Tissue Engineering and Regenerative Medicine: Perspectives and Challenges

Van T. Hoang , Quyen Thi Nguyen , Trang Thi Kieu Phan , Trang H. Pham , Nhung Thi Hong Dinh , Le Phuong Hoang Anh , Lan Thi Mai Dao , Van Dat Bui , Hong-Nhung Dao , Duc Son Le , Anh Thi Lan Ngo , Quang-Duong Le , Liem Nguyen Thanh

MedComm ›› 2025, Vol. 6 ›› Issue (5) : e70192

PDF
MedComm ›› 2025, Vol. 6 ›› Issue (5) : e70192 DOI: 10.1002/mco2.70192
REVIEW

Tissue Engineering and Regenerative Medicine: Perspectives and Challenges

Author information +
History +
PDF

Abstract

From the pioneering days of cell therapy to the achievement of bioprinting organs, tissue engineering, and regenerative medicine have seen tremendous technological advancements, offering solutions for restoring damaged tissues and organs. However, only a few products and technologies have received United States Food and Drug Administration approval. This review highlights significant progress in cell therapy, extracellular vesicle-based therapy, and tissue engineering. Hematopoietic stem cell transplantation is a powerful tool for treating many diseases, especially hematological malignancies. Mesenchymal stem cells have been extensively studied. The discovery of induced pluripotent stem cells has revolutionized disease modeling and regenerative applications, paving the way for personalized medicine. Gene therapy represents an innovative approach to the treatment of genetic disorders. Additionally, extracellular vesicle-based therapies have emerged as rising stars, offering promising solutions in diagnostics, cell-free therapeutics, drug delivery, and targeted therapy. Advances in tissue engineering enable complex tissue constructs, further transforming the field. Despite these advancements, many technical, ethical, and regulatory challenges remain. This review addresses the current bottlenecks, emphasizing novel technologies and interdisciplinary research to overcome these hurdles. Standardizing practices and conducting clinical trials will balance innovation and regulation, improving patient outcomes and quality of life.

Keywords

cell therapy / extracellular vesicle-based therapy / tissue engineering / regenerative medicine / stem cell / bioprinting

Cite this article

Download citation ▾
Van T. Hoang, Quyen Thi Nguyen, Trang Thi Kieu Phan, Trang H. Pham, Nhung Thi Hong Dinh, Le Phuong Hoang Anh, Lan Thi Mai Dao, Van Dat Bui, Hong-Nhung Dao, Duc Son Le, Anh Thi Lan Ngo, Quang-Duong Le, Liem Nguyen Thanh. Tissue Engineering and Regenerative Medicine: Perspectives and Challenges. MedComm, 2025, 6(5): e70192 DOI:10.1002/mco2.70192

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

F. Han, J. Wang, L. Ding, et al., “Tissue Engineering and Regenerative Medicine: Achievements, Future, and Sustainability in Asia,” Frontiers in Bioengineering and Biotechnology 8 (2020): 83.

[2]

I. E. Konstantinov, “In Search of Alexander A. Maximow: The Man Behind the unitarian Theory of Hematopoiesis,” Perspectives in Biology and Medicine 43, no. 2 (2000): 269-276.

[3]

E. D. Thomas, H. L. Lochte, W. C. Lu, and J. W. Ferrebee, “Intravenous Infusion of Bone Marrow in Patients Receiving Radiation and Chemotherapy,” New England Journal of Medicine 257, no. 11 (1957): 491-496.

[4]

A. J. Friedenstein, R. K. Chailakhyan, and U. V. Gerasimov, “Bone Marrow Osteogenic Stem Cells: In Vitro Cultivation and Transplantation in Diffusion Chambers,” Cell and Tissue Kinetics 20, no. 3 (1987): 263-272, https://doi.org/10.1111/j.1365-2184.1987.tb01309.x.

[5]

D. M. Hoang, P. T. Pham, T. Q. Bach, et al., “Stem Cell-based Therapy for human Diseases,” Signal Transduction and Targeted Therapy 7, no. 1 (2022): 1-41.

[6]

K. Takahashi, K. Tanabe, M. Ohnuki, et al., “Induction of Pluripotent Stem Cells From Adult human Fibroblasts by Defined Factors,” Cell 131, no. 5 (2007): 861-872.

[7]

K. Sugai, M. Sumida, T. Shofuda, et al., “First-in-human Clinical Trial of Transplantation of iPSC-derived NS/PCs in Subacute Complete Spinal Cord Injury: Study Protocol,” Regenerative Therapy 18 (2021): 321-333.

[8]

S. Takagi, M. Mandai, K. Gocho, et al., “Evaluation of Transplanted Autologous Induced Pluripotent Stem Cell-Derived Retinal Pigment Epithelium in Exudative Age-Related Macular Degeneration,” Ophthalmology Retina 3, no. 10 (2019): 850-859.

[9]

K. L. McKinley, M. T. Longaker, and S. Naik, “Emerging Frontiers in Regenerative Medicine,” Science 380, no. 6647 (2023): 796-798.

[10]

A. Nagelkerke, M. Ojansivu, L. van der Koog, et al., “Extracellular Vesicles for Tissue Repair and Regeneration: Evidence, Challenges and Opportunities,” Advanced Drug Delivery Reviews 175 (2021): 113775.

[11]

G. van Niel, G. D'Angelo, and G. Raposo, “Shedding Light on the Cell Biology of Extracellular Vesicles,” Nature Reviews Molecular Cell Biology 19, no. 4 (2018): 213-228.

[12]

Y. Couch, E. I. Buzàs, D. D. Vizio, et al., “A Brief History of Nearly EV-erything—The Rise and Rise of Extracellular Vesicles,” Journal of Extracellular Vesicles 10, no. 14 (2021): e12144.

[13]

R. E. Veerman, G. G. Akpinar, M. Eldh, and S. Gabrielsson, “Immune Cell-Derived Extracellular Vesicles—Functions and Therapeutic Applications,” Trends in Molecular Medicine 25, no. 5 (2019): 382-394.

[14]

E. Karnas, P. Dudek, and E. K. Zuba-Surma, “Stem Cell- derived Extracellular Vesicles as New Tools in Regenerative Medicine-Immunomodulatory Role and Future Perspectives,” Frontiers in Immunology 14 (2023): 1120175.

[15]

D. E. Murphy, O. G. de Jong, M. Brouwer, et al., “Extracellular Vesicle-based Therapeutics: Natural versus Engineered Targeting and Trafficking,” Experimental & Molecular Medicine 51 (2019): 32.

[16]

R. C. Lai, F. Arslan, M. M. Lee, et al., “Exosome Secreted by MSC Reduces Myocardial Ischemia/Reperfusion Injury,” Stem Cell Research 4, no. 3 (2010): 214-222.

[17]

M. J. Haney, N. L. Klyachko, Y. L. Zhao, et al., “Exosomes as Drug Delivery Vehicles for Parkinson's Disease Therapy,” Journal of Controlled Release 207 (2015): 18-30.

[18]

Y. You, Y. Tian, Z. G. Yang, et al., “Intradermally Delivered mRNA-encapsulating Extracellular Vesicles for Collagen-replacement Therapy,” Nature Biomedical Engineering 7, no. 7 (2023): 887.

[19]

A. I. Caplan and J. E. Dennis, “Mesenchymal Stem Cells as Trophic Mediators,” Journal of Cellular Biochemistry 98, no. 5 (2006): 1076-1084.

[20]

P. M. P. Lins, E. Pirlet, M. Szymonik, A. Bronckaers, and I. Nelissen, “Manufacture of Extracellular Vesicles Derived From Mesenchymal Stromal Cells,” Trends in Biotechnology 41, no. 7 (2023): 965-981.

[21]

J. F. Burke, I. V. Yannas, W. C. Quinby, C. C. Bondoc, and W. K. Jung, “Successful Use of a Physiologically Acceptable Artificial Skin in the Treatment of Extensive Burn Injury,” Annals of Surgery 194, no. 4 (1981): 413-428.

[22]

R. Langer and J. P. Vacanti, “Tissue Engineering,” Science 260, no. 5110 (1993): 920-926.

[23]

A. Atala, S. B. Bauer, S. Soker, J. J. Yoo, and A. B. Retik, “Tissue-engineered Autologous Bladders for Patients Needing Cystoplasty,” Lancet 367, no. 9518 (2006): 1241-1246.

[24]

M. Kim, Y. J. Kim, Y. S. Kim, et al., “One-Year Results of Ear Reconstruction With 3D Printed Implants,” Yonsei Medical Journal 65, no. 8 (2024): 456-462.

[25]

G. Prindull, B. Prindull, and N. Meulen, “Haematopoietic Stem Cells (CFUc) in human Cord Blood,” Acta Paediatrica Scandinavica 67, no. 4 (1978): 413-416.

[26]

A. I. Caplan, “Mesenchymal Stem Cells,” Journal of Orthopaedic Research 9, no. 5 (1991): 641-650.

[27]

R. L. Rietze and B. A. Reynolds, “Neural Stem Cell Isolation and Characterization,” Methods in Enzymology 419 (2006): 3-23.

[28]

J. A. Thomson, J. Itskovitz-Eldor, S. S. Shapiro, et al., “Embryonic Stem Cell Lines Derived From human Blastocysts,” Science 282, no. 5391 (1998): 1145-1147.

[29]

J. A. Snowden, I. Sanchez-Ortega, S. Corbacioglu, et al., “Indications for Haematopoietic Cell Transplantation for Haematological Diseases, Solid Tumours and Immune Disorders: Current Practice in Europe, 2022,” Bone Marrow Transplantation 57, no. 8 (2022): 1217-1239.

[30]

I. L. Weissman and J. A. Shizuru, “The Origins of the Identification and Isolation of Hematopoietic Stem Cells, and Their Capability to Induce Donor-specific Transplantation Tolerance and Treat Autoimmune Diseases,” Blood 112, no. 9 (2008): 3543-3553.

[31]

S. Pinho and P. S. Frenette, “Haematopoietic Stem Cell Activity and Interactions With the Niche,” Nature Reviews Molecular Cell Biology 20, no. 5 (2019): 303-320.

[32]

N. Granot and R. Storb, “History of Hematopoietic Cell Transplantation: Challenges and Progress,” Haematologica 105, no. 12 (2020): 2716-2729.

[33]

L. Li and P. K. Mandal, “Recent Advancements in Gene Therapy for Sickle Cell Disease and β-Thalassemia,” Frontiers in Hematology 3 (2024).

[34]

X. M. Anguela and K. A. High, “Hemophilia B and Gene Therapy: A New Chapter With Etranacogene dezaparvovec,” Blood Advances 8, no. 7 (2024): 1796-1803.

[35]

D. Jovic, Y. Yu, D. Wang, et al., “A Brief Overview of Global Trends in MSC-Based Cell Therapy,” Stem Cell Reviews and Reports 18, no. 5 (2022): 1525-1545.

[36]

O. Levy, R. Kuai, E. M. J. Siren, et al., “Shattering Barriers Toward Clinically Meaningful MSC Therapies,” Science Advances 6, no. 30 (2020): eaba6884.

[37]

J. Isaković, K. Šerer, B. Barišić, and D. Mitrečić, “Mesenchymal Stem Cell Therapy for Neurological Disorders: The Light or the Dark Side of the Force?,” Frontiers in Bioengineering and Biotechnology 11 (2023): 1139359.

[38]

W. Chen, L. Lv, N. Chen, and E. Cui, “Immunogenicity of Mesenchymal Stromal/Stem Cells,” Scandinavian Journal of Immunology 97, no. 6 (2023): e13267.

[39]

A. K. Berglund, L. A. Fortier, D. F. Antczak, and L. V. Schnabel, “Immunoprivileged no More: Measuring the Immunogenicity of Allogeneic Adult Mesenchymal Stem Cells,” Stem Cell Research & Therapy 8, no. 1 (2017): 288.

[40]

W. Z. Zhuang, Y. H. Lin, L. J. Su, et al., “Mesenchymal Stem/Stromal Cell-based Therapy: Mechanism, Systemic Safety and Biodistribution for Precision Clinical Applications,” Journal of Biomedical Science 28, no. 1 (2021): 28.

[41]

D. Lu, X. Jiao, W. Jiang, et al., “Mesenchymal Stem Cells Influence Monocyte/Macrophage Phenotype: Regulatory Mode and Potential Clinical Applications,” Biomedicine & Pharmacotherapy 165 (2023): 115042.

[42]

N. Song, M. Scholtemeijer, and K. Shah, “Mesenchymal Stem Cell Immunomodulation: Mechanisms and Therapeutic Potential,” Trends in Pharmacological Sciences 41, no. 9 (2020): 653-664.

[43]

L. Muller, A. Tunger, M. Wobus, et al., “Immunomodulatory Properties of Mesenchymal Stromal Cells: An Update,” Frontiers in Cell and Developmental Biology 9 (2021): 637725.

[44]

N. Luque-Campos, F. A. Bustamante-Barrientos, C. Pradenas, et al., “The Macrophage Response Is Driven by Mesenchymal Stem Cell-Mediated Metabolic Reprogramming,” Frontiers in Immunology 12 (2021): 624746.

[45]

H. Min, L. Xu, R. Parrott, et al., “Mesenchymal Stromal Cells Reprogram Monocytes and Macrophages With Processing Bodies,” Stem Cells 39, no. 1 (2021): 115-128.

[46]

T. S. Cheung, A. Galleu, M. von Bonin, M. Bornhauser, and F. Dazzi, “Apoptotic Mesenchymal Stromal Cells Induce Prostaglandin E2 in Monocytes: Implications for the Monitoring of Mesenchymal Stromal Cell Activity,” Haematologica 104, no. 10 (2019): e438-e441.

[47]

A. Galleu, Y. Riffo-Vasquez, C. Trento, et al., “Apoptosis in Mesenchymal Stromal Cells Induces in Vivo Recipient-mediated Immunomodulation,” Science Translational Medicine 9, no. 416 (2017): eaam7828.

[48]

S. H. M. Pang, J. D'Rozario, S. Mendonca, et al., “Mesenchymal Stromal Cell Apoptosis Is Required for Their Therapeutic Function,” Nature Communications 12, no. 1 (2021): 6495.

[49]

F. Mohamad Yusoff and Y. Higashi, “Mesenchymal Stem/Stromal Cells for Therapeutic Angiogenesis,” Cells 12, no. 17 (2023): 2162.

[50]

C. Huang, W. Luo, Q. Wang, et al., “Human Mesenchymal Stem Cells Promote Ischemic Repairment and Angiogenesis of Diabetic Foot Through Exosome miRNA-21-5p,” Stem Cell Research 52 (2021): 102235.

[51]

Y. L. Tan, S. P. Eng, P. Hafez, N. Abdul Karim, J. X. Law, and M. H. Ng, “Mesenchymal Stromal Cell Mitochondrial Transfer as a Cell Rescue Strategy in Regenerative Medicine: A Review of Evidence in Preclinical Models,” Stem Cells Translational Medicine 11, no. 8 (2022): 814-827.

[52]

A. N. Mukkala, M. Jerkic, Z. Khan, K. Szaszi, A. Kapus, and O. Rotstein, “Therapeutic Effects of Mesenchymal Stromal Cells Require Mitochondrial Transfer and Quality Control,” International Journal of Molecular Sciences 24, no. 21 (2023): 15788.

[53]

Y. Cen, G. Lou, J. Qi, M. Zheng, and Y. Liu, “A New Perspective on Mesenchymal Stem Cell-based Therapy for Liver Diseases: Restoring Mitochondrial Function,” Cell Communication and Signaling 21, no. 1 (2023): 214.

[54]

Medicine AfR. Cell Therapy Products.

[55]

US FDA. RYONCIL, https://www.fda.gov/vaccines-blood-biologics/cellular-gene-therapy-products/ryoncil.

[56]

G. Liu, B. T. David, M. Trawczynski, and R. G. Fessler, “Advances in Pluripotent Stem Cells: History, Mechanisms, Technologies, and Applications,” Stem Cell Reviews and Reports 16, no. 1 (2020): 3-32.

[57]

S. Yamanaka, “Pluripotent Stem Cell-Based Cell Therapy-Promise and Challenges,” Cell Stem Cell 27, no. 4 (2020): 523-531.

[58]

D. Liu, L. Bobrovskaya, and X.-F. Zhou, “Cell Therapy for Neurological Disorders: The Perspective of Promising Cells,” Biology (Basel) 10, no. 11 (2021): 1142.

[59]

R. Rahimi Darehbagh, S. A. Seyedoshohadaei, R. Ramezani, R. Ramezani, and N. Rezaei, “Stem Cell Therapies for Neurological Disorders: Current Progress, Challenges, and Future Perspectives,” European Journal of Medical Research 29, no. 1 (2024): 386.

[60]

N. Xie and B. Tang, “The Application of human iPSCs in Neurological Diseases: From Bench to Bedside,” Stem Cells International 2016, no. 1 (2016): 6484713.

[61]

J. Cerneckis, H. Cai, and Y. Shi, “Induced Pluripotent Stem Cells (iPSCs): Molecular Mechanisms of Induction and Applications,” Signal Transduction and Targeted Therapy 9, no. 1 (2024): 112.

[62]

L. Yang, S.-C. Liu, Y.-Y. Liu, et al., “Therapeutic Role of Neural Stem Cells in Neurological Diseases,” Frontiers in Bioengineering and Biotechnology 12 (2024): 1329712.

[63]

M. Singh, P. K. Pandey, A. Bhasin, M. Padma, and S. Mohanty, “Application of Stem Cells in Stroke: A Multifactorial Approach,” Frontiers in Neuroscience 14 (2020): 473.

[64]

W. Li, L. Shi, B. Hu, et al., “Mesenchymal Stem Cell-based Therapy for Stroke: Current Understanding and Challenges,” Frontiers in Cellular Neuroscience 15 (2021): 628940.

[65]

Y. Zhang, N. Dong, H. Hong, J. Qi, S. Zhang, and J. Wang, “Mesenchymal Stem Cells: Therapeutic Mechanisms for Stroke,” International Journal of Molecular Sciences 23, no. 5 (2022): 2550.

[66]

A. Andrzejewska, S. Dabrowska, B. Lukomska, and M. Janowski, “Mesenchymal Stem Cells for Neurological Disorders,” Advanced Science 8, no. 7 (2021): 2002944.

[67]

H. Huang, J. Zhang, J. Lin, and S. Shi, “Efficacy and Safety of Mesenchymal Stem Cells in Patients With Acute Ischemic Stroke: A Meta-analysis,” BMC Neurology 24, no. 1 (2024): 48.

[68]

J. D. Sinden, C. Hicks, P. Stroemer, I. Vishnubhatla, and R. Corteling, “Human Neural Stem Cell Therapy for Chronic Ischemic Stroke: Charting Progress From Laboratory to Patients,” Stem Cells and Development 26, no. 13 (2017): 933-947.

[69]

G. Zhang, Y. Li, J. L. Reuss, et al., “Stable Intracerebral Transplantation of Neural Stem Cells for the Treatment of Paralysis due to Ischemic Stroke,” Stem Cells Translational Medicine 8, no. 10 (2019): 999-1007.

[70]

D. Kalladka, J. Sinden, K. Pollock, et al., “Human Neural Stem Cells in Patients With Chronic Ischaemic Stroke (PISCES): A Phase 1, First-in-man Study,” The Lancet 388, no. 10046 (2016): 787-796.

[71]

K. W. Muir, D. Bulters, M. Willmot, et al., “Intracerebral Implantation of human Neural Stem Cells and Motor Recovery After Stroke: Multicentre Prospective Single-arm Study (PISCES-2),” Journal of Neurology, Neurosurgery & Psychiatry 91, no. 4 (2020): 396-401.

[72]

Y. Xia, G. Hu, Y. Chen, et al., “Embryonic Stem Cell Derived Small Extracellular Vesicles Modulate Regulatory T Cells to Protect Against Ischemic Stroke,” ACS Nano 15, no. 4 (2021): 7370-7385.

[73]

A. A. Taei, S. Nasoohi, G. Hassanzadeh, M. Kadivar, L. Dargahi, and M. Farahmandfar, “Enhancement of Angiogenesis and Neurogenesis by Intracerebroventricular Injection of Secretome From human Embryonic Stem Cell-derived Mesenchymal Stem Cells in Ischemic Stroke Model,” Biomedicine & Pharmacotherapy 140 (2021): 111709.

[74]

S. Yaqubi and M. Karimian, “Stem Cell Therapy as a Promising Approach for Ischemic Stroke Treatment,” Current Research in Pharmacology and Drug Discovery 6 (2024): 100183.

[75]

G. Shroff, “Comparison of nutech Functional Score With European Stroke Scale for Patients With Cerebrovascular Accident Treated With human Embryonic Stem Cells: Nfs for cva Patients Treated With hescs,” Journal of Vascular and Interventional Neurology 9, no. 4 (2017): 35.

[76]

R. Duan, Y. Gao, R. He, et al., “Induced Pluripotent Stem Cells for Ischemic Stroke Treatment,” Frontiers in Neuroscience 15 (2021): 628663.

[77]

K. Zhang, Y. Jiang, B. Wang, T. Li, D. Shang, and X. Zhang, “Mesenchymal Stem Cell Therapy: A Potential Treatment Targeting Pathological Manifestations of Traumatic Brain Injury,” Oxidative Medicine and Cellular Longevity 2022, no. 1 (2022): 4645021.

[78]

J. S. Andreassen, K. Thorsen, K. Søreide, D. Werner, and C. Weber, “Is There a Weekend Effect on Mortality Rate and Outcome for Moderate and Severe Traumatic Brain Injury? A Population-based, Observational Cohort Study,” Brain and Spine 2 (2022): 101699.

[79]

S. H. Haddad and Y. M. Arabi, “Critical Care Management of Severe Traumatic Brain Injury in Adults,” Scandinavian Journal of Trauma, Resuscitation and Emergency Medicine 20 (2012): 1-15.

[80]

Z.-X. Zhang, L.-X. Guan, K. Zhang, Q. Zhang, and L.-J. Dai, “A Combined Procedure to Deliver Autologous Mesenchymal Stromal Cells to Patients With Traumatic Brain Injury,” Cytotherapy 10, no. 2 (2008): 134-139.

[81]

C. Tian, X. Wang, X. Wang, et al., “Autologous Bone Marrow Mesenchymal Stem Cell Therapy in the Subacute Stage of Traumatic Brain Injury by Lumbar Puncture,” Experimental and Clinical Transplantation 11, no. 2 (2013): 176-181.

[82]

M. Kawabori, A. H. Weintraub, H. Imai, et al., “Cell Therapy for Chronic TBI: Interim Analysis of the Randomized Controlled STEMTRA Trial,” Neurology 96, no. 8 (2021): e1202-e1214.

[83]

S. Wang, H. Cheng, G. Dai, et al., “Umbilical Cord Mesenchymal Stem Cell Transplantation Significantly Improves Neurological Function in Patients With Sequelae of Traumatic Brain Injury,” Brain Research 1532 (2013): 76-84.

[84]

Z. Wang, Y. Luo, L. Chen, and W. Liang, “Safety of Neural Stem Cell Transplantation in Patients With Severe Traumatic Brain Injury,” Experimental and Therapeutic Medicine 13, no. 6 (2017): 3613-3618.

[85]

Y. Xia, J. Zhu, R. Yang, H. Wang, Y. Li, and C. Fu, “Mesenchymal Stem Cells in the Treatment of Spinal Cord Injury: Mechanisms, Current Advances and Future Challenges,” Frontiers in Immunology 14 (2023): 1141601.

[86]

L. T. de Araújo, C. T. Macêdo, P. K. F. Damasceno, et al., “Clinical Trials Using Mesenchymal Stem Cells for Spinal Cord Injury: Challenges in Generating Evidence,” Cells 11, no. 6 (2022): 1019.

[87]

S. Muthu, M. Jeyaraman, A. Gulati, and A. Arora, “Current Evidence on Mesenchymal Stem Cell Therapy for Traumatic Spinal Cord Injury: Systematic Review and Meta-analysis,” Cytotherapy 23, no. 3 (2021): 186-197.

[88]

F. Tahmasebi and S. Barati, “Effects of Mesenchymal Stem Cell Transplantation on Spinal Cord Injury Patients,” Cell and Tissue Research 389, no. 3 (2022): 373-384.

[89]

C. Gu, J. Feng, A. Waqas, et al., “Technological Advances of 3D Scaffold-based Stem Cell/Exosome Therapy in Tissues and Organs,” Frontiers in Cell and Developmental Biology 9 (2021): 709204.

[90]

G. Shroff, “Human Embryonic Stem Cell Therapy in Chronic Spinal Cord Injury: A Retrospective Study,” Clinical and Translational Science 9, no. 3 (2016): 168-175.

[91]

G. Shroff and R. Gupta, “Human Embryonic Stem Cells in the Treatment of Patients With Spinal Cord Injury,” Annals of Neurosciences 22, no. 4 (2015): 208.

[92]

S. McIntyre, S. Goldsmith, A. Webb, et al., “Global Prevalence of Cerebral Palsy: A Systematic Analysis,” Developmental Medicine & Child Neurology 64, no. 12 (2022): 1494-1506.

[93]

I. Novak, C. Morgan, M. Fahey, et al., “State of the Evidence Traffic Lights 2019: Systematic Review of Interventions for Preventing and Treating Children With Cerebral Palsy,” Current Neurology and Neuroscience Reports 20 (2020): 1-21.

[94]

B. Xie, M. Chen, R. Hu, W. Han, and S. Ding, “Therapeutic Evidence of Human Mesenchymal Stem Cell Transplantation for Cerebral Palsy: A Meta-Analysis of Randomized Controlled Trials,” Stem Cells International 2020, no. 1 (2020): 5701920.

[95]

Z.-Y. Lv, Y. Li, and J. Liu, “Progress in Clinical Trials of Stem Cell Therapy for Cerebral Palsy,” Neural Regeneration Research 16, no. 7 (2021): 1377-1382.

[96]

G. Shroff, A. Gupta, and J. K. Barthakur, “Therapeutic Potential of human Embryonic Stem Cell Transplantation in Patients With Cerebral Palsy,” Journal of Translational Medicine 12 (2014): 1-9.

[97]

A. Bhatt, H. Bhardwaj, and P. Shrivastava, “Mesenchymal Stem Cell Therapy for Alzheimer's Disease: A Novel Therapeutic Approach for Neurodegenerative Diseases,” Neuroscience 555 (2024): 52-68.

[98]

S. Regmi, D. D. Liu, M. Shen, et al., “Mesenchymal Stromal Cells for the Treatment of Alzheimer's Disease: Strategies and Limitations,” Frontiers in Molecular Neuroscience 15 (2022): 1011225.

[99]

H. J. Kim, S. W. Seo, J. W. Chang, et al., “Stereotactic Brain Injection of human Umbilical Cord Blood Mesenchymal Stem Cells in Patients With Alzheimer's Disease Dementia: A Phase 1 Clinical Trial,” Alzheimer's & Dementia: Translational Research & Clinical Interventions 1, no. 2 (2015): 95-102.

[100]

H. J. Kim, K. R. Cho, H. Jang, et al., “Intracerebroventricular Injection of human Umbilical Cord Blood Mesenchymal Stem Cells in Patients With Alzheimer's disease Dementia: A Phase I Clinical Trial,” Alzheimer's Research & Therapy 13 (2021): 1-11.

[101]

C. Duma, O. Kopyov, A. Kopyov, et al., “Human Intracerebroventricular (ICV) Injection of Autologous, Non-engineered, Adipose-derived Stromal Vascular Fraction (ADSVF) for Neurodegenerative Disorders: Results of a 3-year Phase 1 Study of 113 Injections in 31 Patients,” Molecular Biology Reports 46, no. 5 (2019): 5257-5272.

[102]

M. T. Valenti, M. Serena, L. Dalle Carbonare, and D. Zipeto, “CRISPR/Cas System: An Emerging Technology in Stem Cell Research,” World Journal of Stem Cells 11, no. 11 (2019): 937.

[103]

K. S. Chen, M. H. Noureldein, L. M. McGinley, et al., “Human Neural Stem Cells Restore Spatial Memory in a Transgenic Alzheimer's disease Mouse Model by an Immunomodulating Mechanism,” Frontiers in Aging Neuroscience 15 (2023): 1306004.

[104]

M. H. Tuszynski, L. Thal, M. Pay, et al., “A Phase 1 Clinical Trial of Nerve Growth Factor Gene Therapy for Alzheimer Disease,” Nature Medicine 11, no. 5 (2005): 551-555.

[105]

A. Ortega, B. Chernicki, G. Ou, and M. S. Parmar, “From Lab Bench to Hope: Emerging Gene Therapies in Clinical Trials for Alzheimer's Disease,” Molecular Neurobiology (2024): 1-24.

[106]

P. Tambe, V. Undale, A. Sanap, R. Bhonde, and N. Mante, “The Prospective Role of Mesenchymal Stem Cells in Parkinson's Disease,” Parkinsonism & Related Disorders 127 (2024): 107087.

[107]

R. M. Heris, M. Shirvaliloo, S. Abbaspour-Aghdam, et al., “The Potential Use of Mesenchymal Stem Cells and Their Exosomes in Parkinson's disease Treatment,” Stem Cell Research & Therapy 13, no. 1 (2022): 371.

[108]

A. Unnisa, K. Dua, and M. A. Kamal, “Mechanism of Mesenchymal Stem Cells as a Multitarget Disease-Modifying Therapy for Parkinson's Disease,” Current Neuropharmacology 21, no. 4 (2023): 988.

[109]

N. K. Venkataramana, S. K. Kumar, S. Balaraju, et al., “Open-labeled Study of Unilateral Autologous Bone-marrow-derived Mesenchymal Stem Cell Transplantation in Parkinson's Disease,” Translational Research 155, no. 2 (2010): 62-70.

[110]

M. Schiess, J. Suescun, M. F. Doursout, et al., “Allogeneic Bone Marrow-derived Mesenchymal Stem Cell Safety in Idiopathic Parkinson's Disease,” Movement Disorders 36, no. 8 (2021): 1825-1834.

[111]

Y. Qiu, Z. Wang, and H.-S. Lu, “Umbilical Cord Mesenchymal Stem Cell Transplantation for Treatment of Parkinson's Disease in 8 Cases,” Chinese Journal of Tissue Engineering Research 15, no. 36 (2011): 6833.

[112]

Y. Wang, X.-L. Zhao, J.-Y. Zhang, and J. Tan, “Therapeutic Applications of Umbilical Cord Mesenchymal Stem Cells in Parkinson's Disease,” Chinese Journal of Tissue Engineering Research 18, no. 6 (2014): 932.

[113]

K. Shigematsu, N. Komori, K. Tahara, and H. Yamagishi, “Repeated Infusion of Autologous Adipose Tissue-derived Stem Cells for Parkinson's Disease,” Acta Neurologica Scandinavica 145, no. 1 (2022): 119-122.

[114]

J. Lee, D. Bayarsaikhan, R. Arivazhagan, et al., “CRISPR/Cas9 Edited sRAGE-MSCs Protect Neuronal Death in Parkinson's Disease Model,” International Journal of Stem Cells 12, no. 1 (2019): 114-124.

[115]

L. Leng and T. Zengmin, “Transplantation of Neural Precursor Cells in the Treatment of Parkinson Disease: An Efficacy and Safety Analysis,” Turkish Neurosurgery 26, no. 3 (2016): 378-383.

[116]

I. Madrazo, O. Kopyov, M. Ávila-Rodríguez, et al., “Transplantation of human Neural Progenitor Cells (NPC) Into Putamina of parkinsonian Patients: A Case Series Study, Safety and Efficacy Four Years After Surgery,” Cell Transplantation 28, no. 3 (2019): 269-285.

[117]

J. S. Schweitzer, B. Song, T. M. Herrington, et al., “Personalized iPSC-derived Dopamine Progenitor Cells for Parkinson's Disease,” New England Journal of Medicine 382, no. 20 (2020): 1926-1932.

[118]

WHO. Autism Spectrum Disorder. September 18, 2024. Accessed September 18, 2024. https://www.who.int/news-room/fact-sheets/detail/autism-spectrum-disorders.

[119]

A. Akat and E. Karaöz, “Cell Therapies for Autism Spectrum Disorder: A Systematic Review of Clinical Applications,” Middle East Current Psychiatry 30, no. 1 (2023): 94.

[120]

R. Tamouza, F. Volt, J.-R. Richard, et al., “Possible Effect of the Use of Mesenchymal Stromal Cells in the Treatment of Autism Spectrum Disorders: A Review,” Frontiers in Cell and Developmental Biology 10 (2022): 809686.

[121]

B. Gesundheit, P. Ashwood, A. Keating, D. Naor, M. Melamed, and J. P. Rosenzweig, “Therapeutic Properties of Mesenchymal Stem Cells for Autism Spectrum Disorders,” Medical Hypotheses 84, no. 3 (2015): 169-177.

[122]

Q. Liu, M.-X. Chen, L. Sun, et al., “Rational Use of Mesenchymal Stem Cells in the Treatment of Autism Spectrum Disorders,” World Journal of Stem Cells 11, no. 2 (2019): 55.

[123]

S. Y. Lee, H. Ahn, W.-J. Jung, J. Ahn, and K. H. Lee, “A Case Study on Autism Spectrum Disorder Treatment Using Allogenic Mesenchymal Stem Cells Derived From the Human Umbilical Cord,” Case Reports: Open Access 5 (2020): 1-7.

[124]

J. M. Sun, G. Dawson, L. Franz, et al., “Infusion of human Umbilical Cord Tissue Mesenchymal Stromal Cells in Children With autism Spectrum Disorder,” Stem Cells Translational Medicine 9, no. 10 (2020): 1137-1146.

[125]

N. Sharifzadeh, A. Ghasemi, J. Tavakol Afshari, et al., “Intrathecal Autologous Bone Marrow Stem Cell Therapy in Children With Autism: A Randomized Controlled Trial,” Asia-Pacific Psychiatry 13, no. 2 (2021): e12445.

[126]

E. Deneault, S. H. White, D. C. Rodrigues, et al., “Complete Disruption of Autism-susceptibility Genes by Gene Editing Predominantly Reduces Functional Connectivity of Isogenic human Neurons,” Stem Cell Reports 11, no. 5 (2018): 1211-1225.

[127]

S. Najafi, P. Najafi, N. K. Farkhad, et al., “Mesenchymal Stem Cell Therapy in Amyotrophic Lateral Sclerosis (ALS) Patients: A Comprehensive Review of Disease Information and Future Perspectives,” Iranian Journal of Basic Medical Sciences 26, no. 8 (2023): 872.

[128]

S. H. Kim, K.-W. Oh, M.-Y. Noh, and M.-S. Kwon, “Optimal Therapeutic Strategy of Bone Marrow-Originated Autologous Mesenchymal Stromal/Stem Cells for ALS,” Stem Cells Translational Medicine 13, no. 4 (2024): 309-316.

[129]

L. Mazzini, F. Fagioli, R. Boccaletti, et al., “Stem Cell Therapy in Amyotrophic Lateral Sclerosis: A Methodological Approach in Humans,” Amyotrophic Lateral Sclerosis and Other Motor Neuron Disorders 4, no. 3 (2003): 158-161.

[130]

J. D. Glass, V. S. Hertzberg, N. M. Boulis, et al., “Transplantation of Spinal Cord-derived Neural Stem Cells for ALS: Analysis of Phase 1 and 2 Trials,” Neurology 87, no. 4 (2016): 392-400.

[131]

E. L. Feldman, N. M. Boulis, J. Hur, et al., “Intraspinal Neural Stem Cell Transplantation in amyotrophic Lateral sclerosis: Phase 1 Trial Outcomes,” Annals of Neurology 75, no. 3 (2014): 363-373.

[132]

L. Mazzini, M. Gelati, D. C. Profico, et al., “Human Neural Stem Cell Transplantation in ALS: Initial Results From a Phase I Trial,” Journal of Translational Medicine 13 (2015): 1-16.

[133]

R. H. Baloh, J. P. Johnson, P. Avalos, et al., “Transplantation of human Neural Progenitor Cells Secreting GDNF Into the Spinal Cord of Patients With ALS: A Phase 1/2a Trial,” Nature Medicine 28, no. 9 (2022): 1813-1822.

[134]

T.-T. Li, Z.-R. Wang, W.-Q. Yao, E.-Q. Linghu, F.-S. Wang, and L. Shi, “Stem Cell Therapies for Chronic Liver Diseases: Progress and Challenges,” Stem Cells Translational Medicine 11, no. 9 (2022): 900-911.

[135]

W. Lu, J. Qu, L. Yan, et al., “Efficacy and Safety of Mesenchymal Stem Cell Therapy in Liver Cirrhosis: A Systematic Review and Meta-analysis,” Stem Cell Research & Therapy 14, no. 1 (2023): 301.

[136]

Clinicaltrials.gove. Clinical trials using MSCs for liver disease. September 09, 2024. Accessed September 29, 2024. https://clinicaltrials.gov/search?cond=liver%20disease&intr=mesenchymal%20stem%20cell%20OR%20mesenchymal%20stromal%20cell.

[137]

Y. Nagamoto, K. Takayama, K. Ohashi, et al., “Transplantation of a human iPSC-derived Hepatocyte Sheet Increases Survival in Mice With Acute Liver Failure,” Journal of Hepatology 64, no. 5 (2016): 1068-1075.

[138]

A. Laemmle, M. Poms, B. Hsu, et al., “Aquaporin 9 Induction in human i PSC-derived Hepatocytes Facilitates Modeling of Ornithine Transcarbamylase Deficiency,” Hepatology 76, no. 3 (2022): 646-659.

[139]

M. J. Hossain, M. Al-Mamun, and M. R. Islam, “Diabetes Mellitus, the Fastest Growing Global Public Health Concern: Early Detection Should be Focused,” Health Science Reports 7, no. 3 (2024): e2004.

[140]

M. A. Ghoneim, M. M. Gabr, S. M. El-Halawani, and A. F. Refaie, “Current Status of Stem Cell Therapy for Type 1 Diabetes: A Critique and a Prospective Consideration,” Stem Cell Research & Therapy 15, no. 1 (2024): 23.

[141]

J. Hu, X. Yu, Z. Wang, et al., “Long Term Effects of the Implantation of Wharton's Jelly-derived Mesenchymal Stem Cells From the Umbilical Cord for Newly-onset Type 1 Diabetes Mellitus,” Endocrine Journal 60, no. 3 (2013): 347-357.

[142]

P.-O. Carlsson, E. Schwarcz, O. Korsgren, and K. Le Blanc, “Preserved β-cell Function in Type 1 Diabetes by Mesenchymal Stromal Cells,” Diabetes 64, no. 2 (2015): 587-592.

[143]

D. B. Araujo, J. R. Dantas, K. R. Silva, et al., “Allogenic Adipose Tissue-derived Stromal/Stem Cells and Vitamin D Supplementation in Patients With Recent-onset Type 1 Diabetes Mellitus: A 3-month Follow-up Pilot Study,” Frontiers in Immunology 11 (2020): 993.

[144]

M. Izadi, A. Sadr Hashemi Nejad, M. Moazenchi, et al., “Mesenchymal Stem Cell Transplantation in Newly Diagnosed Type-1 Diabetes Patients: A Phase I/II Randomized Placebo-controlled Clinical Trial,” Stem Cell Research & Therapy 13, no. 1 (2022): 264.

[145]

J. He, D. Kong, Z. Yang, et al., “Clinical Efficacy on Glycemic Control and Safety of Mesenchymal Stem Cells in Patients With Diabetes Mellitus: Systematic Review and Meta-analysis of RCT Data,” PLoS One 16, no. 3 (2021): e0247662.

[146]

T. C. Schulz, “Concise Review: Manufacturing of Pancreatic Endoderm Cells for Clinical Trials in Type 1 Diabetes,” Stem Cells Translational Medicine 4, no. 8 (2015): 927-931.

[147]

R. R. Henry, J. Pettus, J. Wilensky, et al., “Initial Clinical Evaluation of VC-01TM Combination Product—a Stem Cell-derived Islet Replacement for Type 1 Diabetes (T1D),” Diabetes 67, no. Supplement_1 (2018): 138-OR.

[148]

A. J. Shapiro, D. Thompson, T. W. Donner, et al., “Insulin Expression and C-peptide in Type 1 Diabetes Subjects Implanted With Stem Cell-derived Pancreatic Endoderm Cells in an Encapsulation Device,” Cell Reports Medicine 2, no. 12 (2021): 100466.

[149]

J. A. Bluestone, J. H. Buckner, M. Fitch, et al., “Type 1 Diabetes Immunotherapy Using Polyclonal Regulatory T Cells,” Science Translational Medicine 7, no. 315 (2015): 315ra189-315ra189.

[150]

S. Gao, Y. Zhang, K. Liang, R. Bi, and Y. Du, “Mesenchymal Stem Cells (MSCs): A Novel Therapy for Type 2 Diabetes,” Stem Cells International 2022, no. 1 (2022): 8637493.

[151]

K.-S. Park, Y.-S. Kim, J.-H. Kim, et al., “Trophic Molecules Derived From human Mesenchymal Stem Cells Enhance Survival, Function, and Angiogenesis of Isolated Islets After Transplantation,” Transplantation 89, no. 5 (2010): 509-517.

[152]

M. Shrestha, T. T. Nguyen, J. Park, et al., “Immunomodulation Effect of Mesenchymal Stem Cells in Islet Transplantation,” Biomedicine & Pharmacotherapy 142 (2021): 112042.

[153]

D. J. Borg, M. Weigelt, C. Wilhelm, et al., “Mesenchymal Stromal Cells Improve Transplanted Islet Survival and Islet Function in a syngeneic Mouse Model,” Diabetologia 57 (2014): 522-531.

[154]

K. Zhao, H. Hao, J. Liu, et al., “Bone Marrow-derived Mesenchymal Stem Cells Ameliorate Chronic High Glucose-induced β-cell Injury Through Modulation of Autophagy,” Cell Death & Disease 6, no. 9 (2015): e1885-e1885.

[155]

Y. Yuan, L. Yuan, L. Li, et al., “Mitochondrial Transfer From Mesenchymal Stem Cells to Macrophages Restricts Inflammation and Alleviates Kidney Injury in Diabetic Nephropathy Mice via PGC-1α Activation,” Stem Cells 39, no. 7 (2021): 913-928.

[156]

N. Konari, K. Nagaishi, S. Kikuchi, and M. Fujimiya, “Mitochondria Transfer From Mesenchymal Stem Cells Structurally and Functionally Repairs Renal Proximal Tubular Epithelial Cells in Diabetic Nephropathy in Vivo,” Scientific Reports 9, no. 1 (2019): 5184.

[157]

C. L. Rackham, E. L. Hubber, A. Czajka, A. N. Malik, A. J. King, and P. M. Jones, “Optimizing Beta Cell Function Through Mesenchymal Stromal Cell-mediated Mitochondria Transfer,” Stem Cells 38, no. 4 (2020): 574-584.

[158]

Clinicaltrials.gov. October 1st, 2024. Accessed October 1st, 2024. https://clinicaltrials.gov/search?cond=Diabetes%20Type%202&intr=mesenchymal%20stem%20cell%20OR%20mesenchymal%20stromal%20cell&page=1.

[159]

U. E. Habiba, N. Khan, D. L. Greene, K. Ahmad, S. Shamim, and A. Umer, “Meta-analysis Shows That Mesenchymal Stem Cell Therapy Can be a Possible Treatment for Diabetes,” Frontiers in Endocrinology (Lausanne) 15 (2024): 1380443.

[160]

Y. Li, F. Wang, H. Liang, et al., “Efficacy of Mesenchymal Stem Cell Transplantation Therapy for Type 1 and Type 2 Diabetes Mellitus: A Meta-analysis,” Stem Cell Research & Therapy 12, no. 1 (2021): 273.

[161]

J. Wu, T. Li, M. Guo, et al., “Treating a Type 2 Diabetic Patient With Impaired Pancreatic Islet Function by Personalized Endoderm Stem Cell-derived Islet Tissue,” Cell Discovery 10, no. 1 (2024): 45.

[162]

D. Balboa, J. Saarimäki-Vire, D. Borshagovski, et al., “Insulin Mutations Impair Beta-cell Development in a Patient-derived iPSC Model of Neonatal Diabetes,” Elife 7 (2018): e38519.

[163]

S. Safiri, K. Carson-Chahhoud, M. Noori, et al., “Burden of Chronic Obstructive Pulmonary Disease and Its Attributable Risk Factors in 204 Countries and territories, 1990-2019: Results From the Global Burden of Disease Study 2019,” Bmj 378 (2022): e069679.

[164]

Z. N. Wang and X. X. Tang, “New Perspectives on the Aberrant Alveolar Repair of Idiopathic Pulmonary Fibrosis,” Frontiers in Cell and Developmental Biology 8 (2020): 580026.

[165]

A. Daher and M. Dreher, “Supplemental Oxygen Therapy in Chronic Obstructive Pulmonary Disease: Is Less Is More? How Much Is Too Much?,” Current Opinion in Pulmonary Medicine 30, no. 2 (2024): 179-184.

[166]

F. M. Siddiqui and J. M. Diamond, “Lung Transplantation for Chronic Obstructive Pulmonary Disease: Past, Present, and Future Directions,” Current Opinion in Pulmonary Medicine 24, no. 2 (2018): 199-204.

[167]

M. A. Matthay, R. L. Zemans, G. A. Zimmerman, et al., “Acute respiratory Distress Syndrome,” Nature Reviews Disease Primers 5, no. 1 (2019): 18.

[168]

J. C. Grotberg, D. Reynolds, and B. D. Kraft, “Management of Severe Acute respiratory Distress Syndrome: A Primer,” Critical Care 27, no. 1 (2023): 289.

[169]

D. Murugan and L. Rangasamy, “Pooled Evidence From Preclinical and Clinical Studies for Stem Cell-based Therapy in ARDS and COVID-19,” Molecular and Cellular Biochemistry 478, no. 7 (2023): 1487-1518.

[170]

L. Chow, V. Johnson, R. Impastato, J. Coy, A. Strumpf, and S. Dow, “Antibacterial Activity of human Mesenchymal Stem Cells Mediated Directly by Constitutively Secreted Factors and Indirectly by Activation of Innate Immune Effector Cells,” Stem Cells Translational Medicine 9, no. 2 (2020): 235-249.

[171]

O. E. Simonson, D. Mougiakakos, N. Heldring, et al., “In Vivo Effects of Mesenchymal Stromal Cells in Two Patients With Severe Acute respiratory Distress Syndrome,” Stem Cells Translational Medicine 4, no. 10 (2015): 1199-1213.

[172]

J. G. Wilson, K. D. Liu, H. Zhuo, et al., “Mesenchymal Stem (stromal) Cells for Treatment of ARDS: A Phase 1 Clinical Trial,” The Lancet Respiratory Medicine 3, no. 1 (2015): 24-32.

[173]

M. A. Matthay, C. S. Calfee, H. Zhuo, et al., “Treatment With Allogeneic Mesenchymal Stromal Cells for Moderate to Severe Acute respiratory Distress Syndrome (START study): A Randomised Phase 2a Safety Trial,” The Lancet Respiratory Medicine 7, no. 2 (2019): 154-162.

[174]

K. Ichikado, T. Kotani, Y. Kondoh, et al., “Clinical Efficacy and Safety of Multipotent Adult Progenitor Cells (invimestrocel) for Acute respiratory Distress Syndrome (ARDS) Caused by Pneumonia: A Randomized, Open-label, Standard Therapy-controlled, Phase 2 Multicenter Study (ONE-BRIDGE),” Stem Cell Research & Therapy 14, no. 1 (2023): 217.

[175]

B. Thébaud, K. N. Goss, M. Laughon, et al., “Bronchopulmonary Dysplasia,” Nature Reviews Disease Primers 5, no. 1 (2019): 78.

[176]

A. R. Schmidt and C. Ramamoorthy, “Bronchopulmonary Dysplasia,” Pediatric Anesthesia 32, no. 2 (2022): 174-180.

[177]

Y. S. Chang, S. Y. Ahn, H. S. Yoo, et al., “Mesenchymal Stem Cells for Bronchopulmonary Dysplasia: Phase 1 Dose-escalation Clinical Trial,” The Journal of Pediatrics 164, no. 5 (2014): 966-972. e6.

[178]

N. T. Liem, T. L. Anh, T. T. H. Thai, and B. V. Anh, “Bone Marrow Mononuclear Cells Transplantation in Treatment of Established Bronchopulmonary Dysplasia: A Case Report,” The American Journal of Case Reports 18 (2017): 1090.

[179]

S. Y. Ahn, Y. S. Chang, J. H. Kim, S. I. Sung, and W. S. Park, “Two-year Follow-up Outcomes of Premature Infants Enrolled in the Phase I Trial of Mesenchymal Stem Cells Transplantation for Bronchopulmonary Dysplasia,” The Journal of Pediatrics 185 (2017): 49-54. e2.

[180]

S. Y. Ahn, Y. S. Chang, M. H. Lee, et al., “Stem Cells for Bronchopulmonary Dysplasia in Preterm Infants: A Randomized Controlled Phase II Trial,” Stem Cells Translational Medicine 10, no. 8 (2021): 1129-1137.

[181]

M. J. del Cerro Marín, I. G. Ormazábal, A. Gimeno-Navarro, et al., “Repeated Intravenous Doses of human Umbilical Cord-derived Mesenchymal Stromal Cells for Bronchopulmonary Dysplasia: Results of a Phase 1 Clinical Trial With 2-year Follow-up,” Cytotherapy 26, no. 6 (2024): 632-640.

[182]

A. Sharma, I. Ahmad Farouk, and S. K. Lal, “COVID-19: A Review on the Novel Coronavirus Disease Evolution, Transmission, Detection, Control and Prevention,” Viruses. 13, no. 2 (2021): 202.

[183]

E. Arabpour, S. Khoshdel, N. Tabatabaie, A. Akhgarzad, M. Zangiabadian, and M. J. Nasiri, “Stem Cells Therapy for COVID-19: A Systematic Review and Meta-analysis,” Frontiers in Medicine 8 (2021): 737590.

[184]

Y. Feng, J. Huang, J. Wu, et al., “Safety and Feasibility of Umbilical Cord Mesenchymal Stem Cells in Patients With COVID-19 Pneumonia: A Pilot Study,” Cell Proliferation 53, no. 12 (2020): e12947.

[185]

F. Meng, R. Xu, S. Wang, et al., “Human Umbilical Cord-derived Mesenchymal Stem Cell Therapy in Patients With COVID-19: A Phase 1 Clinical Trial,” Signal Transduction and Targeted Therapy 5, no. 1 (2020): 172.

[186]

L. Shu, C. Niu, R. Li, et al., “Treatment of Severe COVID-19 With human Umbilical Cord Mesenchymal Stem Cells,” Stem Cell Research & Therapy 11 (2020): 1-11.

[187]

H. Y. Li, T. Y. Gao, W. Fang, et al., “Global, Regional and National Burden of Chronic Obstructive Pulmonary Disease Over a 30-year Period: Estimates From the 1990 to 2019 Global Burden of Disease Study,” Respirology (Carlton, Vic.) 28, no. 1 (2023): 29-36.

[188]

M. Hikichi, K. Mizumura, S. Maruoka, and Y. Gon, “Pathogenesis of Chronic Obstructive Pulmonary Disease (COPD) Induced by Cigarette Smoke,” Journal of Thoracic Disease 11, no. Suppl 17 (2019): S2129.

[189]

W. Broekman, P. P. Khedoe, K. Schepers, H. Roelofs, J. Stolk, and P. S. Hiemstra, “Mesenchymal Stromal Cells: A Novel Therapy for the Treatment of Chronic Obstructive Pulmonary Disease?,” Thorax 73, no. 6 (2018): 565-574.

[190]

X. Liu, Q. Fang, and H. Kim, “Preclinical Studies of Mesenchymal Stem Cell (MSC) Administration in Chronic Obstructive Pulmonary Disease (COPD): A Systematic Review and Meta-analysis,” PLoS One 11, no. 6 (2016): e0157099.

[191]

H. G. de Oliveira, F. F. Cruz, M. A. Antunes, et al., “Combined Bone Marrow-derived Mesenchymal Stromal Cell Therapy and One-way Endobronchial Valve Placement in Patients With Pulmonary Emphysema: A Phase I Clinical Trial,” Stem Cells Translational Medicine 6, no. 3 (2017): 962-969.

[192]

E. C. van Doorn, J. H. Amesz, A. H. Sadeghi, N. M. de Groot, O. C. Manintveld, and Y. J. Taverne, “Preclinical Models of Cardiac Disease: A Comprehensive Overview for Clinical Scientists,” Cardiovascular Engineering and Technology 15, no. 2 (2024): 232-249.

[193]

S. Funakoshi and Y. Yoshida, “Recent Progress of iPSC Technology in Cardiac Diseases,” Archives of Toxicology 95, no. 12 (2021): 3633-3650.

[194]

J. M. Hare, J. E. Fishman, G. Gerstenblith, et al., “Comparison of Allogeneic vs Autologous Bone Marrow-derived Mesenchymal Stem Cells Delivered by Transendocardial Injection in Patients With Ischemic Cardiomyopathy: The POSEIDON Randomized Trial,” Jama 308, no. 22 (2012): 2369-2379.

[195]

A. B. Mathiasen, A. A. Qayyum, E. Jørgensen, et al., “Bone Marrow-derived Mesenchymal Stromal Cell Treatment in Patients With Severe Ischaemic Heart Failure: A Randomized Placebo-controlled Trial (MSC-HF trial),” European Heart Journal 36, no. 27 (2015): 1744-1753.

[196]

X. He, Q. Wang, Y. Zhao, et al., “Effect of Intramyocardial Grafting Collagen Scaffold With Mesenchymal Stromal Cells in Patients With Chronic Ischemic Heart Disease: A Randomized Clinical Trial,” JAMA Network Open 3 (2020): e2016236.

[197]

J. Bartunek, A. Terzic, B. A. Davison, et al., “Cardiopoietic Cell Therapy for Advanced Ischaemic Heart Failure: Results at 39 Weeks of the Prospective, Randomized, Double Blind, Sham-controlled CHART-1 Clinical Trial,” European Heart Journal 38, no. 9 (2017): 648-660.

[198]

S. Miyagawa, S. Kainuma, T. Kawamura, et al., “Case Report: Transplantation of human Induced Pluripotent Stem Cell-derived Cardiomyocyte Patches for Ischemic Cardiomyopathy,” Frontiers in Cardiovascular Medicine 9 (2022): 950829.

[199]

T. K. Gill, M. M. Mittinty, L. M. March, et al., “Global, Regional, and National Burden of Other Musculoskeletal Disorders, 1990-2020, and Projections to 2050: A Systematic Analysis of the Global Burden of Disease Study 2021,” The Lancet Rheumatology 5, no. 11 (2023): e670-e682.

[200]

B. M. Fullen, H. Wittink, A. De Groef, et al., “Musculoskeletal Pain: Current and Future Directions of Physical Therapy Practice,” Archives of Rehabilitation Research and Clinical Translation 5, no. 1 (2023): 100258.

[201]

V. Molnar, E. Pavelić, K. Vrdoljak, et al., “Mesenchymal Stem Cell Mechanisms of Action and Clinical Effects in Osteoarthritis: A Narrative Review,” Genes 13, no. 6 (2022): 949.

[202]

J. Matas, C. García, D. Poblete, et al., “A Phase I Dose-escalation Clinical Trial to Assess the Safety and Efficacy of Umbilical Cord-derived Mesenchymal Stromal Cells in Knee Osteoarthritis,” Stem Cells Translational Medicine 13, no. 3 (2024): 193-203.

[203]

K.-I. Kim, M. C. Lee, J. H. Lee, et al., “Clinical Efficacy and Safety of the Intra-articular Injection of Autologous Adipose-derived Mesenchymal Stem Cells for Knee Osteoarthritis: A Phase III, Randomized, Double-blind, Placebo-controlled Trial,” The American Journal of Sports Medicine 51, no. 9 (2023): 2243-2253.

[204]

D. Jevotovsky, A. Alfonso, T. Einhorn, and E. Chiu, “Osteoarthritis and Stem Cell Therapy in Humans: A Systematic Review,” Osteoarthritis and Cartilage 26, no. 6 (2018): 711-729.

[205]

T. G. Wiggers, M. Winters, N. A. Van den Boom, H. J. Haisma, and M. H. Moen, “Autologous Stem Cell Therapy in Knee Osteoarthritis: A Systematic Review of Randomised Controlled Trials,” British Journal of Sports Medicine 55, no. 20 (2021): 1161-1169.

[206]

P. Peláez, E. Damiá, M. Torres-Torrillas, et al., “Cell and Cell Free Therapies in Osteoarthritis,” Biomedicines 9, no. 11 (2021): 1726.

[207]

M. Lee, S. Y. Jeong, J. Ha, et al., “Low Immunogenicity of Allogeneic human Umbilical Cord Blood-derived Mesenchymal Stem Cells in Vitro and in Vivo,” Biochemical and Biophysical Research Communications 446, no. 4 (2014): 983-989.

[208]

J.-S. Song, K.-T. Hong, N.-M. Kim, et al., “Implantation of Allogenic Umbilical Cord Blood-derived Mesenchymal Stem Cells Improves Knee Osteoarthritis Outcomes: Two-year Follow-up,” Regenerative Therapy 14 (2020): 32-39.

[209]

C. Albrecht. Arthroscopic cartilage cell transplantation (Spherox).

[210]

P. Behrens, T. Bitter, B. Kurz, and M. Russlies, “Matrix-associated Autologous Chondrocyte Transplantation/Implantation (MACT/MACI)—5-year Follow-up,” The Knee 13, no. 3 (2006): 194-202.

[211]

B. Sadri, M. Hassanzadeh, A. Bagherifard, et al., “Cartilage Regeneration and Inflammation Modulation in Knee Osteoarthritis Following Injection of Allogeneic Adipose-derived Mesenchymal Stromal Cells: A Phase II, Triple-blinded, Placebo Controlled, Randomized Trial,” Stem Cell Research & Therapy 14, no. 1 (2023): 162.

[212]

A. Saito, A. Ooki, T. Nakamura, et al., “Targeted Reversion of Induced Pluripotent Stem Cells From Patients With human Cleidocranial Dysplasia Improves Bone Regeneration in a Rat Calvarial Bone Defect Model,” Stem Cell Research & Therapy 9 (2018): 1-10.

[213]

C. Long, J. R. McAnally, J. M. Shelton, A. A. Mireault, R. Bassel-Duby, and E. N. Olson, “Prevention of Muscular Dystrophy in Mice by CRISPR/Cas9-mediated Editing of Germline DNA,” Science 345, no. 6201 (2014): 1184-1188.

[214]

C. M. van Gelder, A. A. Vollebregt, I. Plug, A. T. van der Ploeg, and A. J. Reuser, “Treatment Options for Lysosomal Storage Disorders: Developing Insights,” Expert Opinion on Pharmacotherapy 13, no. 16 (2012): 2281-2299.

[215]

M. Fukami, “Ovarian Dysfunction in Women With Turner Syndrome,” Frontiers in Endocrinology (Lausanne) 14 (2023): 1160258.

[216]

S. L. Sherman, “Premature Ovarian Failure in the Fragile X Syndrome,” American Journal of Medical Genetics 97, no. 3 (2000): 189-194.

[217]

H. Ke, S. Tang, T. Guo, et al., “Landscape of Pathogenic Mutations in Premature Ovarian Insufficiency,” Nature Medicine 29, no. 2 (2023): 483-492.

[218]

R. Beranger, P. Hoffmann, S. Christin-Maitre, and V. Bonneterre, “Occupational Exposures to Chemicals as a Possible Etiology in Premature Ovarian Failure: A Critical Analysis of the Literature,” Reproductive Toxicology 33, no. 3 (2012): 269-279.

[219]

M. Ebrahimi and F. Akbari Asbagh, “Pathogenesis and Causes of Premature Ovarian Failure: An Update,” International Journal of Fertility and Sterility 5, no. 2 (2011): 54-65.

[220]

H. Gao, L. Gao, and W. Wang, “Advances in the Cellular Immunological Pathogenesis and Related Treatment of Primary Ovarian Insufficiency,” American Journal of Reproductive Immunology 88, no. 5 (2022): e13622.

[221]

C. Yamashiro, K. Sasaki, Y. Yabuta, et al., “Generation of human Oogonia From Induced Pluripotent Stem Cells in Vitro,” Science 362, no. 6412 (2018): 356-360.

[222]

K. Zou, Z. Yuan, Z. Yang, et al., “Production of Offspring From a Germline Stem Cell Line Derived From Neonatal Ovaries,” Nature Cell Biology 11, no. 5 (2009): 631-636.

[223]

Y. A. R. White, D. C. Woods, Y. Takai, O. Ishihara, H. Seki, and J. L. Tilly, “Oocyte Formation by Mitotically Active Germ Cells Purified From Ovaries of Reproductive-age Women,” Nature Medicine 18, no. 3 (2012): 413-421.

[224]

Z. Li, M. Zhang, Y. Tian, Q. Li, and X. Huang, “Mesenchymal Stem Cells in Premature Ovarian Insufficiency: Mechanisms and Prospects,” Frontiers in Cell and Developmental Biology 9 (2021): 718192.

[225]

S. Sadeghi, N. Mosaffa, B. Huang, and F. Ramezani Tehrani, “Protective Role of Stem Cells in POI: Current Status and Mechanism of Action, a Review Article,” Heliyon 10, no. 1 (2024): e23271.

[226]

X. Wang, T. Li, X. Bai, Y. Zhu, M. Zhang, and L. Wang, “Therapeutic Prospect on Umbilical Cord Mesenchymal Stem Cells in Animal Model With Primary Ovarian Insufficiency: A Meta-analysis,” Frontiers in Medicine 10 (2023): 1211070.

[227]

C. Guo, Y. Ma, Y. Situ, et al., “Mesenchymal Stem Cells Therapy Improves Ovarian Function in Premature Ovarian Failure: A Systematic Review and Meta-analysis Based on Preclinical Studies,” Frontiers in Endocrinology 14 (2023): 1165574.

[228]

H. K. Kim and T. J. Kim, “Current Status and Future Prospects of Stem Cell Therapy for Infertile Patients With Premature Ovarian Insufficiency,” Biomolecules 14, no. 2 (2024): 242.

[229]

K. Bahrehbar, M. Rezazadeh Valojerdi, F. Esfandiari, R. Fathi, S. N. Hassani, and H. Baharvand, “Human Embryonic Stem Cell-derived Mesenchymal Stem Cells Improved Premature Ovarian Failure,” World Journal of Stem Cells 12, no. 8 (2020): 857-878.

[230]

R. W. Rebar, “Premature Ovarian Failure,” Obstetrics and Gynecology 113, no. 6 (2009): 1355-1363.

[231]

S. Herraiz, M. Romeu, A. Buigues, et al., “Autologous Stem Cell Ovarian Transplantation to Increase Reproductive Potential in Patients Who Are Poor Responders,” Fertility and Sterility 110, no. 3 (2018): 496-505. e1.

[232]

L. Ding, G. Yan, B. Wang, et al., “Transplantation of UC-MSCs on Collagen Scaffold Activates Follicles in Dormant Ovaries of POF Patients With Long History of Infertility,” Science China Life Sciences 61, no. 12 (2018): 1554-1565.

[233]

L. Yan, Y. Wu, L. Li, et al., “Clinical Analysis of human Umbilical Cord Mesenchymal Stem Cell Allotransplantation in Patients With Premature Ovarian Insufficiency,” Cell Proliferation 53, no. 12 (2020): e12938.

[234]

S. Zafardoust, S. Kazemnejad, M. Darzi, M. Fathi-Kazerooni, H. Rastegari, and A. Mohammadzadeh, “Improvement of Pregnancy Rate and Live Birth Rate in Poor Ovarian Responders by Intraovarian Administration of Autologous Menstrual Blood Derived- Mesenchymal Stromal Cells: Phase I/II Clinical Trial,” Stem Cell Reviews and Reports 16, no. 4 (2020): 755-763.

[235]

D. D. Manavella, L. Cacciottola, S. Pomme, et al., “Two-step Transplantation With Adipose Tissue-derived Stem Cells Increases Follicle Survival by Enhancing Vascularization in Xenografted Frozen-thawed human Ovarian Tissue,” Human Reproduction 33, no. 6 (2018): 1107-1116.

[236]

E. Dreisler and J. J. Kjer, “Asherman's Syndrome: Current Perspectives on Diagnosis and Management,” International Journal of Women's Health 11 (2019): 191-198.

[237]

P. Wangikar, T. Ahmed, and S. Vangala. Chapter 76 - Toxicologic Pathology of the Reproductive System. In: Gupta RC, ed. “Reproductive and Developmental Toxicology”. (Academic Press, 2011): 1003-1026.

[238]

N. Singh, B. Shekhar, S. Mohanty, S. Kumar, T. Seth, and B. Girish, “Autologous Bone Marrow-derived Stem Cell Therapy for Asherman's Syndrome and Endometrial Atrophy: A 5-year Follow-up Study,” Journal of Human Reproductive Sciences 13, no. 1 (2020): 31.

[239]

S. Y. Lee, J. E. Shin, H. Kwon, D. H. Choi, and J. H. Kim, “Effect of Autologous Adipose-Derived Stromal Vascular Fraction Transplantation on Endometrial Regeneration in Patients of Asherman's Syndrome: A Pilot Study,” Reproductive Sciences 27, no. 2 (2020): 561-568.

[240]

H. Ma, M. Liu, Y. Li, et al., “Intrauterine Transplantation of Autologous Menstrual Blood Stem Cells Increases Endometrial Thickness and Pregnancy Potential in Patients With Refractory Intrauterine Adhesion,” Journal of Obstetrics and Gynaecology Research 46, no. 11 (2020): 2347-2355.

[241]

Y. Cao, H. Sun, H. Zhu, et al., “Allogeneic Cell Therapy Using Umbilical Cord MSCs on Collagen Scaffolds for Patients With Recurrent Uterine Adhesion: A Phase I Clinical Trial,” Stem Cell Research & Therapy 9, no. 1 (2018): 192.

[242]

Y. Zhang, L. Shi, X. Lin, et al., “Unresponsive Thin Endometrium Caused by Asherman Syndrome Treated With Umbilical Cord Mesenchymal Stem Cells on Collagen Scaffolds: A Pilot Study,” Stem Cell Research & Therapy 12, no. 1 (2021): 420.

[243]

L. Nguyen Thanh, P. T. M. Dam, H. P. Nguyen, et al., “Can Autologous Adipose-Derived Mesenchymal Stem Cell Transplantation Improve Sexual Function in People With Sexual Functional Deficiency?,” Stem Cell Reviews and Reports 17, no. 6 (2021): 2153-2163.

[244]

I. Mastrolia, E. M. Foppiani, A. Murgia, et al., “Challenges in Clinical Development of Mesenchymal Stromal/Stem Cells: Concise Review,” Stem Cells Translational Medicine 8, no. 11 (2019): 1135-1148.

[245]

A. B. Česnik and U. Švajger, “The Issue of Heterogeneity of MSC-based Advanced Therapy Medicinal Products-a Review,” Frontiers in Cell and Developmental Biology 12 (2024): 1400347.

[246]

S. Chen, B. Liang, and J. Xu, “Unveiling Heterogeneity in MSCs: Exploring Marker-based Strategies for Defining MSC Subpopulations,” Journal of Translational Medicine 22, no. 1 (2024): 459.

[247]

S. Ikehara, Grand challenges in stem cell treatments. (Frontiers Media SA, 2013): 2. biology d.

[248]

A. Musiał-Wysocka, M. Kot, and M. Majka, “The Pros and Cons of Mesenchymal Stem Cell-based Therapies,” Cell Transplantation 28, no. 7 (2019): 801-812.

[249]

G. Moll, I. Rasmusson-Duprez, L. von Bahr, et al., “Are Therapeutic human Mesenchymal Stromal Cells Compatible With human Blood?,” Stem Cells 30, no. 7 (2012): 1565-1574.

[250]

G. Moll, J. A. Ankrum, S. D. Olson, and J. A. Nolta, “Improved MSC Minimal Criteria to Maximize Patient Safety: A Call to Embrace Tissue Factor and Hemocompatibility Assessment of MSC Products,” Stem Cells Translational Medicine 11, no. 1 (2022): 2-13.

[251]

O. Repetto and V. De Re, “Coagulation and Fibrinolysis in Gastric Cancer,” Annals of the New York Academy of Sciences 1404, no. 1 (2017): 27-48.

[252]

H. Caplan, S. D. Olson, A. Kumar, et al., “Mesenchymal Stromal Cell Therapeutic Delivery: Translational Challenges to Clinical Application,” Frontiers in Immunology 10 (2019): 1645.

[253]

V. T. Hoang, D. S. Le, D. M. Hoang, et al., “Impact of Tissue Factor Expression and Administration Routes on Thrombosis Development Induced by Mesenchymal Stem/Stromal Cell Infusions: Re-evaluating the Dogma,” Stem Cell Research & Therapy 15, no. 1 (2024): 56.

[254]

S. M. Ridge, F. J. Sullivan, and S. A. Glynn, “Mesenchymal Stem Cells: Key Players in Cancer Progression,” Molecular Cancer 16, no. 1 (2017): 31.

[255]

T. Lan, M. Luo, and X. J. Wei, “Mesenchymal Stem/Stromal Cells in Cancer Therapy,” Journal of Hematology & Oncology 14 (2021): 1-16.

[256]

R. W. Y. Yeo and S. K. Lim, “Embryonic Stem Cells for Therapies-challenges and Possibilities,” Embryonic Stem Cells—Basic Biology to Bioengineering (2011).

[257]

H. Liu, S. Reiter, X. Zhou, et al., “Insight Into the Mechanisms and the Challenges on Stem Cell-based Therapies for Cerebral Ischemic Stroke,” Frontiers in Cellular Neuroscience 15 (2021): 637210.

[258]

M. Ohnuki and K. Takahashi, “Present and Future Challenges of Induced Pluripotent Stem Cells,” Philosophical Transactions of the Royal Society B: Biological Sciences 370, no. 1680 (2015): 20140367.

[259]

K. Okita and S. Yamanaka, “Induced Pluripotent Stem Cells: Opportunities and Challenges,” Philosophical Transactions of the Royal Society B: Biological Sciences 366, no. 1575 (2011): 2198-2207.

[260]

C. Zhong, M. Liu, X. Pan, and H. Zhu, “Tumorigenicity Risk of iPSCs in Vivo: Nip It in the Bud,” Precision Clinical Medicine 5, no. 1 (2022): pbac004.

[261]

Y. Liang, H. Zhang, Q.-S. Feng, et al., “The Propensity for Tumorigenesis in human Induced Pluripotent Stem Cells Is Related With Genomic Instability,” Chinese Journal of Cancer 32, no. 4 (2013): 205.

[262]

G. Liang and Y. Zhang, “Embryonic Stem Cell and Induced Pluripotent Stem Cell: An Epigenetic Perspective,” Cell Research 23, no. 1 (2013): 49-69.

[263]

M. Yoshihara, Y. Hayashizaki, and Y. Murakawa, “Genomic Instability of iPSCs: Challenges towards Their Clinical Applications,” Stem Cell Reviews and Reports 13 (2017): 7-16.

[264]

A. E. Omole, A. O. J. Fakoya, K. C. Nnawuba, and K. H. Haider, “Common Ethical Considerations of human-induced Pluripotent Stem Cell Research,” Handbook of stem cell therapy. (Springer, 2022): 1-17.

[265]

A. B. Moy, A. Kamath, S. Ternes, and J. Kamath, “The Challenges to Advancing Induced Pluripotent Stem Cell-Dependent Cell Replacement Therapy,” Medical Research Archives 11, no. 11 (2023): 4784.

[266]

Y. Tang, P. Yu, and L. J. Cheng, “Current Progress in the Derivation and Therapeutic Application of Neural Stem Cells,” Cell Death & Disease 8, no. 10 (2017): e3108-e3108.

[267]

S. Pluchino, J. A. Smith, and L. Peruzzotti-Jametti, “Promises and Limitations of Neural Stem Cell Therapies for Progressive Multiple Sclerosis,” Trends in Molecular Medicine 26, no. 10 (2020): 898-912.

[268]

C. Yue, S. Feng, Y. Chen, and N. Jing, “The Therapeutic Prospects and Challenges of human Neural Stem Cells for the Treatment of Alzheimer's Disease,” Cell Regen 11, no. 1 (2022): 28.

[269]

V. Miceli, G. Zito, M. Bulati, et al., “Different Priming Strategies Improve Distinct Therapeutic Capabilities of Mesenchymal Stromal/Stem Cells: Potential Implications for Their Clinical Use,” World Journal of Stem Cells 15, no. 5 (2023): 400.

[270]

E. Nasiri, A. Alizadeh, A. M. Roushandeh, R. Gazor, N. Hashemi-Firouzi, and Z. Golipoor, “Melatonin-pretreated Adipose-derived Mesenchymal Stem Cells Efficeintly Improved Learning, Memory, and Cognition in an Animal Model of Alzheimer's Disease,” Metabolic Brain Disease 34 (2019): 1131-1143.

[271]

M. I. Phillips and Y. L. Tang, “Genetic Modification of Stem Cells for Transplantation,” Advanced Drug Delivery Reviews 60, no. 2 (2008): 160-172.

[272]

N. C. Leite, G. C. Pelayo, and D. A. Melton, “Genetic Manipulation of Stress Pathways Can Protect Stem-cell-derived Islets From Apoptosis in Vitro,” Stem Cell Reports 17, no. 4 (2022): 766-774.

[273]

M. Kot, M. Baj-Krzyworzeka, R. Szatanek, A. Musiał-Wysocka, M. Suda-Szczurek, and M. Majka, “The Importance of HLA Assessment in “off-the-shelf” Allogeneic Mesenchymal Stem Cells Based-therapies,” International Journal of Molecular Sciences 20, no. 22 (2019): 5680.

[274]

Y. Shan, M. Zhang, E. Tao, et al., “Pharmacokinetic Characteristics of Mesenchymal Stem Cells in Translational Challenges,” Signal Transduction and Targeted Therapy 9, no. 1 (2024): 242.

[275]

S. Pellegrini, V. Zamarian, and V. Sordi, “Strategies to Improve the Safety of iPSC-derived β Cells for β Cell Replacement in Diabetes,” Transplant International 35 (2022): 10575.

[276]

S. Park, Y. Gwon, S. A. Khan, K.-J. Jang, and J. Kim, “Engineering Considerations of iPSC-based Personalized Medicine,” Biomaterials Research 27, no. 1 (2023): 67.

[277]

V. Truong, K. Viken, Z. Geng, et al., “Automating human Induced Pluripotent Stem Cell Culture and Differentiation of iPSC-derived Retinal Pigment Epithelium for Personalized Drug Testing,” Slas Technology: Translating Life Sciences Innovation 26, no. 3 (2021): 287-299.

[278]

A. Dimitri, F. Herbst, and J. A. Fraietta, “Engineering the next-generation of CAR T-cells With CRISPR-Cas9 Gene Editing,” Molecular Cancer 21, no. 1 (2022): 78.

[279]

H. Lin, Q. Li, Q. Du, et al., “Integrated Generation of Induced Pluripotent Stem Cells in a Low-cost Device,” Biomaterials 189 (2019): 23-36.

[280]

S. Yamanaka, “Pluripotent Stem Cell-based Cell Therapy—promise and Challenges,” Cell Stem Cell 27, no. 4 (2020): 523-531.

[281]

R. De Gioia, F. Biella, G. Citterio, et al., “Neural Stem Cell Transplantation for Neurodegenerative Diseases,” International Journal of Molecular Sciences 21, no. 9 (2020): 3103.

[282]

G. M. Thomsen, P. Avalos, A. A. Ma, et al., “Transplantation of Neural Progenitor Cells Expressing Glial Cell Line-derived Neurotrophic Factor Into the Motor Cortex as a Strategy to Treat Amyotrophic Lateral sclerosis,” Stem Cells 36, no. 7 (2018): 1122-1131.

[283]

M. Khazaei, C. S. Ahuja, H. Nakashima, et al., “GDNF Rescues the Fate of Neural Progenitor Grafts by Attenuating Notch Signals in the Injured Spinal Cord in Rodents,” Science Translational Medicine 12, no. 525 (2020): eaau3538.

[284]

X. Li, Z. Peng, L. Long, et al., “Wnt4-modified NSC Transplantation Promotes Functional Recovery After Spinal Cord Injury,” The FASEB Journal 34, no. 1 (2020): 82-94.

[285]

D. Park, Y.-H. Yang, D. K. Bae, et al., “Improvement of Cognitive Function and Physical Activity of Aging Mice by human Neural Stem Cells Over-expressing Choline Acetyltransferase,” Neurobiology of Aging 34, no. 11 (2013): 2639-2646.

[286]

S. Mathivanan, P. Fonseka, C. Nedeva, and I. Atukorala. New Frontiers: Extracellular Vesicles. vol 97. Springer; 2021.

[287]

C. Théry, K. W. Witwer, E. Aikawa, et al., “Minimal Information for Studies of Extracellular Vesicles 2018 (MISEV2018): A Position Statement of the International Society for Extracellular Vesicles and Update of the MISEV2014 Guidelines,” Journal of Extracellular Vesicles 7, no. 1 (2018): 1535750.

[288]

G. Berumen Sánchez, K. E. Bunn, H. H. Pua, and M. Rafat, “Extracellular Vesicles: Mediators of Intercellular Communication in Tissue Injury and Disease,” Cell Communication and Signaling 19, no. 1 (2021): 104.

[289]

L. M. Doyle and M. Z. Wang, “Overview of Extracellular Vesicles, Their Origin, Composition, Purpose, and Methods for Exosome Isolation and Analysis,” Cells 8, no. 7 (2019): 727.

[290]

S. Gandham, X. Su, J. Wood, et al., “Technologies and Standardization in Research on Extracellular Vesicles,” Trends in Biotechnology 38, no. 10 (2020): 1066-1098.

[291]

S. Gurung, D. Perocheau, L. Touramanidou, and J. Baruteau, “The Exosome Journey: From Biogenesis to Uptake and Intracellular Signalling,” Cell Communication and Signaling 19, no. 1 (2021): 47.

[292]

V. R. Minciacchi, M. R. Freeman, and D. Di Vizio, “Extracellular Vesicles in Cancer: Exosomes, Microvesicles and the Emerging Role of Large Oncosomes,” Seminars in Cell & Developmental Biology 40 (2015): 41-51.

[293]

X. Zou, Q. Lei, X. Luo, et al., “Advances in Biological Functions and Applications of Apoptotic Vesicles,” Cell Communication and Signaling 21, no. 1 (2023): 260.

[294]

M. M. Shi, Q. Y. Yang, A. Monsel, et al., “Preclinical Efficacy and Clinical Safety of Clinical-grade Nebulized Allogenic Adipose Mesenchymal Stromal Cells-derived Extracellular Vesicles,” Journal of Extracellular Vesicles 10, no. 10 (2021): e12134.

[295]

X. Xie, Q. Song, C. Dai, et al., “Clinical Safety and Efficacy of Allogenic human Adipose Mesenchymal Stromal Cells-derived Exosomes in Patients With Mild to Moderate Alzheimer's Disease: A Phase I/II Clinical Trial,” General Psychiatry 36, no. 5 (2023): e101143.

[296]

M. Akhlaghpasand, R. Tavanaei, M. Hosseinpoor, et al., “Safety and Potential Effects of Intrathecal Injection of Allogeneic human Umbilical Cord Mesenchymal Stem Cell-derived Exosomes in Complete Subacute Spinal Cord Injury: A First-in-human, Single-arm, Open-label, Phase I Clinical Trial,” Stem Cell Research & Therapy 15, no. 1 (2024): 264.

[297]

V. Sengupta, S. Sengupta, A. Lazo, P. Woods, A. Nolan, and N. Bremer, “Exosomes Derived From Bone Marrow Mesenchymal Stem Cells as Treatment for Severe COVID-19,” Stem Cells and Development 29, no. 12 (2020): 747-754.

[298]

Y. G. Zhu, M. M. Shi, A. Monsel, et al., “Nebulized Exosomes Derived From Allogenic Adipose Tissue Mesenchymal Stromal Cells in Patients With Severe COVID-19: A Pilot Study,” Stem Cell Research & Therapy 13, no. 1 (2022): 220.

[299]

M. Chu, H. Wang, L. Bian, et al., “Nebulization Therapy With Umbilical Cord Mesenchymal Stem Cell-Derived Exosomes for COVID-19 Pneumonia,” Stem Cell Reviews and Reports 18, no. 6 (2022): 2152-2163.

[300]

A. L. Lightner, V. Sengupta, S. Qian, et al., “Bone Marrow Mesenchymal Stem Cell-Derived Extracellular Vesicle Infusion for the Treatment of Respiratory Failure from COVID-19: A Randomized, Placebo-Controlled Dosing Clinical Trial,” Chest 164, no. 6 (2023): 1444-1453.

[301]

M. H. Zamanian, A. H. Norooznezhad, Z. Hosseinkhani, et al., “Human Placental Mesenchymal Stromal Cell-derived Small Extracellular Vesicles as a Treatment for Severe COVID-19: A Double-blind Randomized Controlled Clinical Trial,” Journal of Extracellular Vesicles 13, no. 7 (2024): e12492.

[302]

P. Menasche, N. K. Renault, A. Hagege, et al., “First-in-man Use of a Cardiovascular Cell-derived Secretome in Heart Failure. Case Report,” EBioMedicine 103 (2024): 105145.

[303]

J. East and M. Dordevic, “Pilot Safety Study of an Extracellular VesicleIsolate Product for Treatment ofOsteoarthritis in Combat-Related Injuries:One Year Follow up,” Genesis Scientific Journals 2, no. 2 (2021): 1-10.

[304]

H. H. Kwon, S. H. Yang, J. Lee, et al., “Combination Treatment With Human Adipose Tissue Stem Cell-derived Exosomes and Fractional CO2 Laser for Acne Scars: A 12-week Prospective, Double-blind, Randomized, Split-face Study,” Acta Dermato-Venereologica 100, no. 18 (2020): adv00310.

[305]

J. Johnson, S. Q. K. Law, M. Shojaee, et al., “First-in-human Clinical Trial of Allogeneic, Platelet-derived Extracellular Vesicles as a Potential Therapeutic for Delayed Wound Healing,” Journal of Extracellular Vesicles 12, no. 7 (2023): e12332.

[306]

G. H. Park, H. H. Kwon, J. Seok, et al., “Efficacy of Combined Treatment With human Adipose Tissue Stem Cell-derived Exosome-containing Solution and Microneedling for Facial Skin Aging: A 12-week Prospective, Randomized, Split-face Study,” Journal of Cosmetic Dermatology 22, no. 12 (2023): 3418-3426.

[307]

B. Yavuz, E. C. Mutlu, Z. Ahmed, B. Ben-Nissan, and A. Stamboulis, “Applications of Stem Cell-Derived Extracellular Vesicles in Nerve Regeneration,” International Journal of Molecular Sciences 25, no. 11 (2024): 5863.

[308]

B. Wei, M. Wei, H. Huang, T. Fan, Z. Zhang, and X. Song, “Mesenchymal Stem Cell-Derived Exosomes: A Promising Therapeutic Strategy for Age-Related Diseases,” Cell Proliferation (2024): e13795.

[309]

G. Singh, A. Mehra, S. Arora, et al., “Exosome-mediated Delivery and Regulation in Neurological Disease Progression,” International Journal of Biological Macromolecules 264, no. Pt 2 (2024): 130728.

[310]

H. Ni, S. Yang, F. Siaw-Debrah, et al., “Exosomes Derived from Bone Mesenchymal Stem Cells Ameliorate Early Inflammatory Responses Following Traumatic Brain Injury,” Frontiers in Neuroscience 13 (2019): 14.

[311]

T. R. Doeppner, J. Herz, A. Gorgens, et al., “Extracellular Vesicles Improve Post-Stroke Neuroregeneration and Prevent Postischemic Immunosuppression,” Stem Cells Translational Medicine 4, no. 10 (2015): 1131-1143.

[312]

Y. Zhang, M. Chopp, Y. Meng, et al., “Effect of Exosomes Derived From Multipluripotent Mesenchymal Stromal Cells on Functional Recovery and Neurovascular Plasticity in Rats After Traumatic Brain Injury,” Journal of Neurosurgery 122, no. 4 (2015): 856-867.

[313]

Z. Zhu, X. Zhang, H. Hao, et al., “Exosomes Derived from Umbilical Cord Mesenchymal Stem Cells Treat Cutaneous Nerve Damage and Promote Wound Healing,” Frontiers in Cellular Neuroscience 16 (2022): 913009.

[314]

H. R. Hofer and R. S. Tuan, “Secreted Trophic Factors of Mesenchymal Stem Cells Support Neurovascular and Musculoskeletal Therapies,” Stem Cell Research & Therapy 7, no. 1 (2016): 131.

[315]

K. Sankarappan and A. K. Shetty, “Promise of Mesenchymal Stem Cell-derived Extracellular Vesicles for Alleviating Subarachnoid Hemorrhage-induced Brain Dysfunction by Neuroprotective and Antiinflammatory Effects,” Brain, Behavior, & Immunity - Health 40 (2024): 100835.

[316]

X. Xiao, W. Li, D. Rong, et al., “Human Umbilical Cord Mesenchymal Stem Cells-derived Extracellular Vesicles Facilitate the Repair of Spinal Cord Injury via the miR-29b-3p/PTEN/Akt/mTOR Axis,” Cell Death Discovery 7, no. 1 (2021): 212.

[317]

X. Hu, Z. Liu, X. Zhou, et al., “Small Extracellular Vesicles Derived From Mesenchymal Stem Cell Facilitate Functional Recovery in Spinal Cord Injury by Activating Neural Stem Cells via the ERK1/2 Pathway,” Frontiers in Cellular Neuroscience 16 (2022): 954597.

[318]

W. Zhou, M. Silva, C. Feng, et al., “Exosomes Derived From human Placental Mesenchymal Stem Cells Enhanced the Recovery of Spinal Cord Injury by Activating Endogenous Neurogenesis,” Stem Cell Research & Therapy 12, no. 1 (2021): 174.

[319]

H. Wang, H. Zhao, Z. Chen, et al., “Hypoxic Bone Mesenchymal Stem Cell-Derived Exosomes Direct Schwann Cells Proliferation, Migration, and Paracrine to Accelerate Facial Nerve Regeneration via circRNA_Nkd2/miR-214-3p/MED19 Axis,” International Journal of Nanomedicine 19 (2024): 1409-1429.

[320]

J. Xie, B. Jin, D. W. Li, et al., “Effect of Laminin-binding BDNF on Induction of Recurrent Laryngeal Nerve Regeneration by miR-222 Activation of mTOR Signal Pathway,” American Journal of Translational Research 7, no. 6 (2015): 1071-1080.

[321]

J. Yang, B. Wang, Y. Wang, et al., “Exosomes Derived From Adipose Mesenchymal Stem Cells Carrying miRNA-22-3p Promote Schwann Cells Proliferation and Migration Through Downregulation of PTEN,” Disease Markers 2022 (2022): 7071877.

[322]

L. Chen, B. Xiang, X. Wang, and C. Xiang, “Exosomes Derived From human Menstrual Blood-derived Stem Cells Alleviate Fulminant Hepatic Failure,” Stem Cell Research & Therapy: 8, no. 1 (2017): 9.

[323]

Y. Pan, W. F. Tan, M. Q. Yang, J. Y. Li, and D. A. Geller, “The Therapeutic Potential of Exosomes Derived From Different Cell Sources in Liver Diseases,” American Journal of Physiology Gastrointestinal and Liver Physiology 322, no. 4 (2022): G397-G404.

[324]

L. Jiang, S. Zhang, H. Hu, et al., “Exosomes Derived From human Umbilical Cord Mesenchymal Stem Cells Alleviate Acute Liver Failure by Reducing the Activity of the NLRP3 Inflammasome in Macrophages,” Biochemical and Biophysical Research Communications 508, no. 3 (2019): 735-741.

[325]

Y. Jin, J. Wang, H. Li, et al., “Extracellular Vesicles Secreted by Human Adipose-derived Stem Cells (hASCs) Improve Survival Rate of Rats With Acute Liver Failure by Releasing lncRNA H19,” EBioMedicine 34 (2018): 231-242.

[326]

W. Jiang, Y. Tan, M. Cai, et al., “Human Umbilical Cord MSC-Derived Exosomes Suppress the Development of CCl(4)-Induced Liver Injury Through Antioxidant Effect,” Stem Cells International 2018 (2018): 6079642.

[327]

W. Lu, H. Tang, S. Li, L. Bai, and Y. Chen, “Extracellular Vesicles as Potential Biomarkers and Treatment Options for Liver Failure: A Systematic Review up to March 2022,” Frontiers in Immunology 14 (2023): 1116518.

[328]

K. Xie, L. Liu, J. Chen, and F. Liu, “Exosomal miR-1246 Derived From human Umbilical Cord Blood Mesenchymal Stem Cells Attenuates Hepatic Ischemia Reperfusion Injury by Modulating T Helper 17/Regulatory T Balance,” Iubmb Life 71, no. 12 (2019): 2020-2030.

[329]

M. Kou, L. Huang, J. Yang, et al., “Mesenchymal Stem Cell-derived Extracellular Vesicles for Immunomodulation and Regeneration: A next Generation Therapeutic Tool?,” Cell Death & Disease 13, no. 7 (2022): 580.

[330]

J. Janockova, L. Slovinska, D. Harvanova, T. Spakova, and J. Rosocha, “New Therapeutic Approaches of Mesenchymal Stem Cells-derived Exosomes,” Journal of Biomedical Science 28, no. 1 (2021): 39.

[331]

Y. Zhang, Y. Li, Q. Wang, et al., “Attenuation of Hepatic Ischemia‑Reperfusion Injury by Adipose Stem Cell‑Derived Exosome Treatment via ERK1/2 and GSK‑3beta Signaling Pathways,” International Journal of Molecular Medicine 49, no. 2 (2022): 13.

[332]

X. J. Song, L. Zhang, Q. Li, Y. Li, F. H. Ding, and X. Li, “hUCB-MSC Derived Exosomal miR-124 Promotes Rat Liver Regeneration After Partial Hepatectomy via Downregulating Foxg1,” Life Sciences 265 (2021): 118821.

[333]

J. Lee, S. R. Kim, C. Lee, et al., “Extracellular Vesicles From in Vivo Liver Tissue Accelerate Recovery of Liver Necrosis Induced by Carbon Tetrachloride,” Journal of Extracellular Vesicles 10, no. 10 (2021): e12133.

[334]

L. Zhou, H. Luo, and J. W. Lee, “Role of Extracellular Vesicles in Lung Diseases,” Chinese Medical Journal 135, no. 15 (2022): 1765-1780.

[335]

E. F. Landry, J. M. Vaughn, T. J. Vicale, and R. Mann, “Inefficient Accumulation of Low Levels of Monodispersed and Feces-associated Poliovirus in Oysters,” Applied and Environmental Microbiology 44, no. 6 (1982): 1362-1369.

[336]

Q. Hu, S. Zhang, Y. Yang, et al., “Extracellular Vesicles in the Pathogenesis and Treatment of Acute Lung Injury,” Military Medical Research 9, no. 1 (2022): 61.

[337]

N. Basalova, G. Sagaradze, M. Arbatskiy, et al., “Secretome of Mesenchymal Stromal Cells Prevents Myofibroblasts Differentiation by Transferring Fibrosis-Associated microRNAs Within Extracellular Vesicles,” Cells 9, no. 5 (2020): 1272.

[338]

K. Lv, Y. Wang, P. Lou, et al., “Extracellular Vesicles as Advanced Therapeutics for the Resolution of Organ Fibrosis: Current Progress and Future Perspectives,” Frontiers in Immunology 13 (2022): 1042983.

[339]

Q. Li, Q. Feng, H. Zhou, et al., “Mechanisms and Therapeutic Strategies of Extracellular Vesicles in Cardiovascular Diseases,” MedComm 4, no. 6 (2023): e454.

[340]

J. Zhao, X. Li, J. Hu, et al., “Mesenchymal Stromal Cell-derived Exosomes Attenuate Myocardial Ischaemia-reperfusion Injury Through miR-182-regulated Macrophage Polarization,” Cardiovascular Research 115, no. 7 (2019): 1205-1216.

[341]

R. Yue, S. Lu, Y. Luo, et al., “Mesenchymal Stem Cell-derived Exosomal microRNA-182-5p Alleviates Myocardial Ischemia/Reperfusion Injury by Targeting GSDMD in Mice,” Cell Death Discovery 8, no. 1 (2022): 202.

[342]

D. Shen and Z. He, “Mesenchymal Stem Cell-derived Exosomes Regulate the Polarization and Inflammatory Response of Macrophages via miR-21-5p to Promote Repair After Myocardial Reperfusion Injury,” Annals of Translational Medicine 9, no. 16 (2021): 1323.

[343]

C. Liang, Y. Liu, H. Xu, et al., “Exosomes of Human Umbilical Cord MSCs Protect against Hypoxia/Reoxygenation-Induced Pyroptosis of Cardiomyocytes via the miRNA-100-5p/FOXO3/NLRP3 Pathway,” Frontiers in Bioengineering and Biotechnology 8 (2020): 615850.

[344]

X. H. Sun, X. Wang, Y. Zhang, and J. Hui, “Exosomes of Bone-marrow Stromal Cells Inhibit Cardiomyocyte Apoptosis Under Ischemic and Hypoxic Conditions via miR-486-5p Targeting the PTEN/PI3K/AKT Signaling Pathway,” Thrombosis Research 177 (2019): 23-32.

[345]

K. S. Li, Y. Bai, J. Li, et al., “LncRNA HCP5 in hBMSC-derived Exosomes Alleviates Myocardial Ischemia Reperfusion Injury by Sponging miR-497 to Activate IGF1/PI3K/AKT Pathway,” International Journal of Cardiology 342 (2021): 72-81.

[346]

G. Chen, M. Wang, Z. Ruan, L. Zhu, and C. Tang, “Mesenchymal Stem Cell-derived Exosomal miR-143-3p Suppresses Myocardial Ischemia-reperfusion Injury by Regulating Autophagy,” Life Sciences 280 (2021): 119742.

[347]

X. Cui, Z. He, Z. Liang, Z. Chen, H. Wang, and J. Zhang, “Exosomes from Adipose-derived Mesenchymal Stem Cells Protect the Myocardium against Ischemia/Reperfusion Injury through Wnt/Beta-Catenin Signaling Pathway,” Journal of Cardiovascular Pharmacology 70, no. 4 (2017): 225-231.

[348]

T. C. Lai, T. L. Lee, Y. C. Chang, et al., “MicroRNA-221/222 Mediates ADSC-Exosome-Induced Cardioprotection against Ischemia/Reperfusion by Targeting PUMA and ETS-1,” Frontiers in Cell and Developmental Biology 8 (2020): 569150.

[349]

Z. Liu, Y. Xu, Y. Wan, J. Gao, Y. Chu, and J. Li, “Exosomes From Adipose-derived Mesenchymal Stem Cells Prevent Cardiomyocyte Apoptosis Induced by Oxidative Stress,” Cell Death Discovery 5 (2019): 79.

[350]

M. Khan, E. Nickoloff, T. Abramova, et al., “Embryonic Stem Cell-derived Exosomes Promote Endogenous Repair Mechanisms and Enhance Cardiac Function Following Myocardial Infarction,” Circulation Research 117, no. 1 (2015): 52-64.

[351]

Y. Wang, L. Zhang, Y. Li, et al., “Exosomes/Microvesicles From Induced Pluripotent Stem Cells Deliver Cardioprotective miRNAs and Prevent Cardiomyocyte Apoptosis in the Ischemic Myocardium,” International Journal of Cardiology 192 (2015): 61-69.

[352]

L. Zhang, Q. Wang, H. Su, and J. Cheng, “Exosomes From Adipose Derived Mesenchymal Stem Cells Alleviate Diabetic Osteoporosis in Rats Through Suppressing NLRP3 Inflammasome Activation in Osteoclasts,” Journal of Bioscience and Bioengineering 131, no. 6 (2021): 671-678.

[353]

S. Cosenza, M. Ruiz, K. Toupet, C. Jorgensen, and D. Noel, “Mesenchymal Stem Cells Derived Exosomes and Microparticles Protect Cartilage and Bone From Degradation in Osteoarthritis,” Scientific Reports 7, no. 1 (2017): 16214.

[354]

L. Zheng, Y. Wang, P. Qiu, et al., “Primary Chondrocyte Exosomes Mediate Osteoarthritis Progression by Regulating Mitochondrion and Immune Reactivity,” Nanomedicine (London) 14, no. 24 (2019): 3193-3212.

[355]

W. Li, L. Li, R. Cui, X. Chen, H. Hu, and Y. Qiu, “Bone Marrow Mesenchymal Stem Cells Derived Exosomal Lnc TUG1 Promotes Bone Fracture Recovery via miR-22-5p/Anxa8 Axis,” Human Cell 36, no. 3 (2023): 1041-1053.

[356]

X. Yang, J. Yang, P. Lei, and T. Wen, “LncRNA MALAT1 Shuttled by Bone Marrow-derived Mesenchymal Stem Cells-secreted Exosomes Alleviates Osteoporosis Through Mediating microRNA-34c/SATB2 Axis,” Aging (Albany NY) 11, no. 20 (2019): 8777-8791.

[357]

R. Z. Yang, W. N. Xu, H. L. Zheng, et al., “Exosomes Derived From Vascular Endothelial Cells Antagonize Glucocorticoid-induced Osteoporosis by Inhibiting Ferritinophagy With Resultant Limited Ferroptosis of Osteoblasts,” Journal of Cellular Physiology 236, no. 9 (2021): 6691-6705.

[358]

H. Hu, D. Wang, L. Li, H. Yin, G. He, and Y. Zhang, “Role of microRNA-335 Carried by Bone Marrow Mesenchymal Stem Cells-derived Extracellular Vesicles in Bone Fracture Recovery,” Cell Death & Disease 12, no. 2 (2021): 156.

[359]

T. Manzoor, A. Saleem, N. Farooq, et al., “Extracellular Vesicles Derived From Mesenchymal Stem Cells—a Novel Therapeutic Tool in Infectious Diseases,” Inflammation and Regeneration 43, no. 1 (2023): 17.

[360]

J.-Y. Ding, M.-J. Chen, L.-F. Wu, et al., “Mesenchymal Stem Cell-derived Extracellular Vesicles in Skin Wound Healing: Roles, Opportunities and Challenges,” Military Medical Research 10, no. 1 (2023): 36.

[361]

J. Wang, S. Yuan, Y. Tu, Z. Lv, H. Cheng, and X. Ding, “Extracellular Vesicles in Skin Health, Diseases, and Aging,” Interdisciplinary Medicine 2, no. 3 (2024): e20240011.

[362]

J. Y. Hong, T. R. Kwon, J. H. Kim, B. C. Lee, and B. J. Kim, “Prospective, Preclinical Comparison of the Performance Between Radiofrequency Microneedling and Microneedling Alone in Reversing Photoaged Skin,” Journal of Cosmetic Dermatology 19, no. 5 (2020): 1105-1109.

[363]

W. Gao, L.-M. Yuan, Y. Zhang, et al., “miR-1246-overexpressing Exosomes Suppress UVB-induced Photoaging via Regulation of TGF-β/Smad and Attenuation of MAPK/AP-1 Pathway,” Photochemical & Photobiological Sciences 22, no. 1 (2023): 135-146.

[364]

M. Deng, T. Z. Yu, D. Li, et al., “Human Umbilical Cord Mesenchymal Stem Cell-derived and Dermal Fibroblast-derived Extracellular Vesicles Protect Dermal Fibroblasts From Ultraviolet Radiation-induced Photoaging in Vitro,” Photochemical & Photobiological Sciences 19 (2020): 406-414.

[365]

T. Wang, Z. Jian, A. Baskys, et al., “MSC-derived Exosomes Protect Against Oxidative Stress-induced Skin Injury via Adaptive Regulation of the NRF2 Defense System,” Biomaterials 257 (2020): 120264.

[366]

W. Zhang, X. Bai, B. Zhao, et al., “Cell-free Therapy Based on Adipose Tissue Stem Cell-derived Exosomes Promotes Wound Healing via the PI3K/Akt Signaling Pathway,” Experimental Cell Research 370, no. 2 (2018): 333-342.

[367]

J. S. Choi, W. L. Cho, Y. J. Choi, et al., “Functional Recovery in Photo-damaged human Dermal Fibroblasts by human Adipose-derived Stem Cell Extracellular Vesicles,” Journal of Extracellular Vesicles 8, no. 1 (2019): 1565885.

[368]

Y. Shimizu, E. H. Ntege, Y. Inoue, N. Matsuura, H. Sunami, and Y. Sowa, “Optimizing Mesenchymal Stem Cell Extracellular Vesicles for Chronic Wound Healing: Bioengineering, Standardization, and Safety,” Regenerative Therapy 26 (2024): 260-274.

[369]

J. Kim, E. H. Kim, H. Lee, and J. H. Sung, “Bang OYJIJoMS. Clinical-scale Mesenchymal Stem Cell-derived Extracellular Vesicle Therapy for Wound Healing,” International Journal of Molecular Sciences 24, no. 5 (2023): 4273.

[370]

X. He, Z. Dong, Y. Cao, et al., “MSC-derived Exosome Promotes M2 Polarization and Enhances Cutaneous Wound Healing,” Stem Cells International 2019, no. 1 (2019): 7132708.

[371]

L. Pi, L. Yang, B.-R. Fang, X.-X. Meng, L. J. M. Qian, and C. Biochemistry, “Exosomal microRNA-125a-3p From human Adipose-derived Mesenchymal Stem Cells Promotes Angiogenesis of Wound Healing Through Inhibiting PTEN,” Molecular and Cellular Biochemistry 477, no. 1 (2022): 115-127.

[372]

L. Zhang, P. Ouyang, G. He, et al., “Exosomes From microRNA-126 Overexpressing Mesenchymal Stem Cells Promote Angiogenesis by Targeting the PIK3R2-mediated PI3K/Akt Signalling Pathway,” Journal of Cellular and Molecular Medicine 25, no. 4 (2021): 2148-2162.

[373]

Y. Li, J. Zhang, J. Shi, et al., “Exosomes Derived From human Adipose Mesenchymal Stem Cells Attenuate Hypertrophic Scar Fibrosis by miR-192-5p/IL-17RA/Smad Axis,” Stem Cell Research & Therapy 12 (2021): 1-16.

[374]

J. Yang, M. Xiao, K. Ma, et al., “Therapeutic Effects of Mesenchymal Stem Cells and Their Derivatives in Common Skin Inflammatory Diseases: Atopic Dermatitis and Psoriasis,” Frontiers in Immunology 14 (2023): 1092668.

[375]

J. Kim, S. K. Lee, M. Jung, et al., “Extracellular Vesicles From IFN-γ-primed Mesenchymal Stem Cells Repress Atopic Dermatitis in Mice,” Journal of Nanobiotechnology 20, no. 1 (2022): 526.

[376]

W. Zhang, J. Lin, P. Shi, et al., “Small Extracellular Vesicles Derived From MSCs Have Immunomodulatory Effects to Enhance Delivery of ASO-210 for Psoriasis Treatment,” Frontiers in Cell and Developmental Biology 10 (2022): 842813.

[377]

M. Qu, Q. Lin, L. Huang, et al., “Dopamine-loaded Blood Exosomes Targeted to Brain for Better Treatment of Parkinson's Disease,” Journal of Controlled Release 287 (2018): 156-166.

[378]

M. J. Haney, N. L. Klyachko, Y. Zhao, et al., “Exosomes as Drug Delivery Vehicles for Parkinson's Disease Therapy,” Journal of Controlled Release 207 (2015): 18-30.

[379]

Q. Chen, Y. Liu, X. Ding, et al., “Bone Marrow Mesenchymal Stem Cell-secreted Exosomes Carrying microRNA-125b Protect Against Myocardial Ischemia Reperfusion Injury via Targeting SIRT7,” Molecular and Cellular Biochemistry 465, no. 1-2 (2020): 103-114.

[380]

L. Jiang, Y. Zhang, T. Liu, et al., “Exosomes Derived From TSG-6 Modified Mesenchymal Stromal Cells Attenuate Scar Formation During Wound Healing,” Biochimie 177 (2020): 40-49.

[381]

S. C. Tao, T. Yuan, Y. L. Zhang, W. J. Yin, S. C. Guo, and C. Q. Zhang, “Exosomes Derived From miR-140-5p-overexpressing human Synovial Mesenchymal Stem Cells Enhance Cartilage Tissue Regeneration and Prevent Osteoarthritis of the Knee in a Rat Model,” Theranostics 7, no. 1 (2017): 180-195.

[382]

Z. Erana-Perez, M. Igartua, E. Santos-Vizcaino, and R. M. Hernandez, “Genetically Engineered Loaded Extracellular Vesicles for Drug Delivery,” Trends in Pharmacological Sciences 45, no. 4 (2024): 350-365.

[383]

Q. Liu, D. Li, X. Pan, and Y. Liang, “Targeted Therapy Using Engineered Extracellular Vesicles: Principles and Strategies for Membrane Modification,” Journal of Nanobiotechnology 21, no. 1 (2023): 334.

[384]

L. Alvarez-Erviti, Y. Seow, H. Yin, C. Betts, S. Lakhal, and M. J. A. Wood, “Delivery of siRNA to the Mouse Brain by Systemic Injection of Targeted Exosomes,” Nature Biotechnology 29, no. 4 (2011): 341-345.

[385]

M. Kim, G. Kim, D. W. Hwang, and M. Lee, “Delivery of High Mobility Group Box-1 siRNA Using Brain-Targeting Exosomes for Ischemic Stroke Therapy,” Journal of Biomedical Nanotechnology 15, no. 12 (2019): 2401-2412.

[386]

J. Yang, X. Zhang, X. Chen, L. Wang, and G. Yang, “Exosome Mediated Delivery of miR-124 Promotes Neurogenesis After Ischemia,” Molecular Therapy Nucleic Acids 7 (2017): 278-287.

[387]

J. Yang, S. Wu, L. Hou, et al., “Therapeutic Effects of Simultaneous Delivery of Nerve Growth Factor mRNA and Protein via Exosomes on Cerebral Ischemia,” Molecular Therapy Nucleic Acids 21 (2020): 512-522.

[388]

L. Yang, B. Han, Z. Zhang, et al., “Extracellular Vesicle-Mediated Delivery of Circular RNA SCMH1 Promotes Functional Recovery in Rodent and Nonhuman Primate Ischemic Stroke Models,” Circulation 142, no. 6 (2020): 556-574.

[389]

R. Kojima, D. Bojar, G. Rizzi, et al., “Designer Exosomes Produced by Implanted Cells Intracerebrally Deliver Therapeutic Cargo for Parkinson's Disease Treatment,” Nature Communications 9, no. 1 (2018): 1305.

[390]

X. Ren, Y. Zhao, F. Xue, et al., “Exosomal DNA Aptamer Targeting α-Synuclein Aggregates Reduced Neuropathological Deficits in a Mouse Parkinson's Disease Model,” Molecular Therapy Nucleic Acids 17 (2019): 726-740.

[391]

Y. Liang, X. Xu, X. Li, et al., “Chondrocyte-Targeted MicroRNA Delivery by Engineered Exosomes Toward a Cell-Free Osteoarthritis Therapy,” ACS Applied Materials and Interfaces Journal 12, no. 33 (2020): 36938-36947.

[392]

X. Xu, Y. Liang, X. Li, et al., “Exosome-mediated Delivery of Kartogenin for Chondrogenesis of Synovial Fluid-derived Mesenchymal Stem Cells and Cartilage Regeneration,” Biomaterials 269 (2021): 120539.

[393]

Y. Lin, M. Yan, Z. Bai, et al., “Huc-MSC-derived Exosomes Modified With the Targeting Peptide of aHSCs for Liver Fibrosis Therapy,” Journal of Nanobiotechnology 20, no. 1 (2022): 432.

[394]

R. H. Fang, C. M. Hu, K. N. Chen, et al., “Lipid-insertion Enables Targeting Functionalization of Erythrocyte Membrane-cloaked Nanoparticles,” Nanoscale 5, no. 19 (2013): 8884-8888.

[395]

G.-H. Cui, H.-D. Guo, H. Li, et al., “RVG-modified Exosomes Derived From Mesenchymal Stem Cells Rescue Memory Deficits by Regulating Inflammatory Responses in a Mouse Model of Alzheimer's Disease,” Immunity & Ageing 16, no. 1 (2019): 10.

[396]

J. Che, C. I. Okeke, Z. B. Hu, and J. Xu, “DSPE-PEG: A Distinctive Component in Drug Delivery System,” Current Pharmaceutical Design 21, no. 12 (2015): 1598-1605.

[397]

D. G. You, B. H. Oh, V. Q. Nguyen, et al., “Vitamin A-coupled Stem Cell-derived Extracellular Vesicles Regulate the Fibrotic Cascade by Targeting Activated Hepatic Stellate Cells in Vivo,” Journal of Controlled Release 336 (2021): 285-295.

[398]

T. Tian, H. X. Zhang, C. P. He, et al., “Surface Functionalized Exosomes as Targeted Drug Delivery Vehicles for Cerebral Ischemia Therapy,” Biomaterials 150 (2018): 137-149.

[399]

H. Zhang, J. Wu, J. Wu, et al., “Exosome-mediated Targeted Delivery of miR-210 for Angiogenic Therapy After Cerebral Ischemia in Mice,” Journal of Nanobiotechnology 17, no. 1 (2019): 29.

[400]

A. H. Berggreen, J. L. Petersen, L. Lin, K. Benabdellah, and Y. Luo, “CRISPR Delivery With Extracellular Vesicles: Promises and Challenges,” Journal of Extracellular Biology 2, no. 9 (2023): e111.

[401]

L. You, R. Tong, M. Li, Y. Liu, J. Xue, and Y. Lu, “Advancements and Obstacles of CRISPR-Cas9 Technology in Translational Research,” Molecular Therapy Methods & Clinical Development 13 (2019): 359-370.

[402]

B. Yan and Y. Liang, “New Therapeutics for Extracellular Vesicles: Delivering CRISPR for Cancer Treatment,” International Journal of Molecular Sciences 23, no. 24 (2022): 15758.

[403]

D. Lainšček, L. Kadunc, M. M. Keber, I. H. Bratkovič, R. Romih, and R. Jerala, “Delivery of an Artificial Transcription Regulator dCas9-VPR by Extracellular Vesicles for Therapeutic Gene Activation,” ACS Synthetic Biology 7, no. 12 (2018): 2715-2725.

[404]

T. Wan, J. Zhong, Q. Pan, T. Zhou, Y. Ping, and X. Liu, “Exosome-mediated Delivery of Cas9 ribonucleoprotein Complexes for Tissue-specific Gene Therapy of Liver Diseases,” Science Advances 8, no. 37 (2022): eabp9435.

[405]

N. Majeau, A. Fortin-Archambault, C. Gérard, J. Rousseau, P. Yaméogo, and J. P. Tremblay, “Serum Extracellular Vesicles for Delivery of CRISPR-CAS9 Ribonucleoproteins to Modify the Dystrophin Gene,” Molecular Therapy 30, no. 7 (2022): 2429-2442.

[406]

B. Wang, M. Chang, R. Zhang, et al., “Spinal Cord Injury Target-immunotherapy With TNF-α Autoregulated and Feedback-controlled human Umbilical Cord Mesenchymal Stem Cell Derived Exosomes Remodelled by CRISPR/Cas9 Plasmid,” Biomaterials Advances 133 (2022): 112624.

[407]

P. Gee, M. S. Y. Lung, Y. Okuzaki, et al., “Extracellular Nanovesicles for Packaging of CRISPR-Cas9 Protein and sgRNA to Induce Therapeutic Exon Skipping,” Nature Communications 11, no. 1 (2020): 1334.

[408]

X. Osteikoetxea, A. Silva, E. Lázaro-Ibáñez, et al., “Engineered Cas9 Extracellular Vesicles as a Novel Gene Editing Tool,” Journal of Extracellular Vesicles 11, no. 5 (2022): e12225.

[409]

Y. Yuana, A. Sturk, and R. Nieuwland, “Extracellular Vesicles in Physiological and Pathological Conditions,” Blood Reviews 27, no. 1 (2013): 31-39.

[410]

M. A. Kumar, S. K. Baba, H. Q. Sadida, et al., “Extracellular Vesicles as Tools and Targets in Therapy for Diseases,” Signal Transduction and Targeted Therapy 9, no. 1 (2024): 27.

[411]

M. Catalano and L. O'Driscoll, “Inhibiting Extracellular Vesicles Formation and Release: A Review of EV Inhibitors,” Journal of Extracellular Vesicles 9, no. 1 (2020): 1703244.

[412]

K. Trajkovic, C. Hsu, S. Chiantia, et al., “Ceramide Triggers Budding of Exosome Vesicles Into Multivesicular Endosomes,” Science 319, no. 5867 (2008): 1244-1247.

[413]

M. B. Dinkins, S. Dasgupta, G. Wang, G. Zhu, and E. Bieberich, “Exosome Reduction in Vivo Is Associated With Lower Amyloid Plaque Load in the 5XFAD Mouse Model of Alzheimer's Disease,” Neurobiology of Aging 35, no. 8 (2014): 1792-1800.

[414]

K. Essandoh, L. Yang, X. Wang, et al., “Blockade of Exosome Generation With GW4869 Dampens the Sepsis-induced Inflammation and Cardiac Dysfunction,” Biochimica Et Biophysica Acta 1852, no. 11 (2015): 2362-2371.

[415]

L. Lyu, H. Wang, B. Li, et al., “A Critical Role of Cardiac Fibroblast-derived Exosomes in Activating Renin Angiotensin System in Cardiomyocytes,” Journal of Molecular and Cellular Cardiology 89, no. Pt B (2015): 268-279.

[416]

H. Dai, W. Zheng, J. Luo, et al., “Inhibiting Uptake of Extracellular Vesicles Derived From Senescent Bone Marrow Mesenchymal Stem Cells by Muscle Satellite Cells Attenuates Sarcopenia,” Journal of Orthopaedic Translation 35 (2022): 23-36.

[417]

E. Eggenhofer, V. Benseler, A. Kroemer, et al., “Mesenchymal Stem Cells Are Short-lived and Do Not Migrate Beyond the Lungs After Intravenous Infusion,” Frontiers in Immunology 3 (2012): 297.

[418]

M. Sanchez-Diaz, M. I. Quinones-Vico, R. Sanabria de la Torre, et al., “Biodistribution of Mesenchymal Stromal Cells After Administration in Animal Models and Humans: A Systematic Review,” Journal of Clinical Medicine 10, no. 13 (2021): 2925.

[419]

B. Zeng, Y. Li, J. Xia, et al., “Micro Trojan Horses: Engineering Extracellular Vesicles Crossing Biological Barriers for Drug Delivery,” Bioengineering & Translational Medicine 9, no. 2 (2024): e10623.

[420]

D. H. Kim, V. K. Kothandan, H. W. Kim, et al., “Noninvasive Assessment of Exosome Pharmacokinetics in Vivo: A Review,” Pharmaceutics 11, no. 12 (2019): 649.

[421]

M. Van Delen, J. Derdelinckx, K. Wouters, I. Nelissen, and N. Cools, “A Systematic Review and Meta-analysis of Clinical Trials Assessing Safety and Efficacy of human Extracellular Vesicle-based Therapy,” Journal of Extracellular Vesicles 13, no. 7 (2024): e12458.

[422]

H. M. Ramos-Zaldívar, I. Polakovicova, E. Salas-Huenuleo, et al., “Extracellular Vesicles Through the Blood-brain Barrier: A Review,” Fluids and Barriers of the CNS 19, no. 1 (2022): 60.

[423]

W. A. Banks, P. Sharma, K. M. Bullock, K. M. Hansen, N. Ludwig, and T. L. Whiteside, “Transport of Extracellular Vesicles Across the Blood-Brain Barrier: Brain Pharmacokinetics and Effects of Inflammation,” International Journal of Molecular Sciences 21, no. 12 (2020): 4407.

[424]

I. K. Herrmann, M. J. A. Wood, and G. Fuhrmann, “Extracellular Vesicles as a next-generation Drug Delivery Platform,” Nature Nanotechnology 16, no. 7 (2021): 748-759.

[425]

D. T. Harris, “Long-term Frozen Storage of Stem Cells: Challenges and Solutions,” Journal of Biorepository Science for Applied Medicine (2016): 9-20.

[426]

S. Bruno, C. Pasquino, M. B. Herrera Sanchez, et al., “HLSC-Derived Extracellular Vesicles Attenuate Liver Fibrosis and Inflammation in a Murine Model of Non-alcoholic Steatohepatitis,” Molecular Therapy 28, no. 2 (2020): 479-489.

[427]

H. S. Han, H. Lee, D. You, et al., “Human Adipose Stem Cell-derived Extracellular Nanovesicles for Treatment of Chronic Liver Fibrosis,” Journal of Controlled Release 320 (2020): 328-336.

[428]

Z. Yan, T. Zhang, Y. Wang, S. Xiao, and J. Gao, “Extracellular Vesicle Biopotentiated Hydrogels for Diabetic Wound Healing: The Art of Living Nanomaterials Combined With Soft Scaffolds,” Materials Today Bio 23 (2023): 100810.

[429]

X. Geng, Y. Qi, X. Liu, Y. Shi, H. Li, and L. Zhao, “A Multifunctional Antibacterial and Self-healing Hydrogel Laden With Bone Marrow Mesenchymal Stem Cell-derived Exosomes for Accelerating Diabetic Wound Healing,” Biomaterials Advances 133 (2022): 112613.

[430]

P. C. Dinh, D. Paudel, H. Brochu, et al., “Inhalation of Lung Spheroid Cell Secretome and Exosomes Promotes Lung Repair in Pulmonary Fibrosis,” Nature Communications 11, no. 1 (2020): 1064.

[431]

K. Zhang and K. Cheng, “Stem Cell-derived Exosome versus Stem Cell Therapy,” Nature Reviews Bioengineering 1, no. 9 (2023): 608-609.

[432]

S. Hua, M. B. C. de Matos, J. M. Metselaar, and G. Storm, “Current Trends and Challenges in the Clinical Translation of Nanoparticulate Nanomedicines: Pathways for Translational Development and Commercialization,” Frontiers in Pharmacology 9 (2018): 790.

[433]

K. Cheng and R. Kalluri, “Guidelines for Clinical Translation and Commercialization of Extracellular Vesicles and Exosomes Based Therapeutics,” Extracellular Vesicle 2 (2023): 100029.

[434]

T. Driedonks, L. Jiang, B. Carlson, et al., “Pharmacokinetics and Biodistribution of Extracellular Vesicles Administered Intravenously and Intranasally to Macaca nemestrina,” Journal of Extracellular Biology 1, no. 10 (2022): e59.

[435]

V. D. Bui, J. Jeon, V. H. Duong, et al., “Chondroitin Sulfate-based Microneedles for Transdermal Delivery of Stem Cell-derived Extracellular Vesicles to Treat Rheumatoid Arthritis,” Journal of Controlled Release 375 (2024): 105-115.

[436]

V. D. Bui, S. Son, W. Xavier, et al., “Dissolving Microneedles for Long-term Storage and Transdermal Delivery of Extracellular Vesicles,” Biomaterials 287 (2022): 121644.

[437]

C. K. Wang, T. H. Tsai, and C. H. Lee, “Regulation of Exosomes as Biologic Medicines: Regulatory Challenges Faced in Exosome Development and Manufacturing Processes,” Clinical and Translational Science 17, no. 8 (2024): e13904.

[438]

S. Fripont, C. Marneffe, M. Marino, M. Y. Rincon, and M. G. Holt, “Production, Purification, and Quality Control for Adeno-associated Virus-based Vectors,” Journal of Visualized Experiments: JoVE no. 143 (2019).

[439]

S. J. Shepherd, X. Han, A. J. Mukalel, et al., “Throughput-scalable Manufacturing of SARS-CoV-2 mRNA Lipid Nanoparticle Vaccines,” Proceedings of the National Academy of Sciences of the United States of America 120, no. 33 (2023): e2303567120.

[440]

E. E. Kepplinger, “FDA's Expedited Approval Mechanisms for New Drug Products,” Biotechnology Law Report 34, no. 1 (2015): 15-37.

[441]

P. Pakter, “Rare Disease Care in Europe-Gaping Unmet Needs,” Rare 2 (2024): 100018.

[442]

A. E. Eldeeb, S. Salah, and N. A. Elkasabgy, “Biomaterials for Tissue Engineering Applications and Current Updates in the Field: A Comprehensive Review,” Aaps Pharmscitech [Electronic Resource] 23, no. 7 (2022): 267.

[443]

M. J. Evans and M. H. Kaufman, “Establishment in Culture of Pluripotential Cells From Mouse Embryos,” Nature 292, no. 5819 (1981): 154-156.

[444]

L. V. Thomas. Polymeric Biomaterials in Tissue Engineering: Retrospect and Prospects. In: B. Bhaskar, P. Sreenivasa Rao, N. Kasoju, V. Nagarjuna, R. R. Baadhe, eds. “Biomaterials in Tissue Engineering and Regenerative Medicine: From Basic Concepts to State of the Art Approaches” (Singapore: Springer, 2021): 89-118.

[445]

G. G. Gallico, N. E. O'Connor, C. C. Compton, O. Kehinde, and H. Green, “Permanent Coverage of Large Burn Wounds With Autologous Cultured human Epithelium,” New England Journal of Medicine 311, no. 7 (1984): 448-451.

[446]

S. V. Murphy and A. Atala, “3D bioprinting of Tissues and Organs,” Nature Biotechnology 32, no. 8 (2014): 773-785.

[447]

P. S. Gungor-Ozkerim, I. Inci, Y. S. Zhang, A. Khademhosseini, and M. R. Dokmeci, “Bioinks for 3D Bioprinting: An Overview,” Biomaterials Science 6, no. 5 (2018): 915-946.

[448]

H.-W. Kang, S. J. Lee, I. K. Ko, C. Kengla, J. J. Yoo, and A. Atala, “A 3D Bioprinting System to Produce human-scale Tissue Constructs With Structural Integrity,” Nature Biotechnology 34, no. 3 (2016): 312-319.

[449]

S. M. Riha, M. Maarof, and M. B. Fauzi, “Synergistic Effect of Biomaterial and Stem Cell for Skin Tissue Engineering in Cutaneous Wound Healing: A Concise Review,” Polymers (Basel) 13, no. 10 (2021): 1546.

[450]

T. Kitsuka, F. Takahashi, J. Reinhardt, et al., “Advances in Cardiac Tissue Engineering,” Bioengineering 9, no. 11 (2022): 696.

[451]

R. Ramli, M. Reddy, and N. Oliver, “Artificial Pancreas: Current Progress and Future Outlook in the Treatment of Type 1 Diabetes,” Drugs 79, no. 10 (2019): 1089-1101.

[452]

M. A. Lancaster and J. A. Knoblich, “Generation of Cerebral Organoids From human Pluripotent Stem Cells,” Nature Protocols 9, no. 10 (2014): 2329-2340.

[453]

H. Clevers, “Modeling Development and Disease With Organoids,” Cell 165, no. 7 (2016): 1586-1597.

[454]

Y. Kim, H. Ko, I. K. Kwon, and K. Shin, “Extracellular Matrix Revisited: Roles in Tissue Engineering,” International Neurourology Journal 20, no. Suppl 1 (2016): S23-S29.

[455]

M. M. Farag, “Recent Trends on Biomaterials for Tissue Regeneration Applications: Review,” Journal of Materials Science 58, no. 2 (2023): 527-558.

[456]

N. Kasula, P. Dhokare, A. Bhattacharyya, and I. Noh, “Recent Advances in 3D Bioprinting of Polysaccharide-based Bioinks for Fabrication of Bioengineered Tissues,” Molecular Systems Design & Engineering 9 (2024): 977-999.

[457]

P. S. Gungor-Ozkerim, I. Inci, Y. S. Zhang, A. Khademhosseini, and M. R. Dokmeci, “Bioinks for 3D Bioprinting: An Overview,” Biomaterials Science 6, no. 5 (2018): 915-946.

[458]

E. Isaeva, E. Beketov, N. Arguchinskaya, S. Ivanov, P. Shegay, and А. Kaprin, “Decellularized Extracellular Matrix for Tissue Engineering,” Современные технологии в медицине 14, no. 3 (2022): 57-68.

[459]

M. Zhe, X. Wu, P. Yu, et al., “Recent Advances in Decellularized Extracellular Matrix-based Bioinks for 3D Bioprinting in Tissue Engineering,” Materials 16, no. 8 (2023): 3197.

[460]

S. Dabrowska, A. Andrzejewska, M. Janowski, and B. Lukomska, “Immunomodulatory and Regenerative Effects of Mesenchymal Stem Cells and Extracellular Vesicles: Therapeutic Outlook for Inflammatory and Degenerative Diseases,” Frontiers in Immunology 11 (2021): 591065.

[461]

R. Ju, X. Gao, C. Zhang, W. Tang, W. Tian, and M. He, “Exogenous MSC Based Tissue Regeneration: A Review of Immuno-protection Strategies From Biomaterial Scaffolds,” Journal of Materials Chemistry B 12, no. 36 (2024): 8868-8882.

[462]

D. Zujur, Z. Al-Akashi, A. Nakamura, et al., “Enhanced Chondrogenic Differentiation of iPS Cell-derived Mesenchymal Stem/Stromal Cells via Neural Crest Cell Induction for Hyaline Cartilage Repair. Original Research,” Frontiers in Cell and Developmental Biology 11 (2023): 1140717.

[463]

M. Dottori, W.-J. Li, G. Minchiotti, A. Rosa, and F. Sangiuolo, “Editorial: Reviews in Induced Pluripotent Stem Cells,” Frontiers in Cell and Developmental Biology 11 (2023): 1197891.

[464]

Z. Liu, J. Zhou, H. Wang, M. Zhao, and C. Wang, “Current Status of Induced Pluripotent Stem Cells in Cardiac Tissue Regeneration and Engineering,” Regenerative Medicine Research 1, no. 1 (2013): 6.

[465]

Y. Yang, B. Ma, J. Chen, et al., “Epigenetic Regulation and Factors That Influence the Effect of iPSCs-derived Neural Stem/Progenitor Cells (NS/PCs) in the Treatment of Spinal Cord Injury,” Clinical Epigenetics 16, no. 1 (2024): 30.

[466]

S. G. Ozcebe, M. Tristan, and P. Zorlutuna, “Adult Human Heart ECM Improves Human iPSC-CM Function via Mitochondrial and Metabolic Maturation,” BioRxiv (2023), 2023.10.31.565062.

[467]

S. Rashidi, G. Bagherpour, Z. Abbasi-Malati, et al., “Endothelial Progenitor Cells for Fabrication of Engineered Vascular Units and Angiogenesis Induction,” Cell Proliferation 57, no. 9 (2024): e13716.

[468]

S. Ribeiro, A. Watigny, Y. Bayon, M. Biggs, and D. Zeugolis, “It Takes Two to Tango: Controlling Human Mesenchymal Stromal Cell Response via Substrate Stiffness and Surface Topography,” Advanced NanoBiomed Research 4 (2023): 2300042.

[469]

Q. Zhang, Y. Hu, X. Long, et al., “Preparation and Application of Decellularized ECM-Based Biological Scaffolds for Articular Cartilage Repair: A Review,” Frontiers in Bioengineering and Biotechnology 10 (2022): 908082.

[470]

Y.-H. Kim, S. Vijayavenkataraman, and G. Cidonio, “Biomaterials and Scaffolds for Tissue Engineering and Regenerative Medicine,” BMC Methods 1, no. 1 (2024): 2.

[471]

F. Han, Q. Meng, E. Xie, et al., “Engineered Biomimetic Micro/Nano-materials for Tissue Regeneration,” Frontiers in Bioengineering and Biotechnology 11 (2023): 1205792.

[472]

H.-C. Yan, T.-T. Yu, J. Li, et al., “The Delivery of Extracellular Vesicles Loaded in Biomaterial Scaffolds for Bone Regeneration,” Frontiers in Bioengineering and Biotechnology 8 (2020): 1015.

[473]

D. N. Tavakol, S. Fleischer, T. Falcucci, et al., “Emerging Trajectories for Next Generation Tissue Engineers,” ACS Biomaterials Science & Engineering 8, no. 11 (2022): 4598-4604.

[474]

R. Langer, “Chemical and Biological Approaches to Regenerative Medicine and Tissue Engineering,” Molecular Frontiers Journal 03, no. 02 (2019): 122-128.

[475]

L. E. Niklason, J. Gao, W. M. Abbott, et al., “Functional Arteries Grown in Vitro,” Science 284, no. 5413 (1999): 489-493.

[476]

J. H. Lawson, M. H. Glickman, M. Ilzecki, et al., “Bioengineered human Acellular Vessels for Dialysis Access in Patients With End-stage Renal Disease: Two Phase 2 Single-arm Trials,” The Lancet 387, no. 10032 (2016): 2026-2034.

[477]

J. R. Slotkin, C. D. Pritchard, B. Luque, et al., “Biodegradable Scaffolds Promote Tissue Remodeling and Functional Improvement in Non-human Primates With Acute Spinal Cord Injury,” Biomaterials 123 (2017): 63-76.

[478]

Y. D. Teng, E. B. Lavik, X. Qu, et al., “Functional Recovery Following Traumatic Spinal Cord Injury Mediated by a Unique Polymer Scaffold Seeded With Neural Stem Cells,” Proceedings of the National Academy of Sciences of the United States of America 99, no. 5 (2002): 3024-3029.

[479]

S. Sabetkish, P. Currie, and L. Meagher, “Recent Trends in 3D Bioprinting Technology for Skeletal Muscle Regeneration,” Acta Biomaterialia 181: 46-66.

[480]

M. Asadian, K. V. Chan, M. Norouzi, et al., “Fabrication and Plasma Modification of Nanofibrous Tissue Engineering Scaffolds,” Nanomaterials 10, no. 1 (2020): 119.

[481]

O. Omar, T. Engstrand, L. Kihlström Burenstam Linder, et al., “In Situ Bone Regeneration of Large Cranial Defects Using Synthetic Ceramic Implants With a Tailored Composition and Design,” Proceedings of the National Academy of Sciences of the United States of America 117, no. 43 (2020): 26660-26671.

[482]

X. Dou, X. Wei, G. Liu, et al., “Effect of Porous Tantalum on Promoting the Osteogenic Differentiation of Bone Marrow Mesenchymal Stem Cells in Vitro Through the MAPK/ERK Signal Pathway,” Journal of Orthopaedic Translation 19 (2019): 81-93.

[483]

A. R. Edelmann, D. Patel, R. K. Allen, C. J. Gibson, A. M. Best, and S. Bencharit, “Retrospective Analysis of Porous Tantalum Trabecular Metal-enhanced Titanium Dental Implants,” The Journal of Prosthetic Dentistry 121, no. 3 (2019): 404-410.

[484]

F. Re, E. Borsani, R. Rezzani, L. Sartore, and D. Russo, “Bone Regeneration Using Mesenchymal Stromal Cells and Biocompatible Scaffolds: A Concise Review of the Current Clinical Trials,” Gels 9, no. 5 (2023): 389.

[485]

S. Shakir, T. L. Hackett, and L. B. Mostaço-Guidolin, “Bioengineering Lungs: An Overview of Current Methods, Requirements, and Challenges for Constructing Scaffolds,” Frontiers in Bioengineering and Biotechnology 10 (2022): 1011800.

[486]

W. L. Ng, T. C. Ayi, Y. C. Liu, S. L. Sing, W. Y. Yeong, and B. H. Tan, “Fabrication and Characterization of 3D Bioprinted Triple-layered Human Alveolar Lung Models,” International Journal of Bioprinting 7, no. 2 (2021): 332.

[487]

A. Neishabouri, A. Soltani Khaboushan, F. Daghigh, A.-M. Kajbafzadeh, and M. Majidi Zolbin, “Decellularization in Tissue Engineering and Regenerative Medicine: Evaluation, Modification, and Application Methods,” Frontiers in Bioengineering and Biotechnology 10 (2022): 805299.

[488]

Y. H. Song, M. A. Maynes, N. Hlavac, et al., “Development of Novel Apoptosis-assisted Lung Tissue Decellularization Methods,” Biomaterials Science 9, no. 9 (2021): 3485-3498.

[489]

S. Alsobaie, T. Alsobaie, A. Alshammary, and S. Mantalaris, “Differentiation of human Induced Pluripotent Stem Cells Into Functional Lung Alveolar Epithelial Cells in 3D Dynamic Culture,” Frontiers in Bioengineering and Biotechnology 11 (2023): 1173149.

[490]

W. Wruck, N. Graffmann, L.-S. Spitzhorn, and J. Adjaye, “Human Induced Pluripotent Stem Cell-Derived Mesenchymal Stem Cells Acquire Rejuvenation and Reduced Heterogeneity,” Frontiers in Cell and Developmental Biology 9 (2021): 717772.

[491]

J. Li, X. Chen, M. Hu, et al., “The Application of Composite Scaffold Materials Based on Decellularized Vascular Matrix in Tissue Engineering: A Review,” BioMedical Engineering OnLine 22, no. 1 (2023): 62.

[492]

A. H. Nguyen, P. Marsh, L. Schmiess-Heine, et al., “Cardiac Tissue Engineering: State-of-the-art Methods and Outlook,” Journal of Biological Engineering 13, no. 1 (2019): 57.

[493]

M. M. Rana and H. De la Hoz Siegler, “Evolution of Hybrid Hydrogels: Next-Generation Biomaterials for Drug Delivery and Tissue Engineering,” Gels 10, no. 4 (2024): 216.

[494]

M. M. Peters, J. K. Brister, E. M. Tang, et al., “Self-organizing Behaviors of Cardiovascular Cells on Synthetic Nanofiber Scaffolds,” APL Bioengineering 7, no. 4 (2023): 046114.

[495]

W. LaBarge, S. Mattappally, R. Kannappan, et al., “Maturation of Three-dimensional, hiPSC-derived Cardiomyocyte Spheroids Utilizing Cyclic, Uniaxial Stretch and Electrical Stimulation,” PLoS One 14, no. 7 (2019): e0219442.

[496]

K. Zheng, Y. Hao, C. Xia, et al., “Effects and Mechanisms of the Myocardial Microenvironment on Cardiomyocyte Proliferation and Regeneration,” Frontiers in Cell and Developmental Biology 12 (2024): 1429020.

[497]

Z. S. Razavi, M. Soltani, G. Mahmoudvand, et al., “Advancements in Tissue Engineering for Cardiovascular Health: A Biomedical Engineering Perspective,” Frontiers in Bioengineering and Biotechnology 12 (2024): 1385124.

[498]

A. Akbarzadeh, S. Sobhani, A. Soltani Khaboushan, and A.-M. Kajbafzadeh, “Whole-Heart Tissue Engineering and Cardiac Patches: Challenges and Promises,” Bioengineering 10, no. 1 (2023): 106.

[499]

P. Li, J. Hu, J. Wang, J. Zhang, L. Wang, and C. Zhang, “The Role of Hydrogel in Cardiac Repair and Regeneration for Myocardial Infarction: Recent Advances and Future Perspectives,” Bioengineering 10, no. 2 (2023): 165.

[500]

B. Sanders, S. Loerakker, E. S. Fioretta, et al., “Improved Geometry of Decellularized Tissue Engineered Heart Valves to Prevent Leaflet Retraction,” Annals of Biomedical Engineering 44, no. 4 (2016): 1061-1071.

[501]

P. E. Dijkman, A. Driessen-Mol, L. Frese, S. P. Hoerstrup, and F. P. T. Baaijens, “Decellularized Homologous Tissue-engineered Heart Valves as off-the-shelf Alternatives to Xeno- and Homografts,” Biomaterials 33, no. 18 (2012): 4545-4554.

[502]

P. Mohacsi and P. Leprince, “The CARMAT Total Artificial Heart,” European Journal of Cardio-Thoracic Surgery 46, no. 6 (2014): 933-934.

[503]

W. C. DeVries, J. L. Anderson, L. D. Joyce, et al., “Clinical Use of the Total Artificial Heart,” New England Journal of Medicine 310, no. 5 (1984): 273-278.

[504]

G. Torregrossa, A. Anyanwu, F. Zucchetta, and G. Gerosa, “SynCardia: The Total Artificial Heart,” Annals of Cardiothoracic Surgery 3, no. 6 (2014): 612-620.

[505]

G. Torregrossa, M. Morshuis, R. Varghese, et al., “Results with Syncardia Total Artificial Heart beyond 1 Year,” ASAIO Journal 60, no. 6 (2014): 626-634.

[506]

N. Stafford, “Leonard L Bailey: In 1984 he Transplanted a Baboon Heart Into a human Infant Known as “Baby Fae”,” Bmj 366 (2019): l4669.

[507]

L. L. Bailey, S. L. Nehlsen-Cannarella, W. Concepcion, and W. B. Jolley, “Baboon-to-Human Cardiac Xenotransplantation in a Neonate,” Jama 254, no. 23 (1985): 3321-3329.

[508]

M. Mohiuddin, Tiny Matters. (Pig hearts in people: Xenotransplantation's long history, current promise, and the ethical use of brain-dead people in research, 2024). August 21.

[509]

L. Peterson, M. H. Yacoub, D. Ayares, et al., “Physiological Basis for Xenotransplantation From Genetically Modified Pigs to Humans,” Physiological Reviews 104, no. 3 (2024): 1409-1459.

[510]

D. B. Joseph, J. G. Borer, R. E. De Filippo, S. J. Hodges, and G. A. McLorie, “Autologous Cell Seeded Biodegradable Scaffold for Augmentation Cystoplasty: Phase II Study in Children and Adolescents With Spina Bifida,” The Journal of Urology 191, no. 5 (2014): 1389-1395.

[511]

B. Jeon, C. Lee, M. Kim, T. H. Choi, S. Kim, and S. Kim, “Fabrication of Three-dimensional Scan-to-print Ear Model for Microtia Reconstruction,” Journal of Surgical Research 206, no. 2 (2016): 490-497.

[512]

M. S. Mannoor, Z. Jiang, T. James, et al., “3D Printed Bionic Ears,” Nano Letters 13, no. 6 (2013): 2634-2639.

[513]

X. Xie, S. Wu, S. Mou, N. Guo, Z. Wang, and J. Sun, “Microtissue-Based Bioink as a Chondrocyte Microshelter for DLP Bioprinting,” Advanced Healthcare Materials 11, no. 22 (2022): 2201877.

[514]

O. Y. Joo, T. H. Kim, Y. S. Kim, et al., “Fabrication of 3D-Printed Implant for Two-Stage Ear Reconstruction Surgery and Its Clinical Application,” Yonsei Medical Journal 64, no. 4 (2023): 291-296.

[515]

J. J. Song, J. P. Guyette, S. E. Gilpin, G. Gonzalez, J. P. Vacanti, and H. C. Ott, “Regeneration and Experimental Orthotopic Transplantation of a Bioengineered Kidney,” Nature Medicine 19, no. 5 (2013): 646-651.

[516]

C.-Y. Hsu, P.-L. Chi, H.-Y. Chen, et al., “Kidney Bioengineering by Using Decellularized Kidney Scaffold and Renal Progenitor Cells,” Tissue and Cell 74 (2022): 101699.

[517]

G. Satchanska, S. Davidova, and P. D. Petrov, “Natural and Synthetic Polymers for Biomedical and Environmental Applications,” Polymers 16, no. 8 (2024): 1159.

[518]

A. Aazmi, D. Zhang, C. Mazzaglia, et al., “Biofabrication Methods for Reconstructing Extracellular Matrix Mimetics,” Bioactive Materials 31 (2024): 475-496.

[519]

C. Gao, Y. Wang, F. Han, et al., “Antibacterial Activity and Osseointegration of Silver-coated Poly(ether ether ketone) Prepared Using the Polydopamine-assisted Deposition Technique,” Journal of Materials Chemistry B 5, no. 47 (2017): 9326-9336.

[520]

S. Hiromoto and T. Yamazaki, “Micromorphological Effect of Calcium Phosphate Coating on Compatibility of Magnesium Alloy With Osteoblast,” Science and Technology of Advanced Materials 18, no. 1 (2017): 96-109.

[521]

W. Lin, L. Qin, H. Qi, et al., “Long-term in Vivo Corrosion Behavior, Biocompatibility and Bioresorption Mechanism of a Bioresorbable Nitrided Iron Scaffold,” Acta Biomaterialia 54 (2017): 454-468.

[522]

S. Ullah, I. Zainol, and R. H. Idrus, “Incorporation of Zinc Oxide Nanoparticles Into Chitosan-collagen 3D Porous Scaffolds: Effect on Morphology, Mechanical Properties and Cytocompatibility of 3D Porous Scaffolds,” International Journal of Biological Macromolecules 104, no. Pt A (2017): 1020-1029.

[523]

J. Deng, Y. Tang, Q. Zhang, et al., “A Bioinspired Medical Adhesive Derived From Skin Secretion of Andrias davidianus for Wound Healing,” Advanced Functional Materials 29, no. 31 (2019): 1809110.

[524]

N. Mandakhbayar, A. El-Fiqi, J.-H. Lee, and H.-W. Kim, “Evaluation of Strontium-Doped Nanobioactive Glass Cement for Dentin-Pulp Complex Regeneration Therapy,” ACS Biomaterials Science & Engineering 5, no. 11 (2019): 6117-6126.

[525]

S. Petrus-Reurer, M. Romano, S. Howlett, J. L. Jones, G. Lombardi, and K. Saeb-Parsy, “Immunological Considerations and Challenges for Regenerative Cellular Therapies,” Communications Biology 4, no. 1 (2021): 798.

[526]

Q. Li and P. Lan, “Activation of Immune Signals During Organ Transplantation,” Signal Transduction and Targeted Therapy 8, no. 1 (2023): 110.

[527]

C. Y. X. Chua, A. Y. Jiang, T. Eufrasio-da-Silva, et al., “Emerging Immunomodulatory Strategies for Cell Therapeutics,” Trends in Biotechnology 41, no. 3 (2023): 358-373.

[528]

L. C. Kadyk, R. M. Okamura, and S. Talib, “Enabling Allogeneic Therapies: CIRM-funded Strategies for Immune Tolerance and Immune Evasion,” Stem Cells Translational Medicine 9, no. 9 (2020): 959-964.

[529]

D. S. Masson-Meyers and L. Tayebi, “Vascularization Strategies in Tissue Engineering Approaches for Soft Tissue Repair,” Journal of Tissue Engineering and Regenerative Medicine 15, no. 9 (2021): 747-762.

[530]

A. Sanchez-Rubio, V. Jayawarna, E. Maxwell, M. J. Dalby, and M. Salmeron-Sanchez, “Keeping It Organized: Multicompartment Constructs to Mimic Tissue Heterogeneity,” Advanced Healthcare Materials 12, no. 17 (2023): 2202110.

[531]

G. Maroli and T. Braun, “The Long and Winding Road of Cardiomyocyte Maturation,” Cardiovascular Research 117, no. 3 (2021): 712-726.

[532]

A. S. Mao and D. J. Mooney, “Regenerative Medicine: Current Therapies and Future Directions,” Proceedings of the National Academy of Sciences of the United States of America 112, no. 47 (2015): 14452-14459.

[533]

K. D. Nordham and S. Ninokawa, “The History of Organ Transplantation,” Baylor University Medical Center Proceedings 35, no. 1 (2022): 124-128.

[534]

M. Hofer and M. P. Lutolf, “Engineering Organoids,” Nature Reviews Materials 6, no. 5 (2021): 402-420.

[535]

A. Oryan, S. Alidadi, A. Moshiri, and N. Maffulli, “Bone Regenerative Medicine: Classic Options, Novel Strategies, and Future Directions,” Journal of Orthopaedic Surgery and Research 9, no. 1 (2014): 18.

[536]

A. El-Sayed and M. Kamel, “Advances in Nanomedical Applications: Diagnostic, Therapeutic, Immunization, and Vaccine Production,” Environmental Science and Pollution Research 27, no. 16 (2020): 19200-19213.

[537]

R. Augustine, P. Dan, A. Hasan, et al., “Stem Cell-based Approaches in Cardiac Tissue Engineering: Controlling the Microenvironment for Autologous Cells,” Biomedicine & Pharmacotherapy 138 (2021): 111425.

[538]

I.-S. Hong, “Enhancing Stem Cell-Based Therapeutic Potential by Combining Various Bioengineering Technologies,” Frontiers in Cell and Developmental Biology 10 (2022): 901661.

[539]

H. Lin, Y. Tang, T. P. Lozito, N. Oyster, B. Wang, and R. S. Tuan, “Efficient in Vivo Bone Formation by BMP-2 Engineered human Mesenchymal Stem Cells Encapsulated in a Projection Stereolithographically Fabricated Hydrogel Scaffold,” Stem Cell Research & Therapy 10, no. 1 (2019): 254.

[540]

A. S. Cakmak, S. Fuerkaiti, D. Karaguzel, C. Karaaslan, and M. Gumusderelioglu, “Enhanced Osteogenic Potential of Noggin Knockout C2C12 Cells on BMP-2 Releasing Silk Scaffolds,” ACS Biomaterials Science & Engineering Journal 9, no. 11 (2023): 6175-6185.

[541]

S. Prithiviraj, A. G. Garcia, K. Linderfalk, et al., Compositional editing of extracellular matrices by CRISPR/Cas9 engineering of human mesenchymal stem cell lines. (eLife Sciences Publications, Ltd, 2024).

[542]

C. Zhong, S. He, Y. Huang, et al., “Scaffold-based Non-viral CRISPR Delivery Platform for Efficient and Prolonged Gene Activation to Accelerate Tissue Regeneration,” Acta Biomaterialia 173 (2024): 283-297.

[543]

C. Li, Z. Mills, and Z. Zheng, “Novel Cell Sources for Bone Regeneration,” MedComm 2, no. 2 (2021): 145-174.

[544]

P. Chen, L. Cui, G. Chen, et al., “The Application of BMP-12-overexpressing Mesenchymal Stem Cells Loaded 3D-printed PLGA Scaffolds in Rabbit Rotator Cuff Repair,” International Journal of Biological Macromolecules 138 (2019): 79-88.

[545]

D. Hao, J.-M. Lopez, J. Chen, A. M. Iavorovschi, N. M. Lelivelt, and A. Wang, “Engineering Extracellular Microenvironment for Tissue Regeneration,” Bioengineering 9, no. 5 (2022): 202.

[546]

J. Gao, X. Yu, X. Wang, Y. He, and J. Ding, “Biomaterial-Related Cell Microenvironment in Tissue Engineering and Regenerative Medicine,” Engineering 13 (2022): 31-45.

[547]

M. Schot, N. Araújo-Gomes, B. van Loo, T. Kamperman, and J. Leijten, “Scalable Fabrication, Compartmentalization and Applications of Living Microtissues,” Bioactive Materials 19 (2022): 392-405.

[548]

I. Decoene, G. Nasello, R. F. Madeiro de Costa, et al., “Robotics-Driven Manufacturing of Cartilaginous Microtissues for Skeletal Tissue Engineering Applications,” Stem Cells Translational Medicine 13, no. 3 (2024): 278-292.

[549]

B. Zhang, A. Korolj, B. F. L. Lai, and M. Radisic, “Advances in Organ-on-a-chip Engineering,” Nature Reviews Materials 3, no. 8 (2018): 257-278.

[550]

K. Thakare, L. Jerpseth, Z. Pei, A. Elwany, F. Quek, and H. Qin, “Bioprinting of Organ-on-Chip Systems: A Literature Review From a Manufacturing Perspective,” Journal of Manufacturing and Materials Processing 5, no. 3 (2021): 91.

RIGHTS & PERMISSIONS

2025 The Author(s). MedComm published by Sichuan International Medical Exchange & Promotion Association (SCIMEA) and John Wiley & Sons Australia, Ltd.

AI Summary AI Mindmap
PDF

10

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/