1
|
Lin H, Zhou C, Li Q, Xie Q, Xia L, Liu L, Bao W, Xiong X, Zhang H, Zheng Z, Zhao J, Liang W. Nanotechnology-Assisted mesenchymal stem cells treatment for improved cartilage regeneration: A review of current practices. Biochem Pharmacol 2025; 237:116895. [PMID: 40154890 DOI: 10.1016/j.bcp.2025.116895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 02/26/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025]
Abstract
Cartilage tissue does not promptly elicit an inflammatory response upon injury, hence constraining its capacity for healing and self-regeneration. Mesenchymal Stem Cells (MSC) therapy, enhanced by nanotechnology, offers promising advancements in cartilage repair. Injuries to cartilage often cause chronic pain, where current treatments are inadequate. As MSCs can readily differentiate into chondrocytes and secrete soluble factors, they are essential components in tissue engineering of cartilage repair. Although, like other stem cell applications, clinical applications are restricted by poor post implantation survival and differentiation. Recent studies show that nanoparticles (NPs) can further improve MSC outcomes by promoting cell adhesion, and chondrogenic differentiation allowing for sustained growth factor release. In addition, nanomaterials can improve the biological activity of MSCs, by also facilitating the composition of a conducive microenvironment for cartilage repair. In this review, the application of nanofibrous scaffolds, hydrogels and nanoscale particulate matter to improve mechanical properties in cartilage tissue engineering, are discussed. Moreover, the MSCs and nanotechnology synergistic effects present hope of overcoming the limitations of conventional treatments. Nanotechnology greatly enhances the MSC based cartilage regeneration strategies and could provide better treatment for cartilage related diseases in the future. Future research should be aimed at standardizing MSC harvesting and culturing protocols and contrasting their long-term efficacy.
Collapse
Affiliation(s)
- Hongming Lin
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000 Zhejiang Province, China
| | - Chao Zhou
- Department of Orthopedics, Zhoushan Guanghua hospital, Zhoushan 316000 Zhejiang Province, China
| | - Qingping Li
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000 Zhejiang Province, China
| | - Qiong Xie
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000 Zhejiang Province, China
| | - Linying Xia
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000 Zhejiang Province, China
| | - Lu Liu
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000 Zhejiang Province, China
| | - Wenwen Bao
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000 Zhejiang Province, China
| | - Xiaochun Xiong
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000 Zhejiang Province, China
| | - Hao Zhang
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000 Zhejiang Province, China
| | - Zeping Zheng
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000 Zhejiang Province, China
| | - Jiayi Zhao
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000 Zhejiang Province, China.
| | - Wenqing Liang
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000 Zhejiang Province, China.
| |
Collapse
|
2
|
Ross M, Aldred S, Drayson MT, Bosch JA, Turner JE. The magnitude of exercise-induced progenitor cell mobilisation and extravasation is positively associated with cardiorespiratory fitness. Exp Physiol 2025; 110:206-214. [PMID: 39476319 PMCID: PMC11782223 DOI: 10.1113/ep092041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 10/08/2024] [Indexed: 02/01/2025]
Abstract
CD34+ progenitor cells with angiogenic capabilities traffic into blood during exercise and extravasate afterwards but the magnitude of this response varies between people. We examined whether exercise-induced progenitor cell trafficking is influenced by cardiorespiratory fitness (maximum oxygen uptake;V ̇ O 2 max ${{\dot{V}}_{{{{\mathrm{O}}}_2}{\mathrm{max}}}}$ ). Ten males (age: 23 ± 3 years;V ̇ O 2 max ${{\dot{V}}_{{{{\mathrm{O}}}_2}{\mathrm{max}}}}$ : 61.88 ± 4.68 mL kg min-1) undertook 1 h of treadmill running at 80% ofV ̇ O 2 max ${{\dot{V}}_{{{{\mathrm{O}}}_2}{\mathrm{max}}}}$ . Blood samples were collected before exercise (Pre), in the final minute of exercise (0 h) and afterwards at 0.25, 1 and 24 h. Pan-progenitor cells (CD34+, CD34+CD45dim) and putative endothelial progenitor cells (CD34+CD133+, CD34+VEGFR2+, CD34+CD45dimVEGFR2+) were quantified using flow cytometry. Progenitor subpopulations (except for CD34+CD45dimVEGFR2+) increased at 0 h (P < 0.05) and returned to pre-exercise levels by 1 h.V ̇ O 2 max ${{\dot{V}}_{{{{\mathrm{O}}}_2}{\mathrm{max}}}}$ was positively associated with the exercise-induced progenitor cell response and there were statistically significant time ×V ̇ O 2 max ${{\dot{V}}_{{{{\mathrm{O}}}_2}{\mathrm{max}}}}$ interactions for CD34+, CD34+CD45dim and CD34+CD133+ subpopulations but not VEGFR2-expressing progenitor cells. There were statistically significant correlations betweenV ̇ O 2 max ${{\dot{V}}_{{{{\mathrm{O}}}_2}{\mathrm{max}}}}$ and ingress (r > 0.70, P < 0.025) and egress (r > -0.77, P < 0.009) of progenitor cell subsets (CD34+, CD34+CD45dim, CD34+CD133+), showing that cardiorespiratory fitness influences the magnitude of progenitor cell mobilisation into the blood and subsequent extravasation. These data may provide a link between high levels of cardiorespiratory fitness and vascular health.
Collapse
Affiliation(s)
- Mark Ross
- Institue of Life and Earth SciencesHeriot‐Watt UniversityEdinburghUK
| | - Sarah Aldred
- School of Sport, Exercise and Rehabilitation SciencesUniversity of BirminghamBirminghamUK
| | - Mark T. Drayson
- Institute of Immunology and ImmunotherapyUniversity of BirminghamBirminghamUK
| | - Jos A. Bosch
- Department of Clinical PsychologyUniversity of AmsterdamAmsterdamThe Netherlands
| | - James E. Turner
- School of Sport, Exercise and Rehabilitation SciencesUniversity of BirminghamBirminghamUK
| |
Collapse
|
3
|
Xu HR, Le VV, Oprescu SN, Kuang S. Muscle stem cells as immunomodulator during regeneration. Curr Top Dev Biol 2024; 158:221-238. [PMID: 38670707 PMCID: PMC11801201 DOI: 10.1016/bs.ctdb.2024.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
The skeletal muscle is well known for its remarkable ability to regenerate after injuries. The regeneration is a complex and dynamic process that involves muscle stem cells (also called muscle satellite cells, MuSCs), fibro-adipogenic progenitors (FAPs), immune cells, and other muscle-resident cell populations. The MuSCs are the myogenic cell populaiton that contribute nuclei directly to the regenerated myofibers, while the other cell types collaboratively establish a microenvironment that facilitates myogenesis of MuSCs. The myogenic process includes activation, proliferation and differentiationof MuSCs, and subsequent fusion their descendent mononuclear myocytes into multinuclear myotubes. While the contributions of FAPs and immune cells to this microenvironment have been well studied, the influence of MuSCs on other cell types remains poorly understood. This review explores recent evidence supporting the potential role of MuSCs as immunomodulators during muscle regeneration, either through cytokine production or ligand-receptor interactions.
Collapse
Affiliation(s)
- H Rex Xu
- Department of Animal Sciences, Purdue University, West Lafayette, IN, United States
| | - Victor V Le
- Department of Biological Sciences, Purdue University, West Lafayette, IN, United States
| | - Stephanie N Oprescu
- Department of Biological Sciences, Purdue University, West Lafayette, IN, United States
| | - Shihuan Kuang
- Department of Animal Sciences, Purdue University, West Lafayette, IN, United States; Purdue University Institute for Cancer Research, West Lafayette, IN, United States.
| |
Collapse
|
4
|
Archacka K, Grabowska I, Mierzejewski B, Graffstein J, Górzyńska A, Krawczyk M, Różycka AM, Kalaszczyńska I, Muras G, Stremińska W, Jańczyk-Ilach K, Walczak P, Janowski M, Ciemerych MA, Brzoska E. Hypoxia preconditioned bone marrow-derived mesenchymal stromal/stem cells enhance myoblast fusion and skeletal muscle regeneration. Stem Cell Res Ther 2021; 12:448. [PMID: 34372911 PMCID: PMC8351116 DOI: 10.1186/s13287-021-02530-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 07/08/2021] [Indexed: 12/19/2022] Open
Abstract
Background The skeletal muscle reconstruction occurs thanks to unipotent stem cells, i.e., satellite cells. The satellite cells remain quiescent and localized between myofiber sarcolemma and basal lamina. They are activated in response to muscle injury, proliferate, differentiate into myoblasts, and recreate myofibers. The stem and progenitor cells support skeletal muscle regeneration, which could be disturbed by extensive damage, sarcopenia, cachexia, or genetic diseases like dystrophy. Many lines of evidence showed that the level of oxygen regulates the course of cell proliferation and differentiation. Methods In the present study, we analyzed hypoxia impact on human and pig bone marrow-derived mesenchymal stromal cell (MSC) and mouse myoblast proliferation, differentiation, and fusion. Moreover, the influence of the transplantation of human bone marrow-derived MSCs cultured under hypoxic conditions on skeletal muscle regeneration was studied. Results We showed that bone marrow-derived MSCs increased VEGF expression and improved myogenesis under hypoxic conditions in vitro. Transplantation of hypoxia preconditioned bone marrow-derived MSCs into injured muscles resulted in the improved cell engraftment and formation of new vessels. Conclusions We suggested that SDF-1 and VEGF secreted by hypoxia preconditioned bone marrow-derived MSCs played an essential role in cell engraftment and angiogenesis. Importantly, hypoxia preconditioned bone marrow-derived MSCs more efficiently engrafted injured muscles; however, they did not undergo myogenic differentiation. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02530-3.
Collapse
Affiliation(s)
- Karolina Archacka
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Iwona Grabowska
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Bartosz Mierzejewski
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Joanna Graffstein
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Alicja Górzyńska
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Marta Krawczyk
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Anna M Różycka
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Ilona Kalaszczyńska
- Department of Histology and Embryology, Medical University of Warsaw, 02-004, Warsaw, Poland.,Laboratory for Cell Research and Application, Medical University of Warsaw, 02-097, Warsaw, Poland
| | - Gabriela Muras
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Władysława Stremińska
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Katarzyna Jańczyk-Ilach
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Piotr Walczak
- Department of Pathophysiology, Faculty of Medical Sciences, University of Warmia and Mazury, Warszawska 30 St, 10-082, Olsztyn, Poland.,Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, the Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Mirosław Janowski
- Center for Advanced Imaging Research, Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland, Baltimore, MD, 21201, USA.,NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Pawinskiego 5 St, 02-106, Warsaw, Poland
| | - Maria A Ciemerych
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Edyta Brzoska
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland.
| |
Collapse
|
5
|
Wang M, Xu X, Lei X, Tan J, Xie H. Mesenchymal stem cell-based therapy for burn wound healing. BURNS & TRAUMA 2021; 9:tkab002. [PMID: 34212055 PMCID: PMC8240555 DOI: 10.1093/burnst/tkab002] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/14/2020] [Indexed: 02/05/2023]
Abstract
Burns, with their high incidence and mortality rates, have a devastating effect on patients. There are still huge challenges in the management of burns. Mesenchymal stem cells (MSCs), which have multidirectional differentiation potential, have aroused interest in exploring the capacity for treating different intractable diseases due to their strong proliferation, tissue repair, immune tolerance and paracrine abilities, among other features. Currently, several animal studies have shown that MSCs play various roles and have beneficial effects in promoting wound healing, inhibiting burn inflammation and preventing the formation of pathological scars during burn healing process. The substances MSCs secrete can act on peripheral cells and promote burn repair. According to preclinical research, MSC-based treatments can effectively improve burn wound healing and reduce pain. However, due to the small number of patients and the lack of controls, treatment plans and evaluation criteria vary widely, thus limiting the value of these clinical studies. Therefore, to better evaluate the safety and effectiveness of MSC-based burn treatments, standardization of the application scheme and evaluation criteria of MSC therapy in burn treatment is required in the future. In addition, the combination of MSC pretreatment and dressing materials are also conducive to improving the therapeutic effect of MSCs on burns. In this article, we review current animal research and clinical trials based on the use of stem cell therapy for treating burns and discuss the main challenges and coping strategies facing future clinical applications.
Collapse
Affiliation(s)
- Mingyao Wang
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Keyuan Road 4, Gaopeng Street, Chengdu, Sichuan 610041, China
| | - Xinxuan Xu
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Keyuan Road 4, Gaopeng Street, Chengdu, Sichuan 610041, China
| | - Xiongxin Lei
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Keyuan Road 4, Gaopeng Street, Chengdu, Sichuan 610041, China
| | - Jie Tan
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Keyuan Road 4, Gaopeng Street, Chengdu, Sichuan 610041, China
| | - Huiqi Xie
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Keyuan Road 4, Gaopeng Street, Chengdu, Sichuan 610041, China
| |
Collapse
|
6
|
Oprescu SN, Yue F, Qiu J, Brito LF, Kuang S. Temporal Dynamics and Heterogeneity of Cell Populations during Skeletal Muscle Regeneration. iScience 2020; 23:100993. [PMID: 32248062 PMCID: PMC7125354 DOI: 10.1016/j.isci.2020.100993] [Citation(s) in RCA: 160] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 01/08/2020] [Accepted: 03/13/2020] [Indexed: 12/20/2022] Open
Abstract
Mammalian skeletal muscle possesses a unique ability to regenerate, which is primarily mediated by a population of resident muscle stem cells (MuSCs) and requires a concerted response from other supporting cell populations. Previous targeted analysis has described the involvement of various specific populations in regeneration, but an unbiased and simultaneous evaluation of all cell populations has been limited. Therefore, we used single-cell RNA-sequencing to uncover gene expression signatures of over 53,000 individual cells during skeletal muscle regeneration. Cells clustered into 25 populations and subpopulations, including a subpopulation of immune gene enriched myoblasts (immunomyoblasts) and subpopulations of fibro-adipogenic progenitors. Our analyses also uncovered striking spatiotemporal dynamics in gene expression, population composition, and cell-cell interaction during muscle regeneration. These findings provide insights into the cellular and molecular underpinning of skeletal muscle regeneration.
Collapse
Affiliation(s)
- Stephanie N Oprescu
- Department of Biological Sciences, Purdue University, 915 W State St, West Lafayette, IN 47907, USA
| | - Feng Yue
- Department of Animal Sciences, Purdue University, 270 S Russell St, West Lafayette, IN 47907, USA
| | - Jiamin Qiu
- Department of Animal Sciences, Purdue University, 270 S Russell St, West Lafayette, IN 47907, USA
| | - Luiz F Brito
- Department of Animal Sciences, Purdue University, 270 S Russell St, West Lafayette, IN 47907, USA
| | - Shihuan Kuang
- Department of Biological Sciences, Purdue University, 915 W State St, West Lafayette, IN 47907, USA; Department of Animal Sciences, Purdue University, 270 S Russell St, West Lafayette, IN 47907, USA; Center for Cancer Research, Purdue University, 201 S University St, West Lafayette, IN 47907, USA.
| |
Collapse
|
7
|
Kowalski K, Dos Santos M, Maire P, Ciemerych MA, Brzoska E. Induction of bone marrow-derived cells myogenic identity by their interactions with the satellite cell niche. Stem Cell Res Ther 2018; 9:258. [PMID: 30261919 PMCID: PMC6161400 DOI: 10.1186/s13287-018-0993-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 08/22/2018] [Accepted: 08/23/2018] [Indexed: 12/25/2022] Open
Abstract
Background Skeletal muscle regeneration is possible thanks to unipotent stem cells, which are satellite cells connected to the myofibers. Populations of stem cells other than muscle-specific satellite cells are considered as sources of cells able to support skeletal muscle reconstruction. Among these are bone marrow-derived mesenchymal stem cells (BM-MSCs), which are multipotent, self-renewing stem cells present in the bone marrow stroma. Available data documenting the ability of BM-MSCs to undergo myogenic differentiation are not definitive. In the current work, we aimed to check if the satellite cell niche could impact the ability of bone marrow-derived cells to follow a myogenic program. Methods We established a new in-vitro method for the coculture of bone marrow-derived cells (BMCs) that express CXCR4 (CXCR4+BMCs; the stromal-derived factor-1 (Sdf-1) receptor) with myofibers. Using various tests, we analyzed the myogenic identity of BMCs and their ability to fuse with myoblasts in vitro and in vivo. Results We showed that Sdf-1 treatment increased the number of CXCR4+BMCs able to bind the myofiber and occupy the satellite cell niche. Moreover, interaction with myofibers induced the expression of myogenic regulatory factors (MRFs) in CXCR4+BMCs. CXCR4+BMCs, pretreated by the coculture with myofibers and Sdf-1, participated in myotube formation in vitro and also myofiber reconstruction in vivo. We also showed that Sdf-1 overexpression in vivo (in injured and regenerating muscles) supported the participation of CXCR4+BMCs in new myofiber formation. Conclusion We showed that CXCR4+BMC interaction with myofibers (that is, within the satellite cell niche) induced CXCR4+BMC myogenic commitment. CXCR4+BMCs, pretreated using such a method of culture, were able to participate in skeletal muscle regeneration.
Collapse
Affiliation(s)
- Kamil Kowalski
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Matthieu Dos Santos
- Institut Cochin, Université Paris-Descartes, Centre National de la Recherche Scientifique (CNRS), UMR 8104, Paris, France.,Institut National de la Santé et de la Recherche Médicale (INSERM) U1016, Paris, France
| | - Pascal Maire
- Institut Cochin, Université Paris-Descartes, Centre National de la Recherche Scientifique (CNRS), UMR 8104, Paris, France.,Institut National de la Santé et de la Recherche Médicale (INSERM) U1016, Paris, France
| | - Maria A Ciemerych
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Edyta Brzoska
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland.
| |
Collapse
|
8
|
Niemiro GM, Edwards T, Barfield JP, Beals JW, Broad EM, Motl RW, Burd NA, Pilutti LA, DE Lisio M. Circulating Progenitor Cell Response to Exercise in Wheelchair Racing Athletes. Med Sci Sports Exerc 2018; 50:88-97. [PMID: 28806276 DOI: 10.1249/mss.0000000000001402] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
INTRODUCTION Circulating progenitor cells (CPC) are a heterogeneous population of stem/progenitor cells in peripheral blood that participate in tissue repair. CPC mobilization has been well characterized in able-bodied persons but has not been previously investigated in wheelchair racing athletes. The purpose of this study was to characterize CPC and CPC subpopulation mobilization in elite wheelchair racing athletes in response to acute, upper-extremity aerobic exercise to determine whether CPC responses are similar to ambulatory populations. METHODS Eight participants (three females; age = 27.5 ± 4.0 yr, supine height = 162.5 ± 18.6 cm, weight = 53.5 ± 10.9 kg, V˙O2peak = 2.4 ± 0.62 L·min, years postinjury = 21.5 ± 6.2 yr) completed a 25-km time trial on a road course. Blood sampling occurred before and immediately after exercise for quantification of CPC (CD34), hematopoietic stem and progenitor cells (HSPC) (CD34/CD45), hematopoietic stem cells (HSC) (CD34/CD45/CD38), CD34 adipose tissue (AT)-derived mesenchymal stromal cells (MSC) (CD45/CD34/CD105/CD31), CD34 bone marrow (BM)-derived MSC (CD45/CD34/CD105/CD31), and endothelial progenitor cells (EPC) (CD45/CD34/VEGFR2) via flow cytometry. Blood lactate was measured before and after trial as an indicator of exercise intensity. RESULTS CPC concentration increased 5.7-fold postexercise (P = 0.10). HSPC, HSC, EPC, and both MSC populations were not increased postexercise. Baseline HSPC populations were significantly positively correlated to absolute V˙O2peak (rho = 0.71, P < 0.05) with HSC trending to positively correlate to V˙O2peak (rho = 0.62, P = 0.10). AT-MSC populations were trending to be negatively correlated to baseline V˙O2peak (rho = -0.62, P = 0.058). The change in CPC, EPC, and AT-MSC pre- and postexercise significantly positively correlated to the change in lactate concentrations (rho = 0.91 P = 0.002, 0.71 P = 0.047, 0.81 P = 0.02, respectively, all P < 0.05). CONCLUSION These data suggest that CPC content in wheelchair racing athletes is related to cardiorespiratory fitness, and responses to exercise are positively related to exercise intensity.
Collapse
Affiliation(s)
- Grace M Niemiro
- 1Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, IL; 2Department of Health and Human Performance, Radford University, Radford, VA; 3U.S. Paralympics, Colorado Springs, CO; 4Department of Physical Therapy, University of Alabama-Birmingham, Birmingham, AL; 5Interdisciplinary School of Health Sciences, University of Ottawa, Ottawa, ON, CANADA; and 6School of Human Kinetics, Brain and Mind Research Institute, and Centre for Neuromuscular Disease, University of Ottawa, Ottawa, ON, CANADA
| | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Characterisation and immunosuppressive activity of human cartilage-derived mesenchymal stem cells. Cytotechnology 2018; 70:1037-1050. [PMID: 29497876 DOI: 10.1007/s10616-017-0182-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 12/07/2017] [Indexed: 01/04/2023] Open
Abstract
Mesenchymal stem cells (MSCs) exert potent immuno-regulatory activities on various immune cells and also differentiate into various mesodermal lineages besides retaining a distinct self-renewal ability. Such exclusive characteristics had enabled MSCs to be recognised as an ideal source for cell-based treatment in regenerative medicine and immunotherapy. Thus, considering MSCs for treating degenerative disease of organs with limited regenerative potential such as cartilage would serve as an ideal therapy. This study explored the feasibility of generating human cartilage-derived MSCs (hC-MSCs) from sports injured patients and characterised based on multipotent differentiation and immunosuppressive activities. Cartilage tissues harvested from a non-weight bearing region during an arthroscopy procedure were used to generate MSCs. Despite the classic morphology of fibroblast-like cells and a defined immunophenotyping, MSCs expressed early embryonic transcriptional markers (SOX2, REX1, OCT4 and NANOG) and differentiated into chondrocytes, adipocytes and osteocytes when induced accordingly. Upon co-culture with PHA-L activated T-cells, hC-MSCs suppressed the proliferation of the T-cells in a dose-dependent manner. Although, hC-MSCs did not alter the activation profile of T cells significantly, yet prevented the entering of activated T cells into S phase of the cell cycle by cell cycle arrest. The present study has strengthened the evidence of tissue-resident mesenchymal stem cells in human cartilage tissue. The endogenous MSCs could be an excellent tool in treating dysregulated immune response that associated with cartilage since hC-MSCs exerted both immunosuppressive and regenerative capabilities.
Collapse
|
10
|
Agha NH, Baker FL, Kunz HE, Graff R, Azadan R, Dolan C, Laughlin MS, Hosing C, Markofski MM, Bond RA, Bollard CM, Simpson RJ. Vigorous exercise mobilizes CD34+ hematopoietic stem cells to peripheral blood via the β 2-adrenergic receptor. Brain Behav Immun 2018; 68:66-75. [PMID: 29017969 PMCID: PMC6980177 DOI: 10.1016/j.bbi.2017.10.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 09/21/2017] [Accepted: 10/01/2017] [Indexed: 01/03/2023] Open
Abstract
Acute dynamic exercise mobilizes CD34+ hematopoietic stem cells (HSCs) to the bloodstream, potentially serving as an economical adjuvant to boost the collection of HSCs from stem cell transplant donors. The mechanisms responsible for HSC mobilization with exercise are unknown but are likely due to hemodynamic perturbations, endogenous granulocyte-colony stimulating factor (G-CSF), and/or β2-adrenergic receptor (β2-AR) signaling. We characterized the temporal response of HSC mobilization and plasma G-CSF following exercise, and determined the impact of in vivo β-AR blockade on the exercise-induced mobilization of HSCs. Healthy runners (n = 15) completed, in balanced order, two single bouts of steady state treadmill running exercise at moderate (lasting 90-min) or vigorous (lasting 30-min) intensity. A separate cohort of healthy cyclists (n = 12) completed three 30-min cycling ergometer trials at vigorous intensity after ingesting: (i) 10 mg bisoprolol (β1-AR antagonist); (ii) 80 mg nadolol (β1 + β2-AR antagonist); or (iii) placebo, in balanced order with a double-blind design. Blood samples collected before, during (runners only), immediately after, and at several points during exercise recovery were used to determine circulating G-CSF levels (runners only) and enumerate CD34+ HSCs by flow cytometry (runners and cyclists). Steady state vigorous but not moderate intensity exercise mobilized HSCs, increasing the total blood CD34+ count by ∼4.15 ± 1.62 Δcells/µl (+202 ± 92%) compared to resting conditions. Plasma G-CSF increased in response to moderate but not vigorous exercise. Relative to placebo, nadolol and bisoprolol lowered exercising heart rate and blood pressure to comparable levels. The number of CD34+ HSCs increased with exercise after the placebo and bisoprolol trials, but not the nadolol trial, suggesting β2-AR signaling mediated the mobilization of CD34+ cells [Placebo: 2.10 ± 1.16 (207 ± 69.2%), Bisoprolol 1.66 ± 0.79 (+163 ± 29%), Nadolol: 0.68 ± 0.54 (+143 ± 36%) Δcells/µL]. We conclude that the mobilization of CD34+ HSCs with exercise is not dependent on circulating G-CSF and is likely due to the combined actions of β2-AR signaling and hemodynamic shear stress.
Collapse
Affiliation(s)
- Nadia H Agha
- Laboratory of Integrated Physiology, Department of Health and Human Performance, University of Houston, 3875 Holman Street, Houston, TX 77204, USA
| | - Forrest L Baker
- Laboratory of Integrated Physiology, Department of Health and Human Performance, University of Houston, 3875 Holman Street, Houston, TX 77204, USA
| | - Hawley E Kunz
- Laboratory of Integrated Physiology, Department of Health and Human Performance, University of Houston, 3875 Holman Street, Houston, TX 77204, USA
| | - Rachel Graff
- Laboratory of Integrated Physiology, Department of Health and Human Performance, University of Houston, 3875 Holman Street, Houston, TX 77204, USA
| | - Rod Azadan
- Laboratory of Integrated Physiology, Department of Health and Human Performance, University of Houston, 3875 Holman Street, Houston, TX 77204, USA
| | - Chad Dolan
- Laboratory of Integrated Physiology, Department of Health and Human Performance, University of Houston, 3875 Holman Street, Houston, TX 77204, USA
| | - Mitzi S Laughlin
- Laboratory of Integrated Physiology, Department of Health and Human Performance, University of Houston, 3875 Holman Street, Houston, TX 77204, USA
| | - Chitra Hosing
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Melissa M Markofski
- Laboratory of Integrated Physiology, Department of Health and Human Performance, University of Houston, 3875 Holman Street, Houston, TX 77204, USA
| | - Richard A Bond
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, TX, USA
| | - Catherine M Bollard
- Program for Cell Enhancement and Technologies for Immunotherapy, Children's National Health System and The George Washington University, Washington D.C., USA
| | - Richard J Simpson
- Laboratory of Integrated Physiology, Department of Health and Human Performance, University of Houston, 3875 Holman Street, Houston, TX 77204, USA; Department of Behavioral Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Nutritional Sciences, University of Arizona, Tucson, AZ, USA; Department of Pediatrics, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
11
|
Čížková D, Komárková Z, Bezrouk A, Macháčková L, Vávrová J, Filip S, Mokrý J. Bone Marrow-Derived Cells Participate in Composition of the Satellite Cell Niche in Intact and Regenerating Mouse Skeletal Muscle. Folia Biol (Praha) 2018; 64:155-166. [PMID: 30938672 DOI: 10.14712/fb2018064050155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
The cellular components of the satellite cell niche participate in the regulation of skeletal muscle regeneration. Beside myogenic cells at different developmental stages, this niche is formed by cells of the immune system, the interstitial connective tissue and the vascular system. Unambiguous determination of the origin of these cell types could contribute to optimization of the cell-based therapy of skeletal muscle disorders. In our work, we intravenously transplanted mouse GFP+ unseparated bone marrow cells into whole-body lethally irradiated immunocompetent mice four weeks before cardiotoxin-induced injury of the recipients' skeletal muscles. Seven and 28 days after the toxin injection, the injured regenerating and contralateral intact muscles were examined for identification of GFP+ bone marrow-derived cells by direct fluorescence, protein immunohistochemistry and immunogold transmission electron microscopy. In both the intact and injured muscles, GFP positivity was determined in immune cells, mainly in macrophages, and in interstitial spindleshaped cells. Moreover, in the injured muscles, rare GFP+ endothelial cells of the blood vessels and newly formed myotubes and muscle fibres were present. Our results confirmed the ability of bone marrowderived cells to contribute to the cellular component of the satellite cell niche in the intact and regenerating skeletal muscle. These cells originated not only from haematopoietic stem cells, but obviously also from other stem or progenitor cells residing in the bone marrow, such as multipotent mesenchymal stromal cells and endothelial progenitors.
Collapse
Affiliation(s)
- D Čížková
- Department of Histology and Embryology, Faculty of Medicine in Hradec Králové, Charles University, Czech Republic
| | - Z Komárková
- Department of Histology and Embryology, Faculty of Medicine in Hradec Králové, Charles University, Czech Republic
| | - A Bezrouk
- Department of Medical Biophysics, Faculty of Medicine in Hradec Králové, Charles University, Czech Republic
| | - L Macháčková
- Department of Histology and Embryology, Faculty of Medicine in Hradec Králové, Charles University, Czech Republic
| | - J Vávrová
- Department of Radiobiology, Faculty of Military Health Sciences in Hradec Králové, University of Defence, Czech Republic
| | - S Filip
- Department of Oncology and Radiotherapy, Faculty of Medicine in Hradec Králové, Charles University, Czech Republic
| | - J Mokrý
- Department of Histology and Embryology, Faculty of Medicine in Hradec Králové, Charles University, Czech Republic
| |
Collapse
|
12
|
Abstract
The number of clinical trials in regenerative medicine is burgeoning, and stem cell/tissue engineering technologies hold the possibility of becoming the standard of care for a multitude of diseases and injuries. Advances in regenerative biology reveal novel molecular and cellular targets, with potential to optimize tissue healing and functional recovery, thereby refining rehabilitation clinical practice. The purpose of this review is to (1) highlight the potential for synergy between the fields of regenerative medicine and rehabilitation, a convergence of disciplines known as regenerative rehabilitation; (2) provide translational examples of regenerative rehabilitation within the context of neuromuscular injuries and diseases; and (3) offer recommendations for ways to leverage activity dependence via combined therapy and technology, with the goal of enhancing long-term recovery. The potential clinical benefits of regenerative rehabilitation will likely become a critical aspect in the standard of care for many neurological and musculoskeletal disorders.
Collapse
|
13
|
Stearns-Reider KM, D'Amore A, Beezhold K, Rothrauff B, Cavalli L, Wagner WR, Vorp DA, Tsamis A, Shinde S, Zhang C, Barchowsky A, Rando TA, Tuan RS, Ambrosio F. Aging of the skeletal muscle extracellular matrix drives a stem cell fibrogenic conversion. Aging Cell 2017; 16:518-528. [PMID: 28371268 PMCID: PMC5418187 DOI: 10.1111/acel.12578] [Citation(s) in RCA: 170] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/08/2017] [Indexed: 12/13/2022] Open
Abstract
Age‐related declines in skeletal muscle regeneration have been attributed to muscle stem cell (MuSC) dysfunction. Aged MuSCs display a fibrogenic conversion, leading to fibrosis and impaired recovery after injury. Although studies have demonstrated the influence of in vitro substrate characteristics on stem cell fate, whether and how aging of the extracellular matrix (ECM) affects stem cell behavior has not been investigated. Here, we investigated the direct effect of the aged muscle ECM on MuSC lineage specification. Quantification of ECM topology and muscle mechanical properties reveals decreased collagen tortuosity and muscle stiffening with increasing age. Age‐related ECM alterations directly disrupt MuSC responses, and MuSCs seeded ex vivo onto decellularized ECM constructs derived from aged muscle display increased expression of fibrogenic markers and decreased myogenicity, compared to MuSCs seeded onto young ECM. This fibrogenic conversion is recapitulated in vitro when MuSCs are seeded directly onto matrices elaborated by aged fibroblasts. When compared to young fibroblasts, fibroblasts isolated from aged muscle display increased nuclear levels of the mechanosensors, Yes‐associated protein (YAP)/transcriptional coactivator with PDZ‐binding motif (TAZ), consistent with exposure to a stiff microenvironment in vivo. Accordingly, preconditioning of young fibroblasts by seeding them onto a substrate engineered to mimic the stiffness of aged muscle increases YAP/TAZ nuclear translocation and promotes secretion of a matrix that favors MuSC fibrogenesis. The findings here suggest that an age‐related increase in muscle stiffness drives YAP/TAZ‐mediated pathogenic expression of matricellular proteins by fibroblasts, ultimately disrupting MuSC fate.
Collapse
Affiliation(s)
- Kristen M. Stearns-Reider
- Department of Physical Medicine and Rehabilitation; University of Pittsburgh; Kaufmann Medical Building, Suite 201, 3471 Fifth Avenue Pittsburgh PA 15213 USA
- McGowan Institute for Regenerative Medicine; University of Pittsburgh; 450 Technology Drive, Suite 300 Pittsburgh PA 15219 USA
| | - Antonio D'Amore
- Department of Surgery; University of Pittsburgh; 450 Technology Drive, Suite 300 Pittsburgh PA 15219 USA
| | - Kevin Beezhold
- Department of Environmental and Occupational Health; University of Pittsburgh; 100 Technology Drive, Suite 328 Pittsburgh PA 15219 USA
| | - Benjamin Rothrauff
- Center for Cellular and Molecular Engineering; Department of Orthopaedic Surgery; University of Pittsburgh; 450 Technology Drive, Bridgeside Point II, Suite 221 Pittsburgh PA 15219 USA
| | - Loredana Cavalli
- Department of Physical Medicine and Rehabilitation; University of Pittsburgh; Kaufmann Medical Building, Suite 201, 3471 Fifth Avenue Pittsburgh PA 15213 USA
| | - William R. Wagner
- McGowan Institute for Regenerative Medicine; University of Pittsburgh; 450 Technology Drive, Suite 300 Pittsburgh PA 15219 USA
- Department of Surgery; University of Pittsburgh; 450 Technology Drive, Suite 300 Pittsburgh PA 15219 USA
- Center for Vascular Remodeling and Regeneration; Center for Bioengineering (CNBIO); University of Pittsburgh; 300 Technology Drive, Suite 300 Pittsburgh PA 15219 USA
| | - David A. Vorp
- McGowan Institute for Regenerative Medicine; University of Pittsburgh; 450 Technology Drive, Suite 300 Pittsburgh PA 15219 USA
- Department of Surgery; University of Pittsburgh; 450 Technology Drive, Suite 300 Pittsburgh PA 15219 USA
- Center for Vascular Remodeling and Regeneration; Center for Bioengineering (CNBIO); University of Pittsburgh; 300 Technology Drive, Suite 300 Pittsburgh PA 15219 USA
- Department of Bioengineering; University of Pittsburgh; 213 Center for Bioengineering, 300 Technology Drive Pittsburgh PA 15219 USA
| | - Alkiviadis Tsamis
- Department of Engineering; University of Leicester; 127 Michael Atiyah Building, University Road Leicester LE1 7RH UK
| | - Sunita Shinde
- Department of Physical Medicine and Rehabilitation; University of Pittsburgh; Kaufmann Medical Building, Suite 201, 3471 Fifth Avenue Pittsburgh PA 15213 USA
| | - Changqing Zhang
- Department of Physical Medicine and Rehabilitation; University of Pittsburgh; Kaufmann Medical Building, Suite 201, 3471 Fifth Avenue Pittsburgh PA 15213 USA
| | - Aaron Barchowsky
- Department of Environmental and Occupational Health; University of Pittsburgh; 100 Technology Drive, Suite 328 Pittsburgh PA 15219 USA
| | - Thomas A. Rando
- Glenn Center for the Biology of Aging and Department of Neurology and Neurological Sciences; Stanford University School of Medicine; Stanford CA 94305 USA
- RR&D Center; VA Palo Alto Health Care System; Palo Alto CA 94304 USA
| | - Rocky S. Tuan
- McGowan Institute for Regenerative Medicine; University of Pittsburgh; 450 Technology Drive, Suite 300 Pittsburgh PA 15219 USA
- Center for Cellular and Molecular Engineering; Department of Orthopaedic Surgery; University of Pittsburgh; 450 Technology Drive, Bridgeside Point II, Suite 221 Pittsburgh PA 15219 USA
| | - Fabrisia Ambrosio
- Department of Physical Medicine and Rehabilitation; University of Pittsburgh; Kaufmann Medical Building, Suite 201, 3471 Fifth Avenue Pittsburgh PA 15213 USA
- McGowan Institute for Regenerative Medicine; University of Pittsburgh; 450 Technology Drive, Suite 300 Pittsburgh PA 15219 USA
- Department of Bioengineering; University of Pittsburgh; 213 Center for Bioengineering, 300 Technology Drive Pittsburgh PA 15219 USA
| |
Collapse
|
14
|
Synergetic effect of topological cue and periodic mechanical tension-stress on osteogenic differentiation of rat bone mesenchymal stem cells. Colloids Surf B Biointerfaces 2017; 154:1-9. [PMID: 28268191 DOI: 10.1016/j.colsurfb.2017.02.035] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 02/01/2017] [Accepted: 02/27/2017] [Indexed: 02/05/2023]
Abstract
Mesenchymal stem cells (MSCs) are able to self-renew and differentiate into tissues of mesenchymal origin, making them to be significant for cell-based therapies, such as metabolic bone diseases and bone repair. Regulating the differentiation of MSCs is significant for bone regeneration. Electrospun fibers mimicking natural extracellular matrix (ECM), is an effective artificial ECM to regulate the behaviors and fates of MSCs. The aligned electrospun fibers can modulate polar cell pattern of bone mesenchymal stem cells, which leads to more obvious osteogenic differentiation. Apart from the topographic effect of electrospun fibers, mechanical cues can also intervene the cell behaviors. In this study, the osteogenic differentiation of rat bone mesenchymal stem cells was evaluated, which were cultured on aligned/random electrospun fiber mats materials under mechanical tension intervention. Scanning electron microscope and immune-fluorescent staining were used to directly observe the polarity changing of cellular morphology and cytoskeleton. The results proved that aligned electrospun fibers could be more conducive to promote osteogenic differentiation of rat bone mesenchymal stem cells and this promotion of osteogenic differentiation was enhanced by tension intervention. These results were correlated to the quantitative real-time PCR assay. In general, culturing rat bone mesenchymal stem cells on electrospun fibers under the intervention of mechanical tension is an effective way to mimic a more real cellular microenvironment.
Collapse
|
15
|
Niemiro GM, Parel J, Beals J, van Vliet S, Paluska SA, Moore DR, Burd NA, De Lisio M. Kinetics of circulating progenitor cell mobilization during submaximal exercise. J Appl Physiol (1985) 2017; 122:675-682. [DOI: 10.1152/japplphysiol.00936.2016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 12/23/2016] [Accepted: 01/08/2017] [Indexed: 12/28/2022] Open
Abstract
Circulating progenitor cells (CPCs) are a heterogeneous population of stem/progenitor cells in peripheral blood that includes hematopoietic stem and progenitor cells (HSPCs and HSCs), endothelial progenitor cells (EPCs), and mesenchymal stem cells (MSCs) that are involved in tissue repair and adaptation. CPC mobilization during exercise remains uncharacterized in young adults. The purpose of this study was to investigate the kinetics of CPC mobilization during and after submaximal treadmill running and their relationship to mobilization factors. Seven men [age = 25.3 ± 2.4 yr, body mass index = 23.5 ± 1.0 kg/m2, peak O2uptake (V̇o2peak) = 60.9 ± 2.74 ml·kg−1·min−1] ran on a treadmill for 60 min at 70% V̇o2peak. Blood sampling occurred before (Pre), during [20 min (20e), 40 min (40e), 60 min (60e)], and after exercise [15 min (15p), 60 min (60p), 120 min (120p)] for quantification of CPCs (CD34+), HSPCs (CD34+/CD45low), HSCs (CD34+/CD45low/CD38−), CD34+MSCs (CD45−/CD34+/CD31−/CD105+), CD34−MSCs (CD45−/CD34−/CD31−/CD105+), and EPCs (CD45−/CD34+/CD31+) via flow cytometry. CPC concentration increased compared with Pre at 20e and 40e (2.7- and 2.4-fold, respectively, P < 0.05). HSPCs and HSCs increased at 20e compared with 60p (2.7- and 2.8-fold, respectively, P < 0.05), whereas EPCs and both MSC populations did not change. CXC chemokine ligand (CXCL) 12 (1.5-fold; P < 0.05) and stem cell factor (1.3-fold; P < 0.05) were increased at 40e and remained elevated postexercise. The peak increase in CPCs was positively correlated to concentration of endothelial cells during exercise with no relationship to CXCL12 and SCF. Our data show the kinetics of progenitor cell mobilization during exercise that could provide insight into cellular mediators of exercise-induced adaptations, and have implication for the use of exercise as an adjuvant therapy for CPC collection in hematopoietic stem cell transplant.NEW & NOTEWORTHY Using a comprehensive evaluation of circulating progenitor cells (CPCs), we show that CPC mobilization during exercise is related to tissue damage, and not plasma concentrations of CXC chemokine ligand 12 and stem cell factor. These data have implications for the use of exercise interventions as adjuvant therapy for CPC mobilization in the context of hematopoietic stem cell transplant and also support the role of mobilized progenitor cells as cellular mediators of systemic adaptations to exercise.
Collapse
Affiliation(s)
- Grace M. Niemiro
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Champaign, Illinois
| | - Justin Parel
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Champaign, Illinois
| | - Joseph Beals
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Champaign, Illinois
| | - Stephan van Vliet
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Champaign, Illinois
| | - Scott A. Paluska
- Department of Family Medicine, University of Illinois at Urbana-Champaign, Champaign, Illinois
| | - Daniel R. Moore
- Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, Ontario, Canada; and
| | - Nicholas A. Burd
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Champaign, Illinois
| | - Michael De Lisio
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Champaign, Illinois
- School of Human Kinetics, Brain and Mind Institute, Centre for Neuromuscular Disease, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
16
|
Strömberg A, Rullman E, Jansson E, Gustafsson T. Exercise-induced upregulation of endothelial adhesion molecules in human skeletal muscle and number of circulating cells with remodeling properties. J Appl Physiol (1985) 2017; 122:1145-1154. [PMID: 28183821 DOI: 10.1152/japplphysiol.00956.2016] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 01/23/2017] [Accepted: 02/01/2017] [Indexed: 11/22/2022] Open
Abstract
Multipotent cells have received great interest because of their potential capacity to repair and remodel peripheral tissues. We examined the effect of an acute exercise bout on the number of circulating cells with known remodeling properties and the level of factors in plasma and skeletal muscle tissue with potential to recruit these cells. Twenty healthy male subjects performed a 60-min cycling exercise. Blood samples for flow cytometry were drawn from 10 subjects (group 1) before and up to 2 h after exercise, and absolute cell counts of the classical (CD14++CD16-), intermediate (CD14++CD16+), and nonclassical (CD14+CD16++) monocyte (MO) subpopulations and of CD45dimCD34+VEGFR2+ endothelial progenitor cells (EPCs) were measured by bead-based determination. Plasma samples and vastus lateralis muscle biopsies were obtained from the other 10 subjects (group 2). In group 1, all MO subsets were increased directly after exercise, with CD14+CD16++ MOs showing the greatest fold increase. After 2 h, only CD14++CD16- MOs were increased compared with resting levels. The number of EPCs showed a trend toward increasing with exercise (P = 0.08). In group 2, the mRNA levels of the endothelial adhesion molecules ICAM-1, VCAM-1, and E-selectin increased in the skeletal muscle tissue. VEGF-A increased in exercised skeletal muscle and stimulated the expression of VCAM-1 and E-selectin in human umbilical vein endothelial cells. In conclusion, exercise increases MO subsets with different temporal patterns and enhances the capacity of skeletal muscle tissue to recruit circulating cells as shown by increased expression of endothelial adhesion molecules.NEW & NOTEWORTHY In the present study we showed for the first time that the adhesion molecules ICAM-1, VCAM-1, and E-selectin, known to be able to recruit circulating cells to the peripheral tissue, increased in exercised human skeletal muscle concurrently with increased circulating levels of cells shown to have importance for skeletal muscle remodeling. These findings support the concept of cell recruitment from the circulation playing a role in skeletal muscle adaptation to exercise.
Collapse
Affiliation(s)
- Anna Strömberg
- Division of Clinical Physiology, Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Eric Rullman
- Division of Clinical Physiology, Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Eva Jansson
- Division of Clinical Physiology, Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Thomas Gustafsson
- Division of Clinical Physiology, Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| |
Collapse
|
17
|
Lai S, Panarese A, Lawrence R, Boninger ML, Micera S, Ambrosio F. A Murine Model of Robotic Training to Evaluate Skeletal Muscle Recovery after Injury. Med Sci Sports Exerc 2016; 49:840-847. [PMID: 27875498 DOI: 10.1249/mss.0000000000001160] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE In vivo studies have suggested that motor exercise can improve muscle regeneration after injury. Nevertheless, preclinical investigations still lack reliable tools to monitor motor performance over time and to deliver optimal training protocols to maximize force recovery. Here, we evaluated the utility of a murine robotic platform (i) to detect early impairment and longitudinal recovery after acute skeletal muscle injury and (ii) to administer varying intensity training protocols to enhance forelimb motor performance. METHODS A custom-designed robotic platform was used to train mice to perform a forelimb retraction task. After an acute injury to bilateral biceps brachii muscles, animals performed a daily training protocol in the platform at high (HL) or low (LL) loading levels over the course of 3 wk. Control animals were not trained (NT). Motor performance was assessed by quantifying force, time, submovement count, and number of movement attempts to accomplish the task. Myofiber number and cross-sectional area at the injury site were quantified histologically. RESULTS Two days after injury, significant differences in the time, submovement count, number of movement attempts, and exerted force were observed in all mice, as compared with baseline values. Interestingly, the recovery time of muscle force production differed significantly between intervention groups, with HL group showing a significantly accelerated recovery. Three weeks after injury, all groups showed motor performance comparable with baseline values. Accordingly, there were no differences in the number of myofibers or average cross-sectional area among groups after 3 wk. CONCLUSION Our findings demonstrate the utility of our custom-designed robotic device for the quantitative assessment of skeletal muscle function in preclinical murine studies. Moreover, we demonstrate that this device may be used to apply varying levels of resistance longitudinally as a means manipulate physiological muscle responses.
Collapse
Affiliation(s)
- Stefano Lai
- 1Scuola Superiore Sant'Anna, Translational Neural Engineering Area, The BioRobotics Institute, Pisa, ITALY; 2Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA; 3McGowan Institute for Regenerative Medicine, University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, PA; 4Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA; 5Department of Rehabilitation Science and Technology, University of Pittsburgh, Pittsburgh, PA; and 6Ecole Polytechnique Federale de Lausanne (EPFL), Bertarelli Foundation Chair in Translational NeuroEngineering Laboratory, Center for Neuroprosthetics and Institute of Bioengineering, Lausanne, SWITZERLAND
| | | | | | | | | | | |
Collapse
|
18
|
Abstract
Rehabilitation and regenerative medicine therapies has shown improved outcomes for tissue regeneration. Regenerative rehabilitation guides protocols regarding when to start therapy, types of stimuli administered, and graded exercise programs, taking into account biological factors and technologies designed to optimize healing potential. Although there are currently no evidence-based guidelines for rehabilitation, fundamental physical therapy principles likely apply. Immobilization tends to have deleterious effects on musculoskeletal tissues; mechanical loading promotes tissue healing and regeneration. Common physical therapy interventions may provide beneficial effects after the application of regenerative therapies. Research is needed to determine optimal rehabilitation protocols to enhance tissue healing and regeneration.
Collapse
Affiliation(s)
- Penny L Head
- Department of Physical Therapy, University of Tennessee Health Science Center, 930 Madison Avenue, Room 604, Memphis, TN 38163, USA.
| |
Collapse
|
19
|
Badylak SF, Dziki JL, Sicari BM, Ambrosio F, Boninger ML. Mechanisms by which acellular biologic scaffolds promote functional skeletal muscle restoration. Biomaterials 2016; 103:128-136. [PMID: 27376561 DOI: 10.1016/j.biomaterials.2016.06.047] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 06/15/2016] [Accepted: 06/20/2016] [Indexed: 12/31/2022]
|
20
|
Niemiro GM, Raine LB, Khan NA, Emmons R, Little J, Kramer AF, Hillman CH, De Lisio M. Circulating progenitor cells are positively associated with cognitive function among overweight/obese children. Brain Behav Immun 2016; 57:47-52. [PMID: 27132057 PMCID: PMC7404617 DOI: 10.1016/j.bbi.2016.03.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 03/11/2016] [Accepted: 03/23/2016] [Indexed: 12/18/2022] Open
Abstract
Recent evidence has indicated that overweight/obese children may experience cognitive and immune dysfunction, but the underlying mechanisms responsible for the association between overweight/obesity, immune dysfunction, and cognition have yet to be established. The present study aimed to identify a novel link between obesity-induced immune system dysregulation and cognition in preadolescent children. A total of 27 male children (age: 8-10years) were recruited and separated by body mass index (BMI) into healthy weight (HW: 5th-84.9th percentile, n=16) and overweight/obese (OW: ⩾85th percentile, n=11) groups. Adiposity was assessed using dual energy X-ray absorptiometry (DXA), and aspects of executive function were assessed using the Woodcock-Johnson III Tests of Cognitive Abilities. Monocyte populations (CD14(+)CD16(-), CD14(+)CD16(+)) with and without expression of chemokine receptor type 2 (CCR2), and circulating progenitor cells (CPCs: CD34(+)CD45(dim)), in peripheral blood were quantified by flow cytometry. CPCs were isolated by flow sorting and cultured for 24h for collection of conditioned media (CM) that was applied to SH-SY5Y neuroblastomas to examine the paracrine effects of CPCs on neurogenesis. OW had significantly higher quantities of both populations of monocytes (CD14(+)CD16(-): 57% increase; CD14(+)CD16(+): 95% increase, both p<0.01), monocytes expressing CCR2 (CD14(+)CD16(-)CCR2(+): 66% increase; CD14(+)CD16(+)CCR2(+): 168% increase, both p<0.01), and CPCs (47% increase, p<0.05) than HW. CPCs were positively correlated with abdominal adiposity in OW, and negatively correlated in HW with a significant difference between correlations (p<0.05). CPC content was positively correlated with executive processes in OW, and negatively correlated in HW with a significant difference in the strength of the correlations between groups (p<0.05 for correlation between OW and HW). Finally, CPC-CM from OW trended to increase neuroblast viability in vitro relative to HW (1.79 fold, p=0.07). These novel findings indicate that increased content of CPCs among OW children may play a role in preventing decrements in cognitive function via paracrine mechanisms.
Collapse
Affiliation(s)
- Grace M. Niemiro
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, United States
| | - Lauren B. Raine
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, United States
| | - Naiman A. Khan
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, United States
| | - Russell Emmons
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, United States
| | - Jonathan Little
- School of Health and Exercise Sciences, University of British Columbia Okanagan, Canada
| | - Arthur F. Kramer
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, United States
| | - Charles H. Hillman
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, United States,Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, United States
| | - Michael De Lisio
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, United States.
| |
Collapse
|
21
|
Kowalski K, Archacki R, Archacka K, Stremińska W, Paciorek A, Gołąbek M, Ciemerych MA, Brzoska E. Stromal derived factor-1 and granulocyte-colony stimulating factor treatment improves regeneration of Pax7-/- mice skeletal muscles. J Cachexia Sarcopenia Muscle 2016; 7:483-96. [PMID: 27239402 PMCID: PMC4863826 DOI: 10.1002/jcsm.12092] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 10/03/2015] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND The skeletal muscle has the ability to regenerate after injury. This process is mediated mainly by the muscle specific stem cells, that is, satellite cells. In case of extensive damage or under pathological conditions, such as muscular dystrophy, the process of muscle reconstruction does not occur properly. The aim of our study was to test whether mobilized stem cells, other than satellite cells, could participate in skeletal muscle reconstruction. METHODS Experiments were performed on wild-type mice and mice lacking the functional Pax7 gene, that is, characterized by the very limited satellite cell population. Gastrocnemius mice muscles were injured by cardiotoxin injection, and then the animals were treated by stromal derived factor-1 (Sdf-1) with or without granulocyte-colony stimulating factor (G-CSF) for 4 days. The muscles were subjected to thorough assessment of the tissue regeneration process using histological and in vitro methods, as well as evaluation of myogenic factors' expression at the transcript and protein levels. RESULTS Stromal derived factor-1 alone and Sdf-1 in combination with G-CSF significantly improved the regeneration of Pax7-/- skeletal muscles. The Sdf-1 and G-CSF treatment caused an increase in the number of mononucleated cells associated with muscle fibres. Further analysis showed that Sdf-1 and G-CSF treatment led to the rise in the number of CD34+ and Cxcr4+ cells and expression of Cxcr7. CONCLUSIONS Stromal derived factor-1 and G-CSF stimulated regeneration of the skeletal muscles deficient in satellite cells. We suggest that mobilized CD34+, Cxcr4+, and Cxcr7+ cells can efficiently participate in the skeletal muscle reconstruction and compensate for the lack of satellite cells.
Collapse
Affiliation(s)
- Kamil Kowalski
- Department of Cytology, Faculty of Biology University of Warsaw Warsaw Poland
| | - Rafał Archacki
- Laboratory of Systems Biology, Faculty of Biology University of Warsaw Warsaw Poland
| | - Karolina Archacka
- Department of Cytology, Faculty of Biology University of Warsaw Warsaw Poland
| | | | - Anna Paciorek
- Department of Cytology, Faculty of Biology University of Warsaw Warsaw Poland
| | - Magdalena Gołąbek
- Department of Cytology, Faculty of Biology University of Warsaw Warsaw Poland
| | - Maria A Ciemerych
- Department of Cytology, Faculty of Biology University of Warsaw Warsaw Poland
| | - Edyta Brzoska
- Department of Cytology, Faculty of Biology University of Warsaw Warsaw Poland
| |
Collapse
|
22
|
Helal MAM, Shaheen NEM, Abu Zahra FA. Immunomodulatory capacity of the local mesenchymal stem cells transplantation after severe skeletal muscle injury in female rats. Immunopharmacol Immunotoxicol 2016; 38:414-422. [PMID: 27560658 DOI: 10.1080/08923973.2016.1222617] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
CONTEXT Cell therapy technique with stem cells is a very attractive strategy for the treatment of muscle disorders. OBJECTIVE The objective of this study was to investigate the mechanism of local transplantation of mesenchymal stem cells (MSCs) which could contribute to skeletal muscle healing. MATERIALS AND METHODS Female rats were divided into three equal groups as the following: group 1, the negative control group (untreated group), group 2, sham-treated group, rats with muscle injuries involving volumetric muscle loss (VML) of adductor brevis muscle and injected locally with phosphate-buffered saline (PBS) 0.5 ml without stem cells after 7 d of muscle injury, group 3, treated group, rats with VML and injected locally (intramuscular) with 1.5 × 106 bone marrow MSCs suspended in PBS 0.5 ml (1) after 7 d of muscle tissue injury. All animals were sacrificed after 4 weeks of stem cell transplantation. RESULTS In vitro culture the morphology of MSCs reached confluence and appeared as long spindle in shape on 9-14 d. Most of the cells did not express the hematopoietic cell marker, CD34 and CD45 but expressed MSCs marker CD44, CD90 and CD105. The remarkable increase of proliferating cell nuclear antigen positive nucleus was recorded in MSCs group as compared to PBS group. After 28 d of injection, administration of only PBS into the site of muscle injury caused up-regulation in the levels of interleukins IL-1β, IL-6, tumor necrosis factor alpha (TNF-α), transforming growth factor beta (TGF-β1), interferon alpha (IFN-α) and down-regulate the level of IL-10 in muscular tissue comparing to the untreated control. Bone marrow MSCs + PBS injected at the site of muscle injury significantly down-regulate the inflammatory cytokines levels IL-1β and IL-6 and TNF-α, TGF-β1 and IFN-α and up-regulate the level of IL-10. Collagen concentrations in the injured skeletal muscle estimated by enzyme-linked immuno sorbent assay and stained with Masson trichrome stain were increased with PBS group and decreased after transplantation of bone marrow MSCs in the site of injury. Muscle sections stained with H&E showed a higher number of centronucleated regenerating myofibers in the stem-cell-treated group than in the (PBS) and untreated control group. Microvasculature of skeletal muscle was decreased as demonstrated by immunostaining technique for CD34 in PBS group from untreated control. The MSCs group showed angiogenesis and marked increase of skeletal muscle microvasculature than PBS group. CONCLUSION MSCs can modify the local immunological responses and improve muscle regeneration by suppressing of inflammatory cytokines, activating of the anti-inflammatory cytokine, restoration of muscle fibers and angiogenesis. By means of increase in TGF-β production in response to muscle injury prevent the repair of injured fibers and increase connective tissue production (collagen fibers), thus propagating skeletal muscle weakness and fibrosis whereas MSCs + PBS injected at the site of muscle injury significantly down-regulate (TGF-β1) and hence the level of collagen (fibrosis or scar areas). MSCs are able to block the fibrotic signaling cascade by declining TGF-β1 and scar areas in the injured muscle.
Collapse
Affiliation(s)
- Mona A M Helal
- a Department of Zoology, Faculty of Women for Arts, Science & Education , Ain Shams University , Cairo , Egypt
| | - Noura E M Shaheen
- a Department of Zoology, Faculty of Women for Arts, Science & Education , Ain Shams University , Cairo , Egypt
| | - Fatma A Abu Zahra
- b Molecular Biology and Tissue Culture , Medical Research Center, Ain Shams University , Cairo , Egypt
| |
Collapse
|
23
|
Developmental Biology and Regenerative Medicine: Addressing the Vexing Problem of Persistent Muscle Atrophy in the Chronically Torn Human Rotator Cuff. Phys Ther 2016; 96:722-33. [PMID: 26847008 PMCID: PMC4858662 DOI: 10.2522/ptj.20150029] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 01/24/2016] [Indexed: 12/18/2022]
Abstract
Persistent muscle atrophy in the chronically torn rotator cuff is a significant obstacle for treatment and recovery. Large atrophic changes are predictive of poor surgical and nonsurgical outcomes and frequently fail to resolve even following functional restoration of loading and rehabilitation. New insights into the processes of muscle atrophy and recovery gained through studies in developmental biology combined with the novel tools and strategies emerging in regenerative medicine provide new avenues to combat the vexing problem of muscle atrophy in the rotator cuff. Moving these treatment strategies forward likely will involve the combination of surgery, biologic/cellular agents, and physical interventions, as increasing experimental evidence points to the beneficial interaction between biologic therapies and physiologic stresses. Thus, the physical therapy profession is poised to play a significant role in defining the success of these combinatorial therapies. This perspective article will provide an overview of the developmental biology and regenerative medicine strategies currently under investigation to combat muscle atrophy and how they may integrate into the current and future practice of physical therapy.
Collapse
|
24
|
Exercise as an Adjuvant Therapy for Hematopoietic Stem Cell Mobilization. Stem Cells Int 2016; 2016:7131359. [PMID: 27123008 PMCID: PMC4830735 DOI: 10.1155/2016/7131359] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 02/03/2016] [Accepted: 02/07/2016] [Indexed: 12/13/2022] Open
Abstract
Hematopoietic stem cell transplant (HSCT) using mobilized peripheral blood hematopoietic stem cells (HSPCs) is the only curative strategy for many patients suffering from hematological malignancies. HSPC collection protocols rely on pharmacological agents to mobilize HSPCs to peripheral blood. Limitations including variable donor responses and long dosing protocols merit further investigations into adjuvant therapies to enhance the efficiency of HSPCs collection. Exercise, a safe and feasible intervention in patients undergoing HSCT, has been previously shown to robustly stimulate HSPC mobilization from the bone marrow. Exercise-induced HSPC mobilization is transient limiting its current clinical potential. Thus, a deeper investigation of the mechanisms responsible for exercise-induced HSPC mobilization and the factors responsible for removal of HSPCs from circulation following exercise is warranted. The present review will describe current research on exercise and HSPC mobilization, outline the potential mechanisms responsible for exercise-induced HSPC mobilization, and highlight potential sites for HSPC homing following exercise. We also outline current barriers to the implementation of exercise as an adjuvant therapy for HSPC mobilization and suggest potential strategies to overcome these barriers.
Collapse
|
25
|
Emmons R, Niemiro GM, Owolabi O, De Lisio M. Acute exercise mobilizes hematopoietic stem and progenitor cells and alters the mesenchymal stromal cell secretome. J Appl Physiol (1985) 2016; 120:624-32. [DOI: 10.1152/japplphysiol.00925.2015] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 01/02/2016] [Indexed: 12/20/2022] Open
Abstract
Transplantation of hematopoietic stem and progenitor cells (HSPC), collected from peripheral blood, is the primary treatment for many hematological malignancies; however, variable collection efficacy with current protocols merits further examination into factors responsible for HSPC mobilization. HSPCs primarily reside within the bone marrow and are regulated by mesenchymal stromal cells (MSC). Exercise potently and transiently mobilizes HSPCs from the bone marrow into peripheral circulation. Thus the purpose of the present study was to evaluate potential factors in the bone marrow responsible for HSPC mobilization, investigate potential sites of HSPC homing, and assess changes in bone marrow cell populations following exercise. An acute exercise bout increased circulating HSPCs at 15 min (88%, P < 0.001) that returned to baseline at 60 min. Gene expression for HSPC homing factors (CXCL12, vascular endothelial growth factor-a, and angiopoietin-1) were increased at 15 min in skeletal muscle and HSPC content was increased in the spleen 48 h postexercise (45%, P < 0.01). Acute exercise did not alter HSPCs or MSCs quantity in the bone marrow; however, proliferation of HSPCs (40%, P < 0.001), multipotent progenitors (40%, P < 0.001), short-term hematopoietic stem cells (61%, P < 0.001), long-term hematopoietic stem cells (55%, P = 0.002), and MSCs (20%, P = 0.01) increased postexercise. Acute exercise increased the content of the mobilization agent granulocyte-colony stimulating factor, as well as stem cell factor, interleukin-3, and thrombopoeitin in conditioned media collected from bone marrow stromal cells 15 min postexercise. These findings suggest that the MSC secretome is responsible for HSPC mobilization and proliferation; concurrently, HSPCs are homing to extramedullary sites following exercise.
Collapse
Affiliation(s)
- Russell Emmons
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Grace M. Niemiro
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Olatomide Owolabi
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Michael De Lisio
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, Illinois
| |
Collapse
|
26
|
|
27
|
Napolitano M, Gerardi C, Di Lucia A, Accardo PA, Rizzuto L, Ferraro M, Siragusa S, Buscemi F. Hematopoietic peripheral circulating blood stem cells as an independent marker of good transfusion management in patients with β-thalassemia: results from a preliminary study. Transfusion 2016; 56:827-30. [PMID: 26801519 DOI: 10.1111/trf.13452] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 11/08/2015] [Accepted: 11/13/2015] [Indexed: 12/14/2022]
Abstract
BACKGROUND Beyond hemoglobin (Hb) levels and performance status, further surrogate markers of appropriate transfusion management should improve the quality of thalassemia care. We investigated the levels of peripheral circulating CD34+ stem cells as an independent marker of appropriate hematopoietic balance in patients with thalassemia. STUDY DESIGN AND METHODS Peripheral circulating CD34+ stem cells, colony-forming unitgranulocyte, erythrocyte, macrophage, magakaryocyte (CF-GEMM), colony-forming unitgranulocyte/macrophage (CFU-GM), and erythroidburst-forming units (BFU-E) were assayed, according to standard procedures. Patients with thalassemia major (TM) and thalassemia intermedia (TI) were tested and compared to healthy controls. Demographic and clinical data were recorded. RESULTS Overall, 56 patients with TM (median age, 35 years; range, 13-52 years) and 13 with TI (median age, 44 years; range, 27-67 years) were evaluated. Annual red blood cell (RBC) transfusion requirements ranged from 10 to 65 units in all patients except four nontransfused cases. A significant increase in peripheral circulating stem cells was observed in patients, in comparison with healthy controls. Nontransfused patients showed the mean highest levels of stem cells (CD34, 32.5 ± 14.8/μL; BFU-E, 41.3 ± 22.8/mL; CFU-GM, 19.6 ± 5.6/mL; CFU-GEMM, 9.0 ± 6.1/mL). CD34+ cell count was 6.9 ± 4.5/μL in TM (p = 0.014) and 11.8 ± 14.8/μL (p = 0.051) in TI. Furthermore, only in patients with TI was a significant increase in CFU-GEMM (3.0 ± 4.8 vs. 0.75 ± 2.05/mL, p = 0.0001) observed. At multivariate analysis, peripheral circulating CD34+ stem cells did not correlate with age, sex, smoking habit, number of RBCs units transfused, Hb levels, iron chelation therapy, history of splenectomy, and hypothyroidism. CONCLUSION Circulating peripheral CD34 + stem cells are increased in β-thalassemia, in particular in nontransfused patients, compared to healthy controls.
Collapse
Affiliation(s)
| | - Calogera Gerardi
- Banca del Sangue da Cordone Ombelicale, UOC Medicina Trasfusionale, PO "Giovanni Paolo II,", Sciacca, Italy
| | - Anna Di Lucia
- Banca del Sangue da Cordone Ombelicale, UOC Medicina Trasfusionale, PO "Giovanni Paolo II,", Sciacca, Italy
| | | | - Luigi Rizzuto
- UOS Talassemia, UOC Medicina Trasfusionale, PO "Giovanni Paolo II,", Sciacca, Italy
| | - Maria Ferraro
- UOS Talassemia, UOC Medicina Trasfusionale, PO "Giovanni Paolo II,", Sciacca, Italy
| | - Sergio Siragusa
- UOC Ematologia con Trapianto, Università di Palermo, Palermo, Italy; and
| | - Filippo Buscemi
- UOS Talassemia, UOC Medicina Trasfusionale, PO "Giovanni Paolo II,", Sciacca, Italy
| |
Collapse
|
28
|
Andrade BM, Baldanza MR, Ribeiro KC, Porto A, Peçanha R, Fortes FSA, Zapata-Sudo G, Campos-de-Carvalho AC, Goldenberg RCS, Werneck-de-Castro JP. Bone marrow mesenchymal cells improve muscle function in a skeletal muscle re-injury model. PLoS One 2015; 10:e0127561. [PMID: 26039243 PMCID: PMC4454438 DOI: 10.1371/journal.pone.0127561] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 04/16/2015] [Indexed: 02/05/2023] Open
Abstract
Skeletal muscle injury is the most common problem in orthopedic and sports medicine, and severe injury leads to fibrosis and muscle dysfunction. Conventional treatment for successive muscle injury is currently controversial, although new therapies, like cell therapy, seem to be promise. We developed a model of successive injuries in rat to evaluate the therapeutic potential of bone marrow mesenchymal cells (BMMC) injected directly into the injured muscle. Functional and histological assays were performed 14 and 28 days after the injury protocol by isometric tension recording and picrosirius/Hematoxilin & Eosin staining, respectively. We also evaluated the presence and the fate of BMMC on treated muscles; and muscle fiber regeneration. BMMC treatment increased maximal skeletal muscle contraction 14 and 28 days after muscle injury compared to non-treated group (4.5 ± 1.7 vs 2.5 ± 0.98 N/cm2, p<0.05 and 8.4 ± 2.3 vs. 5.7 ± 1.3 N/cm2, p<0.05 respectively). Furthermore, BMMC treatment increased muscle fiber cross-sectional area and the presence of mature muscle fiber 28 days after muscle injury. However, there was no difference in collagen deposition between groups. Immunoassays for cytoskeleton markers of skeletal and smooth muscle cells revealed an apparent integration of the BMMC within the muscle. These data suggest that BMMC transplantation accelerates and improves muscle function recovery in our extensive muscle re-injury model.
Collapse
Affiliation(s)
- Bruno M. Andrade
- Laboratório de Biologia do Exercício, Instituto de Biofísica Carlos Chagas Filho e Escola de Educação Física e Desportos, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
- Instituto de Biofísica Carlos Chagas Filho, Centro de Ciências e Saúde, Universidade Federal do Rio de Janeiro, Bloco G, Ilha do Fundão, Rio de Janeiro, RJ, Brasil
| | - Marcelo R. Baldanza
- Laboratório de Biologia do Exercício, Instituto de Biofísica Carlos Chagas Filho e Escola de Educação Física e Desportos, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - Karla C. Ribeiro
- Instituto de Biofísica Carlos Chagas Filho, Centro de Ciências e Saúde, Universidade Federal do Rio de Janeiro, Bloco G, Ilha do Fundão, Rio de Janeiro, RJ, Brasil
| | - Anderson Porto
- Laboratório de Biologia do Exercício, Instituto de Biofísica Carlos Chagas Filho e Escola de Educação Física e Desportos, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - Ramon Peçanha
- Laboratório de Biologia do Exercício, Instituto de Biofísica Carlos Chagas Filho e Escola de Educação Física e Desportos, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| | - Fabio S. A. Fortes
- Instituto de Biofísica Carlos Chagas Filho, Centro de Ciências e Saúde, Universidade Federal do Rio de Janeiro, Bloco G, Ilha do Fundão, Rio de Janeiro, RJ, Brasil
| | - Gisele Zapata-Sudo
- Departamento de Farmacologia Básica e Clínica, Universidade Federal do Rio de Janeiro, Centro de Ciências e Saúde, Bloco J, Ilha do Fundão, Rio de Janeiro, Brasil
| | - Antonio C. Campos-de-Carvalho
- Instituto de Biofísica Carlos Chagas Filho, Centro de Ciências e Saúde, Universidade Federal do Rio de Janeiro, Bloco G, Ilha do Fundão, Rio de Janeiro, RJ, Brasil
| | - Regina C. S. Goldenberg
- Instituto de Biofísica Carlos Chagas Filho, Centro de Ciências e Saúde, Universidade Federal do Rio de Janeiro, Bloco G, Ilha do Fundão, Rio de Janeiro, RJ, Brasil
| | - João Pedro Werneck-de-Castro
- Laboratório de Biologia do Exercício, Instituto de Biofísica Carlos Chagas Filho e Escola de Educação Física e Desportos, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brasil
| |
Collapse
|
29
|
Mooren FC, Krüger K. Apoptotic lymphocytes induce progenitor cell mobilization after exercise. J Appl Physiol (1985) 2015; 119:135-9. [PMID: 26023229 DOI: 10.1152/japplphysiol.00287.2015] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 05/26/2015] [Indexed: 01/01/2023] Open
Abstract
There is evidence that apoptotic cells and their components have immunmodulatory properties and signaling function. The present study investigated first whether exercise-induced apoptosis and exercise-induced mobilization of progenitor cells are similarly affected by subjects' training status and, second, whether the appearance of dying cells in the circulation might mobilize progenitor cells. CD1 SWISS mice were subjected to a 10-wk endurance training using free wheel running or served as untrained controls. Mice of both groups performed an intensive exercise test after the training period at a velocity corresponding to 80% maximal oxygen uptake for 30 min. Cells from blood and bone marrow were analyzed, and apoptosis and number of progenitor cells determined via flow cytometry. In a second experiment, apoptotic cells were transferred into recipient mice, and mobilization of progenitor cells was analyzed while vital cells served as controls. In untrained animals, the exhaustive exercise was followed by an enhanced rate of annexin V positive CD3(+) cells in blood and bone marrow (P < 0.05), whereas no increase was found in trained mice. Similarly, exercise mobilized Sca-1(+)/c-kit(+) and Sca-1(+)/Flk(+) cells in untrained (P < 0.05) but not trained mice. Furthermore, application of apoptotic cells and their supernatant mobilized Sca-1(+)/c-kit(+) cells into the blood (P < 0.05), whereas Sca-1(+)/Flk(+) cells were not affected. The present study demonstrated that both lymphocyte apoptosis, as well as mobilization of progenitor cells are similarly related to training status. Furthermore, apoptotic cells seem to induce signals that effectively mobilize hematopoietic progenitor cells. The relevance of this effect for the adaptation to exercise stimuli remains to be shown.
Collapse
Affiliation(s)
- Frank C Mooren
- Department of Sports Medicine, Institute of Sports Sciences, Justus-Liebig-University, Giessen, Germany
| | - Karsten Krüger
- Department of Sports Medicine, Institute of Sports Sciences, Justus-Liebig-University, Giessen, Germany
| |
Collapse
|
30
|
Improvement of endurance of DMD animal model using natural polyphenols. BIOMED RESEARCH INTERNATIONAL 2015; 2015:680615. [PMID: 25861640 PMCID: PMC4377377 DOI: 10.1155/2015/680615] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 08/13/2014] [Accepted: 08/27/2014] [Indexed: 12/11/2022]
Abstract
Duchenne muscular dystrophy (DMD), the most common form of muscular dystrophy, is characterized by muscular wasting caused by dystrophin deficiency that ultimately ends in force reduction and premature death. In addition to primary genetic defect, several mechanisms contribute to DMD pathogenesis. Recently, antioxidant supplementation was shown to be effective in the treatment of multiple diseases including muscular dystrophy. Different mechanisms were hypothesized such as reduced hydroxyl radicals, nuclear factor-κB deactivation, and NO protection from inactivation. Following these promising evidences, we investigated the effect of the administration of a mix of dietary natural polyphenols (ProAbe) on dystrophic mdx mice in terms of muscular architecture and functionality. We observed a reduction of muscle fibrosis deposition and myofiber necrosis together with an amelioration of vascularization. More importantly, the recovery of the morphological features of dystrophic muscle leads to an improvement of the endurance of treated dystrophic mice. Our data confirmed that ProAbe-based diet may represent a strategy to coadjuvate the treatment of DMD.
Collapse
|
31
|
Sicari BM, Rubin JP, Dearth CL, Wolf MT, Ambrosio F, Boninger M, Turner NJ, Weber DJ, Simpson TW, Wyse A, Brown EHP, Dziki JL, Fisher LE, Brown S, Badylak SF. An acellular biologic scaffold promotes skeletal muscle formation in mice and humans with volumetric muscle loss. Sci Transl Med 2014; 6:234ra58. [PMID: 24786326 DOI: 10.1126/scitranslmed.3008085] [Citation(s) in RCA: 347] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Biologic scaffolds composed of naturally occurring extracellular matrix (ECM) can provide a microenvironmental niche that alters the default healing response toward a constructive and functional outcome. The present study showed similarities in the remodeling characteristics of xenogeneic ECM scaffolds when used as a surgical treatment for volumetric muscle loss in both a preclinical rodent model and five male patients. Porcine urinary bladder ECM scaffold implantation was associated with perivascular stem cell mobilization and accumulation within the site of injury, and de novo formation of skeletal muscle cells. The ECM-mediated constructive remodeling was associated with stimulus-responsive skeletal muscle in rodents and functional improvement in three of the five human patients.
Collapse
Affiliation(s)
- Brian M Sicari
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Zhang Y, Zhang H, Zhang G, Ka K, Huang W. Combining acellular nerve allografts with brain-derived neurotrophic factor transfected bone marrow mesenchymal stem cells restores sciatic nerve injury better than either intervention alone. Neural Regen Res 2014; 9:1814-9. [PMID: 25422643 PMCID: PMC4239771 DOI: 10.4103/1673-5374.143427] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/06/2014] [Indexed: 01/08/2023] Open
Abstract
In this study, we chemically extracted acellular nerve allografts from bilateral sciatic nerves, and repaired 10-mm sciatic nerve defects in rats using these grafts and brain-derived neurotrophic factor transfected bone marrow mesenchymal stem cells. Experiments were performed in three groups: the acellular nerve allograft bridging group, acellular nerve allograft + bone marrow mesenchymal stem cells group, and the acellular nerve allograft + brain-derived neurotrophic factor transfected bone marrow mesenchymal stem cells group. Results showed that at 8 weeks after bridging, sciatic functional index, triceps wet weight recovery rate, myelin thickness, and number of myelinated nerve fibers were significantly changed in the three groups. Variations were the largest in the acellular nerve allograft + brain-derived neurotrophic factor transfected bone marrow mesenchymal stem cells group compared with the other two groups. Experimental findings suggest that chemically extracted acellular nerve allograft combined nerve factor and mesenchymal stem cells can promote the restoration of sciatic nerve defects. The repair effect seen is better than the single application of acellular nerve allograft or acellular nerve allograft combined mesenchymal stem cell transplantation.
Collapse
Affiliation(s)
- Yanru Zhang
- School of International Education, Zhengzhou University, Zhengzhou, Henan Province, China ; Institute of Clinical Anatomy, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Hui Zhang
- Department of Orthopedic Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Gechen Zhang
- Department of Orthopedic Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Ka Ka
- School of International Education, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Wenhua Huang
- Institute of Clinical Anatomy, Southern Medical University, Guangzhou, Guangdong Province, China
| |
Collapse
|
33
|
Ambrosio F, Boninger ML, Brubaker CE, Delitto A, Wagner WR, Shields RK, Wolf SL, Rando TA. Guest editorial: emergent themes from second annual symposium on regenerative rehabilitation, Pittsburgh, Pennsylvania. ACTA ACUST UNITED AC 2014; 50:vii-xiv. [PMID: 23881770 DOI: 10.1682/jrrd.2013.04.0081] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
34
|
Corona BT, Rathbone CR. Accelerated functional recovery after skeletal muscle ischemia-reperfusion injury using freshly isolated bone marrow cells. J Surg Res 2014; 188:100-9. [PMID: 24485153 DOI: 10.1016/j.jss.2013.12.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Revised: 12/18/2013] [Accepted: 12/30/2013] [Indexed: 12/13/2022]
Abstract
BACKGROUND Relatively little information exists regarding the usefulness of bone marrow-derived cells for skeletal muscle ischemia-reperfusion injury (I/R), especially when compared with I/R that occurs in other tissues. The objectives of this study were to evaluate the ability of freshly isolated bone marrow cells to home to injured skeletal muscle and to determine their effects on muscle regeneration. MATERIALS AND METHODS Freshly isolated lineage-depleted bone marrow cells (Lin(-) BMCs) were injected intravenously 2 d after I/R. Bioluminescent imaging was used to evaluate cell localization for up to 28 d after injury. Muscle function, the percentage of fibers with centrally located nuclei, and the capillary-to-fiber ratio were evaluated 14 d after delivery of either saline (Saline) or saline containing Lin(-) BMCs (Lin(-) BMCs). RESULTS Bioluminescence was higher in the injured leg than the contralateral control leg for up to 7 d after injection (P < 0.05) suggestive of cell homing to the injured skeletal muscle. Fourteen days after injury, there was a significant improvement in maximal tetanic torque (40% versus 22% deficit; P < 0.05), a faster rate of force production (+dP/dt) (123.6 versus 94.5 Nmm/S; P < 0.05), and a reduction in the percentage of fibers containing centrally located nuclei (40 versus 17%; P < 0.05), but no change in the capillary-to-fiber ratio in the Lin(-) BMC as compared with the Saline group. CONCLUSIONS The homing of freshly isolated BMCs to injured skeletal muscle after I/R is associated with an increase in functional outcomes.
Collapse
Affiliation(s)
- Benjamin T Corona
- Department of Extremity Trauma and Regenerative Medicine, United States Army Institute of Surgical Research, Fort Sam Houston, Texas
| | - Christopher R Rathbone
- Department of Extremity Trauma and Regenerative Medicine, United States Army Institute of Surgical Research, Fort Sam Houston, Texas.
| |
Collapse
|
35
|
Strömberg A, Jansson M, Fischer H, Rullman E, Hägglund H, Gustafsson T. Bone marrow derived cells in adult skeletal muscle tissue in humans. Skelet Muscle 2013; 3:12. [PMID: 23680018 PMCID: PMC3668176 DOI: 10.1186/2044-5040-3-12] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Accepted: 04/10/2013] [Indexed: 11/12/2022] Open
Abstract
Background During the past decade, several animal studies have demonstrated that in addition to local cells, cells from the bone marrow (BM) possess the ability to contribute to regeneration of injured skeletal muscle tissue. In addition, in mice, regular physical activity has been displayed to be a sufficient stimulus for BM-derived cell contribution to the muscle, indicating that this is part of the ongoing physiological remodeling of skeletal muscle. However, whether BM-derived cells participate in human skeletal muscle remodeling is not known. To this end, we analyzed the incorporation of BM-derived cells in healthy human skeletal muscle in women transplanted with male BM. Methods Skeletal muscle biopsies were obtained from the m. vastus lateralis of women transplanted with male donor hematopoietic stem cells 6 to 12 years earlier. Healthy women served as controls. Immunohistochemical staining for skeletal muscle fibers, satellite cells (SCs) or endothelial cells (ECs) combined with fluorescent in situ hybridization (FISH) of X and Y chromosomes was used to identify cells of BM origin within the biopsies. Three dimensional confocal imaging was performed to demonstrate colocalization of Y chromosome and DAPI within muscle fibers. To further investigate whether BM-derived cells incorporate into the SC niche, myoblasts were extracted from the biopsies from the transplanted women, cultured, and analyzed using XY FISH and immunocytochemistry. Results Three dimensional confocal imaging indisputably demonstrated colocalization of Y chromosome and DAPI within muscle fibers. Some Y chromosomes were found within centrally located nuclei. No Y chromosomes were detected in CD56+ SCs in the tissue sections nor in the myoblasts cultured from the extracted SCs. Y chromosome+ ECs were found in all sections from the transplanted subjects. No Y chromosomes were found in the skeletal muscle biopsies obtained from healthy control women. Conclusions We demonstrate that BM-derived cells contribute to skeletal muscle fibers and ECs. Our results support that BM contribution to skeletal muscle occurs via direct fusion to muscle fibers, and that the contributing cells derive from the hematopoietic lineage. Thus, the present findings encourage further studies of the importance of this process for the physiological adaptation occurring throughout life.
Collapse
Affiliation(s)
- Anna Strömberg
- Department of Laboratory Medicine, Division of Clinical Physiology, Karolinska Institutet, Karolinska University Hospital Huddinge, 141 86, Stockholm, Sweden.
| | | | | | | | | | | |
Collapse
|
36
|
Meregalli M, Farini A, Belicchi M, Torrente Y. CD133(+) Cells for the Treatment of Degenerative Diseases: Update and Perspectives. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 777:229-43. [PMID: 23161086 DOI: 10.1007/978-1-4614-5894-4_15] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/08/2022]
Abstract
Stem cells are used in cell therapy for degenerative disorders. The main advantage of stem cells is that they can replenish their numbers for long periods through cell division and produce a progeny that can differentiate into multiple cell lineages with specific functions. CD133 is a member of a novel family of cell surface glycoproteins. The expression of human CD133 (AC133 antigen) was originally described in the hematopoietic CD34(+) stem cells, but now it becomes more and more evident that CD133 is a marker of stem and progenitor cell populations originating from various tissues and organs. The main objective of this chapter is to describe the potential sources of CD133(+) stem cells that harbor the ability to engraft, proliferate, and differentiate into functional cells. The characterization of such CD133(+) stem cells unlocks new opportunities in the treatment of degenerative diseases such as Duchenne muscular dystrophy.
Collapse
Affiliation(s)
- Mirella Meregalli
- Stem Cell Laboratory, Dipartimento di Fisiopatologia Medico-Chirurgica e dei Trapianti, Università degli Studi di Milano, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, di Milano, Centro Dino Ferrari, via Francesco Sforza 35, 20122, Milan, Italy
| | | | | | | |
Collapse
|
37
|
Sicari BM, Zhang L, Londono R, Badylak SF. An assay to quantify chemotactic properties of degradation products from extracellular matrix. Methods Mol Biol 2013; 1202:103-10. [PMID: 24155230 DOI: 10.1007/7651_2013_37] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The endogenous chemotaxis of cells toward sites of tissue injury and/or biomaterial implantation is an important component of the host response. Implanted biomaterials capable of recruiting host stem/progenitor cells to a site of interest may obviate challenges associated with cell transplantation. An assay for the identification and quantification of chemotaxis induced by surgically placed biologic scaffolds composed of extracellular matrix is described herein.
Collapse
Affiliation(s)
- Brian M Sicari
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | | |
Collapse
|
38
|
Brzoska E, Kowalewska M, Markowska-Zagrajek A, Kowalski K, Archacka K, Zimowska M, Grabowska I, Czerwińska AM, Czarnecka-Góra M, Stremińska W, Jańczyk-Ilach K, Ciemerych MA. Sdf-1 (CXCL12) improves skeletal muscle regeneration via the mobilisation of Cxcr4 and CD34 expressing cells. Biol Cell 2012; 104:722-37. [PMID: 22978573 DOI: 10.1111/boc.201200022] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Accepted: 09/11/2012] [Indexed: 01/27/2023]
Abstract
BACKGROUND INFORMATION The regeneration of skeletal muscles involves satellite cells, which are muscle-specific precursor cells. In muscles, injured either mechanically or as a consequence of a disease, such as muscular dystrophy, local release of the growth factors and cytokines leads to satellite cells activation, proliferation and differentiation of the resulting myoblasts, followed by the formation of new myofibres. Various cell types, such as stem and progenitor cells, originating from other tissues different than the muscle, are also able to follow a myogenic program. Participation of these cells in the repair process depends on their precise mobilisation to the site of the injury. RESULTS In this study, we showed that stromal-derived factor-1 (Sdf-1) impacts on the mobilisation of CXC chemokine receptor (Cxcr)4-positive cells and improves skeletal muscle regeneration. Analysis of isolated and in vitro cultured satellite cells showed that Sdf-1 did not influence myoblasts proliferation and expression of myogenic regulatory transcription factors but induced migration of the myoblasts in Cxcr4-dependent ways. This phenomenon was also associated with the increased activity of crucial extracellular matrix modifiers, i.e. metalloproteases Mmp-2 and Mmp-9. CONCLUSIONS Thus, positive impact of Sdf-1 on muscle regeneration is related to the mobilisation of endogenous cells, that is satellite cells and myoblasts, as well as non-muscle stem cells, expressing Cxcr4 and CD34.
Collapse
Affiliation(s)
- Edyta Brzoska
- Department of Cytology, Faculty of Biology, University of Warsaw, Warsaw 02-096, Poland.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Frozoni M, Zaia AA, Line SRP, Mina M. Analysis of the contribution of nonresident progenitor cells and hematopoietic cells to reparative dentinogenesis using parabiosis model in mice. J Endod 2012; 38:1214-9. [PMID: 22892738 PMCID: PMC3745019 DOI: 10.1016/j.joen.2012.05.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2011] [Revised: 05/12/2012] [Accepted: 05/17/2012] [Indexed: 11/20/2022]
Abstract
INTRODUCTION The aim of this study was to analyze the contribution of nonresident progenitor/stem cells and hematopoietic cells to reparative dentinogenesis. METHODS Parabiosis was established between C57BL/6-TgN(ACTbEGFP)10sb/J transgenic mice (GFP+) and C57BL/6 wild-type mice (GFP-) to ensure blood cross-circulation between animals. Reparative dentinogenesis was stimulated by pulp exposures and capping on the first maxillary molar in the GFP- mice. Histologic sections of injured molars from GFP- mice were analyzed by epifluorescence microscopy to examine the contributions of GFP+ cells (nonresident progenitor cells and hematopoietic cells originating from GFP+ mice) to reparative dentinogenesis. RESULTS GFP+ cells were detected in close association with reparative dentin formed at the site of pulp exposure in the maxillary first molars of the GFP- mice. CONCLUSIONS The present study suggests the participation of the nonresident progenitor cells and hematopoietic cells in reparative dentinogenesis.
Collapse
Affiliation(s)
- Marcos Frozoni
- Department of Restorative Dentistry, Division of Endodontics, Dental School of Piracicaba, State University of Campinas, São Paulo, São Paulo, Brazil.
| | | | | | | |
Collapse
|
40
|
Horváthy DB, Vácz G, Cselenyák A, Weszl M, Kiss L, Lacza Z. Albumin-coated bioactive suture for cell transplantation. Surg Innov 2012; 20:249-55. [PMID: 22717700 DOI: 10.1177/1553350612451353] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cell therapy holds the promise for a novel modality in the surgical toolkit; however, delivery of cells into damaged soft tissues constitutes a challenge. The authors hypothesized that growing stem cells on the surface of absorbable sutures in vitro and then implanting them via stitching would be a suitable delivery route for cell therapy. Fibronectin, poly-L-lysine, and albumin coatings were used to increase attachment of human and rat bone-marrow-derived mesenchymal stem cells (BMSC) to polyfilament absorbable sutures in vitro. Fluorescence microscopy was performed to localize the cells on the suture. After 48 hours of incubation, the albumin-coated sutures had the highest cell number, and after 168 hours cell number reached confluency. In the in vivo experiments, a 10-mm incision was made on the triceps surae muscle of male Wistar rats and rat BMSC coated sutures were placed into the muscle. Two days after the implantation, cells were seen on the surface of the sutures as well as in the surrounding muscle tissue. Long-term results at 5 weeks showed that transplanted cells survived and the sutures were partly absorbed. In conclusion, coating absorbable sutures with proteins, especially serum albumin, improves attachment and proliferation of cells, and only 48 hours in culture is enough to cover the sutures sufficiently. Using these stitches in vivo resulted in short-term and long-term survival of cells. As a result, albumin-coated suture can be a vehicle for stem cell therapy in soft tissues such as muscle, tendon, or peripheral nerves.
Collapse
Affiliation(s)
- Dénes Balázs Horváthy
- Institute of Human Physiology and Clinical Experimental Research, Semmelweis University, Budapest, Hungary.
| | | | | | | | | | | |
Collapse
|
41
|
Frozoni M, Balic A, Sagomonyants K, Zaia AA, Line SRP, Mina M. A feasibility study for the analysis of reparative dentinogenesis in pOBCol3.6GFPtpz transgenic mice. Int Endod J 2012; 45:907-14. [PMID: 22551423 DOI: 10.1111/j.1365-2591.2012.02047.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
AIM To examine the feasibility of using the pOBCol3.6GFPtpz [3.6-green fluorescent protein (GFP)] transgenic mice as an in vivo model for studying the biological sequence of events during pulp healing and reparative dentinogenesis. METHODOLOGY Pulp exposures were created in the first maxillary molar of 12-16-week-old 3.6-GFP transgenic mice with CD1 and C57/Bl6 genetic background. Direct pulp capping on exposed teeth was performed using mineral trioxide aggregate followed by restoration with a light-cured adhesive system (AS) and composite resin. In control teeth, the AS was placed in direct contact with the pulp. Animals were euthanized at various time points after pulp exposure and capping. The maxillary arch was isolated, fixed and processed for histological and epifluorescence analysis to examine reparative dentinogenesis. RESULTS Analysis of teeth immediately after pulp exposure revealed absence of odontoblasts expressing 3.6-GFP at the injury site. Evidence of reparative dentinogenesis was apparent at 4 weeks in 3.6-GFP mice in CD1 background and at 8 weeks in 3.6-GFP mice with C57/Bl6 background. The reparative dentine with both groups contained newly formed atubular-mineralized tissue resembling a dentine bridge and/or osteodentine that was lined by cells expressing 3.6-GFP as well as 3.6-GFP expressing cells embedded within the atubular matrix. CONCLUSION This study was conducted in a few animals and did not allow statistical analysis. The results revealed that the 3.6-GFP transgenic animals provide a unique model for direct analysis of cellular and molecular mechanisms of pulp repair and tertiary dentinogenesis in vivo. The study also shows the effects of the capping material and the genetic background of the mice in the sequence and timing of reparative dentinogenesis.
Collapse
Affiliation(s)
- M Frozoni
- Division of Endodontics, Department of Restorative Dentistry, Piracicaba Dental School, University of Campinas, São Paulo, Brazil
| | | | | | | | | | | |
Collapse
|
42
|
Valero MC, Huntsman HD, Liu J, Zou K, Boppart MD. Eccentric exercise facilitates mesenchymal stem cell appearance in skeletal muscle. PLoS One 2012; 7:e29760. [PMID: 22253772 PMCID: PMC3256189 DOI: 10.1371/journal.pone.0029760] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Accepted: 12/03/2011] [Indexed: 01/13/2023] Open
Abstract
Eccentric, or lengthening, contractions result in injury and subsequently stimulate the activation and proliferation of satellite stem cells which are important for skeletal muscle regeneration. The discovery of alternative myogenic progenitors in skeletal muscle raises the question as to whether stem cells other than satellite cells accumulate in muscle in response to exercise and contribute to post-exercise repair and/or growth. In this study, stem cell antigen-1 (Sca-1) positive, non-hematopoetic (CD45-) cells were evaluated in wild type (WT) and α7 integrin transgenic (α7Tg) mouse muscle, which is resistant to injury yet liable to strain, 24 hr following a single bout of eccentric exercise. Sca-1+CD45− stem cells were increased 2-fold in WT muscle post-exercise. The α7 integrin regulated the presence of Sca-1+ cells, with expansion occurring in α7Tg muscle and minimal cells present in muscle lacking the α7 integrin. Sca-1+CD45− cells isolated from α7Tg muscle following exercise were characterized as mesenchymal-like stem cells (mMSCs), predominantly pericytes. In vitro multiaxial strain upregulated mMSC stem cells markers in the presence of laminin, but not gelatin, identifying a potential mechanistic basis for the accumulation of these cells in muscle following exercise. Transplantation of DiI-labeled mMSCs into WT muscle increased Pax7+ cells and facilitated formation of eMHC+DiI− fibers. This study provides the first demonstration that mMSCs rapidly appear in skeletal muscle in an α7 integrin dependent manner post-exercise, revealing an early event that may be necessary for effective repair and/or growth following exercise. The results from this study also support a role for the α7 integrin and/or mMSCs in molecular- and cellular-based therapeutic strategies that can effectively combat disuse muscle atrophy.
Collapse
MESH Headings
- Animals
- Antigens, CD/metabolism
- Ataxin-1
- Ataxins
- Biomarkers/metabolism
- Cell Proliferation/drug effects
- Cell Separation
- Connective Tissue Cells/cytology
- Female
- Gelatin/pharmacology
- Integrin alpha Chains/metabolism
- Laminin/metabolism
- Leukocyte Common Antigens/metabolism
- Mesenchymal Stem Cells/cytology
- Mesenchymal Stem Cells/drug effects
- Mesenchymal Stem Cells/metabolism
- Mice
- Mice, Transgenic
- Multipotent Stem Cells/cytology
- Multipotent Stem Cells/drug effects
- Muscle Development/drug effects
- Muscle Fibers, Skeletal/cytology
- Muscle Fibers, Skeletal/drug effects
- Muscle Fibers, Skeletal/metabolism
- Muscle, Skeletal/blood supply
- Muscle, Skeletal/cytology
- Muscle, Skeletal/drug effects
- Nerve Tissue Proteins/metabolism
- Nuclear Proteins/metabolism
- PAX7 Transcription Factor/metabolism
- Pericytes/cytology
- Pericytes/drug effects
- Physical Conditioning, Animal
- Stem Cell Transplantation
- Stress, Mechanical
- Up-Regulation/drug effects
Collapse
Affiliation(s)
- M. Carmen Valero
- Department of Kinesiology and Community Health, and Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana, Illinois, United States of America
| | - Heather D. Huntsman
- Department of Kinesiology and Community Health, and Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana, Illinois, United States of America
| | - Jianming Liu
- Department of Kinesiology and Community Health, and Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana, Illinois, United States of America
| | - Kai Zou
- Department of Kinesiology and Community Health, and Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana, Illinois, United States of America
| | - Marni D. Boppart
- Department of Kinesiology and Community Health, and Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana, Illinois, United States of America
- * E-mail:
| |
Collapse
|
43
|
Asakura A. Skeletal Muscle-derived Hematopoietic Stem Cells: Muscular Dystrophy Therapy by Bone Marrow Transplantation. ACTA ACUST UNITED AC 2012; Suppl 11. [PMID: 24524008 PMCID: PMC3918728 DOI: 10.4172/2157-7633.s11-005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
For postnatal growth and regeneration of skeletal muscle, satellite cells, a self-renewing pool of muscle stem cells, give rise to daughter myogenic precursor cells that contribute to the formation of new muscle fibers. In addition to this key myogenic cell class, adult skeletal muscle also contains hematopoietic stem cell and progenitor cell populations which can be purified as a side population (SP) fraction or as a hematopoietic marker CD45-positive cell population. These muscle-derived hematopoietic stem/progenitor cell populations are surprisingly capable of differentiation into hematopoietic cells both after transplantation into irradiated mice and during in vitro colony formation assay. Therefore, these muscle-derived hematopoietic stem/progenitor cells appear to have characteristics similar to classical hematopoietic stem/progenitor cells found in bone marrow. This review outlines recent findings regarding hematopoietic stem/progenitor cell populations residing in adult skeletal muscle and discusses their myogenic potential along with their role in the stem cell niche and related cell therapies for approaching treatment of Duchenne muscular dystrophy.
Collapse
Affiliation(s)
- Atsushi Asakura
- Stem Cell Institute, Paul and Sheila Wellstone Muscular Dystrophy Center, Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, USA
| |
Collapse
|
44
|
Turner NJ, Badylak SF. Regeneration of skeletal muscle. Cell Tissue Res 2011; 347:759-74. [PMID: 21667167 DOI: 10.1007/s00441-011-1185-7] [Citation(s) in RCA: 196] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2011] [Accepted: 04/20/2011] [Indexed: 01/12/2023]
Abstract
Skeletal muscle has a robust capacity for regeneration following injury. However, few if any effective therapeutic options for volumetric muscle loss are available. Autologous muscle grafts or muscle transposition represent possible salvage procedures for the restoration of mass and function but these approaches have limited success and are plagued by associated donor site morbidity. Cell-based therapies are in their infancy and, to date, have largely focused on hereditary disorders such as Duchenne muscular dystrophy. An unequivocal need exists for regenerative medicine strategies that can enhance or induce de novo formation of functional skeletal muscle as a treatment for congenital absence or traumatic loss of tissue. In this review, the three stages of skeletal muscle regeneration and the potential pitfalls in the development of regenerative medicine strategies for the restoration of functional skeletal muscle in situ are discussed.
Collapse
Affiliation(s)
- Neill J Turner
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Bridgeside Point 2, 450 Technology Drive, Pittsburgh, PA 15219, USA
| | | |
Collapse
|
45
|
Merritt EK, Cannon MV, Hammers DW, Le LN, Gokhale R, Sarathy A, Song TJ, Tierney MT, Suggs LJ, Walters TJ, Farrar RP. Repair of traumatic skeletal muscle injury with bone-marrow-derived mesenchymal stem cells seeded on extracellular matrix. Tissue Eng Part A 2010; 16:2871-81. [PMID: 20412030 DOI: 10.1089/ten.tea.2009.0826] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Skeletal muscle injury resulting in tissue loss poses unique challenges for surgical repair. Despite the regenerative potential of skeletal muscle, if a significant amount of tissue is lost, skeletal myofibers will not grow to fill the injured area completely. Prior work in our lab has shown the potential to fill the void with an extracellular matrix (ECM) scaffold, resulting in restoration of morphology, but not functional recovery. To improve the functional outcome of the injured muscle, a muscle-derived ECM was implanted into a 1 x 1 cm(2), full-thickness defect in the lateral gastrocnemius (LGAS) of Lewis rats. Seven days later, bone-marrow-derived mesenchymal stem cells (MSCs) were injected directly into the implanted ECM. Partial functional recovery occurred over the course of 42 days when the LGAS was repaired with an MSC-seeded ECM producing 85.4 +/- 3.6% of the contralateral LGAS. This was significantly higher than earlier recovery time points (p < 0.05). The specific tension returned to 94 +/- 9% of the contralateral limb. The implanted MSC-seeded ECM had more blood vessels and regenerating skeletal myofibers than the ECM without cells (p < 0.05). The data suggest that the repair of a skeletal muscle defect injury by the implantation of a muscle-derived ECM seeded with MSCs can improve functional recovery after 42 days.
Collapse
Affiliation(s)
- Edward K Merritt
- Department of Kinesiology, The University of Texas, Austin, Texas 78712, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
Dramatic changes in the health care landscape over the next few decades undoubtedly will affect rehabilitation specialists' practice. In the multidisciplinary field of regenerative medicine, cell, tissue, or organ substitutes are used to enhance the healing potential of the body. Given that the restoration of normal functioning of injured or diseased tissues is expected to be the ultimate goal of these therapies, the future of regenerative medicine is, undeniably, tightly intertwined with that of rehabilitation. Rehabilitation specialists not only must be aware of cutting-edge medical advances as they relate to regenerative medicine but also must work closely with basic scientists to guide the development of clinically relevant protocols. The purposes of this article are to provide a current perspective on biological approaches to the management of musculoskeletal disorders and to highlight the needed integration of physical therapeutics with regenerative medicine.
Collapse
|
47
|
Horváthy DB, Nardai PP, Major T, Schandl K, Cselenyák A, Vácz G, Kiss L, Szendrői M, Lacza Z. Muscle regeneration is undisturbed by repeated polytraumatic injury. Eur J Trauma Emerg Surg 2010; 37:161-7. [PMID: 21837257 PMCID: PMC3150816 DOI: 10.1007/s00068-010-0034-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2010] [Accepted: 05/31/2010] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Clinical observations suggest that repeated injury within a week after a traumatic event impairs the regeneration of tissues. Our aim was to investigate the effect of repeated trauma on the proliferation of satellite cells in skeletal muscle tissue. MATERIALS AND METHODS Cold lesion injury was performed in the soleus muscle and in the motor cortex of anesthetized male Wistar rats 0, 1, or 2 times with 7 day intervals between the interventions. Following the last operation, 5-bromo-2'-deoxyuridine was injected i.p. for 6 or 12 days to label dividing cells. Gut epithelium was used as positive control. Immunohistochemistry was performed 1 and 5 weeks after the last injury and the sections were analyzed with confocal microscopy. RESULTS In the case of repeated trauma, the percentage of proliferating cells remained the same compared to single hit animals after 1 week (28.0 ± 2.5% and 29.6 ± 3.0%) as well as after 5 weeks (13.9 ± 1.8% and 14.5 ± 2.2%). CONCLUSION The second hit phenomenon is probably due to systemic factors rather than to a diminished regenerating potential of injured soft tissues.
Collapse
Affiliation(s)
- D. B. Horváthy
- Institute of Human Physiology and Clinical Experimental Research, Semmelweis University, Tűzoltó utca 37-47, Budapest, 1094 Hungary
| | - P. P. Nardai
- Institute of Human Physiology and Clinical Experimental Research, Semmelweis University, Tűzoltó utca 37-47, Budapest, 1094 Hungary
| | - T. Major
- Institute of Human Physiology and Clinical Experimental Research, Semmelweis University, Tűzoltó utca 37-47, Budapest, 1094 Hungary
| | - K. Schandl
- Institute of Human Physiology and Clinical Experimental Research, Semmelweis University, Tűzoltó utca 37-47, Budapest, 1094 Hungary
| | - A. Cselenyák
- Institute of Human Physiology and Clinical Experimental Research, Semmelweis University, Tűzoltó utca 37-47, Budapest, 1094 Hungary
| | - G. Vácz
- Institute of Human Physiology and Clinical Experimental Research, Semmelweis University, Tűzoltó utca 37-47, Budapest, 1094 Hungary
| | - L. Kiss
- Institute of Human Physiology and Clinical Experimental Research, Semmelweis University, Tűzoltó utca 37-47, Budapest, 1094 Hungary
| | - M. Szendrői
- Department of Orthopedics, Semmelweis University, Budapest, Hungary
| | - Z. Lacza
- Institute of Human Physiology and Clinical Experimental Research, Semmelweis University, Tűzoltó utca 37-47, Budapest, 1094 Hungary
- Department of Orthopedics, Semmelweis University, Budapest, Hungary
| |
Collapse
|
48
|
Merritt EK, Hammers DW, Tierney M, Suggs LJ, Walters TJ, Farrar RP. Functional assessment of skeletal muscle regeneration utilizing homologous extracellular matrix as scaffolding. Tissue Eng Part A 2010; 16:1395-405. [PMID: 19929169 DOI: 10.1089/ten.tea.2009.0226] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The loss of a portion of skeletal muscle poses a unique challenge for the normal regeneration of muscle tissue. A transection injury with tissue loss will not heal due to the gap between muscle segments. A damage model was developed by removing a portion of the lateral gastrocnemius (GAS) of Sprague-Dawley rats. Maximal isometric, tetanic tension (P(o)) was measured after the removal of either a small defect (0.5 x 1.0 cm) or a large defect (1.0 x 1.0 cm) piece of the GAS. In situ P(o) immediately after creation of the defect was 88.3 +/- 2.0% of the nonoperated contralateral GAS force for small defect and 76.9 +/- 3.2% of control for large defect. No functional recovery occurred in either group over the course of 28 days. To enhance recovery, a homologous, decellularized, muscle extracellular matrix (ECM) was implanted into the 1 x 1 cm defect of the lateral GAS of Lewis rats. After 42 days, growth of blood vessels and myofibers into the ECM was apparent, but no restoration of P(o) occurred. These data demonstrate the ability of the ECM to support muscle and blood vessel regeneration, but full recovery of function does not occur after 42 days.
Collapse
Affiliation(s)
- Edward K Merritt
- Department of Kinesiology, The University of Texas at Austin , Austin, TX, USA
| | | | | | | | | | | |
Collapse
|
49
|
Sancricca C, Mirabella M, Gliubizzi C, Broccolini A, Gidaro T, Morosetti R. Vessel-associated stem cells from skeletal muscle: From biology to future uses in cell therapy. World J Stem Cells 2010; 2:39-49. [PMID: 21607121 PMCID: PMC3097924 DOI: 10.4252/wjsc.v2.i3.39] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2010] [Revised: 05/18/2010] [Accepted: 05/25/2010] [Indexed: 02/06/2023] Open
Abstract
Over the last years, the existence of different stem cells with myogenic potential has been widely investigated. Besides the classical skeletal muscle progenitors represented by satellite cells, numerous multipotent and embryologically unrelated progenitors with a potential role in muscle differentiation and repair have been identified. In order to conceive a therapeutic approach for degenerative muscle disorders, it is of primary importance to identify an ideal stem cell endowed with all the features for a possible use in vivo. Among all emerging populations, vessel-associated stem cells are a novel and promising class of multipotent progenitors of mesodermal origin and with high myogenic potential which seem to best fit all the requirements for a possible cell therapy. In vitro and in vivostudies have already tested the effectiveness and safety of vessel-associated stem cells in animal models. This leads to the concrete possibility in the future to start pilot human clinical trials, hopefully opening the way to a turning point in the treatment of genetic and acquired muscle disorders.
Collapse
Affiliation(s)
- Cristina Sancricca
- Cristina Sancricca, Massimiliano Mirabella, Carla Gliubizzi, Aldobrando Broccolini, Teresa Gidaro, Roberta Morosetti, Department of Neurosciences, Catholic University School of Medicine, Largo A. Gemelli 8, 00168 Rome, Italy
| | | | | | | | | | | |
Collapse
|
50
|
Ambrosio F, Ferrari RJ, Distefano G, Plassmeyer JM, Carvell GE, Deasy BM, Boninger ML, Fitzgerald GK, Huard J. The synergistic effect of treadmill running on stem-cell transplantation to heal injured skeletal muscle. Tissue Eng Part A 2010; 16:839-49. [PMID: 19788347 DOI: 10.1089/ten.tea.2009.0113] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Muscle-derived stem-cell (MDSC) transplantation presents a promising method for the treatment of muscle injuries. This study investigated the ability of exercise to enhance MDSC transplantation into the injured muscle. Mice were divided into four groups: contusion + phosphate-buffered saline (C + PBS; n = 14 muscles), C + MDSC transplantation (n = 12 muscles), C + PBS + treadmill running (C + PBS + TM; n = 17 muscles), and C + MDSC + TM (n = 13 muscles). One day after injury, the TM groups began running for 1 or 5 weeks. Two days after injury, muscles of C + MDSC and C + MDSC + TM groups were injected with MDSCs. One or 5 weeks later, the number and differentiation of transplanted MDSCs, myofiber regeneration, collagen I formation, and vascularity were assessed histologically. In vitro, MDSCs were subjected to mechanical stimulation, and growth kinetics were quantified. In vitro, mechanical stimulation decreased the MDSC population doubling time (18.6 +/- 1.6 h) and cell division time (10.9 +/- 0.7 h), compared with the controls (population doubling time: 23.0 +/- 3.4 h; cell division time: 13.3 +/- 1.1 h) (p = 0.01 and 0.03, respectively). In vivo, 5 weeks of TM increased the myogenic contribution of transplanted MDSCs, compared with the controls (p = 0.02). C + MDSC, C + PBS + TM, and C + MDSC + TM demonstrated decreased fibrosis at 5 weeks, compared with the C + PBS controls (p = 0.00, p = 0.03, and p = 0.02, respectively). Results suggest that the mechanical stimulation favors MDSC proliferation, both in vitro and in vivo, and that exercise enhances MDSC transplantation after injury.
Collapse
Affiliation(s)
- Fabrisia Ambrosio
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|