1
|
Harris A, Gilbert F. Need for greater post-trial support for clinical trial participants assessing high-risk, irreversible treatments. JOURNAL OF MEDICAL ETHICS 2025; 51:268-273. [PMID: 38834240 DOI: 10.1136/jme-2023-109719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 05/18/2024] [Indexed: 06/06/2024]
Abstract
There are increasing numbers of clinical trials assessing high-risk, irreversible treatments. Trial participants should only expect knowledge gain to society, no personal therapeutic benefit. However, participation may lead to long-term harms and prevent future therapeutic options. While some discussion has occurred around post-trial access to treatments for participants who received therapeutic benefit, there are no post-trial support requirements for those suffering long-term consequences from trial participation. Participants may be left with significant medical, psychological, social, technical or financial needs. All trials will end at some point, regardless of their success. Subsequently, they should be designed to take into account the post-trial period including the impact on the ongoing health of a participant and their post-trial needs.
Collapse
Affiliation(s)
- Alex Harris
- Department of Biomedical Engineering, The University of Melbourne, Melbourne, Victoria, Australia
| | - Frederic Gilbert
- EthicsLab, School of Humanities, University of Tasmania, Hobart, Tasmania, Australia
| |
Collapse
|
2
|
Dalton J, Narayanan R, Oris RJ, Ezeonu T, Bradley E, Canseco JA, Vaccaro AR, Koerner JD, Markova D, Kepler C. Can Treatment with Human Mesenchymal Stem Cells Rescue the Degenerative Disc Phenotype? An in Vitro Pilot Study of Induced Cytokine Expression. Spine J 2025:S1529-9430(25)00176-7. [PMID: 40154630 DOI: 10.1016/j.spinee.2025.03.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 01/07/2025] [Accepted: 03/23/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND CONTEXT Given the relatively low cell density in degenerative discs, strategies intended to bolster disc cellularity through stem cell injections have come into clinical use. Stem cell therapy is meant to provide a source of viable disc cells that can promote a healthy disc phenotype. Nevertheless, there is a limited understanding of the mechanisms through which stem cell therapy impacts degeneration. PURPOSE The objectives of this pilot study were: 1) to evaluate gene expression changes associated with an in vitro induced degenerative phenotype in human nucleus pulposus (NP) cells, 2) to co-culture these degenerative NP cells with human mesenchymal stem cells (hMSCs) and investigate the impact this has on gene expression, 3) to investigate possible mechanisms by which hMSCs may impact the degenerative phenotype. STUDY DESIGN Laboratory study. METHODS NP cells were isolated and cultured from patients undergoing anterior lumbar interbody fusion for degenerative disc disease. A degenerative phenotype was induced in cultured NP cells by treatment with an inflammatory protocol (10pg/ml IL-1β and 100pg/ml TNF-α) for 7 days. Gene expression of Treated NP cells was compared to Untreated NP cells via reverse transcriptase polymerase chain reaction. NP cells were then co-cultured with hMSCs in vitro and treated with the inflammatory protocol. Gene expression of Treated NP cells co-cultured with hMSCs was compared to Treated NP cells alone. Preliminary co-culture data demonstrated that IL-10 was uniquely and dramatically upregulated. Therefore, gene expression of Treated NP cells exposed to IL-10 for 24 hours was compared to Treated NP cells alone. RESULTS Treated NP cells compared to Control NP cells showed upregulation of numerous pro-inflammatory cytokines, including CXCL5, IL-8, and IL-6 and downregulation of several anti-inflammatory cytokines, including IL-10. After co-culture of Treated NP cells with hMSCs, a significant increase in gene expression was identified in IL-10 (+15.34 fold), BMP-6 (+2.32 fold), and LIF (+2.14 fold). A significant decrease in gene expression (p < 0.05) was seen in CCL7 (-2.03) and CXCL12 (-1.67). Exposure of Treated NP cells to IL-10 resulted in upregulation of COL-2 (+1.55 fold, p=0.013) and downregulation of IL-8 (-1.4 fold), CXCL-5 (-1.58 fold,), and MMP-3 (-2.02 fold). CONCLUSION This in vitro pilot study shows that co-culture of degenerative phenotype NP cells with hMSCs produces multiple gene regulatory changes associated with an anti-inflammatory phenotype. Additionally, exposure of degenerative phenotype NP cells to IL-10 produces gene regulation associated with both anti-inflammatory and pro-extracellular matrix effects. CLINICAL SIGNIFICANCE These findings provide mechanistic support for the use of stem cell therapy as a strategy to decrease the pro-inflammatory molecular environment associated with disc degeneration. Additionally, given the challenges with the viability of hMSCs in the disc microenvironment, IL-10 may be another potential candidate for future targeted therapies for disc degeneration.
Collapse
Affiliation(s)
- Jonathan Dalton
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, PA 19107.
| | - Rajkishen Narayanan
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, PA 19107
| | - Robert J Oris
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, PA 19107
| | - Teeto Ezeonu
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, PA 19107
| | - Evan Bradley
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, PA 19107
| | - Jose A Canseco
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, PA 19107
| | - Alexander R Vaccaro
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, PA 19107; Department of Orthopaedic Surgery, Rothman Orthopaedic Institute, Philadelphia, PA 19107
| | - John D Koerner
- Hackensack Hospital, Department of Orthopaedic Surgery, Hackensack, NJ
| | - Dessislava Markova
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, PA 19107
| | - Christopher Kepler
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, PA 19107; Department of Orthopaedic Surgery, Rothman Orthopaedic Institute, Philadelphia, PA 19107
| |
Collapse
|
3
|
Chang MM, Chu DT, Lin SC, Lee JS, Vu TD, Vu HT, Ramasamy TS, Lin SP, Wu CC. Enhanced mitochondrial function and delivery from adipose-derived stem cell spheres via the EZH2-H3K27me3-PPARγ pathway for advanced therapy. Stem Cell Res Ther 2025; 16:129. [PMID: 40069892 PMCID: PMC11899936 DOI: 10.1186/s13287-025-04164-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 01/21/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Microenvironmental alterations induce significant genetic and epigenetic changes in stem cells. Mitochondria, essential for regenerative capabilities, provide the necessary energy for stem cell function. However, the specific roles of histone modifications and mitochondrial dynamics in human adipose-derived stem cells (ASCs) during morphological transformations remain poorly understood. In this study, we aim to elucidate the mechanisms by which ASC sphere formation enhances mitochondrial function, delivery, and rescue efficiency. METHODS ASCs were cultured on chitosan nano-deposited surfaces to form 3D spheres. Mitochondrial activity and ATP production were assessed using MitoTracker staining, Seahorse XF analysis, and ATP luminescence assays. Single-cell RNA sequencing, followed by Ingenuity Pathway Analysis (IPA), was conducted to uncover key regulatory pathways, which were validated through molecular techniques. Pathway involvement was confirmed using epigenetic inhibitors or PPARγ-modulating drugs. Mitochondrial structural integrity and delivery efficiency were evaluated after isolation. RESULTS Chitosan-induced ASC spheres exhibited unique compact mitochondrial morphology, characterized by condensed cristae, enhanced mitochondrial activity, and increased ATP production through oxidative phosphorylation. High expressions of mitochondrial complex I genes and elevated levels of mitochondrial complex proteins were observed without an increase in reactive oxygen species (ROS). Epigenetic modification of H3K27me3 and PPARγ involvement were discovered and confirmed by inhibiting H3K27me3 with the specific EZH2 inhibitor GSK126 and by adding the PPARγ agonist Rosiglitazone (RSG). Isolated mitochondria from ASC spheres showed improved structural stability and delivery efficiency, suppressed the of inflammatory cytokines in LPS- and TNFα-induced inflamed cells, and rescued cells from damage, thereby enhancing function and promoting recovery. CONCLUSION Enhancing mitochondrial ATP production via the EZH2-H3K27me3-PPARγ pathway offers an alternative strategy to conventional cell-based therapies. High-functional mitochondria and delivery efficiency show significant potential for regenerative medicine applications.
Collapse
Affiliation(s)
- Ming-Min Chang
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, No. 1, University Road, Tainan, 70101, Taiwan
- Medical Device Innovation Center, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Dinh Toi Chu
- Faculty of Applied Sciences, International School, Vietnam National University, Hanoi, 1000, Vietnam
| | - Sheng-Che Lin
- Division of Plastic and Reconstructive Surgery, Tainan Municipal An-Nan Hospital-China Medical University, Tainan, 70965, Taiwan
| | - Jung-Shun Lee
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, No. 1, University Road, Tainan, 70101, Taiwan
- Division of Neurosurgery, Department of Surgery, National Cheng Kung University Hospital, Tainan, 701401, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Thuy Duong Vu
- Faculty of Applied Sciences, International School, Vietnam National University, Hanoi, 1000, Vietnam
| | - Hue Thi Vu
- Faculty of Applied Sciences, International School, Vietnam National University, Hanoi, 1000, Vietnam
| | - Thamil Selvee Ramasamy
- Stem Cell Biology Laboratory, Department of Molecular Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, 50603, Malaysia
| | - Shau-Ping Lin
- Institute of Biotechnology, College of Bio-Resources and Agriculture, National Taiwan University, Taipei, 10672, Taiwan
| | - Chia-Ching Wu
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, No. 1, University Road, Tainan, 70101, Taiwan.
- Medical Device Innovation Center, National Cheng Kung University, Tainan, 70101, Taiwan.
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan.
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, 70101, Taiwan.
- International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan, 70101, Taiwan.
| |
Collapse
|
4
|
Rajalekshmi R, Agrawal DK. Synergistic potential of stem cells and microfluidics in regenerative medicine. Mol Cell Biochem 2025; 480:1481-1493. [PMID: 39285093 PMCID: PMC11842489 DOI: 10.1007/s11010-024-05108-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 08/27/2024] [Indexed: 02/21/2025]
Abstract
Regenerative medicine has immense potential to revolutionize healthcare by using regenerative capabilities of stem cells. Microfluidics, a cutting-edge technology, offers precise control over cellular microenvironments. The integration of these two fields provides a deep understanding of stem cell behavior and enables the development of advanced therapeutic strategies. This critical review explores the use of microfluidic systems to culture and differentiate stem cells with precision. We examined the use of microfluidic platforms for controlled nutrient supply, mechanical stimuli, and real-time monitoring, providing an unprecedented level of detail in studying cellular responses. The convergence of stem cells and microfluidics holds immense promise for tissue repair, regeneration, and personalized medicine. It offers a unique opportunity to revolutionize the approach to regenerative medicine, facilitating the development of advanced therapeutic strategies and enhancing healthcare outcomes.
Collapse
Affiliation(s)
- Resmi Rajalekshmi
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766, USA
| | - Devendra K Agrawal
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766, USA.
| |
Collapse
|
5
|
Boopathy K, Palaniyandi T, Ravi M, Wahab MRA, Baskar G, Rab SO, Saeed M, Balaramnavar VM. Exploring the potential of stem cell therapy: Applications, types, and future directions. Acta Histochem 2025; 127:152237. [PMID: 40020616 DOI: 10.1016/j.acthis.2025.152237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/14/2025] [Accepted: 02/18/2025] [Indexed: 03/03/2025]
Abstract
One of the most significant treatment approaches now accessible is stem cell therapy. Over the last few decades, a lot of study has been done in this field, and this fascinating feature of plasticity could have therapeutic uses. The potential of stem cells to restore function lost as a result of disease, trauma, congenital defects, and age has made stem cell research a key priority for scientific and medical organizations across the world. Stem cells are a crucial topic of study in regenerative medicine because of their capacity to replace, repair, or regenerate damaged cells, tissues, or organs. As a result, stem cell therapy is being used as a treatment strategy for a number of illnesses. Because stem cells may proliferate indefinitely and generate vast quantities of differentiated cells needed for transplantation, they hold enormous promise for regenerative medicine. Stem cells can be reprogrammed from adult cell types or originate from embryonic or fetal origins. Depending on their availability and place of origin, stem cells can be totipotent, pluripotent, multipotent, oligopotent, or unipotent. With stem cell treatment, many ailments, including diabetes, liver disease, infertility, wounds and traumas, neurological disorders, cardiovascular disease, and cancer, might be cured. Various types of stem cell treatment are described in this review along with their applications in different therapeutic fields, ethical considerations, and advantages and disadvantages.
Collapse
Affiliation(s)
- KeerthiShri Boopathy
- Department of Biotechnology, Dr. M.G.R Educational and Research Institute, Chennai 600095, India
| | - Thirunavukkarasu Palaniyandi
- Department of Biotechnology, Dr. M.G.R Educational and Research Institute, Chennai 600095, India; ACS-Advanced Medical Research Institute, Dr. M.G.R Educational and Research Institute, Chennai 600077, India.
| | - Maddaly Ravi
- Department of Human Genetics, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu 600 116, India
| | | | - Gomathy Baskar
- Department of Biotechnology, Dr. M.G.R Educational and Research Institute, Chennai 600095, India
| | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Mohd Saeed
- Department of Biology, College of Science, University of Hail, Hail, Saudi Arabia
| | - Vishal M Balaramnavar
- School of Pharmacy and Research Centre, Sanskriti University, Chhata, Mathura, Uttar Pradesh 281401, India
| |
Collapse
|
6
|
Kim M, Kong D, Kim NG, Kim MJ, Kim HY, Choi JJ, Choi YS, Lee HE, Farzaneh KS, Kwon D, Lee S, Kang KS. Therapeutic effect of long-interval repeated subcutaneous administration of canine amniotic membrane-derived mesenchymal stem cells in atopic dermatitis mouse model. BMC Vet Res 2025; 21:115. [PMID: 40011929 DOI: 10.1186/s12917-025-04554-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 01/31/2025] [Indexed: 02/28/2025] Open
Abstract
Atopic dermatitis (AD) is a chronic and inflammatory disease. According to a recent study, administration of canine MSCs is a potential therapy for immunological diseases. However, most related studies involve short-term experiments and acute atopic dermatitis animal models. Thus, studies of repeated subcutaneous injection of canine MSCs for ameliorating long-term inflammatory skin disorders have not yet been established. In this study, we evaluated the effects of long-term canine amniotic mesenchymal stem cells (cAM-MSCs) and calcineurin inhibitors (CNIs) treatments in mouse AD model for up to 8 weeks and compared the differences in therapeutic effect through canine peripheral blood mononuclear cells (PBMCs). Using a mouse model, we validated the therapeutic impact of cAM-MSCs in comparison to pimecrolimus (Pime), the most widely used CNIs, as a therapy for canine AD. Based on our results, we verified that the cAM-MSC treatment group exhibited substantially lower scores for tissue pathologic alterations, inflammatory cytokines, and dermatologic symptoms than the PBS control group. Importantly, compared with Pime, cAM-MSCs were more effective at preventing wound dysfunction and regulating mast cell activity. Additionally, we confirmed that immune modulation proteins (TGF-β1, IDO1, and COX-2) were increased in the cAM-MSCs treatment group. Furthermore, we examined the immunoregulatory effect of cAM-MSCs through the proliferation of T lymphocytes from activated canine PBMCs. As a result, cAM-MSCs suppressed the proliferative capacity of effector T cells from canine PBMCs more effectively than Pime. In conclusion, this study suggested that the cAM-MSCS could be an effective canine treatment for long-term canine AD through regeneration and immunomodulation.
Collapse
Grants
- 23A0101L1 The Ministry of Science and ICT, the Ministry of Health & Welfare
- 23A0101L1 The Ministry of Science and ICT, the Ministry of Health & Welfare
- 23A0101L1 The Ministry of Science and ICT, the Ministry of Health & Welfare
- 23A0101L1 The Ministry of Science and ICT, the Ministry of Health & Welfare
- 23A0101L1 The Ministry of Science and ICT, the Ministry of Health & Welfare
- 23A0101L1 The Ministry of Science and ICT, the Ministry of Health & Welfare
- 23A0101L1 The Ministry of Science and ICT, the Ministry of Health & Welfare
- 23A0101L1 The Ministry of Science and ICT, the Ministry of Health & Welfare
- 23A0101L1 The Ministry of Science and ICT, the Ministry of Health & Welfare
- 23A0101L1 The Ministry of Science and ICT, the Ministry of Health & Welfare
- 23A0101L1 The Ministry of Science and ICT, the Ministry of Health & Welfare
Collapse
Affiliation(s)
- Minsoo Kim
- The Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Dasom Kong
- The Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Nam Gyo Kim
- The Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Min-Ji Kim
- The Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hee-Yeong Kim
- The Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jung-Ju Choi
- The Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Yu-Seung Choi
- The Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Ha-Eun Lee
- The Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Khaligh Seyedeh Farzaneh
- The Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Dohyung Kwon
- The Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Seunghee Lee
- Stem Cell and Regenerative Bioengineering Institute, Global R&D Center, Kangstem Biotech Co. Ltd., Ace Highend Tower 8, 84, Gasan digital 1-ro, Geumcheon-gu, Seoul, 08590, Republic of Korea
| | - Kyung-Sun Kang
- The Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
7
|
Greșiță A, Hermann DM, Boboc IKS, Doeppner TR, Petcu E, Semida GF, Popa-Wagner A. Glial Cell Reprogramming in Ischemic Stroke: A Review of Recent Advancements and Translational Challenges. Transl Stroke Res 2025:10.1007/s12975-025-01331-7. [PMID: 39904845 DOI: 10.1007/s12975-025-01331-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 01/16/2025] [Accepted: 01/18/2025] [Indexed: 02/06/2025]
Abstract
Ischemic stroke, the second leading cause of death worldwide and the leading cause of long-term disabilities, presents a significant global health challenge, particularly in aging populations where the risk and severity of cerebrovascular events are significantly increased. The aftermath of stroke involves neuronal loss in the infarct core and reactive astrocyte proliferation, disrupting the neurovascular unit, especially in aged brains. Restoring the balance between neurons and non-neuronal cells within the perilesional area is crucial for post-stroke recovery. The aged post-stroke brain mounts a fulminant proliferative astroglial response, leading to gliotic scarring that prevents neural regeneration. While countless therapeutic techniques have been attempted for decades with limited success, alternative strategies aim to transform inhibitory gliotic tissue into an environment conducive to neuronal regeneration and axonal growth through genetic conversion of astrocytes into neurons. This concept gained momentum following discoveries that in vivo direct lineage reprogramming in the adult mammalian brain is a feasible strategy for reprogramming non-neuronal cells into neurons, circumventing the need for cell transplantation. Recent advancements in glial cell reprogramming, including transcription factor-based methods with factors like NeuroD1, Ascl1, and Neurogenin2, as well as small molecule-induced reprogramming and chemical induction, show promise in converting glial cells into functional neurons. These approaches leverage the brain's intrinsic plasticity for neuronal replacement and circuit restoration. However, applying these genetic conversion therapies in the aged, post-stroke brain faces significant challenges, such as the hostile inflammatory environment and compromised regenerative capacity. There is a critical need for safe and efficient delivery methods, including viral and non-viral vectors, to ensure targeted and sustained expression of reprogramming factors. Moreover, addressing the translational gap between preclinical successes and clinical applications is essential, emphasizing the necessity for robust stroke models that replicate human pathophysiology. Ethical considerations and biosafety concerns are critically evaluated, particularly regarding the long-term effects and potential risks of genetic reprogramming. By integrating recent research findings, this comprehensive review provides an in-depth understanding of the current landscape and future prospects of genetic conversion therapy for ischemic stroke rehabilitation, highlighting the potential to enhance personalized stroke management and regenerative strategies through innovative approaches.
Collapse
Affiliation(s)
- Andrei Greșiță
- Experimental Research Center for Normal and Pathological Aging, University of Medicine and Pharmacy Craiova, 200349, Craiova, Romania
- Department of Biomedical Sciences, College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, NY, 11568, USA
| | - Dirk M Hermann
- Chair of Vascular Neurology and Dementia, Department of Neurology, University Hospital Essen, 45147, Essen, Germany
- Experimental Research Center for Normal and Pathological Aging, University of Medicine and Pharmacy Craiova, 200349, Craiova, Romania
| | - Ianis Kevyn Stefan Boboc
- Experimental Research Center for Normal and Pathological Aging, University of Medicine and Pharmacy Craiova, 200349, Craiova, Romania
| | - Thorsten R Doeppner
- Department of Neurology, University Medical Center Göttingen, 37075, Göttingen, Germany
- Department of Neurology, University of Giessen Medical School, 35392, Giessen, Germany
| | - Eugen Petcu
- Department of Biomedical Sciences, College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, NY, 11568, USA
- Department of Biological & Chemical Sciences, New York Institute of Technology, Old Westbury, NY, 11568, USA
| | - Ghinea Flavia Semida
- Experimental Research Center for Normal and Pathological Aging, University of Medicine and Pharmacy Craiova, 200349, Craiova, Romania.
| | - Aurel Popa-Wagner
- Chair of Vascular Neurology and Dementia, Department of Neurology, University Hospital Essen, 45147, Essen, Germany.
- Experimental Research Center for Normal and Pathological Aging, University of Medicine and Pharmacy Craiova, 200349, Craiova, Romania.
| |
Collapse
|
8
|
Lee HJ, Hossain R, Baek CH, Lee CJ, Hwang SC. Intra-Articular Injection of Stem Cells for the Regeneration of Knee Joint Cartilage: a Therapeutic Option for Knee Osteoarthritis - a Narrative Review. Biomol Ther (Seoul) 2025; 33:86-94. [PMID: 39632656 PMCID: PMC11704397 DOI: 10.4062/biomolther.2024.139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/22/2024] [Accepted: 10/30/2024] [Indexed: 12/07/2024] Open
Abstract
Current approaches to regulating osteoarthritis primarily focus on symptom management; however, these methods often have significant side effects and may not be suitable for long-term care. As an alternative to conventional treatments, injecting stem cells into knee joint cartilage is a promising option for repairing damaged cartilage. In this review, we outline the general procedure for stem cell treatment of knee joint cartilage regeneration, emphasizing the potential of intra-articular stem cell injections as a therapeutic option for osteoarthritis. We examined and summarized patient evaluation and preparation for knee joint stem cell therapy, stem cell harvesting, stem cell preparation, injection procedures for stem cell therapy, post-injection care and monitoring, potential outcomes of stem cell therapy, and considerations and risks associated with stem cell therapy. Overall, stem cell injections for knee joint cartilage damage represent a promising frontier in orthopedic care. They offer potential benefits such as pain and inflammation reduction, promotion of cartilage repair and regeneration, and the possibility of avoiding more invasive treatments such as knee surgery. Ongoing collaboration among researchers, clinicians, and regulatory organizations is crucial for advancing this field and translating scientific discoveries into effective clinical applications.
Collapse
Affiliation(s)
- Hyun Jae Lee
- Smith Liberal Arts College and Department of Addiction Science, Graduate School, Sahmyook University, Seoul 01795, Republic of Korea
| | - Rajib Hossain
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
- Brain Korea 21 FOUR Project for Medical Science, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Chang-Heon Baek
- Department of Orthopaedic Surgery and Institute of Health Sciences, Gyeongsang National University College of Medicine and Gyeongsang National University Hospital, Jinju 52727, Republic of Korea
| | - Choong Jae Lee
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
- Brain Korea 21 FOUR Project for Medical Science, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Sun-Chul Hwang
- Department of Orthopaedic Surgery and Institute of Health Sciences, Gyeongsang National University College of Medicine and Gyeongsang National University Hospital, Jinju 52727, Republic of Korea
| |
Collapse
|
9
|
Pandey K, Srivastava P, Pandey SK, Johari S, Bhatnagar P, Sonane M, Mishra A. Stem Cells as a Novel Source for Regenerative Medicinal Applications in Alzheimer's Disease: An Update. Curr Mol Med 2025; 25:146-166. [PMID: 39318206 DOI: 10.2174/0115665240334785240913071442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 08/08/2024] [Accepted: 08/26/2024] [Indexed: 09/26/2024]
Abstract
Alzheimer's Disease (AD) is a progressive neurodegenerative disorder characterized by loss of the neurons, excessive accumulation of misfolded Aβ and Tau proteins, and degeneration of neural synapses, primarily occurring in the neocortex and the hippocampus regions of the brain. AD Progression is marked by cognitive deterioration, memory decline, disorientation, and loss of problem-solving skills, as well as language. Due to limited comprehension of the factors contributing to AD and its severity due to neuronal loss, even today, the medications approved by the U.S. Food and Drug Administration (FDA) are not precisely efficient and curative. Stem cells possess great potential in aiding AD due to their self-renewal, proliferation, and differentiation properties. Stem cell therapy can aid by replacing the lost neurons, enhancing neurogenesis, and providing an enriched environment to the pre-existing neural cells. Stem cell therapy has provided us with promising results in regard to the animal AD models, and even pre-clinical studies have shown rather positive results. Cell replacement therapies are potential curative means to treat AD, and there are a number of undergoing human clinical trials to make Stem Cell therapy accessible for AD patients. In this review, we aim to discuss the AD pathophysiology and varied stem cell types and their application.
Collapse
Affiliation(s)
- Kratika Pandey
- School of Biosciences, Institute of Management Studies Ghaziabad 9 (University Courses Campus), NH09, Adhyatmik Nagar, Ghaziabad, Uttar Pradesh, 201015, India
| | - Priyanka Srivastava
- School of Biosciences, Institute of Management Studies Ghaziabad 9 (University Courses Campus), NH09, Adhyatmik Nagar, Ghaziabad, Uttar Pradesh, 201015, India
| | - Swaroop Kumar Pandey
- Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Mathura, 281406, India
| | - Surabhi Johari
- School of Biosciences, Institute of Management Studies Ghaziabad 9 (University Courses Campus), NH09, Adhyatmik Nagar, Ghaziabad, Uttar Pradesh, 201015, India
| | - Priyanka Bhatnagar
- Department of Pharmacology, All India Institute of Medical Sciences, Ansari Nagar, Delhi, 110029, India
| | - Madhavi Sonane
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Lucknow, 226025, India
| | - Anuja Mishra
- Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Mathura, 281406, India
| |
Collapse
|
10
|
Shiri H, Javan M. Sox2-mediated transdifferentiation of hAT-MSCs into induced neural progenitor-like cells for remyelination therapies. Tissue Cell 2024; 91:102553. [PMID: 39255744 DOI: 10.1016/j.tice.2024.102553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/03/2024] [Accepted: 09/03/2024] [Indexed: 09/12/2024]
Abstract
Mesenchymal stem cells (MSCs) are converted to neural cells using growth factors and chemicals. Although these neural cells are effective at modulating disease symptoms, they are less effective at replacing lost neural cells. Direct transdifferentiation seems to be a promising method for generating the required cells for regenerative medicine applications. Sox2 is a key transcription factor in neural progenitor (NP) fate determination and has been frequently used for transdifferentiating different cell types to NPs. Here, we demonstrated that the overexpression of a single transcription factor, Sox2, in human adipose tissue-derived mesenchymal stem cells (hAT-MSCs) led to the generation of induced NPs-like cells that were clonogenic, proliferative and passageable, and showed the potential to differentiate into three neural lineages. NPs are known as progenitors with the potential to differentiate into oligodendrocytes. In vivo, following transplantation into demyelinated adult mouse brains, they survived, differentiated and integrated into the adult brain while participating in the remyelination process and behavioral improvement. This report introduces a beneficial, low-cost and effective approach for generating NPs from an accessible adult source for autologous applications in treating neurodegenerative diseases, including remyelination therapies for multiple sclerosis and other demyelinating diseases.
Collapse
Affiliation(s)
- Hamed Shiri
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Javan
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; Institute for Brain and Cognition, Tarbiat Modares University, Tehran, Iran; Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
11
|
Aly RM, Abohashem RS, Ahmed HH, Halim ASA. Combinatorial intervention with dental pulp stem cells and sulfasalazine in a rat model of ulcerative colitis. Inflammopharmacology 2024; 32:3863-3879. [PMID: 39078564 PMCID: PMC11550242 DOI: 10.1007/s10787-024-01532-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 07/08/2024] [Indexed: 07/31/2024]
Abstract
BACKGROUND Ulcerative colitis is an inflammatory bowel disease (IBD) that involves inflammation of the colon lining and rectum. Although a definitive cure for IBD has not been identified, various therapeutic approaches have been proposed to mitigate the symptomatic presentation of this disease, primarily focusing on reducing inflammation. The aim of the present study was to evaluate the therapeutic potential of combining dental pulp stem cells (DPSCs) with sulfasalazine in an acetic acid-induced ulcerative colitis rat model and to assess the impact of this combination on the suppression of inflammatory cytokines and the regulation of oxidative stress in vivo. METHODS Ulcerative colitis was induced in rats through transrectal administration of 3% acetic acid. The therapeutic effect of combining DPSCs and sulfasalazine on UC was evaluated by measuring the colonic weight/length ratio and edema markers; performing histopathological investigations of colon tissue; performing immunohistochemical staining for NF-κB-P65 and IL-1β; and evaluating oxidative stress and antioxidant indices via ELISA. Moreover, the proinflammatory markers NF-κB-P65, TNF-α and TLR-4 were assessed in colon tissue via ELISA. Furthermore, qRT‒PCR was used to estimate the expression levels of the TLR-4, NF-κB-P65, and MYD88 genes in colon tissue. RESULTS The investigated macroscopic and microscopic signs of inflammation were markedly improved after the combined administration of sulfasalazine and DPSCs, where a noticeable improvement in histological structure, with an intact mucosal epithelium and mild inflammatory infiltration in the mucosa and submucosa, with slight hemorrhage. The administration of either DPSCs or sulfasalazine, either individually or in combination, significantly reduced ROS levels and significantly increased XOD activity. The immunohistochemical results demonstrated that the combined administration of DPSCs and sulfasalazine attenuated NFκB-p65 and IL-1β expression. Finally, the combined administration of DPSCs and sulfasalazine significantly downregulated MyD88, NF-κB and TLR4 gene expression. CONCLUSIONS Cotreatment with DPSCs and sulfasalazine had synergistic effects on ulcerative colitis, and these effects were relieved.
Collapse
Affiliation(s)
- Riham M Aly
- Basic Dental Science Department, Oral & Dental Research Institute, National Research Centre, 33 El Bohouth St, Dokki, Giza, Egypt.
- Stem Cells Lab, Center of Excellence for Advanced Sciences, National Research Centre, Dokki, Giza, Egypt.
| | - Rehab S Abohashem
- Stem Cells Lab, Center of Excellence for Advanced Sciences, National Research Centre, Dokki, Giza, Egypt
- Hormones Department, Medical Research and Clinical Studies Institute, National Research Centre, Dokki, Giza, Egypt
| | - Hanaa H Ahmed
- Stem Cells Lab, Center of Excellence for Advanced Sciences, National Research Centre, Dokki, Giza, Egypt
- Hormones Department, Medical Research and Clinical Studies Institute, National Research Centre, Dokki, Giza, Egypt
| | - Alyaa S Abdel Halim
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| |
Collapse
|
12
|
Granjeiro JM, Borchio PGDM, Ribeiro IPB, Paiva KBS. Bioengineering breakthroughs: The impact of stem cell models on advanced therapy medicinal product development. World J Stem Cells 2024; 16:860-872. [PMID: 39493828 PMCID: PMC11525646 DOI: 10.4252/wjsc.v16.i10.860] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/22/2024] [Accepted: 09/23/2024] [Indexed: 10/25/2024] Open
Abstract
The burgeoning field of bioengineering has witnessed significant strides due to the advent of stem cell models, particularly in their application in advanced therapy medicinal products (ATMPs). In this review, we examine the multifaceted impact of these developments, emphasizing the potential of stem cell models to enhance the sophistication of ATMPs and to offer alternatives to animal testing. Stem cell-derived tissues are particularly promising because they can reshape the preclinical landscape by providing more physiologically relevant and ethically sound platforms for drug screening and disease modelling. We also discuss the critical challenges of reproducibility and accuracy in measurements to ensure the integrity and utility of stem cell models in research and application. Moreover, this review highlights the imperative of stem cell models to align with regulatory standards, ensuring using stem cells in ATMPs translates into safe and effective clinical therapies. With regulatory approval serving as a gateway to clinical adoption, the collaborative efforts between scientists and regulators are vital for the progression of stem cell applications from bench to bedside. We advocate for a balanced approach that nurtures innovation within the framework of rigorous validation and regulatory compliance, ensuring that stem cell-base solutions are maximized to promote public trust and patient health in ATMPs.
Collapse
Affiliation(s)
- José Mauro Granjeiro
- Division of Biological Metrology, The National Institute of Metrology, Quality, and Technology, Duque de Caxias 25250020, Rio de Janeiro, Brazil.
| | | | - Icaro Paschoal Brito Ribeiro
- Laboratory of Extracellular Matrix Biology and Cellular Interaction, Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508000, São Paulo, Brazil
| | - Katiucia Batista Silva Paiva
- Laboratory of Extracellular Matrix Biology and Cellular Interaction, Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508000, São Paulo, Brazil
| |
Collapse
|
13
|
Song Y, Yin C, Kong N. Stem Cell-Derived Exosomes: Natural Intercellular Messengers with Versatile Mechanisms for the Treatment of Diabetic Retinopathy. Int J Nanomedicine 2024; 19:10767-10784. [PMID: 39469447 PMCID: PMC11514697 DOI: 10.2147/ijn.s475234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 10/05/2024] [Indexed: 10/30/2024] Open
Abstract
Diabetic retinopathy is one of the complications of diabetes mellitus that occurs in the early stages. It is a disease that has a serious impact, and may lead to blindness when the disease progresses to advanced stages. Currently, treatments for diabetic retinopathy are mainly limited to its advanced stages of the disease, being restricted to a single therapeutic mechanism. Stem cells hold the promise of regenerative therapy and have the potential to comprehensively improve diabetic retinopathy. However, direct stem cell therapy carries some risk of carcinogenesis. Exosomes secreted by stem cells have shown a similar overall improvement in disease as stem cells. Exosomes can carry a number of biologically active materials from donor cells to recipient cells or distant organs, regulating intercellular signaling. Exosomes have shown remarkable efficacy in alleviating oxidative stress, inhibiting inflammatory responses, suppressing angiogenesis, reducing apoptosis and protecting neural tissues. Currently, the experimental literature using stem cell exosomes in the treatment of diabetic retinopathy tends to converge on the above experimental results. With this in mind, we have chosen to explore exosomes in depth from a subtle molecular perspective. We will elaborate on this perspective in this paper and propose to advocate exosome therapy as one promising approach for the treatment of diabetic retinopathy to ameliorate the lesions through multiple mechanisms.
Collapse
Affiliation(s)
- Yameng Song
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, Changchun, People’s Republic of China
- National Health Commission (NHC) Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, People’s Republic of China
| | - Caiyun Yin
- National Health Commission (NHC) Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, People’s Republic of China
| | - Ning Kong
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, Changchun, People’s Republic of China
| |
Collapse
|
14
|
Deng C, Aldali F, Luo H, Chen H. Regenerative rehabilitation: a novel multidisciplinary field to maximize patient outcomes. MEDICAL REVIEW (2021) 2024; 4:413-434. [PMID: 39444794 PMCID: PMC11495474 DOI: 10.1515/mr-2023-0060] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 05/15/2024] [Indexed: 10/25/2024]
Abstract
Regenerative rehabilitation is a novel and rapidly developing multidisciplinary field that converges regenerative medicine and rehabilitation science, aiming to maximize the functions of disabled patients and their independence. While regenerative medicine provides state-of-the-art technologies that shed light on difficult-to-treated diseases, regenerative rehabilitation offers rehabilitation interventions to improve the positive effects of regenerative medicine. However, regenerative scientists and rehabilitation professionals focus on their aspects without enough exposure to advances in each other's field. This disconnect has impeded the development of this field. Therefore, this review first introduces cutting-edge technologies such as stem cell technology, tissue engineering, biomaterial science, gene editing, and computer sciences that promote the progress pace of regenerative medicine, followed by a summary of preclinical studies and examples of clinical investigations that integrate rehabilitative methodologies into regenerative medicine. Then, challenges in this field are discussed, and possible solutions are provided for future directions. We aim to provide a platform for regenerative and rehabilitative professionals and clinicians in other areas to better understand the progress of regenerative rehabilitation, thus contributing to the clinical translation and management of innovative and reliable therapies.
Collapse
Affiliation(s)
- Chunchu Deng
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Fatima Aldali
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hongmei Luo
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hong Chen
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
15
|
Chakra MA, Bailly H, Klampke F, Boaz J, Jida M, Yassine AA, McElree IM, Moussa M. An update on the use of stem cell therapy for erectile dysfunction. Asian J Urol 2024; 11:530-544. [PMID: 39534008 PMCID: PMC11551375 DOI: 10.1016/j.ajur.2023.07.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 07/24/2023] [Indexed: 11/16/2024] Open
Abstract
Objective This systematic review aimed to analyze animal and human trial data to better understand the efficacy of stem cell therapy (SCT) for erectile dysfunction (ED) and the obstacles that may hinder its application in this field. Methods We searched electronic databases, including PubMed and Scopus, for published studies with the Medical Subject Heading terms of "erectile dysfunction" (AND) "stem cell therapy" (OR) "erectile dysfunction" (AND) "clinical trial of stem cell therapy" (OR) "stem cell therapy" (AND) "sexual dysfunction". The search was limited to English-language journals and full papers only. The initial search resulted in 450 articles, of which 90 relevant to our aims were included in the analysis. Results ED is a multifactorial disease. Current treatment options rely on pharmacotherapy as well as surgical options. Patients may have side effects or unsatisfactory results following the use of these treatment options. SCT may restore pathophysiological changes leading to ED rather than treating the symptoms. It has been evaluated in animal models and shown promising results in humans. Results confirm that SCT does improve erectile function in animals with different types of SC use. In humans, evidence showed promising results, but the trials were heterogeneous and limited mainly by a lack of randomization and the small sample size. Many challenges could limit future research in this field, including ethical dilemmas, regulation, patient recruitment, the cost of therapy, and the lack of a standardized SCT regimen. Repairing and possibly replacing diseased cells, tissue, or organs and eventually retrieving normal function should always be the goals of any therapy, and this can only be guaranteed by SCT. Conclusion SCT is a potential and successful treatment for ED, particularly in patients who are resistant to the classic therapy. SCT may promote nerve regeneration and vascular cell regeneration, not only symptomatic treatment.
Collapse
Affiliation(s)
| | - Hugo Bailly
- Department of Urology, Vivantes Klinikum, Berlin, Germany
| | - Fabian Klampke
- Department of Urology, Vivantes Klinikum, Berlin, Germany
| | - Johann Boaz
- Department of Urology, Royal Liverpool University Hospital, Liverpool, UK
| | | | - Ahmad Abou Yassine
- Department of Internal Medicine, Staten Island University Hospital, Staten Island, NY, USA
| | - Ian M. McElree
- Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Mohamad Moussa
- Department of Urology, Lebanese University, Beirut, Lebanon
| |
Collapse
|
16
|
Puspita R, Jusuf AA, Antarianto RD, Sianipar IR. A systematic review of the anti-inflammatory and anti-fibrotic potential of human umbilical cord mesenchymal stem cells-derived exosomes in experimental models of liver regeneration. Mol Biol Rep 2024; 51:999. [PMID: 39302506 DOI: 10.1007/s11033-024-09929-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024]
Abstract
Chronic liver injuries and their complications are leading causes of death, especially in developing countries (Sharma and Nagalli in Sex/Gender-Specific Medicine in the Gastrointestinal Diseases, StatPearls Publishing, 2023). The available and effective treatment plans are limited, implicating the need for innovative treatment approaches (Tsuchiya et al. in Inflamm Regener, 2019;Sharma and Nagalli in Sex/Gender-Specific Medicine in the Gastrointestinal Diseases, StatPearls Publishing, 2023;Younossi et al. in Clin Gastroenterol Hepatol 21:1978-1991, 2023;). This paper aims to summarize the effects and mechanisms of hUC-MSC-exo on liver injuries and its complications; it also suggests future directions for future research. The outcomes of interest are the morphology and histology of the liver, pathology score, liver function enzyme, glucose and lipid metabolism, and the effect hUC-MSC-exo had on gene regulation regarding liver diseases. A comprehensive review of nineteen studies was conducted to assess the effectiveness of the implementation of the hUC-MSC-Exo, instilling confidence in the validity of the findings. Regarding the morphology and histology of the liver and pathology score, hUC-MSC-exo treatment resulted in improved liver morphology post-treatment, as indicated by the reduction in pathology scores. However, these observed improvements in the liver surface are not directly attributed to the hUC-MSC-Exo itself but to the overall healing processes stimulated by the treatment. In physiological outcomes, hUC-MSC-exo also improves glucose and lipid metabolism, especially in diet-induced liver injury and its complications. In gene regulation, one interesting gene in this intervention is the fat mass and obesity-associated (FTO), in which hUC-MSC-exo combined with miRNAs can suppress FTO. HUC-MSC-Exo can improve by utilizing several possible pathways, targeting pinpoints in the pathogenesis of liver disease or glucose and lipid metabolism. This study presents hUC-MSC-exo better in all outcomes of interest compared to the control or sham group. Further specification of indications of the hUC-MSC-exo method may be beneficial and essential to be analyzed in future reviews to better understand the effectiveness of each hUC-MSC-exo dose, duration, and medium.
Collapse
Affiliation(s)
- Ratna Puspita
- Doctoral Program in Biomedical Science, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Department of Biochemistry, Faculty of Medicine, Universitas Pembangunan Nasional Veteran Jakarta, Jakarta, Indonesia
| | - Ahmad Aulia Jusuf
- Department of Histology, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia.
| | | | | |
Collapse
|
17
|
Lu X, Perr E, Naqvi T, Galitz D, Andersen M, Grabowski D, Person A, Kalyuzhny A, Flynn KC. A Novel Recombinant Vitronectin Variant Supports the Expansion and Differentiation of Pluripotent Stem Cells in Defined Animal-Free Workflows. Cells 2024; 13:1566. [PMID: 39329750 PMCID: PMC11429963 DOI: 10.3390/cells13181566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 09/04/2024] [Accepted: 09/09/2024] [Indexed: 09/28/2024] Open
Abstract
An essential aspect of harnessing the potential of pluripotent stem cells (PSCs) and their derivatives for regenerative medicine is the development of animal-free and chemically defined conditions for ex vivo cultivation. PSCs, including embryonic and induced PSCs (iPSCs), are in the early stages of clinical trials for various indications, including degenerative diseases and traumatic injury. A key step in the workflows generating these cells for more widespread clinical use is their safe and robust ex vivo cultivation. This entails optimization of cell culture media and substrates that are safe and consistent while maintaining robust functionality. Here, we describe the design of a human vitronectin (hVTN) variant with improved manufacturability in a bacterial expression system along with improved function in comparison to wild-type VTN and other previously characterized polypeptide fragments. In conjunction with an animal component-free media formulation, our hVTN fragment provides animal-free conditions for the enhanced expansion of iPSCs. This hVTN variant also supports the reprogramming of PBMCs into iPSCs. Furthermore, we show that these iPSCs can be efficiently differentiated into the three major germ layers and cortical neurons, thereby closing the loop on a completely defined animal-free workflow for cell types relevant for regenerative medicine.
Collapse
Affiliation(s)
- Xi Lu
- Stem Cell & Gene Therapy, Bio-Techne, Minneapolis, MN 55413, USA; (X.L.); (E.P.); (T.N.); (D.G.); (M.A.)
| | - Eli Perr
- Stem Cell & Gene Therapy, Bio-Techne, Minneapolis, MN 55413, USA; (X.L.); (E.P.); (T.N.); (D.G.); (M.A.)
| | - Tahmina Naqvi
- Stem Cell & Gene Therapy, Bio-Techne, Minneapolis, MN 55413, USA; (X.L.); (E.P.); (T.N.); (D.G.); (M.A.)
| | - David Galitz
- Stem Cell & Gene Therapy, Bio-Techne, Minneapolis, MN 55413, USA; (X.L.); (E.P.); (T.N.); (D.G.); (M.A.)
| | - Marnelle Andersen
- Stem Cell & Gene Therapy, Bio-Techne, Minneapolis, MN 55413, USA; (X.L.); (E.P.); (T.N.); (D.G.); (M.A.)
| | - David Grabowski
- Protein Development, Bio-Techne, Minneapolis, MN 55413, USA; (D.G.); (A.P.)
| | - Anthony Person
- Protein Development, Bio-Techne, Minneapolis, MN 55413, USA; (D.G.); (A.P.)
| | - Alex Kalyuzhny
- Antibody Applications, Bio-Techne, Minneapolis, MN 55413, USA;
| | - Kevin C. Flynn
- Stem Cell & Gene Therapy, Bio-Techne, Minneapolis, MN 55413, USA; (X.L.); (E.P.); (T.N.); (D.G.); (M.A.)
- Department of Biochemistry & Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
18
|
Penny TR, Jenkin G, Miller SL, McDonald CA. Umbilical cord blood derived cell expansion: a potential neuroprotective therapy. Stem Cell Res Ther 2024; 15:234. [PMID: 39075614 PMCID: PMC11287950 DOI: 10.1186/s13287-024-03830-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 07/02/2024] [Indexed: 07/31/2024] Open
Abstract
Umbilical cord blood (UCB) is a rich source of beneficial stem and progenitor cells with known angiogenic, neuroregenerative and immune-modulatory properties. Preclinical studies have highlighted the benefit of UCB for a broad range of conditions including haematological conditions, metabolic disorders and neurological conditions, however clinical translation of UCB therapies is lacking. One barrier for clinical translation is inadequate cell numbers in some samples meaning that often a therapeutic dose cannot be achieved. This is particularly important when treating adults or when administering repeat doses of cells. To overcome this, UCB cell expansion is being explored to increase cell numbers. The current focus of UCB cell expansion is CD34+ haematopoietic stem cells (HSCs) for which the main application is treatment of haematological conditions. Currently there are 36 registered clinical trials that are examining the efficacy of expanded UCB cells with 31 of these being for haematological malignancies. Early data from these trials suggest that expanded UCB cells are a safe and feasible treatment option and show greater engraftment potential than unexpanded UCB. Outside of the haematology research space, expanded UCB has been trialled as a therapy in only two preclinical studies, one for spinal cord injury and one for hind limb ischemia. Proteomic analysis of expanded UCB cells in these studies showed that the cells were neuroprotective, anti-inflammatory and angiogenic. These findings are also supported by in vitro studies where expanded UCB CD34+ cells showed increased gene expression of neurotrophic and angiogenic factors compared to unexpanded CD34+ cells. Preclinical evidence demonstrates that unexpanded CD34+ cells are a promising therapy for neurological conditions where they have been shown to improve multiple indices of injury in rodent models of stroke, Parkinson's disease and neonatal hypoxic ischemic brain injury. This review will highlight the current application of expanded UCB derived HSCs in transplant medicine, and also explore the potential use of expanded HSCs as a therapy for neurological conditions. It is proposed that expanded UCB derived CD34+ cells are an appropriate cellular therapy for a range of neurological conditions in children and adults.
Collapse
Affiliation(s)
- Tayla R Penny
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia.
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia.
| | - Graham Jenkin
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Suzanne L Miller
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Courtney A McDonald
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| |
Collapse
|
19
|
Mahmoudi A, Meidany P, Almahmeed W, Jamialahmadi T, Sahebkar A. Stem Cell Therapy as a Potential Treatment of Non-Alcoholic Steatohepatitis-Related End-Stage Liver Disease: A Narrative Review. CURRENT STEM CELL REPORTS 2024; 10:85-107. [DOI: 10.1007/s40778-024-00241-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/17/2024] [Indexed: 01/04/2025]
|
20
|
Rolsma JL, Darch W, Higgins NC, Morgan JT. The tardigrade-derived mitochondrial abundant heat soluble protein improves adipose-derived stem cell survival against representative stressors. Sci Rep 2024; 14:11834. [PMID: 38783150 PMCID: PMC11116449 DOI: 10.1038/s41598-024-62693-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 05/20/2024] [Indexed: 05/25/2024] Open
Abstract
Human adipose-derived stem cell (ASC) grafts have emerged as a powerful tool in regenerative medicine. However, ASC therapeutic potential is hindered by stressors throughout their use. Here we demonstrate the transgenic expression of the tardigrade-derived mitochondrial abundant heat soluble (MAHS) protein for improved ASC resistance to metabolic, mitochondrial, and injection shear stress. In vitro, MAHS-expressing ASCs demonstrate up to 61% increased cell survival following 72 h of incubation in phosphate buffered saline containing 20% media. Following up to 3.5% DMSO exposure for up to 72 h, a 14-49% increase in MAHS-expressing ASC survival was observed. Further, MAHS expression in ASCs is associated with up to 39% improved cell viability following injection through clinically relevant 27-, 32-, and 34-gauge needles. Our results reveal that MAHS expression in ASCs supports survival in response to a variety of common stressors associated with regenerative therapies, thereby motivating further investigation into MAHS as an agent for stem cell stress resistance. However, differentiation capacity in MAHS-expressing ASCs appears to be skewed in favor of osteogenesis over adipogenesis. Specifically, activity of the early bone formation marker alkaline phosphatase is increased by 74% in MAHS-expressing ASCs following 14 days in osteogenic media. Conversely, positive area of the neutral lipid droplet marker BODIPY is decreased by up to 10% in MAHS-transgenic ASCs following 14 days in adipogenic media. Interestingly, media supplementation with up to 40 mM glucose is sufficient to restore adipogenic differentiation within 14 days, prompting further analysis of mechanisms underlying interference between MAHS and differentiation processes.
Collapse
Affiliation(s)
- Jordan L Rolsma
- Department of Bioengineering, University of California, 900 University Ave, Riverside, CA, 92521, USA
| | - William Darch
- Department of Bioengineering, University of California, 900 University Ave, Riverside, CA, 92521, USA
| | - Nicholas C Higgins
- Department of Bioengineering, University of California, 900 University Ave, Riverside, CA, 92521, USA
| | - Joshua T Morgan
- Department of Bioengineering, University of California, 900 University Ave, Riverside, CA, 92521, USA.
| |
Collapse
|
21
|
Kumar R, Mishra N, Tran T, Kumar M, Vijayaraghavalu S, Gurusamy N. Emerging Strategies in Mesenchymal Stem Cell-Based Cardiovascular Therapeutics. Cells 2024; 13:855. [PMID: 38786076 PMCID: PMC11120430 DOI: 10.3390/cells13100855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 05/13/2024] [Accepted: 05/15/2024] [Indexed: 05/25/2024] Open
Abstract
Cardiovascular diseases continue to challenge global health, demanding innovative therapeutic solutions. This review delves into the transformative role of mesenchymal stem cells (MSCs) in advancing cardiovascular therapeutics. Beginning with a historical perspective, we trace the development of stem cell research related to cardiovascular diseases, highlighting foundational therapeutic approaches and the evolution of cell-based treatments. Recognizing the inherent challenges of MSC-based cardiovascular therapeutics, which range from understanding the pro-reparative activity of MSCs to tailoring patient-specific treatments, we emphasize the need to refine the pro-regenerative capacity of these cells. Crucially, our focus then shifts to the strategies of the fourth generation of cell-based therapies: leveraging the secretomic prowess of MSCs, particularly the role of extracellular vesicles; integrating biocompatible scaffolds and artificial sheets to amplify MSCs' potential; adopting three-dimensional ex vivo propagation tailored to specific tissue niches; harnessing the promise of genetic modifications for targeted tissue repair; and institutionalizing good manufacturing practice protocols to ensure therapeutic safety and efficacy. We conclude with reflections on these advancements, envisaging a future landscape redefined by MSCs in cardiovascular regeneration. This review offers both a consolidation of our current understanding and a view toward imminent therapeutic horizons.
Collapse
Affiliation(s)
- Rishabh Kumar
- Department of Biochemistry, Faculty of Science, University of Allahabad, Prayagraj 211002, India
| | - Nitin Mishra
- Department of Biochemistry, Faculty of Science, University of Allahabad, Prayagraj 211002, India
| | - Talan Tran
- Department of Pharmaceutical Sciences, Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, 3200 South University Drive, Fort Lauderdale, FL 33328-2018, USA
| | - Munish Kumar
- Department of Biochemistry, Faculty of Science, University of Allahabad, Prayagraj 211002, India
| | | | - Narasimman Gurusamy
- Department of Pharmaceutical Sciences, Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, 3200 South University Drive, Fort Lauderdale, FL 33328-2018, USA
| |
Collapse
|
22
|
Olatunji G, Kokori E, Yusuf I, Ayanleke E, Damilare O, Afolabi S, Adetunji B, Mohammed S, Akinmoju O, Aboderin G, Aderinto N. Stem cell-based therapies for heart failure management: a narrative review of current evidence and future perspectives. Heart Fail Rev 2024; 29:573-598. [PMID: 37733137 DOI: 10.1007/s10741-023-10351-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/13/2023] [Indexed: 09/22/2023]
Abstract
Heart failure (HF) is a prevalent and debilitating global cardiovascular condition affecting around 64 million individuals, placing significant strain on healthcare systems and diminishing patients' quality of life. The escalating prevalence of HF underscores the urgent need for innovative therapeutic approaches that target the root causes and aim to restore normal cardiac function. Stem cell-based therapies have emerged as promising candidates, representing a fundamental departure from conventional treatments focused primarily on symptom management. This review explores the evolving landscape of stem cell-based therapies for HF management. It delves into the mechanisms of action, clinical evidence from both positive and negative outcomes, ethical considerations, and regulatory challenges. Key findings include the potential for improved cardiac function, enhanced quality of life, and long-term benefits associated with stem cell therapies. However, adverse events and patient vulnerabilities necessitate stringent safety assessments. Future directions in stem cell-based HF therapies include enhancing efficacy and safety through optimized stem cell types, delivery techniques, dosing strategies, and long-term safety assessments. Personalized medicine, combining therapies, addressing ethical and regulatory challenges, and expanding access while reducing costs are crucial aspects of the evolving landscape.
Collapse
Affiliation(s)
- Gbolahan Olatunji
- Department of Medicine and Surgery, University of Ilorin, Ilorin, Nigeria
| | - Emmanuel Kokori
- Department of Medicine and Surgery, University of Ilorin, Ilorin, Nigeria
| | - Ismaila Yusuf
- Department of Medicine and Surgery, Obafemi Awolowo University, Osun, Nigeria
| | - Emmanuel Ayanleke
- Department of Medicine and Surgery, University of Ilorin, Ilorin, Nigeria
| | - Olakanmi Damilare
- Department of Medicine and Surgery, Ladoke Akintola University Teaching Hospital, Ogbomoso, Nigeria
| | - Samson Afolabi
- Department of Medicine and Surgery, Ladoke Akintola University Teaching Hospital, Ogbomoso, Nigeria
| | - Busayo Adetunji
- Department of Medicine and Surgery, Ladoke Akintola University Teaching Hospital, Ogbomoso, Nigeria
| | - Saad Mohammed
- Al-Kindy College of Medicine, University of Baghdad, Baghdad, Iraq
| | | | - Gbolahan Aboderin
- Department of Medicine and Surgery, Ladoke Akintola University Teaching Hospital, Ogbomoso, Nigeria
| | - Nicholas Aderinto
- Department of Medicine and Surgery, Ladoke Akintola University Teaching Hospital, Ogbomoso, Nigeria.
| |
Collapse
|
23
|
Recktenwald M, Hutt E, Davis L, MacAulay J, Daringer NM, Galie PA, Staehle MM, Vega SL. Engineering transcriptional regulation for cell-based therapies. SLAS Technol 2024; 29:100121. [PMID: 38340892 DOI: 10.1016/j.slast.2024.100121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/10/2024] [Accepted: 02/07/2024] [Indexed: 02/12/2024]
Abstract
A major aim in the field of synthetic biology is developing tools capable of responding to user-defined inputs by activating therapeutically relevant cellular functions. Gene transcription and regulation in response to external stimuli are some of the most powerful and versatile of these cellular functions being explored. Motivated by the success of chimeric antigen receptor (CAR) T-cell therapies, transmembrane receptor-based platforms have been embraced for their ability to sense extracellular ligands and to subsequently activate intracellular signal transduction. The integration of transmembrane receptors with transcriptional activation platforms has not yet achieved its full potential. Transient expression of plasmid DNA is often used to explore gene regulation platforms in vitro. However, applications capable of targeting therapeutically relevant endogenous or stably integrated genes are more clinically relevant. Gene regulation may allow for engineered cells to traffic into tissues of interest and secrete functional proteins into the extracellular space or to differentiate into functional cells. Transmembrane receptors that regulate transcription have the potential to revolutionize cell therapies in a myriad of applications, including cancer treatment and regenerative medicine. In this review, we will examine current engineering approaches to control transcription in mammalian cells with an emphasis on systems that can be selectively activated in response to extracellular signals. We will also speculate on the potential therapeutic applications of these technologies and examine promising approaches to expand their capabilities and tighten the control of gene regulation in cellular therapies.
Collapse
Affiliation(s)
- Matthias Recktenwald
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ 08028, USA
| | - Evan Hutt
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ 08028, USA
| | - Leah Davis
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ 08028, USA
| | - James MacAulay
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ 08028, USA
| | - Nichole M Daringer
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ 08028, USA
| | - Peter A Galie
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ 08028, USA
| | - Mary M Staehle
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ 08028, USA
| | - Sebastián L Vega
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ 08028, USA; Department of Orthopaedic Surgery, Cooper Medical School of Rowan University, Camden, NJ 08103, USA.
| |
Collapse
|
24
|
Lee J, Nguyen S, Bhattacharya S. Optic nerve regeneration: Potential treatment approaches. Curr Opin Pharmacol 2024; 74:102428. [PMID: 38171063 PMCID: PMC10922496 DOI: 10.1016/j.coph.2023.102428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/13/2023] [Accepted: 12/14/2023] [Indexed: 01/05/2024]
Abstract
The optic nerve, predominantly constituted by the axons of retinal ganglion cells (RGCs), lacks the ability to regenerate and re-establish function after injury. RGCs are crucial for visual function, and thus, RGC death contributes to the development of numerous progressive neurodegenerative optic neuropathies including glaucoma, ischemic optic neuropathy, and optic neuritis. Regenerating optic nerve axons poses numerous challenges due to factors such as the intricate and inhibitory conditions that exist within their environment, intrinsic breaks to regeneration, and the geometric tortuosity that offers physical hindrance to axon growth. However, recent research advancements offer hope for clinically meaningful regeneration for those who suffer from optic nerve damage. In this review, we highlight the current treatment approaches for optic nerve axon regeneration.
Collapse
Affiliation(s)
- Jessica Lee
- Bascom Palmer Eye Institute, Miller School of Medicine at University of Miami, Miami, FL, USA; College of Medicine, Northeast Ohio Medical University (NEOMED), Rootstown, OH, USA
| | - Sherilyn Nguyen
- Bascom Palmer Eye Institute, Miller School of Medicine at University of Miami, Miami, FL, USA; College of Osteopathic Medicine, Nova Southeastern University, Tampa, FL, USA
| | - Sanjoy Bhattacharya
- Bascom Palmer Eye Institute, Miller School of Medicine at University of Miami, Miami, FL, USA.
| |
Collapse
|
25
|
Bharuka T, Reche A. Advancements in Periodontal Regeneration: A Comprehensive Review of Stem Cell Therapy. Cureus 2024; 16:e54115. [PMID: 38487109 PMCID: PMC10938178 DOI: 10.7759/cureus.54115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 02/13/2024] [Indexed: 03/17/2024] Open
Abstract
Periodontal disease, characterized by inflammation and infection of the supporting structures of teeth, presents a significant challenge in dentistry and public health. Current treatment modalities, while effective to some extent, have limitations in achieving comprehensive periodontal tissue regeneration. This comprehensive review explores the potential of stem cell therapy in advancing the field of periodontal regeneration. Stem cells, including mesenchymal stem cells (MSCs) and induced pluripotent stem cells (iPSCs), hold promise due to their immunomodulatory effects, differentiation potential into periodontal tissues, and paracrine actions. Preclinical studies using various animal models have revealed encouraging outcomes, though standardization and long-term assessment remain challenges. Clinical trials and case studies demonstrate the safety and efficacy of stem cell therapy in real-world applications, especially in personalized regenerative medicine. Patient selection criteria, ethical considerations, and standardized treatment protocols are vital for successful clinical implementation. Stem cell therapy is poised to revolutionize periodontal regeneration, offering more effective, patient-tailored treatments while addressing the systemic health implications of periodontal disease. This transformative approach holds the potential to significantly impact clinical practice and improve the overall well-being of individuals affected by this prevalent oral health concern. Responsible regulatory compliance and a focus on ethical considerations will be essential as stem cell therapy evolves in periodontal regeneration.
Collapse
Affiliation(s)
- Tanvi Bharuka
- Dentistry, Sharad Pawar Dental College and Hospital, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Amit Reche
- Public Health Dentistry, Sharad Pawar Dental College and Hospital, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
26
|
Marto CM, Laranjo M, Gonçalves AC, Paula A, Jorge J, Caetano-Oliveira R, Sousa MI, Oliveiros B, Ramalho-Santos J, Sarmento-Ribeiro AB, Marques-Ferreira M, Cabrita A, Botelho MF, Carrilho E. In Vitro Characterization of Reversine-Treated Gingival Fibroblasts and Their Safety Evaluation after In Vivo Transplantation. Pharmaceutics 2024; 16:207. [PMID: 38399261 PMCID: PMC10892828 DOI: 10.3390/pharmaceutics16020207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/18/2024] [Accepted: 01/25/2024] [Indexed: 02/25/2024] Open
Abstract
Reversine is a purine derivative that has been investigated with regard to its biological effects, such as its anticancer properties and, mostly, its ability to induce the dedifferentiation of adult cells, increasing their plasticity. The obtained dedifferentiated cells have a high potential for use in regenerative procedures, such as regenerative dentistry (RD). Instead of replacing the lost or damaged oral tissues with synthetic materials, RD uses stem cells combined with matrices and an appropriate microenvironment to achieve tissue regeneration. However, the currently available stem cell sources present limitations, thus restricting the potential of RD. Based on this problem, new sources of stem cells are fundamental. This work aims to characterize mouse gingival fibroblasts (GFs) after dedifferentiation with reversine. Different administration protocols were tested, and the cells obtained were evaluated regarding their cell metabolism, protein and DNA contents, cell cycle changes, morphology, cell death, genotoxicity, and acquisition of stem cell characteristics. Additionally, their teratoma potential was evaluated after in vivo transplantation. Reversine caused toxicity at higher concentrations, with decreased cell metabolic activity and protein content. The cells obtained displayed polyploidy, a cycle arrest in the G2/M phase, and showed an enlarged size. Additionally, apoptosis and genotoxicity were found at higher reversine concentrations. A subpopulation of the GFs possessed stem properties, as supported by the increased expression of CD90, CD105, and TERT, the existence of a CD106+ population, and their trilineage differentiation capacity. The dedifferentiated cells did not induce teratoma formation. The extensive characterization performed shows that significant functional, morphological, and genetic changes occur during the dedifferentiation process. The dedifferentiated cells have some stem-like characteristics, which are of interest for RD.
Collapse
Affiliation(s)
- Carlos Miguel Marto
- Institute of Experimental Pathology, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Institute of Biophysics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Institute of Integrated Clinical Practice and Laboratory of Evidence-Based and Precision Dentistry, Faculty of Medicine, University of Coimbra, 3000-075 Coimbra, Portugal (E.C.)
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Area of Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (A.C.G.); (B.O.); (M.M.-F.)
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3004-561 Coimbra, Portugal
| | - Mafalda Laranjo
- Institute of Biophysics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Area of Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (A.C.G.); (B.O.); (M.M.-F.)
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3004-561 Coimbra, Portugal
| | - Ana Cristina Gonçalves
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Area of Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (A.C.G.); (B.O.); (M.M.-F.)
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3004-561 Coimbra, Portugal
- Laboratory of Oncobiology and Hematology (LOH) and University Clinic of Hematology, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Anabela Paula
- Institute of Integrated Clinical Practice and Laboratory of Evidence-Based and Precision Dentistry, Faculty of Medicine, University of Coimbra, 3000-075 Coimbra, Portugal (E.C.)
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Area of Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (A.C.G.); (B.O.); (M.M.-F.)
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3004-561 Coimbra, Portugal
| | - Joana Jorge
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Area of Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (A.C.G.); (B.O.); (M.M.-F.)
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3004-561 Coimbra, Portugal
- Laboratory of Oncobiology and Hematology (LOH) and University Clinic of Hematology, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Rui Caetano-Oliveira
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Area of Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (A.C.G.); (B.O.); (M.M.-F.)
- Pathology Department, Centro Hospitalar e Universitário de Coimbra, 3000-075 Coimbra, Portugal
- Germano de Sousa—Centro de Diagnóstico Histopatológico CEDAP, University of Coimbra, 3000-377 Coimbra, Portugal
| | - Maria Inês Sousa
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Department of Life Sciences, University of Coimbra, 3000-456 Coimbra, Portugal
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Bárbara Oliveiros
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Area of Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (A.C.G.); (B.O.); (M.M.-F.)
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3004-561 Coimbra, Portugal
- Laboratory of Biostatistics and Medical Informatics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - João Ramalho-Santos
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Department of Life Sciences, University of Coimbra, 3000-456 Coimbra, Portugal
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Ana Bela Sarmento-Ribeiro
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Area of Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (A.C.G.); (B.O.); (M.M.-F.)
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3004-561 Coimbra, Portugal
- Laboratory of Oncobiology and Hematology (LOH) and University Clinic of Hematology, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Manuel Marques-Ferreira
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Area of Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (A.C.G.); (B.O.); (M.M.-F.)
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3004-561 Coimbra, Portugal
- Institute of Endodontics, Faculty of Medicine, University of Coimbra, 3000-075 Coimbra, Portugal
| | - António Cabrita
- Institute of Experimental Pathology, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Maria Filomena Botelho
- Institute of Biophysics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Area of Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (A.C.G.); (B.O.); (M.M.-F.)
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3004-561 Coimbra, Portugal
| | - Eunice Carrilho
- Institute of Integrated Clinical Practice and Laboratory of Evidence-Based and Precision Dentistry, Faculty of Medicine, University of Coimbra, 3000-075 Coimbra, Portugal (E.C.)
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Area of Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (A.C.G.); (B.O.); (M.M.-F.)
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3004-561 Coimbra, Portugal
| |
Collapse
|
27
|
Wilson JE, Today BA, Salazar M, Kuo J, Ransom JT, Lightner AL, Chen G, Wong A. Safety of bone marrow derived mesenchymal stem cell extracellular vesicle injection for lumbar facet joint pain. Regen Med 2024; 19:19-26. [PMID: 38327218 DOI: 10.2217/rme-2023-0110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2024] Open
Abstract
Aim: A 3-month pilot study to evaluate the safety of injecting a bone marrow-derived mesenchymal stem cell extracellular vesicle advanced investigational product (IP) into the lumbar facet joint space as a treatment for chronic low back pain. Methods: 20 healthy adults were treated with IP injections (0.5 ml/joint) and evaluated by three functional assessments 1, 3, 7, 14, 30, 60 and 90 days later. Results: No adverse effects or complications occurred across the 3-month follow-up. There were no reports of worsening pain. After 3 months group average scores improved significantly (p < 0.0001) in the Severity Index (65.04%), Interference Index (72.09%) and Oswestry Disability Index (58.43%) assessments. Conclusion: IP injections were safe and associated with significant functional improvements.
Collapse
Affiliation(s)
- James E Wilson
- Interventional Pain Specialists, 28604 Ravine Circle, Covert, MI 49043, USA
| | - Bobbie A Today
- Interventional Pain Specialists, 28604 Ravine Circle, Covert, MI 49043, USA
| | - Maria Salazar
- Interventional Pain Specialists, 28604 Ravine Circle, Covert, MI 49043, USA
| | | | - John T Ransom
- Direct Biologics, 5301 Southwest Parkway, Building 1, Suite 415, Austin, TX 78735, USA
| | - Amy L Lightner
- Direct Biologics, 5301 Southwest Parkway, Building 1, Suite 415, Austin, TX 78735, USA
| | - Grace Chen
- Hudson Medical Group, 160 7th Ave S, NY 10014, USA
| | - Anita Wong
- Hudson Medical Group, 160 7th Ave S, NY 10014, USA
| |
Collapse
|
28
|
Lee CS, Lee M, Na K, Hwang HS. Stem Cell-Derived Extracellular Vesicles for Cancer Therapy and Tissue Engineering Applications. Mol Pharm 2023; 20:5278-5311. [PMID: 37867343 DOI: 10.1021/acs.molpharmaceut.3c00376] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2023]
Abstract
Recently, stem cells and their secretomes have attracted great attention in biomedical applications, particularly extracellular vesicles (EVs). EVs are secretomes of cells for cell-to-cell communication. They play a role as intercellular messengers as they carry proteins, nucleic acids, lipids, and therapeutic agents. They have also been utilized as drug-delivery vehicles due to their biocompatibility, low immunogenicity, stability, targetability, and engineerable properties. The therapeutic potential of EVs can be further enhanced by surface engineering and modification using functional molecules such as aptamers, peptides, and antibodies. As a consequence, EVs hold great promise as effective delivery vehicles for enhancing treatment efficacy while avoiding side effects. Among various cell types that secrete EVs, stem cells are ideal sources of EVs because stem cells have unique properties such as self-renewal and regenerative potential for transplantation into damaged tissues that can facilitate their regeneration. However, challenges such as immune rejection and ethical considerations remain significant hurdles. Stem cell-derived EVs have been extensively explored as a cell-free approach that bypasses many challenges associated with cell-based therapy in cancer therapy and tissue regeneration. In this review, we summarize and discuss the current knowledge of various types of stem cells as a source of EVs, their engineering, and applications of EVs, focusing on cancer therapy and tissue engineering.
Collapse
Affiliation(s)
- Chung-Sung Lee
- Department of Pharmaceutical Engineering, Soonchunhyang University, Asan 31538, Republic of Korea
| | - Min Lee
- Division of Advanced Prosthodontics, University of California, Los Angeles, California 90095, United States
- Department of Bioengineering, University of California, Los Angeles, California 90095, United States
| | - Kun Na
- Department of BioMedical-Chemical Engineering, The Catholic University of Korea, Bucheon 14662, Republic of Korea
- Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Republic of Korea
| | - Hee Sook Hwang
- Department of Pharmaceutical Engineering, Dankook University, Cheonan 31116, Republic of Korea
| |
Collapse
|
29
|
Jhunjhunwala A, Kim J, Kubelick KP, Ethier CR, Emelianov SY. In Vivo Photoacoustic Monitoring of Stem Cell Location and Apoptosis with Caspase-3-Responsive Nanosensors. ACS NANO 2023; 17:17931-17945. [PMID: 37703202 PMCID: PMC10540261 DOI: 10.1021/acsnano.3c04161] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 09/06/2023] [Indexed: 09/15/2023]
Abstract
Stem cell therapy has immense potential in a variety of regenerative medicine applications. However, clinical stem cell therapy is severely limited by challenges in assessing the location and functional status of implanted cells in vivo. Thus, there is a great need for longitudinal, noninvasive stem cell monitoring. Here we introduce a multidisciplinary approach combining nanosensor-augmented stem cell labeling with ultrasound guided photoacoustic (US/PA) imaging for the spatial tracking and functional assessment of transplanted stem cell fate. Specifically, our nanosensor incorporates a peptide sequence that is selectively cleaved by caspase-3, the primary effector enzyme in mammalian cell apoptosis; this cleavage event causes labeled cells to show enhanced optical absorption in the first near-infrared (NIR) window. Optimization of labeling protocols and spectral characterization of the nanosensor in vitro showed a 2.4-fold increase in PA signal from labeled cells during apoptosis while simultaneously permitting cell localization. We then successfully tracked the location and apoptotic status of mesenchymal stem cells in a mouse hindlimb ischemia model for 2 weeks in vivo, demonstrating a 4.8-fold increase in PA signal and spectral slope changes in the first NIR window under proapoptotic (ischemic) conditions. We conclude that our nanosensor allows longitudinal, noninvasive, and nonionizing monitoring of stem cell location and apoptosis, which is a significant improvement over current end-point monitoring methods such as biopsies and histological staining of excised tissue.
Collapse
Affiliation(s)
- Anamik Jhunjhunwala
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, Georgia 30332, United States
| | - Jinhwan Kim
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, Georgia 30332, United States
- School
of Electrical & Computer Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Kelsey P. Kubelick
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, Georgia 30332, United States
- School
of Electrical & Computer Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - C. Ross Ethier
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, Georgia 30332, United States
| | - Stanislav Y. Emelianov
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, Georgia 30332, United States
- School
of Electrical & Computer Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| |
Collapse
|
30
|
Sacco AM, Castaldo C, Di Meglio FD, Nurzynska D, Palermi S, Spera R, Gnasso R, Zinno G, Romano V, Belviso I. The Long and Winding Road to Cardiac Regeneration. APPLIED SCIENCES 2023; 13:9432. [DOI: 10.3390/app13169432] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Cardiac regeneration is a critical endeavor in the treatment of heart diseases, aimed at repairing and enhancing the structure and function of damaged myocardium. This review offers a comprehensive overview of current advancements and strategies in cardiac regeneration, with a specific focus on regenerative medicine and tissue engineering-based approaches. Stem cell-based therapies, which involve the utilization of adult stem cells and pluripotent stem cells hold immense potential for replenishing lost cardiomyocytes and facilitating cardiac tissue repair and regeneration. Tissue engineering also plays a prominent role employing synthetic or natural biomaterials, engineering cardiac patches and grafts with suitable properties, and fabricating upscale bioreactors to create functional constructs for cardiac recovery. These constructs can be transplanted into the heart to provide mechanical support and facilitate tissue healing. Additionally, the production of organoids and chips that accurately replicate the structure and function of the whole organ is an area of extensive research. Despite significant progress, several challenges persist in the field of cardiac regeneration. These include enhancing cell survival and engraftment, achieving proper vascularization, and ensuring the long-term functionality of engineered constructs. Overcoming these obstacles and offering effective therapies to restore cardiac function could improve the quality of life for individuals with heart diseases.
Collapse
Affiliation(s)
- Anna Maria Sacco
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy
| | - Clotilde Castaldo
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy
| | - Franca Di Di Meglio
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy
| | - Daria Nurzynska
- Department of Medicine, Surgery and Dentistry, Scuola Medica Salernitana, University of Salerno, 84081 Baronissi, Italy
| | - Stefano Palermi
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy
| | - Rocco Spera
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy
| | - Rossana Gnasso
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy
| | - Giorgio Zinno
- Department of Clinical and Surgical Medicine, University of Naples Federico II, 80131 Naples, Italy
| | - Veronica Romano
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy
| | - Immacolata Belviso
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy
| |
Collapse
|
31
|
Bragança J, Pinto R, Silva B, Marques N, Leitão HS, Fernandes MT. Charting the Path: Navigating Embryonic Development to Potentially Safeguard against Congenital Heart Defects. J Pers Med 2023; 13:1263. [PMID: 37623513 PMCID: PMC10455635 DOI: 10.3390/jpm13081263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/11/2023] [Accepted: 08/14/2023] [Indexed: 08/26/2023] Open
Abstract
Congenital heart diseases (CHDs) are structural or functional defects present at birth due to improper heart development. Current therapeutic approaches to treating severe CHDs are primarily palliative surgical interventions during the peri- or prenatal stages, when the heart has fully developed from faulty embryogenesis. However, earlier interventions during embryonic development have the potential for better outcomes, as demonstrated by fetal cardiac interventions performed in utero, which have shown improved neonatal and prenatal survival rates, as well as reduced lifelong morbidity. Extensive research on heart development has identified key steps, cellular players, and the intricate network of signaling pathways and transcription factors governing cardiogenesis. Additionally, some reports have indicated that certain adverse genetic and environmental conditions leading to heart malformations and embryonic death may be amendable through the activation of alternative mechanisms. This review first highlights key molecular and cellular processes involved in heart development. Subsequently, it explores the potential for future therapeutic strategies, targeting early embryonic stages, to prevent CHDs, through the delivery of biomolecules or exosomes to compensate for faulty cardiogenic mechanisms. Implementing such non-surgical interventions during early gestation may offer a prophylactic approach toward reducing the occurrence and severity of CHDs.
Collapse
Affiliation(s)
- José Bragança
- Algarve Biomedical Center-Research Institute (ABC-RI), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Champalimaud Research Program, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal
| | - Rute Pinto
- Algarve Biomedical Center-Research Institute (ABC-RI), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
| | - Bárbara Silva
- Algarve Biomedical Center-Research Institute (ABC-RI), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- PhD Program in Biomedical Sciences, Faculty of Medicine and Biomedical Sciences, Universidade do Algarve, 8005-139 Faro, Portugal
| | - Nuno Marques
- Algarve Biomedical Center-Research Institute (ABC-RI), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
| | - Helena S. Leitão
- Algarve Biomedical Center-Research Institute (ABC-RI), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
| | - Mónica T. Fernandes
- Algarve Biomedical Center-Research Institute (ABC-RI), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- School of Health, University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
| |
Collapse
|
32
|
Aderinto N, Abdulbasit MO, Olatunji D. Stem cell-based combinatorial therapies for spinal cord injury: a narrative review of current research and future directions. Ann Med Surg (Lond) 2023; 85:3943-3954. [PMID: 37554849 PMCID: PMC10406006 DOI: 10.1097/ms9.0000000000001034] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 06/22/2023] [Indexed: 08/10/2023] Open
Abstract
Spinal cord injury (SCI) is a devastating condition that can result in lifelong disability. Despite significant progress in SCI research, current treatments only offer limited functional recovery. Stem cell-based combinatorial therapies have emerged promising to enhance neural repair and regeneration after SCI. Combining stem cells with growth factors, biomaterials, and other therapeutic agents can improve outcomes by providing a multifaceted approach to neural repair. However, several challenges must be addressed before these therapies can be widely adopted in clinical practice. Standardisation of stem cell isolation, characterisation, and production protocols ensures consistency and safety in clinical trials. Developing appropriate animal models that accurately mimic human SCI is crucial for successfully translating these therapies. Additionally, optimal delivery methods and biomaterials that support the survival and integration of stem cells into injured tissue must be identified. Despite these challenges, stem cell-based combinatorial therapies for SCI hold great promise. Innovative approaches such as gene editing and the use of neural tissue engineering may further enhance the efficacy of these therapies. Further research and development in this area are critical to advancing the field and providing effective therapies for SCI patients. This paper discusses the current evidence and challenges from the literature on the potential of stem cell-based combinatorial therapies for SCI.
Collapse
Affiliation(s)
- Nicholas Aderinto
- Department of Medicine and Surgery, Ladoke Akintola University of Technology, Ogbomoso
| | | | - Deji Olatunji
- Department of Medicine and Surgery, University of Ilorin, Ilorin, Nigeria
| |
Collapse
|
33
|
Han P, Moran CS, Liu C, Griffiths R, Zhou Y, Ivanovski S. Engineered adult stem cells: Current clinical trials status of disease treatment. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 199:33-62. [PMID: 37678978 DOI: 10.1016/bs.pmbts.2023.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/09/2023]
Abstract
Regenerative medicine is an interdisciplinary field involving the process of replacing and regenerating cells/tissues or organs by integrating medicine, science, and engineering principles to enhance the intrinsic regenerative capacity of the host. Recently, engineered adult stem cells have gained attention for their potential use in regenerative medicine by reducing inflammation and modulating the immune system. This chapter introduces adult stem cell engineering and chimeric antigen receptor T cells (CAR T) gene therapy and summarises current engineered stem cell- and extracellular vesicles (EVs)-focused clinical trial studies that provide the basis for the proposal of a personalised medicine approach to diseases diagnosis and treatment.
Collapse
Affiliation(s)
- Pingping Han
- Center for Oral-facial Regeneration, Rehabilitation and Reconstruction (COR3), Brisbane, QLD, Australia; The University of Queensland, School of Dentistry, Brisbane, QLD, Australia
| | - Corey Stephan Moran
- Center for Oral-facial Regeneration, Rehabilitation and Reconstruction (COR3), Brisbane, QLD, Australia; The University of Queensland, School of Dentistry, Brisbane, QLD, Australia
| | - Chun Liu
- Center for Oral-facial Regeneration, Rehabilitation and Reconstruction (COR3), Brisbane, QLD, Australia; The University of Queensland, School of Dentistry, Brisbane, QLD, Australia
| | | | - Yinghong Zhou
- Center for Oral-facial Regeneration, Rehabilitation and Reconstruction (COR3), Brisbane, QLD, Australia; The University of Queensland, School of Dentistry, Brisbane, QLD, Australia.
| | - Sašo Ivanovski
- Center for Oral-facial Regeneration, Rehabilitation and Reconstruction (COR3), Brisbane, QLD, Australia; The University of Queensland, School of Dentistry, Brisbane, QLD, Australia.
| |
Collapse
|
34
|
Yin J, Qi TF, Yang YY, Vera-Colón M, Zur Nieden NI, Wang Y. Temporal Profiling of Epitranscriptomic Modulators during Osteogenic Differentiation of Human Embryonic Stem Cells. J Proteome Res 2023; 22:2179-2185. [PMID: 37348120 PMCID: PMC10330632 DOI: 10.1021/acs.jproteome.3c00215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/24/2023]
Abstract
Osteogenesis is modulated by multiple regulatory networks. Recent studies showed that RNA modifications and their reader, writer, and eraser (RWE) proteins are involved in regulating various biological processes. Few studies, however, were conducted to investigate the functions of RNA modifications and their RWE proteins in osteogenesis. By using LC-MS/MS in parallel-reaction monitoring (PRM) mode, we performed a comprehensive quantitative assessment of 154 epitranscriptomic RWE proteins throughout the entire time course of osteogenic differentiation in H9 human embryonic stem cells (ESCs). We found that approximately half of the 127 detected RWE proteins were down-regulated during osteogenic differentiation, and they included mainly proteins involved in RNA methylation and pseudouridylation. Protein-protein interaction (PPI) network analysis unveiled significant associations between the down-regulated epitranscriptomic RWE proteins and osteogenesis-related proteins. Gene set enrichment analysis (GSEA) of publicly available RNA-seq data obtained from osteogenesis imperfecta patients suggested a potential role of METTL1 in osteogenesis through the cytokine network. Together, this is the first targeted profiling of epitranscriptomic RWE proteins during osteogenic differentiation of human ESCs, and our work unveiled potential regulatory roles of these proteins in osteogenesis. LC-MS/MS data were deposited on ProteomeXchange (PXD039249).
Collapse
Affiliation(s)
- Jiekai Yin
- Environmental Toxicology Graduate Program, University of California Riverside, Riverside, California 92521-0403, United States
| | - Tianyu F Qi
- Environmental Toxicology Graduate Program, University of California Riverside, Riverside, California 92521-0403, United States
| | - Yen-Yu Yang
- Department of Chemistry, University of California Riverside, Riverside, California 92521-0403, United States
| | - Madeline Vera-Colón
- Environmental Toxicology Graduate Program, University of California Riverside, Riverside, California 92521-0403, United States
| | - Nicole I Zur Nieden
- Environmental Toxicology Graduate Program, University of California Riverside, Riverside, California 92521-0403, United States
- Department of Molecular, Cell, and Systems Biology, University of California, Riverside, California 92521-0403, United States
| | - Yinsheng Wang
- Environmental Toxicology Graduate Program, University of California Riverside, Riverside, California 92521-0403, United States
- Department of Chemistry, University of California Riverside, Riverside, California 92521-0403, United States
| |
Collapse
|
35
|
Maytum A, Edginton-White B, Bonifer C. Identification and characterization of enhancer elements controlling cell type-specific and signalling dependent chromatin programming during hematopoietic development. Stem Cell Investig 2023; 10:14. [PMID: 37404470 PMCID: PMC10316067 DOI: 10.21037/sci-2023-011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 05/24/2023] [Indexed: 07/06/2023]
Abstract
The development of multi-cellular organisms from a single fertilized egg requires to differentially execute the information encoded in our DNA. This complex process is regulated by the interplay of transcription factors with a chromatin environment, both of which provide the epigenetic information maintaining cell-type specific gene expression patterns. Moreover, transcription factors and their target genes form vast interacting gene regulatory networks which can be exquisitely stable. However, all developmental processes originate from pluripotent precursor cell types. The production of terminally differentiated cells from such cells, therefore, requires successive changes of cell fates, meaning that genes relevant for the next stage of differentiation must be switched on and genes not relevant anymore must be switched off. The stimulus for the change of cell fate originates from extrinsic signals which set a cascade of intracellular processes in motion that eventually terminate at the genome leading to changes in gene expression and the development of alternate gene regulatory networks. How developmental trajectories are encoded in the genome and how the interplay between intrinsic and extrinsic processes regulates development is one of the major questions in developmental biology. The development of the hematopoietic system has long served as model to understand how changes in gene regulatory networks drive the differentiation of the various blood cell types. In this review, we highlight the main signals and transcription factors and how they are integrated at the level of chromatin programming and gene expression control. We also highlight recent studies identifying the cis-regulatory elements such as enhancers at the global level and explain how their developmental activity is regulated by the cooperation of cell-type specific and ubiquitous transcription factors with extrinsic signals.
Collapse
Affiliation(s)
- Alexander Maytum
- Institute of Cancer and Genomic Sciences, School of Medicine and Dentistry, University of Birmingham, Birmingham, UK
| | - Ben Edginton-White
- Institute of Cancer and Genomic Sciences, School of Medicine and Dentistry, University of Birmingham, Birmingham, UK
| | - Constanze Bonifer
- Institute of Cancer and Genomic Sciences, School of Medicine and Dentistry, University of Birmingham, Birmingham, UK
| |
Collapse
|
36
|
Alanazi RF, Alhwity BS, Almahlawi RM, Alatawi BD, Albalawi SA, Albalawi RA, Albalawi AA, Abdel-Maksoud MS, Elsherbiny N. Multilineage Differentiating Stress Enduring (Muse) Cells: A New Era of Stem Cell-Based Therapy. Cells 2023; 12:1676. [PMID: 37443710 PMCID: PMC10340735 DOI: 10.3390/cells12131676] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 06/03/2023] [Accepted: 06/17/2023] [Indexed: 07/15/2023] Open
Abstract
Stem cell transplantation has recently demonstrated a significant therapeutic efficacy in various diseases. Multilineage-differentiating stress-enduring (Muse) cells are stress-tolerant endogenous pluripotent stem cells that were first reported in 2010. Muse cells can be found in the peripheral blood, bone marrow and connective tissue of nearly all body organs. Under basal conditions, they constantly move from the bone marrow to peripheral blood to supply various body organs. However, this rate greatly changes even within the same individual based on physical status and the presence of injury or illness. Muse cells can differentiate into all three-germ-layers, producing tissue-compatible cells with few errors, minimal immune rejection and without forming teratomas. They can also endure hostile environments, supporting their survival in damaged/injured tissues. Additionally, Muse cells express receptors for sphingosine-1-phosphate (S1P), which is a protein produced by damaged/injured tissues. Through the S1P-S1PR2 axis, circulating Muse cells can preferentially migrate to damaged sites following transplantation. In addition, Muse cells possess a unique immune privilege system, facilitating their use without the need for long-term immunosuppressant treatment or human leucocyte antigen matching. Moreover, they exhibit anti-inflammatory, anti-apoptotic and tissue-protective effects. These characteristics circumvent all challenges experienced with mesenchymal stem cells and induced pluripotent stem cells and encourage the wide application of Muse cells in clinical practice. Indeed, Muse cells have the potential to break through the limitations of current cell-based therapies, and many clinical trials have been conducted, applying intravenously administered Muse cells in stroke, myocardial infarction, neurological disorders and acute respiratory distress syndrome (ARDS) related to novel coronavirus (SARS-CoV-2) infection. Herein, we aim to highlight the unique biological properties of Muse cells and to elucidate the advantageous difference between Muse cells and other types of stem cells. Finally, we shed light on their current therapeutic applications and the major obstacles to their clinical implementation from laboratory to clinic.
Collapse
Affiliation(s)
- Raghad F. Alanazi
- Pharm D Program, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (R.F.A.); (B.S.A.); (R.M.A.); (B.D.A.); (S.A.A.); (R.A.A.); (A.A.A.)
| | - Basma S. Alhwity
- Pharm D Program, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (R.F.A.); (B.S.A.); (R.M.A.); (B.D.A.); (S.A.A.); (R.A.A.); (A.A.A.)
| | - Raghad M. Almahlawi
- Pharm D Program, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (R.F.A.); (B.S.A.); (R.M.A.); (B.D.A.); (S.A.A.); (R.A.A.); (A.A.A.)
| | - Bashayer D. Alatawi
- Pharm D Program, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (R.F.A.); (B.S.A.); (R.M.A.); (B.D.A.); (S.A.A.); (R.A.A.); (A.A.A.)
| | - Shatha A. Albalawi
- Pharm D Program, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (R.F.A.); (B.S.A.); (R.M.A.); (B.D.A.); (S.A.A.); (R.A.A.); (A.A.A.)
| | - Raneem A. Albalawi
- Pharm D Program, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (R.F.A.); (B.S.A.); (R.M.A.); (B.D.A.); (S.A.A.); (R.A.A.); (A.A.A.)
| | - Amaal A. Albalawi
- Pharm D Program, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (R.F.A.); (B.S.A.); (R.M.A.); (B.D.A.); (S.A.A.); (R.A.A.); (A.A.A.)
| | - Mohamed S. Abdel-Maksoud
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia;
| | - Nehal Elsherbiny
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
37
|
Zavaro M, Dangot A, Bar-Lev TH, Amit O, Avivi I, Ram R, Aharon A. The Role of Extracellular Vesicles (EVs) in Chronic Graft vs. Host Disease, and the Potential Function of Placental Cell-Derived EVs as a Therapeutic Tool. Int J Mol Sci 2023; 24:ijms24098126. [PMID: 37175831 PMCID: PMC10179565 DOI: 10.3390/ijms24098126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/19/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
Chronic graft-versus-host disease (cGVHD) presents with dermal inflammation and fibrosis. We investigated the characteristics of extracellular vesicles (EVs) obtained from cGVHD patients, and their potential effects on human dermal fibroblast (NHDF) cells. The anti-inflammatory and anti-fibrotic effects of placental EVs were also explored given their known anti-inflammatory properties. Fourteen cGVHD patients' EVs contained higher levels of fibrosis-related proteins, TGFβ and α-smooth muscle actin (αSMA), compared to EVs from thirteen healthy subjects. The exposure of NHDF cells to the patients' EVs increased the NHDF cells' TGFβ and αSMA expressions. Placental EVs derived from placental-expanded cells (PLX) (Pluri Inc.) and human villous trophoblast (HVT) cells expressing the mesenchymal markers CD29, CD73, and CD105, penetrated into both the epidermal keratinocytes (HACATs) and NHDF cells. Stimulation of the HACAT cells with cytokine TNFα/INFγ (0.01-0.1 ng/µL) reduced cell proliferation, while the addition of placental EVs attenuated this effect, increasing and normalizing cell proliferation. The treatment of NHDF cells with a combination of TGFβ and placental HVT EVs reduced the stimulatory effects of TGFβ on αSMA production by over 40% (p = 0.0286). In summary, EVs from patients with cGVHD can serve as a biomarker for the cGVHD state. Placental EVs may be used to regulate dermal inflammation and fibrosis, warranting further investigation of their therapeutic potential.
Collapse
Affiliation(s)
- Mor Zavaro
- Hematology Research Laboratory, Hematology Division, Tel-Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel
- The Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6195001, Israel
| | - Ayelet Dangot
- Hematology Research Laboratory, Hematology Division, Tel-Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel
- The Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6195001, Israel
| | - Tali Hana Bar-Lev
- Hematology Research Laboratory, Hematology Division, Tel-Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel
| | - Odelia Amit
- The BMT Unit, Tel Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel
| | - Irit Avivi
- The Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6195001, Israel
- Hematology Department, Tel Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel
| | - Ron Ram
- The Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6195001, Israel
- The BMT Unit, Tel Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel
| | - Anat Aharon
- Hematology Research Laboratory, Hematology Division, Tel-Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel
- The Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6195001, Israel
| |
Collapse
|
38
|
Al-Shammary AA, Hassan SUN. Inconsistencies in Pregnant Mothers' Attitudes and Willingness to Donate Umbilical Cord Stem Cells: A Cross-Sectional Analysis from Saudi Arabia. J Clin Med 2023; 12:jcm12093079. [PMID: 37176520 PMCID: PMC10179412 DOI: 10.3390/jcm12093079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/31/2023] [Accepted: 04/18/2023] [Indexed: 05/15/2023] Open
Abstract
The collection and storage of umbilical cord stem cells (UCSCs) have a crucial role in improving and expanding stem cell-based therapies, which are becoming popular in Saudi Arabia and other Middle East countries. Many patients and families in Saudi Arabia depend on private cord banks in foreign countries to purchase stem cells, which has financial and medical implications. The current study aims at determining the predictors of current registration status and willingness to donate cord blood stem cells among expectant mothers in Saudi Arabia. A cross-sectional study collected data from 714 expectant mothers from all thirteen regions of Saudi Arabia in December 2022. The online survey questionnaire assessed women's awareness, direct and indirect exposure to stem-cell therapy, sources of knowledge, willingness, reluctance, and current registration status to donate cord blood. Although women demonstrated higher acceptance and lower rejection towards the donation of UCSCs, just one percent (n = 7; 1%) of expectant mothers in this sample are registered with the Saudi Stem Cell Registry. Overall, 48% indicated their willingness to register in the future. Both correlational analysis and multiple regression analysis demonstrated that awareness significantly predicted willingness to donate (p < 0.01), and rejection attitudes were negatively related to willingness to donate (p < 0.001). Although the mean scores on acceptance were high, they were not found to be significantly associated with willingness to donate. Prior direct and indirect exposure to stem cell therapy appeared to be the strongest predictor of pregnant women's willingness to register (p < 0.001). Findings suggest that acceptance attitudes do not have a symmetrical relationship with intention. Women's prior exposure to stem cell therapy was the most significant factor; therefore, findings demonstrate that currently women are relying on their firsthand experience to decide about cord blood donation rather than the information obtained from other sources, such as social media and the internet. Though attitudes were not identified as significant predictors in the statistical models, awareness was a relevant factor, and the findings signify increasing awareness in various target populations to enhance the probability of intention to donate cord stem cells.
Collapse
Affiliation(s)
- Asma Ayyed Al-Shammary
- Department of Public Health, College of Public Health and Health Informatics, University of Ha'il, Ha'il 81451, Saudi Arabia
| | - Sehar Un-Nisa Hassan
- Department of Public Health, College of Public Health and Health Informatics, University of Ha'il, Ha'il 81451, Saudi Arabia
| |
Collapse
|
39
|
Xu Y, Rothe R, Voigt D, Sayed A, Huang C, Hauser S, Lee PW, Cui M, Sáenz JP, Boccaccini AR, Zheng K, Pietzsch J, Zhang Y. A self-assembled dynamic extracellular matrix-like hydrogel system with multi-scale structures for cell bioengineering applications. Acta Biomater 2023; 162:211-225. [PMID: 36931420 DOI: 10.1016/j.actbio.2023.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 03/04/2023] [Accepted: 03/09/2023] [Indexed: 03/17/2023]
Abstract
Extracellular matrix (ECM) provides various types of direct interactions with cells and a dynamic environment, which can be remodeled through different assembly/degradation mechanisms to adapt to different biological processes. Herein, through introducing polyphosphate-modified hyaluronic acid and bioactive glass (BG) nano-fibril into a self-assembled hydrogel system with peptide-polymer conjugate, we can realize many new ECM-like functions in a synthetic polymer network. The hydrogel network formation is mediated by coacervation, followed by a gradual transition of peptide structure from α-helix to β-sheet. The ECM-like hydrogels can be degraded through a number of orthogonal mechanisms, including treatments with protease, hyaluronidase, alkaline phosphatase, and calcium ion. As 2D coating, the ECM-like hydrogels can be used to modify the planar surface to promote the adhesion of mesenchymal stromal cells, or to coat the cell surface in a layer-by-layer fashion to shield the interaction with the substrate. As ECM-like hydrogels for 3D cell culture, the system is compatible with injection and cell encapsulation. Upon incorporating fragmented electrospun bioactive glass nano-fibril into the hydrogels, the synergetic effects of soft hydrogel and stiff reinforcement nanofibers on recapitulating ECM functions result in reduced cell circularity in 3D. Finally, by injecting the ECM-like hydrogels into mice, gradual degradations over a time period of one month and high biocompatibility have been shown in vivo. The contribution of complex network dynamics and hierarchical structures to cell-biomatrix interaction can be investigated multi-dimensionally, as many mechanisms are orthogonal to each other and can be regulated individually. STATEMENT OF SIGNIFICANCE: A list of native ECM features has attracted the most interest and attention in the research of synthetic biomaterials. In this research, we have described a simple ECM-like hydrogel system in which the complex and elegant functions of native ECM can be recapitulated in a chemically defined synthetic system. The ECM-like hydrogel systems were developed to avoid undesired features of biological substances (e.g., ethical concerns, batch-to-batch variation, immunogenicity, and potential risk of contamination), as well as gaining new functions to facilitate bioengineering applications (e.g., 3D cell culture, injection, and high stability). To this end, we have developed an ECM-like hydrogel system and provide evidence that this purely synthetic biomaterial is a promising candidate for cell bioengineering applications.
Collapse
Affiliation(s)
- Yong Xu
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, P. R. China; Orthopaedic Institute, Medical College, Soochow University, Suzhou 215006, P. R. China; B CUBE Center for Molecular Bioengineering, Technische Universität Dresden, Dresden 01307, Germany.
| | - Rebecca Rothe
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research Department of Radiopharmaceutical and Chemical Biology, Dresden 01328, Germany; Technische Universität Dresden, School of Science, Faculty of Chemistry and Food Chemistry, Dresden 01062, Germany
| | - Dagmar Voigt
- Institute for Botany, Faculty of Biology, Technische Universität Dresden, Dresden 01062, Germany
| | - Ahmed Sayed
- B CUBE Center for Molecular Bioengineering, Technische Universität Dresden, Dresden 01307, Germany
| | - Can Huang
- Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, P. R. China
| | - Sandra Hauser
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research Department of Radiopharmaceutical and Chemical Biology, Dresden 01328, Germany
| | - Pao-Wan Lee
- B CUBE Center for Molecular Bioengineering, Technische Universität Dresden, Dresden 01307, Germany
| | - Meiying Cui
- B CUBE Center for Molecular Bioengineering, Technische Universität Dresden, Dresden 01307, Germany
| | - James P Sáenz
- B CUBE Center for Molecular Bioengineering, Technische Universität Dresden, Dresden 01307, Germany
| | - Aldo R Boccaccini
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Erlangen 91058, Germany
| | - Kai Zheng
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing 210029, P. R. China; Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, P. R. China.
| | - Jens Pietzsch
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research Department of Radiopharmaceutical and Chemical Biology, Dresden 01328, Germany; Technische Universität Dresden, School of Science, Faculty of Chemistry and Food Chemistry, Dresden 01062, Germany.
| | - Yixin Zhang
- B CUBE Center for Molecular Bioengineering, Technische Universität Dresden, Dresden 01307, Germany; Cluster of Excellence Physics of Life, Technische Universität Dresden, Dresden 01062, Germany.
| |
Collapse
|
40
|
A Simplified and Effective Approach for the Isolation of Small Pluripotent Stem Cells Derived from Human Peripheral Blood. Biomedicines 2023; 11:biomedicines11030787. [PMID: 36979766 PMCID: PMC10045871 DOI: 10.3390/biomedicines11030787] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 02/24/2023] [Accepted: 03/03/2023] [Indexed: 03/08/2023] Open
Abstract
Pluripotent stem cells are key players in regenerative medicine. Embryonic pluripotent stem cells, despite their significant advantages, are associated with limitations such as their inadequate availability and the ethical dilemmas in their isolation and clinical use. The discovery of very small embryonic-like (VSEL) stem cells addressed the aforementioned limitations, but their isolation technique remains a challenge due to their small cell size and their efficiency in isolation. Here, we report a simplified and effective approach for the isolation of small pluripotent stem cells derived from human peripheral blood. Our approach results in a high yield of small blood stem cell (SBSC) population, which expresses pluripotent embryonic markers (e.g., Nanog, SSEA-3) and the Yamanaka factors. Further, a fraction of SBSCs also co-express hematopoietic markers (e.g., CD45 and CD90) and/or mesenchymal markers (e.g., CD29, CD105 and PTH1R), suggesting a mixed stem cell population. Finally, quantitative proteomic profiling reveals that SBSCs contain various stem cell markers (CD9, ITGA6, MAPK1, MTHFD1, STAT3, HSPB1, HSPA4), and Transcription reg complex factors (e.g., STAT5B, PDLIM1, ANXA2, ATF6, CAMK1). In conclusion, we present a novel, simplified and effective isolating process that yields an abundant population of small-sized cells with characteristics of pluripotency from human peripheral blood.
Collapse
|
41
|
Hu L, Morganti S, Nguyen U, Benavides OR, Walsh AJ. Label-free optical imaging of cell function and collagen structure for cell-based therapies. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2023; 25:100433. [PMID: 36642995 PMCID: PMC9836225 DOI: 10.1016/j.cobme.2022.100433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cell-based therapies harness functional cells or tissues to mediate healing and treat disease. Assessment of cellular therapeutics requires methods that are non-destructive to ensure therapies remain viable and uncontaminated for use in patients. Optical imaging of endogenous collagen, by second-harmonic generation, and the metabolic coenzymes NADH and FAD, by autofluorescence microscopy, provides tissue structure and cellular information. Here, we review applications of label-free nonlinear optical imaging of cellular metabolism and collagen second-harmonic generation for assessing cell-based therapies. Additionally, we discuss the potential of label-free imaging for quality control of cell-based therapies, as well as the current limitations and potential future directions of label-free imaging technologies.
Collapse
|
42
|
Azam M, Ghufran H, Tasneem S, Mehmood A, Malik K, Yousaf MA, Tarar MN, Akram SJ, Riazuddin S. Priming of adipose-derived stem cells with curcumin prior to cryopreservation preserves their functional potency: Towards an 'Off-the-shelf' therapy for burns. Cryobiology 2023; 110:69-78. [PMID: 36470459 DOI: 10.1016/j.cryobiol.2022.12.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/18/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022]
Abstract
Stem cells-based treatment for burn wounds require frozen cells as an off-the-shelf therapy; however, cryopreservation-induced oxidative stress resulted in post-thaw cell death or loss of cell functions, thus arrested their clinical practicality. Although antioxidant priming to stem cells increase their resistant to oxidative stress, but this strategy is still unexplored on cryopreserved cells. Herein, we investigated whether curcumin priming before cryopreservation could preserve the therapeutic potency of thawed stem cells. For this, unprimed and curcumin-primed adipose-derived stem cells (ASCs) were cryopreserved for one month. Post-thawing, cells were assessed for viability by trypan blue assay; metabolic activity by MTT assay; senescence by senescence-associated (SA)-β-galactosidase activity staining assay; migration by scratch healing assay and; mRNA expression by real-time PCR. Subsequently, the healing potential was examined by injecting cells around the wound periphery of acidic burn in rats. Post-healing, skin architecture was histologically examined. Results demonstrated that, curcumin-primed frozen cells (Cryo/Cur-ASCs) showed better post-thaw viability, metabolic activity, migration ability and lower percent of senescence comparative to unprimed frozen cells (Cryo/ASCs). Curcumin priming alleviated the oxidative damage by activating the ROS-reducing cellular antioxidant system as shown by the evident increase in GSH levels and upregulated mRNA expression of glutathione peroxidase (GPx), superoxide dismutases (SOD1, SOD2), and catalase (CAT). Further, invivo findings revealed that Cryo/Cur-ASCs-treated wounds exhibited earlier wound closure with an improved architecture comparative to Cryo/ASCs and depicted healing capacity almost similar to Fresh/ASCs. Our findings suggested that curcumin priming could be effective to alleviate the cryo-induced oxidative stress in post-thawed cells.
Collapse
Affiliation(s)
- Maryam Azam
- National Centre of Excellence in Molecular Biology, 87-West Canal Bank Road, University of the Punjab, Lahore, Pakistan
| | - Hafiz Ghufran
- National Centre of Excellence in Molecular Biology, 87-West Canal Bank Road, University of the Punjab, Lahore, Pakistan
| | - Saba Tasneem
- National Centre of Excellence in Molecular Biology, 87-West Canal Bank Road, University of the Punjab, Lahore, Pakistan
| | - Azra Mehmood
- National Centre of Excellence in Molecular Biology, 87-West Canal Bank Road, University of the Punjab, Lahore, Pakistan.
| | - Kausar Malik
- National Centre of Excellence in Molecular Biology, 87-West Canal Bank Road, University of the Punjab, Lahore, Pakistan
| | | | - Moazzam N Tarar
- Jinnah Burn and Reconstructive Surgery Centre, Lahore, Pakistan
| | | | - Sheikh Riazuddin
- National Centre of Excellence in Molecular Biology, 87-West Canal Bank Road, University of the Punjab, Lahore, Pakistan; Jinnah Burn and Reconstructive Surgery Centre, Lahore, Pakistan
| |
Collapse
|
43
|
Shuster-Hyman H, Siddiqui F, Gallagher D, Gauthier-Fisher A, Librach CL. Time course and mechanistic analysis of human umbilical cord perivascular cell mitigation of lipopolysaccharide-induced systemic and neurological inflammation. Cytotherapy 2023; 25:125-137. [PMID: 36473795 DOI: 10.1016/j.jcyt.2022.10.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 10/11/2022] [Accepted: 10/20/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND AIMS Because of their potent immunomodulatory and anti-inflammatory properties, mesenchymal stromal cells are a major focus in the field of stem cell therapy. However, the precise mechanisms underlying this are not entirely understood. Human umbilical cord perivascular cells (HUCPVCs) are a promising cell therapy candidate. This study was designed to evaluate the time course and mechanisms by which HUCPVCs mitigate lipopolysaccharide (LPS)-induced systemic and neurological inflammation in immunocompetent mice. To explore the underlying mechanisms, the authors investigated the biodistribution and fate of HUCPVCs. METHODS Male C57BL/6 mice were randomly allocated to four groups: control, LPS, HUCPVCs or LPS + HUCPVCs. Quantitative polymerase chain reaction, enzyme-linked immunosorbent assay and cytokine arrays were used to assess changes in pro-inflammatory mediators systemically and in the brain. Depressive-like behavioral changes were evaluated via a forced swim test. Quantum dot (qDot) labeling and immunohistochemistry were used to assess the biodistribution and fate of HUCPVCs and interactions with recipient innate immune cells. RESULTS A single intravenously delivered dose of HUCPVCs significantly reduced the systemic inflammation induced by LPS within the first 24 h after administration. HUCPVC treatment abrogated the upregulated expression of pro-inflammatory genes in the hippocampus and cortex and attenuated depressive-like behavior induced by LPS treatment. Biodistribution analysis revealed that HUCPVC-derived qDots rapidly accumulated in the lungs and demonstrated limited in vivo persistence. Furthermore, qDot signals were associated with major recipient innate immune cells and promoted a shift in macrophages toward a regulatory phenotype in the lungs. CONCLUSIONS Overall, this study demonstrates that HUCPVCs can successfully reduce systemic and neurological inflammation induced by LPS within the first 24 h after administration. Biodistribution and fate analyses suggest a critical role for the innate immune system in the HUCPVC-based immunomodulation mechanism.
Collapse
Affiliation(s)
- Hannah Shuster-Hyman
- CReATe Fertility Center, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | - Clifford L Librach
- CReATe Fertility Center, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Department of Obstetrics and Gynecology, University of Toronto, Toronto, Ontario, Canada; Department of Physiology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
44
|
Hamad L, Anthias C, Machin L. How do we forecast tomorrow's transfusion: The future of hematopoietic stem cell donation. Transfus Clin Biol 2023; 30:52-55. [PMID: 36372195 DOI: 10.1016/j.tracli.2022.11.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Lina Hamad
- Lancaster Medical School, Lancaster University, Lancaster, United Kingdom
| | | | - Laura Machin
- Lancaster Medical School, Lancaster University, Lancaster, United Kingdom; Imperial College London.
| |
Collapse
|
45
|
Maracaja L, Khanna AK, Murphy SV, Maracaja DL, Lane MR, Khoury O, Tan J, Damuka N, Crawford FF, Bottoms JA, Miller MD, Kaczka DW, Jordam JE, Sai KKS. Positron Emission Tomography-Computed Tomography Imaging of Selective Lobar Delivery of Stem Cells in Ex Vivo Lung Model of Mechanical Ventilation. J Aerosol Med Pulm Drug Deliv 2023; 36:20-26. [PMID: 36594924 PMCID: PMC9942179 DOI: 10.1089/jamp.2022.0013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Introduction: The delivery of cell therapies may be an important frontier to treat different respiratory diseases in the near future. However, the cell size, delivery conditions, cell viability, and effect in the pulmonary function are critical factors. We performed a proof-of-concept experiment using ex vivo lungs and novel subglottic airway device that allows for selective lobar isolation and administration of drugs and biologics in liquid solution deep into the lung tissues, while simultaneously ventilating the rest of the lung lobes. Methods: We used radiolabeled cells and positron emission tomography-computed tomography (PET-CT) imaging to demonstrate the feasibility of high-yield cell delivery to a specifically targeted lobe. This study proposes an alternative delivery method of live cells labeled with radioactive isotope into the lung parenchyma and tracks the cell delivery using PET-CT imaging. The technique combines selective lobar isolation and lobar infusion to carry large particles distal to the trachea, subtending bronchial segments and reaching alveoli in targeted regions. Results: The solution with cells and carrier achieved a complete and homogeneous lobar distribution. An increase in tissue density was shown on the computed tomography (CT) scan, and the PET-CT imaging demonstrated retention of the activity at central, peripheral lung parenchyma, and pleural surface. The increase in CT density and metabolic activity of the isotope was restricted to the desired lobe only without leak to other lobes. Conclusion: The selective lobe delivery is targeted and imaging-guided by bronchoscopy and CT to a specific diseased lobe during mechanical ventilation. The feasibility of high-yield cell delivery demonstrated in this study will lead to the development of potential novel therapies that contribute to lung health.
Collapse
Affiliation(s)
- Luiz Maracaja
- Department of Anesthesiology, Wake Forest Baptist Medical Center–Wake Forest School of Medicine, Winston-Salem, North Carolina, USA.,Address correspondence to: Luiz Maracaja, MD, Wake Forest Baptist Medical Center, Winston-Salem, NC 27157-0001, USA
| | - Ashish K. Khanna
- Department of Anesthesiology, Wake Forest Baptist Medical Center–Wake Forest School of Medicine, Winston-Salem, North Carolina, USA.,Outcomes Research Consortium, Cleveland, Ohio, USA
| | - Sean V. Murphy
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Danielle L.V. Maracaja
- Department of Pathology, Wake Forest Baptist Medical Center–Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Magan R. Lane
- Department of Cardiothoracic Surgery, Wake Forest Baptist Medical Center–Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Oula Khoury
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Josh Tan
- Department of Radiology, Wake Forest Baptist Medical Center–Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Naresh Damuka
- Department of Radiology, Wake Forest Baptist Medical Center–Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Freda F. Crawford
- Department of Radiology, Wake Forest Baptist Medical Center–Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Joseph A. Bottoms
- Department of Radiology, Wake Forest Baptist Medical Center–Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Mack D. Miller
- Department of Radiology, Wake Forest Baptist Medical Center–Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - David W. Kaczka
- Department of Anesthesia, The University of Iowa Hospital and Clinics, The University of Iowa, Iowa City, Iowa, USA
| | - James Eric Jordam
- Department of Cardiothoracic Surgery, Wake Forest Baptist Medical Center–Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Kiran Kumar Solingapuram Sai
- Department of Radiology, Wake Forest Baptist Medical Center–Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
46
|
Karami Z, Moradi S, Eidi A, Soleimani M, Jafarian A. Induced pluripotent stem cells: Generation methods and a new perspective in COVID-19 research. Front Cell Dev Biol 2023; 10:1050856. [PMID: 36733338 PMCID: PMC9887183 DOI: 10.3389/fcell.2022.1050856] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 11/22/2022] [Indexed: 01/18/2023] Open
Abstract
Induced pluripotent stem cells (iPSCs) exhibit an unlimited ability to self-renew and produce various differentiated cell types, thereby creating high hopes for both scientists and patients as a great tool for basic research as well as for regenerative medicine purposes. The availability and safety of iPSCs for therapeutic purposes require safe and highly efficient methods for production of these cells. Different methods have been used to produce iPSCs, each of which has advantages and disadvantages. Studying these methods would be very helpful in developing an easy, safe, and efficient method for the generation of iPSCs. Since iPSCs can be generated from somatic cells, they can be considered as valuable cellular resources available for important research needs and various therapeutic purposes. Coronavirus disease 2019 (COVID-19) is a disease that has endangered numerous human lives worldwide and currently has no definitive cure. Therefore, researchers have been rigorously studying and examining all aspects of COVID-19 and potential treatment modalities and various drugs in order to enable the treatment, control, and prevention of COVID-19. iPSCs have become one of the most attractive and promising tools in this field by providing the ability to study COVID-19 and the effectiveness of drugs on this disease outside the human body. In this study, we discuss the different methods of generation of iPSCs as well as their respective advantages and disadvantages. We also present recent applications of iPSCs in the study and treatment of COVID-19.
Collapse
Affiliation(s)
- Zahra Karami
- 1Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Sharif Moradi
- 2Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Akram Eidi
- 1Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Masoud Soleimani
- 3Hematology and Cell Therapy Department, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran,4Department of Tissue Engineering and Applied Cell Science, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arefeh Jafarian
- 5Iranian Tissue Bank and Research Center, Tehran University of Medical Sciences, Tehran, Iran,*Correspondence: Arefeh Jafarian,
| |
Collapse
|
47
|
Valeri A, Mazzon E. State of the Art and Future of Stem Cell Therapy in Ischemic Stroke: Why Don't We Focus on Their Administration? BIOENGINEERING (BASEL, SWITZERLAND) 2023; 10:bioengineering10010118. [PMID: 36671691 PMCID: PMC9854993 DOI: 10.3390/bioengineering10010118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 01/17/2023]
Abstract
Stroke is one of the leading causes of death and disability worldwide, so there is an urgent need to find a therapy for the tragic outcomes of this cerebrovascular disease. Stem cells appeared to be a good solution for many conditions, so different experiments were made to establish stem cells as a feasible therapy for stroke. The aim of this review is to analyze the state of the art of stem cell therapy for stroke and if the route of administration could represent a valid adjusting point for ameliorating the therapy's outcome. To obtain this, we searched the scientific literature of the last 10 years for relevant in vitro and in vivo evidence regarding stem cells' potential in stroke therapy. In vitro evidence points to hypoxia, among the preconditioning strategies, as the most used and probably efficient method to enhance cells qualities, while in vivo results raise the question if it is the type of cells or how they are administrated which can make the difference in terms of efficiency. Unfortunately, despite the number of clinical trials, only a few were successfully concluded, demonstrating how urgent the necessity is to translate pre-clinical results into clinics. Since any type of stem cell seems suitable for therapy, the chosen route of administration corresponds to different engraftment rates, distribution and efficiency in terms of the beneficial effects of stem cells. Intravenous administration was widely used for delivering stem cells into the human body, but recently intranasal administration has given promising results in vivo. It allows stem cells to efficiently reach the brain that was precluded to intravenous administration, so it is worth further investigation.
Collapse
|
48
|
Wilton-Clark H, Yokota T. Biological and genetic therapies for the treatment of Duchenne muscular dystrophy. Expert Opin Biol Ther 2023; 23:49-59. [PMID: 36409820 DOI: 10.1080/14712598.2022.2150543] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Duchenne muscular dystrophy is a lethal genetic disease which currently has no cure, and poor standard treatment options largely focused on symptom relief. The development of multiple biological and genetic therapies is underway across various stages of clinical progress which could markedly affect how DMD patients are treated in the future. AREAS COVERED The purpose of this review is to provide an introduction to the different therapeutic modalities currently being studied, as well as a brief description of their progress to date and relative advantages and disadvantages for the treatment of DMD. This review discusses exon skipping therapy, microdystrophin therapy, stop codon readthrough therapy, CRISPR-based gene editing, cell-based therapy, and utrophin upregulation. Secondary therapies addressing nonspecific symptoms of DMD were excluded. EXPERT OPINION Despite the vast potential held by gene replacement therapy options such as microdystrophin production and utrophin upregulation, safety risks inherent to the adeno-associated virus delivery vector might hamper the clinical viability of these approaches until further improvements can be made. Of the mutation-specific therapies, exon skipping therapy remains the most extensively validated and explored option, and the cell-based CAP-1002 therapy may prove to be a suitable adjunct therapy filling the urgent need for cardiac-specific therapies.
Collapse
Affiliation(s)
- Harry Wilton-Clark
- Faculty of Medicine and Dentistry, Department of Medical Genetics, University of Alberta, Edmonton, Alberta, Canada
| | - Toshifumi Yokota
- Faculty of Medicine and Dentistry, Department of Medical Genetics, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
49
|
Unnisa A, Dua K, Kamal MA. Mechanism of Mesenchymal Stem Cells as a Multitarget Disease- Modifying Therapy for Parkinson's Disease. Curr Neuropharmacol 2023; 21:988-1000. [PMID: 35339180 PMCID: PMC10227913 DOI: 10.2174/1570159x20666220327212414] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 02/10/2022] [Accepted: 03/12/2022] [Indexed: 02/08/2023] Open
Abstract
Parkinson's disease (PD) is one of the most prevalent neurodegenerative disorders, affecting the basal nuclei, causing impairment of motor and cognitive functions. Loss of dopaminergic (DAergic) neurons or their degeneration and the aggregation of Lewy bodies is the hallmark of this disease. The medications used to treat PD relieve the symptoms and maintain quality of life, but currently, there is no cure. There is a need for the development of therapies that can cease or perhaps reverse neurodegeneration effectively. With the rapid advancements in cell replacement therapy techniques, medical professionals are trying to find a cure by which restoration of dopamine neurotransmitters can occur. Researchers have started focusing on cell-based therapies using mesenchymal stem cells (MSCs) due to their abundance in the body, the ability of proliferation, and immunomodulation. Here we review the MSC-based treatment in Parkinson's disease and the various mechanisms it repairs DAergic neurons in parkinsonian patients.
Collapse
Affiliation(s)
- Aziz Unnisa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Hail, Hail, Kingdom Saudi Arabia
| | - Kamal Dua
- Graduate School of Health, University of Technology Sydney, NSW 2007, Australia
| | - Mohammad Amjad Kamal
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, China
- King Fahd Medical Research Center, King Abdulaziz University, Saudi Arabia
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Bangladesh
- Enzymoics, 7 Peterlee Place, Hebersham, NSW 2770; Novel Global Community Educational Foundation, Australia
| |
Collapse
|
50
|
Chawla S, Das A. Preclinical-to-clinical innovations in stem cell therapies for liver regeneration. Curr Res Transl Med 2023; 71:103365. [PMID: 36427419 DOI: 10.1016/j.retram.2022.103365] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 08/03/2022] [Accepted: 09/14/2022] [Indexed: 02/06/2023]
Abstract
Acute and chronic liver diseases are the major cause of high morbidity and mortality globally. Liver transplantation is a widely used therapeutic option for liver failure. However, the shortage of availability of liver donors has encouraged research on the alternative approach to liver regeneration. Cell-based regenerative medicine is the best alternative therapy to cater to this need. To date, advanced preclinical approaches have been undertaken on stem cell differentiation and their use in liver tissue engineering for generating efficacious and promising regenerative therapies. Advancements in the bioengineering of stem cells, and organoid generation are the way forward to efficient therapies against liver injury. This review summarizes the recent approaches for stem cell therapy-based liver regeneration and their proof of concepts for clinical application, bioengineering liver organoids to alleviate the liver failure caused due to chronic liver diseases.
Collapse
Affiliation(s)
- Shilpa Chawla
- Department of Applied Biology, Council of Scientific & Industrial Research-Indian Institute of Chemical Technology (CSIR-IICT), Uppal Road, Tarnaka, Hyderabad, TS 500 007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, UP 201 002, India
| | - Amitava Das
- Department of Applied Biology, Council of Scientific & Industrial Research-Indian Institute of Chemical Technology (CSIR-IICT), Uppal Road, Tarnaka, Hyderabad, TS 500 007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, UP 201 002, India.
| |
Collapse
|