissue-derived mesenchymal stem cells in the treatment of paralysis due to traumatic spinal cord injury . Mayo Clinic Proceedings. 2020;95:406.

Refer a patient to Mayo Clinic.

Receive Mayo Clinic news in your inbox.

Related content.

research article on stem cell therapy

  • Medical Professionals

Help transform healthcare

Your donation can make a difference in the future of healthcare. Give now to support Mayo Clinic's research.

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
  • My Bibliography
  • Collections
  • Citation manager

Save citation to file

Email citation, add to collections.

  • Create a new collection
  • Add to an existing collection

Add to My Bibliography

Your saved search, create a file for external citation management software, your rss feed.

  • Search in PubMed
  • Search in NLM Catalog
  • Add to Search

Stem cells: past, present, and future

Affiliations.

  • 1 Department of Experimental Surgery and Biomaterials Research, Wroclaw Medical University, Bujwida 44, Wrocław, 50-345, Poland. [email protected].
  • 2 Department of Conservative Dentistry and Pedodontics, Krakowska 26, Wrocław, 50-425, Poland.
  • 3 Department of Experimental Surgery and Biomaterials Research, Wroclaw Medical University, Bujwida 44, Wrocław, 50-345, Poland.
  • PMID: 30808416
  • PMCID: PMC6390367
  • DOI: 10.1186/s13287-019-1165-5

In recent years, stem cell therapy has become a very promising and advanced scientific research topic. The development of treatment methods has evoked great expectations. This paper is a review focused on the discovery of different stem cells and the potential therapies based on these cells. The genesis of stem cells is followed by laboratory steps of controlled stem cell culturing and derivation. Quality control and teratoma formation assays are important procedures in assessing the properties of the stem cells tested. Derivation methods and the utilization of culturing media are crucial to set proper environmental conditions for controlled differentiation. Among many types of stem tissue applications, the use of graphene scaffolds and the potential of extracellular vesicle-based therapies require attention due to their versatility. The review is summarized by challenges that stem cell therapy must overcome to be accepted worldwide. A wide variety of possibilities makes this cutting edge therapy a turning point in modern medicine, providing hope for untreatable diseases.

Keywords: Differentiation; Growth media; Induced pluripotent stem cell (iPSC); Pluripotency; Stem cell derivation; Stem cells; Teratoma formation assay; Tissue banks; Tissue transplantation.

PubMed Disclaimer

Conflict of interest statement

Ethics approval and consent to participate.

Not applicable.

Consent for publication

Competing interests.

The authors declare that they have no competing interests.

Publisher’s Note

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

Oocyte development and formation of…

Oocyte development and formation of stem cells: the blastocoel, which is formed from…

Changes in the potency of…

Changes in the potency of stem cells in human body development. Potency ranges…

Spontaneous differentiation of hESCs causes…

Spontaneous differentiation of hESCs causes the formation of a heterogeneous cell population. There…

Culturing of pluripotent stem cells…

Culturing of pluripotent stem cells in vitro. Three days after fertilization, totipotent cells…

Retroviral-mediated transduction induces pluripotency in…

Retroviral-mediated transduction induces pluripotency in isolated patient somatic cells. Target cells lose their…

Stem cell experiments on animals.…

Stem cell experiments on animals. These experiments are one of the many procedures…

Localization of stem cells in…

Localization of stem cells in dental tissues. Dental pulp stem cells (DPSCs) and…

Use of inner cell mass…

Use of inner cell mass pluripotent stem cells and their stimulation to differentiate…

Similar articles

  • Derivation of Human Induced Pluripotent Stem Cell (iPSC) Lines and Mechanism of Pluripotency: Historical Perspective and Recent Advances. Chhabra A. Chhabra A. Stem Cell Rev Rep. 2017 Dec;13(6):757-773. doi: 10.1007/s12015-017-9766-9. Stem Cell Rev Rep. 2017. PMID: 28918520 Review.
  • Future prospects for regenerated heart using induced pluripotent stem cells. Fujita J, Fukuda K. Fujita J, et al. J Pharmacol Sci. 2014;125(1):1-5. doi: 10.1254/jphs.14r01cp. Epub 2014 Apr 16. J Pharmacol Sci. 2014. PMID: 24739283 Review.
  • Induced pluripotent stem cells for the treatment of stroke: the potential and the pitfalls. Yu F, Li Y, Morshead CM. Yu F, et al. Curr Stem Cell Res Ther. 2013 Sep;8(5):407-14. doi: 10.2174/1574888x113089990052. Curr Stem Cell Res Ther. 2013. PMID: 23895059 Review.
  • Integration of induced pluripotent stem cell-derived endothelial cells with polycaprolactone/gelatin-based electrospun scaffolds for enhanced therapeutic angiogenesis. Tan RP, Chan AHP, Lennartsson K, Miravet MM, Lee BSL, Rnjak-Kovacina J, Clayton ZE, Cooke JP, Ng MKC, Patel S, Wise SG. Tan RP, et al. Stem Cell Res Ther. 2018 Mar 21;9(1):70. doi: 10.1186/s13287-018-0824-2. Stem Cell Res Ther. 2018. PMID: 29562916 Free PMC article.
  • 3D Printed Graphene-PLA Scaffolds Promote Cell Alignment and Differentiation. Gasparotto M, Bellet P, Scapin G, Busetto R, Rampazzo C, Vitiello L, Shah DI, Filippini F. Gasparotto M, et al. Int J Mol Sci. 2022 Feb 3;23(3):1736. doi: 10.3390/ijms23031736. Int J Mol Sci. 2022. PMID: 35163657 Free PMC article.
  • Proneurogenic actions of follicle-stimulating hormone on neurospheres derived from ovarian cortical cells in vitro. González-Gil A, Sánchez-Maldonado B, Rojo C, Flor-García M, Queiroga FL, Ovalle S, Ramos-Ruiz R, Fuertes-Recuero M, Picazo RA. González-Gil A, et al. BMC Vet Res. 2024 Aug 20;20(1):372. doi: 10.1186/s12917-024-04203-8. BMC Vet Res. 2024. PMID: 39160565
  • Identification of Meibomian gland stem cell populations and mechanisms of aging. Zhu X, Xu M, Portal C, Lin Y, Ferdinand A, Peng T, Morrisey EE, Dlugosz AA, Castellano JM, Lee V, Seykora JT, Iomini C, Millar SE. Zhu X, et al. bioRxiv [Preprint]. 2024 Aug 10:2024.08.09.607015. doi: 10.1101/2024.08.09.607015. bioRxiv. 2024. PMID: 39149265 Free PMC article. Preprint.
  • Therapeutic potential of Chinese medicinal herbs stimulating osteogenic differentiation of bone marrow-derived mesenchymal stem cells in osteoporosis. Wang H, Shan K, Li Y, Wu S, Zhou C, Tao S, Wang M, Kang X, Zhou L, Lyu Z, Li N. Wang H, et al. Front Pharmacol. 2024 Jul 31;15:1423555. doi: 10.3389/fphar.2024.1423555. eCollection 2024. Front Pharmacol. 2024. PMID: 39144620 Free PMC article. Review.
  • Emerging Landscape of Mesenchymal Stem Cell Senescence Mechanisms and Implications on Therapeutic Strategies. Wang J, Zhang M, Wang H. Wang J, et al. ACS Pharmacol Transl Sci. 2024 Jul 17;7(8):2306-2325. doi: 10.1021/acsptsci.4c00284. eCollection 2024 Aug 9. ACS Pharmacol Transl Sci. 2024. PMID: 39144566 Review.
  • Examining the potentials of stem cell therapy in reducing the burden of selected non-communicable diseases in Africa. Atewologun FA, Okesanya OJ, Okon II, Kayode HH, Ukoaka BM, Olaleke NO, Ogaya JB, Okikiola LA, Manirambona E, Lucero-Prisno Iii DE. Atewologun FA, et al. Stem Cell Res Ther. 2024 Aug 13;15(1):253. doi: 10.1186/s13287-024-03864-4. Stem Cell Res Ther. 2024. PMID: 39135088 Free PMC article. Review.
  • Sukoyan MA, Vatolin SY, et al. Embryonic stem cells derived from morulae, inner cell mass, and blastocysts of mink: comparisons of their pluripotencies. Embryo Dev. 1993;36(2):148–58 - PubMed
  • Larijani B, Esfahani EN, Amini P, Nikbin B, Alimoghaddam K, Amiri S, Malekzadeh R, Yazdi NM, Ghodsi M, Dowlati Y, Sahraian MA, Ghavamzadeh A. Stem cell therapy in treatment of different diseases. Acta Medica Iranica. 2012:79–96 https://www.ncbi.nlm.nih.gov/pubmed/22359076 . - PubMed
  • Sullivan S, Stacey GN, Akazawa C, et al. Quality guidelines for clinical-grade human induced pluripotent stem cell lines. Regenerative Med. 2018; 10.2217/rme-2018-0095. - PubMed
  • Amps K, Andrews PW, et al. Screening ethnically diverse human embryonic stem cells identifies a chromosome 20 minimal amplicon conferring growth advantage. Nat. Biotechnol. 2011;29(12):1121–1144. - PMC - PubMed
  • Amit M, Itskovitz-Eldor J. Atlas of human pluripotent stem cells: derivation and culturing. New York: Humana Press; 2012.

Publication types

  • Search in MeSH

Related information

Linkout - more resources, full text sources.

  • BioMed Central
  • Europe PubMed Central
  • PubMed Central

Other Literature Sources

  • The Lens - Patent Citations
  • MedlinePlus Health Information

Miscellaneous

  • NCI CPTAC Assay Portal

full text provider logo

  • Citation Manager

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Home

Study at Cambridge

About the university, research at cambridge.

  • For Cambridge students
  • For our researchers
  • Business and enterprise
  • Colleges and Departments
  • Email and phone search
  • Give to Cambridge
  • Museums and collections
  • Events and open days
  • Fees and finance
  • Postgraduate courses
  • How to apply
  • Fees and funding
  • Postgraduate events
  • International students
  • Continuing education
  • Executive and professional education
  • Courses in education
  • How the University and Colleges work
  • Visiting the University
  • Annual reports
  • Equality and diversity
  • A global university
  • Public engagement

New way to extend ‘shelf life’ of blood stem cells will improve gene therapy

  • Research home
  • About research overview
  • Animal research overview
  • Overseeing animal research overview
  • The Animal Welfare and Ethical Review Body
  • Animal welfare and ethics
  • Report on the allegations and matters raised in the BUAV report
  • What types of animal do we use? overview
  • Guinea pigs
  • Equine species
  • Naked mole-rats
  • Non-human primates (marmosets)
  • Other birds
  • Non-technical summaries
  • Animal Welfare Policy
  • Alternatives to animal use
  • Further information
  • Funding Agency Committee Members
  • Research integrity
  • Horizons magazine
  • Strategic Initiatives & Networks
  • Nobel Prize
  • Interdisciplinary Research Centres
  • Open access
  • Energy sector partnerships
  • Podcasts overview
  • S2 ep1: What is the future?
  • S2 ep2: What did the future look like in the past?
  • S2 ep3: What is the future of wellbeing?
  • S2 ep4 What would a more just future look like?

Test tubes in a lab

Researchers have discovered a way to extend the shelf life of blood stem cells outside the body for use in gene therapy, providing patients with better options and improving their outcomes.

We were able to identify a key molecular pathway...that can be targeted by a drug which is already in use and is safe to use. Elisa Laurenti

Researchers have identified a drug already used for cancer patients, that, when applied to current gene therapy protocols can improve blood stem cell function threefold.

One trillion blood cells are produced every day in humans, and blood stem cells are the only cell types in our body capable of producing all blood cell types over our lifespan, giving them immense regenerative potential.

Blood stem cell gene therapy is a ground-breaking treatment that currently provides the only cure to more than ten life-debilitating genetic diseases and has already saved the lives of more than two million people with blood cancers and other diseases.

These therapies take blood stem cell samples from patients, where their genetic defect is corrected in a dish before being delivered back to the patient. However, limitations persist in blood stem cell therapies because of the shelf life of the cells outside the body. When removed from their environment in the human body and cultured in a dish, most blood stem cells lose their function. The exact timing and cause of this function loss has not previously been well understood.

Now, scientists in the Laurenti Group and others at the University of Cambridge’s Cambridge Stem Cell Institute (CSCI) and Department of Haematology have pinpointed a timeline for the blood stem cells under the current gene therapy protocols, which typically take place over three days. After the first 24 hours in a dish, more than 50% of the blood stem cells can no longer sustain life-long blood production, which is before therapy would even begin in a clinical setting.

During those first 24 hours, the cells activate a complex molecular stress response in order to adapt to the dish. By studying this stress response, the team identified a solution. Through repurposing a cancer growth blocker drug (Ruxolitinib), already in use for cancer treatments, they were able to improve stem cell function in a dish by three times its former capabilities.

The group is now aiming to modify current gene therapy protocols to include this drug, providing patients with the highest number of high-quality blood stem cells and improving their outcomes.

The study is published today in the journal Blood .

Professor Elisa Laurenti at the University of Cambridge Stem Cell Institute, and senior author of the study, said: “This is really exciting because we are now in a position where we can begin to understand the huge stress that these stem cells sense when they are manipulated outside of our body. Biologically it is really fascinating because it affects every aspect of their biology. Luckily, we were able to identify a key molecular pathway which governs many of these responses, and that can be targeted by a drug which is already in use and is safe to use.

“I hope our findings will enable safer treatments for gene therapy patients. Our discovery also opens up many possibilities to better expand blood stem cells ex vivo and expand the set of disease where we can use blood stem cells to improve patients’ lives.”

Dr Carys Johnson at the University of Cambridge Stem Cell Institute, and first author of the study, said: “Although we expected that removing these cells from the body and culturing them on a plastic surface would alter gene expression, the extent of change we found was surprising, with over 10,000 genes altered and a significant stress response detected. It was also striking to discover that the majority of blood stem cells are functionally lost during gene therapy protocols, before transplantation back to the patient.

“We have identified a key bottleneck where function is lost and clinical culture could be improved. I hope that our work will drive advancements in culture protocols to better harness the power of blood stem cells and improve the safety and efficacy of clinical approaches.”

C.S. Johnson, M.J. Williams, K. Sham, et al. ‘ Adaptation to ex vivo culture reduces human hematopoietic stem cell activity independently of cell cycle .’ Blood 2024; DOI: 10.1182/blood.2023021426

Story written by Laura Puhl, Cambridge Stem Cell Institute.

Creative Commons License.

Read this next

A colour-enhanced image showing a clump of prostate cancer cells

Combined forces

Cancer researcher Tom Else sitting in a lab wearing a white lab coat

Sight and sound

Dr Alex Cagan outside the Department of Genetics

How 'superstar species' solve healthier ageing and cancer resistance

research article on stem cell therapy

Black excellence in cancer research at Cambridge

Media enquiries.

Credit: Philippe Delavie from Pixabay

research article on stem cell therapy

Search research

Sign up to receive our weekly research email.

Our selection of the week's biggest Cambridge research news sent directly to your inbox. Enter your email address, confirm you're happy to receive our emails and then select 'Subscribe'.

I wish to receive a weekly Cambridge research news summary by email.

The University of Cambridge will use your email address to send you our weekly research news email. We are committed to protecting your personal information and being transparent about what information we hold. Please read our email privacy notice for details.

  • Elisa Laurenti
  • Carys Johnson
  • Cambridge Stem Cell Institute
  • Department of Haematology
  • School of Clinical Medicine

Connect with us

Cambridge University

© 2024 University of Cambridge

  • Contact the University
  • Accessibility statement
  • Freedom of information
  • Privacy policy and cookies
  • Statement on Modern Slavery
  • Terms and conditions
  • University A-Z
  • Undergraduate
  • Postgraduate
  • Cambridge University Press & Assessment
  • Research news
  • About research at Cambridge
  • Spotlight on...

research article on stem cell therapy

  • Open access
  • Published: 28 July 2022

Clinical application of mesenchymal stem cell in regenerative medicine: a narrative review

  • Ria Margiana 1 , 2 , 3 ,
  • Alexander Markov 4 , 5 ,
  • Angelina O. Zekiy 6 ,
  • Mohammed Ubaid Hamza 7 ,
  • Khalid A. Al-Dabbagh 8 ,
  • Sura Hasan Al-Zubaidi 9 ,
  • Noora M. Hameed 10 ,
  • Irshad Ahmad 11 ,
  • R. Sivaraman 12 ,
  • Hamzah H. Kzar 13 ,
  • Moaed E. Al-Gazally 14 ,
  • Yasser Fakri Mustafa 15 &
  • Homayoon Siahmansouri 16  

Stem Cell Research & Therapy volume  13 , Article number:  366 ( 2022 ) Cite this article

23k Accesses

116 Citations

19 Altmetric

Metrics details

The multipotency property of mesenchymal stem cells (MSCs) has attained worldwide consideration because of their immense potential for immunomodulation and their therapeutic function in tissue regeneration. MSCs can migrate to tissue injury areas to contribute to immune modulation, secrete anti-inflammatory cytokines and hide themselves from the immune system. Certainly, various investigations have revealed anti-inflammatory, anti-aging, reconstruction, and wound healing potentials of MSCs in many in vitro and in vivo models. Moreover, current progresses in the field of MSCs biology have facilitated the progress of particular guidelines and quality control approaches, which eventually lead to clinical application of MSCs. In this literature, we provided a brief overview of immunoregulatory characteristics and immunosuppressive activities of MSCs. In addition, we discussed the enhancement, utilization, and therapeutic responses of MSCs in neural, liver, kidney, bone, heart diseases, and wound healing.

Introduction

In the last decade, stem cells are increasingly applied as a therapeutic method for numerous disorders. Stem cell therapy, traditionally applied for hematopoietic disorders, nonetheless, is now established for the treatment of non-hematologic disorders [ 1 , 2 ].

Accumulating evidence has shown that mesenchymal stem cells (MSCs) offer an encouraging option for cell treatment and reconstruction of human tissues because of their differentiation multipotency, self‐renewal capacity, long‐term ex vivo proliferation, paracrine potentials, and immunoregulatory effect [ 3 ]. Furthermore, MSCs have the capability to support the progression and differentiation of other stem cells. They can release bioactive molecules, which is a key benefit in tissue regeneration [ 4 , 5 ]. These properties result in progression of treatments for a wide range of diseases, such as diseases affecting the bone, neuron, lung, liver, heart, kidney, etc. [ 4 ]. Due to these features, it is obvious that MSCs will hold a major therapeutic role in clinical trials. Because of these properties, we provided a general overview of the latest trials that studied the effectiveness of MSCs in several diseases such as neural, liver, kidney, bone, heart diseases, and wound healing.

Stem cells in regenerative medicine

In the last years, numerous studies have demonstrated that cellular therapy has exhibited great development in both in vitro and in vivo researches. Stem cells have the capability to self-renew, and also to differentiate into all cell types and are involved in physiological regeneration [ 6 ]. There are multiple stem cell sources of adult and pluripotent stem cells (PSCs) such as embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) for tissue regeneration. PSCs have a high potential for pluripotency and self-renewal, which makes these cells an important option for treatment of diseases. However, there are ethical issues when using these cells, in which ESCs are separated from blastocyst-stage embryos, requiring destruction of the embryo [ 7 , 8 , 9 ]. The results of studies have revealed the regenerative ability of iPSCs in preclinical setting and conducted the first clinical study for treatment of age-associated with macular deterioration [ 10 , 11 ]. Nonetheless, the tumorigenicity risk remains unsolved. Because of these limitations, researchers began to investigate adult stem cells, the multipotent stem cells found in tissues and organs of adults. Various investigations have reported that stem cell therapy can regenerate and repair injured organs in vivo, including bone repair, cutaneous wound, pulpitis, and ischemic cardiac tissue through stem cell differentiation and production of new particular cells [ 12 , 13 , 14 , 15 ]. Moreover, some investigations have demonstrated that cultured adult stem cells release many molecular factors with anti-apoptotic, immunoregulatory, angiogenic, and chemoattractant features that stimulate regeneration [ 16 , 17 , 18 ]. Hematopoietic stem cells (HSCs) and MSCs are part of adult stem cells, which are the most widely used, generally because they can be isolated from individuals in diseased conditions.

Mesenchymal stem cell

In the late 1960s, Friedenstein and colleagues discovered MSCs as multipotent stem cells for the first time [ 19 ]. MSCs are non-hematopoietic cells and have the capability to differentiate into various lineage including mesodermal (adipocytes, osteocytes, and chondrocytes), ectodermal (neurocytes), and endodermal lineage (hepatocytes) [ 20 , 21 ]. At the beginning, it was thought that MSCs are “stromal” cells instead of stem cells [ 22 ]. Several investigators tried to alter the name of MSCs to medicinal signaling cells due to their function in secretion of some metabolites molecules in the sites of diseases, injuries, and inflammations [ 23 , 24 ]. After that, some studies have stated that MSCs can release prostaglandin E2 (PGE2), which plays a major role in the self-renewal ability, immunomodulation of MSCs, and generating a cascade of events, that demonstrates the stemness of MSCs [ 25 ]. Therefore, the term mesenchymal stem cells is justified.

MSCs chiefly found in the bone marrow (BM) possess the ability of self-renewal and also display multilineage differentiation [ 8 , 26 , 27 ]. They were obtained from various tissues and organs including BM, adipose tissue, Wharton’s jelly, peripheral blood, umbilical cord, placenta, amniotic fluid, and dental pulp [ 3 , 28 , 29 , 30 ]. MSCs can express a wide range of surface markers and cytokine profiles according to the origin of isolation [ 31 ]. Nevertheless, the common characterization markers of MSCs are CD73, CD105, CD90 and lacking expression of CD45, CD34, CD14 or CD11b, CD79α or CD19, and HLA-DR [ 32 , 33 , 34 ]. During the last decades, MSCs have shown various biological roles such as multilineage differentiation, immunomodulation, angiogenesis, anti-apoptotic and anti-fibrotic activity, chemo-attraction, and tissue repair development [ 35 , 36 , 37 ]. The MSCs have broad properties that make them a suitable source for cell therapy, such as stemness potency, easily isolation from different sources, they can be rapidly expanded in a large scale for clinical use, have less ethical issues as compared to ESCs, unlike iPSCs, MSCs transport a lower risk of teratoma formation, and they are beneficial for a wide scale of therapeutic applications due to their capability to migrate to injured tissue through chemo-attraction [ 38 , 39 , 40 ]. In addition, MSCs can release a variety of bioactive components including proteins, growth factors chemokines, microRNAs (miRNAs), and cytokines which can suggest their acceptable application [ 41 ].

The biological roles of MSCs

MSCs have the ability to inhibit the immune response in inflammatory cytokine-rich situations, including infections, wounds, or immune-mediated disorders. These immunomodulatory properties were discovered in preclinical and clinical trials, where MSCs effectively suppressed T cell activation and proliferation along with stimulation of macrophages shift from M1 to M2 [ 42 , 43 , 44 ]. This specific performance of MSCs in the presence and absence of inflammatory mediators is termed MSC polarization. MSCs have the ability to migrate to damaged areas after systemic infusion and consequently exert a beneficial effect by various mechanisms, chiefly immunoregulation, and angiogenesis [ 45 , 46 ]. Although the related mechanism-mediated MSC immunosuppression has not been entirely clear, it appears that cellular interaction, accompanied by many factors, performs the principal function in this process. In the presence of high levels of inflammatory cytokines, e.g., TNF-α and IFN-γ, MSCs release several cytokines including TGF-β and hepatocyte growth factor (HGF) and produce soluble factors including indoleamine 2,3-dioxygenase (IDO), PGE2, and nitric oxide (NO). These mediators suppress T effector cells and enhance the expression of FOXP3, CTLA4, and GITR in regulatory T cells (Tregs) to increase their immunomodulation effects [ 47 , 48 , 49 ]. Moreover, cell-to-cell communication facilitates the stimulation of Tregs by cytokine-primed MSCs [ 50 ]. Overexpression of inducible co-stimulator ligands (ICOSL) induces the stimulation of efficient Tregs [ 51 ].

In addition, MSCs can enhance the generation of Treg cells indirectly. According to the literature, MSCs stimulate M2 macrophage and alter the phenotype through secretion of extracellular vesicles in an in vitro study [ 52 ]. Also, M2 cells that are activated by MSCs express CCL-18 and induce Treg cells [ 53 ]. Moreover, MSCs increase the expression of cyclooxygenase 2 (COX2) and IDO, resulting in expression of CD206 and CD163 in M2 cells, as well as enhance the expression of IL-6 and IL-10 in the microenvironment [ 54 ]. The overexpression of IL-10 that is produced by dendritic cells (DCs) and M2 cells upon MSCs co-culture leads to further immunomodulation via inhibition of effector T cells [ 55 , 56 ]. Furthermore, the secretion of IDO from MSCs can induce the proliferation, activation, and IgG releasing of B cells, thereby suppressing T effector cells [ 57 , 58 ].

One of the typical properties of MSCs is their multipotency capacity in which these stem cells are able to differentiate into a number of tissues in vitro [ 59 ]. Chondrogenic differentiation of MSCs in vitro occurs commonly via culturing them in the existence of TGF-β1 or TGF-β3, IGF-1, FGF-2, or BMP-2 [ 60 , 61 , 62 , 63 ]. MSC differentiation into chondroblasts is characterized by the increasing of various genes such as collagen type II, IX, aggrecan, and proliferation of chondroblast cell morphology. During the process of chondrogenesis, FGF-2 promotes the MSCs induced with TGF-β1 or TGF-β3 and/ or IGF-1 [ 64 ]. According to the literature works, several molecular pathways such as hedgehog, Wnt/β-catenin, TGF-βs, BMPs, and FGFs can regulate chondrogenesis [ 65 ]. In addition, MSCs can exert the osteogenesis function by inducing MSCs with ascorbic acid, β-glycerophosphate, vitamin D3, and/or BMP-2, BMP-4, BMP-6, and BMP-7 [ 66 ].

One of the major abilities of MSCs is anti-fibrotic activity. These cells can differentiate into various cell lineages such as hepatocytes, both in vivo and in vitro [ 67 ]. MSCs contain multiple trophic factors which induce cells and matrix remodeling to stimulate progenitor cells and the recovery of damaged cells. MSCs can decrease myofibroblasts and reverse the fibrotic activity of injured tissues [ 68 ]. Furthermore, these cells release pro-angiogenic factors including VEGF, IGF-1, and anti-inflammatory factors that participate in the recovery of tissue function. For instance, MSCs can increase neovascularization of ischemic myocardium through VEGF in a mice model of heart disease [ 69 ]; also, IGF-1 exerts an advantageous effect on the survival and proliferation of cardiomyocytes [ 70 ].

Bone marrow mesenchymal stem cell-based regenerative medicine

So far, increasing data have lately studied the effects of MSCs in the treatment or regeneration of various disorders (Table 1 ). In this section, we reviewed the latest clinical studies that investigate the potential contribution of MSCs in the regenerative medicine, as shown in Fig.  1 .

figure 1

Effect of bone marrow mesenchymal stem cell-based regenerative medicine

Neural regeneration

The application of BMSCs has demonstrated promising therapeutic results in the treatment of neurological diseases. Amyotrophic lateral sclerosis (ALS), also known as motor neuron disease, is a neurodegenerative disorder that leads to degeneration of the motor neurons that causes paralysis and muscle weakness [ 138 , 139 ]. Syková et al. [ 71 ] carried out a study that intrathecally injected 15 ± 4.5 × 10 6 autologous BMSCs into 26 patients with ALS. After mesenchymal stem cells transplantation (MSCT), ALS functional rating scale (ALSFRS) significantly reduced, forced vital capacity (FVC) remained stable or above 70%, and weakness scales (WSs) were stable in 75% of patients. They have shown that the intrathecal BMSCs intervention in ALS patients is a safe method and it can slow down the development of the disease. There were no significant adverse events related to the trial during and after transplantation of BMSCs. Barczewska and colleagues indicated that three intrathecal injections of 30 × 10 6 Wharton’s jelly-MSCs (WJ-MSCs) improved ALSFRS [ 77 ]. They showed that WJ-MSCs are safe and effective in individuals that suffer from ALS. However, one other group found that intrathecal injection of autologous adipose MSCs does not improve clinical symptoms of ALS patients [ 76 ]. Their results indicated that the levels of CSF protein and nucleated cells were increased and ALSFRS-R showed development of disease in all treated patients. In the trial by OH et al., autologous BMSCs were injected to treat seven participants that suffer from ALS [ 75 ]. The participants were injected twice with autologous BMSCs (one million cells per kg) and followed up for 12 months. No serious adverse events were reported during the follow-up period. Furthermore, during the 12-month follow-up, there was no acceleration in the decrease in the ALSFRS-Revised (ALSFRS-R) score, Appel ALS score, and FVC. Moreover, CSF analysis showed that the levels of TGF-β and IL-10 were evaluated, while MCP-1, which is chemokine-related and exacerbates the motor neuron damage in ALS, was decreased. Their results exhibited that two repeated MSC infusions have safety and feasibility for at least 1 year in seven individuals; nevertheless, the study has some limitations such as low number of participants and short-time follow-up. In another study [ 73 ], 15 ALS patients were transplanted with autologous BMSCs. These 15 patients were divided into two groups (group 1: patients who had ALS with an inherently slow course, group 2: individuals who had ALS with an inherently rapid course) and received three intrathecal infusions of MSCs. There were no significant adverse events in the course of multiple intrathecal injections of MSCs. In group 1, there were no major changes in the rate of disease development and in group 2 ameliorating of the disease was indicated following MSCs therapy. According to their observation, the response of patients with ALS to treatment with MSCs was variable. Also, the authors indicated that due to the small number of patients, less subgroups were available for statistical analysis, limiting their ability to draw conclusions from the data.

Spinal cord injury (SCI) is usually related to devastating results. The damage to the spinal cord leads to injury to the motor, sensory, and autonomic roles of the spinal cord that affects patients’ well-being such as their physical and psychological state [ 140 , 141 ]. In a phase I, nonrandomized, uncontrolled study by Mendonça et al. [ 84 ], 15 SCI patients were administered 1 × 10 7 cells/ml MSCs. The results of the investigation revealed that SCI symptoms were meaningfully decreased by MSCT, all participants showed variable improvements in tactile sensitivity, and eight participants improved lower limb motor functional gains, chiefly in the hip flexors. Seven patients revealed sacral sparing and developed American Spinal Injury Association impairment scale (AIS) grades B or C – partial damage. Nine participants had developments in urologic function and one patient showed alterations in somatosensory evoked potentials (SSEP) 3 and 6 months after MSCT. These results stated that treatment with MSCs ameliorated the organ malfunction in people with SCI and has clinical safety, because no serious adverse effects were reported. The authors indicated that their results should be confirmed in larger and controlled clinical trials. Albu and colleagues have been demonstrated that intrathecal administration of WJ-MSCs considerably improved the pinprick sensation in the dermatomes below the level of damage [ 88 ]. Further results showed that bladder maximum capacity was elevated and bladder neurogenic hyperactivity and external sphincter dyssynergy were reduced. In another study [ 85 ], ten SCI subjects received four subarachnoid injections of 30 × 10 6 autologous BMSCs, maintained in autologous plasma, at weeks 1, 16, 28, and 40 of the trial and followed up for 12 months. There were no adverse events and all participants tolerated the therapy. Vaquero et al. [ 86 ] demonstrated that MSCT is safe and improves sensitivity, motor power, spasms, spasticity, neuropathic pain, sexual function, or sphincter dysfunction in the SCI patients. The results of their study have shown that 55.5% of patients improved in SSEP and 44.4% of patients ameliorated in voluntary muscle contraction together with intralesional active muscle reinnervation. Hur et al. carried out a study in which 14 patients with SCI were administered intrathecally 9 × 10 7 adipose MSCs [ 87 ]. Their observations showed mild progresses in neurological function. No serious adverse events were observed. In a phase 2 study, 13 patients with SCI were intravenously administered a single dose of autologous MSCs cultured in auto-serum [ 82 ]. The results of this trial revealed that SCI symptoms were considerably declined by MSC therapy, ASI, International Standards for Neurological and Functional Classification of Spinal Cord (ISCSCI-92), and Spinal Cord Independence Measure (SCIM-III) demonstrated functional improvements after MSC injection. No severe adverse effects were related to MSC administration.

Parkinson’s disease (PD) is a neurological disorder principally characterized by the deterioration of motor activities due to the impairment of the dopaminergic nigrostriatal system [ 142 , 143 ]. It has been indicated that MSCs improved the symptoms of PD. In a phase I controlled, randomized clinical study, patients that suffer from progressive supranuclear palsy were administered autologous BMSCs via intra-arterial injection [ 78 ]. The results of the study exhibited that autologous BMSCs are safe and reduce disease progression. Canesi et al. [ 79 ] have demonstrated that injection of MSCs into cerebral arteries of PD patients led to positive results in 17 PD participants: all treated participants were alive and motor function rating scales remained stable for at least 6 months during the 12-month follow-up period. One patient died 9 months after the injection for reasons not associated with cell infusion or to disease development.

In a study conducted by Jaillard and colleagues in 2019 [ 89 ], 31 individuals with subacute stroke were administered the intravenous injections of autologous BMSCs. The results of the trial exhibited significant improvements in motor-National Institute of the Health Stroke Scale (NIHSS) score, motor-Fugl-Meyer scores, and task-related functional MRI activity in motor cortex-4a. However, there was no remarkable progress in Barthel Index, NIHSS, and modified Rankin scores. In general, their results suggested that BMSCs improved motor recovery via sensorimotor neuroplasticity. In another study, 17 patients with subacute middle cerebral artery infarct received two million cells/kg autologous BMSCs [ 92 ]. During the follow-up process, NIHSS score, modified Rankin Scale or Barthel Index did not improve after the transplantation. Nonetheless, there was a significant improvement in absolute change in median infarct volume, but no treatment-related adverse effects were observed.

In sum, these outcomes suppose that BMSCs can safely and efficiently treat neural diseases, inhibit disease development, and considerably ameliorate the quality of life and clinical manifestations of patients. Consequently, BMSCs can become a new option for the clinical treatment of neural diseases.

Liver regeneration

The potential of BMSCs to differentiate into the endodermal lineage, such as hepatocyte‐like cells, makes them an attractive alternative for the treatment of liver diseases [ 144 ]. Some clinical studies have demonstrated the efficacy and feasibility of BMSC therapy in patients with liver diseases. The effect of BMSCs has been studied in individuals suffering from liver cirrhosis by Suk et al. [ 98 ]. Seventy-two patients were enrolled in this trial and randomly classified into three groups: one control group and two autologous BMSC groups that received one-time or two-time hepatic arterial administrations of fifty million autologous BMSCs 30 days after BM aspiration. Fibrosis quantification exhibited that in one-time and two-time BMSC groups there are a reduction of 25% and 37% in the proportion of collagen, respectively. In addition, the Child–Pugh (CP) scores of both test groups were meaningfully improved following BMSC administration in comparison with the control group. No serious adverse events were associated with MSC injection during the 12-month follow-up. Wang and coworkers have found that intravenous injection of UC-MSCs (0.5 × 10 6 cells/kg) is feasible and well tolerated in patients with primary biliary cirrhosis (PBC) [ 93 ]. They exhibited that MSCs significantly decreased the level of ALP and GGT; however, there were no considerable changes in serum AST, ALT, total bilirubin, albumin, prothrombin time activity, or immunoglobulin M levels. Similarly, Zhang et al. [ 94 ] have demonstrated that intravenous administration of 1.0 × 10 6 cells/kg UC-MSCs is safe and efficient for patients with ischemic-type biliary lesions after liver transplantation. According to their results, MSCs therapy reduced the serum ALP, GGT, and total bilirubin. In a randomized placebo-controlled phase I–II single-center study, nine patients that suffer from acute-on-chronic liver failure (ACLF) grades 2 and 3 were enrolled [ 95 ]. The experiment group (n = 4) received standard medical therapy along with five injections of 1 × 10 6 cells/kg of BMSC for 3 weeks. There were no transplant-related adverse events; however, one patient in the experiment group showed hypernatremia and a gastric ulcer, after the third and fifth administrations, respectively. Furthermore, MSCT revealed a considerable improvement in CP, model for end-stage liver disease (MELD), and ACLF (grade 3 to 0). Thus, MSCT is safe and viable in individuals with ACLF. In an open-label non-blinded randomized controlled study conducted by Lin et al. [ 96 ], 110 patients with hepatitis B virus (HBV)-related ACLF were enrolled in this trial. These patients were divided into two groups: control group (N = 54) was treated with standard medical therapy only and the intervention group (N = 56) was injected four times with 1.0–10 × 10 5 cells/kg allogeneic BMSCs, and then followed up for 6 months. There were no serious adverse events associated with transplantation. The results of that study demonstrated that MSCT significantly improved clinical laboratory measurements, such as serum total bilirubin, and MELD scores in comparison with control group. In addition, mortality from multiple organ failure and prevalence rate of serious infection in the intervention group was lower than that in the control group. Their results clearly established the safety and feasibility of the clinical use of peripheral administration of allogeneic BMSCs for subjects with HBV-associated ACLF, and markedly enhanced the survival rate through enhancing liver function and reducing the prevalence of severe infections.

In summary, MSCT can meaningfully ameliorate the clinical manifestations of these patients, reduce the liver fibrosis, and inhibit the development of disease.

Kidney regeneration

Hurt to renal cells can occur because of a wide range of ischemic and toxic insults and results in inflammation and cell death, which can lead to kidney damage. Inflammation has a significant role in the damage of renal cells, as well as following cellular regeneration processes [ 3 , 145 ]. Various investigations have consistently demonstrated a supportive effect of MSC on acute and chronic renal injury [ 146 ]. Makhlough et al. declared that intravenous administration of 1–2 × 10 6 cells/kg into seven patients with chronic kidney disease failed to induce remission [ 101 ]. They indicated that variations in estimated glomerular filtration rate (eGFR) and serum creatinine during the 18-month follow-up were not statistically significant. Nonetheless, no severe adverse events were reported, and they could not assess the efficacy because of their study design. Authors postulated that limited sample size and lack of a control group led to the lack of success. A study conducted by Swaminathan et al. in 2021, has displayed the effect of allogeneic BMSCs in acute kidney injury patients. They have shown that treatment of MSCs with SBI-101 stimulated an immunotherapeutic response that initiated an enhanced phenotypic alteration from tissue injury to tissue repair [ 102 ]. In a single-arm phase I clinical trial carried out by Makhlough et al. [ 100 ], six patients with autosomal dominant polycystic kidney disease (ADPKD) were intravenously injected 2 × 10 6 cells/kg autologous BMSCs. The results of the study showed that the mean eGFR value declined and the level of serum creatinine enhanced during the 1-year follow-up. Moreover, no remarkable modifications in renal function parameters and blood pressure were observed during the year after intervention. However, there were no severe adverse events after 1-year follow-up. In addition, the authors indicated that there are some reasons for the lack of success, including small number of patients, absence of a comparison group, limited follow-up period, single dose administration, and they did not utilize htTKV as a surrogate endpoint. Abumoawad and colleagues have established that adipose MSCs enhanced blood flow, GFR and reduced inflammatory injury in poststenotic kidneys of individuals that suffer from atherosclerotic renovascular disease (ARVD) [ 99 ]. Their results illustrated that mean renal blood flow was considerably enhanced, and hypoxia, renal vein inflammatory cytokines, and angiogenic factors were considerably attenuated.

Heart regeneration

Heart disease is the first and most frequently diagnosed disease and the leading cause of disease death [ 147 ]. When cardiomyocytes are damaged via ischemic and other factors, the remaining viable cardiomyocytes have a restricted ability to proliferate and dead cardiomyocytes are changed by non-contractile fibrous tissue, leading to functional impairment that elicits the progression of heart failure. According to the developing number of patients with heart disease, there is a vital need to expand an innovative remedy to rescue deteriorating hearts. Regenerative medicine and cell therapy are the upcoming therapeutic opportunities for heart diseases. According to the literature, the transplantation of BM-derived cells and cardiac stem cells into deteriorating hearts appeared to provide functional benefits [ 148 , 149 ].

In a study by Yagyu et al. [ 110 ], 8 individuals with symptomatic heart failure were infused with BMSCs. During the follow-up period, no serious adverse events were observed. There were no major differences in B-type natriuretic peptide, left ventricular ejection fraction (LVEF), and peak oxygen uptake at 2 months. The results of this study recommend further research regarding the feasibility and efficacy of MSCs. In a study by Gao et al. [ 107 ], 116 patients with acute myocardial infarction randomly received an intracoronary injection of WJ-MSCs. They indicated that MSCs therapy elevated the myocardial viability and perfusion within the infarcted territory. In addition, the LVEF was elevated and LV end-systolic volumes and end-diastolic volumes were decreased in the WJ-MSCs group.

Chan et al. demonstrated that intramyocardial infusion of autologous BMSCs in conjunction with transmyocardial revascularization or coronary artery bypass graft surgery was technically feasible and could be performed safely. The results showed that regional contractility in the cell-treated regions improved during the 1-year follow-up; also, the quality of life was improved along with a substantial decrease in angina scores at 12 month post-treatment [ 104 ]. In a study by Kaushal et al. [ 113 ], 12 participants with hypoplastic left heart syndrome were transplanted with allogeneic human MSCs (2.5 × 10 5 cells/kg). This study determined the safety, feasibility, and usefulness of MSC administration into the left ventricular myocardium. No serious adverse effects were reported during the trial. Mathiasen et al. observed that after BM-MSCT, left ventricular end-systolic volume was significantly reduced, also LVEF, stroke volume, and myocardial mass remarkably improved [ 103 ]. In addition, a major decrease in the amount of scar tissue and quality of life score was observed. No side effects were identified. In a randomized, double-blind, placebo-controlled, multicenter, phase II study, 100 patients with anterior ST elevation myocardial infarction received autologous BMSCs and atorvastatin (ATV) treatment. The results of that study represented the absolute change of LEVF within 12 months, improvement in cardiac function, induction of remodeling and regeneration, and improvement in quality of life [ 108 ]. Recently, Celis-Ruiz and coworkers conducted a study in which intravenous administration of adipose MSCs within the first 2 weeks of ischemic stroke onset is safe at 24 months of follow-up [ 106 ]. In a study conducted by Hare et al. [ 112 ], 37 non-ischemic dilated cardiomyopathy patients were divided into two groups and received 10 × 10 7 allogeneic and autologous BMSCs. Minnesota Living with Heart Failure Questionnaire score decreased in both groups. The major adverse cardiac event rate was lower in allo vs. auto. Also, TNF-α decreased, to a greater extent in allo vs. auto at 6 months. These results suggested the clinically meaningful efficacy of allogeneic vs. autologous BMSCs in non-ischemic dilated cardiomyopathy patients. Qayyum et al. have found that intra‑myocardial injections of autologous adipose MSCs ameliorated cardiac functions and unchanged exercise capacity, in contrast to deterioration in the placebo group [ 115 ].

Levy et al. indicated that after allogeneic BMSCs in patients with chronic stroke, Barthel Index scores increased. Moreover, electrocardiograms, laboratory tests, and computed tomography scans of chest/abdomen/pelvis suggest that BMSCs could alleviate the clinical symptoms in patients with stroke [ 90 ].

In sum, BMSC therapy can be an effective, achievable, and safe process that remarkably improves cardiac function and promotes patients’ quality of life.

Bone regeneration

Bone regeneration is a hot topic of research in clinical studies. Bone regeneration is a crucial problem in numerous cases, including bone fracture, defect, osteoarthritis, and osteoporosis, which should be resolved [ 150 , 151 , 152 ]. Autogenous bone grafts are considered the standard approach for bone formation by means of the participants’ own cells that stimulate osteoinductive, bone conductivity, and histocompatibility in bone diseases [ 153 ]. Nevertheless, there are some shortcomings of this procedure such as unpredictable absorption, extended recovery time, and patients commonly experience pain and nerve injury at the harvest area [ 154 , 155 , 156 ]. With the development of understanding bone tissue biology as well as recent approaches in the improvement in tissue regeneration, the application of MSC has become an attractive subject in augmenting bone tissue forming [ 157 , 158 ].

In a pilot study by Jayankura and coworkers, allogeneic BMSCs were applied to treat 22 participants with bone fractures [ 128 ]. All participants received percutaneous implantation of autologous BMSCs (5 to 10 × 10 7 cells) into the fracture area. After intervention, Tomographic Union Score (TUS) and Global Disease Evaluation (GDE) score were improved, and pain at palpation at the fracture site was reduced. In addition, the ratio of blood samples comprising donor-specific anti-HLA antibodies enhanced at 6 months post-intervention. Three serious cell-related adverse events were reported. In another study by Shim and coworkers [ 129 ], intramedullary (4 × 10 7 cells) and intravenous (2 × 10 8 cells) infusion of WJ-MSCs in combination with teriparatide showed beneficial results in individuals with osteoporotic vertebral compression fractures. Their observation displayed that the mean visual analog scale, Oswestry Disability Index, and Short Form-36 scores meaningfully improved. They stated that WJ-MSCs in combination with teriparatide are viable and have a clinical profit for fracture healing by stimulating bone architecture.

Several studies investigated the effect of BMSCs in osteoarthritis (OA) patients. Chahal et al. carried out a clinical phase I/IIa trial that involved 12 individuals with late-stage Kellgren–Lawrence knee OA. These 12 patients were injected with a single intra-articular of 1 × 10 6 , 10 × 10 6 , and 50 × 10 6 BMSCs. The results showed that patients had improved Knee Injury and Osteoarthritis Outcome Score (KOOS) pain, symptoms, quality of life, and Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) stiffness relative to baseline. Moreover, cartilage catabolic biomarkers and MRI synovitis were meaningfully lower at higher doses and the levels of pro-inflammatory monocytes/macrophages and IL-2 reduced in the synovial fluid after intervention. No serious events had occurred [ 116 ]. Dilogo et al. have reported that UC-MSCs (10 × 10 6 cells) significantly decreased the WOMAC and could be a potentially new regenerative treatment for patients with knee OA [ 127 ]. In a study conducted by Hernigou et al. [ 117 ], 140 patients with OA received a subchondral infusion of BMSCs on one side and received total knee arthroplasty (TKA) on the contralateral knee. They demonstrated that subchondral MSCs had a significant effect on pain to postpone or avoid the TKA in the contralateral joint of patients with OA. In a phase II multicenter randomized controlled clinical trial, 60 OA patients received 10 × 10 7 cells of autologous BMSCs along with platelet-rich plasma and followed up for 12 months [ 119 ]. No serious adverse effects were observed after MSCs injection or during follow‑up. According to the observations, treatment with BMSC related to platelet-rich plasma was demonstrated to be a feasible alternative treatment for individuals with OA, along with clinical development at the end of follow-up. Similarly, Bastos et al. have reported that MSCs alone or in combination with platelet-rich plasma are safe and have an advantageous effect on symptoms in OA individuals [ 121 ]. They found that MSCs group and MSCs + platelet-rich plasma group can improve the pain, function and daily living activities, and quality of life subscales. Ten adverse events were reported in three participants in the MSCs group and in two of the MSCs + platelet-rich plasma group. PERS and colleagues reported another clinical phase Ia study that involved 19 individuals suffering from knee OA [ 123 ]. These 18 individuals were classified into three groups and received a single intra-articular administration of 2 × 10 6 , 10 × 10 6 , and 50 × 10 6 adipose MSCs. According to their results, individuals had experienced significant improvement in pain levels and function. There were no severe adverse events; however, 4 individuals experienced transient knee joint pain and swelling after local administration. In a long-term follow-up of a multicenter randomized controlled clinical trial by Espinosa et al. [ 120 ], 30 OA patients were administered the intra-articular infusion of two diverse doses of autologous BMSCs cells (10 × 10 6 or 10 × 10 7 ) versus hyaluronic acid in the treatment of OA. No adverse effects occurred after MSCT or during the 4-year follow‑up. Their results showed that intra-articular infusion of BMSCs together with hyaluronic acid is a safe and viable process that leads to a clinical and functional improvement in knee OA.

Overall, these data display that BMSCs can be a promising, safe and effective alternative for bone regeneration, significantly improve the clinical manifestation of patients, and inhibit development of diseases.

Wound regeneration

The skin has several layers along with different compounds and roles that work together to support internal organs and serve various biological roles. It has three main layers, the epidermis, the dermis, and the subcutaneous layer [ 159 ]. Generally, skin wound healing, triggered by tissue injury, includes four stages: hemostasis, inflammation, proliferation, and maturation. MSCs can assist in all stages of the wound healing process. The use of MSCs for the treatment of skin can improve the regeneration of skin and reduce scarring. MSCs exert their functions through migration into the skin damage site, suppressing inflammation, and increasing the growth and differentiation ability of fibroblasts, epidermal cells, and endothelial cells [ 160 , 161 ]. As MSCs have exhibited wound healing in many preclinical studies, the application of MSCs for chronic wounds contributes to progress toward clinical trials. Falanga et al. have demonstrated that autologous BMSCs are an impressive and safe treatment method for wound healing [ 131 ]. The results of the study indicated a trend toward a reduction in ulcer size or complete wound closure by 4–5 months. No adverse events were noted. In a study by Zhou et al., 346 patients with skin wounds were administered adipose MSCs [ 132 ]. There were no adverse events during the trial. They reported that the granulation tissue coverage rate and thickness of granulation tissue were considerably ameliorated. In an open-label phase I/II study, sixteen participants with vocal fold scarring were administered a single dose of 0.5–2 × 10 6 cells autologous MSCs [ 137 ]. Video ratings of vocal fold vibrations and digitized analysis of high-speed laryngoscopy and phonation pressure threshold were considerably enhanced for 62–75% of the participants. Voice Handicap Index was meaningfully enhanced in eight participants, with the remaining experiencing no remarkable alteration. No serious adverse events or minor side effects were reported. Lonardi et al. observed that micro-fragmented adipose tissue improved skin tropism in patients with diabetic foot ulcer [ 135 ]. Furthermore, the results of studies have shown that adipose-derived stem cells had a beneficial effect on the full-thickness foot dorsal skin wound in diabetic mice with a considerably decreased ulcer area [ 162 ]. Recently, Huang et al. carried out a clinical study in which six subjects with intrauterine adhesion and four with cesarean scar diverticulum enrolled in this trial [ 136 ]. They found that intrauterine injection of UC-MSCs improved the endometrial thickness, cesarean scar diverticulum, and the volume of the uterus.

In the last decades, optimizations of isolation, culture, and differentiation procedures have permitted MSCs to improve closer to clinical uses for improving disorders and various tissue regeneration. MSCs have some important characteristics that make them preferred candidates to use for regenerative medicine: immunomodulatory capability valuable to improve immune system abnormalities, paracrine or autocrine roles that produce growth factors, and the vital potential to differentiate into various cells. Several clinical trials have reported that both autologous and allogeneic MSCs are valuable sources for tissue forming. Particularly, autologous MSCs signify the chief sources examined safe for administration and minimization of immunological threat, regardless of the lack of reported grievances concerning allogeneic MSC-based therapy. According to the studies described in this literature, administration of MSCs appear to be more effective and the usefulness of MSC therapy in bone and heart disorders has been broadly established. In terms of safety, no significant relationship was found between the MSC therapy and incidence of cancer and infection. Intravenous injection of MSCs is the most widely used form of administration and the dosage commonly fluctuates between 1 × 10 6 cells/kg and 2 × 10 8 cells/kg. According to the literature works mentioned in this review, the repeated administration of MSCs suggests being more beneficial than a single injection. In addition, the effectiveness of MSCs therapy in osteoarthritis disorder has been widely established. Long-term follow-up studies exhibited that serum tumor markers did not enhance before and 3 years after MSCs therapy. Nevertheless, there is still a lack of reliable scientific data on the mechanisms whereby the MSC therapy improves the numerous disorders that can develop the MSC modification and increase their prospective clinical application.

Availability of data and materials

Not applicable.

Abbreviations

Amyotrophic lateral sclerosis

Association impairment scale

ALS functional rating scale

ALSFRS-revised

Acute-on-chronic liver failure

Autosomal dominant polycystic kidney disease

Atorvastatin

Bone marrow

Bone marrow mesenchymal stem cells

Cyclooxygenase 2

Dendritic cells

Embryonic stem cells

Estimated glomerular filtration rate

Forced vital capacity

Global Disease Evaluation

Hematopoietic stem cells

Hepatitis B virus

Hepatocyte growth factor

Induced pluripotent stem cells

Indoleamine 2,3-dioxygenase

Inducible co-stimulator ligands

International Standards for Neurological and Functional Classification of Spinal Cord

Knee injury and osteoarthritis outcome score

Left ventricular ejection fraction

Mesenchymal stem cells

Mesenchymal stem cells transplantation

Model for end-stage liver disease

Nitric oxide

Osteoarthritis

Pluripotent stem cells

Prostaglandin E2

Spinal cord injury

Somatosensory evoked potentials

Spinal cord independence measure

Regulatory T cells

Tomographic Union Score

Total knee arthroplasty

Weakness scales

Western Ontario and McMaster Universities Osteoarthritis Index

Fugger L, Jensen LT, Rossjohn J. Challenges, progress, and prospects of developing therapies to treat autoimmune diseases. Cell. 2020;181(1):63–80.

Article   CAS   PubMed   Google Scholar  

Swart JF, et al. Haematopoietic stem cell transplantation for autoimmune diseases. Nat Rev Rheumatol. 2017;13(4):244–56.

Abbaszadeh H, et al. Regenerative potential of Wharton’s jelly-derived mesenchymal stem cells: a new horizon of stem cell therapy. J Cell Physiol. 2020;235(12):9230–40.

Saeedi P, Halabian R, Imani Fooladi AA. A revealing review of mesenchymal stem cells therapy, clinical perspectives and Modification strategies. Stem Cell Investig. 2019;6:34.

Article   CAS   PubMed   PubMed Central   Google Scholar  

Patel DM, Shah J, Srivastava AS. Therapeutic potential of mesenchymal stem cells in regenerative medicine. Stem Cells Int. 2013;2013: 496218.

Article   PubMed   PubMed Central   CAS   Google Scholar  

Rao M. Stem cells and regenerative medicine. Stem Cell Res Ther. 2012;3(4):27.

Article   PubMed   PubMed Central   Google Scholar  

Ilic D, Ogilvie C. Concise Review: Human Embryonic Stem Cells-What Have We Done? What Are We Doing? Where Are We Going? Stem Cells. 2017;35(1):17–25.

Zakrzewski W, et al. Stem cells: past, present, and future. Stem Cell Res Ther. 2019;10(1):68.

Chen Y, et al. Dental-derived mesenchymal stem cell sheets: a prospective tissue engineering for regenerative medicine. Stem Cell Res Ther. 2022;13(1):38.

Kanemura H, et al. Tumorigenicity studies of induced pluripotent stem cell (iPSC)-derived retinal pigment epithelium (RPE) for the treatment of age-related macular degeneration. PLoS ONE. 2014;9(1):e85336–e85336.

Souied E, Pulido J, Staurenghi G. Autologous induced stem-cell-derived retinal cells for macular degeneration. N Engl J Med. 2017;377(8):792–3.

Article   PubMed   Google Scholar  

Rong X, et al. Antler stem cell-conditioned medium stimulates regenerative wound healing in rats. Stem Cell Res Ther. 2019;10(1):326.

Hong H, et al. Dental follicle stem cells rescue the regenerative capacity of inflamed rat dental pulp through a paracrine pathway. Stem Cell Res Ther. 2020;11(1):333.

Chimutengwende-Gordon M, Khan WS. Advances in the use of stem cells and tissue engineering applications in bone repair. Curr Stem Cell Res Ther. 2012;7(2):122–6.

Yu Y, et al. Human embryonic stem cell-derived cardiomyocyte therapy in mouse permanent ischemia and ischemia-reperfusion models. Stem Cell Res Ther. 2019;10(1):167.

Jin L, et al. Mesenchymal stem cells ameliorate myocardial fibrosis in diabetic cardiomyopathy via the secretion of prostaglandin E2. Stem Cell Res Ther. 2020;11(1):122.

Chugh RM, et al. Mesenchymal stem cell therapy ameliorates metabolic dysfunction and restores fertility in a PCOS mouse model through interleukin-10. Stem Cell Res Ther. 2021;12(1):388.

Saldaña L, et al. Immunoregulatory potential of mesenchymal stem cells following activation by macrophage-derived soluble factors. Stem Cell Res Ther. 2019;10(1):58.

Friedenstein AJ, Piatetzky S II, Petrakova KV. Osteogenesis in transplants of bone marrow cells. J Embryol Exp Morphol. 1966;16(3):381–90.

CAS   PubMed   Google Scholar  

Abbaszadeh H, et al. Human umbilical cord mesenchymal stem cell-derived extracellular vesicles: a novel therapeutic paradigm. J Cell Physiol. 2020;235(2):706–17.

Chang D, et al. Application of mesenchymal stem cell sheet to treatment of ischemic heart disease. Stem Cell Res Ther. 2021;12(1):384.

Horwitz EM, et al. Clarification of the nomenclature for MSC: the international society for cellular therapy position statement. Cytotherapy. 2005;7(5):393–5.

Caplan AI. What’s in a name? Tissue Eng Part A. 2010;16(8):2415–7.

Caplan AI. Mesenchymal stem cells: time to change the name! Stem Cells Transl Med. 2017;6(6):1445–51.

Lee BC, et al. PGE2 maintains self-renewal of human adult stem cells via EP2-mediated autocrine signaling and its production is regulated by cell-to-cell contact. Sci Rep. 2016;6:26298.

Jiang Y, et al. Pluripotency of mesenchymal stem cells derived from adult marrow. Nature. 2002;418(6893):41–9.

Ding DC, Shyu WC, Lin SZ. Mesenchymal stem cells. Cell Transplant. 2011;20(1):5–14.

Meirelles LDS, Chagastelles PC, Nardi NB. Mesenchymal stem cells reside in virtually all post-natal organs and tissues. J Cell Sci. 2006;119(11):2204–13.

Article   CAS   Google Scholar  

Ghorbani F, et al. Renoprotective effects of extracellular vesicles: a systematic review. Gene Reports. 2022;26: 101491.

Article   Google Scholar  

Tang Y, Zhou Y, Li H-J. Advances in mesenchymal stem cell exosomes: a review. Stem Cell Res Ther. 2021;12(1):71.

Wu Y, et al. Adipose tissue-derived mesenchymal stem cells have a heterogenic cytokine secretion profile. Stem Cells Int. 2017;2017:4960831.

Mushahary D, et al. Isolation, cultivation, and characterization of human mesenchymal stem cells. Cytometry A. 2018;93(1):19–31.

Barberini DJ, et al. Equine mesenchymal stem cells from bone marrow, adipose tissue and umbilical cord: immunophenotypic characterization and differentiation potential. Stem Cell Res Ther. 2014;5(1):25.

Abbaszadeh H, et al. Chronic obstructive pulmonary disease and asthma: mesenchymal stem cells and their extracellular vesicles as potential therapeutic tools. Stem Cell Res Ther. 2022;13(1):262.

Jiang XX, et al. Human mesenchymal stem cells inhibit differentiation and function of monocyte-derived dendritic cells. Blood. 2005;105(10):4120–6.

Marinescu C-I, Preda MB, Burlacu A. A procedure for in vitro evaluation of the immunosuppressive effect of mouse mesenchymal stem cells on activated T cell proliferation. Stem Cell Res Ther. 2021;12(1):319.

Malekpour K, et al. The potential use of mesenchymal stem cells and their derived exosomes for orthopedic diseases treatment. Stem Cell Rev Reports. 2022;18(3):933–51.

Steens J, Klein D. Current strategies to generate human mesenchymal stem cells in vitro. Stem Cells Int. 2018;2018:6726185.

Beeravolu N, et al. Isolation and characterization of mesenchymal stromal cells from human umbilical cord and fetal placenta. J Vis Exp. 2017;122:e55224.

Google Scholar  

Hmadcha A, et al. Therapeutic potential of mesenchymal stem cells for cancer therapy. Front Bioeng Biotechnol. 2020;8:43.

Aravindhan S, et al. Mesenchymal stem cells and cancer therapy: insights into targeting the tumour vasculature. Cancer Cell Int. 2021;21(1):158.

Di Nicola M, et al. Human bone marrow stromal cells suppress T-lymphocyte proliferation induced by cellular or nonspecific mitogenic stimuli. Blood. 2002;99(10):3838–43.

Bartholomew A, et al. Mesenchymal stem cells suppress lymphocyte proliferation in vitro and prolong skin graft survival in vivo. Exp Hematol. 2002;30(1):42–8.

Djouad F, et al. Immunosuppressive effect of mesenchymal stem cells favors tumor growth in allogeneic animals. Blood. 2003;102(10):3837–44.

Swartzlander MD, et al. Immunomodulation by mesenchymal stem cells combats the foreign body response to cell-laden synthetic hydrogels. Biomaterials. 2015;41:79–88.

Rigotti G, et al. Expanded stem cells, stromal-vascular fraction, and platelet-rich plasma enriched fat: comparing results of different facial rejuvenation approaches in a clinical trial. Aesthet Surg J. 2016;36(3):261–70.

Djouad F, et al. Reversal of the immunosuppressive properties of mesenchymal stem cells by tumor necrosis factor α in collagen-induced arthritis. Arthritis Rheum. 2005;52(5):1595–603.

Ge W, et al. Infusion of mesenchymal stem cells and rapamycin synergize to attenuate alloimmune responses and promote cardiac allograft tolerance. Am J Transplant. 2009;9(8):1760–72.

Waterman RS, et al. A new mesenchymal stem cell (MSC) paradigm: polarization into a pro-inflammatory MSC1 or an Immunosuppressive MSC2 phenotype. PLoS ONE. 2010;5(4): e10088.

Miyagawa I, et al. Induction of regulatory T cells and its regulation with insulin-like growth factor/insulin-like growth factor binding protein-4 by human mesenchymal stem cells. J Immunol. 2017;199(5):1616–25.

Lee H-J, et al. ICOSL expression in human bone marrow-derived mesenchymal stem cells promotes induction of regulatory T cells. Sci Rep. 2017;7(1):1–15.

CAS   Google Scholar  

Heo JS, Choi Y, Kim HO. Adipose-derived mesenchymal stem cells promote M2 macrophage phenotype through exosomes. Stem Cells Int. 2019;2019:7921760.

Morrison TJ, et al. Mesenchymal stromal cells modulate macrophages in clinically relevant lung injury models by extracellular vesicle mitochondrial transfer. Am J Respir Crit Care Med. 2017;196(10):1275–86.

Melief SM, et al. Multipotent stromal cells induce human regulatory T cells through a novel pathway involving skewing of monocytes toward anti-inflammatory macrophages. Stem Cells. 2013;31(9):1980–91.

Aggarwal S, Pittenger MF. Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood. 2005;105(4):1815–22.

Beyth S, et al. Human mesenchymal stem cells alter antigen-presenting cell maturation and induce T-cell unresponsiveness. Blood. 2005;105(5):2214–9.

Corcione A, et al. Human mesenchymal stem cells modulate B-cell functions. Blood. 2006;107(1):367–72.

Glennie S, et al. Bone marrow mesenchymal stem cells induce division arrest anergy of activated T cells. Blood. 2005;105(7):2821–7.

Naji A, et al. Biological functions of mesenchymal stem cells and clinical implications. Cell Mol Life Sci. 2019;76(17):3323–48.

Pittenger MF, et al. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284(5411):143–7.

Pelttari K, Steck E, Richter W. The use of mesenchymal stem cells for chondrogenesis. Injury. 2008;39(Suppl 1):S58-65.

Pourakbari R, et al. Identification of genes and miRNAs associated with angiogenesis, metastasis, and apoptosis in colorectal cancer. Gene Reports. 2020;18: 100552.

Tuli R, et al. Transforming growth factor-beta-mediated chondrogenesis of human mesenchymal progenitor cells involves N-cadherin and mitogen-activated protein kinase and Wnt signaling cross-talk. J Biol Chem. 2003;278(42):41227–36.

Longobardi L, et al. Effect of IGF-I in the chondrogenesis of bone marrow mesenchymal stem cells in the presence or absence of TGF-beta signaling. J Bone Miner Res. 2006;21(4):626–36.

Chen Q, et al. Fate decision of mesenchymal stem cells: adipocytes or osteoblasts? Cell Death Differ. 2016;23(7):1128–39.

Friedman MS, Long MW, Hankenson KD. Osteogenic differentiation of human mesenchymal stem cells is regulated by bone morphogenetic protein-6. J Cell Biochem. 2006;98(3):538–54.

Eom YW, Shim KY, Baik SK. Mesenchymal stem cell therapy for liver fibrosis. Korean J Intern Med. 2015;30(5):580–9.

Quintanilha LF, et al. Canine mesenchymal stem cells show antioxidant properties against thioacetamide-induced liver injury in vitro and in vivo. Hepatol Res. 2014;44(10):E206–17.

Tang JM, et al. VEGF/SDF-1 promotes cardiac stem cell mobilization and myocardial repair in the infarcted heart. Cardiovasc Res. 2011;91(3):402–11.

Troncoso R, et al. New insights into IGF-1 signaling in the heart. Trends Endocrinol Metab. 2014;25(3):128–37.

Syková E, et al. Transplantation of Mesenchymal stromal cells in patients with amyotrophic lateral sclerosis: results of phase I/IIa clinical trial. Cell Transplant. 2017;26(4):647–58.

Mazzini L, et al. Mesenchymal stem cell transplantation in amyotrophic lateral sclerosis: A phase I clinical trial. Exp Neurol. 2010;223(1):229–37.

Siwek T, et al. Repeat Administration of bone marrow-derived mesenchymal stem cells for treatment of amyotrophic lateral sclerosis. Med Sci Monit. 2020;26: e927484.

Petrou P, et al. Safety and clinical effects of mesenchymal stem cells secreting neurotrophic factor transplantation in patients with amyotrophic lateral sclerosis: results of phase 1/2 and 2a clinical trials. JAMA Neurol. 2016;73(3):337–44.

Oh KW, et al. Phase I trial of repeated intrathecal autologous bone marrow-derived mesenchymal stromal cells in amyotrophic lateral sclerosis. Stem Cells Transl Med. 2015;4(6):590–7.

Staff NP, et al. Safety of intrathecal autologous adipose-derived mesenchymal stromal cells in patients with ALS. Neurology. 2016;87(21):2230–4.

Barczewska M, et al. Umbilical cord mesenchymal stem cells in amyotrophic lateral sclerosis: an original study. Stem Cell Rev Rep. 2020;16(5):922–32.

Giordano R, et al. Autologous mesenchymal stem cell therapy for progressive supranuclear palsy: translation into a phase I controlled, randomized clinical study. J Transl Med. 2014;12:14.

Canesi M, et al. Finding a new therapeutic approach for no-option Parkinsonisms: mesenchymal stromal cells for progressive supranuclear palsy. J Transl Med. 2016;14(1):127.

Venkataramana NK, et al. Open-labeled study of unilateral autologous bone-marrow-derived mesenchymal stem cell transplantation in Parkinson’s disease. Transl Res. 2010;155(2):62–70.

Zamani H, et al. Safety and feasibility of autologous olfactory ensheathing cell and bone marrow mesenchymal stem cell co-transplantation in chronic human spinal cord injury: a clinical trial. Spinal Cord. 2022;60(1):63–70.

Honmou O, et al. Intravenous infusion of auto serum-expanded autologous mesenchymal stem cells in spinal cord injury patients: 13 case series. Clin Neurol Neurosurg. 2021;203: 106565.

Satti HS, et al. Autologous mesenchymal stromal cell transplantation for spinal cord injury: a phase I pilot study. Cytotherapy. 2016;18(4):518–22.

Mendonça MV, et al. Safety and neurological assessments after autologous transplantation of bone marrow mesenchymal stem cells in subjects with chronic spinal cord injury. Stem Cell Res Ther. 2014;5(6):126.

Vaquero J, et al. Repeated subarachnoid administrations of autologous mesenchymal stromal cells supported in autologous plasma improve quality of life in patients suffering incomplete spinal cord injury. Cytotherapy. 2017;19(3):349–59.

Vaquero J, et al. Intrathecal administration of autologous mesenchymal stromal cells for spinal cord injury: safety and efficacy of the 100/3 guideline. Cytotherapy. 2018;20(6):806–19.

Hur JW, et al. Intrathecal transplantation of autologous adipose-derived mesenchymal stem cells for treating spinal cord injury: a human trial. J Spinal Cord Med. 2016;39(6):655–64.

Albu S, et al. Clinical effects of intrathecal administration of expanded Wharton jelly mesenchymal stromal cells in patients with chronic complete spinal cord injury: a randomized controlled study. Cytotherapy. 2021;23(2):146–56.

Jaillard A, et al. Autologous mesenchymal stem cells improve motor recovery in subacute ischemic stroke: a randomized clinical trial. Transl Stroke Res. 2020;11(5):910–23.

Levy ML, et al. Phase I/II study of safety and preliminary efficacy of intravenous allogeneic mesenchymal stem cells in chronic stroke. Stroke. 2019;50(10):2835–41.

Shichinohe H, et al. Research on advanced intervention using novel bone marrOW stem cell (RAINBOW): a study protocol for a phase I, open-label, uncontrolled, dose-response trial of autologous bone marrow stromal cell transplantation in patients with acute ischemic stroke. BMC Neurol. 2017;17(1):179.

Law ZK, et al. The effects of intravenous infusion of autologous mesenchymal stromal cells in patients with subacute middle cerebral artery infarct: a phase 2 randomized controlled trial on safety, tolerability and efficacy. Cytotherapy. 2021;23(9):833–40.

Wang L, et al. Pilot study of umbilical cord-derived mesenchymal stem cell transfusion in patients with primary biliary cirrhosis. J Gastroenterol Hepatol. 2013;28(Suppl 1):85–92.

Zhang YC, et al. Therapeutic potentials of umbilical cord-derived mesenchymal stromal cells for ischemic-type biliary lesions following liver transplantation. Cytotherapy. 2017;19(2):194–9.

Schacher FC, et al. Bone marrow mesenchymal stem cells in acute-on-chronic liver failure grades 2 and 3: a phase I-II randomized clinical trial. Can J Gastroenterol Hepatol. 2021;2021:3662776.

Lin BL, et al. Allogeneic bone marrow-derived mesenchymal stromal cells for hepatitis B virus-related acute-on-chronic liver failure: A randomized controlled trial. Hepatology. 2017;66(1):209–19.

Lanthier N, et al. Autologous bone marrow-derived cell transplantation in decompensated alcoholic liver disease: what is the impact on liver histology and gene expression patterns? Stem Cell Res Ther. 2017;8(1):88.

Suk KT, et al. Transplantation with autologous bone marrow-derived mesenchymal stem cells for alcoholic cirrhosis: Phase 2 trial. Hepatology. 2016;64(6):2185–97.

Abumoawad A, et al. In a Phase 1a escalating clinical trial, autologous mesenchymal stem cell infusion for renovascular disease increases blood flow and the glomerular filtration rate while reducing inflammatory biomarkers and blood pressure. Kidney Int. 2020;97(4):793–804.

Makhlough A, et al. Safety and tolerability of autologous bone marrow mesenchymal stromal cells in ADPKD patients. Stem Cell Res Ther. 2017;8(1):116.

Makhlough A, et al. Bone marrow-mesenchymal stromal cell infusion in patients with chronic kidney disease: A safety study with 18 months of follow-up. Cytotherapy. 2018;20(5):660–9.

Swaminathan M, et al. Pharmacological effects of ex vivo mesenchymal stem cell immunotherapy in patients with acute kidney injury and underlying systemic inflammation. Stem Cells Transl Med. 2021;10(12):1588–601.

Mathiasen AB, et al. Bone marrow-derived mesenchymal stromal cell treatment in patients with ischaemic heart failure: final 4-year follow-up of the MSC-HF trial. Eur J Heart Fail. 2020;22(5):884–92.

Chan JL, et al. Intramyocardial bone marrow stem cells in patients undergoing cardiac surgical revascularization. Ann Thorac Surg. 2020;109(4):1142–9.

Bolli R, et al. A Phase II study of autologous mesenchymal stromal cells and c-kit positive cardiac cells, alone or in combination, in patients with ischaemic heart failure: the CCTRN CONCERT-HF trial. Eur J Heart Fail. 2021;23(4):661–74.

de Celis-Ruiz E, et al. Final results of allogeneic adipose tissue-derived mesenchymal stem cells in acute ischemic stroke (AMASCIS): a phase II, randomized, double-blind, placebo-controlled, single-center, pilot clinical trial. Cell Transplant. 2022;31:9636897221083864.

PubMed   Google Scholar  

Gao LR, et al. Intracoronary infusion of Wharton’s jelly-derived mesenchymal stem cells in acute myocardial infarction: double-blind, randomized controlled trial. BMC Med. 2015;13:162.

Xu JY, et al. Transplantation efficacy of autologous bone marrow mesenchymal stem cells combined with atorvastatin for acute myocardial infarction (TEAM-AMI): rationale and design of a randomized, double-blind, placebo-controlled, multi-center, Phase II TEAM-AMI trial. Regen Med. 2019;14(12):1077–87.

Bartolucci J, et al. Safety and efficacy of the intravenous infusion of umbilical cord mesenchymal stem cells in patients with heart failure: a phase 1/2 randomized controlled trial (RIMECARD Trial [Randomized Clinical Trial of Intravenous Infusion Umbilical Cord Mesenchymal Stem Cells on Cardiopathy]). Circ Res. 2017;121(10):1192–204.

Yagyu T, et al. Long-term results of intracardiac mesenchymal stem cell transplantation in patients with cardiomyopathy. Circ J. 2019;83(7):1590–9.

Florea V, et al. The impact of patient sex on the response to intramyocardial mesenchymal stem cell administration in patients with non-ischaemic dilated cardiomyopathy. Cardiovasc Res. 2020;116(13):2131–41.

Hare JM, et al. Randomized comparison of allogeneic versus autologous mesenchymal stem cells for nonischemic dilated cardiomyopathy: POSEIDON-DCM trial. J Am Coll Cardiol. 2017;69(5):526–37.

Kaushal S, et al. Study design and rationale for ELPIS: A phase I/IIb randomized pilot study of allogeneic human mesenchymal stem cell injection in patients with hypoplastic left heart syndrome. Am Heart J. 2017;192:48–56.

Xiao W, et al. A randomized comparative study on the efficacy of intracoronary infusion of autologous bone marrow mononuclear cells and mesenchymal stem cells in patients with dilated cardiomyopathy. Int Heart J. 2017;58(2):238–44.

Qayyum AA, et al. Autologous adipose-derived stromal cell treatment for patients with refractory angina (MyStromalCell Trial): 3-years follow-up results. J Transl Med. 2019;17(1):360.

Chahal J, et al. Bone marrow mesenchymal stromal cell treatment in patients with osteoarthritis results in overall improvement in pain and symptoms and reduces synovial inflammation. Stem Cells Transl Med. 2019;8(8):746–57.

Hernigou P, et al. Human bone marrow mesenchymal stem cell injection in subchondral lesions of knee osteoarthritis: a prospective randomized study versus contralateral arthroplasty at a mean fifteen year follow-up. Int Orthop. 2021;45(2):365–73.

Hernigou P, et al. Subchondral bone or intra-articular injection of bone marrow concentrate mesenchymal stem cells in bilateral knee osteoarthritis: what better postpone knee arthroplasty at fifteen years? A randomized study. Int Orthop. 2021;45(2):391–9.

Lamo-Espinosa JM, et al. Phase II multicenter randomized controlled clinical trial on the efficacy of intra-articular injection of autologous bone marrow mesenchymal stem cells with platelet rich plasma for the treatment of knee osteoarthritis. J Transl Med. 2020;18(1):356.

Lamo-Espinosa JM, et al. Intra-articular injection of two different doses of autologous bone marrow mesenchymal stem cells versus hyaluronic acid in the treatment of knee osteoarthritis: long-term follow up of a multicenter randomized controlled clinical trial (phase I/II). J Transl Med. 2018;16(1):213.

Bastos R, et al. Intra-articular injections of expanded mesenchymal stem cells with and without addition of platelet-rich plasma are safe and effective for knee osteoarthritis. Knee Surg Sports Traumatol Arthrosc. 2018;26(11):3342–50.

Al-Najar M, et al. Intra-articular injection of expanded autologous bone marrow mesenchymal cells in moderate and severe knee osteoarthritis is safe: a phase I/II study. J Orthop Surg Res. 2017;12(1):190.

Pers YM, et al. Adipose mesenchymal stromal cell-based therapy for severe osteoarthritis of the knee: a phase I dose-escalation trial. Stem Cells Transl Med. 2016;5(7):847–56.

Freitag J, et al. Adipose-derived mesenchymal stem cell therapy in the treatment of knee osteoarthritis: a randomized controlled trial. Regen Med. 2019;14(3):213–30.

Lee WS, et al. Intra-articular injection of autologous adipose tissue-derived mesenchymal stem cells for the treatment of knee osteoarthritis: a phase IIb, randomized, placebo-controlled clinical trial. Stem Cells Transl Med. 2019;8(6):504–11.

Matas J, et al. Umbilical cord-derived mesenchymal stromal cells (MSCs) for knee osteoarthritis: repeated MSC dosing is superior to a single MSC dose and to hyaluronic acid in a controlled randomized phase I/II trial. Stem Cells Transl Med. 2019;8(3):215–24.

Dilogo IH, et al. Umbilical cord-derived mesenchymal stem cells for treating osteoarthritis of the knee: a single-arm, open-label study. Eur J Orthop Surg Traumatol. 2020;30(5):799–807.

Jayankura M, et al. Percutaneous administration of allogeneic bone-forming cells for the treatment of delayed unions of fractures: a pilot study. Stem Cell Res Ther. 2021;12(1):363.

Shim J, et al. Safety and efficacy of Wharton’s jelly-derived mesenchymal stem cells with teriparatide for osteoporotic vertebral fractures: a phase I/IIa study. Stem Cells Transl Med. 2021;10(4):554–67.

Talaat WM, et al. Autologous bone marrow concentrates and concentrated growth factors accelerate bone regeneration after enucleation of mandibular pathologic lesions. J Craniofac Surg. 2018;29(4):992–7.

Falanga V, et al. Autologous bone marrow-derived cultured mesenchymal stem cells delivered in a fibrin spray accelerate healing in murine and human cutaneous wounds. Tissue Eng. 2007;13(6):1299–312.

Zhou L, et al. Efficacy of human adipose derived mesenchymal stem cells in promoting skin wound healing. J Healthc Eng. 2022;2022:6590025.

Moon KC, et al. Potential of allogeneic adipose-derived stem cell-hydrogel complex for treating diabetic foot ulcers. Diabetes. 2019;68(4):837–46.

Qin HL, et al. Clinical evaluation of human umbilical cord mesenchymal stem cell transplantation after angioplasty for diabetic foot. Exp Clin Endocrinol Diabetes. 2016;124(8):497–503.

Lonardi R, et al. Autologous micro-fragmented adipose tissue for the treatment of diabetic foot minor amputations: a randomized controlled single-center clinical trial (MiFrAADiF). Stem Cell Res Ther. 2019;10(1):223.

Huang J, et al. Intrauterine infusion of clinically graded human umbilical cord-derived mesenchymal stem cells for the treatment of poor healing after uterine injury: a phase I clinical trial. Stem Cell Res Ther. 2022;13(1):85.

Hertegård S, et al. Treatment of vocal fold scarring with autologous bone marrow-derived human mesenchymal stromal cells-first phase I/II human clinical study. Stem Cell Res Ther. 2020;11(1):128.

Hardiman O, et al. Amyotrophic lateral sclerosis. Nat Rev Dis Primers. 2017;3:17071.

van Es MA, et al. Amyotrophic lateral sclerosis. Lancet. 2017;390(10107):2084–98.

Eli I, Lerner DP, Ghogawala Z. Acute traumatic spinal cord injury. Neurol Clin. 2021;39(2):471–88.

McDonald JW, Sadowsky C. Spinal-cord injury. Lancet. 2002;359(9304):417–25.

Pajares M, et al. Inflammation in Parkinson’s disease: mechanisms and therapeutic implications. Cells. 2020;9(7):1687.

Article   CAS   PubMed Central   Google Scholar  

Kalia LV, Lang AE. Parkinson’s disease. Lancet. 2015;386(9996):896–912.

Hu C, et al. Mesenchymal stem cell-based cell-free strategies: safe and effective treatments for liver injury. Stem Cell Res Ther. 2020;11(1):377.

Baer PC, Koch B, Geiger H. Kidney Inflammation, Injury and Regeneration. Int J Mol Sci. 2020;21(3):1164.

Article   PubMed Central   Google Scholar  

Fleig SV, Humphreys BD. Rationale of mesenchymal stem cell therapy in kidney injury. Nephron Clin Pract. 2014;127(1–4):75–80.

Virani SS, et al. Heart disease and stroke statistics—2021 update. Circulation. 2021;143(8):e254–743.

Chien KR, et al. Regenerating the field of cardiovascular cell therapy. Nat Biotechnol. 2019;37(3):232–7.

Murry CE, MacLellan WR. Stem cells and the heart-the road ahead. Science. 2020;367(6480):854–5.

Oryan A, Alidadi S. Reconstruction of radial bone defect in rat by calcium silicate biomaterials. Life Sci. 2018;201:45–53.

Qaseem A, et al. Treatment of low bone density or osteoporosis to prevent fractures in men and women: a clinical practice guideline update from the American College of Physicians. Ann Intern Med. 2017;166(11):818–39.

Čamernik K, et al. Comprehensive analysis of skeletal muscle- and bone-derived mesenchymal stem/stromal cells in patients with osteoarthritis and femoral neck fracture. Stem Cell Res Ther. 2020;11(1):146.

Raghoebar GM, et al. Resorbable screws for fixation of autologous bone grafts. Clin Oral Implants Res. 2006;17(3):288–93.

Felice P, et al. Inlay versus onlay iliac bone grafting in atrophic posterior mandible: a prospective controlled clinical trial for the comparison of two techniques. Clin Implant Dent Relat Res. 2009;11(Suppl 1):e69-82.

Swan MC, Goodacre TE. Morbidity at the iliac crest donor site following bone grafting of the cleft alveolus. Br J Oral Maxillofac Surg. 2006;44(2):129–33.

Amini AR, Laurencin CT, Nukavarapu SP. Bone tissue engineering: recent advances and challenges. Crit Rev Biomed Eng. 2012;40(5):363–408.

Fu J, et al. Systemic therapy of MSCs in bone regeneration: a systematic review and meta-analysis. Stem Cell Res Ther. 2021;12(1):377.

Gjerde C, et al. Cell therapy induced regeneration of severely atrophied mandibular bone in a clinical trial. Stem Cell Res Ther. 2018;9(1):213.

Hsu YC, Li L, Fuchs E. Emerging interactions between skin stem cells and their niches. Nat Med. 2014;20(8):847–56.

Hu MS, et al. Mesenchymal Stromal Cells and Cutaneous Wound Healing: A Comprehensive Review of the Background, Role, and Therapeutic Potential. Stem Cells Int. 2018;2018:6901983.

Marfia G, et al. Mesenchymal stem cells: potential for therapy and treatment of chronic non-healing skin wounds. Organogenesis. 2015;11(4):183–206.

Shi R, et al. Localization of human adipose-derived stem cells and their effect in repair of diabetic foot ulcers in rats. Stem Cell Res Ther. 2016;7(1):155.

Download references

Acknowledgements

The authors express their gratitude to the Deanship of Scientific Research at King Khalid University for funding this work through the Research Group Program under grant number RGP. 2/122/43.

Author information

Authors and affiliations.

Department of Anatomy, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia

Ria Margiana

Master’s Programme Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia

Dr. Soetomo General Academic Hospital, Surabaya, Indonesia

Tyumen State Medical University, Tyumen, Russian Federation

Alexander Markov

Tyumen Industrial University, Tyumen, Russian Federation

Department of Prosthetic Dentistry, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia

Angelina O. Zekiy

Medical Technical College, Al-Farahidi University, Baghdad, Iraq

Mohammed Ubaid Hamza

Department of Dentistry, Al-Hadba University College, Mosul, Iraq

Khalid A. Al-Dabbagh

Anesthesia Techniques Department, Al-Mustaqbal University College, Babylon, Iraq

Sura Hasan Al-Zubaidi

Anesthesia Techniques, Al–Nisour University College, Baghdad, Iraq

Noora M. Hameed

Department of Medical Rehabilitation Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia

Irshad Ahmad

Department of Mathematics, Dwaraka Doss Goverdhan Doss Vaishnav College, Arumbakkam, University of Madras, Chennai, India

R. Sivaraman

Veterinary Medicine College, Al-Qasim Green University, Al-Qasim, Iraq

Hamzah H. Kzar

College of Medicine, University of Al-Ameed, Karbala, Iraq

Moaed E. Al-Gazally

Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, 41001, Iraq

Yasser Fakri Mustafa

Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran

Homayoon Siahmansouri

You can also search for this author in PubMed   Google Scholar

Contributions

RM, AM, AOZ, and MUH performed and wrote the manuscript; KHA, SHA, and NMH collected the references and designed the table and figures; IA, HHA, HHK, and RS modified the manuscript; and MEA, YFM, and HS designed the manuscript and approved the final manuscript for publication. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Homayoon Siahmansouri .

Ethics declarations

Ethics approval and consent to participate, consent for publication, competing interests.

The authors indicated no potential conflicts of interest.

Additional information

Publisher's note.

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

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/ . The Creative Commons Public Domain Dedication waiver ( http://creativecommons.org/publicdomain/zero/1.0/ ) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Cite this article.

Margiana, R., Markov, A., Zekiy, A.O. et al. Clinical application of mesenchymal stem cell in regenerative medicine: a narrative review. Stem Cell Res Ther 13 , 366 (2022). https://doi.org/10.1186/s13287-022-03054-0

Download citation

Received : 10 June 2022

Accepted : 18 July 2022

Published : 28 July 2022

DOI : https://doi.org/10.1186/s13287-022-03054-0

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

  • Mesenchymal stromal cell
  • Regenerative medicine
  • Stem cell therapy
  • Tissue regeneration
  • Cell therapy

Stem Cell Research & Therapy

ISSN: 1757-6512

  • Submission enquiries: Access here and click Contact Us
  • General enquiries: [email protected]

research article on stem cell therapy

Will Stem Cells Help You Live Longer?

By Jennifer Chesak

Medically Reviewed by Natalie Kunsman, M.D.

Share this article

  • facebook icon
  • twitter icon
  • linkedin icon

On their never-ending quest for the proverbial fountain of youth, biohackers and celebrities alike have latched onto stem cell anti-aging therapies. Do these treatments, reportedly used by longevity influencer Bryan Johnson and actors John Cleese and William Shatner, have any merit? Can they really turn back the hands of time and help you look and feel more youthful?

A harsh, but also beautiful fact of life is that we age. We say goodbye to the emotional angst of our teens and 20s, only for our joints begin to express their angst via aches, pains, and creaks.

Ultimately, aging happens because of various inflammatory processes that fall under the umbrella term inflammaging . Inflammaging affects our stem cells, the body’s innate repair system. Stem cell therapy, although still controversial as the research is in its infancy, may hold promise for reducing the impact of inflammaging, thereby boosting longevity.

“As we age the number of circulating stem cells decreases,” says stem cell scientist Christian Drapeau, M.S.c. By the age of 30, he adds, the decline reaches about 90 percent. “This reduction means that at some point in our 30s, we don’t have enough stem cells to keep up with cellular loss, and that’s when aging begins,” he says.

About the Experts

Christian Drapeau, M.S.c ., is a stem cell scientist and the founder and chief science officer at Stemregen . He is also the bestselling author of Cracking the Stem Cell Code . Drapeau is credited as first proposing and publishing the hypothesis that stem cells are the body’s repair system.

Fouad Ghaly, M.D. , is the founder of the Ghaly Center for Regenerative Medicine in California. He focuses on stem cell research and regenerative medicine therapies in his practice.

What Are Stem Cells, Anyway?

Stem cells are unique cells found in nearly all bodily tissues. They help keep tissue healthy and repair tissue that’s damaged.

Though we still have a long way to go, researchers have made substantial progress in stem cell research in the 21st century. “Historically, stem cells were primarily recognized as precursors to blood cells, including white and red blood cells, as well as platelets.”

But, as Drapeau notes, the early 2000s led to a promising discovery. “Stem cells are remarkable cells with the unique ability to renew and differentiate into any cell type in the body throughout a person’s lifetime,” Drapeau says. Their ability to differentiate means they can convert or morph. This enables them to aid in the repair and regeneration of any bodily organ ( 2 ). No other cells in the body have stem cells’ natural abilities ( 1 ).

“This groundbreaking finding has propelled stem cells into today’s therapeutic limelight and suggested new applications,” Drapeau says.

How stem cells function

Stem cells have two important functions: to make more stem cells and to convert into other types of cells that do different jobs ( 3 ).

Let’s say you seriously tweak your ankle on a trail run. The injured tissue sends signals to your bone marrow to release stem cells, Drapeau explains. Circulating stem cells will increase 3- to 10-fold ( 4 ). “The stem cells move to the site of damage, multiply, and transform into the necessary cells to repair the tissue,” he adds.

Stem cells also replace cells we naturally lose every day in all our organs and tissues ( 5 ). “To stay healthy, we need to constantly replace these lost cells, and that’s the role of stem cells,” Drapeau says. 

Current application for stem cell therapies

You’ve likely heard of stem cell therapies to treat diseases. Stem cell therapy is currently approved by the U.S. Food and Drug Administration (FDA) for treating certain cancers and disorders that affect the blood and immune system.

Stem cell treatments have different processes, depending on the condition. Blood cancer treatment, for example, may involve a stem cell transplant. In these cases, healthy stem cells are harvested from either the blood or bone marrow of a donor or the patient, and then transplanted into the patient ( 6 ).

Can Stem Cells Reverse Aging?

The number of circulating stem cells available to help us repair and renew tissue is crucial for our health and vitality ( 7 ). And those stem cells decline with age. Where things get tricky with this type of treatment is who gets to have access to them, as they are extremely expensive. 

“Currently, stem cell treatments for anti-aging are mostly accessible to the wealthy,” Drapeau says. However, endogenous stem cell mobilization may be more accessible. “This process involves stimulating the release of one’s own stem cells from the bone marrow, rather than relying on injections to increase circulating stem cells,” he says.

How a stem cell transplant for anti-aging works

Stem cell transplants are not currently available for anti-aging, at least not in the sense that such transplants are available in the United States to treat certain immune system conditions, blood disorders, or cancers.

However, some clinics in the country do offer therapies that harness stem cells from one’s own body. Regenerative medicine specialist Fouad Ghaly, M.D., explains one of the regenerative therapy options he offers at his clinic in California. 

“I can take fat from a man or a woman, isolate the stem cells, activate the stem cells, and inject them into the body,” he says. The stem cells go to the damaged areas and create “a field of regeneration,” he adds. (Critics of stem cell research say that stem cells may migrate to areas on the body that don’t need them.)

But not many practitioners in the U.S. offer the treatment. When you read about biohackers and celebrities getting stem cell therapies, they probably traveled elsewhere to get it. Bryan Johnson, for example, said in a post on X that he participated in a clinical trial in the Bahamas in which bone marrow-derived stem cells were injected into his knees, shoulders, and hips.

Live Longer

research article on stem cell therapy

Age-Related Issues Stem Cell Therapy May Help

Over time, inflammaging and other factors can lead to age-related diseases including type 2 diabetes, cardiovascular disease, neurodegenerative diseases, cancer, musculoskeletal disorders, sexual dysfunction, and more ( 8 ).

Age-related diseases are linked to a reduction in stem cells. “People with conditions affecting the heart, liver, kidneys, pancreas, lungs, and cardiovascular system, as well as those with high blood pressure or erectile dysfunction typically have less than 50 percent of the circulating stem cells found in healthy individuals of the same age,” Drapeau says. 

Not everyone develops these diseases as they age. The conditions are simply more prevalent in older populations. Lifestyle, environmental, and genetic factors all play a role in disease development ( 9 ).

“Harnessing the power of endogenous stem cell mobilization—releasing your own stem cells—or utilizing stem cell treatments can significantly impact age-related health issues,” Drapeau says.

Why Stem Cell Treatments Are Controversial

Over the past five years or so, the FDA, the Centers for Disease Control and Prevention (CDC), and the National Institutes of Health (NIH), have repeatedly warned the public about clinics and companies that market stem cell products or therapies ( 10 , 11 , 12 ). 

Stem cell products and treatments are not currently approved to treat any orthopedic conditions, neurological disorders, cardiopulmonary disorders, chronic pain, fatigue, macular degeneration, or autism, according to the FDA ( 10 ).

The federal agency also warns of severe health issues that have come about from unapproved therapies. “FDA has received reports of blindness, tumor formation, infections, and more … due to the use of these unapproved products,” the agency states ( 10 ). According to a 2017 report, patients who received stem cell eye injections at a Florida clinic became blind shortly after treatment ( 13 ).

And while the CDC and NIH both remain hopeful about stem cell therapy’s potential to treat various medical conditions and diseases, further research is needed to rule out acute and long-term health complications ( 11 , 12 ).

Where Stem Cell Research Is Heading

Research around stem cell therapy’s capabilities have, in many ways, only just begun ( 14 ). However, Drapeau and Ghaly say the evidence is growing around how stem cell therapies can be used strategically to combat aging and support longevity. 

“It will become clear that supporting stem cell function is the first step in addressing any type of tissue damage or degeneration,” Drapeau says. “As research progresses and clinical evidence accumulates, endogenous stem cell mobilization is likely to become standard practice for treating a wide range of conditions, from cardiovascular diseases to musculoskeletal injuries.”

But more than that, stem cells could potentially be used for organ replacement. “Can we induce a regular cell to become a liver? Yes,” Ghaly says. “Can we induce it to become a kidney? Yes. So now there will come a day when they can manufacture organs.”

Researchers have already regenerated fully functional urinary bladder tissue in a nonhuman primate by using stem cells from the animal’s bone marrow ( 15 ). The research serves as a preclinical model for humans.

Ultimately, stem cell research may shape how we treat and prevent age-related diseases, how we repair damaged tissue and organs, and how we recover from illness and injury. Such changes could help us theoretically feel and look better, despite our chronological age .

By using regenerative therapies, such as those related to stem cells, we may even be able to extend our lifespan, which is how long we live, and our healthspan, which is how long we generally feel healthy and have good quality of life.

Poliwoda S, et al. (2022.) Stem cells: a comprehensive review of origins and emerging clinical roles in medical practice.

Eguizabal C, et al. (2019.) Two decades of embryonic stem cells: a historical overview.

Tian Z, et al. (2023. ) Introduction to stem cells.

Wosczyna, et al. (2018.) A muscle stem cell support group: coordinated cellular responses in muscle regeneration.

Zakrzewski, et al. (2019.) Stem cells: past, present, and future.

Bair SM, et al. (2020.) Hematopoietic stem cell transplantation for blood cancers in the era of precision medicine and immunotherapy.

Ahmed A, et al. (2017.) Effect of aging on stem cells.

Li Z, et al. (2021.) Aging and age‐related diseases: from mechanisms to therapeutic strategies.

Farhud D, et al. (2022.) Hypothetical strategies of gene and environmental influence on life expectancy: A brief review.

U.S. Food & Drug Administration (FDA). (2020.) Consumer Alert on Regenerative Medicine Products Including Stem Cells and Exosomes.

Centers for Disease Control and Prevention (CDC). (2019.) Stem Cell and Exosome Products.

National Institutes of Health (NIH). (2022.) Putting Stem Cell-Based Therapies in Context.

Ajay E Kuriyan, et al. (2017.) Vision Loss after Intravitreal Injection of Autologous “Stem Cells” for AMD.

Duc M. Hoang, et al. (2022.) Stem cell-based therapy for human diseases.

Bury M, et al. (2024.) Multipotent bone marrow cell–seeded polymeric composites drive long-term, definitive urinary bladder tissue regeneration.

About the author

Jennifer Chesak is a freelance science and medical journalist and editor who covers the pandemic, chronic health issues, medical rights, healthcare, and the scientific evidence around health and wellness trends. Her work has appeared in Washington Post, Prevention, Healthline, Health, The Daily Beast, Runner's World, Greatist, Real Simple, and more.

Related Articles

Dec 28, 2023

NAD+ vs. NADH—What’s the Difference?

The two molecules facilitate important metabolic functions in the body—and give you energy.

Jan 03, 2024

David Sinclair Got a $2,500 “Preventative” MRI. Here’s What It Revealed

By Rebekah Harding

For starters, the 53-year-old longevity expert’s prostate is in 20-something shape.

Dec 22, 2023

Are Genetic Methylation Tests Actually Worth It?

This assessment may be able to tell you a lot about yourself, from your potential risk of chronic disease to your estimated biological age.

Nov 22, 2023

Does Dairy Cause Inflammation? Your Final, Expert-Backed Answer

Your favorite ice cream could irritate your gut—but not for the reason you expect.

Apr 02, 2024

Is Functional Medicine Legit?

The alternative approach promises to fill gaps missed by mainstream medicine. But does it deliver?

Nov 24, 2022

12 Interesting Facts From Limitless With Chris Hemsworth’s “Stress-Proof” Episode

By Sean Evans

Finally, a show that nails healthspan and lifespan science.

Apr 24, 2023

If You Don’t Want to Look Old, You Should Wash Your Face (With an Actual Cleanser)

By Brianna Lapolla

It takes the same amount of time as splashing some water on there.

Apr 17, 2023

Why Peter Attia Is Literally Counting the Days Until He Dies

His use of a life calendar is morbid, but effective

research article on stem cell therapy

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
  • HHS Author Manuscripts

Logo of nihpa

Introduction to Stem Cell Therapy

Jesse k. biehl.

1 Department of Bioengineering, University of Illinois at Chicago

Brenda Russell

2 Department of Physiology and Biophysics and Department of Bioengineering, University of Illinois at Chicago

Stem cells have the ability to differentiate into specific cell types. The two defining characteristics of a stem cell are perpetual self-renewal and the ability to differentiate into a specialized adult cell type. There are two major classes of stem cells: pluripotent that can become any cell in the adult body, and multipotent that are restricted to becoming a more limited population of cells. Cell sources, characteristics, differentiation and therapeutic applications are discussed. Stem cells have great potential in tissue regeneration and repair but much still needs to be learned about their biology, manipulation and safety before their full therapeutic potential can be achieved.

Introduction

Stem cells have the ability to build every tissue in the human body, hence have great potential for future therapeutic uses in tissue regeneration and repair. In order for cells to fall under the definition of “stem cells,” they must display two essential characteristics. First, stem cells must have the ability of unlimited self-renewal to produce progeny exactly the same as the originating cell. This trait is also true of cancer cells that divide in an uncontrolled manner whereas stem cell division is highly regulated. Therefore, it is important to note the additional requirement for stem cells; they must be able to give rise to a specialized cell type that becomes part of the healthy animal. 1

The general designation, “stem cell” encompasses many distinct cell types. Commonly, the modifiers, “embryonic,” and “adult” are used to distinguish stem cells by the developmental stage of the animal from which they come, but these terms are becoming insufficient as new research has discovered how to turn fully differentiated adult cells back into embryonic stem cells and, conversely, adult stem cells, more correctly termed “somatic” stem cells meaning “from the body”, are found in the fetus, placenta, umbilical cord blood and infants. 2 Therefore, this review will sort stem cells into two categories based on their biologic properties - pluripotent stem cells and multipotent stem cells. Their sources, characteristics, differentiation and therapeutic applications are discussed.

Pluripotent stem cells are so named because they have the ability to differentiate into all cell types in the body. In natural development, pluripotent stem cells are only present for a very short period of time in the embryo before differentiating into the more specialized multipotent stem cells that eventually give rise to the specialized tissues of the body ( Figure 1 ). These more limited multipotent stem cells come in several subtypes: some can become only cells of a particular germ line (endoderm, mesoderm, ectoderm) and others, only cells of a particular tissue. In other words, pluripotent cells can eventually become any cell of the body by differentiating into multipotent stem cells that themselves go through a series of divisions into even more restricted specialized cells.

An external file that holds a picture, illustration, etc.
Object name is nihms100185f1.jpg

During natural embryo development, cells undergo proliferation and specialization from the fertilized egg, to the blastocyst, to the gastrula during natural embryo development (left side of panel). Pluripotent, embryonic stem cells are derived from the inner cell mass of the blastoctyst (lightly shaded). Multipotent stem cells (diamond pattern, diagonal lines, and darker shade) are found in the developing gastrula or derived from pluripotent stem cells and are restricted to give rise to only cells of their respective germ layer.

Stem Cell Fates

Based on the two defining characteristics of stem cells (unlimited self-renewal and ability to differentiate), they can be described as having four outcomes or fates 3 ( Figure 2 ). A common fate for multipotent stem cells is to remain quiescent without dividing or differentiating, thus maintaining its place in the stem cell pool. An example of this is stem cells in the bone marrow that await activating signals from the body. A second fate of stem cells is symmetric self-renewal in which two daughter stem cells, exactly like the parent cell, arise from cell division. This does not result in differentiated progeny but does increase the pool of stem cells from which specialized cells can develop in subsequent divisions. The third fate, asymmetric self-renewal, occurs when a stem cell divides into two daughter cells, one a copy of the parent, the other a more specialized cell, named a somatic or progenitor cell. Asymmetric self-renewal results in the generation of differentiated progeny needed for natural tissue development/regeneration while also maintaining the stem cell pool for the future. The fourth fate is that in which a stem cell divides to produce two daughters both different from the parent cell. This results in greater proliferation of differentiated progeny with a net loss in the stem cell pool.

An external file that holds a picture, illustration, etc.
Object name is nihms100185f2.jpg

Four potential outcomes of stem cells. A) Quiescence in which a stem cell does not divide but maintains the stem cell pool. B) Symmetric self-renewal where a stem cell divides into two daughter stem cells increasing the stem cell pool. C) Asymmetric self-renewal in which a stem cell divides into one differentiated daughter cell and one stem cell, maintaining the stem cell pool. D) Symmetric division without self-renewal where there is a loss in the stem cell pool but results in two differentiated daughter cells. (SC- Stem cell, DP-Differentiated progeny)

The factors that determine the fate of stem cells is the focus of intense research. Knowledge of the details could be clinically useful. For example, clinicians and scientists might direct a stem cell population to expand several fold through symmetrical self-renewal before differentiation into multipotent or more specialized progenitor cells. This would ensure a large, homogeneous population of cells at a useful differentiation stage that could be delivered to patients for successful tissue regeneration.

Sources of Stem Cells

Pluripotent.

Pluripotent stem cells being used in research today mainly come from embryos, hence the name, “embryonic stem cells”. Pre-implantation embryos a few days old contain only 10-15% pluripotent cells in the “inner cell mass” ( Figure 1 ). Those pluripotent cells can be isolated, then cultured on a layer of “feeder” cells which provide unknown cues for many rounds of proliferation while sustaining their pluripotency.

Recently, two different groups of scientists induced adult cells back into the pluripotent state by molecular manipulation to yield “induced pluripotent stem cells” (iPS) that share some of the same characteristics as embryonic stem cells such as proliferation, morphology and gene expression (in the form of distinct surface markers and proteins being expressed). 4 - 8 Both groups used retroviruses to carry genes for transcription factors into the adult cells. These genes are transcribed and translated into proteins that regulate the expression of other genes designed to reprogram the adult nucleus back into its embryonic state. Both introduced the embryonic transcription factors known as Sox2 and Oct4. One group also added Klf4 and c-Myc 4 , and the other group added Lin28 and Nanog. 6 Other combinations of factors would probably also work, but, unfortunately, neither the retroviral carrier method nor the use of the oncogenic transcription factor c-Myc are likely to be approved for human therapy. Consequently, a purely chemical approach to deliver genes into the cells, and safer transcription factors are being tried. Results of these experiments look promising. 9

Multipotent

Multipotent stem cells may be a viable option for clinical use. These cells have the plasticity to become all the progenitor cells for a particular germ layer or can be restricted to become only one or two specialized cell types of a particular tissue. The multipotent stem cells with the highest differentiating potential are found in the developing embryo during gastrulation (day 14-15 in humans, day 6.5-7 in mice). These cells give rise to all cells of their particular germ layer, thus, they still have flexibility in their differentiation capacity. They are not pluripotent stem cells because they have lost the ability to become cells of all three germ layers ( Figure 1 ). On the low end of the plasticity spectrum are the unipotent cells that can become only one specialized cell type such as skin stem cells or muscle stem cells. These stem cells are typically found within their organ and although their differentiation capacity is restricted, these limited progenitor cells play a vital role in maintaining tissue integrity by replenishing aging or injured cells. There are many other sub-types of multipotent stem cells occupying a range of differentiation capacities. For example, multipotent cells derived from the mesoderm of the gastrula undergo a differentiation step limiting them to muscle and connective tissue; however, further differentiation results in increased specialization towards only connective tissue and so on until the cells can give rise to only cartilage or only bone.

Multipotent stem cells found in bone marrow are best known, because these have been used therapeutically since the 1960’s 10 (their potential will be discussed in greater detail in a later section). Recent research has found new sources for multipotent stem cells of greater plasticity such as the placenta and umbilical cord blood. 11 Further, the heart, until recently considered void of stem cells, is now known to contain stem cells with the potential to become cardiac myocytes. 12 Similarly, neuro-progenitor cells have been found within the brain. 13

The cardiac stem cells are present in such small numbers, that they are difficult to study and their function has not been fully determined. The second review in this series will discuss their potential in greater detail.

Characteristics that Identify Stem Cells

Since Federal funding for human embryonic stem cells is restricted in the United States, many scientists use the mouse model instead. Besides their ability to self-renew indefinitely and differentiate into cell types of all three germ layers, murine and human pluripotent stem cells have much in common. It should not be surprising that so many pluripotency traits are conserved between species given the shared genomic sequences and intra-cellular structure in mammals. Both mouse and human cells proliferate indefinitely in culture, have a high nucleus to cytoplasm ratio, need the support of growth factors derived from other live cells, and display similar surface antigens, transcription factors and enzymatic activity (i.e. high alkaline phosphatase activity). 14 However, differences between mouse and human pluripotent cells, while subtle, are very important. Although the transcription factors mentioned above to induce pluripotency from adult cells (Oct3/4 and Sox2) are shared, the extracellular signals needed to regulate them differ. Mouse embryonic stem cells need the leukemia inhibitory factor and bone morphogenic proteins while human require the signaling proteins Noggin and Wnt for sustained pluripotency. 15 Surface markers used to identify pluripotent cells also differ slightly between the two species as seen in the variants of the adhesion molecule SSEA (SSEA-1 in mouse, SSEA-3 & 4 in humans). 16 Thus, while pluripotency research in mouse cells is valuable, a direct correlation to the human therapy is not likely.

Last, but certainly not least, a big difference between mouse and human stem cells are the moral and ethical dilemmas that accompany the research. Some people consider working with human embryonic stem cells to be ethically problematic while very few people have reservations on working with the mouse models. However, given the biological differences between human and mouse cells, most scientists believe that data relevant for human therapy will be missed by working only on rodents.

Cell surface markers are typically also used to identify multipotent stem cells. For example, mesenchymal stem cells can be purified from the whole bone marrow aspirate by eliminating cells that express markers of committed cell types, a step referred to as lineage negative enrichment, and then further separating the cells that express the sca-1 and c-Kit surface markers signifying mesenchymal stem cells. Both the lineage negative enrichment step and the sca-1/c-Kit isolation can be achieved by using flow cytometry and is discussed in further detail in the following review. The c-Kit surface marker also is used to distinguish the recently discovered cardiac stem cells from the rest of the myocardium. A great deal of recent work in cardiovascular research has centered on trying to find which markers indicate early multipotent cells that will give rise to pre-cardiac myocytes. Cells with the specific mesodermal marker, Kdr, give rise to the progenitor cells of the cardiovascular system including contracting cardiac myocytes, endothelial cells and vascular smooth muscle cells and are therefore considered to be the earliest cells with specification towards the cardiovascular lineage. 17 Cells at this early stage still proliferate readily and yet are destined to become cells of the cardiovascular system and so may be of great value therapeutically.

Differentiation

Scientists are still struggling to reliably direct differentiation of stem cells into specific cell types. They have used a virtual alphabet soup of incubation factors toward that end (including trying a variety of growth factors, chemicals and complex substrates on which the cells are grown), with, so far, only moderate success. As an example of this complexity, one such approach to achieve differentiation towards cardiac myocytes is to use the chemical activin A and the growth factor BMP-4. When these two factors are administered to pluripotent stem cells in a strictly controlled manner, both in concentration and temporally, increased efficiency is seen in differentiation towards cardiac myocytes, but still, only 30% of cells can be expected to become cardiac. 18

Multipotent cells have also been used as the starting point for cell therapy, again with cocktails of growth factors and/or chemicals to induce differentiation toward a specific, desired lineage. Some recipes are simple, such as the use of retinoic acid to induce mesenchymal stem cells into neuronal cells, 19 or transforming growth factor-β to make bone marrow-derived stem cells express cardiac myocyte markers. 20 Others are complicated or ill-defined such as addition of the unknown factors secreted by cells in culture. Physical as well as chemical cues cause differentiation of stem cells. Simply altering the stiffness of the substrate on which cells are cultured can direct stem cells to neuronal, myogenic or osteogenic lineages. 21 Cells evolve in physical and chemical environments so a combination of both will probably be necessary for optimal differentiation of stem cells. The importance of physical cues in the cell’s environment will be discussed in greater detail in the final review of this series. Ideally, for stem cells to be used therapeutically, efficient, uniform protocols must be established so that cells are a well-controlled and well-defined entity.

Stem Cell Therapy

Pluripotent stem cells.

Pluripotent stem cells have not yet been used therapeutically in humans because many of the early animal studies resulted in the undesirable formation of unusual solid tumors, called teratomas. Teratomas are made of a mix of cell types from all the early germ layers. Later successful animal studies used pluripotent cells modified to a more mature phenotype which limits this proliferative capacity. Cells derived from pluripotent cells have been used to successfully treat animals. For example, animals with diabetes have been treated by the creation of insulin-producing cells responsive to glucose levels. Also, animals with acute spinal cord injury or visual impairment have been treated by creation of new myelinated neurons or retinal epithelial cells, respectively. Commercial companies are currently in negotiations with the FDA regarding the possibility of advancing to human trials. Other animal studies have been conducted to treat several maladies such as Parkinson’s disease, muscular dystrophy and heart failure. 18 , 22 , 23

Scientists hope that stem cell therapy can improve cardiac function by integration of newly formed beating cardiac myocytes into the myocardium to produce greater force. Patches of cardiac myocytes derived from human embryonic stem cells can form viable human myocardium after transplantation into animals, 24 with some showing evidence of electrical integration. 25 , 26 Damaged rodent hearts showed slightly improved cardiac function after injection of cardiac myocytes derived from human embryonic stem cells. 21 The mechanisms for the gain in function are not fully understood but it may be only partially due to direct integration of new beating heart cells. It is more likely due to paracrine effects that benefit other existing heart cells (see next review).

Multipotent stem cells

Multipotent stem cells harvested from bone marrow have been used since the 1960’s to treat leukemia, myeloma and lymphoma. Since cells there give rise to lymphocytes, megakaryocytes and erythrocytes, the value of these cells is easily understood in treating blood cancers. Recently, some progress has been reported in the use of cells derived from bone marrow to treat other diseases. For example, the ability to form whole joints in mouse models 27 has been achieved starting with mesenchymal stem cells that give rise to bone and cartilage. In the near future multipotent stem cells are likely to benefit many other diseases and clinical conditions. Bone marrow-derived stem cells are in clinical trials to remedy heart ailments. This is discussed in detail in the next review of this series.

Pluripotent vs. Multipotent

Pluripotent and multipotent stem cells have their respective advantages and disadvantages. The capacity of pluripotent cells to become any cell type is an obvious therapeutic advantage over their multipotent kin. Theoretically, they could be used to treat diseased or aging tissues in which multipotent stem cells are insufficient. Also, pluripotent stem cells proliferate more rapidly so can yield higher numbers of useful cells. However, use of donor pluripotent stem cells would require immune suppressive drugs for the duration of the graft 28 while use of autologous multipotent stem cells (stem cells from ones’ self) would not. This ability to use one’s own cells is a great advantage of multipotent stem cells. The immune system recognizes specific surface proteins on cells/objects that tell them whether the cell is from the host and is healthy. Autologous, multipotent stem cells have the patient’s specific surface proteins that allow it to be accepted by the host’s immune system and avoid an immunological reaction. Pluripotent stem cells, on the other hand, are not from the host and therefore, lack the proper signals required to stave off rejection from the immune system. Research is ongoing trying to limit the immune response caused by pluripotent cells and is one possible advantage that iPS cells may have.

The promises of cures for human ailments by stem cells have been much touted but many obstacles must still be overcome. First, more human pluripotent and multipotent cell research is needed since stem cell biology differs in mice and men. Second, the common feature of unlimited cell division shared by cancer cells and pluripotent stem cells must be better understood in order to avoid cancer formation. Third, the ability to acquire large numbers of the right cells at the right stage of differentiation must be mastered. Fourth, specific protocols must be developed to enhance production, survival and integration of transplanted cells. Finally, clinical trials must be completed to assure safety and efficacy of the stem cell therapy. When it comes to stem cells, knowing they exist is a long way from using them therapeutically.

Acknowledgments

Supported by NIH (HL 62426 and T32 HL 007692)

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
  • Published: 14 August 2024

Lab & life

Stem cell science starters

  • Vivien Marx 1  

Nature Methods ( 2024 ) Cite this article

130 Accesses

5 Altmetric

Metrics details

  • Induced pluripotent stem cells
  • Neural stem cells
  • Pluripotent stem cells

Early-career scientists shared some of their plans, hopes and dreams about being a principal investigator at the 2024 annual meeting of the International Society for Stem Cell Research.

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

Access options

Access Nature and 54 other Nature Portfolio journals

Get Nature+, our best-value online-access subscription

24,99 € / 30 days

cancel any time

Subscribe to this journal

Receive 12 print issues and online access

251,40 € per year

only 20,95 € per issue

Buy this article

  • Purchase on SpringerLink
  • Instant access to full article PDF

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

Author information

Authors and affiliations.

Nature Methods https://www.nature.com/nmeth/

Vivien Marx

You can also search for this author in PubMed   Google Scholar

Corresponding author

Correspondence to Vivien Marx .

Rights and permissions

Reprints and permissions

About this article

Cite this article.

Marx, V. Stem cell science starters. Nat Methods (2024). https://doi.org/10.1038/s41592-024-02398-0

Download citation

Published : 14 August 2024

DOI : https://doi.org/10.1038/s41592-024-02398-0

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 newsletter — what matters in science, free to your inbox daily.

research article on stem cell therapy

COMMENTS

  1. Current state of stem cell-based therapies: an overview

    Current state of stem cell-based therapies: an overview. Recent research reporting successful translation of stem cell therapies to patients have enriched the hope that such regenerative strategies may one day become a treatment for a wide range of vexing diseases. In fact, the past few years witnessed, a rather exponential advancement in ...

  2. Stem cell-based therapy for human diseases

    Abstract. Recent advancements in stem cell technology open a new door for patients suffering from diseases and disorders that have yet to be treated. Stem cell-based therapy, including human ...

  3. Articles

    Offering a unique focus on translational research into stem cell therapies, Stem Cell Research & Therapy acts as a platform for global debate and discussion. ...

  4. Clarifying Stem-Cell Therapy's Benefits and Risks

    To ensure that the emerging field of stem-cell therapy fulfills its promise to patients, we must first understand its risks and benefits and develop therapeutic approaches based on sound science. T...

  5. Progress and challenges in stem cell biology

    These abilities make stem cells an incredibly promising tool in therapeutics and have turned stem cell biology into a fast-evolving field.

  6. Stem cells: past, present, and future

    In recent years, stem cell therapy has become a very promising and advanced scientific research topic. The development of treatment methods has evoked great expectations. This paper is a review focused on the discovery of different stem cells and the potential therapies based on these cells. The genesis of stem cells is followed by laboratory steps of controlled stem cell culturing and ...

  7. Advances in stem cell research and therapeutic development

    In this Review, the authors survey key advances in stem cell research and describe the cell types that are currently being used in the clinic or are close to clinical trials.

  8. Home page

    An international peer-reviewed journal, it publishes high-quality open access research articles with a special emphasis on basic, translational and clinical research into stem cell therapeutics and regenerative therapies, including animal models and clinical trials. The journal also provides reviews, viewpoints, commentaries, reports and methods.

  9. Stem cells: a comprehensive review of origins and emerging clinical

    Stem cell therapies are at present indicated for a range of clinical conditions beyond traditional origins to treat genetic blood diseases and have seen substantial success. In this regard, emerging use for stem cells is their potential to treat pain states and neurodegenerative diseases such as Parkinson's and Alzheimer's disease.

  10. Stem cell-based therapy for human diseases

    Learn how stem cell-based therapy can treat various human diseases, such as cancer, neurodegeneration, and diabetes, from this PMC article.

  11. Stem Cells in the Treatment of Disease

    The derivation of induced pluripotent stem cells (iPSCs) has revolutionized stem-cell research (see the box for a list of the abbreviations used in this article).

  12. Stem-cell therapies

    Stem-cell therapies articles from across Nature Portfolio Stem cell therapies are a type of cell therapy in which the cells used are either stem cells (as in the case of bone marrow ...

  13. Study finds stem cell therapy is safe and may benefit people with

    May 23, 2024. Mayo Clinic researchers have demonstrated the safety and potential benefit of stem cell regenerative medicine therapy for patients with subacute and chronic spinal cord injury. The results of the phase 1 Clinical Trial of Autologous Adipose-Derived Mesenchymal Stem Cells in the Treatment of Paralysis Due to Traumatic Spinal Cord ...

  14. Stem cells: past, present, and future

    In recent years, stem cell therapy has become a very promising and advanced scientific research topic. The development of treatment methods has evoked great expectations. This paper is a review focused on the discovery of different stem cells and the potential therapies based on these cells. The gen …

  15. Recent Progress of Stem Cell Therapy in Cancer Treatment: Molecular

    The insufficient and unspecific target of traditional therapeutic approaches in cancer treatment often leads to therapy resistance and cancer recurrence. Over the past decades, accumulating discoveries about stem cell biology have provided new potential ...

  16. Association between Mesenchymal Stem Cells and COVID‐19 Therapy

    As per a new declaration of the International Society for Stem Cell Research (ISSCR), at present, there are no endorsed stem cell-based methodologies for the counteraction of coronavirus disease. In any case, as of late, mesenchymal stem cells (MSCs) have presented one of the restorative methodologies for utilizing in the treatment of COVID-19 .

  17. Welcome to Stem Cell Research & Therapy

    Stem Cell Research & Therapy aims to be the major forum for translational research into stem cell therapies. The journal has a special emphasis on basic, translational, and clinical research into stem cell therapeutics, including animal models, and clinical trials.

  18. Next-generation stem cells

    However, the advent of cell-engineering approaches is ushering in a new generation of stem cell-based therapies, greatly expanding their therapeutic utility.

  19. Stem Cell Therapy: From Idea to Clinical Practice

    In addition, regenerative medicine using modified stem cells and combinations of stem cells with scaffolds and chemicals to overcome stem cell therapy challenges and increase the associated efficiency are two important future directions of research.

  20. New way to extend 'shelf life' of blood stem cells will improve gene

    Researchers have discovered a way to extend the shelf life of blood stem cells outside the body for use in gene therapy, providing patients with better options and improving their outcomes.

  21. Clinical application of mesenchymal stem cell in regenerative medicine

    Introduction In the last decade, stem cells are increasingly applied as a therapeutic method for numerous disorders. Stem cell therapy, traditionally applied for hematopoietic disorders, nonetheless, is now established for the treatment of non-hematologic disorders [ 1, 2 ].

  22. Are Stem Cells the Secret to Anti-Aging?

    And while the CDC and NIH both remain hopeful about stem cell therapy's potential to treat various medical conditions and diseases, further research is needed to rule out acute and long-term health complications (11, 12). Where Stem Cell Research Is Heading. Research around stem cell therapy's capabilities have, in many ways, only just ...

  23. Stem-cell research

    Stem-cell research articles from across Nature Portfolio Stem-cell research is the area of research that studies the properties of stem cells and their potential use in medicine.

  24. Stem Cell Therapies for Ischemic Stroke: A Systematic Review

    Many studies have demonstrated the benefits of stem cell therapy in stroke patients. In a meta-analysis by Jeong et al. (2014), stem cell therapy involving bone marrow mononuclear cells, mesenchymal and fetal stem cells in stroke patients was shown to be effective in improving many domains of clinical outcomes [ 20 ].

  25. Cancer stem cell

    De-differentiation of mutated cells may create stem cell-like characteristics, suggesting that any cell might become a cancer stem cell. In other words, fully differentiated cell undergoes mutations or extracellular signals that drive it back to a stem-like state.

  26. Introduction to Stem Cell Therapy

    Introduction to Stem Cell Therapy. Stem cells have the ability to differentiate into specific cell types. The two defining characteristics of a stem cell are perpetual self-renewal and the ability to differentiate into a specialized adult cell type. There are two major classes of stem cells: pluripotent that can become any cell in the adult ...

  27. Stem cell science starters

    Early-career scientists shared some of their plans, hopes and dreams about being a principal investigator at the 2024 annual meeting of the International Society for Stem Cell Research.