1
|
Li J, Zhai X, Yu C. Spatial distribution-based progression of spinal cord injury pathology: a key role for neuroimmune cells. Front Immunol 2025; 15:1505755. [PMID: 39850888 PMCID: PMC11754049 DOI: 10.3389/fimmu.2024.1505755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 12/19/2024] [Indexed: 01/25/2025] Open
Abstract
An external trauma, illness, or other pathological cause can harm the structure and function of the spinal cord, resulting in a significant neurological disorder known as spinal cord injury (SCI). In addition to impairing movement and sensory functions, spinal cord injury (SCI) triggers complex pathophysiological responses, with the spatial dynamics of immune cells playing a key role. The inflammatory response and subsequent healing processes following SCI are profoundly influenced by the spatial distribution and movement of immune cells. Despite significant advances in both scientific and clinical research, SCI therapy still faces several challenges. These challenges primarily stem from our limited understanding of the spatial dynamics of immune cell distribution and the processes that regulate their interactions within the microenvironment following injury. Therefore, a comprehensive investigation into the spatial dynamics of immune cells following SCI is essential to uncover their mechanisms in neuroinflammation and repair, and to develop novel therapeutic strategies.
Collapse
Affiliation(s)
- Jian Li
- Shanghai TCM-Integrated Hospital, Shanghai University of TCM, Department of Neurosurgery, Shanghai, China
| | - Xiaolei Zhai
- Department of Neurosurgery, Shuyang Hospital of Traditional Chinese Medicine, Affiliated Shuyang Hospital of Nanjing University of Chinese Medicine, Shuyang, China
| | - Chaochun Yu
- Shanghai TCM-Integrated Hospital, Shanghai University of TCM, Department of Neurosurgery, Shanghai, China
| |
Collapse
|
2
|
Mitani K, Ito Y, Takene Y, Inaba T. Evaluation of the quality of life-enhancing effect of allogeneic feline adipose mesenchymal stem cells in cats with osteoarthritis: A pilot study. Res Vet Sci 2025; 182:105470. [PMID: 39612738 DOI: 10.1016/j.rvsc.2024.105470] [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: 02/01/2024] [Revised: 11/20/2024] [Accepted: 11/20/2024] [Indexed: 12/01/2024]
Abstract
Osteoarthritis (OA) is a progressive degenerative disease in older cats, and often leads to decreased quality of life (QOL). Mesenchymal stem cells (MSCs) have been used in novel therapies for inflammatory diseases. We aimed to evaluate quantitatively allogeneic adipose-derived MSC (ADSC) therapy in cats with naturally occurring OA, based on QOL assessment resources. To characterize the in vitro properties of ADSCs, we estimated ADSCs from four healthy cats with respect to morphology, differentiation potential, and immunomodulatory potential. Six cats with OA were administered a single intravenous injection of allogeneic ADSCs. Based on the feline musculoskeletal pain index (FMPI), the outcome measure was QOL. The cultured cells were adherent, exhibited a spindle shape without becoming flattened or large, and maintained doubling time until passage 5. After induction, the cells had osteogenic, adipogenic, and chondrogenic phenotypes. These cells expressed CD44 and CD90 and lacked expression of CD14 and CD45, had significantly suppressed the production of interferon -ɤ released from mitogen-stimulated lymphocytes (P < 0.05). The FMPI of all cats with OA significantly increased one month after ADSC therapy (P < 0.05). No adverse effects associated with ADSC administration were observed during follow-up in any of the cats. In conclusion, ADSC therapy with immunomodulatory potential could have beneficial effects on the QOL in cats with OA. Further research is necessary to carry out larger studies of the effectiveness of ADSC therapy.
Collapse
Affiliation(s)
- Kosuke Mitani
- Research and Development Department, J-ARM Co., Ltd., Osaka, Japan
| | - Yuki Ito
- Research and Development Department, J-ARM Co., Ltd., Osaka, Japan
| | - Yukio Takene
- Research and Development Department, J-ARM Co., Ltd., Osaka, Japan
| | - Toshio Inaba
- Faculty of Veterinary Medicine, Okayama University of Science, Ehime, Japan.
| |
Collapse
|
3
|
Abdelbaset S, Mohamed Sob MA, Mutawa G, El-Dein MA, Abou-El-Naga AM. Therapeutic Potential of Different Injection Methods for Bone Marrow Mesenchymal Stem Cell Transplantation in Buslfan-Induced Male Rat Infertility. J Stem Cells Regen Med 2024; 20:26-46. [PMID: 39845505 PMCID: PMC11750064 DOI: 10.46582/jsrm.2002005] [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: 11/06/2023] [Accepted: 03/10/2024] [Indexed: 01/24/2025]
Abstract
Background: In recent years, bone marrow derived mesenchymal stem cells (BM-derived MSCs) have emerged as a powerful cell-based therapy for various diseases, including male infertility. Aim: Demonstrating the efficiency of BM-derived MSCs transplantation by different routes of injection to home and repair testis of busulfan-induced azoospermic rats. Material and methods: In the present study, rat BM-derived MSC was isolated and characterized for mesenchymal &hematopoietic markers using flow-cytometry. Induction of infertility was induced by two successive doses of 10 mg/kg of busulfan. Azoospermic rats were treated by BM-derived MSCs which were injected via various routes (IP, IV, and local in testis). After 60 days; sperm analyses were performed beside mainly Biochemical, histopathological, immunohistological, and ultrastructural investigations. Results: BM-derived MSCs were expressed by CD44+ve, CD105+ve, CD106+ve, CD73+ve, CD34-ve, and CD45-ve. Sperm analysis showed a substantial improvement in sperm morphology, motility, and count following treatment with BM-derived MSCs. Caspase-3 and PCNA immunoexp ression accompanied with the levels of FSH, LH, testosterone, SOD, GSH and MDA depicted a considerable restoration of healthy levels after BM-derived MSCs treatment. The seminiferous tubules showed healthy morphology and spermatozoa were detected in their lumen according to the histopathological and ultrastructural analysis of BM-derived MSCs treated rats. Interestingly, BM-derived MSCs intravenous injection revealed the most significant infertility repair outcomes (P<0.05). Conclusion: Transplanted BM-derived MSCs had the potential to home in rat azoospermic testes and restore spermatogenesis. Consequently, the distinctive characteristics of BM-derived MSCs, such as their ability to differentiate and home, make them a promising cell-based therapeutic option for male infertility.
Collapse
Affiliation(s)
- Samar Abdelbaset
- Mansoura University, Faculty of Science, Zoology department, Mansoura, Dakahlia, Egypt
| | | | - Ghada Mutawa
- Department of Basic Science, Faculty of Dentistry, Horus University-Egypt (HUE), New Damietta 34518, Egypt
| | - Mai Alaa El-Dein
- Mansoura University, Faculty of Science, Zoology department, Mansoura, Dakahlia, Egypt
| | | |
Collapse
|
4
|
Mili B, Choudhary OP. Advancements and mechanisms of stem cell-based therapies for spinal cord injury in animals. Int J Surg 2024; 110:6182-6197. [PMID: 38265419 PMCID: PMC11486964 DOI: 10.1097/js9.0000000000001074] [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: 10/23/2023] [Accepted: 12/24/2023] [Indexed: 01/25/2024]
Abstract
Spinal cord injury (SCI) is a neurodegenerative disorder of the central nervous system that can lead to permanent loss of sensation and voluntary movement beyond the affected area. Extensive preclinical and clinical trials have been conducted to evaluate the safety and effectiveness of stem cells for the treatment of various central nervous system diseases or disorders, including SCI. However, several challenges hinder nerve cell regeneration in the injured spinal cord, such as extensive cell loss, limited neural cell regeneration capacity, axonal disruption, and the presence of growth-inhibiting molecules, particularly astroglial scarring or glial scars at the injury site in chronic cases. These obstacles pose significant challenges for physicians in restoring normal motor and sensory nerve function in both humans and animals following SCI. This review focuses on SCI pathogenesis, the mechanisms underlying the therapeutic potential of mesenchymal stem cells in SCI, and the potential of stem cell-based therapies as promising avenues for treatment. This review article also included relevant preclinical and clinical data from animal studies.
Collapse
Affiliation(s)
- Bhabesh Mili
- Department of Veterinary Physiology and Biochemistry, College of Veterinary Sciences and Animal Husbandry, Central Agricultural University (I), Jalukie, Peren, Nagaland
| | - Om Prakash Choudhary
- Department of Veterinary Anatomy, College of Veterinary Science, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Rampura Phul, Bathinda, Punjab, India
| |
Collapse
|
5
|
Wang X, Hong CG, Duan R, Pang ZL, Zhang MN, Xie H, Liu ZZ. Transplantation of olfactory mucosa mesenchymal stromal cells repairs spinal cord injury by inducing microglial polarization. Spinal Cord 2024; 62:429-439. [PMID: 38849489 DOI: 10.1038/s41393-024-01004-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 05/27/2024] [Accepted: 06/03/2024] [Indexed: 06/09/2024]
Abstract
STUDY DESIGN Animal studies OBJECTIVES: To evaluate the therapeutic effect of olfactory mucosa mesenchymal stem cell (OM-MSCs) transplantation in mice with spinal cord injury (SCI) and to explore the mechanism by which OM-MSCs inhibit neuroinflammation and improve SCI. SETTING Xiangya Hospital, Central South University; Affiliated Hospital of Guangdong Medical University. METHODS Mice (C57BL/6, female, 6-week-old) were randomly divided into sham, SCI, and SCI + OM-MSC groups. The SCI mouse model was generated using Allen's method. OM-MSCs were immediately delivered to the lateral ventricle after SCI using stereotaxic brain injections. One day prior to injury and on days 1, 5, 7, 14, 21, and 28 post-injury, the Basso Mouse Scale and Rivlin inclined plate tests were performed. Inflammation and microglial polarization were evaluated using histological staining, immunofluorescence, and qRT-PCR. RESULTS OM-MSCs originating from the neuroectoderm have great potential in the management of SCI owing to their immunomodulatory effects. OM-MSCs administration improved motor function, alleviated inflammation, promoted the transformation of the M1 phenotype of microglia into the M2 phenotype, facilitated axonal regeneration, and relieved spinal cord injury in SCI mice. CONCLUSIONS OM-MSCs reduced the level of inflammation in the spinal cord tissue, protected neurons, and repaired spinal cord injury by regulating the M1/M2 polarization of microglia.
Collapse
Affiliation(s)
- Xin Wang
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Chun-Gu Hong
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Ran Duan
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Zhi-Lin Pang
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Min-Na Zhang
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Hui Xie
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Zheng-Zhao Liu
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, China.
| |
Collapse
|
6
|
Alqasoumi A, Alsharidah M, Mahmood A, Elsafadi M, Al Rugaie O, Mohany KM, Al-Regaiey KA, Alyahya KI, Alanteet AA, Algarzae NK, AlGhibiwi HK, AlHomaidi A, Abumaree M. Mesenchymal Stem Cell-Conditioned Media Modulate HUVEC Response to H 2O 2: Impact on Gene Expression and Potential for Atherosclerosis Intervention. BIOMED RESEARCH INTERNATIONAL 2024; 2024:7726493. [PMID: 39050921 PMCID: PMC11268959 DOI: 10.1155/2024/7726493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 05/31/2024] [Accepted: 06/13/2024] [Indexed: 07/27/2024]
Abstract
Background: We studied the potential of human bone marrow-derived mesenchymal stem cell conditioned media (hBMSC CM) in protecting endothelial cell properties (viability, proliferation, and migrations) from the deleterious effects produced by the inflammatory environment of H2O2. Additionally, we investigated their impact on the endothelial cells' gene expression of some inflammatory-related genes, namely, TGF-β1, FOS, ATF3, RAF-1, and SMAD3. Methods: Human umbilical vein endothelial cells (HUVECs) were cultured individually under three conditions: alone, with varying concentrations of H2O2, or with varying concentrations of H2O2 and hBMSC CM. HUVEC adhesion, proliferation, and migration were evaluated using the xCELLigence system. The HUVECs' gene expressions were evaluated by real-time polymerase chain reaction (RT-PCR). Results: Generally, we observed enhanced HUVEC viability, proliferation, and migration when cultured in media supplemented with H2O2 and hBMSC CM. Furthermore, the CM modulated the expressions of the studied inflammatory-related genes in HUVECs, promoting a more robust cellular response. Conclusion: This study has illuminated the protective role of hBMSC CM in mitigating the damaging effects of H2O2 on endothelial cell function. Our data demonstrate that hBMSC CM enhances the viability, proliferation, and migration of HUVECs even under oxidative stress conditions. Additionally, the conditioned medium was found to modulate the gene expression of pivotal markers related to inflammation, suggesting a favorable influence on cellular response mechanisms.
Collapse
Affiliation(s)
- Abdulmajeed Alqasoumi
- Department of Pharmacy PracticeCollege of PharmacyQassim University, Qassim, Saudi Arabia
| | - Mansour Alsharidah
- Department of PhysiologyCollege of MedicineQassim University, Qassim, Saudi Arabia
| | - Amer Mahmood
- Stem Cell Unit Department of AnatomyCollege of MedicineKing Saud University, Riyadh, Saudi Arabia
| | - Mona Elsafadi
- Stem Cell Unit Department of AnatomyCollege of MedicineKing Saud University, Riyadh, Saudi Arabia
| | - Osamah Al Rugaie
- Department of Biology and ImmunologyCollege of MedicineQassim University, Qassim, Saudi Arabia
| | - Khalid M. Mohany
- Department of Medical BiochemistryFaculty of MedicineAssiut University, El Gamma Street, Assiut City 71515, Egypt
| | - Khalid A. Al-Regaiey
- Department of PhysiologyCollege of MedicineKing Saud University, Riyadh, Saudi Arabia
| | - Khaleel I. Alyahya
- Department of AnatomyCollege of MedicineKing Saud University, Riyadh, Saudi Arabia
| | - Alaa A. Alanteet
- Department of Pharmacology and ToxicologyCollege of PharmacyKing Saud University, Riyadh, Saudi Arabia
| | - Norah K. Algarzae
- Department of PhysiologyCollege of MedicineKing Saud University, Riyadh, Saudi Arabia
| | - Hanan K. AlGhibiwi
- Department of Pharmacology and ToxicologyCollege of PharmacyKing Saud University, Riyadh, Saudi Arabia
| | - Adel AlHomaidi
- Department of PathologyCollege of MedicineQassim University, Qassim, Saudi Arabia
| | - Mohammad Abumaree
- Stem Cells and Regenerative MedicineCell Therapy and Cancer Research (CTCR)King Abdullah International Medical Research Center (KAIMRC)King Saud Bin Abdulaziz University for Health Sciences (KSAU)King Abdulaziz Medical CityMinistry of National Guard Health Affairs (MNGHA), Riyadh, Saudi Arabia
| |
Collapse
|
7
|
Yuce M, Albayrak E. Paracrine Factors Released from Tonsil-Derived Mesenchymal Stem Cells Inhibit Proliferation of Hematological Cancer Cells Under Hyperthermia in Co-culture Model. Appl Biochem Biotechnol 2024; 196:4105-4124. [PMID: 37897623 DOI: 10.1007/s12010-023-04757-7] [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] [Accepted: 10/17/2023] [Indexed: 10/30/2023]
Abstract
Mesenchymal stem cells (MSCs) are promising biological therapeutic candidates in cancer treatment. As a source of MSCs, palatine tonsil tissue is one of the secondary lymphoid organs that form an essential part of the immune system, and the relation between the secondary lymphoid organs and cancer progression leads us to investigate the effect of tonsil-derived MSCs (T-MSC) on cancer treatment. We aimed to determine the anti-tumoral effects of T-MSCs cultured at the febrile temperature (40 °C) on hematological cancer cell lines. The co-culture of cancer cells with T-MSCs was carried out under fever and normal culture conditions, and then the cell viability was determined by cell counting. In addition, apoptosis rate and cell cycle arrest were determined by flow cytometry. We confirmed the apoptotic effect of T-MSC co-culture at the transcriptional level by using real-time polymerase chain reaction (RT-PCR). We found that co-culture of cancer cells with T-MSCs significantly decreased the viable cell number under the febrile and normal culture conditions. Besides, the T-MSC co-culture induced apoptosis on K562 and MOLT-4 cells and induced the cell cycle arrest at the G2/M phase on MOLT-4 cells. The apoptotic effect of T-MSC co-culture under febrile stimulation was confirmed at the transcriptional level. Our study has highlighted the anti-tumoral effect of the cellular interaction between the T-MSCs and human hematological cancer cells during in vitro co-culture under hyperthermia.
Collapse
Affiliation(s)
- Melek Yuce
- Stem Cell Research & Application Center, Ondokuz Mayıs University, Kurupelit Campus, 55139, Atakum, Samsun, Turkey.
| | - Esra Albayrak
- Stem Cell Research & Application Center, Ondokuz Mayıs University, Kurupelit Campus, 55139, Atakum, Samsun, Turkey
| |
Collapse
|
8
|
Agostini F, Vicinanza C, Lombardi E, Da Ros F, Marangon M, Massarut S, Mazzucato M, Durante C. Ex vivo expansion in a clinical grade medium, containing growth factors from human platelets, enhances migration capacity of adipose stem cells. Front Immunol 2024; 15:1404228. [PMID: 38812519 PMCID: PMC11135042 DOI: 10.3389/fimmu.2024.1404228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 04/30/2024] [Indexed: 05/31/2024] Open
Abstract
Introduction Adipose tissue mesenchymal stem/stromal cells (ASC) can be used as advanced therapy medicinal product in regenerative and cancer medicine. We previously demonstrated Supernatant Rich in Growth Factors (SRGF) can replace fetal bovine serum (FBS) to expand ASC by a clinical grade compliant protocol. The therapeutic potential of ASC is based also on their homing capacity toward inflammatory/cancer sites: oriented cell migration is a fundamental process in this scenario. We investigated the impact of SRGF on ASC migration properties. Methods The motility/migration potential of ASC expanded in 5% SRGF was analyzed, in comparison to 10% FBS, by standard wound healing, bidimensional chemotaxis and transwell assays, and by millifluidic transwell tests. Mechanisms involved in the migration process were investigated by transient protein overexpression. Results In comparison to standard 10% FBS, supplementation of the cell culture medium with 5% SRGF, strongly increased migration properties of ASC along the chemotactic gradient and toward cancer cell derived soluble factors, both in static and millifluidic conditions. We showed that, independently from applied migratory stimulus, SRGF expanded ASC were characterized by far lower expression of α-smooth muscle actin (αSMA), a protein involved in the cell migration machinery. Overexpression of αSMA induced a significant and marked decrease in migration capacity of SRGF expanded ASC. Discussion In conclusion, 5% SRGF addition in the cell culture medium increases the migration potential of ASC, reasonably through appropriate downregulation of αSMA. Thus, SRGF could potentially improve the therapeutic impact of ASC, both as modulators of the immune microenviroment or as targeted drug delivery vehicles in oncology.
Collapse
Affiliation(s)
- Francesco Agostini
- Stem Cell Unit, CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy
| | - Carla Vicinanza
- Stem Cell Unit, CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy
| | | | - Francesco Da Ros
- Stem Cell Unit, CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy
| | - Miriam Marangon
- Stem Cell Unit, CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy
| | - Samuele Massarut
- Breast Cancer Surgery Unit, CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy
| | - Mario Mazzucato
- Stem Cell Unit, CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy
| | - Cristina Durante
- Stem Cell Unit, CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy
| |
Collapse
|
9
|
Poomani MS, Regurajan R, Perumal R, Ramachandran A, Mariappan I, Muthan K, Subramanian V. Differentiation of placenta-derived MSCs cultured in human platelet lysate: a xenofree supplement. 3 Biotech 2024; 14:116. [PMID: 38524240 PMCID: PMC10959853 DOI: 10.1007/s13205-024-03966-z] [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: 03/16/2022] [Accepted: 02/22/2024] [Indexed: 03/26/2024] Open
Abstract
In the last few decades, mesenchymal stem cells (MSCs)-based regenerative therapies in clinical applications have gradually become a hot topic due to their long-term self-renewal and multilineage differentiation ability. In this scenario, placenta (p) has been considered as a good source of MSCs. As a tissue of fetal origin with abundant number of stem cells compared to other sources, their non-invasive acquisition, strong immunosuppression, and lack of ethical concerns make placenta an indispensable source of MSC in stem cell research and therapy. The mesenchymal stem cells were derived from human term placenta (p-MSCs) in xenofree condition using platelet lysate (PL) as a suitable alternative to fetal bovine serum (FBS). Upon isolation, p-MSCs showed plastic adherence with spindle-shaped, fibroblast-like morphology under microscope. p-MSCs flourished well in PL-containing media. Immunophenotyping showed classical MSC markers (> 90%) and lack expression of hematopoietic and HLA-DR (< 1%). Surprisingly, differentiation study showed differentiation of p-MSCs to mature adipocytes in both induced cells and control (spontaneous differentiation), as observed via oil red staining. This is in line with gene expression data where both control and induced cells were positive for visfatin and leptin. Thus, we propose that p-MSCs can be used for clinical applications in the treatment of various chronic and degenerative diseases.
Collapse
Affiliation(s)
- Merlin Sobia Poomani
- Genetic Engineering and Regenerative Biology Lab, Department of Biotechnology, Manonmaniam Sundaranar University, Tirunelveli, Tamil Nadu 627012 India
| | - Rathika Regurajan
- Centre for Marine Science and Technology, Manonmaniam Sundaranar University, Tirunelveli, Tamil Nadu 627012 India
| | | | | | - Iyyadurai Mariappan
- Genetic Engineering and Regenerative Biology Lab, Department of Biotechnology, Manonmaniam Sundaranar University, Tirunelveli, Tamil Nadu 627012 India
| | - Krishnaveni Muthan
- Department of Animal Science, Manonmaniam Sundaranar University, Tirunelveli, Tamil Nadu 627012 India
| | - Venkatesh Subramanian
- Genetic Engineering and Regenerative Biology Lab, Department of Biotechnology, Manonmaniam Sundaranar University, Tirunelveli, Tamil Nadu 627012 India
| |
Collapse
|
10
|
Lee YS, Jun YH, Lee J. Oral administration of bone marrow-derived mesenchymal stem cells attenuates intestinal injury in necrotizing enterocolitis. Clin Exp Pediatr 2024; 67:152-160. [PMID: 38369803 PMCID: PMC10915455 DOI: 10.3345/cep.2023.01151] [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: 08/28/2023] [Revised: 11/01/2023] [Accepted: 11/03/2023] [Indexed: 02/20/2024] Open
Abstract
BACKGROUND Necrotizing enterocolitis (NEC) is a major cause of morbidity in premature infants. However, effective treatment options for NEC are currently lacking. PURPOSE This study aimed to determine the optimal dose of intraperitoneally administered bone marrow-derived mesenchymal stem cells (BM-MSCs) and investigate the therapeutic potential of orally administered BM-MSCs in NEC. METHODS Neonatal mice were fed maternal breast milk for the first 2 days of life. On day 3, the neonatal mice were randomly divided into control, negative control, and BM-MSC-treated groups. Lipopolysaccharide (LPS) was administered for 3 days, and cold stress (4°C, 10 minutes) was applied 3 times a day to induce NEC. High-dose (1×106 cells) or low-dose (1×105 cells) BM-MSCs were administered intraperitoneally 1 or 3 times between days 6 and 8 to treat the NEC. The orally administered group received a low dose of BM-MSCs on day 6. Furthermore, except for the control group, intraepithelial cells (IECs) of the small intestine of neonatal mice were treated with LPS and exposed to 5% O2/95% N2 hypoxic stress for 2 hours. Thereafter, each was treated with BM-MSCs. RESULTS Tissue injury, apoptosis, and inflammatory marker levels were significantly reduced after BM-MSC administration. Oral administration was as effective as intraperitoneal administration, even at a low dose (1×105 cells) of BM-MSCs. The efficacy of high (1×106 cells) or multiple divided doses of BM-MSCs did not differ from that of low-dose treatment. Significantly improved wound healing was observed after BM-MSC administration to injured IECs. CONCLUSION The oral administration of BM-MSCs is a promising treatment option for NEC in infants. Further human studies of BM-MSCs are necessary to determine the optimal dose required to achieve safe and effective outcomes.
Collapse
Affiliation(s)
- Yeong Seok Lee
- Department of Pediatrics, School of Medicine, Inha University, Incheon, Korea
- Department of Medicine, Yesan Public Health Center, Yesan, Korea
| | - Yong Hoon Jun
- Department of Pediatrics, School of Medicine, Inha University, Incheon, Korea
| | - Juyoung Lee
- Department of Pediatrics, School of Medicine, Inha University, Incheon, Korea
| |
Collapse
|
11
|
Najar M, Bouhtit F, Rahmani S, Bouali A, Melki R, Najimi M, Lewalle P, Merimi M. The immunogenic profile and immunomodulatory function of mesenchymal stromal / stem cells in the presence of Ptychotis verticillata. Heliyon 2024; 10:e24822. [PMID: 38317994 PMCID: PMC10838760 DOI: 10.1016/j.heliyon.2024.e24822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 01/15/2024] [Accepted: 01/15/2024] [Indexed: 02/07/2024] Open
Abstract
Mesenchymal stromal/stem cells (MSCs) are considered to be a promising immunotherapeutic tool due to their easy accessibility, culture expansion possibilities, safety profile, and immunomodulatory properties. Although several studies have demonstrated the therapeutic effects of MSCs, their efficacy needs to be improved while also preserving their safety. It has been suggested that cell homeostasis may be particularly sensitive to plant extracts. The impact of natural compounds on immunity is thus a fascinating and growing field. Ptychotis verticillata and its bioactive molecules, carvacrol and thymol, are potential candidates for improving MSC therapeutic effects. They can be used as immunotherapeutic agents to regulate MSC functions and behavior during immunomodulation. Depending on their concentrations and incubation time, these compounds strengthened the immunomodulatory functions of MSCs while maintaining their immune-evasive profile. Incubating MSCs with carvacrol and thymol does not alter their hypoimmunogenicity, as no induction of the allogeneic immune response was observed. MSCs also showed enhanced abilities to reduce the proliferation of activated T cells. Thus, MSCs are immunologically responsive to bioactive molecules derived from PV. The bioactivity may depend on the whole phyto-complex of the oil. These findings may contribute to the development of safe and efficient immunotherapeutic MSCs by using medicinal plant-derived active molecules.
Collapse
Affiliation(s)
- Mehdi Najar
- Faculty of Medicine, ULB721, Université Libre de Bruxelles, 1070 Brussels, Belgium
- Osteoarthritis Research Unit, Department of Medicine, University of Montreal Hospital Research Center (CRCHUM), University of Montreal, Montreal H2X 0A9, QC, Canada
| | - Fatima Bouhtit
- Laboratory of Experimental Hematology, Jules Bordet Institute, Université Libre de Bruxelles, 1000 Brussels, Belgium
- LBBES Laboratory, Genetics and Immune Cell Therapy Unit, Faculty of Sciences, University Mohammed Premier, Oujda 60000, Morocco
| | - Saida Rahmani
- Laboratory of Experimental Hematology, Jules Bordet Institute, Université Libre de Bruxelles, 1000 Brussels, Belgium
| | - Abderrahim Bouali
- LBBES Laboratory, Genetics and Immune Cell Therapy Unit, Faculty of Sciences, University Mohammed Premier, Oujda 60000, Morocco
| | - Rahma Melki
- LBBES Laboratory, Genetics and Immune Cell Therapy Unit, Faculty of Sciences, University Mohammed Premier, Oujda 60000, Morocco
| | - Mustapha Najimi
- Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Philippe Lewalle
- Laboratory of Experimental Hematology, Jules Bordet Institute, Université Libre de Bruxelles, 1000 Brussels, Belgium
| | - Makram Merimi
- LBBES Laboratory, Genetics and Immune Cell Therapy Unit, Faculty of Sciences, University Mohammed Premier, Oujda 60000, Morocco
| |
Collapse
|
12
|
Cao JK, Hong XY, Feng ZC, Li QP. Mesenchymal stem cells-based therapies for severe ARDS with ECMO: a review. Intensive Care Med Exp 2024; 12:12. [PMID: 38332384 PMCID: PMC10853094 DOI: 10.1186/s40635-024-00596-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 01/11/2024] [Indexed: 02/10/2024] Open
Abstract
Acute respiratory distress syndrome (ARDS) is the primary cause of respiratory failure in critically ill patients. Despite remarkable therapeutic advances in recent years, ARDS remains a life-threatening clinical complication with high morbidity and mortality, especially during the global spread of the coronavirus disease 2019 (COVID-19) pandemic. Previous studies have demonstrated that mesenchymal stem cell (MSC)-based therapy is a potential alternative strategy for the treatment of refractory respiratory diseases including ARDS, while extracorporeal membrane oxygenation (ECMO) as the last resort treatment to sustain life can help improve the survival of ARDS patients. In recent years, several studies have explored the effects of ECMO combined with MSC-based therapies in the treatment of ARDS, and some of them have demonstrated that this combination can provide better therapeutic effects, while others have argued that some critical issues need to be solved before it can be applied to clinical practice. This review presents an overview of the current status, clinical challenges and future prospects of ECMO combined with MSCs in the treatment of ARDS.
Collapse
Affiliation(s)
- Jing-Ke Cao
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Xiao-Yang Hong
- Department of Pediatric Intensive Care Unit, Senior Department of Pediatrics, the Seventh Medical Center of PLA General Hospital, NO.5 Nanmencang, Dongcheng District, 100700, Beijing, China
| | - Zhi-Chun Feng
- Department of Neonatology, Senior Department of Pediatrics, the Seventh Medical Center of PLA General Hospital, NO. 5 Nanmencang, Dongcheng District, Beijing, 100700, China
| | - Qiu-Ping Li
- Department of Neonatology, Senior Department of Pediatrics, the Seventh Medical Center of PLA General Hospital, NO. 5 Nanmencang, Dongcheng District, Beijing, 100700, China.
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
13
|
Kang WY, Shin EK, Kim EH, Kang MH, Bang CY, Bang OY, Cha JM. Lyoprotectant Constituents Suited for Lyophilization and Reconstitution of Stem-Cell-Derived Extracellular Vesicles. Biomater Res 2024; 28:0005. [PMID: 38327614 PMCID: PMC10845601 DOI: 10.34133/bmr.0005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 01/01/2024] [Indexed: 02/09/2024] Open
Abstract
Stem-cell-derived extracellular vesicles (EVs) are emerging as an alternative approach to stem cell therapy. Successful lyophilization of EVs could enable convenient storage and distribution of EV medicinal products at room temperature for long periods, thus considerably increasing the accessibility of EV therapeutics to patients. In this study, we aimed to identify an appropriate lyoprotectant composition for the lyophilization and reconstitution of stem-cell-derived EVs. MSC-derived EVs were lyophilized using different lyoprotectants, such as dimethyl sulfoxide, mannitol, trehalose, and sucrose, at varying concentrations. Our results revealed that a mixture of trehalose and sucrose at high concentrations could support the formation of amorphous ice by enriching the amorphous phase of the solution, which successfully inhibited the acceleration of buffer component crystallization during lyophilization. Lyophilized and reconstituted EVs were thoroughly evaluated for concentration and size, morphology, and protein and RNA content. The therapeutic effects of the reconstituted EVs were examined using a tube formation assay with human umbilical vein endothelial cells. After rehydration of the lyophilized EVs, most of their generic characteristics were well-maintained, and their therapeutic capacity recovered to levels similar to those of freshly collected EVs. The concentrations and morphologies of the lyophilized EVs were similar to the initial features of the fresh EV group until day 30 at room temperature, although their therapeutic capacity appeared to decrease after 7 days. Our study suggests an appropriate composition of lyoprotectants, particularly for EV lyophilization, which could encourage the applications of stem-cell-derived EV therapeutics in the health industry.
Collapse
Affiliation(s)
- Wu Young Kang
- Department of Biomedical & Robotics Engineering, College of Engineering,
Incheon National University, Incheon 22012, Republic of Korea
- 3D Stem Cell Bioengineering Laboratory, Research Institute for Engineering and Technology,
Incheon National University, Incheon 22012, Republic of Korea
| | | | - Eun Hee Kim
- S&E bio Co., Ltd., Seoul 06351, Republic of Korea
| | - Min-Ho Kang
- Department of BioMedical-Chemical Engineering (BMCE),
The Catholic University of Korea, Bucheon 14662, Republic of Korea
- Department of Biotechnology,
The Catholic University of Korea, Bucheon 14662, Republic of Korea
| | - Chi Young Bang
- Department of Plastic and Reconstructive Surgery,
Kangwon National University Hospital, Chuncheon 24341, Republic of Korea
| | - Oh Young Bang
- S&E bio Co., Ltd., Seoul 06351, Republic of Korea
- Department of Neurology, Samsung Medical Center,
Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Jae Min Cha
- Department of Biomedical & Robotics Engineering, College of Engineering,
Incheon National University, Incheon 22012, Republic of Korea
- 3D Stem Cell Bioengineering Laboratory, Research Institute for Engineering and Technology,
Incheon National University, Incheon 22012, Republic of Korea
| |
Collapse
|
14
|
Xie Q, Gu J. Therapeutic and Safety Promise of Mesenchymal Stem Cells for Liver Failure: From Preclinical Experiment to Clinical Application. Curr Stem Cell Res Ther 2024; 19:1351-1368. [PMID: 37807649 DOI: 10.2174/011574888x260690230921174343] [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: 05/04/2023] [Revised: 08/11/2023] [Accepted: 08/17/2023] [Indexed: 10/10/2023]
Abstract
Liver failure (LF) is serious liver damage caused by multiple factors, resulting in severe impairment or decompensation of liver synthesis, detoxification, metabolism, and biotransformation. The general prognosis of LF is poor with high mortality in non-transplant patients. The clinical treatments for LF are mainly internal medicine comprehensive care, artificial liver support system, and liver transplantation. However, none of the above treatment strategies can solve the problems of all liver failure patients and has its own limitations. Mesenchymal stem cells (MSCs) are a kind of stem cells with multidirectional differentiation potential and paracrine function, which play an important role in immune regulation and tissue regeneration. In recent years, MSCs have shown multiple advantages in the treatment of LF in pre-clinical experiments and clinical trials. In this work, we reviewed the biological characteristics of MSCs, the possible molecular mechanisms of MSCs in the treatment of liver failure, animal experiments, and clinical application, and also discussed the existing problems of MSCs in the treatment of liver failure.
Collapse
Affiliation(s)
- Qiong Xie
- National Engineering Research Center of Cell Products, AmCellGene Engineering Co., Ltd, Tianjin, 300457, China
| | - Jundong Gu
- National Engineering Research Center of Cell Products, AmCellGene Engineering Co., Ltd, Tianjin, 300457, China
| |
Collapse
|
15
|
Sarvari A, Niasari-Naslaji A, Shirazi A, Heidari B, Boroujeni SB, Moradi MH, Naderi MM, Behzadi B, Mehrazar MM, Dehghan MM. Effect of Intra-ovarian Injection of Mesenchymal Stem Cells or its Conditioned Media on Repeated OPU-IVEP Outcomes in Jersey Heifers and Its Relationship with Follicular Fluid Inflammatory Markers. Avicenna J Med Biotechnol 2024; 16:16-28. [PMID: 38605741 PMCID: PMC11005394 DOI: 10.18502/ajmb.v16i1.14167] [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] [Received: 07/27/2023] [Accepted: 08/26/2023] [Indexed: 04/13/2024] Open
Abstract
Background Repeated Ovum Pick Up (OPU) could have a detrimental effect on ovarian function, reducing In Vitro Embryo Production (IVEP). The present study examined the therapeutic effect of adipose-derived Mesenchymal Stem Cells (MSCs) or its Conditioned Medium (ConM) on ovarian trauma following repeated OPU. Resolvin E1 (RvE1) and Interleukin-12 (IL-12) were investigated as biomarkers. Methods Jersey heifers (n=8) experienced 11 OPU sessions including 5 pre-treatment and 6 treatment sessions. Heifers received intra-ovarian administration of MSCs or ConM (right ovary) and Dulbecco's Modified Phosphate Buffer Saline (DMPBS; left ovary) after OPU in sessions 5 and 8 and 2 weeks after session 11. The concentrations of RvE1 and IL-12 in follicular fluid was evaluated on sessions 1, 5, 6, 9, and 4 weeks after session 11. Following each OPU session, the IVEP parameters were recorded. Results Intra-ovarian administration of MSCs, ConM, and DMPBS did not affect IVEP parameters (p>0.05). The concentration of IL-12 in follicular fluid increased at the last session of pre-treatment (Session 5; p<0.05) and remained elevated throughout the treatment period. There was no correlation between IL-12 and IVEP parameters (p>0.05). However, RvE1 remained relatively high during the pre-treatment and decreased toward the end of treatment period (p<0.05). This in turn was associated with decline in some IVEP parameters (p<0.05). Conclusion Intra-ovarian administration of MSCs or ConM during repeated OPU did not enhance IVEP outcomes in Bos taurus heifers. The positive association between RvE1 and some of IVEP parameters could nominate RvE1 as a promising biomarker to predict IVEP parameters following repeated OPU.
Collapse
Affiliation(s)
- Ali Sarvari
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Amir Niasari-Naslaji
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Abolfazl Shirazi
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Banafsheh Heidari
- Department of Photo Healing and Regeneration, Medical Laser Research Center, Yara Institute, ACECR, Tehran, Iran
| | - Sara Borjian Boroujeni
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Mohammad Hossein Moradi
- Department of Animal Sciences, Faculty of Agriculture and Natural Resources, Arak University, Arak, Iran
| | - Mohammad-Mahdi Naderi
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Bahareh Behzadi
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Mohammad-Mahdi Mehrazar
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Mohammad Mehdi Dehghan
- Department of Surgery and Radiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| |
Collapse
|
16
|
Wang X, Tian H, Yang X, Zhao H, Liang X, Li Y. Mesenchymal Stem Cells‐Derived Extracellular Vesicles in Orthopedic Diseases: Recent Advances and Therapeutic Potential. ADVANCED THERAPEUTICS 2023; 6. [DOI: 10.1002/adtp.202300193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Indexed: 01/06/2025]
Abstract
AbstractEver since the first application of mesenchymal stem cell (MSC) transplantation treating human hematologic malignancies in 1995, MSC‐based treatments have demonstrated great therapeutic potential in clinical settings. However, only a few MSC‐based cell therapy products have been clinically approved. Accumulating evidence suggests that the beneficial effects of MSCs are mainly attributed to the release of paracrine factors or extracellular vesicles (EVs) rather than their mesodermal differentiation potential. Therefore, MSC‐derived EVs (MSC‐EVs), such as exosomes and microvesicles, have merged as promising alternatives to traditional cell‐based therapeutics in clinical practice. They offer several advantages such as better safety, lower immunogenicity, protection of cargoes from degradation, and the ability to overcome biological barriers. Moreover, there have been multiple clinical studies exploring the potential of MSC‐EVs for treating various diseases, including orthopedic disorders. However, there is no definitive “cure” for conditions such as osteoporosis and other bone disorders, but MSC‐EVs have displayed significant therapeutic potential for these orthopedic ailments. Therefore, the objective of this study is to conduct a systematic review of current knowledge related to MSC‐EVs and emphasize their potential application in treating orthopedic diseases, such as bone defects, osteoarthritis, osteoporosis, intervertebral disc degeneration, osteosarcoma, and osteoradionecrosis.
Collapse
Affiliation(s)
- Xinwen Wang
- Department of Foot and Ankle Surgery, Honghui Hospital Xi'an Jiaotong University Xi'an Shaanxi Province 710054 P. R. China
| | - Haodong Tian
- Department of Foot and Ankle Surgery, Honghui Hospital Xi'an Jiaotong University Xi'an Shaanxi Province 710054 P. R. China
| | - Xinquan Yang
- Department of Foot and Ankle Surgery, Honghui Hospital Xi'an Jiaotong University Xi'an Shaanxi Province 710054 P. R. China
| | - Hongmou Zhao
- Department of Foot and Ankle Surgery, Honghui Hospital Xi'an Jiaotong University Xi'an Shaanxi Province 710054 P. R. China
| | - Xiaojun Liang
- Department of Foot and Ankle Surgery, Honghui Hospital Xi'an Jiaotong University Xi'an Shaanxi Province 710054 P. R. China
| | - Yi Li
- Department of Foot and Ankle Surgery, Honghui Hospital Xi'an Jiaotong University Xi'an Shaanxi Province 710054 P. R. China
| |
Collapse
|
17
|
Chen SY, Yang RL, Wu XC, Zhao DZ, Fu SP, Lin FQ, Li LY, Yu LM, Zhang Q, Zhang T. Mesenchymal Stem Cell Transplantation: Neuroprotection and Nerve Regeneration After Spinal Cord Injury. J Inflamm Res 2023; 16:4763-4776. [PMID: 37881652 PMCID: PMC10595983 DOI: 10.2147/jir.s428425] [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: 07/01/2023] [Accepted: 10/03/2023] [Indexed: 10/27/2023] Open
Abstract
Spinal Cord Injury (SCI), with its morbidity characteristics of high disability rate and high mortality rate, is a disease that is highly destructive to both the physiology and psychology of the patient, and for which there is still a lack of effective treatment. Following spinal cord injury, a cascade of secondary injury reactions known as ischemia, peripheral inflammatory cell infiltration, oxidative stress, etc. create a microenvironment that is unfavorable to neural recovery and ultimately results in apoptosis and necrosis of neurons and glial cells. Mesenchymal stem cell (MSC) transplantation has emerged as a more promising therapeutic options in recent years. MSC can promote spinal cord injury repair through a variety of mechanisms, including immunomodulation, neuroprotection, and nerve regeneration, giving patients with spinal cord injury hope. In this paper, it is discussed the neuroprotection and nerve regeneration components of MSCs' therapeutic method for treating spinal cord injuries.
Collapse
Affiliation(s)
- Si-Yu Chen
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China
| | - Rui-Lin Yang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China
| | - Xiang-Chong Wu
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China
| | - De-Zhi Zhao
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China
| | - Sheng-Ping Fu
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China
| | - Feng-Qin Lin
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China
| | - Lin-Yan Li
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China
| | - Li-Mei Yu
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China
| | - Qian Zhang
- Department of Human Anatomy, Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China
| | - Tao Zhang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China
| |
Collapse
|
18
|
Guarnier LP, Moro LG, Lívero FADR, de Faria CA, Azevedo MF, Roma BP, Albuquerque ER, Malagutti-Ferreira MJ, Rodrigues AGD, da Silva AA, Sekiya EJ, Ribeiro-Paes JT. Regenerative and translational medicine in COPD: hype and hope. Eur Respir Rev 2023; 32:220223. [PMID: 37495247 PMCID: PMC10369169 DOI: 10.1183/16000617.0223-2022] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 05/23/2023] [Indexed: 07/28/2023] Open
Abstract
COPD is a common, preventable and usually progressive disease associated with an enhanced chronic inflammatory response in the airways and lung, generally caused by exposure to noxious particles and gases. It is a treatable disease characterised by persistent respiratory symptoms and airflow limitation due to abnormalities in the airways and/or alveoli. COPD is currently the third leading cause of death worldwide, representing a serious public health problem and a high social and economic burden. Despite significant advances, effective clinical treatments have not yet been achieved. In this scenario, cell-based therapies have emerged as potentially promising therapeutic approaches. However, there are only a few published studies of cell-based therapies in human patients with COPD and a small number of ongoing clinical trials registered on clinicaltrials.gov Despite the advances and interesting results, numerous doubts and questions remain about efficacy, mechanisms of action, culture conditions, doses, timing, route of administration and conditions related to homing and engraftment of the infused cells. This article presents the state of the art of cell-based therapy in COPD. Clinical trials that have already been completed and with published results are discussed in detail. We also discuss the questions that remain unanswered about cell-based regenerative and translational medicine for COPD.
Collapse
Affiliation(s)
- Lucas Pires Guarnier
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, Brazil
- Laboratory of Genetics and Cell Therapy - GenTe Cel, Department of Biotechnology, São Paulo State University (UNESP), Assis, Brazil
| | - Lincoln Gozzi Moro
- Laboratory of Genetics and Cell Therapy - GenTe Cel, Department of Biotechnology, São Paulo State University (UNESP), Assis, Brazil
- Biomedical Sciences Institute, Butantan Institute, Technological Research Institute, University of São Paulo (USP), São Paulo, Brazil
| | | | | | - Mauricio Fogaça Azevedo
- Laboratory of Genetics and Cell Therapy - GenTe Cel, Department of Biotechnology, São Paulo State University (UNESP), Assis, Brazil
| | - Beatriz Pizoni Roma
- Laboratory of Genetics and Cell Therapy - GenTe Cel, Department of Biotechnology, São Paulo State University (UNESP), Assis, Brazil
| | | | - Maria José Malagutti-Ferreira
- Laboratory of Genetics and Cell Therapy - GenTe Cel, Department of Biotechnology, São Paulo State University (UNESP), Assis, Brazil
| | | | - Adelson Alves da Silva
- São Lucas Research and Education Institute (IEP - São Lucas), TechLife, São Paulo, Brazil
| | - Eliseo Joji Sekiya
- São Lucas Research and Education Institute (IEP - São Lucas), TechLife, São Paulo, Brazil
| | - João Tadeu Ribeiro-Paes
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, Brazil
- Laboratory of Genetics and Cell Therapy - GenTe Cel, Department of Biotechnology, São Paulo State University (UNESP), Assis, Brazil
| |
Collapse
|
19
|
Tieu A, Stewart DJ, Chwastek D, Lansdell C, Burger D, Lalu MM. Biodistribution of mesenchymal stromal cell-derived extracellular vesicles administered during acute lung injury. Stem Cell Res Ther 2023; 14:250. [PMID: 37705086 PMCID: PMC10500845 DOI: 10.1186/s13287-023-03472-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 08/23/2023] [Indexed: 09/15/2023] Open
Abstract
BACKGROUND Mesenchymal stromal cell-derived extracellular vesicles (MSC-EVs) are a promising cell-free therapy for acute lung injury (ALI). To date, no studies have investigated their biodistribution in ALI or discerned the timing of administration for maximal lung targeting, which are crucial considerations for clinical translation. Our study aimed to characterize a mouse model of ALI and establish the distribution kinetics and optimal timing of MSC-EV delivery during lung injury. METHODS MSC-EVs were isolated by ultracentrifugation alone (U/C) or tangential flow filtration with ultracentrifugation (TFF-U/C) and characterized by nanoparticle tracking analysis and western blot. A lipopolysaccharide (LPS)-induced mouse model of ALI was established to study the inflammatory response over 72 h. ALI was assessed by histological lung injury score, bronchoalveolar lavage fluid cell count and inflammatory cytokines. For biodistribution studies, ALI mice were intravenously administered fluorescently labeled MSC-EVs to determine the optimal timing of administration and organ-specific biodistribution. Live in vivo and ex vivo fluorescence imaging was conducted at various timepoints post-EV injection. RESULTS EVs isolated by either ultracentrifugation alone or TFF-U/C displayed comparable size distribution (~ 50-350 nm) and EV marker expression (CD63/81). TFF-U/C generated a 5.4-fold higher particle concentration and 3.9-fold higher total protein when compared to ultracentrifugation alone. From the inflammatory time-course study, cell count and IL-1β peaked in bronchoalveolar lavage fluid at 24 h after ALI induction. MSC-EVs delivered at 24 h (as opposed to 0.5 h, 5 h or 10 h) after disease induction resulted in a 2.7-4.4-fold higher lung uptake of EVs. Biodistribution studies comparing organ-specific MSC-EV uptake showed progressive lung accumulation up to 48 h post-delivery (threefold higher than the spleen/liver), with a decline at 72 h. Importantly, lung EV fluorescence at 48 h in ALI mice was significantly elevated as compared to control mice. The lung tropism of MSC-EVs was further validated as therapeutically inert EVs derived from HEK293T cells accumulated mainly to the spleen and liver with a 5.5-fold lower distribution to the lungs as compared to MSC-EVs. CONCLUSION MSC-EVs exhibit maximal lung accumulation when administered during heightened inflammation at 24 h after ALI induction. This lung tropism suggests that MSC-EVs may serve as a practical rescue treatment for acute inflammatory respiratory conditions.
Collapse
Affiliation(s)
- Alvin Tieu
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8L1, Canada
- Department of Anesthesiology and Pain Medicine, University of Ottawa, Ottawa, ON, K1H 8L1, Canada
- Clinical Epidemiology Program, BLUEPRINT Translational Research Group, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
| | - Duncan J Stewart
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8L1, Canada
- Department of Medicine, University of Ottawa, Ottawa, ON, K1H 8L1, Canada
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
| | - Damian Chwastek
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8L1, Canada
- Clinical Epidemiology Program, BLUEPRINT Translational Research Group, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
| | - Casey Lansdell
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8L1, Canada
- Clinical Epidemiology Program, BLUEPRINT Translational Research Group, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
| | - Dylan Burger
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8L1, Canada
- Chronic Disease Program, Kidney Research Centre, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
- School of Pharmaceutical Sciences, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Manoj M Lalu
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8L1, Canada.
- Department of Anesthesiology and Pain Medicine, University of Ottawa, Ottawa, ON, K1H 8L1, Canada.
- Clinical Epidemiology Program, BLUEPRINT Translational Research Group, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada.
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada.
| |
Collapse
|
20
|
Shimizu Y, Ntege EH, Azuma C, Uehara F, Toma T, Higa K, Yabiku H, Matsuura N, Inoue Y, Sunami H. Management of Rheumatoid Arthritis: Possibilities and Challenges of Mesenchymal Stromal/Stem Cell-Based Therapies. Cells 2023; 12:1905. [PMID: 37508569 PMCID: PMC10378234 DOI: 10.3390/cells12141905] [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: 05/06/2023] [Revised: 06/28/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
Rheumatoid arthritis (RA) is a highly prevalent, chronic, and progressive autoimmune disorder primarily affecting joints and muscles. The associated inflammation, pain, and motor restriction negatively impact patient quality of life (QOL) and can even contribute to premature mortality. Further, conventional treatments such as antiinflammatory drugs are only symptomatic. Substantial progress has been made on elucidating the etiopathology of overt RA, in particular the contributions of innate and adaptive immune system dysfunction to chronic inflammation. Although the precise mechanisms underlying onset and progression remain elusive, the discovery of new drug targets, early diagnosis, and new targeted treatments have greatly improved the prognosis and QOL of patients with RA. However, a sizable proportion of patients develop severe adverse effects, exhibit poor responses, or cannot tolerate long-term use of these drugs, necessitating more effective and safer therapeutic alternatives. Mounting preclinical and clinical evidence suggests that the transplantation of multipotent adult stem cells such as mesenchymal stromal/stem cells is a safe and effective treatment strategy for controlling chronic inflammation and promoting tissue regeneration in patients with intractable diseases, including RA. This review describes the current status of MSC-based therapies for RA as well as the opportunities and challenges to broader clinical application.
Collapse
Affiliation(s)
- Yusuke Shimizu
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, Nishihara 903-0215, Japan
| | - Edward Hosea Ntege
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, Nishihara 903-0215, Japan
| | - Chinatsu Azuma
- Department of Orthopedic Surgery, Graduate School of Medicine, University of the Ryukyus, Nishihara 903-0215, Japan
| | - Fuminari Uehara
- Department of Orthopedic Surgery, Graduate School of Medicine, University of the Ryukyus, Nishihara 903-0215, Japan
| | - Takashi Toma
- Department of Orthopedic Surgery, Graduate School of Medicine, University of the Ryukyus, Nishihara 903-0215, Japan
| | - Kotaro Higa
- Department of Orthopedic Surgery, Graduate School of Medicine, University of the Ryukyus, Nishihara 903-0215, Japan
| | - Hiroki Yabiku
- Department of Orthopedic Surgery, Graduate School of Medicine, University of the Ryukyus, Nishihara 903-0215, Japan
| | - Naoki Matsuura
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, Nishihara 903-0215, Japan
| | - Yoshikazu Inoue
- Department of Plastic and Reconstructive Surgery, School of Medicine, Fujita Health University, Toyoake 470-1192, Japan
| | - Hiroshi Sunami
- Center for Advanced Medical Research, School of Medicine, University of the Ryukyus, Nishihara 903-0215, Japan
| |
Collapse
|
21
|
Chen W, Lv L, Chen N, Cui E. Immunogenicity of mesenchymal stromal/stem cells. Scand J Immunol 2023; 97:e13267. [PMID: 39007962 DOI: 10.1111/sji.13267] [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: 08/29/2022] [Revised: 03/06/2023] [Accepted: 03/17/2023] [Indexed: 04/07/2023]
Abstract
Mesenchymal stromal/stem cells (MSCs) possess the ability to self-renew and differentiate into other cell types. Because of their anti-inflammatory and immunomodulatory abilities, as well as their more ready availability compared to other stem cell sources, MSCs hold great promise for the treatment of many diseases, such as haematological defects, acute respiratory distress syndrome, autoimmunity, cardiovascular diseases, etc. However, immune rejection remains an important problem. MSCs are considered to have low immunogenicity, but they do not have full immunological privilege. This review analyzes and discusses the safety of MSCs from the perspective of their immunogenicity, with the aim of providing a reference for future research and clinical application.
Collapse
Affiliation(s)
- Wenyan Chen
- Department of Respiratory and Critical Care Medicine, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, 313000, Zhejiang, No. 1558, Third Ring North Road, Huzhou, China
| | - Lu Lv
- Department of Respiratory and Critical Care Medicine, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, 313000, Zhejiang, No. 1558, Third Ring North Road, Huzhou, China
| | - Na Chen
- Department of Respiratory and Critical Care Medicine, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, 313000, Zhejiang, No. 1558, Third Ring North Road, Huzhou, China
| | - Enhai Cui
- Department of Respiratory and Critical Care Medicine, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, 313000, Zhejiang, No. 1558, Third Ring North Road, Huzhou, China
| |
Collapse
|
22
|
Ulpiano C, da Silva CL, Monteiro GA. Bioengineered Mesenchymal-Stromal-Cell-Derived Extracellular Vesicles as an Improved Drug Delivery System: Methods and Applications. Biomedicines 2023; 11:biomedicines11041231. [PMID: 37189850 DOI: 10.3390/biomedicines11041231] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/30/2023] [Accepted: 04/12/2023] [Indexed: 05/17/2023] Open
Abstract
Extracellular vesicles (EVs) are cell-derived nano-sized lipid membranous structures that modulate cell-cell communication by transporting a variety of biologically active cellular components. The potential of EVs in delivering functional cargos to targeted cells, their capacity to cross biological barriers, as well as their high modification flexibility, make them promising drug delivery vehicles for cell-free therapies. Mesenchymal stromal cells (MSCs) are known for their great paracrine trophic activity, which is largely sustained by the secretion of EVs. MSC-derived EVs (MSC-EVs) retain important features of the parental cells and can be bioengineered to improve their therapeutic payload and target specificity, demonstrating increased therapeutic potential in numerous pre-clinical animal models, including in the treatment of cancer and several degenerative diseases. Here, we review the fundamentals of EV biology and the bioengineering strategies currently available to maximize the therapeutic value of EVs, focusing on their cargo and surface manipulation. Then, a comprehensive overview of the methods and applications of bioengineered MSC-EVs is presented, while discussing the technical hurdles yet to be addressed before their clinical translation as therapeutic agents.
Collapse
Affiliation(s)
- Cristiana Ulpiano
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Cláudia L da Silva
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Gabriel A Monteiro
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| |
Collapse
|
23
|
Foreman M, Maddy K, Patel A, Reddy A, Costello M, Lucke-Wold B. Differentiating Lumbar Spinal Etiology from Peripheral Plexopathies. Biomedicines 2023; 11:756. [PMID: 36979737 PMCID: PMC10044821 DOI: 10.3390/biomedicines11030756] [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: 02/04/2023] [Revised: 02/20/2023] [Accepted: 02/25/2023] [Indexed: 03/06/2023] Open
Abstract
Clinicians have managed and treated lower back pain since the earliest days of practice. Historically, lower back pain and its accompanying symptoms of radiating leg pain and muscle weakness have been recognized to be due to any of the various lumbar spine pathologies that lead to the compression of the lumbar nerves at the root, the most common of which is the radiculopathy known as sciatica. More recently, however, with the increased rise in chronic diseases, the importance of differentially diagnosing a similarly presenting pathology, known as lumbosacral plexopathy, cannot be understated. Given the similar clinical presentation of lumbar spine pathologies and lumbosacral plexopathies, it can be difficult to differentiate these two diagnoses in the clinical setting. Resultingly, the inappropriate diagnosis of either pathology can result in ineffective clinical management. Thus, this review aims to aid in the clinical differentiation between lumbar spine pathology and lumbosacral plexopathy. Specifically, this paper delves into spine and plexus anatomy, delineates the clinical assessment of both pathologies, and highlights powerful diagnostic tools in the hopes of bolstering appropriate diagnosis and treatment. Lastly, this review will describe emerging treatment options for both pathologies in the preclinical and clinical realms, with a special emphasis on regenerative nerve therapies.
Collapse
Affiliation(s)
- Marco Foreman
- Department of Neurosurgery, University of Florida, Gainesville, FL 32610, USA
| | - Krisna Maddy
- Department of Neurosurgery, University of Miami, Miami, FL 33136, USA
| | - Aashay Patel
- Department of Neurosurgery, University of Florida, Gainesville, FL 32610, USA
| | - Akshay Reddy
- Department of Neurosurgery, University of Florida, Gainesville, FL 32610, USA
| | - Meredith Costello
- Department of Neurosurgery, University of Miami, Miami, FL 33136, USA
| | - Brandon Lucke-Wold
- Department of Neurosurgery, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
24
|
Zhang B, Chen G, Wu X, Li Y, Xiao Y, Li J, He L, Li Y, Wang S, Zhao J, Liu C, Zhou H, Li Y, Pei X. Biomimetic Prussian blue nanozymes with enhanced bone marrow-targeting for treatment of radiation-induced hematopoietic injury. Biomaterials 2023; 293:121980. [PMID: 36580722 DOI: 10.1016/j.biomaterials.2022.121980] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 12/18/2022] [Accepted: 12/21/2022] [Indexed: 12/24/2022]
Abstract
There is an urgent medical need to develop effective therapies that can ameliorate damage to the radiation-exposed hematopoietic system. Nanozymes with robust antioxidant properties have a therapeutic potential for mitigating radiation-induced hematopoietic injury. However, enhancing nanozyme recruitment to injured tissues in vivo while maintaining their catalytic activity remains a great challenge. Herein, we present the design and preparation of a biomimetic nanoparticle, a mesenchymal stem cell membrane camouflaged Prussian blue nanozyme (PB@MSCM), which exhibits biocompatible surface properties and demonstrates enhanced injury site-targeting towards the irradiated murine bone marrow niche. Notably, the constructed PB@MSCM possessed redox enzyme-mimic catalytic activity and could scavenge overproduced reactive oxygen species in the irradiated bone marrow cells, both in vitro and ex vivo. More importantly, the administration of PB@MSCM significantly mitigated hematopoietic cell apoptosis and accelerated the regeneration of hematopoietic stem and progenitor cells. Our findings provide a new targeted strategy to improve nanozyme therapy in vivo and mitigate radiation-induced hematopoietic injury.
Collapse
Affiliation(s)
- Bowen Zhang
- Beijing Institute of Radiation Medicine, Beijing 100850, China; South China Research Center for Stem Cell & Regenerative Medicine, SCIB, Guangzhou 510005, China
| | - Gan Chen
- Institute of Health Service and Transfusion Medicine, Beijing 100850, China
| | - Xumin Wu
- Beijing Institute of Radiation Medicine, Beijing 100850, China; South China Research Center for Stem Cell & Regenerative Medicine, SCIB, Guangzhou 510005, China
| | - Yunxing Li
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Yao Xiao
- Institute of Health Service and Transfusion Medicine, Beijing 100850, China
| | - Jisheng Li
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Lijuan He
- Institute of Health Service and Transfusion Medicine, Beijing 100850, China; South China Research Center for Stem Cell & Regenerative Medicine, SCIB, Guangzhou 510005, China
| | - Yunqiao Li
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Sihan Wang
- Beijing Institute of Radiation Medicine, Beijing 100850, China; South China Research Center for Stem Cell & Regenerative Medicine, SCIB, Guangzhou 510005, China
| | - Jiahui Zhao
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Chuanli Liu
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Hong Zhou
- Institute of Health Service and Transfusion Medicine, Beijing 100850, China.
| | - Yanhua Li
- Beijing Institute of Radiation Medicine, Beijing 100850, China; South China Research Center for Stem Cell & Regenerative Medicine, SCIB, Guangzhou 510005, China.
| | - Xuetao Pei
- Beijing Institute of Radiation Medicine, Beijing 100850, China; South China Research Center for Stem Cell & Regenerative Medicine, SCIB, Guangzhou 510005, China.
| |
Collapse
|
25
|
Zhang B, Chen G, Wu X, Li Y, Xiao Y, Li J, He L, Li Y, Wang S, Zhao J, Liu C, Zhou H, Li Y, Pei X. Biomimetic Prussian blue nanozymes with enhanced bone marrow-targeting for treatment of radiation-induced hematopoietic injury. Biomaterials 2023; 293:121980. [DOI: https:/doi.org/10.1016/j.biomaterials.2022.121980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2023]
|
26
|
Placental mesenchymal stem cells restore glucose and energy homeostasis in obesogenic adipocytes. Cell Tissue Res 2023; 391:127-144. [PMID: 36227376 DOI: 10.1007/s00441-022-03693-y] [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: 12/08/2021] [Accepted: 09/14/2022] [Indexed: 01/18/2023]
Abstract
Obesity (Ob) depicts a state of energy imbalance(s) being characterized by the accumulation of excessive fat and which predisposes to several metabolic diseases. Mesenchymal stem cells (MSCs) represent a promising option for addressing obesity and its associated metabolic co-morbidities. The present study aims at assessing the beneficial effects of human placental MSCs (P-MSCs) in mitigating Ob-associated insulin resistance (IR) and mitochondrial dysfunction both in vivo and in vitro. Under obesogenic milieu, adipocytes showed a significant reduction in glucose uptake, and impaired insulin signaling with decreased expression of UCP1 and PGC1α, suggestive of dysregulated non-shivering thermogenesis vis-a-vis mitochondrial biogenesis respectively. Furthermore, obesogenic adipocytes demonstrated impaired mitochondrial respiration and energy homeostasis evidenced by reduced oxygen consumption rate (OCR) and blunted ATP/NAD+/NADP+ production respectively. Interestingly, co-culturing adipocytes with P-MSCs activated PI3K-Akt signaling, improved glucose uptake, diminished ROS production, enhanced mitochondrial OCR, improved ATP/NAD+/NADP+ production, and promoted beiging of adipocytes evidenced by upregulated expression of PRDM16, UCP1, and PGC1α expression. In vivo, P-MSCs administration increased the peripheral blood glucose uptake and clearance, and improved insulin sensitivity and lipid profile with a coordinated increase in the ratio of ATP/ADP and NAD+ and NADP+ in the white adipose tissue (WAT), exemplified in WNIN/GR-Ob obese mutant rats. In line with in vitro findings, there was a significant reduction in adipocyte hypertrophy, increased mitochondrial staining, and thermogenesis. Our findings advocate for a therapeutic application of P-MSCs for improving glucose and energy homeostasis, i.e., probably restoring non-shivering thermogenesis towards obesity management.
Collapse
|
27
|
Li M, Chen H, Zhu M. Mesenchymal stem cells for regenerative medicine in central nervous system. Front Neurosci 2022; 16:1068114. [PMID: 36583105 PMCID: PMC9793714 DOI: 10.3389/fnins.2022.1068114] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 11/28/2022] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent stem cells, whose paracrine and immunomodulatory potential has made them a promising candidate for central nervous system (CNS) regeneration. Numerous studies have demonstrated that MSCs can promote immunomodulation, anti-apoptosis, and axon re-extension, which restore functional neural circuits. The therapeutic effects of MSCs have consequently been evaluated for application in various CNS diseases including spinal cord injury, cerebral ischemia, and neurodegenerative disease. In this review, we will focus on the research works published in the field of mechanisms and therapeutic effects of MSCs in CNS regeneration.
Collapse
Affiliation(s)
- Man Li
- Department of Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Chen
- Department of Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mingxin Zhu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,*Correspondence: Mingxin Zhu,
| |
Collapse
|
28
|
Hall A, Fortino T, Spruance V, Niceforo A, Harrop JS, Phelps PE, Priest CA, Zholudeva LV, Lane MA. Cell transplantation to repair the injured spinal cord. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2022; 166:79-158. [PMID: 36424097 PMCID: PMC10008620 DOI: 10.1016/bs.irn.2022.09.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Adam Hall
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States
| | - Tara Fortino
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States
| | - Victoria Spruance
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States; Division of Kidney, Urologic, & Hematologic Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Alessia Niceforo
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States
| | - James S Harrop
- Department of Neurological and Orthopedic Surgery, Thomas Jefferson University, Philadelphia, PA, United States
| | - Patricia E Phelps
- Department of Integrative Biology & Physiology, UCLA, Los Angeles, CA, United States
| | | | - Lyandysha V Zholudeva
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States; Gladstone Institutes, San Francisco, CA, United States
| | - Michael A Lane
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States.
| |
Collapse
|
29
|
Ezdakova MI, Matveeva DK, Andreeva ER. Short-Term Interaction with Endothelial Cells Enhances Angiogenic Activity of Growth-Arrested Mesenchymal Stromal Cells In Vitro and In Ovo. Bull Exp Biol Med 2022; 174:125-130. [PMID: 36437339 DOI: 10.1007/s10517-022-05660-7] [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: 04/05/2022] [Indexed: 11/29/2022]
Abstract
We compared angiogenic effects of conditioned medium from mesenchymal stromal cell (MSC) monoculture and co-culture of MSC with endothelial cells (EC). Conditioned medium from 24-h EC-MSC co-cultures significantly stimulated the proliferation and migration of EC in monoculture and growth of the vascular network of the chorioallantoic membrane of the quail embryo in ovo in comparison with the conditioned medium from MSC monoculture. Conditioned medium from the co-culture contained increased levels of angiogenic factors (FGF-2, MCP-1, PDGF-AB/BB, IL-6, IL-8, etc.), which could explain the revealed effects. We hypothesized that a similar mechanism of EC-mediated enhancement of functional activity of MSC could be involved in reparative angiogenesis in the target tissues in vivo.
Collapse
Affiliation(s)
- M I Ezdakova
- Institute of Biomedical Problems, State Research Center, Russian Academy of Sciences, Moscow, Russia.
| | - D K Matveeva
- Institute of Biomedical Problems, State Research Center, Russian Academy of Sciences, Moscow, Russia
| | - E R Andreeva
- Institute of Biomedical Problems, State Research Center, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
30
|
Hoang DM, Pham PT, Bach TQ, Ngo ATL, Nguyen QT, Phan TTK, Nguyen GH, Le PTT, Hoang VT, Forsyth NR, Heke M, Nguyen LT. Stem cell-based therapy for human diseases. Signal Transduct Target Ther 2022; 7:272. [PMID: 35933430 PMCID: PMC9357075 DOI: 10.1038/s41392-022-01134-4] [Citation(s) in RCA: 457] [Impact Index Per Article: 152.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 07/19/2022] [Accepted: 07/21/2022] [Indexed: 02/07/2023] Open
Abstract
Recent advancements in stem cell technology open a new door for patients suffering from diseases and disorders that have yet to be treated. Stem cell-based therapy, including human pluripotent stem cells (hPSCs) and multipotent mesenchymal stem cells (MSCs), has recently emerged as a key player in regenerative medicine. hPSCs are defined as self-renewable cell types conferring the ability to differentiate into various cellular phenotypes of the human body, including three germ layers. MSCs are multipotent progenitor cells possessing self-renewal ability (limited in vitro) and differentiation potential into mesenchymal lineages, according to the International Society for Cell and Gene Therapy (ISCT). This review provides an update on recent clinical applications using either hPSCs or MSCs derived from bone marrow (BM), adipose tissue (AT), or the umbilical cord (UC) for the treatment of human diseases, including neurological disorders, pulmonary dysfunctions, metabolic/endocrine-related diseases, reproductive disorders, skin burns, and cardiovascular conditions. Moreover, we discuss our own clinical trial experiences on targeted therapies using MSCs in a clinical setting, and we propose and discuss the MSC tissue origin concept and how MSC origin may contribute to the role of MSCs in downstream applications, with the ultimate objective of facilitating translational research in regenerative medicine into clinical applications. The mechanisms discussed here support the proposed hypothesis that BM-MSCs are potentially good candidates for brain and spinal cord injury treatment, AT-MSCs are potentially good candidates for reproductive disorder treatment and skin regeneration, and UC-MSCs are potentially good candidates for pulmonary disease and acute respiratory distress syndrome treatment.
Collapse
Affiliation(s)
- Duc M Hoang
- Department of Research and Development, Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Healthcare System, Hanoi, Vietnam.
| | - Phuong T Pham
- Department of Cellular Therapy, Vinmec High-Tech Center, Vinmec Healthcare System, Hanoi, Vietnam
| | - Trung Q Bach
- Department of Research and Development, Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Healthcare System, Hanoi, Vietnam
| | - Anh T L Ngo
- Department of Cellular Therapy, Vinmec High-Tech Center, Vinmec Healthcare System, Hanoi, Vietnam
| | - Quyen T Nguyen
- Department of Research and Development, Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Healthcare System, Hanoi, Vietnam
| | - Trang T K Phan
- Department of Research and Development, Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Healthcare System, Hanoi, Vietnam
| | - Giang H Nguyen
- Department of Research and Development, Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Healthcare System, Hanoi, Vietnam
| | - Phuong T T Le
- Department of Research and Development, Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Healthcare System, Hanoi, Vietnam
| | - Van T Hoang
- Department of Research and Development, Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Healthcare System, Hanoi, Vietnam
| | - Nicholas R Forsyth
- Institute for Science & Technology in Medicine, Keele University, Keele, UK
| | - Michael Heke
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Liem Thanh Nguyen
- Department of Research and Development, Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Healthcare System, Hanoi, Vietnam
| |
Collapse
|
31
|
Interaction between Mesenchymal Stem Cells and the Immune System in Rheumatoid Arthritis. Pharmaceuticals (Basel) 2022; 15:ph15080941. [PMID: 36015088 PMCID: PMC9416102 DOI: 10.3390/ph15080941] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/22/2022] [Accepted: 07/26/2022] [Indexed: 02/04/2023] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease that causes damage to joints. This review focuses on the possibility of influencing the disease through immunomodulation by mesenchymal stem cells (MSCs). There is an occurrence of rheumatoid factor and RA-specific autoantibodies to citrullinated proteins in most patients. Citrulline proteins have been identified in the joints of RA patients, and are considered to be the most suitable candidates for the stimulation of anti-citrulline protein antibodies production. Fibroblast-like proliferating active synoviocytes actively promote inflammation and destruction in the RA joint, in association with pro-inflammatory cells. The inflammatory process may be suppressed by MSCs, which are a population of adherent cells with the following characteristic phenotype: CD105+, CD73+, CD90+, CD45−, CD34− and HLA DR−. Following the stimulation process, MSCs are capable of immunomodulatory action through the release of bioactive molecules, as well as direct contact with the cells of the immune system. Furthermore, MSCs show the ability to suppress natural killer cell activation and dendritic cells maturation, inhibit T cell proliferation and function, and induce T regulatory cell formation. MSCs produce factors that suppress inflammatory processes, such as PGE2, TGF-β, HLA-G5, IDO, and IL-10. These properties suggest that MSCs may affect and suppress the excessive inflammation that occurs in RA. The effect of MSCs on rheumatoid arthritis has been proven to be a suitable alternative treatment thanks to successful experiments and clinical studies.
Collapse
|
32
|
Wiese DM, Wood CA, Ford BN, Braid LR. Cytokine Activation Reveals Tissue-Imprinted Gene Profiles of Mesenchymal Stromal Cells. Front Immunol 2022; 13:917790. [PMID: 35924240 PMCID: PMC9341285 DOI: 10.3389/fimmu.2022.917790] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/10/2022] [Indexed: 11/30/2022] Open
Abstract
Development of standardized metrics to support manufacturing and regulatory approval of mesenchymal stromal cell (MSC) products is confounded by heterogeneity of MSC populations. Many reports describe fundamental differences between MSCs from various tissues and compare unstimulated and activated counterparts. However, molecular information comparing biological profiles of activated MSCs across different origins and donors is limited. To better understand common and source-specific mechanisms of action, we compared the responses of 3 donor populations each of human umbilical cord (UC) and bone marrow (BM) MSCs to TNF-α, IL-1β or IFN-γ. Transcriptome profiles were analysed by microarray and select secretome profiles were assessed by multiplex immunoassay. Unstimulated (resting) UC and BM-MSCs differentially expressed (DE) 174 genes. Signatures of TNF-α-stimulated BM and UC-MSCs included 45 and 14 new DE genes, respectively, while all but 7 of the initial 174 DE genes were expressed at comparable levels after licensing. After IL-1β activation, only 5 of the 174 DE genes remained significantly different, while 6 new DE genes were identified. IFN-γ elicited a robust transcriptome response from both cell types, yet nearly all differences (171/174) between resting populations were attenuated. Nine DE genes predominantly corresponding to immunogenic cell surface proteins emerged as a BM-MSC signature of IFN-γ activation. Changes in protein synthesis of select analytes correlated modestly with transcript levels. The dynamic responses of licensed MSCs documented herein, which attenuated heterogeneity between unstimulated populations, provide new insight into common and source-imprinted responses to cytokine activation and can inform strategic development of meaningful, standardized assays.
Collapse
Affiliation(s)
| | | | - Barry N. Ford
- Defence Research and Development Canada Suffield Research Centre, Casualty Management Section, Medicine Hat, AB, Canada
| | - Lorena R. Braid
- Aurora BioSolutions Inc., Medicine Hat, AB, Canada
- Simon Fraser University, Department of Molecular Biology and Biochemistry, Burnaby, BC, Canada
- *Correspondence: Lorena R. Braid, ;
| |
Collapse
|
33
|
Hernandez JC, Yeh DW, Marh J, Choi HY, Kim J, Chopra S, Ding L, Thornton M, Grubbs B, Makowka L, Sher L, Machida K. Activated and nonactivated MSCs increase survival in humanized mice after acute liver injury through alcohol binging. Hepatol Commun 2022; 6:1549-1560. [PMID: 35246968 PMCID: PMC9234635 DOI: 10.1002/hep4.1924] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 01/15/2022] [Accepted: 01/31/2022] [Indexed: 11/08/2022] Open
Abstract
The ability of the liver to regenerate after injury makes it an ideal organ to study for potential therapeutic interventions. Mesenchymal stem cells (MSCs) possess self-renewal and differentiation properties, as well as anti-inflammatory properties that make them an ideal candidate for therapy of acute liver injury. The primary aim of this study is to evaluate the potential for reversal of hepatic injury using human umbilical cord-derived MSCs. Secondary aims include comparison of various methods of administration as well as comparison of activated versus nonactivated human umbilical cord stem cells. To induce liver injury, humanized mice were fed high-cholesterol high-fat liquid diet with alcohol binge drinking. Mice were then treated with either umbilical cord MSCs, activated umbilical cord MSCs, or a placebo and followed for survival. Blood samples were obtained at the end of the binge drinking and at the time of death to measure alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels. Histology of all mouse livers was reported at time of death. Activated MSCs that were injected intravenously, intraperitoneally, or both routes had superior survival compared with nonactivated MSCs and with placebo-treated mice. AST and ALT levels were elevated in all mice before treatment and improved in the mice treated with stem cells. Conclusion: Activated stem cells resulted in marked improvement in survival and in recovery of hepatic chemistries. Activated umbilical cord MSCs should be considered an important area of investigation in acute liver injury.
Collapse
Affiliation(s)
- Juan Carlos Hernandez
- Departments of Molecular Microbiology and ImmunologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Da-Wei Yeh
- Departments of Molecular Microbiology and ImmunologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Joel Marh
- PrimeGenUS Inc.Santa AnaCaliforniaUSA
| | - Hye Yeon Choi
- Departments of Molecular Microbiology and ImmunologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Julia Kim
- PrimeGenUS Inc.Santa AnaCaliforniaUSA
| | - Shefali Chopra
- Department of PathologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Li Ding
- Department of Population and PublicHealth Sciences University of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Matthew Thornton
- Department of SurgeryUniversity of Southern CaliforniaLos AngelesCaliforniaUSA.,Childrens Hospital Los AngelesLos AngelesCaliforniaUSA
| | - Brendan Grubbs
- Department of SurgeryUniversity of Southern CaliforniaLos AngelesCaliforniaUSA.,Childrens Hospital Los AngelesLos AngelesCaliforniaUSA
| | | | - Linda Sher
- PrimeGenUS Inc.Santa AnaCaliforniaUSA.,Department of SurgeryUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Keigo Machida
- Departments of Molecular Microbiology and ImmunologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA.,Southern California Research Center for ALPD and CirrhosisLos AngelesCaliforniaUSA
| |
Collapse
|
34
|
Xu X, Liang Z, Lin Y, Rao J, Lin F, Yang Z, Wang R, Chen C. Comparing the Efficacy and Safety of Cell Transplantation for Spinal Cord Injury: A Systematic Review and Bayesian Network Meta-Analysis. Front Cell Neurosci 2022; 16:860131. [PMID: 35444516 PMCID: PMC9013778 DOI: 10.3389/fncel.2022.860131] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 03/11/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectiveTo compare the safety and effectiveness of transplanted cells from different sources for spinal cord injury (SCI).DesignA systematic review and Bayesian network meta-analysis.Data SourcesMedline, Embase, and the Cochrane Central Register of Controlled Trials.Study SelectionWe included randomized controlled trials, case–control studies, and case series related to cell transplantation for SCI patients, that included at least 1 of the following outcome measures: American Spinal Cord Injury Association (ASIA) Impairment Scale (AIS grade), ASIA motor score, ASIA sensory score, the Functional Independence Measure score (FIM), International Association of Neurorestoratology Spinal Cord Injury Functional Rating Scale (IANR-SCIFRS), or adverse events. Follow-up data were analyzed at 6 and 12 months.ResultsForty-four eligible trials, involving 1,266 patients, investigated 6 treatments: olfactory ensheathing cells (OECs), neural stem cells/ neural progenitor cells (NSCs), mesenchymal stem cells (MSCs), Schwann cells, macrophages, and combinations of cells (MSCs plus Schwann cells). Macrophages improved the AIS grade at 12 months (mean 0.42, 95% credible interval: 0–0.91, low certainty) and FIM score at 12 months (42.83, 36.33–49.18, very low certainty). MSCs improved the AIS grade at 6 months (0.42, 0.15–0.73, moderate certainty), the motor score at 6 months (4.43, 0.91–7.78, moderate certainty), light touch at 6 (10.01, 5.81–13.88, moderate certainty) and 12 months (11.48, 6.31–16.64, moderate certainty), pinprick score at 6 (14.54, 9.76–19.46, moderate certainty) and 12 months (12.48, 7.09–18.12, moderate certainty), and the IANR-SCIFRS at 6 (3.96, 0.62–6.97, moderate certainty) and 12 months (5.54, 2.45–8.42, moderate certainty). OECs improved the FIM score at 6 months (9.35, 1.71–17.00, moderate certainty). No intervention improved the motor score significantly at 12 months. The certainty of other interventions was low or very low. Overall, the number of adverse events associated with transplanted cells was low.ConclusionsPatients with SCI who receive transplantation of macrophages, MSCs, NSCs, or OECs may have improved disease prognosis. MSCs are the primary recommendations. Further exploration of the mechanism of cell transplantation in the treatment of SCI, transplantation time window, transplantation methods, and monitoring of the number of transplanted cells and cell survival is needed.Systematic Review Registrationhttps://www.crd.york.ac.uk/PROSPERO/#recordDetails, identifier: CRD 42021282043.
Collapse
|
35
|
Biomimetic approaches for targeting tumor inflammation. Semin Cancer Biol 2022; 86:555-567. [DOI: 10.1016/j.semcancer.2022.04.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 03/31/2022] [Accepted: 04/20/2022] [Indexed: 02/08/2023]
|
36
|
Jeon H, Kim HJ, Doo HM, Chang EH, Kwak G, Mo WM, Jang SY, Lee MW, Choi BO, Hong YB. Cytokines secreted by mesenchymal stem cells reduce demyelination in an animal model of Charcot-Marie-Tooth disease. Biochem Biophys Res Commun 2022; 597:1-7. [PMID: 35121177 DOI: 10.1016/j.bbrc.2022.01.098] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 01/25/2022] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Demyelinating Charcot-Marie-Tooth disease (CMT) is caused by mutations in the genes that encode myelinating proteins or their transcription factors. Our study thus sought to assess the therapeutic effects of cytokines secreted from mesenchymal stem cells (MSCs) on this disease. METHODS The therapeutic potential of Wharton's jelly MSCs (WJ-MSCs) and cytokines secreted by WJ-MSCs was evaluated on Schwann cells (SCs) exhibiting demyelination features, as well as a mouse model of demyelinating CMT. RESULTS Co-culture with WJ-MSC protected PMP22-overexpressing SCs from apoptotic cell death. Using a cytokine array, the secretion of growth differentiation factor-15 (GDF-15) and amphiregulin (AREG) was found to be elevated in WJ-MSCs when co-incubated with the PMP22-overexpressing SCs. Administration of both cytokines into trembler-J (Tr-J) mice, an animal model of CMT, significantly enhanced motor nerve conduction velocity compared to the control group. More importantly, this treatment alleviated the demyelinating phenotype of Tr-J mice, as demonstrated by an improvement in the mean diameter and g-ratio of the myelinated axons. CONCLUSIONS Our findings demonstrated that WJ-MSCs alleviate the demyelinating phenotype of CMT via the secretion of several cytokines. Further elucidation of the underlying mechanisms of GDF-15 and AREG in myelination might provide a robust basis for the development of effective therapies against demyelinating CMT.
Collapse
Affiliation(s)
- Hyeonjin Jeon
- Department of Translational Biomedical Sciences, Graduate School of Dong-A University, Busan, South Korea
| | - Hye Jin Kim
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, 06351, South Korea
| | - Hyun Myung Doo
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, 06351, South Korea
| | - Eun Hyuk Chang
- Samsung Biomedical Research Institute, Samsung Advanced Institute of Technology, Samsung Electronics Co, Ltd, Seoul, 06351, South Korea
| | - Geon Kwak
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, 06351, South Korea
| | - Won Min Mo
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, South Korea
| | - So Young Jang
- Department of Biochemistry, College of Medicine, Dong-A University, Busan, 49201, South Korea
| | | | - Byung-Ok Choi
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, 06351, South Korea; Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, South Korea.
| | - Young Bin Hong
- Department of Translational Biomedical Sciences, Graduate School of Dong-A University, Busan, South Korea; Department of Biochemistry, College of Medicine, Dong-A University, Busan, 49201, South Korea.
| |
Collapse
|
37
|
Yan S, Ye P, Aleem MT, Chen X, Xie N, Zhang Y. Mesenchymal Stem Cells Overexpressing ACE2 Favorably Ameliorate LPS-Induced Inflammatory Injury in Mammary Epithelial Cells. Front Immunol 2022; 12:796744. [PMID: 35095873 PMCID: PMC8795506 DOI: 10.3389/fimmu.2021.796744] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 12/22/2021] [Indexed: 01/15/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are capable of homing injury sites to exert anti-inflammatory as well as anti-damage effects and can be used as a vehicle for gene therapy. Angiotensin-converting enzyme 2 (ACE2) plays an important role in numerous inflammatory diseases, but fewer studies have been reported in animal mastitis. We hypothesized that MSCs overexpressing ACE2 is more effective in ameliorating lipopolysaccharide (LPS)-induced inflammatory injury in mammary epithelial cells compared to MSCs alone. The results showed that MSC-ACE2 inhibited the LPS induction by upregulation of TNF-α, IL-Iβ, IL-6, and iNOS mRNA expression levels in EpH4-Ev cells compared with MSCs. Furthermore, results showed that both MSC and MSC-ACE2 were significantly activated IL-10/STAT3/SOCS3 signaling pathway as well as inhibited TLR4/NF-κB and MAPK signaling pathways, but MSC-ACE2 had more significant effects. Meanwhile, MSC-ACE2 promoted the expression of proliferation-associated proteins and inhibited the expression of the apoptosis-associated proteins in EpH4-Ev cells. In addition, MSC and MSC-ACE2 reversed the LPS-induced downregulation expression levels of the tight junction proteins in mammary epithelial cells, indicating that both MSC as well as MSC-ACE2 could promote blood-milk barrier repair, and MSC-ACE2 was more effective. These results suggested that MSCs overexpressing ACE2 were more anti-inflammatory as well as anti-injurious action into LPS-induced inflammatory injury in the EpH4-Ev cells. Thus, MSCs overexpressing ACE2 is expected to serve as a potential strategy for mastitis treatment.
Collapse
Affiliation(s)
- Shuping Yan
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Pingsheng Ye
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Muhammad Tahir Aleem
- Ministry of Education (MOE) Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xi Chen
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Nana Xie
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yuanshu Zhang
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
38
|
Zhang L, Xi Y, Guo R, Miao Y, Chen H, Zhang M, Li B. Bone Marrow Mesenchymal Stem Cells Mediated Radiosensitive Promoter-Combined Sodium Iodide Symporter for the Treatment of Breast Cancer. Hum Gene Ther 2022; 33:638-648. [PMID: 35171716 DOI: 10.1089/hum.2021.144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES To develop a genetically engineered bone marrow mesenchymal stem cells (BMSCs) that carries a radiotherapy gene to target triple-negative breast cancer (TNBC) and to evaluate the efficacy of radiation damage within the tumor microenvironment (TME). METHODS The early growth response protein 1 (Egr1)-human sodium iodide symporter (hNIS) gene was transfected into BMSCs by lentiviral transfection and the expression levels were evaluated by RT-PCR. Transwell and adipogenesis and osteogenesis assays were performed to determine the targeting properties and adipogenic and osteogenic characteristics of the transgenic stem cells. The uptake of radioiodine and the efflux characteristics of the transgenic stem cells were determined by iodine uptake experiments. 131I-SPECT imaging was used to determine the characteristics of targeting to TNBC and to quantify the iodine uptake of transgenic stem cells in vivo. The effects of 131I treatment on BMSCs were characterized using tumor growth, immune cell infiltration and tumor invasion endpoints based on immunohistochemistry and flow cytometry analysis of tumor samples. RESULTS BMSCs-Egr1-hNIS cells abundantly express hNIS after radiation induction and are chemotactically attracted to TNBC tumors. Iodine uptake of BMSCs-Egr1-hNIS gradually increased with increasing induction concentrations and times. When the inductive concentration of 131I was > 100 μCi/mL and lasted for 36 h, the rate of iodine uptake in cells increased. In vitro, the radioiodine quickly flowed out from cells within 20 minutes but in vivo, the rate of radioiodine loss was significantly slower and occurred over 24 hours. After 131I therapy, tumor growth was inhibited, white blood cells infiltrated into tumor site and the levels of invasion-related cytokines significantly decreased. CONCLUSIONS BMSCs-Egr1-hNIS mediates 131I therapy can achieve precisely targeted radiotherapy to inhibit tumor growth, promote immune cells infiltration to the tumor sites and reduce the invasiveness and metastasis characteristics of tumor cells.
Collapse
Affiliation(s)
- Lu Zhang
- Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, 66281, No.197, Ruijin Er Road, Huangpu District, Shanghai, Shanghai, Shanghai, China, 021;
| | - Yue Xi
- Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, 66281, Shanghai, China;
| | - Rui Guo
- Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, 66281, Shanghai, China;
| | - Ying Miao
- Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, 66281, Shanghai, China;
| | - Hong Chen
- Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, 66281, Shanghai, China;
| | - Min Zhang
- Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, 66281, Shanghai, China;
| | - Biao Li
- Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, 66281, Department of Nuclear Medicine, Shanghai, China;
| |
Collapse
|
39
|
Jansen LE, Kim H, Hall CL, McCarthy TP, Lee MJ, Peyton SR. A poly(ethylene glycol) three-dimensional bone marrow hydrogel. Biomaterials 2022; 280:121270. [PMID: 34890973 PMCID: PMC8890749 DOI: 10.1016/j.biomaterials.2021.121270] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 11/19/2021] [Accepted: 11/22/2021] [Indexed: 01/03/2023]
Abstract
Three-dimensional (3D) hydrogels made from synthetic polymers have emerged as in vitro cell culture platforms capable of representing the extracellular geometry, modulus, and water content of tissues in a tunable fashion. Hydrogels made from these otherwise non-bioactive polymers can be decorated with short peptides derived from proteins naturally found in tissues to support cell viability and direct phenotype. We identified two key limitations that limit the ability of this class of materials to recapitulate real tissue. First, these environments typically display between 1 and 3 bioactive peptides, which vastly underrepresents the diversity of proteins found in the extracellular matrix (ECM) of real tissues. Second, peptides chosen are ubiquitous in ECM and not derived from proteins found in specific tissues, per se. To overcome this critical limitation in hydrogel design and functionality, we developed an approach to incorporate the complex and specific protein signature of bone marrow into a poly (ethylene glycol) (PEG) hydrogel. This bone marrow hydrogel mimics the elasticity of marrow and has 20 bone marrow-specific and cell-instructive peptides. We propose this tissue-centric approach as the next generation of 3D hydrogel design for applications in tissue engineering and beyond.
Collapse
Affiliation(s)
- Lauren E Jansen
- Department of Chemical Engineering, University of Massachusetts Amherst, USA
| | - Hyuna Kim
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, USA
| | - Christopher L Hall
- Department of Chemical Engineering, University of Massachusetts Amherst, USA
| | - Thomas P McCarthy
- Department of Chemical Engineering, University of Massachusetts Amherst, USA
| | - Michael J Lee
- Department of Systems Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Shelly R Peyton
- Department of Chemical Engineering, University of Massachusetts Amherst, USA; Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, USA; Institute for Applied Life Sciences, University of Massachusetts Amherst 240 Thatcher Way, Life Sciences Laboratory N531, Amherst, MA, 01003, USA.
| |
Collapse
|
40
|
Zhao Y, Sun Q, Huo B. Focal adhesion regulates osteogenic differentiation of mesenchymal stem cells and osteoblasts. BIOMATERIALS TRANSLATIONAL 2021; 2:312-322. [PMID: 35837413 PMCID: PMC9255797 DOI: 10.12336/biomatertransl.2021.04.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/29/2021] [Accepted: 12/10/2021] [Indexed: 11/18/2022]
Abstract
Focal adhesions are large macromolecular assemblies through which cells are connected with the extracellular matrix so that extracellular signals can be transmitted inside cells. Some studies have focused on the effect of cell shape on the differentiation of stem cells, but little attention has been paid to focal adhesion. In the present study, mesenchymal stem cells (MSCs) and osteoblast-like MC3T3-E1 cells were seeded onto micropatterned substrates on which circular adhesive islands with different spacing and area were created for focal adhesion. Results showed that the patterns of focal adhesion changed cell morphology but did not affect cell survival. For MSCs cultured for 3 days, patterns with small circles and large spacing promoted osteogenesis. For MSCs cultured for 7 days, patterns with large circles and spacing enhanced osteogenesis. For MC3T3-E1 cells, the patterns of focal adhesion had no effect on cell differentiation after 3 days of culture, but patterns with small circles and spacing improved osteogenic differentiation after 7 days. Moreover, the assembly of F-actin, phosphorylation of myosin, and nuclear translocation of yes-associated proteins (YAP) were consistent with the expression of differentiation markers, indicating that the pattern of focal adhesion may affect the osteogenesis of MSCs and osteoblasts through changes in cytoskeletal tension and nuclear localisation of YAP.
Collapse
Affiliation(s)
| | | | - Bo Huo
- Corresponding author: Bo Huo,
| |
Collapse
|
41
|
Huang M, Mehrabi Nasab E, Athari SS. Immunoregulatory effect of mesenchymal stem cell via mitochondria signaling pathways in allergic asthma. Saudi J Biol Sci 2021; 28:6957-6962. [PMID: 34866995 PMCID: PMC8626264 DOI: 10.1016/j.sjbs.2021.07.071] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 06/23/2021] [Accepted: 07/26/2021] [Indexed: 12/05/2022] Open
Abstract
Asthma is a complicated lung disease, which has increased morbidity and mortality rates in worldwide. There is an overlap between asthma pathophysiology and mitochondrial dysfunction and MSCs may have regulatory effect on mitochondrial dysfunction and treats asthma. Therefore, immune-modulatory effect of MSCs and mitochondrial signaling pathways in asthma was studied. After culturing of MSCs and producing asthma animal model, the mice were treated with MSCs via IV via IT. BALf's eosinophil Counting, The levels of IL-4, −5, −13, −25, –33, INF-γ, Cys-LT, LTB4, LTC4, mitochondria genes expression of COX-1, COX-2, ND1, Nrf2, Cytb were measured and lung histopathological study were done. BALf's eosinophils, the levels of IL-4, −5, −13, −25, –33, LTB4, LTC4, Cys-LT, the mitochondria genes expression (COX-1, COX-2, Cytb and ND-1), perivascular and peribronchial inflammation, mucus hyper-production and hyperplasia of the goblet cell in pathological study were significantly decreased in MSCs-treated asthma mice and reverse trend was found about Nrf-2 gene expression, IFN-γ level and ratio of the INF-γ/IL-4. MSC therapy can control inflammation, immune-inflammatory factors in asthma and mitochondrial related genes, and prevent asthma immune-pathology.
Collapse
Key Words
- AHR, Airway hyperresponsiveness
- ATP, Adenosine triphosphate
- Allergy
- BALF, Bronchoalveolar lavage fluid
- BM, Bone marrow
- CCL, Chemokine (C-C motif) ligand
- CD, Cluster of differentiation
- COX, Cyclooxygenase
- Cys-LT, Cysteinyl Leukotriene
- Cytb, Cytochrome b
- Drp1, Mitochondrial fission depends on the cytosolic GTPase dynamin-related protein 1
- ELISA, Enzyme-linked immunosorbent assay
- FIS1, Mitochondrial fission 1 protein
- H&E, Haemotoxylin and eosin
- HGF, Hepatocyte growth factor
- HLA, Human leukocyte antigen
- HO, Heme oxygenase
- IDO, Indoleamine 2,3-dioxygenase
- IFN, Interferon
- IL, Interleukin
- IP, Intraperitoneal injection
- IT, Intratrachea administration
- Ig, Immunoglobulin
- Immune system
- Inflammation
- LT, Leukotriene
- MFN, Mitofusin
- MIP, macrophage inflammatory protein
- MMP, Matrix metalloproteinase
- MSC
- MSC, mesenchymal stem cell
- MSC/BI, mesenchymal stem cell bronchial administration
- MSC/IV, mesenchymal stem cell intravenous injection
- ND1, NADH-ubiquinone oxidoreductase chain 1
- NO, Nitric oxide
- Nrf, Nuclear erythroid 2 p45-related factor
- OPA1, Mitochondrial dynamin like GTPase
- OVA, Ovalbumin
- PAS, Periodic-acid-Schiff
- PBS, Phosphate-buffered saline
- PGC1a, Peroxisome proliferator-activated receptor gamma coactivator 1-alpha
- PGE2, Prostaglandin E2
- ROS, Reactive oxygen species
- TFAM, Transcription factor A mitochondrial
- TGF, Transforming growth factor
- TNF, Tumor necrosis factor
- Th, T helper
- iPSC, induced pluripotent stem cells
Collapse
Affiliation(s)
- Minmin Huang
- Department of ICU, People's Hospital of Haimen District, Nantong City, Jiangsu Province, Haimen District, Nantong City, Jiangsu Province 226100, China
| | - Entezar Mehrabi Nasab
- Department of Cardiology, School of Medicine, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyyed Shamsadin Athari
- Department of Immunology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
42
|
Yang Y, Wang K, Pan Y, Rao L, Luo G. Engineered Cell Membrane-Derived Nanoparticles in Immune Modulation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2102330. [PMID: 34693653 PMCID: PMC8693058 DOI: 10.1002/advs.202102330] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 08/19/2021] [Indexed: 05/26/2023]
Abstract
Immune modulation is one of the most effective approaches in the therapy of complex diseases, including public health emergency. However, most immune therapeutics such as drugs, vaccines, and cellular therapy suffer from the limitations of poor efficacy and adverse side effects. Fortunately, cell membrane-derived nanoparticles (CMDNs) have superior compatibility with other therapeutics and offer new opportunities to push the limits of current treatments in immune modulation. As the interface between cells and outer surroundings, cell membrane contains components which instruct intercellular communication and the plasticity of cytomembrane has significantly potentiated CMDNs to leverage our immune system. Therefore, cell membranes employed in immunomodulatory CMDNs have gradually shifted from natural to engineered. In this review, unique properties of immunomodulatory CMDNs and engineering strategies of emerging CMDNs for immune modulation, with an emphasis on the design logic are summarized. Further, this review points out some pressing problems to be solved during clinical translation and put forward some suggestions on the prospect of immunoregulatory CMDNs. It is anticipated that this review can provide new insights on the design of immunoregulatory CMDNs and expand their potentiation in the precise control of the dysregulated immune system.
Collapse
Affiliation(s)
- Yixiao Yang
- Institute of Burn ResearchThe First Affiliated HospitalState Key Lab of TraumaBurn and Combined InjuryChongqing Key Laboratory for Disease ProteomicsThird Military Medical University (Army Medical University)Chongqing400038China
| | - Kai Wang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS)School of Basic Medical Sciences and Shanghai Public Health Clinical CenterShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Yuanwei Pan
- Institute of Biomedical Health Technology and EngineeringShenzhen Bay LaboratoryShenzhen518132China
| | - Lang Rao
- Institute of Biomedical Health Technology and EngineeringShenzhen Bay LaboratoryShenzhen518132China
| | - Gaoxing Luo
- Institute of Burn ResearchThe First Affiliated HospitalState Key Lab of TraumaBurn and Combined InjuryChongqing Key Laboratory for Disease ProteomicsThird Military Medical University (Army Medical University)Chongqing400038China
| |
Collapse
|
43
|
Paprocka M, Kraskiewicz H, Bielawska-Pohl A, Krawczenko A, Masłowski L, Czyżewska-Buczyńska A, Witkiewicz W, Dus D, Czarnecka A. From Primary MSC Culture of Adipose Tissue to Immortalized Cell Line Producing Cytokines for Potential Use in Regenerative Medicine Therapy or Immunotherapy. Int J Mol Sci 2021; 22:ijms222111439. [PMID: 34768869 PMCID: PMC8584013 DOI: 10.3390/ijms222111439] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/18/2021] [Accepted: 10/18/2021] [Indexed: 11/27/2022] Open
Abstract
For twenty-five years, attempts have been made to use MSCs in the treatment of various diseases due to their regenerative and immunomodulatory properties. However, the results are not satisfactory. Assuming that MSCs can be replaced in some therapies by the active factors they produce, the immortalized MSCs line was established from human adipose tissue (HATMSC1) to produce conditioned media and test its regenerative potential in vitro in terms of possible clinical application. The production of biologically active factors by primary MSCs was lower compared to the HATMSC1 cell line and several factors were produced only by the cell line. It has been shown that an HATMSC1-conditioned medium increases the proliferation of various cell types, augments the adhesion of cells and improves endothelial cell function. It was found that hypoxia during culture resulted in an augmentation in the pro-angiogenic factors production, such as VEGF, IL-8, Angiogenin and MCP-1. The immunomodulatory factors caused an increase in the production of GM-CSF, IL-5, IL-6, MCP-1, RANTES and IL-8. These data suggest that these factors, produced under different culture conditions, could be used for different medical conditions, such as in regenerative medicine, when an increased concentration of pro-angiogenic factors may be beneficial, or in inflammatory diseases with conditioned media with a high concentration of immunomodulatory factors.
Collapse
Affiliation(s)
- Maria Paprocka
- Laboratory of Biology of Stem and Neoplastic Cells, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (M.P.); (H.K.); (A.B.-P.); (A.K.); (D.D.)
| | - Honorata Kraskiewicz
- Laboratory of Biology of Stem and Neoplastic Cells, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (M.P.); (H.K.); (A.B.-P.); (A.K.); (D.D.)
| | - Aleksandra Bielawska-Pohl
- Laboratory of Biology of Stem and Neoplastic Cells, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (M.P.); (H.K.); (A.B.-P.); (A.K.); (D.D.)
| | - Agnieszka Krawczenko
- Laboratory of Biology of Stem and Neoplastic Cells, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (M.P.); (H.K.); (A.B.-P.); (A.K.); (D.D.)
| | - Leszek Masłowski
- Regional Specialist Hospital, Research and Development Centre, 51-154 Wroclaw, Poland; (L.M.); (A.C.-B.); (W.W.)
| | | | - Wojciech Witkiewicz
- Regional Specialist Hospital, Research and Development Centre, 51-154 Wroclaw, Poland; (L.M.); (A.C.-B.); (W.W.)
| | - Danuta Dus
- Laboratory of Biology of Stem and Neoplastic Cells, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (M.P.); (H.K.); (A.B.-P.); (A.K.); (D.D.)
| | - Anna Czarnecka
- Regional Specialist Hospital, Research and Development Centre, 51-154 Wroclaw, Poland; (L.M.); (A.C.-B.); (W.W.)
- Faculty of Physiotherapy, University School of Physical Education, 51-612 Wroclaw, Poland
- Correspondence:
| |
Collapse
|
44
|
Najar M, Merimi M, Faour WH, Lombard CA, Moussa Agha D, Ouhaddi Y, Sokal EM, Lagneaux L, Fahmi H. In Vitro Cellular and Molecular Interplay between Human Foreskin-Derived Mesenchymal Stromal/Stem Cells and the Th17 Cell Pathway. Pharmaceutics 2021; 13:1736. [PMID: 34684029 PMCID: PMC8537928 DOI: 10.3390/pharmaceutics13101736] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 10/06/2021] [Accepted: 10/13/2021] [Indexed: 12/13/2022] Open
Abstract
Foreskin, considered a biological waste material, has been shown to be a reservoir of therapeutic cells. The immunomodulatory properties of mesenchymal stromal/stem cells (MSCs) from the foreskin (FSK-MSCs) are being evaluated in cell-based therapy for degenerative, inflammatory and autoimmune disorders. Within the injured/inflamed tissue, proinflammatory lymphocytes such as IL-17-producing T helper cells (Th17) may interact with the stromal microenvironment, including MSCs. In this context, MSCs may encounter different levels of T cells as well as specific inflammatory signals. Uncovering the cellular and molecular changes during this interplay is central for developing an efficient and safe immunotherapeutic tool. To this end, an in vitro human model of cocultures of FSK-MSCs and T cells was established. These cocultures were performed at different cell ratios in the presence of an inflammatory setting. After confirming that FSK-MSCs respond to ISCT criteria by showing a typical phenotype and multilineage potential, we evaluated by flow cytometry the expression of Th17 cell markers IL-17A, IL23 receptor and RORγt within the lymphocyte population. We also measured 15 human Th17 pathway-related cytokines. Regardless of the T cell/MSC ratio, we observed a significant increase in IL-17A expression associated with an increase in IL-23 receptor expression. Furthermore, we observed substantial modulation of IL-1β, IL-4, IL-6, IL-10, IL-17A, IL-17F, IL-21, IL-22, IL-23, IL-25, IL-31, IL-33, INF-γ, sCD40, and TNF-α secretion. These findings suggest that FSK-MSCs are receptive to their environment and modulate the T cell response accordingly. The changes within the secretome of the stromal and immune environment are likely relevant for the therapeutic effect of MSCs. FSK-MSCs represent a valuable cellular product for immunotherapeutic purposes that needs to be further clarified and developed.
Collapse
Affiliation(s)
- Mehdi Najar
- Laboratory of Clinical Cell Therapy, Jules Bordet Institute, Université Libre de Bruxelles, 1070 Brussels, Belgium; (M.N.); (L.L.)
- Osteoarthritis Research Unit, Department of Medicine, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC H2X 0A9, Canada
| | - Makram Merimi
- Laboratory of Experimental Hematology, Jules Bordet Institute, Université Libre de Bruxelles, 1000 Brussels, Belgium; (M.M.); (D.M.A.)
- LBBES Laboratory, Genetics and Immune Cell Therapy Unit, Faculty of Sciences, University Mohammed Premier, Oujda 60000, Morocco
| | - Wissam H. Faour
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, P.O. Box 36, Byblos 5053, Lebanon;
| | - Catherine A. Lombard
- Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, 1200 Brussels, Belgium; (C.A.L.); (E.M.S.)
| | - Douâa Moussa Agha
- Laboratory of Experimental Hematology, Jules Bordet Institute, Université Libre de Bruxelles, 1000 Brussels, Belgium; (M.M.); (D.M.A.)
| | - Yassine Ouhaddi
- Orthopaedics Division, Department of Surgery, Faculty of Medicine, McGill University, Montreal General Hospital (MGH), The Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, QC H3G 1A4, Canada;
| | - Etienne M. Sokal
- Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, 1200 Brussels, Belgium; (C.A.L.); (E.M.S.)
| | - Laurence Lagneaux
- Laboratory of Clinical Cell Therapy, Jules Bordet Institute, Université Libre de Bruxelles, 1070 Brussels, Belgium; (M.N.); (L.L.)
| | - Hassan Fahmi
- Osteoarthritis Research Unit, Department of Medicine, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC H2X 0A9, Canada
| |
Collapse
|
45
|
Zhao Y, Yang X, Li S, Zhang B, Li S, Wang X, Wang Y, Jia C, Chang Y, Wei W. sTNFRII-Fc modification protects human UC-MSCs against apoptosis/autophagy induced by TNF-α and enhances their efficacy in alleviating inflammatory arthritis. Stem Cell Res Ther 2021; 12:535. [PMID: 34627365 PMCID: PMC8502322 DOI: 10.1186/s13287-021-02602-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 09/07/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Tumor necrosis factor (TNF)-α inhibitors represented by Etanercept (a fusion protein containing soluble TNF receptor II (sTNFRII) and the Fc segment of human IgG1) play a pivotal role in Rheumatoid arthritis (RA) treatment. However, long-term use increases the risk of infection and tumors for their systemic inhibition of TNF-α, which disrupts the regular physiological function of this molecular. Mesenchymal stem cells (MSCs)-based delivery system provides new options for RA treatment with their "homing" and immune-regulation capacities, whereas inflammatory environment (especially TNF-α) is not conducive to MSCs' therapeutic effects by inducing apoptosis/autophagy. Here, we constructed a strain of sTNFRII-Fc-expressing MSCs (sTNFRII-MSC), aiming to offset the deficiency of those two interventions. METHODS Constructed sTNFRII-Fc lentiviral vector was used to infect human umbilical cord-derived MSCs, and sTNFRII-MSC stable cell line was generated by monoclonal cultivation. In vitro and vivo characteristics of sTNFRII-MSC were assessed by coculture assay and an acute inflammatory model in NOD/SCID mice. The sTNFRII-MSC were transplanted into CIA model, pathological and immunological indicators were detected to evaluate the therapeutic effects of sTNFRII-MSC. The distribution of sTNFRII-MSC was determined by immunofluorescence assay. Apoptosis and autophagy were analyzed by flow cytometry, western blot and immunofluorescence. RESULTS sTNFRII-Fc secreted by sTNFRII-MSC present biological activity both in vitro and vivo. sTNFRII-MSC transplantation effectively alleviates mice collagen-induced arthritis (CIA) via migrating to affected area, protecting articular cartilage destruction, modulating immune balance and sTNFRII-MSC showed prolonged internal retention via resisting apoptosis/autophagy induced by TNF-α. CONCLUSION sTNFRII-Fc modification protects MSCs against apoptosis/autophagy induced by TNF-α, in addition to releasing sTNFRII-Fc neutralizing TNF-α to block relevant immune-inflammation cascade, and thus exert better therapeutic effects in alleviating inflammatory arthritis.
Collapse
Affiliation(s)
- Yingjie Zhao
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, 230032, China.,Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, 230601, China
| | - Xuezhi Yang
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, 230032, China
| | - Siyu Li
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, 230032, China
| | - Bingjie Zhang
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, 230032, China
| | - Susu Li
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, 230032, China
| | - Xinwei Wang
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, 230032, China
| | - Yueye Wang
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, 230032, China
| | - Chengyan Jia
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, 230032, China
| | - Yan Chang
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, 230032, China.
| | - Wei Wei
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, 230032, China.
| |
Collapse
|
46
|
A COVID-19 Overview and Potential Applications of Cell Therapy. Biologics 2021. [DOI: 10.3390/biologics1020011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The COVID-19 pandemic has already reaped thousands of lives, although many scientific studies already showed the possibility of this scenario. Currently, further attention is provided to patients depicting comorbidities such as respiratory or immunocompromised diseases, hypertension, and diabetes, as these individuals show a worse prognosis. Cell therapies using stem cells and/or defense cells, combined or not with traditional treatment, could be an outstanding strategy for COVID-19 management since these treatments can act by modulating the immune system, reducing proliferation, and favoring the complete elimination of the virus. In this review, we highlight the main molecular characteristics of this novel coronavirus, as well as the main pathognomonic signs of COVID-19. Furthermore, possible cell therapies are pointed out to show alternative treatments against COVID-19 and its sequels.
Collapse
|
47
|
Hartwig O, Shetab Boushehri MA, Shalaby KS, Loretz B, Lamprecht A, Lehr CM. Drug delivery to the inflamed intestinal mucosa - targeting technologies and human cell culture models for better therapies of IBD. Adv Drug Deliv Rev 2021; 175:113828. [PMID: 34157320 DOI: 10.1016/j.addr.2021.113828] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/09/2021] [Accepted: 06/10/2021] [Indexed: 12/19/2022]
Abstract
Current treatment strategies for inflammatory bowel disease (IBD) seek to alleviate the undesirable symptoms of the disorder. Despite the higher specificity of newer generation therapeutics, e.g. monoclonal antibodies, adverse effects still arise from their interference with non-specific systemic immune cascades. To circumvent such undesirable effects, both conventional and newer therapeutic options can benefit from various targeting strategies. Of course, both the development and the assessment of the efficiency of such targeted delivery systems necessitate the use of suitable in vivo and in vitro models representing relevant pathophysiological manifestations of the disorder. Accordingly, the current review seeks to provide a comprehensive discussion of the available preclinical models with emphasis on human in vitro models of IBD, along with their potentials and limitations. This is followed by an elaboration on the advancements in the field of biology- and nanotechnology-based targeted drug delivery systems and the potential rooms for improvement to facilitate their clinical translation.
Collapse
Affiliation(s)
- Olga Hartwig
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), D-66123 Saarbrücken, Germany; Department of Pharmacy, Saarland University, D-66123 Saarbrücken, Germany
| | | | - Karim S Shalaby
- Department of Pharmaceutics, University of Bonn, D-53121 Bonn, Germany; Department of Pharmaceutics and Industrial Pharmacy, Ain Shams University, Cairo, Egypt
| | - Brigitta Loretz
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), D-66123 Saarbrücken, Germany
| | - Alf Lamprecht
- Department of Pharmaceutics, University of Bonn, D-53121 Bonn, Germany.
| | - Claus-Michael Lehr
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), D-66123 Saarbrücken, Germany; Department of Pharmacy, Saarland University, D-66123 Saarbrücken, Germany.
| |
Collapse
|
48
|
Lauriano ER, Capillo G, Icardo JM, Fernandes JMO, Kiron V, Kuciel M, Zuwala K, Guerrera MC, Aragona M, Germana' A, Zaccone G. Neuroepithelial cells (NECs) and mucous cells express a variety of neurotransmitters and neurotransmitter receptors in the gill and respiratory air-sac of the catfish Heteropneustes fossilis (Siluriformes, Heteropneustidae): a possible role in local immune defence. ZOOLOGY 2021; 148:125958. [PMID: 34399394 DOI: 10.1016/j.zool.2021.125958] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 07/20/2021] [Accepted: 07/22/2021] [Indexed: 11/19/2022]
Abstract
Heteropneustes fossilis is an air-breathing teleost inhabiting environments with very poor O2 conditions, and so it has evolved to cope with hypoxia. In the gills and respiratory air-sac, the sites for O2 sensing and the response to hypoxia rely on the expression of acetylcholine (Ach) acting via its nicotinic receptor (nAChR). This study examined the expression patterns of neuronal markers and some compounds in the NECs of the gills and respiratory air sac having an immunomodulatory function in mammalian lungs. Mucous cells, epithelial cells and neuroepithelial cells (NECs) were immunopositive to a variety of both neuronal markers (VAChT, nAChR, GABA-B-R1 receptor, GAD679) and the antimicrobial peptide piscidin, an evolutionary conserved humoral component of the mucosal immune system in fish. We speculate that Ach release via nAChR from mucous cells may be modulated by GABA production in the NECs and it is required for the induction of mucus production in both normoxic and hypoxic conditions. The presence of piscidin in mucous cells may act in synergy with the autocrine/paracrine signals of Ach and GABA binding to GABA B R1B receptor that may play a local immunomodulatory function in the mucous epithelia of the gills and the respiratory air sac. The potential role of the NECs in the immunobiological behaviour of the gill/air-sac is at moment a matter of speculation. The extent to which the NECs as such may participate is elusive at this stage and waits investigation.
Collapse
Affiliation(s)
- Eugenia Rita Lauriano
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d'Alcontres 31, 98166, Messina, Italy
| | - Gioele Capillo
- Department of Veterinary Sciences, University of Messina, Polo Universitario dell'Annunziata, 98168, Messina, Italy; Institute of Marine Biological Resources and Biotechnology - National Research Council (IRBIM, CNR), Spianata S. Raineri, 86, 98122, Messina, Italy.
| | - Jose Manuel Icardo
- Department of Anatomy and Cell Biology, Poligono de Cazona, Faculty of Medicine, University of Cantabria, Santander, 39011, Spain
| | | | - Viswanath Kiron
- Faculty of Biosciences and Aquaculture, Nord University, 8049, Bodo, Norway
| | - Michal Kuciel
- Poison Information Centre, Department of Toxicology and Environmental Disease, Faculty of Medicine, Jagellonian University, Kopernika 15, 30501, Cracow, Poland
| | - Krystyna Zuwala
- Department of Comparative Anatomy, Faculty of Biology, Institute of Zoology and Biomedical Research, Jagiellonian University, Cracow, 30387, Poland
| | - Maria Cristina Guerrera
- Department of Veterinary Sciences, University of Messina, Polo Universitario dell'Annunziata, 98168, Messina, Italy
| | - Marialuisa Aragona
- Department of Veterinary Sciences, University of Messina, Polo Universitario dell'Annunziata, 98168, Messina, Italy
| | - Antonino Germana'
- Department of Veterinary Sciences, University of Messina, Polo Universitario dell'Annunziata, 98168, Messina, Italy
| | - Giacomo Zaccone
- Department of Veterinary Sciences, University of Messina, Polo Universitario dell'Annunziata, 98168, Messina, Italy
| |
Collapse
|
49
|
Guo S, Redenski I, Levenberg S. Spinal Cord Repair: From Cells and Tissue Engineering to Extracellular Vesicles. Cells 2021; 10:cells10081872. [PMID: 34440641 PMCID: PMC8394921 DOI: 10.3390/cells10081872] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 06/29/2021] [Accepted: 07/19/2021] [Indexed: 02/05/2023] Open
Abstract
Spinal cord injury (SCI) is a debilitating condition, often leading to severe motor, sensory, or autonomic nervous dysfunction. As the holy grail of regenerative medicine, promoting spinal cord tissue regeneration and functional recovery are the fundamental goals. Yet, effective regeneration of injured spinal cord tissues and promotion of functional recovery remain unmet clinical challenges, largely due to the complex pathophysiology of the condition. The transplantation of various cells, either alone or in combination with three-dimensional matrices, has been intensively investigated in preclinical SCI models and clinical trials, holding translational promise. More recently, a new paradigm shift has emerged from cell therapy towards extracellular vesicles as an exciting "cell-free" therapeutic modality. The current review recapitulates recent advances, challenges, and future perspectives of cell-based spinal cord tissue engineering and regeneration strategies.
Collapse
Affiliation(s)
- Shaowei Guo
- The First Affiliated Hospital, Shantou University Medical College, Shantou 515041, China
- Correspondence: (S.G.); (S.L.)
| | - Idan Redenski
- Department of Biomedical Engineering, Technion—Israel Institute of Technology, Haifa 32000, Israel;
| | - Shulamit Levenberg
- Department of Biomedical Engineering, Technion—Israel Institute of Technology, Haifa 32000, Israel;
- Correspondence: (S.G.); (S.L.)
| |
Collapse
|
50
|
Zupan J, Strazar K, Kocijan R, Nau T, Grillari J, Marolt Presen D. Age-related alterations and senescence of mesenchymal stromal cells: Implications for regenerative treatments of bones and joints. Mech Ageing Dev 2021; 198:111539. [PMID: 34242668 DOI: 10.1016/j.mad.2021.111539] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 06/28/2021] [Accepted: 07/01/2021] [Indexed: 12/18/2022]
Abstract
The most common clinical manifestations of age-related musculoskeletal degeneration are osteoarthritis and osteoporosis, and these represent an enormous burden on modern society. Mesenchymal stromal cells (MSCs) have pivotal roles in musculoskeletal tissue development. In adult organisms, MSCs retain their ability to regenerate tissues following bone fractures, articular cartilage injuries, and other traumatic injuries of connective tissue. However, their remarkable regenerative ability appears to be impaired through aging, and in particular in age-related diseases of bones and joints. Here, we review age-related alterations of MSCs in musculoskeletal tissues, and address the underlying mechanisms of aging and senescence of MSCs. Furthermore, we focus on the properties of MSCs in osteoarthritis and osteoporosis, and how their changes contribute to onset and progression of these disorders. Finally, we consider current treatments that exploit the enormous potential of MSCs for tissue regeneration, as well as for innovative cell-free extracellular-vesicle-based and anti-aging treatment approaches.
Collapse
Affiliation(s)
- Janja Zupan
- Department of Clinical Biochemistry, Faculty of Pharmacy, University of Ljubljana, 1000, Ljubljana, Slovenia
| | - Klemen Strazar
- Department of Orthopaedic Surgery, University Medical Centre Ljubljana, 1000, Ljubljana, Slovenia; Faculty of Medicine, University of Ljubljana, 1000, Ljubljana, Slovenia
| | - Roland Kocijan
- Ludwig Boltzmann Institute of Osteology at Hanusch Hospital of OEGK and AUVA Trauma Center Meidling, 1st Medical Department, Hanusch Hospital, Vienna, Austria; Medical Faculty of Bone Diseases, Sigmund Freud University Vienna, 1020, Vienna, Austria
| | - Thomas Nau
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in the AUVA Trauma Research Centre, 1200, Vienna, Austria; Austrian Cluster for Tissue Regeneration, 1200, Vienna, Austria; Building 14, Mohamed Bin Rashid University of Medicine and Health Sciences Dubai, Dubai Healthcare City, Dubai, United Arab Emirates
| | - Johannes Grillari
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in the AUVA Trauma Research Centre, 1200, Vienna, Austria; Austrian Cluster for Tissue Regeneration, 1200, Vienna, Austria; Department of Biotechnology, University of Natural Resources and Life Sciences Vienna, 1180, Vienna, Austria
| | - Darja Marolt Presen
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in the AUVA Trauma Research Centre, 1200, Vienna, Austria; Austrian Cluster for Tissue Regeneration, 1200, Vienna, Austria.
| |
Collapse
|