351
|
Qi F, Deng Z, Ma Y, Wang S, Liu C, Lyu F, Wang T, Zheng Q. From the perspective of embryonic tendon development: various cells applied to tendon tissue engineering. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:131. [PMID: 32175424 DOI: 10.21037/atm.2019.12.78] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
There is a high risk of injury from damage to the force-bearing tissue of the tendon. Due to its poor self-healing ability, clinical interventions for tendon injuries are limited and yield unsatisfying results. Tissue engineering might supply an alternative to this obstacle. As one of the key elements of tissue engineering, various cell sources have been used for tendon engineering, but there is no consensue concerning a single optimal source. In this review, we summarized the development of tendon tissue from the embryonic stage and categorized the used cell sources in tendon engineering. By comparing various cell sources as the candidates for tendon regeneration, each cell type was found to have its advantages and limitations; therefore, it is difficult to define the best cell source for tendon engineering. The microenvironment cells located is also crucial for cell growth and differentiation; so, the optimal cells are unlikely to be the same for each patient. In the future, the clinical application of tendon engineering might be more precise and customized in contrast to the current use of a standardized/generic one-size-fits-all procedure. The best cell source for tendon engineering will require a case-based assessment.
Collapse
Affiliation(s)
- Fangjie Qi
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou Higher Education Mega Centre, Guangzhou 510006, China
| | - Zhantao Deng
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou Higher Education Mega Centre, Guangzhou 510006, China
| | - Yuanchen Ma
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou Higher Education Mega Centre, Guangzhou 510006, China
| | - Shuai Wang
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou Higher Education Mega Centre, Guangzhou 510006, China
| | - Chang Liu
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou Higher Education Mega Centre, Guangzhou 510006, China
| | - Fengjuan Lyu
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou Higher Education Mega Centre, Guangzhou 510006, China
| | - Tao Wang
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou Higher Education Mega Centre, Guangzhou 510006, China.,Centre for Orthopaedic Translational Research, School of Biomedical Sciences, University of Western Australia, Nedlands, Western Australia, Australia
| | - Qiujian Zheng
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou Higher Education Mega Centre, Guangzhou 510006, China.,Centre for Orthopaedic Translational Research, School of Biomedical Sciences, University of Western Australia, Nedlands, Western Australia, Australia
| |
Collapse
|
352
|
A Randomized Controlled Trial of the Treatment of Rotator Cuff Tears with Bone Marrow Concentrate and Platelet Products Compared to Exercise Therapy: A Midterm Analysis. Stem Cells Int 2020; 2020:5962354. [PMID: 32399045 PMCID: PMC7204132 DOI: 10.1155/2020/5962354] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 12/19/2019] [Accepted: 12/26/2019] [Indexed: 12/13/2022] Open
Abstract
Injectable regenerative therapies such as bone marrow concentrate (BMC) and platelet-rich plasma (PRP) may represent a safe alternative in the treatment of rotator cuff tears. This is a midterm review of a randomized, crossover trial comparing autologous BMC and platelet product injections versus exercise therapy in the treatment of partial and full-thickness supraspinatus tears. Patients enrolled into the study were between 18 and 65 years of age presenting to an outpatient orthopedic clinic with partial to full thickness, nonretracted supraspinatus tendon tears. Enrolled patients were randomized to either ultrasound-guided autologous BMC with PRP and platelet lysate (PL) percutaneous injection treatment or exercise therapy. Patients could cross over to BMC treatment after at least 3 months of exercise therapy. Patients completed the Disability of the Arm, Shoulder and Hand (DASH) scores as the primary outcome measure. Secondary outcomes included the numeric pain scale (NPS), a modified Single Assessment Numeric Evaluation (SANE), and a blinded MRI review. At this midterm review, results from 25 enrolled patients who have reached at least 12-month follow-up are presented. No serious adverse events were reported. Significant differences were seen in patient reported outcomes for the BMC treatment compared to exercise therapy at 3 and 6 months for pain, and for function and reported improvement (SANE) at 3 months (p < .05). Patients reported a mean 89% improvement at 24 months, with sustained functional gains and pain reduction. MRI review showed a size decrease of most tears post-BMC treatment. These findings suggest that ultrasound-guided BMC and platelet product injections are a safe and useful alternative to conservative exercise therapy of torn, nonretracted supraspinatus tendons. This trial is registered with NCT01788683.
Collapse
|
353
|
Luo Q, Liu R, Wang L, Hou Y, Zhang H. The Effects of Inhibin B in the Chemotherapy Drug-Induced Premature Ovarian Insufficiency Mice and hPMSCs Treatment. Reprod Sci 2020; 27:1148-1155. [PMID: 31993999 DOI: 10.1007/s43032-019-00128-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 09/25/2019] [Indexed: 12/18/2022]
Abstract
Human placenta mesenchymal stem cells (hPMSCs), have been extensively investigated on the treatment of many diseases. This study was to explore the effects of hPMSCs treatment on the chemotherapy drug-induced premature ovarian insufficiency (POI) mice. Cyclophosphamide (120 mg/kg) and busulfan (30 mg/kg) or cyclophosphamide (70 mg/kg) induced POI mice were used and hPMSCs were injected through tail vein. The hormone levels of serum, morphological changes, the expression and quantitative analysis of inhibin B (INHBB) and FSHR protein, and apoptosis of granulosa cells in ovary were detected. The granulosa cells (GCs) were detected from ovaries of mice and the different concentration of cyclophosphamide on GCs were detected by MTT assay. The apoptosis of GCs was detected by FITC Annexin V Apoptosis Detection Kit. The significant increase in FSH and decrease in E2 and INHBB were observed. Expression of human nuclei was observed in the stroma of ovaries. INHBB and FSHR levels of ovaries were reduced in the POI mice. Following hPMSCs treatment, the amounts of INHBB and FSHR significantly increased close to normal levels. The granulosa cells apoptosis increased in the POI ovaries but decreased after hPMSCs treatment. Moreover, cyclophosphamide has no effect on the GCs and no statistic difference was measured in vitro. The effects of hPMSCs treatment reduce the apoptosis of granulosa cells and restore the ovarian reserve capacity in chemotherapeutic drug-induced POI mice. The data help to further explore new potential clinical therapeutic approach for POI patients.
Collapse
Affiliation(s)
- Qianqian Luo
- School of Basic Medical Sciences&Institute of Reproductive Diseases, Binzhou Medical University, Yantai, Shandong, China
| | - Ranran Liu
- Department of Reproductive Medicine, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| | - Li Wang
- Obstetrical [Maternity] Department, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| | - Yun Hou
- School of Basic Medical Sciences, Binzhou Medical University, Yantai, Shandong, China.
| | - Hongqin Zhang
- School of Basic Medical Sciences&Institute of Reproductive Diseases, Binzhou Medical University, Yantai, Shandong, China.
| |
Collapse
|
354
|
Papa PDM, Guasti PN, De Vita B, Nakazato NG, Maia L, Freitas Dell'Aqua CDP, Scheeren VFDC, Segabinazzi LGTM, de Andrade Junior LRP, Silva LFMC, Araújo EAB, Oliveira SN, Papa FO, Landim-Alvarenga FDC, Alvarenga MA. Clinical safety of intratesticular transplantation of allogeneic bone marrow multipotent stromal cells in stallions. Reprod Domest Anim 2020; 55:429-437. [PMID: 31916315 DOI: 10.1111/rda.13624] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 01/04/2020] [Indexed: 12/27/2022]
Abstract
Although stem cell therapy is a promising alternative for treatment of degenerative diseases, there are just few reports on the use of stem cells therapy in horse's reproductive system. This study aims to evaluate the effect of intratesticular injection of bone marrow mesenchymal stromal/stem cells (MSCs) in healthy stallions, and its outcome on seminal parameters and fertility. In Experiment 1, 24 stallions were divided into treatment group (TG) and control group (CG). In the TG, an intratesticular application of MSC was performed, and in the CG, only PBS was used. Measurements of testicular volume, surface temperature and Doppler ultrasonography were performed 24 and 48 hr after treatments. Fifteen days after application, the testicles were removed and submitted to histological analysis. In Experiment 2, 3 fertile stallions received similarly treatment with MSCs. Physical examination and sperm analysis were performed weekly during 60 days after treatment, and at the end, semen from one of them was used for artificial inseminations of 6 healthy mares. In Experiment 1, clinical examinations showed no signals of acute inflammation on both groups according to the analysed variables (p > .05). Also, no signal of chronic inflammation was observed on histological evaluation. In Experiment 2, stallions presented no physical alterations or changes in sperm parameters, and a satisfactory fertility rate (83%; 5/6) was observed after AI. The results support the hypothesis that intratesticular application of bone marrow MSCs is a safe procedure, and this could be a promising alternative to treat testicular degenerative conditions.
Collapse
Affiliation(s)
- Patrícia de Mello Papa
- Department of Animal Reproduction and Veterinary Radiology, College of Veterinary Medicine and Animal Science, Sao Paulo State University, Botucatu, Brazil
| | - Priscilla Nascimento Guasti
- Department of Animal Reproduction and Veterinary Radiology, College of Veterinary Medicine and Animal Science, Sao Paulo State University, Botucatu, Brazil
| | - Bruna De Vita
- Department of Animal Reproduction and Veterinary Radiology, College of Veterinary Medicine and Animal Science, Sao Paulo State University, Botucatu, Brazil
| | - Nathalia Genú Nakazato
- Department of Animal Reproduction and Veterinary Radiology, College of Veterinary Medicine and Animal Science, Sao Paulo State University, Botucatu, Brazil
| | - Leandro Maia
- Department of Animal Reproduction and Veterinary Radiology, College of Veterinary Medicine and Animal Science, Sao Paulo State University, Botucatu, Brazil
| | - Camila de Paula Freitas Dell'Aqua
- Department of Animal Reproduction and Veterinary Radiology, College of Veterinary Medicine and Animal Science, Sao Paulo State University, Botucatu, Brazil
| | - Verônica Flores da Cunha Scheeren
- Department of Animal Reproduction and Veterinary Radiology, College of Veterinary Medicine and Animal Science, Sao Paulo State University, Botucatu, Brazil
| | | | - Luiz Roberto Pena de Andrade Junior
- Department of Animal Reproduction and Veterinary Radiology, College of Veterinary Medicine and Animal Science, Sao Paulo State University, Botucatu, Brazil
| | - Luis Fernando Mercês Chaves Silva
- Department of Animal Reproduction and Veterinary Radiology, College of Veterinary Medicine and Animal Science, Sao Paulo State University, Botucatu, Brazil
| | - Endrigo Adonis Braga Araújo
- Department of Animal Reproduction and Veterinary Radiology, College of Veterinary Medicine and Animal Science, Sao Paulo State University, Botucatu, Brazil
| | - Sidnei Nunes Oliveira
- Department of Animal Reproduction and Veterinary Radiology, College of Veterinary Medicine and Animal Science, Sao Paulo State University, Botucatu, Brazil
| | - Frederico Ozanam Papa
- Department of Animal Reproduction and Veterinary Radiology, College of Veterinary Medicine and Animal Science, Sao Paulo State University, Botucatu, Brazil
| | - Fernanda da Cruz Landim-Alvarenga
- Department of Animal Reproduction and Veterinary Radiology, College of Veterinary Medicine and Animal Science, Sao Paulo State University, Botucatu, Brazil
| | - Marco Antonio Alvarenga
- Department of Animal Reproduction and Veterinary Radiology, College of Veterinary Medicine and Animal Science, Sao Paulo State University, Botucatu, Brazil
| |
Collapse
|
355
|
Hu C, Wu Z, Li L. Mesenchymal stromal cells promote liver regeneration through regulation of immune cells. Int J Biol Sci 2020; 16:893-903. [PMID: 32071558 PMCID: PMC7019139 DOI: 10.7150/ijbs.39725] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 12/26/2019] [Indexed: 02/06/2023] Open
Abstract
The liver is sensitive to pathogen-induced acute or chronic liver injury, and liver transplantation (LT) is the only effective strategy for end-stage liver diseases. However, the clinical application is limited by a shortage of liver organs, immunological rejection and high cost. Mesenchymal stromal cell (MSC)-based therapy has gradually become a hot topic for promoting liver regeneration and repairing liver injury in various liver diseases, since MSCs are reported to migrate toward injured tissues, undergo hepatogenic differentiation, inhibit inflammatory factor release and enhance the proliferation of liver cells in vivo. MSCs exert immunoregulatory effects through cell-cell contact and the secretion of anti-inflammatory factors to inhibit liver inflammation and promote liver regeneration. In addition, MSCs are reported to effectively inhibit the activation of cells of the innate immune system, including macrophages, natural killer (NK) cells, dendritic cells (DCs), monocytes and other immune cells, and inhibit the activation of cells of the adaptive immune system, including T lymphocytes, B lymphocytes and subsets of T cells or B cells. In the current review, we mainly focus on the potential effects and mechanisms of MSCs in inhibiting the activation of immune cells to attenuate liver injury in models or patients with acute liver failure (ALF), nonalcoholic fatty liver disease (NAFLD), and liver fibrosis and in patients or models after LT. We highlight that MSC transplantation may replace general therapies for eliminating acute or chronic liver injury in the near future.
Collapse
Affiliation(s)
- Chenxia Hu
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Zhongwen Wu
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Lanjuan Li
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, PR China
| |
Collapse
|
356
|
Joseph A, Baiju I, Bhat IA, Pandey S, Bharti M, Verma M, Pratap Singh A, Ansari MM, Chandra V, Saikumar G, Amarpal, Taru Sharma G. Mesenchymal stem cell-conditioned media: A novel alternative of stem cell therapy for quality wound healing. J Cell Physiol 2020; 235:5555-5569. [PMID: 31960454 DOI: 10.1002/jcp.29486] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 01/03/2020] [Indexed: 12/16/2022]
Abstract
Mesenchymal stem cells-conditioned media (MSCs-CM) contains several growth factors and cytokines, thus may be used as a better alternative to stem cell therapy, which needs to be elucidated. The present study was conducted to evaluate the therapeutic potential of caprine, canine, and guinea pig bone marrow-derived MSCs-CM in excision wound healing in a guinea pig model. MSCs were obtained from bone marrow, expanded ex vivo and characterized as per ISCT criteria. CM was collected assayed by western blot to ascertain the presence of important secretory biomolecules. Quantitative estimation by enzyme-linked immunosorbent assay was done for a vascular epidermal growth factor (VEGF) and interleukin-6 (IL-6) in caprine MSCs-CM and optimum time for collection of CM was decided as 72 hr. CM from all the species was lyophilized by freeze-drying method. Full-thickness (2 × 2 cm2 ) excision skin wounds were created in guinea pigs (six animals in each group) and respective lyophilized CM mixed with laminin gel was applied topically at weekly interval. On Day 28, histopathological examinations of healed skin were done by hemotoxylin and eosin staining. MSCs were found to secrete important growth factors and cytokines (i.e., VEGF, transforming growth factor-β1, fibroblast growth factor-2, insulin-like growth factor-1, stem cell factor, and IL-6) as demonstrated by immunohistochemistry and western blot assay. It was found that allogenic and xenogenic application of CM significantly improved quality wound healing with minimal scar formation. Thus, MSCs-CM can be used allogenically as well as xenogenically for quality wound healing.
Collapse
Affiliation(s)
- Anand Joseph
- Division of Physiology and Climatology, ICAR-Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh, India
| | - Indu Baiju
- Division of Physiology and Climatology, ICAR-Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh, India
| | - Irfan A Bhat
- Division of Physiology and Climatology, ICAR-Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh, India
| | - Sriti Pandey
- Division of Physiology and Climatology, ICAR-Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh, India
| | - Mukesh Bharti
- Division of Physiology and Climatology, ICAR-Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh, India
| | - Megha Verma
- Division of Physiology and Climatology, ICAR-Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh, India
| | - Anuj Pratap Singh
- Division of Veterinary Surgery, ICAR-Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh, India
| | - Matin M Ansari
- ICAR-National Research Center on Camel, Jorebeer, Bikaner, Rajasthan, India
| | - Vikash Chandra
- Division of Physiology and Climatology, ICAR-Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh, India
| | - Gutulla Saikumar
- Division of Veterinary Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh, India
| | - Amarpal
- Division of Veterinary Surgery, ICAR-Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh, India
| | - Gutulla Taru Sharma
- Division of Physiology and Climatology, ICAR-Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh, India
| |
Collapse
|
357
|
MacDonald ES, Barrett JG. The Potential of Mesenchymal Stem Cells to Treat Systemic Inflammation in Horses. Front Vet Sci 2020; 6:507. [PMID: 32039250 PMCID: PMC6985200 DOI: 10.3389/fvets.2019.00507] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 12/20/2019] [Indexed: 12/13/2022] Open
Abstract
One hallmark of mesenchymal stem cells (MSCs) is the ability to differentiate into multiple tissue types which assists in tissue regeneration. Another hallmark of MSCs is their potent anti-inflammatory and immunomodulatory properties and the potential to treat inflammatory, immune-mediated, and ischemic conditions. In equine practice, MSCs have shown efficacy in the treatment of musculoskeletal disorders such as tendinopathy, meniscal tears and cartilage injury. However, there are many equine disease processes and conditions that may benefit from the immunomodulatory properties of MSCs. Examples include conditions associated with overwhelming acute inflammatory response such as systemic inflammatory response syndrome to chronic diseases characterized by a prolonged low level of inflammation such as equine asthma and recurrent uveitis. For the acute inflammatory response processes, there is often high morbidity and mortality with no effective immunomodulatory treatment to prevent the overwhelming synthesis of proinflammatory mediators. For chronic inflammatory disease processes, frequently long-term corticosteroid treatment is the therapeutic mainstay, with serious potential complications. Thus, there is an unmet need for alternative anti-inflammatory treatments for both acute and chronic illnesses in horses. While MSCs show promise for such conditions, much research is needed before a clinically safe and effective treatment will be available. Optimal MSC tissue source, patient vs. donor source (autologous vs. allogeneic) and cell growth conditions need to be determined for each problem. For immediate use, allogeneic MSC treatments is preferable, but immune tolerance and adequate safety require further study. MSC collection and cryopreservation from horses before they are injured or ill, whether from umbilical cord tissue, bone marrow or adipose might become more widespread. Once these fundamental approaches to treating specific diseases with MSCs are determined, the route of administration, dose and timing of administration also need to be studied. To provide a framework for development of MSC immunomodulatory treatments, this article reviews the current understanding of equine MSC anti-inflammatory and immunomodulatory properties and proposes how MSC therapy may be further developed to treat acute onset systemic inflammatory processes and chronic inflammatory diseases.
Collapse
Affiliation(s)
- Elizabeth S MacDonald
- Marion duPont Scott Equine Medical Center, Virginia Maryland College of Veterinary Medicine, Virginia Tech, Leesburg, VA, United States
| | - Jennifer G Barrett
- Marion duPont Scott Equine Medical Center, Virginia Maryland College of Veterinary Medicine, Virginia Tech, Leesburg, VA, United States
| |
Collapse
|
358
|
The impact of prenatal environment on postnatal life and performance: Future perspectives for prevention and treatment. Theriogenology 2020; 150:15-19. [PMID: 31983467 DOI: 10.1016/j.theriogenology.2020.01.029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 01/13/2020] [Indexed: 12/16/2022]
Abstract
The present review aims to offer a non-comprehensive outline of the current state-of-the-art and future perspectives on management and therapeutic tools for intrauterine growth restriction (IUGR) and associated prenatal programming in both human and animal species. Animals are used as models for the study of phenomena related to IUGR, but also for research on prenatal therapies with the main objective of designing and developing preventive and therapeutic strategies. The research is currently paying attention on maternal-focused pharmacological treatments and nutritional strategies but also on fetal-focused treatments. Fetal-focused treatments, administered either directly at the fetus or by using infusion of umbilical cord, amniotic sac or placenta, which avoids the administration of substances at high doses to the mother for allowing their availability at the fetoplacental level. The results obtained in this area of research using large animals (rabbits, pigs and ruminants) have a dual interest, for translational biomedicine and for veterinary medicine and animal production.
Collapse
|
359
|
da Silva Meirelles L, Marson RF, Solari MIG, Nardi NB. Are Liver Pericytes Just Precursors of Myofibroblasts in Hepatic Diseases? Insights from the Crosstalk between Perivascular and Inflammatory Cells in Liver Injury and Repair. Cells 2020; 9:cells9010188. [PMID: 31940814 PMCID: PMC7017158 DOI: 10.3390/cells9010188] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 12/31/2019] [Accepted: 01/08/2020] [Indexed: 12/13/2022] Open
Abstract
Cirrhosis, a late form of liver disease, is characterized by extensive scarring due to exacerbated secretion of extracellular matrix proteins by myofibroblasts that develop during this process. These myofibroblasts arise mainly from hepatic stellate cells (HSCs), liver-specific pericytes that become activated at the onset of liver injury. Consequently, HSCs tend to be viewed mainly as myofibroblast precursors in a fibrotic process driven by inflammation. Here, the molecular interactions between liver pericytes and inflammatory cells such as macrophages and neutrophils at the first moments after injury and during the healing process are brought into focus. Data on HSCs and pericytes from other tissues indicate that these cells are able to sense pathogen- and damage-associated molecular patterns and have an important proinflammatory role in the initial stages of liver injury. On the other hand, further data suggest that as the healing process evolves, activated HSCs play a role in skewing the initial proinflammatory (M1) macrophage polarization by contributing to the emergence of alternatively activated, pro-regenerative (M2-like) macrophages. Finally, data suggesting that some HSCs activated during liver injury could behave as hepatic progenitor or stem cells will be discussed.
Collapse
Affiliation(s)
- Lindolfo da Silva Meirelles
- PPGBioSaúde and School of Medicine, Lutheran University of Brazil, Av. Farroupilha 8001, 92425-900 Canoas, RS, Brazil
| | - Renan Fava Marson
- PPGBioSaúde, Lutheran University of Brazil, Av. Farroupilha 8001, 92425-900 Canoas, RS, Brazil
| | - Maria Inês Gonzalez Solari
- Institute of Cardiology of Rio Grande do Sul, Av Princesa Isabel 370, 90620-001 Porto Alegre, RS, Brazil
| | - Nance Beyer Nardi
- Institute of Cardiology of Rio Grande do Sul, Av Princesa Isabel 370, 90620-001 Porto Alegre, RS, Brazil
- Correspondence: ; Tel.: +55-51-3230-3600
| |
Collapse
|
360
|
Clinical potential and current progress of mesenchymal stem cells for Parkinson's disease: a systematic review. Neurol Sci 2020; 41:1051-1061. [PMID: 31919699 DOI: 10.1007/s10072-020-04240-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Accepted: 01/04/2020] [Indexed: 12/11/2022]
Abstract
Parkinson's disease (PD) is the second most prevalent neurodegenerative disease characterized by severe dyskinesia due to a progressive loss of dopaminergic neurons along the nigro-striatal pathway. The current focus of treatment is to relieve symptoms through administration of levodopa, such as L-3,4-dihydroxy phenylalanine replacement therapy, dopaminergic agonist administration, functional neurosurgery, and gene therapy, rather than preventing dopaminergic neuronal damage. Hence, the application and development of neuroprotective/disease modification strategies is absolutely necessary. Currently, stem cell therapy has been considered for PD treatment. As for the stem cells, mesenchymal stem cells (MSCs) seem to be the most promising. In this review, we analyze the mechanisms of action of MSCs in Parkinson's disease, including growth factor secretion, exocytosis, and attenuation of neuroinflammation. To determine efficacy and protect patients from possible adverse effects, ongoing rigorous and controlled studies of MSC treatment will be critical.
Collapse
|
361
|
Kingery MT, Schoof L, Strauss EJ, Bosco JA, Halbrecht J. Online Direct-to-Consumer Advertising of Stem Cell Therapy for Musculoskeletal Injury and Disease: Misinformation and Violation of Ethical and Legal Advertising Parameters. J Bone Joint Surg Am 2020; 102:2-9. [PMID: 31770294 DOI: 10.2106/jbjs.19.00714] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND There has been a recent surge in health-care providers offering stem cell therapy (SCT) to patients with musculoskeletal disease. The purpose of this study was to identify and quantify the misinformation present in online direct-to-consumer (DTC) advertising of SCT targeting patients with musculoskeletal disease in the U.S. It was hypothesized that DTC advertising of SCT contains substantial misinformation. METHODS A list of keywords was used to identify web sites of practices advertising SCT directly to patients with musculoskeletal disease. Web sites were evaluated to determine the specialties of providers offering SCT, types of SCT being advertised, and misinformation presented. Categories of misinformation included false general claims, inaccurate statements regarding mechanism of action, unfounded results, and scare tactics. RESULTS Of the 896 practice web sites included in the analysis, 95.9% contained at least 1 statement of misinformation, with a mean of 4.65 ± 3.66 statements of misinformation among the sites. Practices associated with an orthopaedic surgeon provided 22% fewer statements of misinformation than practices without an orthopaedic surgeon when we controlled for the effects of other specialties. Practices associated with a podiatrist also provided 22% fewer statements of misinformation. CONCLUSIONS Nearly all practices failed to accurately represent the clinical efficacy of SCT in DTC advertising. While practices associated with an orthopaedic surgeon were less likely to provide misinformation, the majority of all web sites contained some type of misinformation, ranging from errors in the basic science of stem cells to outright false and misleading claims of their clinical effectiveness.
Collapse
Affiliation(s)
- Matthew T Kingery
- Department of Orthopedic Surgery, NYU Langone Orthopedic Hospital, NYU Langone Health, New York, NY
| | - Lauren Schoof
- Department of Orthopedic Surgery, NYU Langone Orthopedic Hospital, NYU Langone Health, New York, NY
| | - Eric J Strauss
- Department of Orthopedic Surgery, NYU Langone Orthopedic Hospital, NYU Langone Health, New York, NY
| | - Joseph A Bosco
- Department of Orthopedic Surgery, NYU Langone Orthopedic Hospital, NYU Langone Health, New York, NY
| | | |
Collapse
|
362
|
Dehghanian F, Soltani Z, Khaksari M. Can Mesenchymal Stem Cells Act Multipotential in Traumatic Brain Injury? J Mol Neurosci 2020; 70:677-688. [PMID: 31897971 DOI: 10.1007/s12031-019-01475-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 12/26/2019] [Indexed: 12/22/2022]
Abstract
Traumatic brain injury (TBI), a leading cause of morbidity and mortality throughout the world, will probably become the third cause of death in the world by the year 2020. Lack of effective treatments approved for TBI is a major health problem. TBI is a heterogeneous disease due to the different mechanisms of injury. Therefore, it requires combination therapies or multipotential therapy that can affect multiple targets. In recent years, mesenchymal stem cells (MSCs) transplantation has considered one of the most promising therapeutic strategies to repair of brain injuries including TBI. In these studies, it has been shown that MSCs can migrate to the site of injury and differentiate into the cells secreting growth factors and anti-inflammatory cytokines. The reduction in brain edema, neuroinflammation, microglia accumulation, apoptosis, ischemia, the improvement of motor and cognitive function, and the enhancement in neurogenesis, angiogenesis, and neural stem cells survival, proliferation, and differentiation have been indicated in these studies. However, translation of MSCs research in TBI into a clinical setting will require additional preclinical trials.
Collapse
Affiliation(s)
- Fatemeh Dehghanian
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
- Bam University of Medical Sciences, Bam, Iran
| | - Zahra Soltani
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| | - Mohammad Khaksari
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
363
|
Khan MR, Smith RK, David F, Lam R, Hughes G, De Godoy R, Carr AJ, Goodship AE, Dudhia J. Evaluation of the Effects of Synovial Multipotent Cells on Deep Digital Flexor Tendon Repair in a Large Animal Model of Intra-Synovial Tendinopathy. J Orthop Res 2020; 38:128-138. [PMID: 31329308 PMCID: PMC6973225 DOI: 10.1002/jor.24423] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 07/10/2019] [Indexed: 02/04/2023]
Abstract
Intra-synovial tendon injuries are a common orthopedic problem with limited treatment options. The synovium is a specialized connective tissue forming the inner encapsulating lining of diarthrodial joints and intra-synovial tendons. It contains multipotent mesenchymal stromal cells that render it a viable source of progenitors for tendon repair. This study evaluated the effects of autologous implantation of cells derived from normal synovium (synovial membrane cells [SMCs]) in augmenting repair in an ovine model of intra-synovial tendon injury. For this purpose, synovial biopsies were taken from the right digital flexor tendon sheath following creation of a defect to the lateral deep digital flexor tendon. Mononuclear cells were isolated by partial enzymatic digestion and assessed for MSC characteristics. Cell tracking and tendon repair were assessed by implanting 5 × 106 cells into the digital flexor tendon sheath under ultrasound guidance with the effects evaluated using magnetic resonance imaging and histopathology. Synovial biopsies yielded an average 4.0 × 105 ± 2.7 × 105 SMCs that exhibited a fibroblastic morphology, variable osteogenic, and adipogenic responses but were ubiquitously strongly chondrogenic. SMCs displayed high expression of CD29 with CD271NEGATIVE and MHC-IILOW cell-surface marker profiles, and variable expression of CD73, CD90, CD105, CD166, and MHC-I. Implanted SMCs demonstrated engraftment within the synovium, though a lack of repair of the tendon lesion over 24 weeks was observed. We conclude healthy synovium is a viable source of multipotent cells, but that the heterogeneity of synovium underlies the variability between different SMC populations, which while capable of engraftment and persistence within the synovium exhibit limited capacity of influencing tendon repair. © 2019 The Authors. Journal of Orthopaedic Research® published by Wiley Periodicals, Inc. on behalf of Orthopaedic Research Society J Orthop Res 38:128-138, 2020.
Collapse
Affiliation(s)
- Mohammad R. Khan
- Clinical Sciences and ServicesRoyal Veterinary CollegeHawkshead LaneHertfordshireAL9 7TAUnited Kingdom
| | - Roger K. Smith
- Clinical Sciences and ServicesRoyal Veterinary CollegeHawkshead LaneHertfordshireAL9 7TAUnited Kingdom
| | - Frederic David
- Clinical Sciences and ServicesRoyal Veterinary CollegeHawkshead LaneHertfordshireAL9 7TAUnited Kingdom
| | - Richard Lam
- Clinical Sciences and ServicesRoyal Veterinary CollegeHawkshead LaneHertfordshireAL9 7TAUnited Kingdom
| | - Gillian Hughes
- Clinical Sciences and ServicesRoyal Veterinary CollegeHawkshead LaneHertfordshireAL9 7TAUnited Kingdom
| | - Roberta De Godoy
- Writtle Agricultural CollegeLordship RoadChelmsfordEssexCM1 3RRUnited Kingdom
| | - Andrew J. Carr
- Botnar Research Centre, Institute of Musculoskeletal SciencesUniversity of OxfordOxfordOX3 7LDUnited Kingdom
| | - Allen E. Goodship
- UCL Institute of Orthopaedics and Musculoskeletal Science (IOMS)StanmoreHA7 4LPUnited Kingdom
| | - Jayesh Dudhia
- Clinical Sciences and ServicesRoyal Veterinary CollegeHawkshead LaneHertfordshireAL9 7TAUnited Kingdom
| |
Collapse
|
364
|
Fundueanu G, Constantin M, Bucatariu S, Nicolescu A, Ascenzi P, Moise LG, Tudor L, Trusca VG, Gafencu AV, Ficai D, Ficai A, Andronescu E. Simple and dual cross-linked chitosan millicapsules as a particulate support for cell culture. Int J Biol Macromol 2020; 143:200-212. [DOI: 10.1016/j.ijbiomac.2019.12.045] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/29/2019] [Accepted: 12/05/2019] [Indexed: 02/08/2023]
|
365
|
Shin K, Cha Y, Ban YH, Seo DW, Choi EK, Park D, Kang SK, Ra JC, Kim YB. Anti-osteoarthritis effect of a combination treatment with human adipose tissue-derived mesenchymal stem cells and thrombospondin 2 in rabbits. World J Stem Cells 2019; 11:1115-1129. [PMID: 31875872 PMCID: PMC6904861 DOI: 10.4252/wjsc.v11.i12.1115] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 10/14/2019] [Accepted: 11/04/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Osteoarthritis (OA), a chronic age-related disease characterized by the slowly progressive destruction of articular cartilage, is one of the leading causes of disability. As a new strategy for treatment of OA, mesenchymal stem cells (MSCs) have the potential for articular cartilage regeneration. Meanwhile, thrombospondin 2 (TSP2) promotes the chondrogenic differentiation of MSCs. AIM To investigate whether TSP2 induces chondrogenic differentiation of human adipose-derived MSCs (hADMSCs) and potentiates the therapeutic effects of hADMSCs in OA rabbits. METHODS We investigated the chondrogenic potential of TSP2 in hADMSCs by analyzing the expression of chondrogenic markers as well as NOTCH signaling genes in normal and TSP2 small interfering RNA (siRNA)-treated stem cells. Anterior cruciate ligament transection surgery was performed in male New Zealand white rabbits, and 8 wk later, hADMSCs (1.7 × 106 or 1.7 × 107 cells) were injected into the injured knees alone or in combination with intra-articular injection of TSP2 (100 ng/knee) at 2-d intervals. OA progression was monitored by gross, radiological, and histological examinations. RESULTS In hADMSC culture, treatment with TSP2 increased the expression of chondrogenic markers (SOX9 and collagen II) as well as NOTCH signaling genes (JAGGED1 and NOTCH3), which were inhibited by TSP2 siRNA treatment. In vivo, OA rabbits treated with hADMSCs or TSP2 alone exhibited lower degree of cartilage degeneration, osteophyte formation, and extracellular matrix loss 8 wk after cell transplantation. Notably, such cartilage damage was further alleviated by the combination of hADMSCs and TSP2. In addition, synovial inflammatory cytokines, especially tumor-necrosis factor-α, markedly decreased following the combination treatment. CONCLUSION The results indicate that TSP2 enhances chondrogenic differentiation of hADMSCs via JAGGED1/NOTCH3 signaling, and that combination therapy with hADMSCs and TSP2 exerts synergistic effects in the cartilage regeneration of OA joints.
Collapse
Affiliation(s)
- Kyungha Shin
- College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Chungbuk, South Korea
| | - Yeseul Cha
- College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Chungbuk, South Korea
| | - Young-Hwan Ban
- College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Chungbuk, South Korea
| | - Da Woom Seo
- College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Chungbuk, South Korea
| | - Ehn-Kyoung Choi
- College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Chungbuk, South Korea
| | - Dongsun Park
- Department of Biology Education, Korea National University of Education, Cheongju 28173, Chungbuk, South Korea
| | - Sung Keun Kang
- Biostar Stem Cell Research Institute, R-BIO Co., Ltd., Seoul 07238, South Korea
| | - Jeong Chan Ra
- Biostar Stem Cell Research Institute, R-BIO Co., Ltd., Seoul 07238, South Korea
| | - Yun-Bae Kim
- College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Chungbuk, South Korea.
| |
Collapse
|
366
|
Short and Long Term Clinical and Immunologic Follow up after Bone Marrow Mesenchymal Stromal Cell Therapy in Progressive Multiple Sclerosis-A Phase I Study. J Clin Med 2019; 8:jcm8122102. [PMID: 31810187 PMCID: PMC6947442 DOI: 10.3390/jcm8122102] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 11/20/2019] [Accepted: 11/21/2019] [Indexed: 12/20/2022] Open
Abstract
Bone marrow derived mesenchymal stromal cells (BM-MSCs) have emerged as a possible new therapy for Multiple Sclerosis (MS), however studies regarding efficacy and in vivo immune response have been limited and inconclusive. We conducted a phase I clinical study assessing safety and clinical and peripheral immune responses after MSC therapy in MS. Seven patients with progressive MS were intravenously infused with a single dose of autologous MSC (1–2 × 106 MSCs/kg body weight). The infusions were safe and well tolerated when given during clinical remission. Five out of seven patients completed the follow up of 48 weeks post-infusion. Brain magnetic resonance imaging (MRI) showed the absence of new T2 lesions at 12 weeks in 5/6 patients, while 3/5 had accumulated new T2 lesions at 48 weeks. Patient expanded disability status scales (EDSS) were stable in 6/6 at 12 weeks but declined in 3/5 patients at 48 weeks. Early changes of circulating microRNA levels (2 h) and increased proportion of FOXP3+ Tregs were detected at 7 days post-infusion compared to baseline levels. In conclusion, MSC therapy was safe and well tolerated and is associated with possible transient beneficial clinical and peripheral immunotolerogenic effects.
Collapse
|
367
|
Abstract
PURPOSE OF REVIEW The emergence of cell-based therapies has brought much excitement to the field of orthopedic sports medicine. However, the significant inconsistency of reporting has led to the poor understanding, misinformation, and false expectations for patients and clinicians alike. In this paper, we aim to clarify the available cell-therapy treatments and summarize some of the latest research. RECENT FINDINGS Although this technology is in early development, our understanding of cell biology has grown significantly over the last decade. Furthermore, it is becoming evident that tissue specificity may play a significant role in determining the effectiveness and overall clinical benefit attributed to cell therapy. Cell therapy is an emerging field with tremendous potential for clinically significant benefit. However, in its current state, clinical application of these treatments is limited by federal regulations, variability in formulation, and limited understanding of the biologic activity of various cell formulations.
Collapse
Affiliation(s)
- Bijan Dehghani
- Hospital for Special Surgery, 525 East 71 Street, 1st floor, New York, NY 10021 USA
| | - Scott Rodeo
- Hospital for Special Surgery, 525 East 71 Street, 1st floor, New York, NY 10021 USA
| |
Collapse
|
368
|
Allogeneic Versus Autologous Injectable Mesenchymal Stem Cells for Knee Osteoarthritis: Review and Current Status. Tech Orthop 2019. [DOI: 10.1097/bto.0000000000000357] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
369
|
Caseiro AR, Santos Pedrosa S, Ivanova G, Vieira Branquinho M, Almeida A, Faria F, Amorim I, Pereira T, Maurício AC. Mesenchymal Stem/ Stromal Cells metabolomic and bioactive factors profiles: A comparative analysis on the umbilical cord and dental pulp derived Stem/ Stromal Cells secretome. PLoS One 2019; 14:e0221378. [PMID: 31774816 PMCID: PMC6881058 DOI: 10.1371/journal.pone.0221378] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 10/21/2019] [Indexed: 12/21/2022] Open
Abstract
Mesenchymal Stem/ Stromal Cells assume a supporting role to the intrinsic mechanisms of tissue regeneration, a feature mostly assigned to the contents of their secretome. A comparative study on the metabolomic and bioactive molecules/factors content of the secretome of Mesenchymal Stem/ Stromal Cells derived from two expanding sources: the umbilical cord stroma and the dental pulp is presented and discussed. The metabolic profile (Nuclear Magnetic Resonance Spectroscopy) evidenced some differences in the metabolite dynamics through the conditioning period, particularly on the glucose metabolism. Despite, overall similar profiles are suggested. More prominent differences are highlighted for the bioactive factors (Multiplexing Laser Bear Analysis), in which Follistatin, Growth Regulates Protein, Hepatocyte Growth Factor, Interleukin-8 and Monocyte Chemotactic Protein-1 dominate in Umbilical Cord Mesenchymal Stem/ Stromal Cells secretion, while in Dental Pulp Stem/ Stromal Cells the Vascular Endothelial Growth Factor-A and Follistatin are more evident. The distinct secretory cocktail did not result in significantly different effects on endothelial cell populations dynamics including proliferation, migration, tube formation capacity and in vivo angiogenesis, or in chemotaxis for both Mesenchymal Stem/ Stromal Cells populations.
Collapse
Affiliation(s)
- Ana Rita Caseiro
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, Porto, Portugal
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado, Porto, Portugal
- Escola Universitária Vasco da Gama (EUVG), Lordemão, Coimbra, Portugal
| | - Sílvia Santos Pedrosa
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, Porto, Portugal
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado, Porto, Portugal
| | - Galya Ivanova
- REQUIMTE- LAQV, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre, Porto, Portugal
| | - Mariana Vieira Branquinho
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, Porto, Portugal
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado, Porto, Portugal
| | - André Almeida
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado, Porto, Portugal
- Indústria Transformadora de Subprodutos—I.T.S, SA, Grupo ETSA, Rua Padre Adriano, Olivais do Machio, Santo Antão do Tojal, Loures, Portugal
| | - Fátima Faria
- Departamento de Patologia e Imunologia Molecular, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, Porto, Portugal
| | - Irina Amorim
- Departamento de Patologia e Imunologia Molecular, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, Porto, Portugal
- i3S - Instituto de Investigação e Inovação da Universidade do Porto, Rua Alfredo Allen, Porto, Portugal
| | - Tiago Pereira
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, Porto, Portugal
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado, Porto, Portugal
| | - Ana Colette Maurício
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, Porto, Portugal
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado, Porto, Portugal
- * E-mail: ,
| |
Collapse
|
370
|
MSC-secreted TGF-β regulates lipopolysaccharide-stimulated macrophage M2-like polarization via the Akt/FoxO1 pathway. Stem Cell Res Ther 2019; 10:345. [PMID: 31771622 PMCID: PMC6878630 DOI: 10.1186/s13287-019-1447-y] [Citation(s) in RCA: 206] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 10/04/2019] [Accepted: 10/09/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND An uncontrolled inflammatory response is a critical pathophysiological feature of sepsis. Mesenchymal stem cells (MSCs) induce macrophage phenotype polarization and reduce inflammation in sepsis. MSC-secreted transforming growth factor beta (TGF-β) participated in the immune modulatory function of MSCs. However, the underlying mechanism of MSC-secreted TGF-β was not fully elucidated in regulation macrophage M2-like polarization. METHODS The paracrine effects of MSCs on macrophage polarization were studied using a co-culture protocol with LPS-stimulated RAW264.7 cells/mouse peritoneal macrophages and MSCs. The effect of TGF-β in the co-culture system was blocked by the TGF-β receptor inhibitor. To determine the role of MSC-secreted TGF-β, we used recombinant TGF-β to culture with LPS-stimulated RAW264.7 cells. In addition, we employed antibody microarray analysis to determine the mechanisms of MSC secreted TGF-β on LPS-stimulated RAW264.7 cell/mouse peritoneal macrophage M2-like polarization. Furthermore, we used an Akt inhibitor and a FoxO1 inhibitor to inhibit the Akt/FoxO1 pathway. The nuclear translocation of FoxO1 was detected by Western blot. RESULTS MSCs induced LPS-stimulated RAW264.7 cell/mouse peritoneal macrophage polarization towards the M2-like phenotype and significantly reduced pro-inflammatory cytokine levels via paracrine, which was inhibited by TGF-β receptor inhibitor. Furthermore, we found that MSC-secreted TGF-β enhanced the macrophage phagocytic ability. The antibody microarray analysis and Western blot verified that TGF-β treatment activated the Akt/FoxO1 pathway in LPS-stimulated macrophages, TGF-β-induced FoxO1 nuclear translocation and obviously expressed in the cytoplasm, the effects of TGF-β regulatory effects on LPS-stimulated macrophage were inhibited by pre-treatment with Akt inhibitor and FoxO1 inhibitor. CONCLUSIONS TGF-β secreted by MSCs could skew LPS-stimulated macrophage polarization towards the M2-like phenotype, reduce inflammatory reactions, and improve the phagocytic ability via the Akt/FoxO1 pathway, providing potential therapeutic strategies for sepsis.
Collapse
|
371
|
Saidova AA, Vorobjev IA. Lineage Commitment, Signaling Pathways, and the Cytoskeleton Systems in Mesenchymal Stem Cells. TISSUE ENGINEERING PART B-REVIEWS 2019; 26:13-25. [PMID: 31663422 DOI: 10.1089/ten.teb.2019.0250] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mesenchymal stem cells (MSCs) from adult tissues are promising candidates for personalized cell therapy and tissue engineering. Significant progress was achieved in our understanding of the regulation of MSCs proliferation and differentiation by different cues during the past years. Proliferation and differentiation of MSCs are sensitive to the extracellular matrix (ECM) properties, physical cues, and chemical signaling. Sheath stress, matrix stiffness, surface adhesiveness, and micro- and nanotopography define cell shape and dictate lineage commitment of MSCs even in the absence of specific chemical signals. We discuss mechanotransduction as the major route from ECM through the cytoskeleton toward signaling pathways and gene expression. All components of the cytoskeleton from primary cilium and focal adhesions (FAs) to actin, microtubules (MTs), and intermediate filaments (IFs) are involved in the mechanotransduction. Differentiation of MSCs is regulated via the complex network of interrelated signaling pathways, including RhoA/ROCK, Akt/Erk, and YAP/TAZ effectors of Hippo pathway. These pathways could be regulated both by chemical and mechanical stimuli. Attenuation of these pathways in MSCs results in specific changes in FAs and actin cytoskeleton. Besides, differentiation of MSCs affects MTs and IFs. Recent findings highlight the role of intranuclear actin in the regulation of transcription factors in response to mechanical environmental stimuli. Alterations of cytoskeletal components reflect the MSC senescence state and their migratory capacity. In this review, we discuss the relationships between the molecular interactions in signaling pathways and morphological response of cytoskeletal components and reveal the complex interrelations between cytoskeleton systems and signaling pathways during lineage commitment of MSCs. Impact Statement This review describes the complex network of relationships between mechanical and biochemical stimuli in mesenchymal stem cells (MSC) and their balance which defines the morphological changes of cell shape due to rearrangement of cytoskeletal systems during lineage commitment of MSCs.
Collapse
Affiliation(s)
- Aleena A Saidova
- Biological Faculty, M.V. Lomonosov Moscow State University, Moscow, Russia.,Center of Experimental Embryology and Reproductive Biotechnology, Moscow, Russia
| | - Ivan A Vorobjev
- Biological Faculty, M.V. Lomonosov Moscow State University, Moscow, Russia.,A.N. Belozersky Institute of Physico-Chemical Biology, M.V. Lomonosov Moscow State University, Moscow, Russia.,Department of Biology, School of Science and Humanities and National Laboratory Astana, Nazarbayev University, Nur-Sultan, Kazakhstan
| |
Collapse
|
372
|
Holan V, Hermankova B, Krulova M, Zajicova A. Cytokine interplay among the diseased retina, inflammatory cells and mesenchymal stem cells - a clue to stem cell-based therapy. World J Stem Cells 2019; 11:957-967. [PMID: 31768222 PMCID: PMC6851013 DOI: 10.4252/wjsc.v11.i11.957] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 09/02/2019] [Accepted: 09/14/2019] [Indexed: 02/06/2023] Open
Abstract
Retinal degenerative disorders, such as diabetic retinopathy, retinitis pigmentosa, age-related macular degeneration or glaucoma, represent the most common causes of loss of vision and blindness. In spite of intensive research, treatment options to prevent, stop or cure these diseases are limited. Newer therapeutic approaches are offered by stem cell-based therapy. To date, various types of stem cells have been evaluated in a range of models. Among them, mesenchymal stem/stromal cells (MSCs) derived from bone marrow or adipose tissue and used as autologous cells have been proposed to have the potential to attenuate the negative manifestations of retinal diseases. MSCs delivered to the vicinity of the diseased retina can exert local anti-inflammatory and repair-promoting/regenerative effects on retinal cells. However, MSCs also produce numerous factors that could have negative impacts on retinal regeneration. The secretory activity of MSCs is strongly influenced by the cytokine environment. Therefore, the interactions among the molecules produced by the diseased retina, cytokines secreted by inflammatory cells and factors produced by MSCs will decide the development and propagation of retinal diseases. Here we discuss the interactions among cytokines and other factors in the environment of the diseased retina treated by MSCs, and we present results supporting immunoregulatory and trophic roles of molecules secreted in the vicinity of the retina during MSC-based therapy.
Collapse
Affiliation(s)
- Vladimir Holan
- Department of Transplantation Immunology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague 14220, Czech Republic
- Department of Cell Biology, Faculty of Science, Charles University, Prague 12843, Czech Republic
| | - Barbora Hermankova
- Department of Transplantation Immunology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague 14220, Czech Republic
- Department of Cell Biology, Faculty of Science, Charles University, Prague 12843, Czech Republic
| | - Magdalena Krulova
- Department of Transplantation Immunology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague 14220, Czech Republic
- Department of Cell Biology, Faculty of Science, Charles University, Prague 12843, Czech Republic
| | - Alena Zajicova
- Department of Transplantation Immunology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague 14220, Czech Republic
| |
Collapse
|
373
|
Park JH, Kim OH, Kim KH, Hong HE, Seo H, Choi HJ, Ahn J, Lee TY, Kim SJ. Isolation of Secretome with Enhanced Antifibrotic Properties from miR-214-Transfected Adipose-Derived Stem Cells. J Korean Med Sci 2019; 34:e273. [PMID: 31760709 PMCID: PMC6875435 DOI: 10.3346/jkms.2019.34.e273] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 09/23/2019] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Secretome refers to the total set of molecules secreted or surface-shed by stem cells. The limitations of stem cell research have led numerous investigators to turn their attention to the use of secretome instead of stem cells. In this study, we intended to reinforce antifibrotic properties of the secretome released from adipose-derived stem cells (ASCs) transfected with miR-214. METHODS We generated miR-214-transfected ASCs, and extracted the secretome (miR214-secretome) from conditioned media of the transfected ASCs through a series of ultrafiltrations. Subsequently, we intravenously injected the miR-214-secretome into mice with liver fibrosis, and determined the effects of miR-214-secretome on liver fibrosis. RESULTS Compared with that by naïve secretome, liver fibrosis was ameliorated by intravenous infusion of miR-214-secretome into mice with liver fibrosis, which was demonstrated by significantly lower expression of fibrosis-related markers (alpha-smooth muscle actin, transforming growth factor-β, and metalloproteinases-2) in the livers as well as lower fibrotic scores in the special stained livers compared with naïve secretome. The infusion of miR-214-secretome also led to lesser local and systemic inflammation, higher expression of an antioxidant enzyme (superoxide dismutase), and higher liver proliferative and synthetic function. CONCLUSION MicroRNA-214 transfection stimulates ASCs to release the secretome with higher antifibrotic and anti-inflammatory properties. miR-214-secretome is thus expected to be one of the prominent ways of overcoming liver fibrosis, if further studies consistently validate its safety and efficiency.
Collapse
Affiliation(s)
- Jung Hyun Park
- Department of Surgery, Eunpyeong St. Mary's Hospital, College of Medicine, the Catholic University of Korea, Seoul, Korea
| | - Ok Hee Kim
- Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, the Catholic University of Korea, Seoul, Korea
- Catholic Central Laboratory of Surgery, Institute of Biomedical Industry, College of Medicine, the Catholic University of Korea, Seoul, Korea
| | - Kee Hwan Kim
- Department of Surgery, Uijeongbu St. Mary's Hospital, College of Medicine, the Catholic University of Korea, Seoul, Korea
| | - Ha Eun Hong
- Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, the Catholic University of Korea, Seoul, Korea
- Catholic Central Laboratory of Surgery, Institute of Biomedical Industry, College of Medicine, the Catholic University of Korea, Seoul, Korea
| | - Haeyeon Seo
- Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, the Catholic University of Korea, Seoul, Korea
- Catholic Central Laboratory of Surgery, Institute of Biomedical Industry, College of Medicine, the Catholic University of Korea, Seoul, Korea
| | - Ho Joong Choi
- Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, the Catholic University of Korea, Seoul, Korea
| | - Joseph Ahn
- Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, the Catholic University of Korea, Seoul, Korea
| | - Tae Yun Lee
- Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, the Catholic University of Korea, Seoul, Korea
| | - Say June Kim
- Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, the Catholic University of Korea, Seoul, Korea
- Catholic Central Laboratory of Surgery, Institute of Biomedical Industry, College of Medicine, the Catholic University of Korea, Seoul, Korea.
| |
Collapse
|
374
|
Kargozar S, Lotfibakhshaeish N, Ebrahimi-Barough S, Nazari B, Hill RG. Stimulation of Osteogenic Differentiation of Induced Pluripotent Stem Cells (iPSCs) Using Bioactive Glasses: An in vitro Study. Front Bioeng Biotechnol 2019; 7:355. [PMID: 31850324 PMCID: PMC6901961 DOI: 10.3389/fbioe.2019.00355] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 11/08/2019] [Indexed: 11/13/2022] Open
Abstract
Selection and use of an optimal cell source for bone tissue engineering (BTE) remain a challenging issue; the invention of induced pluripotent stem cells (iPSCs) have created new hopes on this regard. At the present study, we attempted to show the usability of iPSCs in combination with bioactive glasses (BGs) for bone regeneration applications. For this aim, iPSCs were cultured and incubated with the strontium and cobalt-containing BGs for different intervals (1, 5, and 7 days). The cell cytotoxicity and attachment were assessed using MTT assay and scanning electron microscopy (SEM), respectively. Moreover, the osteogenic differentiation of iPSCs seeded onto the glasses was evaluated using alkaline phosphatase (ALP) activity assay and real-time PCR. The obtained results clarified that although the cell viability is decreased during a 7 day period, the iPSCs could adhere and expand onto the BGs particles and over-express the osteogenic markers, including osteocalcin, osteonectin, and Runx2. Based on the data, we conclude that iPSCs in a combination of BGs can be considered as a potential candidate for BTE strategies.
Collapse
Affiliation(s)
- Saeid Kargozar
- Tissue Engineering Research Group, Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Nasrin Lotfibakhshaeish
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Somayeh Ebrahimi-Barough
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahareh Nazari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Robert G. Hill
- Unit of Dental Physical Sciences, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
375
|
Strategies to Enhance Mesenchymal Stem Cell-Based Therapies for Acute Respiratory Distress Syndrome. Stem Cells Int 2019; 2019:5432134. [PMID: 31885615 PMCID: PMC6893276 DOI: 10.1155/2019/5432134] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Accepted: 10/28/2019] [Indexed: 02/07/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a multifaced disease characterized by the acute onset of hypoxemia, worsened pulmonary compliance, and noncardiogenic pulmonary edema. Despite over five decades of research, specific treatments for established ARDS are still lacking. MSC-based therapies have the advantage of targeting nearly all pathophysiological components of ARDS by means of a variety of secreted trophic factors, exerting anti-inflammatory, antioxidative, immunomodulatory, antiapoptotic, and proangiogenic effects, resulting in significant structural and functional recovery following ARDS in various preclinical models. However, the therapeutic efficacy of transplanted MSCs is limited by their poor engraftment and low survival rate in the injured tissues, major barriers to clinical translation. Accordingly, several strategies have been explored to improve MSC retention in the lung and enhance the innate properties of MSCs in preclinical models of ARDS. To provide a comprehensive and updated view, we summarize a large body of experimental evidence for a variety of strategies directed towards strengthening the therapeutic potential of MSCs in ARDS.
Collapse
|
376
|
Mianehsaz E, Mirzaei HR, Mahjoubin-Tehran M, Rezaee A, Sahebnasagh R, Pourhanifeh MH, Mirzaei H, Hamblin MR. Mesenchymal stem cell-derived exosomes: a new therapeutic approach to osteoarthritis? Stem Cell Res Ther 2019; 10:340. [PMID: 31753036 PMCID: PMC6873475 DOI: 10.1186/s13287-019-1445-0] [Citation(s) in RCA: 194] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 10/05/2019] [Accepted: 10/09/2019] [Indexed: 12/16/2022] Open
Abstract
Degenerative disorders of joints, especially osteoarthritis (OA), result in persistent pain and disability and high costs to society. Nevertheless, the molecular mechanisms of OA have not yet been fully explained. OA is characterized by destruction of cartilage and loss of extracellular matrix (ECM). It is generally agreed that there is an association between pro-inflammatory cytokines and the development of OA. There is increased expression of matrix metalloproteinase (MMP) and “a disintegrin and metalloproteinase with thrombospondin motifs” (ADAMTS). Mesenchymal stem cells (MSCs) have been explored as a new treatment for OA during the last decade. It has been suggested that paracrine secretion of trophic factors, in which exosomes have a crucial role, contributes to the mechanism of MSC-based treatment of OA. The paracrine secretion of exosomes may play a role in the repair of joint tissue as well as MSC-based treatments for other disorders. Exosomes isolated from various stem cells may contribute to tissue regeneration in the heart, limbs, skin, and other tissues. Recent studies have indicated that exosomes (or similar particles) derived from MSCs may suppress OA development. Herein, for first time, we summarize the recent findings of studies on various exosomes derived from MSCs and their effectiveness in the treatment of OA. Moreover, we highlight the likely mechanisms of actions of exosomes in OA.
Collapse
Affiliation(s)
- Elaheh Mianehsaz
- Department of Physical Medicine and Rehabilitation, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran.,Trauma Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Hamid Reza Mirzaei
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Mahjoubin-Tehran
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Rezaee
- Department of Physical Medicine and Rehabilitation, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Roxana Sahebnasagh
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, I.R, Iran.
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, 40 Blossom Street, Boston, MA, 02114, USA.
| |
Collapse
|
377
|
Kingery MT, Manjunath AK, Anil U, Strauss EJ. Bone Marrow Mesenchymal Stem Cell Therapy and Related Bone Marrow-Derived Orthobiologic Therapeutics. Curr Rev Musculoskelet Med 2019; 12:451-459. [PMID: 31749105 DOI: 10.1007/s12178-019-09583-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW The purpose of the current article is to review the available literature related to bone marrow-derived mesenchymal stem cell therapy in the management of musculoskeletal pathologies and demonstrate the critical need for additional well-designed clinical studies. RECENT FINDINGS In recent years, there has been a rapid increase in interest regarding the use of bone marrow-derived mesenchymal stem cells in the treatment of musculoskeletal injury and disease. The clinical use of BM-MSCs and other forms of stem cell therapy has far outpaced the basic and translational science evidence required to elucidate the potential efficacy of this orthobiologic treatment approach. Early studies have demonstrated potential clinical benefit of utilizing bone marrow-derived mesenchymal stem cell therapy in the management of knee osteoarthritis, focal chondral lesions, shoulder pathology including rotator cuff tears and glenohumeral arthritis, and degenerative disk disease in the spine. To date, most published studies are small case series often lacking a control group or a standardized method of treatment. Bone marrow-derived mesenchymal stem cell therapy is becoming an increasingly common treatment for musculoskeletal injuries and disease. Although early clinical studies have shown promising outcomes, methodological flaws and lack of standardization among trials have limited the conclusions that can be drawn from the existing literature. A better understanding of the underlying mechanism of action and more carefully designed clinical trials will help reveal the efficacy and utility of BM-MSCs as a treatment modality for various orthopedic pathologies.
Collapse
Affiliation(s)
- Matthew T Kingery
- Division of Sports Medicine, Department of Orthopedic Surgery, NYU Langone Health, New York, NY, USA
| | - Amit K Manjunath
- Division of Sports Medicine, Department of Orthopedic Surgery, NYU Langone Health, New York, NY, USA
| | - Utkarsh Anil
- Division of Sports Medicine, Department of Orthopedic Surgery, NYU Langone Health, New York, NY, USA
| | - Eric J Strauss
- Division of Sports Medicine, Department of Orthopedic Surgery, NYU Langone Health, New York, NY, USA. .,NYU Langone Orthopedics, 333 East 38th Street, 4th Floor, New York, NY, 10016, USA.
| |
Collapse
|
378
|
Cheng RJ, Xiong AJ, Li YH, Pan SY, Zhang QP, Zhao Y, Liu Y, Marion TN. Mesenchymal Stem Cells: Allogeneic MSC May Be Immunosuppressive but Autologous MSC Are Dysfunctional in Lupus Patients. Front Cell Dev Biol 2019; 7:285. [PMID: 31799252 PMCID: PMC6874144 DOI: 10.3389/fcell.2019.00285] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 11/04/2019] [Indexed: 02/05/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have a potently immunosuppressive capacity in both innate and adaptive immune responses. Consequently, MSCs transplantation has emerged as a potential beneficial therapy for autoimmune diseases even though the mechanisms underlying the immunomodulatory activity of MSCs is incompletely understood. Transplanted MSCs from healthy individuals with no known history of autoimmune disease are immunosuppressive in systemic lupus erythematosus (SLE) patients and can ameliorate SLE disease symptoms in those same patients. In contrast, autologous MSCs from SLE patients are not immunosuppressive and do not ameliorate disease symptoms. Recent studies have shown that MSCs from SLE patients are dysfunctional in both proliferation and immunoregulation and phenotypically senescent. The senescent phenotype has been attributed to multiple genes and signaling pathways. In this review, we focus on the possible mechanisms for the defective phenotype and function of MSCs from SLE patients and summarize recent research on MSCs in autoimmune diseases.
Collapse
Affiliation(s)
- Rui-Juan Cheng
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - An-Ji Xiong
- Department of Rheumatology and Immunology, Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College, Nanchong, China
| | - Yan-Hong Li
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Shu-Yue Pan
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Qiu-Ping Zhang
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Zhao
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Tony N Marion
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China.,Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
379
|
Chen W, Cai J, Sun Y, Chen J, Chen S. [Research progress in treatment of knee osteoarthritis by paracrine effect of stem cells]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2019; 33:1446-1451. [PMID: 31650764 PMCID: PMC8337460 DOI: 10.7507/1002-1892.201903074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 07/25/2019] [Indexed: 02/05/2023]
Abstract
OBJECTIVE To review the advances in utilizing paracrine effect of stem cells in knee osteoarthritis (OA) treatment. METHODS The researches in applying stem cells derived conditioned medium, extracellular matrix, exosomes, and microvesicles in knee OA treatment and cartilage repair were reviewed and analyzed. RESULTS The satisfying outcomes of using different products of stem cells paracrine effect in knee OA condition as well as cartilage defect is revealed in studies in vitro and in vivo. The mechanism including suppressing the intraarticular inflammation, the apoptosis of chondrocytes, and the degradation of cartilage matrix, while enhancing the synthesis of cartilage matrix, the differentiation of in-situ stem cells into chondrocytes and the migration to the affected area. The effectiveness can be further improved supplemented with the tissue engineering methods or gene modification. CONCLUSION Compared with the traditional stem cell therapy, applying the products from paracrine effect of stem cells in knee OA treatment is more economical and safer, presenting great potential in clinical practice.
Collapse
Affiliation(s)
- Wenbo Chen
- Department of Sports Medicine and Arthroscopy, Huashan Hospital, Fudan University, Shanghai, 200040, P.R.China
| | - Jiangyu Cai
- Department of Sports Medicine and Arthroscopy, Huashan Hospital, Fudan University, Shanghai, 200040, P.R.China
| | - Yaying Sun
- Department of Sports Medicine and Arthroscopy, Huashan Hospital, Fudan University, Shanghai, 200040, P.R.China
| | - Jun Chen
- Department of Sports Medicine and Arthroscopy, Huashan Hospital, Fudan University, Shanghai, 200040, P.R.China
| | - Shiyi Chen
- Department of Sports Medicine and Arthroscopy, Huashan Hospital, Fudan University, Shanghai, 200040,
| |
Collapse
|
380
|
Kot M, Baj-Krzyworzeka M, Szatanek R, Musiał-Wysocka A, Suda-Szczurek M, Majka M. The Importance of HLA Assessment in "Off-the-Shelf" Allogeneic Mesenchymal Stem Cells Based-Therapies. Int J Mol Sci 2019; 20:E5680. [PMID: 31766164 PMCID: PMC6888380 DOI: 10.3390/ijms20225680] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 10/31/2019] [Accepted: 11/05/2019] [Indexed: 02/07/2023] Open
Abstract
The need for more effective therapies of chronic and acute diseases has led to the attempts of developing more adequate and less invasive treatment methods. Regenerative medicine relies mainly on the therapeutic potential of stem cells. Mesenchymal stem cells (MSCs), due to their immunosuppressive properties and tissue repair abilities, seem to be an ideal tool for cell-based therapies. Taking into account all available sources of MSCs, perinatal tissues become an attractive source of allogeneic MSCs. The allogeneic MSCs provide "off-the-shelf" cellular therapy, however, their allogenicity may be viewed as a limitation for their use. Moreover, some evidence suggests that MSCs are not as immune-privileged as it was previously reported. Therefore, understanding their interactions with the recipient's immune system is crucial for their successful clinical application. In this review, we discuss both autologous and allogeneic application of MSCs, focusing on current approaches to allogeneic MSCs therapies, with a particular interest in the role of human leukocyte antigens (HLA) and HLA-matching in allogeneic MSCs transplantation. Importantly, the evidence from the currently completed and ongoing clinical trials demonstrates that allogeneic MSCs transplantation is safe and seems to cause no major side-effects to the patient. These findings strongly support the case for MSCs efficacy in treatment of a variety of diseases and their use as an "off-the-shelf" medical product.
Collapse
Affiliation(s)
- Marta Kot
- Department of Transplantation, Faculty of Medicine, Medical College, Jagiellonian University, Wielicka 265, 30-663 Kraków, Poland; (M.K.); (A.M.-W.); (M.S.-S.)
| | - Monika Baj-Krzyworzeka
- Department of Clinical Immunology, Medical College, Jagiellonian University, Wielicka 265, 30-663 Kraków, Poland; (M.B.-K.); (R.S.)
| | - Rafał Szatanek
- Department of Clinical Immunology, Medical College, Jagiellonian University, Wielicka 265, 30-663 Kraków, Poland; (M.B.-K.); (R.S.)
| | - Aleksandra Musiał-Wysocka
- Department of Transplantation, Faculty of Medicine, Medical College, Jagiellonian University, Wielicka 265, 30-663 Kraków, Poland; (M.K.); (A.M.-W.); (M.S.-S.)
| | - Magdalena Suda-Szczurek
- Department of Transplantation, Faculty of Medicine, Medical College, Jagiellonian University, Wielicka 265, 30-663 Kraków, Poland; (M.K.); (A.M.-W.); (M.S.-S.)
| | - Marcin Majka
- Department of Transplantation, Faculty of Medicine, Medical College, Jagiellonian University, Wielicka 265, 30-663 Kraków, Poland; (M.K.); (A.M.-W.); (M.S.-S.)
| |
Collapse
|
381
|
Lee J, Kim OH, Lee SC, Kim KH, Shin JS, Hong HE, Choi HJ, Kim SJ. Enhanced Therapeutic Potential of the Secretome Released from Adipose-Derived Stem Cells by PGC-1α-Driven Upregulation of Mitochondrial Proliferation. Int J Mol Sci 2019; 20:5589. [PMID: 31717375 PMCID: PMC6888642 DOI: 10.3390/ijms20225589] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 11/05/2019] [Accepted: 11/07/2019] [Indexed: 12/12/2022] Open
Abstract
Peroxisome proliferator activated receptor λ coactivator 1α (PGC-1α) is a potent regulator of mitochondrial biogenesis and energy metabolism. In this study, we investigated the therapeutic potential of the secretome released from the adipose-derived stem cells (ASCs) transfected with PGC-1α (PGC-secretome). We first generated PGC-1α-overexpressing ASCs by transfecting ASCs with the plasmids harboring the gene encoding PGC-1α. Secretory materials released from PGC-1α-overexpressing ASCs were collected and their therapeutic potential was determined using in vitro (thioacetamide (TAA)-treated AML12 cells) and in vivo (70% partial hepatectomized mice) models of liver injury. In the TAA-treated AML12 cells, the PGC-secretome significantly increased cell viability, promoted expression of proliferation-related markers, such as PCNA and p-STAT, and significantly reduced the levels of reactive oxygen species (ROS). In the mice, PGC-secretome injections significantly increased liver tissue expression of proliferation-related markers more than normal secretome injections did (p < 0.05). We demonstrated that the PGC-secretome does not only have higher antioxidant and anti-inflammatory properties, but also has the potential of significantly enhancing liver regeneration in both in vivo and in vitro models of liver injury. Thus, reinforcing the mitochondrial antioxidant potential by transfecting ASCs with PGC-1α could be one of the effective strategies to enhance the therapeutic potential of ASCs.
Collapse
Affiliation(s)
- Jaeim Lee
- Department of Surgery, Uijeongbu St. Mary’s Hospital, College of Medicine, the Catholic University of Korea, Seoul 11765, Korea; (J.L.); (K.-H.K.)
| | - Ok-Hee Kim
- Department of Surgery, Seoul St. Mary’s Hospital, College of Medicine, the Catholic University of Korea, Seoul 06591, Korea; (O.-H.K.); (J.S.S.); (H.-E.H.); (H.J.C.)
- Catholic Central Laboratory of Surgery, College of Medicine, the Catholic University of Korea, Seoul 06591, Korea
| | - Sang Chul Lee
- Department of Surgery, Daejeon St. Mary’s Hospital, College of Medicine, the Catholic University of Korea, Seoul 34943, Korea;
| | - Kee-Hwan Kim
- Department of Surgery, Uijeongbu St. Mary’s Hospital, College of Medicine, the Catholic University of Korea, Seoul 11765, Korea; (J.L.); (K.-H.K.)
- Catholic Central Laboratory of Surgery, College of Medicine, the Catholic University of Korea, Seoul 06591, Korea
| | - Jin Sun Shin
- Department of Surgery, Seoul St. Mary’s Hospital, College of Medicine, the Catholic University of Korea, Seoul 06591, Korea; (O.-H.K.); (J.S.S.); (H.-E.H.); (H.J.C.)
- Catholic Central Laboratory of Surgery, College of Medicine, the Catholic University of Korea, Seoul 06591, Korea
| | - Ha-Eun Hong
- Department of Surgery, Seoul St. Mary’s Hospital, College of Medicine, the Catholic University of Korea, Seoul 06591, Korea; (O.-H.K.); (J.S.S.); (H.-E.H.); (H.J.C.)
- Catholic Central Laboratory of Surgery, College of Medicine, the Catholic University of Korea, Seoul 06591, Korea
| | - Ho Joong Choi
- Department of Surgery, Seoul St. Mary’s Hospital, College of Medicine, the Catholic University of Korea, Seoul 06591, Korea; (O.-H.K.); (J.S.S.); (H.-E.H.); (H.J.C.)
| | - Say-June Kim
- Department of Surgery, Seoul St. Mary’s Hospital, College of Medicine, the Catholic University of Korea, Seoul 06591, Korea; (O.-H.K.); (J.S.S.); (H.-E.H.); (H.J.C.)
- Catholic Central Laboratory of Surgery, College of Medicine, the Catholic University of Korea, Seoul 06591, Korea
| |
Collapse
|
382
|
Adipose-Derived Mesenchymal Stem Cells Promote M2 Macrophage Phenotype through Exosomes. Stem Cells Int 2019; 2019:7921760. [PMID: 31781246 PMCID: PMC6875419 DOI: 10.1155/2019/7921760] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/30/2019] [Accepted: 10/09/2019] [Indexed: 12/13/2022] Open
Abstract
Accumulating evidence has shown that the paracrine factors derived from mesenchymal stem cells (MSCs) are capable of regulating the immune system via interaction with various immune cells. In this study, adipose-derived MSCs (AdMSCs) and human peripheral blood monocytes (PBMCs) were isolated and cultured to examine the effects of MSC-induced macrophages (iMΦ) on inflammation and immune modulation. Indirect coculture with MSCs increased the expression of arginase-1 and mannose receptor (CD206), markers of activated M2 macrophages, in the PBMCs demonstrating that MSC-secreted factors promoted M2-MΦ polarization. Additionally, iMΦ exhibited a similar higher inhibitory effect on the growth of activated T cells compared to that in the other groups (AdMSCs only, AdMSCs plus iMΦ), implying that iMΦ can play a sufficient functional role. Interestingly, the population of FoxP3 Treg cells significantly increased when cocultured with iMΦ, suggesting that iMΦ have an immunomodulatory effect on the Treg cells through the modulation of the FoxP3 expression. Notably, iMΦ expressed high levels of immunosuppressive and anti-inflammatory cytokines, namely IL-10 and TSG-6. Furthermore, we confirmed that the AdMSC-derived exosomes modulated macrophage polarization by upregulating the expression of M2 macrophage markers. Conclusively, our results suggest that iMΦ play a significant role in regulating the immunomodulatory- and inflammatory-mediated responses. Thus, iMΦ may be used as a novel stem cell-based cell-free therapy for the treatment of immune-mediated inflammatory disorders.
Collapse
|
383
|
Gauthier-Fisher A, Kauffman A, Librach CL. Potential use of stem cells for fertility preservation. Andrology 2019; 8:862-878. [PMID: 31560823 DOI: 10.1111/andr.12713] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 09/19/2019] [Accepted: 09/23/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND Infertility and gonadal dysfunction can result from gonadotoxic therapies, environmental exposures, aging, or genetic conditions. In men, non-obstructive azoospermia (NOA) results from defects in the spermatogenic process that can be attributed to spermatogonial stem cells (SSC) or their niche, or both. While assisted reproductive technologies and sperm banking can enable fertility preservation (FP) in men of reproductive age who are at risk for infertility, FP for pre-pubertal patients remains experimental. Therapeutic options for NOA are limited. The rapid advance of stem cell research and of gene editing technologies could enable new FP options for these patients. Induced pluripotent stem cells (iPSC), SSC, and testicular niche cells, as well as mesenchymal stromal cells (aka medicinal signaling cells, MSCs), have been investigated for their potential use in male FP strategies. OBJECTIVE Here, we review the benefits and challenges for three types of stem cell-based approaches under investigation for male FP, focusing on the role that promising sources of MSC derived from human umbilical cord, specifically human umbilical cord perivascular cells (HUCPVC), could fulfill. These approaches are as follows: 1. isolation and ex vivo expansion of autologous SSC for in vivo transplantation or in vitro spermatogenesis; 2. in vitro differentiation toward germ cell and testicular somatic cell lineages using autologous SSC, or stem cells such iPSC or MSC; and 3. protection or regeneration of the spermatogenic niche after gonadotoxic insults in vivo. CONCLUSION Our studies suggest that HUCPVC are promising sources of cells that could be utilized in multiple aspects of male FP strategies.
Collapse
Affiliation(s)
| | - A Kauffman
- CReATe Fertility Centre, Toronto, ON, Canada
| | - C L Librach
- CReATe Fertility Centre, Toronto, ON, Canada.,Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada.,Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada.,Department of Gynecology, Women's College Hospital, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
384
|
Bolamperti S, Guidobono F, Rubinacci A, Villa I. The Role of Growth Hormone in Mesenchymal Stem Cell Commitment. Int J Mol Sci 2019; 20:ijms20215264. [PMID: 31652811 PMCID: PMC6862273 DOI: 10.3390/ijms20215264] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 10/18/2019] [Accepted: 10/21/2019] [Indexed: 12/16/2022] Open
Abstract
Growth hormone (GH) is best known for its prominent role in promoting prepubertal growth and in regulating body composition and metabolism during adulthood. In recent years, the possible role of GH in the modulation of mesenchymal stem cell (MSC) commitment has gained interest. MSCs, characterized by active self-renewal and differentiation potential, express GH receptors. In MSCs derived from different adult tissues, GH induces an inhibition of adipogenic differentiation and favors MSC differentiation towards osteogenesis. This activity of GH indicates that regulation of body composition by GH has already started in the tissue progenitor cells. These findings have fostered research on possible uses of MSCs treated with GH in those pathologies, where a lack of or delays in bone repair occur. After an overview of GH activities, this review will focus on the research that has characterized GH’s effects on MSCs and on preliminary studies on the possible application of GH in bone regenerative medicine.
Collapse
Affiliation(s)
- Simona Bolamperti
- Bone Metabolism Unit, Division of Genetics & Cell Biology, IRCCS Ospedale San Raffaele, 20132 Milan, Italy.
| | - Francesca Guidobono
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20129 Milan, Italy.
| | - Alessandro Rubinacci
- Bone Metabolism Unit, Division of Genetics & Cell Biology, IRCCS Ospedale San Raffaele, 20132 Milan, Italy.
| | - Isabella Villa
- Bone Metabolism Unit, Division of Genetics & Cell Biology, IRCCS Ospedale San Raffaele, 20132 Milan, Italy.
| |
Collapse
|
385
|
Pean CA, Kingery MT, Strauss E, Bosco JA, Halbrecht J. Direct-to-Consumer Advertising of Stem Cell Clinics: Ethical Considerations and Recommendations for the Health-Care Community. J Bone Joint Surg Am 2019; 101:e103. [PMID: 31577688 DOI: 10.2106/jbjs.19.00266] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
| | | | | | | | - Joanne Halbrecht
- NYU Langone Orthopedic Hospital, New York, NY.,Boulder Regenerative Medicine, Boulder, Colorado
| |
Collapse
|
386
|
Han YH, Kim KH, Abdi S, Kim TK. Stem cell therapy in pain medicine. Korean J Pain 2019; 32:245-255. [PMID: 31569916 PMCID: PMC6813895 DOI: 10.3344/kjp.2019.32.4.245] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 09/16/2019] [Accepted: 09/17/2019] [Indexed: 12/31/2022] Open
Abstract
Stem cells are attracting attention as a key element in future medicine, satisfying the desire to live a healthier life with the possibility that they can regenerate tissue damaged or degenerated by disease or aging. Stem cells are defined as undifferentiated cells that have the ability to replicate and differentiate themselves into various tissues cells. Stem cells, commonly encountered in clinical or preclinical stages, are largely classified into embryonic, adult, and induced pluripotent stem cells. Recently, stem cell transplantation has been frequently applied to the treatment of pain as an alternative or promising approach for the treatment of severe osteoarthritis, neuropathic pain, and intractable musculoskeletal pain which do not respond to conventional medicine. The main idea of applying stem cells to neuropathic pain is based on the ability of stem cells to release neurotrophic factors, along with providing a cellular source for replacing the injured neural cells, making them ideal candidates for modulating and possibly reversing intractable neuropathic pain. Even though various differentiation capacities of stem cells are reported, there is not enough knowledge and technique to control the differentiation into desired tissues in vivo. Even though the use of stem cells is still in the very early stages of clinical use and raises complicated ethical problems, the future of stem cells therapies is very bright with the help of accumulating evidence and technology.
Collapse
Affiliation(s)
- Yong Hee Han
- Department of Anesthesia and Pain Medicine, Pusan National University School of Medicine, Yangsan, Korea
| | - Kyung Hoon Kim
- Department of Anesthesia and Pain Medicine, Pusan National University School of Medicine, Yangsan, Korea
| | - Salahadin Abdi
- Division of Anesthesia and Critical Care, Department of Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tae Kyun Kim
- Department of Anesthesia and Pain Medicine, Pusan National University School of Medicine, Yangsan, Korea
| |
Collapse
|
387
|
Qazi TH, Tytgat L, Dubruel P, Duda GN, Van Vlierberghe S, Geissler S. Extrusion Printed Scaffolds with Varying Pore Size As Modulators of MSC Angiogenic Paracrine Effects. ACS Biomater Sci Eng 2019; 5:5348-5358. [PMID: 33464076 DOI: 10.1021/acsbiomaterials.9b00843] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cell encapsulation in confining 3D hydrogels typically prevents encapsulated cells from spreading and establishing cell-cell contacts. Interactions with neighboring cells or with the extracellular matrix (ECM) influence the paracrine activity of mesenchymal stromal cells (MSCs), but how these interactions are regulated by structural properties of biomaterial scaffolds remains insufficiently explored. Here, we describe the use of extrusion-based 3D printing to fabricate acellular, gelatin-based scaffolds with programmed strut spacings of 400 (small), 500 (medium), and 600 μm (large). These scaffolds showed similar effective Young's moduli in the range of 2-5 kPa, and varied based on average pore size which ranged from ∼200 μm (small pore: SP) through ∼302 μm (medium pore: MP) to ∼382 μm (large pore: LP). When seeded with MSCs, pore size guided cell distribution on the scaffolds, with smaller pores preventing cell infiltration, medium ones causing cells to aggregate in between struts, and large ones causing cells to flow through after attachment on the struts. These changes in cell distribution regulated cell-cell and cell-matrix interactions at the gene level, as assessed by pathway focused PCR arrays. Medium pore size scaffolds stimulated the highest paracrine secretion of a panel of angiogenic cytokines. This enhancement of paracrine activity substantially improved endothelial cell migration in a chemotaxis assay, increased single cell migration kinetics such as velocity, and stimulated the formation of robust tubular structures. Together, these findings not only provide new insights on cellular interactions in scaffold environments but also demonstrate how 3D biomaterial design can instruct and enhance the regenerative paracrine activities of MSCs.
Collapse
Affiliation(s)
- Taimoor H Qazi
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Liesbeth Tytgat
- Polymer Chemistry & Biomaterials Group - Centre of Macromolecular Chemistry (CMaC) - Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281, S4-Bis, 9000 Ghent, Belgium.,Brussels Photonics (B-PHOT) - Department of Applied Physics and Photonics, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium
| | - Peter Dubruel
- Polymer Chemistry & Biomaterials Group - Centre of Macromolecular Chemistry (CMaC) - Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281, S4-Bis, 9000 Ghent, Belgium.,Brussels Photonics (B-PHOT) - Department of Applied Physics and Photonics, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium
| | - Georg N Duda
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Sandra Van Vlierberghe
- Polymer Chemistry & Biomaterials Group - Centre of Macromolecular Chemistry (CMaC) - Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281, S4-Bis, 9000 Ghent, Belgium.,Brussels Photonics (B-PHOT) - Department of Applied Physics and Photonics, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium
| | - Sven Geissler
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies, Charité Universitätsmedizin Berlin, Louisa-Karsch-Str. 2, 10178 Berlin, Germany
| |
Collapse
|
388
|
Park KS, Bandeira E, Shelke GV, Lässer C, Lötvall J. Enhancement of therapeutic potential of mesenchymal stem cell-derived extracellular vesicles. Stem Cell Res Ther 2019; 10:288. [PMID: 31547882 PMCID: PMC6757418 DOI: 10.1186/s13287-019-1398-3] [Citation(s) in RCA: 177] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 08/19/2019] [Accepted: 08/26/2019] [Indexed: 02/06/2023] Open
Abstract
After the initial investigations into applications of mesenchymal stem cells (MSCs) for cell therapy, there was increased interest in their secreted soluble factors. Following studies of MSCs and their secreted factors, extracellular vesicles (EVs) released from MSCs have emerged as a new mode of intercellular crosstalk. MSC-derived EVs have been identified as essential signaling mediators under both physiological and pathological conditions, and they appear to be responsible for many of the therapeutic effects of MSCs. In several in vitro and in vivo models, EVs have been observed to have supportive functions in modulating the immune system, mainly mediated by EV-associated proteins and nucleic acids. Moreover, stimulation of MSCs with biophysical or biochemical cues, including EVs from other cells, has been shown to influence the contents and biological activities of subsequent MSC-derived EVs. This review provides on overview of the contents of MSC-derived EVs in terms of their supportive effects, and it provides different perspectives on the manipulation of MSCs to improve the secretion of EVs and subsequent EV-mediated activities. In this review, we discuss the possibilities for manipulating MSCs for EV-based cell therapy and for using EVs to affect the expression of elements of interest in MSCs. In this way, we provide a clear perspective on the state of the art of EVs in cell therapy focusing on MSCs, and we raise pertinent questions and suggestions for knowledge gaps to be filled.
Collapse
Affiliation(s)
- Kyong-Su Park
- Krefting Research Centre, Institute of Medicine at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Elga Bandeira
- Krefting Research Centre, Institute of Medicine at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ganesh V Shelke
- Krefting Research Centre, Institute of Medicine at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Cecilia Lässer
- Krefting Research Centre, Institute of Medicine at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jan Lötvall
- Krefting Research Centre, Institute of Medicine at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
389
|
Viswanathan S, Shi Y, Galipeau J, Krampera M, Leblanc K, Martin I, Nolta J, Phinney DG, Sensebe L. Mesenchymal stem versus stromal cells: International Society for Cell & Gene Therapy (ISCT®) Mesenchymal Stromal Cell committee position statement on nomenclature. Cytotherapy 2019; 21:1019-1024. [PMID: 31526643 DOI: 10.1016/j.jcyt.2019.08.002] [Citation(s) in RCA: 509] [Impact Index Per Article: 84.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 08/19/2019] [Indexed: 02/06/2023]
Abstract
The International Society for Cell & Gene Therapy (ISCT®) Mesenchymal Stromal Cell (ISCT MSC) committee offers a position statement to clarify the nomenclature of mesenchymal stromal cells (MSCs). The ISCT MSC committee continues to support the use of the acronym "MSCs" but recommends this be (i) supplemented by tissue-source origin of the cells, which would highlight tissue-specific properties; (ii) intended as MSCs unless rigorous evidence for stemness exists that can be supported by both in vitro and in vivo data; and (iii) associated with robust matrix of functional assays to demonstrate MSC properties, which are not generically defined but informed by the intended therapeutic mode of actions.
Collapse
Affiliation(s)
- S Viswanathan
- Arthritis Program, University Health Network, Krembil Research Institute, University Health Network, Cell Therapy Program, University Health Network, Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada.
| | - Y Shi
- The First Affiliated Hospital, Soochow University Institutes for Translational Medicine, Suzhou, China; Institute of Health Sciences, Chinese Academy of Sciences, Shanghai, China.
| | - J Galipeau
- Department of Medicine, University of Wisconsin Carbone Cancer Center, Madison, Wisconsin, USA
| | - M Krampera
- Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - K Leblanc
- Division of Clinical Immunology and Transfusion Medicine, Karolinska Institutet, Stockholm, Sweden
| | - I Martin
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - J Nolta
- Department of Internal Medicine, Stem Cell Program and Institute for Regenerative Cures, University of California Davis, Sacramento, California, USA
| | - D G Phinney
- Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, Florida, USA
| | - L Sensebe
- UMR5273 STROMALab CNRS/EFS/UPS-INSERM U1031, Toulouse, France
| |
Collapse
|
390
|
Szydlak R, Majka M, Lekka M, Kot M, Laidler P. AFM-based Analysis of Wharton's Jelly Mesenchymal Stem Cells. Int J Mol Sci 2019; 20:E4351. [PMID: 31491893 PMCID: PMC6769989 DOI: 10.3390/ijms20184351] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 09/01/2019] [Accepted: 09/02/2019] [Indexed: 12/16/2022] Open
Abstract
Wharton's jelly mesenchymal stem cells (WJ-MSCs) are multipotent stem cells that can be used in regenerative medicine. However, to reach the high therapeutic efficacy of WJ-MSCs, it is necessary to obtain a large amount of MSCs, which requires their extensive in vitro culturing. Numerous studies have shown that in vitro expansion of MSCs can lead to changes in cell behavior; cells lose their ability to proliferate, differentiate and migrate. One of the important measures of cells' migration potential is their elasticity, determined by atomic force microscopy (AFM) and quantified by Young's modulus. This work describes the elasticity of WJ-MSCs during in vitro cultivation. To identify the properties that enable transmigration, the deformability of WJ-MSCs that were able to migrate across the endothelial monolayer or Matrigel was analyzed by AFM. We showed that WJ-MSCs displayed differences in deformability during in vitro cultivation. This phenomenon seems to be strongly correlated with the organization of F-actin and reflects the changes characteristic for stem cell maturation. Furthermore, the results confirm the relationship between the deformability of WJ-MSCs and their migration potential and suggest the use of Young's modulus as one of the measures of competency of MSCs with respect to their possible use in therapy.
Collapse
Affiliation(s)
- Renata Szydlak
- Chair of Medical Biochemistry, Faculty of Medicine, Jagiellonian University Medical College, Kopernika 7, 31-034 Krakow, Poland.
| | - Marcin Majka
- Department of Transplantation, Institute of Pediatrics, Jagiellonian University Medical College, Wielicka 265, 30-663 Kraków, Poland.
| | - Małgorzata Lekka
- Institute of Nuclear Physics, Polish Academy of Sciences, Radzikowskiego 152, 31-342 Kraków, Poland.
| | - Marta Kot
- Department of Transplantation, Institute of Pediatrics, Jagiellonian University Medical College, Wielicka 265, 30-663 Kraków, Poland.
| | - Piotr Laidler
- Chair of Medical Biochemistry, Faculty of Medicine, Jagiellonian University Medical College, Kopernika 7, 31-034 Krakow, Poland.
| |
Collapse
|
391
|
Koh J, Griffin DR, Archang MM, Feng AC, Horn T, Margolis M, Zalazar D, Segura T, Scumpia PO, Di Carlo D. Enhanced In Vivo Delivery of Stem Cells using Microporous Annealed Particle Scaffolds. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1903147. [PMID: 31410986 PMCID: PMC6761037 DOI: 10.1002/smll.201903147] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 07/25/2019] [Indexed: 05/14/2023]
Abstract
Delivery to the proper tissue compartment is a major obstacle hampering the potential of cellular therapeutics for medical conditions. Delivery of cells within biomaterials may improve localization, but traditional and newer void-forming hydrogels must be made in advance with cells being added into the scaffold during the manufacturing process. Injectable, in situ cross-linking microporous scaffolds are recently developed that demonstrate a remarkable ability to provide a matrix for cellular proliferation and growth in vitro in three dimensions. The ability of these scaffolds to deliver cells in vivo is currently unknown. Herein, it is shown that mesenchymal stem cells (MSCs) can be co-injected locally with microparticle scaffolds assembled in situ immediately following injection. MSC delivery within a microporous scaffold enhances MSC retention subcutaneously when compared to cell delivery alone or delivery within traditional in situ cross-linked nanoporous hydrogels. After two weeks, endothelial cells forming blood vessels are recruited to the scaffold and cells retaining the MSC marker CD29 remain viable within the scaffold. These findings highlight the utility of this approach in achieving localized delivery of stem cells through an injectable porous matrix while limiting obstacles of introducing cells within the scaffold manufacturing process.
Collapse
Affiliation(s)
- Jaekyung Koh
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Donald R Griffin
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22904, USA
| | - Maani M Archang
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - An-Chieh Feng
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Thomas Horn
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Michael Margolis
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - David Zalazar
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Tatiana Segura
- Department of Biomedical Engineering, Neurology, Dermatology, Duke University, Durham, NC, 27708, USA
| | - Philip O Scumpia
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Dermatology, VA Greater Los Angeles Healthcare System, Los Angeles, CA, 90073, USA
| | - Dino Di Carlo
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- California NanoSystems Institute (CNSI), University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| |
Collapse
|
392
|
Chen W, Sun Y, Gu X, Hao Y, Liu X, Lin J, Chen J, Chen S. Conditioned medium of mesenchymal stem cells delays osteoarthritis progression in a rat model by protecting subchondral bone, maintaining matrix homeostasis, and enhancing autophagy. J Tissue Eng Regen Med 2019; 13:1618-1628. [PMID: 31210406 DOI: 10.1002/term.2916] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 06/07/2019] [Accepted: 06/07/2019] [Indexed: 02/05/2023]
Abstract
Evidence accumulated that mesenchymal stem cell (MSC) therapy ameliorated osteoarthritis (OA) via paracrine effect, whereas conditioned medium (CM) of MSCs contains all the secretomes. In vitro studies have proved its therapeutic effect in OA, but few in vivo evidences were unveiled. This study investigated the effect of MSCs-CM in an animal model of OA. OA was induced by anterior cruciate ligament transaction and destabilization of the medial meniscus in 12 rats bilaterally. The CM group (N = 6) was administered with intraarticular injection of MSCs-CM weekly, whereas the phosphate-buffered saline (PBS) group (N = 6) was injected with PBS. Six rats served as normal control and received sham operation with weekly PBS injection. Rats were sacrificed 8 weeks postoperatively. Gross and histological morphology were analysed. Microcomputed tomography was applied to assess the subchondral bone. Components of extracellular matrix (ECM) including type II collagen (Col II) and aggrecan, and ECM homeostasis-related enzymes (metalloproteinase-13 [MMP-13] and tissue inhibitor of metalloproteinase-1 [TIMP-1]), as well as autophagy markers (Beclin-1 and microtubule-associated protein light chain 3) were evaluated immunohistochemically. Chondrocyte apoptosis was measured by terminal deoxynucleotidyl transferase dUTP nick-end labelling staining. Gene expression of Col II, aggrecan, MMP-13, and TIMP-1 was confirmed by real-time polymerase chain reaction. Morphological outcomes demonstrated remarkable articular-protective effect of MSCs-CM. Well-maintained subchondral bone structure, significantly more abundant cartilage matrix, notably decreased ratio of MMP-13 to TIMP-1, and inhibited chondrocyte apoptosis with enhanced autophagy were observed in the CM group compared with the PBS group. In conclusion, MSCs-CM demonstrated satisfactory effect in alleviating OA in rats via protecting the microarchitecture of subchondral bone, balancing the ratio of MMP-13 to TIMP-1 in cartilage, and enhancing autophagy, which might provide a new remedy against OA.
Collapse
Affiliation(s)
- Wenbo Chen
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yaying Sun
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Xueping Gu
- Department of Orthopaedics and Sports Medicine, The Northern Branch of Suzhou Municipal Hospital, Suzhou, China
| | - Yuefeng Hao
- Department of Orthopaedics and Sports Medicine, The Northern Branch of Suzhou Municipal Hospital, Suzhou, China
| | - Xingwang Liu
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jinrong Lin
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jiwu Chen
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Shiyi Chen
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
393
|
He B, Chen J, Liu L, Wang H, Wang S, Li P, Zhou J. Knockdown of Tcf3 enhances the wound healing effect of bone marrow mesenchymal stem cells in rats. Biosci Rep 2019; 39:BSR20180369. [PMID: 31085716 PMCID: PMC6712438 DOI: 10.1042/bsr20180369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 01/15/2019] [Accepted: 02/06/2019] [Indexed: 11/17/2022] Open
Abstract
The aim of the present study was to explore the wound healing effect of Tcf3 in rat bone marrow mesenchymal stem cells (BMSCs) and their effects on wound healing. Epidermal growth factor (EGF) and fibroblast growth factors (FGFs) were used to induce BMSCs differentiation into epithelial-like cells. Western Blotting analysis and RT-qPCR were performed to assess the expression levels of Tcf3 and the markers of epithelial-like cells, such as Cytokeratin-18 (CK-18), CK-19 and P63. Cell counting kit-8 (CCK-8) and clone formation assay were carried out to detect cell viability. Immunohistochemistry and HE staining were used to assess the level of Tcf3 protein and skin repair degree, respectively. Rat wound healing model was built to evaluate the effects of BMSCs with altered expression of Tcf3 on wound healing. Results showed that EGF and FGFs stimulation increased the expression of CK-18, CK-19 and P63, improved BMSCs viability, but decreased the expression of Tcf3. Knockdown of Tcf3 in BMSCs increased CK-18, CK-19 and P63 expression and improved cell proliferation, as well as accelerated wound healing process. Moreover, inhibition of Wnt/β-catenin signaling weakened the effect of Tcf3 down-regulation on BMSCs proliferation enhancement. And inhibition of Notch1 signaling impeded the epithelial-like cell differentiation of BMSCs induced by Tcf3 down-regulation. Our study reveals that knockdown of Tcf3 enhances the wound healing process of BMSCs in rat, which provides new approach for accelerating skin regeneration.
Collapse
Affiliation(s)
- Bin He
- Department of Plastic and Reconstructive Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
- Department of Burn and Plastic Surgery, Ningxiang People's Hospital, Ningxiang, Hunan 410600, P.R. China
| | - Jia Chen
- Department of Plastic and Reconstructive Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Liang Liu
- Department of Burn and Plastic Surgery, Ningxiang People's Hospital, Ningxiang, Hunan 410600, P.R. China
| | - Hao Wang
- Department of Burn and Plastic Surgery, Ningxiang People's Hospital, Ningxiang, Hunan 410600, P.R. China
| | - Shaohua Wang
- Department of Plastic and Reconstructive Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Ping Li
- Department of Plastic and Reconstructive Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Jianda Zhou
- Department of Plastic and Reconstructive Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
394
|
Abdik H, Avsar Abdik E, Hızlı Deniz AA, Taşlı PN, Şahin F. A Novel Virtue in Stem Cell Research: Exosomes and Their Role in Differentiation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1144:133-146. [PMID: 30729448 DOI: 10.1007/5584_2019_339] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In the past decade a number of different stem cell types have entered the clinical applications increasingly as a therapeutic option, due to their tissue maintenance capacity at the site where they localize. Although it was initially thought that conferral of resilience to damaged tissue largely depends on the stem cells themselves through orchestration of signaling among the local epithelial and immune systems at the injury site, recent findings point out that the remarkable regenerative capacity of stem cells is rather due to their nanovesicular products that emerge as the new active players of tissue repair processes. Among these extracellular vesicles exosomes generated particularly by stem cells have been receiving a substantial interest both in the fields of stem cell biology and extracellular vesicles. In this chapter fundamental facts about stem cell biology, biogenesis of extracellular vesicles and exosomes, their structure, and function are summarized. Moreover, properties of both tumor-derived exosomes as well as those derived from stem cells are discussed relatively in-depth in terms of their influence on proximal and distal tissue physiology. Last but not the least, among countless studies in an exploding field, we summarize those that attempt to unravel the complex signaling networks through which stem cell-derived exosomes alter the fate of differentiating stem cells as well as the molecular make-up of exosomes released from differentiating stem cells by conducting thorough proteomic and genomic analyses with the ultimate goal of identifying effector gene products mediating exosomal cues in stem cell biology.
Collapse
Affiliation(s)
- Hüseyin Abdik
- Department of Genetics and Bioengineering, Faculty of Engineering and Architecture, Yeditepe University, Istanbul, Turkey.
| | - Ezgi Avsar Abdik
- Department of Genetics and Bioengineering, Faculty of Engineering and Architecture, Yeditepe University, Istanbul, Turkey
| | | | - Pakize Neslihan Taşlı
- Department of Genetics and Bioengineering, Faculty of Engineering and Architecture, Yeditepe University, Istanbul, Turkey
| | - Fikrettin Şahin
- Department of Genetics and Bioengineering, Faculty of Engineering and Architecture, Yeditepe University, Istanbul, Turkey
| |
Collapse
|
395
|
Lam PK, Wang KKW, Chin DWC, Tong CSW, Wang Y, Lo KKY, Lai PBS, Ma H, Zheng VZY, Poon WS, Wong GKC. Topically applied adipose-derived mesenchymal stem cell treatment in experimental focal cerebral ischemia. J Clin Neurosci 2019; 71:226-233. [PMID: 31431402 DOI: 10.1016/j.jocn.2019.08.051] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 08/06/2019] [Indexed: 12/16/2022]
Abstract
In this study, the neuro-modulation effect of topical mesenchymal stem cells (MSCs) was tested in a rodent middle carotid artery occlusion (MCAO) model. Twenty-four hours after MCAO, craniotomy was made and 0.8 × 106 GFP-MSCs were topically applied to the exposed parietal cortex. The MSCs were fixed in position by a thin layer of fibrin glue (N = 30). In the control group, saline were topically applied to the ipsilateral parietal cortex (N = 30). Three days after topical application, few GFP-positive cells were found in the ischemic penumbra. They expressed GFAP and NeuN. Topical MSCs triggered microglial activation, astrocytosis and cellular proliferation at day 3. The recovery of neurological functions were significantly enhanced as determined in Rotarod test and Morris Water Maze test with smaller infarct volume. PCR array showed that expressions of ten genes of neurogenesis were altered in the penumbra region (fold change > 1.25, p < 0.05) in MSCs group: Apoe, Ascl1, Efnb1, Mef2c, Nog, A100a6 and B2m were up-regulated; Pax2, Pax3 and Th were down-regulated. In conclusion, topical application provided a direct and effective transplant method for the delivery of MSCs to the surface of ipsilateral cerebral cortex and the topical MSCs could improve the neurological function from cerebral ischemia resulting from a major cerebral artery occlusion in a rodent experimental model.
Collapse
Affiliation(s)
- Ping Kuen Lam
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong Special Administrative Region; Chow Tai Fook-Cheng Yu Tung Surgical Stem Cell Research Center, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Kevin Ka Wang Wang
- Program for Neurotrauma, Neuroproteomics & Biomarkers Research, Departments of Psychiatry, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Don Wai Ching Chin
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong Special Administrative Region; Chow Tai Fook-Cheng Yu Tung Surgical Stem Cell Research Center, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Cindy See Wai Tong
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong Special Administrative Region; Chow Tai Fook-Cheng Yu Tung Surgical Stem Cell Research Center, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Yixiang Wang
- Department of Imaging and Interventional Radiology, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Kin Ki Yan Lo
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong Special Administrative Region; Chow Tai Fook-Cheng Yu Tung Surgical Stem Cell Research Center, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Paul Bo San Lai
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong Special Administrative Region; Chow Tai Fook-Cheng Yu Tung Surgical Stem Cell Research Center, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Hui Ma
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong Special Administrative Region; Chow Tai Fook-Cheng Yu Tung Surgical Stem Cell Research Center, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Vera Zhi Yuan Zheng
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong Special Administrative Region; Chow Tai Fook-Cheng Yu Tung Surgical Stem Cell Research Center, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Wai Sang Poon
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong Special Administrative Region; Chow Tai Fook-Cheng Yu Tung Surgical Stem Cell Research Center, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - George Kwok Chu Wong
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong Special Administrative Region; Chow Tai Fook-Cheng Yu Tung Surgical Stem Cell Research Center, The Chinese University of Hong Kong, Hong Kong Special Administrative Region.
| |
Collapse
|
396
|
Human placenta-derived mesenchymal stem cells ameliorate orbital adipogenesis in female mice models of Graves' ophthalmopathy. Stem Cell Res Ther 2019; 10:246. [PMID: 31399042 PMCID: PMC6688254 DOI: 10.1186/s13287-019-1348-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 07/03/2019] [Accepted: 07/18/2019] [Indexed: 12/11/2022] Open
Abstract
Background Graves’ ophthalmopathy (GO) is a complication of Graves’ disease (GD), in which orbital connective tissues become inflamed and increase in volume and orbital fibroblasts within the orbital fat and extraocular muscles differentiate into adipocytes in vitro when stimulated by hormones, several cytokines, and growth factors including TSH, IGF-1, IL-1, interferon γ, and platelet-derived growth factor. Human placental mesenchymal stem cells (hPMSCs) have immunomodulatory effects in disease pathogenesis. Although a number of studies have reported that hPMSCs can elicit therapeutic effects, these are not sufficient. Therefore, we constructed a GO animal model in order to find out the hPMSCs recovery effect. Methods We investigated their anti-adipogenic effects in in vitro cultures of orbital fibroblasts established from GO patients. Primary orbital fibroblasts were exposed to differentiation medium for 10 days. After being co-cultured with hPMSCs, the characteristics of orbital fibroblast were determined by Oil Red O stain and real-time PCR. Then, we explored the in vivo regulatory effects of hPMSCs in an experimental mouse model of GO. We developed the GO mouse model using immunization by leg muscle electroporation of pTriEx1.1Neo-hTSHR A-subunit plasmid. Human PMSC injection was performed into the left orbit. We also analyzed the effects of hPMSCs in the GO animal model. Result We found that hPMSCs inhibited a lipid accumulation and activated factors, such as ADIPONECTIN, PPARγ, C/EBPα, and TGFβ2 genes in adipogenesis-induced primary orbital fibroblasts from GO patients. Moreover, hPMSCs were highly effective at ameliorating adipogenesis in the orbital tissue of the model. Conclusion These data indicate that hPMSCs recover pathogenic activation of orbital fibroblasts in animals undergoing experimental GO and confirm the feasibility of applying hPMSCs as a novel treatment for GO patients.
Collapse
|
397
|
Joshi J, Abnavi MD, Kothapalli CR. Synthesis and secretome release by human bone marrow mesenchymal stem cell spheroids within three-dimensional collagen hydrogels: Integrating experiments and modelling. J Tissue Eng Regen Med 2019; 13:1923-1937. [PMID: 31350819 DOI: 10.1002/term.2943] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 07/09/2019] [Accepted: 07/17/2019] [Indexed: 12/12/2022]
Abstract
Myocardial infarction results in loss of cardiac cell types, inflammation, extracellular matrix (ECM) degradation, and fibrotic scar. Transplantation of bone marrow-derived mesenchymal stem cells (BM-MSCs) is being explored as they could differentiate into cardiomyocyte-like cells, integrate into host tissue, and enhance resident cell activity. The ability of these cells to restore lost ECM, remodel the inflammatory scar tissue, and repair the injured myocardium remains unexplored. We here elucidated the synthesis and deposition of ECM (e.g., elastin, sulfated glycosaminoglycans, hyaluronan, collagen type III, laminin, fibrillin, lysyl oxidase, and nitric oxide synthases), matrix metalloproteinases (MMPs) and their inhibitors (TIMPs), and other secretome (cytokines, chemokines, and growth factors) in adult human BM-MSC spheroid cultures within three-dimensional collagen gels. The roles of species-specific type I collagen and 5-azacytadine were assessed over a 28-day period. Results revealed that human collagen (but not rat-derived) suppressed MSC proliferation and survival, and MSCs synthesized and released a variety of ECM proteins and secretome over the 28 days. Matrix deposition is at least an order of magnitude lower than their release levels at every time point, most possibly due to elevated MMP levels and interleukins with a concomitant decrease in TIMPs. Matrix synthesis over the 28-day period was fitted to a competitive inhibition form of Michaelis-Menten kinetics, and the production and decay rates of ECM, MMPs, and TIMPs, along with the kinetic model parameters quantified. Such an integrated experimental and modelling approach would help elucidate the critical roles of various parameters (e.g., cell encapsulation and delivery vehicles) in stem cell-based transplantation therapies.
Collapse
Affiliation(s)
- Jyotsna Joshi
- Department of Chemical and Biomedical Engineering, Cleveland State University, Cleveland, OH, USA
| | | | | |
Collapse
|
398
|
Khalifa J, François S, Rancoule C, Riccobono D, Magné N, Drouet M, Chargari C. Gene therapy and cell therapy for the management of radiation damages to healthy tissues: Rationale and early results. Cancer Radiother 2019; 23:449-465. [PMID: 31400956 DOI: 10.1016/j.canrad.2019.06.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 06/06/2019] [Indexed: 12/14/2022]
Abstract
Nowadays, ionizing radiations have numerous applications, especially in medicine for diagnosis and therapy. Pharmacological radioprotection aims at increasing detoxification of free radicals. Radiomitigation aims at improving survival and proliferation of damaged cells. Both strategies are essential research area, as non-contained radiation can lead to harmful effects. Some advances allowing the comprehension of normal tissue injury mechanisms, and the discovery of related predictive biomarkers, have led to developing several highly promising radioprotector or radiomitigator drugs. Next to these drugs, a growing interest does exist for biotherapy in this field, including gene therapy and cell therapy through mesenchymal stem cells. In this review article, we provide an overview of the management of radiation damages to healthy tissues via gene or cell therapy in the context of radiotherapy. The early management aims at preventing the occurrence of these damages before exposure or just after exposure. The late management offers promises in the reversion of constituted late damages following irradiation.
Collapse
Affiliation(s)
- J Khalifa
- Départment de radiothérapie, institut Claudius-Regaud, institut universitaire du cancer de Toulouse - Oncopole, 1, avenue Irène-Joliot-Curie, 31100 Toulouse, France.
| | - S François
- Institut de recherche biomédicale des armées, BP73, 91223 Brétigny-sur-Orge cedex, France
| | - C Rancoule
- Département de radiothérapie, institut de cancérologie de la Loire Lucien-Neuwirth, 108 bis, avenue Albert-Raimond, 42270 Saint-Priest-en-Jarez, France; Laboratoire de radiobiologie cellulaire et moléculaire, UMR 5822, institut de physique nucléaire de Lyon (IPNL), 69622 Villeurbanne, France; UMR 5822, CNRS, domaine scientifique de la Doua, 4, rue Enrico-Fermi, 69622 Villeurbanne cedex, France; UMR 5822, université Lyon 1, domaine scientifique de la Doua, 4, rue Enrico-Fermi, 69622 Villeurbanne cedex, France; UMR 5822, université de Lyon, domaine scientifique de la Doua, 4, rue Enrico-Fermi, 69622 Villeurbanne cedex, France
| | - D Riccobono
- Institut de recherche biomédicale des armées, BP73, 91223 Brétigny-sur-Orge cedex, France
| | - N Magné
- Département de radiothérapie, institut de cancérologie de la Loire Lucien-Neuwirth, 108 bis, avenue Albert-Raimond, 42270 Saint-Priest-en-Jarez, France; Laboratoire de radiobiologie cellulaire et moléculaire, UMR 5822, institut de physique nucléaire de Lyon (IPNL), 69622 Villeurbanne, France; UMR 5822, CNRS, domaine scientifique de la Doua, 4, rue Enrico-Fermi, 69622 Villeurbanne cedex, France; UMR 5822, université Lyon 1, domaine scientifique de la Doua, 4, rue Enrico-Fermi, 69622 Villeurbanne cedex, France; UMR 5822, université de Lyon, domaine scientifique de la Doua, 4, rue Enrico-Fermi, 69622 Villeurbanne cedex, France
| | - M Drouet
- Institut de recherche biomédicale des armées, BP73, 91223 Brétigny-sur-Orge cedex, France
| | - C Chargari
- Institut de recherche biomédicale des armées, BP73, 91223 Brétigny-sur-Orge cedex, France; Service de santé des armées, école du Val-de-Grâce, 74, boulevard de Port-Royal, 75005 Paris, France; Département de radiothérapie, Gustave-Roussy Cancer Campus, 114, rue Édouard-Vailant, 94805 Villejuif, France
| |
Collapse
|
399
|
Arteaga Cabeza O, Mikrogeorgiou A, Kannan S, Ferriero DM. Advanced nanotherapies to promote neuroregeneration in the injured newborn brain. Adv Drug Deliv Rev 2019; 148:19-37. [PMID: 31678359 DOI: 10.1016/j.addr.2019.10.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 09/19/2019] [Accepted: 10/23/2019] [Indexed: 12/16/2022]
Abstract
Neonatal brain injury affects thousands of babies each year and may lead to long-term and permanent physical and neurological problems. Currently, therapeutic hypothermia is standard clinical care for term newborns with moderate to severe neonatal encephalopathy. Nevertheless, it is not completely protective, and additional strategies to restore and promote regeneration are urgently needed. One way to ensure recovery following injury to the immature brain is to augment endogenous regenerative pathways. However, novel strategies such as stem cell therapy, gene therapies and nanotechnology have not been adequately explored in this unique age group. In this perspective review, we describe current efforts that promote neuroprotection and potential targets that are unique to the developing brain, which can be leveraged to facilitate neuroregeneration.
Collapse
|
400
|
Xie P, Deng M, Sun QG, Ma YG, Zhou Y, Ming JH, Chen Q, Liu SQ, Liu JQ, Cai J, Wu F. Therapeutic effect of transplantation of human bone marrow‑derived mesenchymal stem cells on neuron regeneration in a rat model of middle cerebral artery occlusion. Mol Med Rep 2019; 20:3065-3074. [PMID: 31432152 PMCID: PMC6755237 DOI: 10.3892/mmr.2019.10536] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 05/31/2019] [Indexed: 12/15/2022] Open
Abstract
Human bone marrow-derived mesenchymal stromal cells (hBMSCs) have been revealed to be beneficial for the regeneration of tissues and cells in several diseases. The present study aimed to elucidate the mechanisms underlying the effect of hBMSC transplantation on neuron regeneration in a rat model of middle cerebral artery occlusion (MCAO). The hBMSCs were isolated, cultured and identified. A rat model of MCAO was induced via the modified Longa method. Neurological severity scores (NSS) were adopted for the evaluation of neuronal function in the model rats after cell transplantation. Next, the expression levels of nestin, β-III-tubulin (β-III-Tub), glial fibrillary acidic protein (GFAP), HNA and neuronal nuclear antigen (NeuN) were examined, as well as the positive expression rates of human neutrophil alloantigen (HNA), nestin, NeuN, β-III-Tub and GFAP. The NSS, as well as the mRNA and protein expression of nestin, decreased at the 1st, 2nd, 4 and 8th weeks, while the mRNA and protein expression of NeuN, β-III-Tub and GFAP increased with time. In addition, after treatment, the MCAO rats showed decreased NSS and mRNA and protein expression of nestin, but elevated mRNA and protein expression of NeuN, β-III-Tub and GFAP at the 2nd, 4 and 8th weeks, and decreased positive expression of HNA and nestin with enhanced expression of NeuN, β-III-Tub and GFAP. Therefore, the present findings demonstrated that hBMSC transplantation triggered the formation of nerve cells and enhanced neuronal function in a rat model of MCAO.
Collapse
Affiliation(s)
- Ping Xie
- Department of Chinese Traditional Medicine, Tongren Hospital of Wuhan University (Wuhan Third Hospital), Wuhan, Hubei 430060, P.R. China
| | - Ming Deng
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qin-Guo Sun
- Department of Chinese Traditional Medicine, Tongren Hospital of Wuhan University (Wuhan Third Hospital), Wuhan, Hubei 430060, P.R. China
| | - Yong-Gang Ma
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yan Zhou
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Jiang-Hua Ming
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qing Chen
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Shi-Qing Liu
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Jun-Qi Liu
- Department of Radiation Oncology, The First of Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 45003, P.R. China
| | - Jun Cai
- Department of Emergency and Trauma Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Fei Wu
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|