1
|
Lijten OW, Rosero Salazar DH, van Erp M, Bronkhorst E, Von den Hoff JW. Effect of niche components on masseter satellite cell differentiation on fibrin coatings. Eur J Oral Sci 2022; 130:e12849. [PMID: 35020959 PMCID: PMC9303748 DOI: 10.1111/eos.12849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 12/07/2021] [Indexed: 11/29/2022]
Abstract
In skeletal muscles, niche factors stimulate satellite cells to activate and induce muscle regeneration after injury. In vitro, matrigel is widely used for myoblast differentiation, however, is unsuitable for clinical applications. Therefore, this study aimed to analyze attachment and differentiation of satellite cells into myotubes on fibrin coatings with selected niche components. The attachment of satellite cells to fibrin alone and fibrin with niche components (laminin, collagen‐IV, laminin‐entactin complex [LEC]) were compared to matrigel. Only on matrigel and fibrin with LEC, Pax7‐positive cells attached well. Then, LEC was selected to analyze proliferation, differentiation, and fusion indices. The proliferation index at day 1 on fibrin‐LEC (22.5%, SD 9.1%) was similar to that on matrigel (30.8% [SD 11.1%]). The differentiation index on fibrin‐LEC (28.7% [SD 6.1%] at day 5 and 32.8% [SD 6.7%] at day 7) was similar to that on matrigel (40.1% [5.1%] at day 5 and 27.1% [SD 4.3%] at day 7). On fibrin‐LEC, the fusion index at day 9 (26.9% [SD 11.5%]) was similar to that on matrigel (25.5% [SD 4.7%]). Our results showed that the addition of LEC enhances the formation of myotubes on fibrin. Fibrin with LEC might be suitable to enhance muscle regeneration after surgery such as cleft palate repair and other muscle defects.
Collapse
Affiliation(s)
- Olivier Willem Lijten
- Department of Orthodontics and Craniofacial Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Doris Haydee Rosero Salazar
- Department of Orthodontics and Craniofacial Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Medical Basic Sciences, Faculty of Health, Universidad Icesi, Cali, Colombia
| | - Merijn van Erp
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ewald Bronkhorst
- Department of Dentistry, Radboud University Medical Center, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
| | - Johannes W Von den Hoff
- Department of Orthodontics and Craniofacial Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
2
|
Godbout M, Vargas A, Hélie P, Bullone M, Lavoie JP. Use of a biopolymer delivery system to investigate the influence of interleukin-4 on recruitment of neutrophils in equids. Am J Vet Res 2020; 81:344-354. [PMID: 32228258 DOI: 10.2460/ajvr.81.4.344] [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: 11/20/2022]
Abstract
OBJECTIVE To use a biopolymer delivery system to investigate the ability of interleukin (IL)-4 to recruit neutrophils into subcutaneous tissues of equids. ANIMALS 16 horses and 2 ponies. PROCEDURES Animals were assigned to 3 experiments (6/experiment). Effects of recombinant equine (Req) IL-4 (100, 250, or 500 ng/site) versus a positive control (ReqIL-8; 100 ng, 250 ng, or 1 μg/site) and a negative control (Dulbecco PBSS or culture medium) on neutrophil chemotaxis were assessed after SC injection into the neck with an injectable biopolymer used as the vehicle. Tissue samples including the biopolymer plug were collected by biopsy at various time points from 3 hours to 7 days after injection. Neutrophil infiltration was evaluated by histologic scoring (experiments 1, 2, and 3) or flow cytometry (experiment 3). RESULTS Histologic neutrophil infiltration scores did not differ significantly among treatments at most evaluated time points. On flow cytometric analysis, log-transformed neutrophil counts in biopsy specimens were significantly greater for the ReqIL-8 treatment (1 μg/site) than the negative control treatment at 3 but not 6 hours after injection; results did not differ between ReqIL-4 and control treatments at either time point. Negative control treatments induced an inflammatory response in most equids in all experiments. CONCLUSIONS AND CLINICAL RELEVANCE Flow cytometry was a more reliable method to estimate neutrophil migration than histologic score analysis. The ReqIL-4 treatment did not induce a detectable neutrophil response, compared with the negative control treatment in this study. Evidence of inflammation in negative control samples suggested the biopolymer is not a suitable vehicle for use in equids.
Collapse
|
3
|
High glucose inhibits myogenesis and induces insulin resistance by down-regulating AKT signaling. Biomed Pharmacother 2019; 120:109498. [DOI: 10.1016/j.biopha.2019.109498] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 09/21/2019] [Accepted: 09/26/2019] [Indexed: 12/13/2022] Open
|
4
|
Wang D, Gao CQ, Chen RQ, Jin CL, Li HC, Yan HC, Wang XQ. Focal adhesion kinase and paxillin promote migration and adhesion to fibronectin by swine skeletal muscle satellite cells. Oncotarget 2017; 7:30845-54. [PMID: 27127174 PMCID: PMC5058722 DOI: 10.18632/oncotarget.9010] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Accepted: 04/08/2016] [Indexed: 12/18/2022] Open
Abstract
The focal adhesion kinase (FAK) signaling pathway contributes to the cell migration and adhesion that is critical for wound healing and regeneration of damaged muscle, but its function in skeletal muscle satellite cells (SCs) is less clear. We compared the migration and adhesion of SCs derived from two species of pig (Lantang and Landrace) in vitro, and explored how FAK signaling modulates the two processes. The results showed that Lantang SCs had greater ability to migrate and adhere to fibronection (P < 0.05) than Landrace SCs. Compared to Landrace SCs, Lantang SCs expressed many more focal adhesion (FA) sites, which were indicated by the presence of p-paxillin (Tyr118), and exhibited less F-actin reorganization 24 h after seeding onto fibronectin. Levels of p-FAK (Tyr397) and p-paxillin (Tyr118) were greater (P < 0.05) in Lantang SCs than Landrace SCs after migration for 24 h. Similarly, Lantang SCs showed much higher levels of p-FAK (Tyr397), p-paxillin (Tyr118) and p-Akt (Ser473) than Landrace SCs 2 h after adhesion. Treatment with the FAK inhibitor PF-573228 (5 or 10 μmol/L) inhibited Lantang SC migration and adhesion to fibronectin (P < 0.05), decreased levels of p-paxillin (Tyr118) and p-Akt (Ser473) (P < 0.05), and suppressed the formation of FA sites on migrating SCs. Thus FAK appears to play a key role in the regulation of SC migration and adhesion necessary for muscle regeneration.
Collapse
Affiliation(s)
- Dan Wang
- College of Animal Science, South China Agricultural University/National Engineering Research Center for Breeding Swine Industry, Guangzhou, Guangdong Province, China
| | - Chun-Qi Gao
- College of Animal Science, South China Agricultural University/National Engineering Research Center for Breeding Swine Industry, Guangzhou, Guangdong Province, China
| | - Rong-Qiang Chen
- College of Animal Science, South China Agricultural University/National Engineering Research Center for Breeding Swine Industry, Guangzhou, Guangdong Province, China
| | - Cheng-Long Jin
- College of Animal Science, South China Agricultural University/National Engineering Research Center for Breeding Swine Industry, Guangzhou, Guangdong Province, China
| | - Hai-Chang Li
- Davis Heart and Lung Research Institute, Wexner Medical Center at the Ohio State University, Columbus, OH, USA
| | - Hui-Chao Yan
- College of Animal Science, South China Agricultural University/National Engineering Research Center for Breeding Swine Industry, Guangzhou, Guangdong Province, China
| | - Xiu-Qi Wang
- College of Animal Science, South China Agricultural University/National Engineering Research Center for Breeding Swine Industry, Guangzhou, Guangdong Province, China
| |
Collapse
|
5
|
González MN, de Mello W, Butler-Browne GS, Silva-Barbosa SD, Mouly V, Savino W, Riederer I. HGF potentiates extracellular matrix-driven migration of human myoblasts: involvement of matrix metalloproteinases and MAPK/ERK pathway. Skelet Muscle 2017; 7:20. [PMID: 29017538 PMCID: PMC5635537 DOI: 10.1186/s13395-017-0138-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 09/22/2017] [Indexed: 12/31/2022] Open
Abstract
Background The hepatocyte growth factor (HGF) is required for the activation of muscle progenitor cells called satellite cells (SC), plays a role in the migration of proliferating SC (myoblasts), and is present as a soluble factor during muscle regeneration, along with extracellular matrix (ECM) molecules. In this study, we aimed at determining whether HGF is able to interact with ECM proteins, particularly laminin 111 and fibronectin, and to modulate human myoblast migration. Methods We evaluated the expression of the HGF-receptor c-Met, laminin, and fibronectin receptors by immunoblotting, flow cytometry, or immunofluorescence and used Transwell assays to analyze myoblast migration on laminin 111 and fibronectin in the absence or presence of HGF. Zymography was used to check whether HGF could modulate the production of matrix metalloproteinases by human myoblasts, and the activation of MAPK/ERK pathways was evaluated by immunoblotting. Results We demonstrated that human myoblasts express c-Met, together with laminin and fibronectin receptors. We observed that human laminin 111 and fibronectin have a chemotactic effect on myoblast migration, and this was synergistically increased when low doses of HGF were added. We detected an increase in MMP-2 activity in myoblasts treated with HGF. Conversely, MMP-2 inhibition decreased the HGF-associated stimulation of cell migration triggered by laminin or fibronectin. HGF treatment also induced in human myoblasts activation of MAPK/ERK pathways, whose specific inhibition decreased the HGF-associated stimulus of cell migration triggered by laminin 111 or fibronectin. Conclusions We demonstrate that HGF induces ERK phosphorylation and MMP production, thus stimulating human myoblast migration on ECM molecules. Conceptually, these data state that the mechanisms involved in the migration of human myoblasts comprise both soluble and insoluble moieties. This should be taken into account to optimize the design of therapeutic cell transplantation strategies by improving the migration of donor cells within the host tissue, a main issue regarding this approach. Electronic supplementary material The online version of this article (10.1186/s13395-017-0138-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mariela Natacha González
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Av. Brasil 4365, Manguinhos, Rio de Janeiro, 21045-900, Brazil.,Brazilian National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Av. Brasil 4365, Manguinhos, 21045-900, Rio de Janeiro, Brasil
| | - Wallace de Mello
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Av. Brasil 4365, Manguinhos, Rio de Janeiro, 21045-900, Brazil
| | - Gillian S Butler-Browne
- Sorbonne Universités, Université Pierre et Marie Curie, INSERM UMRS974, CNRS FRE3617, Center for Research in Myology, 47 Boulevard de l'hôpital, 75013, Paris, France
| | - Suse Dayse Silva-Barbosa
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Av. Brasil 4365, Manguinhos, Rio de Janeiro, 21045-900, Brazil.,Department of Clinical Research, National Cancer Institute (INCA), Rio de Janeiro, Brazil
| | - Vincent Mouly
- Sorbonne Universités, Université Pierre et Marie Curie, INSERM UMRS974, CNRS FRE3617, Center for Research in Myology, 47 Boulevard de l'hôpital, 75013, Paris, France
| | - Wilson Savino
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Av. Brasil 4365, Manguinhos, Rio de Janeiro, 21045-900, Brazil.,Brazilian National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Av. Brasil 4365, Manguinhos, 21045-900, Rio de Janeiro, Brasil
| | - Ingo Riederer
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Av. Brasil 4365, Manguinhos, Rio de Janeiro, 21045-900, Brazil. .,Brazilian National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Av. Brasil 4365, Manguinhos, 21045-900, Rio de Janeiro, Brasil.
| |
Collapse
|
6
|
Amer MH, Rose FRAJ, Shakesheff KM, Modo M, White LJ. Translational considerations in injectable cell-based therapeutics for neurological applications: concepts, progress and challenges. NPJ Regen Med 2017; 2:23. [PMID: 29302358 PMCID: PMC5677964 DOI: 10.1038/s41536-017-0028-x] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 06/27/2017] [Accepted: 07/12/2017] [Indexed: 12/11/2022] Open
Abstract
Significant progress has been made during the past decade towards the clinical adoption of cell-based therapeutics. However, existing cell-delivery approaches have shown limited success, with numerous studies showing fewer than 5% of injected cells persisting at the site of injection within days of transplantation. Although consideration is being increasingly given to clinical trial design, little emphasis has been given to tools and protocols used to administer cells. The different behaviours of various cell types, dosing accuracy, precise delivery, and cell retention and viability post-injection are some of the obstacles facing clinical translation. For efficient injectable cell transplantation, accurate characterisation of cellular health post-injection and the development of standardised administration protocols are required. This review provides an overview of the challenges facing effective delivery of cell therapies, examines key studies that have been carried out to investigate injectable cell delivery, and outlines opportunities for translating these findings into more effective cell-therapy interventions.
Collapse
Affiliation(s)
- Mahetab H. Amer
- School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD UK
| | | | | | - Michel Modo
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA USA
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA USA
| | - Lisa J. White
- School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD UK
| |
Collapse
|
7
|
Grasman JM, Page RL, Pins GD. * Design of an In Vitro Model of Cell Recruitment for Skeletal Muscle Regeneration Using Hepatocyte Growth Factor-Loaded Fibrin Microthreads. Tissue Eng Part A 2017; 23:773-783. [PMID: 28351217 DOI: 10.1089/ten.tea.2016.0440] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Large skeletal muscle defects that result in volumetric muscle loss (VML) result in the destruction of the basal lamina, which removes key signaling molecules such as hepatocyte growth factor (HGF) from the wound site, eliminating the endogenous capacity of these injuries to regenerate. We recently showed that HGF-loaded fibrin microthreads increased the force production in muscle tissues after 60 days in a mouse VML model. In this study, we created an in vitro, three-dimensional (3D) microscale outgrowth assay system designed to mimic cell recruitment in vivo, and investigated the effect of HGF-loaded, cross-linked fibrin microthreads on myoblast recruitment to predict the results observed in vivo. This outgrowth assay discretely separated the cellular and molecular functions (migration, proliferation, and chemotaxis) that direct outgrowth from the wound margin, creating a powerful platform to model cell recruitment in axially aligned tissues, such as skeletal muscle. The degree of cross-linking was controlled by pH and microthreads cross-linked using physiologically neutral pH (EDCn) facilitated the release of active HGF; increasing the two-dimensional migration and 3D outgrowth of myoblasts twofold. While HGF adsorbed to uncross-linked microthreads, it did not enhance myoblast migration, possibly due to the low concentrations that were adsorbed. Regardless of the amount of HGF adsorbed on the microthreads, myoblast proliferation increased significantly on stiffer, cross-linked microthreads. Together, the results of these studies show that HGF loaded onto EDCn microthreads supported enhanced myoblast migration and recruitment and suggest that our novel outgrowth assay system is a robust in vitro screening tool that predicts the performance of fibrin microthreads in vivo.
Collapse
Affiliation(s)
- Jonathan M Grasman
- 1 Department of Biomedical Engineering, Worcester Polytechnic Institute , Worcester, Massachusetts.,2 Bioengineering Institute, Worcester Polytechnic Institute , Worcester, Massachusetts
| | - Raymond L Page
- 1 Department of Biomedical Engineering, Worcester Polytechnic Institute , Worcester, Massachusetts.,2 Bioengineering Institute, Worcester Polytechnic Institute , Worcester, Massachusetts
| | - George D Pins
- 1 Department of Biomedical Engineering, Worcester Polytechnic Institute , Worcester, Massachusetts.,2 Bioengineering Institute, Worcester Polytechnic Institute , Worcester, Massachusetts
| |
Collapse
|
8
|
Hepatocyte Growth Factor and Satellite Cell Activation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 900:1-25. [PMID: 27003394 DOI: 10.1007/978-3-319-27511-6_1] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Satellite cells are the "currency" for the muscle growth that is critical to meat production in many species, as well as to phenotypic distinctions in development at the level of species or taxa, and for human muscle growth, function and regeneration. Careful research on the activation and behaviour of satellite cells, the stem cells in skeletal muscle, including cross-species comparisons, has potential to reveal the mechanisms underlying pathological conditions in animals and humans, and to anticipate implications of development, evolution and environmental change on muscle function and animal performance.
Collapse
|
9
|
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: 22] [Impact Index Per Article: 2.4] [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
- * E-mail:
| |
Collapse
|
10
|
Xiang J, Zhao Y, Chen J, Zhou J. Expression of basic fibroblast growth factor, protein kinase C and members of the apoptotic pathway in skeletal muscle of streptozotocin-induced diabetic rats. Tissue Cell 2013; 46:1-8. [PMID: 24008114 DOI: 10.1016/j.tice.2013.07.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 07/22/2013] [Accepted: 07/22/2013] [Indexed: 01/11/2023]
Abstract
This study investigated the potential mechanisms that may underlie diabetes induced amyoatrophy. Sprague-Dawley rats were either injected intraperiotneally with STZ (test group; N=8) to induce diabetic-like symptoms (blood glucose level ≥16.65mmol/L) or with buffer (control group; N=8). Differences in muscle structure between the STZ-induced diabetic and control groups were evaluated by histochemistry. Protein and mRNA levels of basic FGF (bFGF), bax, bcl-2, and caspase 3 in skeletal muscle were compared between the 2 groups using immunohistochemistry and quantitative PCR, respectively. Serum level of insulin and protein kinase C (PKC) were measured by competitive RIA and ELISA, respectively. Unlike control animals, the skeletal muscle fibers from STZ-induced diabetic animals were broken and pyknotic, the sarcomeric structure disrupted, and mild hyperplasia of interstitial adipose tissues was detected. The serum level of PKC was higher (P=0.003) and the protein and mRNA levels of bFGF in skeletal muscle were lower (P=0.001) in STZ-induced diabetic versus control animals. Protein and mRNA levels of the apoptosis promoting genes caspase-3 and bax were higher in skeletal muscle from STZ-induced diabetic rats as compared to control animals (P<0.001 and P=0.037, respectively), while mRNA and protein levels of bcl-2, an inhibitor of apoptosis, was lower in STZ-induced diabetic rats versus control animals (P=0.026). Increasing apoptosis in skeletal muscle from STZ-induced diabetic rats was further demonstrated by TNNEL assay. Our findings suggest that enhanced PKC levels, reduction of bFGF expression, and increased in apoptosis might be associated with the development of diabetes-induced myoatrophy.
Collapse
Affiliation(s)
- Jingyan Xiang
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Yuwu Zhao
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China.
| | - Jingjiong Chen
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Jian Zhou
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| |
Collapse
|
11
|
Grefte S, Vullinghs S, Kuijpers-Jagtman AM, Torensma R, Von den Hoff JW. Matrigel, but not collagen I, maintains the differentiation capacity of muscle derived cells in vitro. Biomed Mater 2012; 7:055004. [PMID: 22791687 DOI: 10.1088/1748-6041/7/5/055004] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Satellite cells are key cells for post-natal muscle growth and regeneration and they play a central role in the search for therapies to treat muscle injuries. In this study the proliferation and differentiation capacity of muscle progenitor cells was studied in 2D and 3D cultures with collagen type I and Matrigel, which contain the niche factors laminin and collagen type IV. Muscle progenitor cells were cultured to induce proliferation and differentiation in collagen- or Matrigel-coated surfaces (2D) or in gels (3D). In the 2D cultures, muscle progenitor cells proliferated faster in Matrigel than in collagen. The numbers of Pax7(+) and MyoD(+) cells were also significantly higher in Matrigel than in collagen. During differentiation, muscle progenitor cells formed more and larger MyoD(+) and myogenin(+) myotubes in Matrigel. In the 3D cultures, muscle progenitor cells in Matrigel expressed higher mRNA levels of MyoD and myogenin, and formed elongated myotubes expressing myogenin and myosin. In collagen gels, the myotubes were short and rounded. In conclusion, muscle progenitor cells, both in 2D and 3D, lose their differentiation capacity in collagen but not in Matrigel. Although Matrigel contains growth factors, our results indicate that the kind of biomaterial steers the maintenance of the myogenic potential and their proper differentiation to achieve optimal skeletal muscle restoration.
Collapse
Affiliation(s)
- S Grefte
- Department of Orthodontics and Craniofacial Biology, Radboud University Nijmegen Medical Centre, 6500 HB Nijmegen, The Netherlands
| | | | | | | | | |
Collapse
|
12
|
Ishido M, Kasuga N. In Vivo Real-Time Imaging of Exogenous HGF-Triggered Cell Migration in Rat Intact Soleus Muscles. Acta Histochem Cytochem 2012; 45:193-9. [PMID: 22829713 PMCID: PMC3395305 DOI: 10.1267/ahc.11058] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2011] [Accepted: 04/13/2012] [Indexed: 11/22/2022] Open
Abstract
The transplantation of myogenic cells is a potentially effective therapy for muscular dystrophy. However, this therapy has achieved little success because the diffusion of transplanted myogenic cells is limited. Hepatocyte growth factor (HGF) is one of the primary triggers to induce myogenic cell migration in vitro. However, to our knowledge, whether exogenous HGF can trigger the migration of myogenic cells (i.e. satellite cells) in intact skeletal muscles in vivo has not been reported. We previously reported a novel in vivo real-time imaging method in rat skeletal muscles. Therefore, the present study examined the relationship between exogenous HGF treatment and cell migration in rat intact soleus muscles using this imaging method. As a result, it was indicated that the cell migration velocity was enhanced in response to increasing exogenous HGF concentration in skeletal muscles. Furthermore, the expression of MyoD was induced in satellite cells in response to HGF treatment. We first demonstrated in vivo real-time imaging of cell migration triggered by exogenous HGF in intact soleus muscles. The experimental method used in the present study will be a useful tool to understand further the regulatory mechanism of HGF-induced satellite cell migration in skeletal muscles in vivo.
Collapse
Affiliation(s)
- Minenori Ishido
- Faculty of Education, Creative Arts and Sciences, Aichi University of Education
- Faculty of Education, Creative Arts and Sciences, Aichi University of Education
| | - Norikatsu Kasuga
- Department of Health and Physical Education, Aichi University of Education
- Department of Health and Physical Education, Aichi University of Education
| |
Collapse
|
13
|
Kim WH, Jung DW, Kim J, Im SH, Hwang SY, Williams DR. Small molecules that recapitulate the early steps of urodele amphibian limb regeneration and confer multipotency. ACS Chem Biol 2012; 7:732-43. [PMID: 22270490 DOI: 10.1021/cb200532v] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
In urodele amphibians, an early step in limb regeneration is skeletal muscle fiber dedifferentiation into a cellulate that proliferates to contribute new limb tissue. However, mammalian muscle cannot dedifferentiate after injury. We have developed a novel, small-molecule-based method to induce dedifferentiation in mammalian skeletal muscle. Muscle cellularization was induced by the small molecule myoseverin. Candidate small molecules were tested for the induction of proliferation in the cellulate. We observed that treatment with the small molecules BIO (glycogen synthase-3 kinase inhibitor), lysophosphatidic acid (pleiotropic activator of G-protein-coupled receptors), SB203580 (p38 MAP kinase inhibitor), or SQ22536 (adenylyl cyclase inhibitor) induced proliferation. Moreover, these proliferating cells were multipotent, as confirmed by the chemical induction of mesodermal-derived cell lineages. Microarray analysis showed that the multipotent, BIO-treated cellulate possessed a markedly different gene expression pattern than lineage-restricted C2C12 myoblasts, especially for genes related to signal transduction and differentiation. Sequential small molecule treatment of the muscle cellulate with BIO, SB203580, or SQ22536 and the aurora B kinase inhibitor, reversine, induced the formation of cells with neurogenic potential (ectodermal lineage), indicating the acquirement of pluripotency. This is the first demonstration of a small molecule method that induces mammalian muscle to undergo dedifferentiation and rededifferentiation into alternate cell lineages. This method induces dedifferentiation in a simple, stepwise approach and has therapeutic potential to enhance tissue regeneration in mammals.
Collapse
Affiliation(s)
| | | | | | | | - Seung Yong Hwang
- Department of Biochemistry, Hanyang University and GenoCheck Co., Ltd., Sa-Dong, Sangrok-Gu, Ansan, Gyeonggi-Do, 426-791,
Republic of Korea
| | | |
Collapse
|
14
|
Palladino M, Gatto I, Neri V, Stigliano E, Smith RC, Pola E, Straino S, Gaetani E, Capogrossi M, Leone G, Hlatky L, Pola R. Combined Therapy with Sonic Hedgehog Gene Transfer and Bone Marrow-Derived Endothelial Progenitor Cells Enhances Angiogenesis and Myogenesis in the Ischemic Skeletal Muscle. J Vasc Res 2012; 49:425-31. [DOI: 10.1159/000337921] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Accepted: 02/23/2012] [Indexed: 01/16/2023] Open
|
15
|
Bayoussef Z, Dixon JE, Stolnik S, Shakesheff KM. Aggregation promotes cell viability, proliferation, and differentiation in an in vitro model of injection cell therapy. J Tissue Eng Regen Med 2011; 6:e61-73. [PMID: 21932267 DOI: 10.1002/term.482] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2010] [Revised: 02/23/2011] [Accepted: 07/11/2011] [Indexed: 11/11/2022]
Abstract
Many cell therapy approaches aim to deliver high-density single-cell suspensions to diseased or injured sites in the body. Long term clinical success will in part be dependent on the cells that remain viable and that assume correct functionality post-administration. The research presented in this paper focuses on the potential of cell aggregate delivery to generate a more supportive environment for cells than single cell suspensions. An in vitro model of injection delivery of C2C12 myoblast cells showed a significant difference in cell function and phenotype between adhesive collagen and non-adhesive alginate, indicating that in vitro assays based on this approach can discriminate between cell-cell/cell-matrix interactions and could be valuable when assessing cell therapy systems. Contrary to single cells, aggregates maintain viability, cellular activity, and phenotype beyond that of single cells, even in non-adhesive matrices, enabling delivery of higher cell densities with enhanced proliferative and differentiation capacity.
Collapse
Affiliation(s)
- Zahia Bayoussef
- Tissue Engineering, Wolfson Centre for Stem Cells, Tissue Engineering, Modelling (STEM), Centre for Biomolecular Sciences, School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, UK
| | | | | | | |
Collapse
|
16
|
Grimaldi A, Banfi S, Vizioli J, Tettamanti G, Noonan DM, de Eguileor M. Cytokine loaded biopolymers as a novel strategy to study stem cells during wound-healing processes. Macromol Biosci 2011; 11:1008-19. [PMID: 21400659 DOI: 10.1002/mabi.201000452] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2010] [Revised: 02/10/2011] [Indexed: 12/12/2022]
Abstract
The biopolymer matrigel loaded with cytokine can be used for the recruitment in vivo of specific cell populations and as a vector for the preparation of cell cultures. Data demonstrate that the injection of the matrigel biopolymer supplemented with interleukin-8 (IL-8) in the leech Hirudo medicinalis can be used to purify cell populations showing the same morphofunctional and molecular mechanisms of specific populations of vertebrate hematopoietic precursor cells involved in tissue repair. These cells spontaneously differentiated into myofibroblasts. This approach highlights how the innovative use of a cytokine-loaded biopolymer for an in vivo cell sorting method, applied to a simple invertebrate model, can be a tool for studying myofibroblast cell biology and its regulation, step by step.
Collapse
Affiliation(s)
- Annalisa Grimaldi
- Department of Biotechnology and Molecular Sciences, University of Insubria, Varese, Italy.
| | | | | | | | | | | |
Collapse
|
17
|
Ishido M, Kasuga N. In situ real-time imaging of the satellite cells in rat intact and injured soleus muscles using quantum dots. Histochem Cell Biol 2010; 135:21-6. [PMID: 21132508 DOI: 10.1007/s00418-010-0767-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/19/2010] [Indexed: 12/26/2022]
Abstract
The recruitment of satellite cells, which are located between the basement membrane and the plasma membrane in myofibers, is required for myofiber repair after muscle injury or disease. In particular, satellite cell migration has been focused on as a satellite cell response to muscle injury because satellite cell motility has been revealed in cell culture. On the other hand, in situ, it is poorly understood how satellite cell migration is involved in muscle regeneration after injury because in situ it has been technically very difficult to visualize living satellite cells localized within skeletal muscle. In the present study, using quantum dots conjugated to anti-M-cadherin antibody, we attempted the visualization of satellite cells in both intact and injured skeletal muscle of rat in situ. As a result, the present study is the first to demonstrate in situ real-time imaging of satellite cells localized within the skeletal muscle. Moreover, it was indicated that satellite cell migration toward an injured site was induced in injured muscle while spatiotemporal change in satellite cells did not occur in intact muscle. Thus, it was suggested that the satellite cell migration may play important roles in the regulation of muscle regeneration after injury. Moreover, the new method used in the present study will be a useful tool to develop satellite cell-based therapies for muscle injury or disease.
Collapse
Affiliation(s)
- Minenori Ishido
- Faculty of Education, Creative Arts and Sciences, Aichi University of Education, Igaya-cho, Kariya, Aichi, Japan.
| | | |
Collapse
|
18
|
Zhao W, Zhang C, Jin C, Zhang Z, Kong D, Xu W, Xiu Y. Periurethral injection of autologous adipose-derived stem cells with controlled-release nerve growth factor for the treatment of stress urinary incontinence in a rat model. Eur Urol 2010; 59:155-63. [PMID: 21050657 DOI: 10.1016/j.eururo.2010.10.038] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2010] [Accepted: 10/15/2010] [Indexed: 12/11/2022]
Abstract
BACKGROUND Stem cell therapy is a promising therapeutic strategy for stress urinary incontinence (SUI). However, its current efficacy is insufficient. OBJECTIVE We designed a stem cell transplantation system that contains autologous adipose-derived stem cells (ADSC) and controlled-release nerve growth factor (NGF). We evaluated whether this system could enhance the therapeutic efficacy of ADSCs by periurethral coinjection in SUI rats. DESIGN, SETTING, AND PARTICIPANTS We first tested for the presence of NGF receptors in rat ADSCs and observed the effect of NGF on ADSCs in vitro and in vivo. NGF was encapsulated within poly(lactic-co-glycolic acid-PLGA) microspheres (PLGA/NGF) to control its release. SUI was created in rats, and ADSCs were harvested, cultured from fat tissue, and retained for later transplantation. SUI rats then received different forms of periurethral injection therapy. Their urodynamic index was monitored. Eight weeks after injection, the SUI rats were sacrificed and their urethra removed for histologic evaluation. INTERVENTION Forty SUI rats were allocated to five groups for receiving periurethral injection with phosphate-buffered saline (PBS), ADSC, ADSC+PLGA, ADSC+NGF, or ADSC+PLGA/NGF. Bladder capacities, abdominal leak point pressure (ALPP), and retrograde urethral perfusion pressure (RUPP) were reassessed at 2, 6, and 8 wk after injection. MEASUREMENTS The rat SUI model was generated by bilateral pudendal nerve transection (PNT). Real-time polymerase chain reaction (RT-PCR) and western blotting detected the NGF receptor Ark-A. The regeneration of muscles and peripheral nerves was evaluated by Masson's trichrome and immunohistochemical staining. RESULTS AND LIMITATIONS Results revealed the presence of the NGF receptor Trk-A on rat ADSCs. Short-term observations showed that NGF could improve ADSCs' viability in vitro and in vivo. ADSCs delivered intramuscularly into the urethra in combination with PLGA/NGF resulted in significant improvements in ALPP and RUPP as well as the amount of muscle and ganglia. There was a significant difference between the ADSC+PLGA/NGF group and other groups. CONCLUSIONS Periurethral coinjection of autologous ADSCs with controlled-release NGF may be a potential strategy for SUI treatment.
Collapse
Affiliation(s)
- Weiming Zhao
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | | | | | | | | | | | | |
Collapse
|
19
|
Yanagiuchi A, Miyake H, Nomi M, Takenaka A, Fujisawa M. Modulation of the microenvironment by growth factors regulates thein vivogrowth of skeletal myoblasts. BJU Int 2009; 103:1569-73. [DOI: 10.1111/j.1464-410x.2008.08318.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
20
|
Grimaldi A, Bianchi C, Greco G, Tettamanti G, Noonan DM, Valvassori R, de Eguileor M. In vivo isolation and characterization of stem cells with diverse phenotypes using growth factor impregnated biomatrices. PLoS One 2008; 3:e1910. [PMID: 18382683 PMCID: PMC2270903 DOI: 10.1371/journal.pone.0001910] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2008] [Accepted: 02/27/2008] [Indexed: 11/18/2022] Open
Abstract
Background The stimulation to differentiate into specific cell types for somatic stem cells is largely due to a series of internal and external signals coming from the microenvironment that surrounds the stem cell. Even though intensive research has been made, the basic mechanisms of plasticity and/or the molecules regulating stem cells proliferation and differentiation are not completely determined. Potential answers concerning the problems could be derived from the studies of stem cells in culture. Methodology/Principle Findings We combine a new procedure (using the matrigel biopolymer supplemented with a selected cytokine/growth factor) with classic techniques such as light, confocal and electron microscopy, immunohistochemistry and cell culture, to perform an analysis on stem cells involved in the leech (Hirudo medicinalis) repair tissues. The leech has a relative anatomical simplicity and is a reliable model for studying a variety of basic events, such as tissue repair, which has a striking similarity with vertebrate responses. Our data demonstrate that the injection of an appropriate combination of the matrigel biopolymer supplemented with a selected cytokine/growth factor in the leech Hirudo medicinalis is a remarkably effective tool for isolating a specific cell population in vivo. A comparative analysis of biopolymer in vivo sorted stem cells indicates that VEGF recruited cells of a hematopoietic/endothelial phenotype whereas MCP-1/CCL2 isolated cells that were of an early myeloid lineage. Conclusion Our paper describes, for the first time, a method allowing not only the isolation of a specific cell population in relation to the cytokine utilized but also the possibility to culture a precise cell type whose isolation is otherwise quite difficult. This approach could be broadly applied to isolate stem cells of diverse origins based on the recruitment stimuli employed.
Collapse
Affiliation(s)
- Annalisa Grimaldi
- Department of Structural and Functional Biology, University of Insubria, Varese, Italy.
| | | | | | | | | | | | | |
Collapse
|
21
|
Dinsmore JH, Dib N. Stem cells and cardiac repair: a critical analysis. J Cardiovasc Transl Res 2008; 1:41-54. [PMID: 20559957 DOI: 10.1007/s12265-007-9008-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2007] [Accepted: 12/27/2007] [Indexed: 01/11/2023]
Abstract
Utilizing stem cells to repair the damaged heart has seen an intense amount of activity over the last 5 years or so. There are currently multiple clinical studies in progress to test the efficacy of various different cell therapy approaches for the repair of damaged myocardium that were only just beginning to be tested in preclinical animal studies a few years earlier. This rapid transition from preclinical to clinical testing is striking and is not typical of the customary timeframe for the progress of a therapy from bench-to-bedside. Doubtless, there will be many more trials to follow in the upcoming years. With the plethora of trials and cell alternatives, there has come not only great enthusiasm for the potential of the therapy, but also great confusion about what has been achieved. Cell therapy has the potential to do what no drug can: regenerate and replace damaged tissue with healthy tissue. Drugs may be effective at slowing the progression of heart failure, but none can stop or reverse the process. However, tissue repair is not a simple process, although the idea on its surface is quite simple. Understanding cells, the signals that they respond to, and the keys to appropriate survival and tissue formation are orders of magnitude more complicated than understanding the pathways targeted by most drugs. Drugs and their metabolites can be monitored, quantified, and their effects correlated to circulating levels in the body. Not so for most cell therapies. It is quite difficult to measure cell survival except through ex vivo techniques like histological analysis of the target organ. This makes the emphasis on preclinical research all the more important because it is only in the animal studies that research has the opportunity to readily harvest the target tissues and perform the detailed analyses of what has happened with the cells. This need for detailed and usually time-intensive research in animal studies stands in contrast to the rapidity with which therapies have progressed to the clinic. It is now becoming clear through a number of notable examples that progress to the clinic may have occurred too quickly, before adequate testing and independent verification of results could be completed (Check, Nature 446:485-486, 2007; Chien, J Clin Investig 116:1838-1840, 2006; Giles, Nature 442:344-347, 2006). Broad reproducibility and transfer of results from one lab to another has been and always will be essential for the successful application of any cell therapy. So, what is the prognosis for cell therapy to repair heart damage? Will there be an approved cell therapy, or multiple ones, or will it require combinations of more than one cell type to be successful? These are questions often asked. The answers are difficult to know and even more difficult to predict because there are so many variables associated with cell-based therapies. There is much about the biology of cell systems that we still do not understand. Much of the pluripotency or transdifferentiation phenomena (see below) being observed go against accepted and well-tested principles for cell development and fate choice, and has caused a reevaluation of long-accepted theories. Clearly, new pathways for tissue repair and regeneration have been uncovered, but will these new pathways be sufficient to effect significant tissue repair and regeneration? Despite the false starts so far, there is the strong likelihood one or possibly multiple cell therapies will succeed. Clearly, important information has been gained, which should better guide the field to achieving success. When there is the successful verification in patients of a cell therapy, there will be an explosion of technological advances around the approach(es) that succeed. Whatever cells get approved accompanying them will be: more effective delivery methods; growth and storage methods; combination therapies, mixes of cells or cells + gene therapies; combinations with biomaterials and technologies for immune protection, allowing allografting. There are many parallel paths of technology development waiting to be brought together once there is an effective cellular approach. The coming years will no doubt bring some exciting developments.
Collapse
Affiliation(s)
- Jonathan H Dinsmore
- Advanced Cell Technology and Mytogen, Inc., Bldg. 96, 13th St., Charlestown, MA 02129, USA.
| | | |
Collapse
|
22
|
Kroehne V, Heschel I, Schügner F, Lasrich D, Bartsch JW, Jockusch H. Use of a novel collagen matrix with oriented pore structure for muscle cell differentiation in cell culture and in grafts. J Cell Mol Med 2008; 12:1640-8. [PMID: 18194451 PMCID: PMC2680279 DOI: 10.1111/j.1582-4934.2008.00238.x] [Citation(s) in RCA: 118] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Tissue engineering of skeletal muscle from cultured cells has been attempted using a variety of synthetic and natural macromolecular scaffolds. Our study describes the application of artificial scaffolds (collagen sponges, CS) consisting of collagen-I with parallel pores (width 20–50 μm) using the permanent myogenic cell line C2C12. CS were infiltrated with a high-density cell suspension, incubated in medium for proliferation of myoblasts prior to further culture in fusion medium to induce differentiation and formation of multinucleated myotubes. This resulted in a parallel arrangement of myotubes within the pore structures. CS with either proliferating cells or with myotubes were grafted into the beds of excised anterior tibial muscles of immunodeficient host mice. The recipient mice were transgenic for enhanced green fluorescent protein (eGFP) to determine a host contribution to the regenerated muscle tissue. Histological analysis 14–50 days after surgery showed that donor muscle fibres had formed in situ with host contributions in the outer portions of the regenerates. The function of the regenerates was assessed by direct electrical stimulation which resulted in the generation of mechanical force. Our study demonstrated that biodegradable CS with parallel pores support the formation of oriented muscle fibres and are compatible with force generation in regenerated muscle.
Collapse
Affiliation(s)
- V Kroehne
- Developmental Biology & Molecular Pathology, Bielefeld University, Bielefeld, Germany.
| | | | | | | | | | | |
Collapse
|
23
|
Bouchentouf M, Benabdallah BF, Rousseau J, Schwartz LM, Tremblay JP. Induction of Anoikis following myoblast transplantation into SCID mouse muscles requires the Bit1 and FADD pathways. Am J Transplant 2007; 7:1491-505. [PMID: 17511679 DOI: 10.1111/j.1600-6143.2007.01830.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Seventy-five percent of the myoblasts transplanted in the mouse muscle die during the first 4 days following transplantation. The purpose of this study was to determine if anoikis plays a role in this phenomenon. Survival and proliferation of myoblasts in vitro were determined by Hoescht-PI labeling and cell counts respectively. In vivo cell survival and proliferation were quantified by injecting human male myoblasts labeled with (14)C-thymidine in SCID mouse muscles. Survival and proliferation of the transplanted myoblasts were evaluated by scintigraphy and quantitative PCR of human Y chromosomal DNA. Inclusion of the extracellular matrix protein fibronectin enhanced transplanted myoblast survival by 1.7-fold while vitronectin improved their proliferation by 1.8-fold. Reductions in FADD and Bit1 expression reduced anoikis in vitro and improved the injected myoblast survival in vivo. Ectopic expression of the anti-apoptotic protein Bcl-2 completely abolished myoblast anoikis in vitro and enhanced cell survival by 3.1-fold in vivo. Cell death following transplantation appears to me mediated in part by anoikis. Inclusion of extracellular matrix proteins enhanced both survival and proliferation. Reduced expression of the proapoptotic proteins Bit1 and FADD or overexpression of Bcl-2 improved myoblast survival.
Collapse
Affiliation(s)
- M Bouchentouf
- Department of Human Genetics, CHUQ-CHUL, Laval University, Ste-Foy, Canada
| | | | | | | | | |
Collapse
|
24
|
Abstract
There are a number of promising cell therapy products under development for the treatment of heart failure, whether due to myocardial infarction or cardiomyopathy. Looking forward beyond current products in development, there are a multitude of possibilities that hold significant promise; however, cell-based therapies present challenges that are unique to this platform. Results from transplant studies can often be misleading and need to be interpreted in the context of fundamental biologic properties of cells and development. Provided here is a summary of the current and future developments in the field of cell therapy for cardiac regeneration along with some critical insights to interpret the multitude of studies recently undertaken. Summarized are both clinical and preclinical studies that should serve as a useful entrée into this exciting new field of therapeutic development.
Collapse
Affiliation(s)
- Nabil Dib
- Arizona Heart Institute, Phoenix, AZ 85006, USA.
| | | |
Collapse
|
25
|
Esmaeili F, Tiraihi T, Movahedin M, Mowla SJ. Selegiline Induces Neuronal Phenotype and Neurotrophins Expression in Embryonic Stem Cells. Rejuvenation Res 2006; 9:475-84. [PMID: 17105388 DOI: 10.1089/rej.2006.9.475] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The antiaging effect of selegiline was reported by several investigators; therefore, there is a growing interest in the potential use of stem cell therapy in aging. In this investigation, selegiline was used to induce neuronal differentiation in undifferentiated pluripotent embryonic stem cells (ESCs). The results show that selegiline can induce neuronal phenotype associated with neurotrophic factor expression. Morphologic and immunohistochemical techniques were used to evaluate the differentiation of the CCE cells, Cresyl violet for the morphologic study, anti-synaptophysin and antityrosine hydroxylase antibodies for characterizing the neuronal phenotype of ESCs, and RT-PCR to study the neurotrophins. The results showed that selegiline can induce dose-dependent ESC differentiation into neurons. Moreover, selegiline can induce neurotrophin expression. This study suggests the potential use of combined selegiline and stem cell therapy to improve deficits in neurodegenerative diseases in aging.
Collapse
Affiliation(s)
- Fariba Esmaeili
- Department of Anatomical Sciences, School of Medical Sciences, Tarbiat Modarres University, Tehran, Iran
| | | | | | | |
Collapse
|
26
|
Ozeki N, Lim M, Yao CC, Tolar M, Kramer RH. alpha7 integrin expressing human fetal myogenic progenitors have stem cell-like properties and are capable of osteogenic differentiation. Exp Cell Res 2006; 312:4162-80. [PMID: 17054947 PMCID: PMC2766282 DOI: 10.1016/j.yexcr.2006.09.017] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2006] [Revised: 09/13/2006] [Accepted: 09/15/2006] [Indexed: 10/24/2022]
Abstract
During muscle development, precursor cells fuse to form myofibers. Following injury in adult muscle, quiescent satellite cells become activated to regenerate muscle in a fashion similar to fetal development. Recent studies indicate that murine skeletal myoblasts can differentiate along multiple cell lineages including the osteoblastic pathway. However, little is known about the multipotency of human myogenic cells. Here, we isolate myogenic precursor cells from human fetal and adult muscle by sorting for the laminin-binding alpha7 integrin and demonstrate their differentiation potential and alteration in adhesive behavior. The alpha7-positive human fetal progenitors were efficient at forming myotubes and a majority expressed known muscle markers including M-cadherin and c-Met, but were heterogeneous for desmin and MyoD expression. To test their pluripotent differentiation potential, enriched populations of alpha7-positive fetal cells were subjected to inductive protocols. Although the myoblasts appeared committed to a muscle lineage, they could be converted to differentiate along the osteoblastic pathway in the presence of BMP-2. Interestingly, osteogenic cells showed altered adhesion and migratory activity that reflected growth factor-induced changes in integrin expression. These results indicate that alpha7-expressing fetal myoblasts are capable of differentiation to osteoblast lineage with a coordinated switch in integrin profiles and may represent a mechanism that promotes homing and recruitment of myogenic stem cells for tissue repair and remodeling.
Collapse
Affiliation(s)
- Nobuaki Ozeki
- Department of Cell and Tissue Biology, University of California at San Francisco, San Francisco, CA 94143-0640, USA
- Department of Endodontics, School of Dentistry, Aichigakuin University, 2–11 Suemori-dori Chikusa-ku, Nagoya, 464–8651, Japan
| | - Moon Lim
- Department of Cell and Tissue Biology, University of California at San Francisco, San Francisco, CA 94143-0640, USA
| | - Chung-Chen Yao
- Department of Cell and Tissue Biology, University of California at San Francisco, San Francisco, CA 94143-0640, USA
| | - Mirek Tolar
- Department of Cell and Tissue Biology, University of California at San Francisco, San Francisco, CA 94143-0640, USA
| | - Randall H. Kramer
- Department of Cell and Tissue Biology, University of California at San Francisco, San Francisco, CA 94143-0640, USA
| |
Collapse
|
27
|
Stern-Straeter J, Bach AD, Stangenberg L, Foerster VT, Horch RE, Stark GB, Beier JP. Impact of electrical stimulation on three-dimensional myoblast cultures - a real-time RT-PCR study. J Cell Mol Med 2006; 9:883-92. [PMID: 16364197 PMCID: PMC6740088 DOI: 10.1111/j.1582-4934.2005.tb00386.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Several focal skeletal muscle diseases, including tumours and trauma lead to a limited loss of functional muscle tissue. There is still no suitable clinical approach for treating such defects. A promising approach could be the tissue engineering of skeletal muscle. However, a clinically reliable differentiation stimulus for three-dimensional (3-D) cultures is necessary for this process, and this condition has not yet been established. In order to quantify and analyze the differentiation potential of electrical cell stimulation, primary myoblasts were stimulated within a 3-D fibrin- matrix. Gene expression of MyoD, myogenin and AChR-epsilon were measured by real-time RT-PCR over a time period of eight days, showing immediate down-regulation of all marker genes. For tissue engineering approaches, cell multiplication is crucial for acquisition of sufficient tissue volumes for reconstruction. Therefore, all experiments were performed with high and low passaged myoblasts, demonstrating higher transcript rates of marker genes in lowpassage cells. Our findings strongly suggest a reconsideration of electrical stimulation in muscle tissue engineering.
Collapse
Affiliation(s)
- J Stern-Straeter
- Department of Plastic and Hand Surgery, Tissue Engineering Laboratory University of Freiburg Medical Center, Germany
| | | | | | | | | | | | | |
Collapse
|
28
|
Becciolini L, Meacci E, Donati C, Cencetti F, Rapizzi E, Bruni P. Sphingosine 1-phosphate inhibits cell migration in C2C12 myoblasts. Biochim Biophys Acta Mol Cell Biol Lipids 2006; 1761:43-51. [PMID: 16510307 DOI: 10.1016/j.bbalip.2006.01.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2005] [Revised: 01/11/2006] [Accepted: 01/11/2006] [Indexed: 11/21/2022]
Abstract
This study shows that sphingosine 1-phosphate (S1P) exerts an anti-migratory action in C2C12 myoblasts by reducing directional cell motility and fully abrogating the chemotactic response to insulin-like growth factor-1. The anti-migratory response to S1P required ligation to S1P(2), being attenuated in myoblasts where the receptor was down-regulated by specific antisense oligodeoxyribonucleotides or small interfering RNA (siRNA) and conversely potentiated in S1P(2)-overexpressing myoblasts. The investigation of RhoA and Rac GTPases, critically implicated in cell motility regulation, demonstrated that RhoA was rapidly activated by S1P, while Rac1 was unaffected within the first 5 min but stimulated thereafter. RhoA, but not Rac activation, was identified as a S1P(2)-dependent pathway in experiments in which receptor expression was attenuated by siRNA treatment or up-regulated by S1P(2)-encoding plasmid transfection. Finally, by expression of the dominant negative mutant of RhoA, the GTPase was found implicated in the anti-migratory action of S1P, whereas modulation of Rac1 functionality unaffected the anti-chemotactic effect of S1P, ruling out a role for this protein in the biological response. Since S1P was previously shown to inhibit myoblast proliferation and stimulate myogenesis, the here identified novel biological activity is in favour of a complex physiological role of the sphingolipid in the process of muscle repair.
Collapse
Affiliation(s)
- Laura Becciolini
- Dipartimento di Scienze Biochimiche, Università degli Studi di Firenze, Viale G.B. Morgagni 50, 50134 Firenze, Italy
| | | | | | | | | | | |
Collapse
|
29
|
Messina A, Bortolotto SK, Cassell OCS, Kelly J, Abberton KM, Morrison WA. Generation of a vascularized organoid using skeletal muscle as the inductive source. FASEB J 2005; 19:1570-2. [PMID: 16014398 DOI: 10.1096/fj.04-3241fje] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The technology required for creating an in vivo microenvironment and a neovasculature that can grow with and service new tissue is lacking, precluding the possibility of engineering complex three-dimensional organs. We have shown that when an arterio-venous (AV) loop is constructed in vivo in the rat groin, and placed inside a semisealed chamber, an extensive functional vasculature is generated. To test whether this unusually angiogenic environment supports the survival and growth of implanted tissue or cells, we inserted various preparations of rat and human skeletal muscle. We show that after 6 weeks incubation of muscle tissue, the chamber filled with predominantly well-vascularized recipient-derived adipose tissue, but some new donor-derived skeletal muscle and connective tissue were also evident. When primary cultured myoblasts were inserted into the chamber with the AV loop, they converted to mature striated muscle fibers. Furthermore, we identify novel adipogenesis-inducing properties of skeletal muscle. This represents the first report of a specific three-dimensional tissue grown on its own vascular supply.
Collapse
Affiliation(s)
- Aurora Messina
- Bernard O'Brien Institute of Microsurgery, St. Vincent's Hospital Melbourne, Fitzroy, Victoria, Australia.
| | | | | | | | | | | |
Collapse
|
30
|
Shah R, Sinanan ACM, Knowles JC, Hunt NP, Lewis MP. Craniofacial muscle engineering using a 3-dimensional phosphate glass fibre construct. Biomaterials 2005; 26:1497-505. [PMID: 15522751 DOI: 10.1016/j.biomaterials.2004.04.049] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2003] [Accepted: 04/30/2004] [Indexed: 11/22/2022]
Abstract
The current technique to replace missing craniofacial skeletal muscle is the surgical transfer of local or free flaps. This is associated with donor site morbidity, possible tissue rejection and limited supply. The alternative is to engineer autologous skeletal muscle in vitro, which can then be re-implanted into the patient. A variety of biomaterials have been used to engineer skeletal muscle with limited success. This study investigated the use of phosphate-based glass fibres as a potential scaffold material for the in vitro engineering of craniofacial skeletal muscle. Human masseter (one of the muscles of mastication)--derived cell cultures were used to seed the glass fibres, which were arranged into various configurations. Growth factors and matrix components were to used to manipulate the in vitro environment. Outcome was determined with the aid of microscopy, time-lapse footage, immunofluorescence imaging and CyQUANT proliferation, creatine kinase and protein assays. A 3-dimensional mesh arrangement of the glass fibres was the best at encouraging cell attachment and proliferation. In addition, increasing the density of the seeded cells and using Matrigel and insulin-like growth factor I enhanced the formation of prototypic muscle fibres. In conclusion, phosphate-based glass fibres can support the in vitro engineering of human craniofacial muscle.
Collapse
Affiliation(s)
- R Shah
- Division of Biomaterials and Tissue Engineering, Eastman Dental Institute, University College London, 256 Gray's Inn Road, London, WC1X 8LD, UK
| | | | | | | | | |
Collapse
|
31
|
Brimah K, Ehrhardt J, Mouly V, Butler-Browne GS, Partridge TA, Morgan JE. Human muscle precursor cell regeneration in the mouse host is enhanced by growth factors. Hum Gene Ther 2005; 15:1109-24. [PMID: 15610611 DOI: 10.1089/hum.2004.15.1109] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The aim of this study was to optimize human muscle formation in vivo from implanted human muscle precursor cells. We transplanted donor muscle precursor cells (MPCs) prepared from postnatal or fetal human muscle into immunodeficient host mice and showed that irradiation of host muscle significantly enhanced muscle formation by donor cells. The amount of donor muscle formed in cryodamaged host muscle was increased by exposure of donor cells to growth factors before their implantation into injured host muscle. Insulin-like growth factor type I (IGF-I) significantly increased the amount of muscle formed by postnatal human muscle cells, but not by fetal human MPCs. However, treatment of fetal muscle cells with IGF-I, in combination with basic fibroblast growth factor and plasmin, significantly increased the amount of donor muscle formed. In vivo, human MPCs formed mosaic human-mouse muscle fibers, in which each human myonucleus was associated with a zone of human sarcolemmal protein spectrin.
Collapse
Affiliation(s)
- K Brimah
- Muscle Cell Biology Group, MRC Clinical Sciences Centre, Imperial College, London W12 ONN, United Kingdom
| | | | | | | | | | | |
Collapse
|
32
|
Suzuki K, Murtuza B, Beauchamp JR, Brand NJ, Barton PJR, Varela-Carver A, Fukushima S, Coppen SR, Partridge TA, Yacoub MH. Role of interleukin-1beta in acute inflammation and graft death after cell transplantation to the heart. Circulation 2005; 110:II219-24. [PMID: 15364866 DOI: 10.1161/01.cir.0000138388.55416.06] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Poor survival of grafted cells is a major factor hindering the therapeutic effect of cell transplantation; however, the causes of cell death remain unclear. We hypothesized that interleukin-1beta (IL-1beta) might play a role in the acute inflammatory response and graft death after cell transplantation and that inhibition of IL-1beta might improve graft survival. METHODS AND RESULTS 14C-labeled male skeletal muscle precursor cells were implanted into female mouse hearts by direct intramuscular injection. The amount of 14C-label provides an estimate of the surviving cell number, whereas the amount of male-specific Smcy gene measured by polymerase chain reaction indicates the total (surviving+proliferated) number of donor-derived cells. At 10 minutes after implantation, 44.8+/-2.4% of the grafted cells survived and this steadily decreased to 14.6+/-1.1% by 24 hours, and to 7.9+/-0.6% by 72 hours (n=6 in each point). Proliferation of the surviving cells, which began after 24 hours, resulted in an increase in the total cell number from 15.5+/-0.8% at 24 hours to 24.4+/-1.6% at 72 hours. Acute inflammation was prominent at 24 hours and was reduced by 72 hours, in parallel with IL-1beta expression. Administration of anti-IL-1beta antibody improved graft survival at both 24 (25.6+/-1.6%) and 72 hours (14.8+/-1.1%) and resulted in a 2-fold increase in the total cell number at 72 hours (45.8+/-2.4%). The effects of IL-1beta inhibition corresponded with a reduced inflammatory response. CONCLUSIONS IL-1beta is involved in acute inflammation and graft death after direct intramyocardial cell transplantation. Targeted inhibition of IL-1beta may be a useful strategy to improve graft survival.
Collapse
Affiliation(s)
- Ken Suzuki
- Cell and Gene Therapy Group, Harefield Heart Science Centre, National Heart and Lung Institute, Imperial College London, Harefield, Middlesex, United Kingdom.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Brennan FR, Shaw L, Wing MG, Robinson C. Preclinical safety testing of biotechnology-derived pharmaceuticals: understanding the issues and addressing the challenges. Mol Biotechnol 2004; 27:59-74. [PMID: 15122047 DOI: 10.1385/mb:27:1:59] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The unique and complex nature of biotechnology-derived pharmaceuticals has meant that it is often not possible to follow the conventional safety testing programs used for chemicals, and hence they are evaluated on a case-by-case basis. Nonclinical safety testing programs must be rationally designed with a strong scientific understanding of the product, including its method of manufacture, purity, sequence, structure, species specificity, pharmacological and immunological effects, and intended clinical use. This knowledge, coupled with a firm understanding of the regulatory requirements for particular product types, will ensure that the most sensitive and regulatory-compliant test systems are used to optimize the chances of gaining regulatory approval for clinical testing or marketing authorization in the shortest possible time frame.
Collapse
Affiliation(s)
- Frank R Brennan
- Huntingdon Life Sciences, Woolley Rd, Alconbury, Huntingdon, Cambridgeshire PE28 4HS, UK.
| | | | | | | |
Collapse
|
34
|
Suzuki K, Murtuza B, Beauchamp JR, Smolenski RT, Varela-Carver A, Fukushima S, Coppen SR, Partridge TA, Yacoub MH. Dynamics and mediators of acute graft attrition after myoblast transplantation to the heart. FASEB J 2004; 18:1153-5. [PMID: 15155562 DOI: 10.1096/fj.03-1308fje] [Citation(s) in RCA: 142] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Survival and proliferation of skeletal myoblasts within the cardiac environment are crucial to the therapeutic efficacy of myoblast transplantation to the heart. We have analyzed the early dynamics of myoblasts implanted into the myocardium and investigated the mechanisms underlying graft attrition. At 10 min after implantation of [14C]thymidine-labeled male myoblasts into female mice hearts, 14C measurement showed that 39.2 +/- 3.0% of the grafted cells survived, and this steadily decreased to 16.0 +/- 1.7% by 24 h and to 7.4 +/- 0.9% by 72 h. PCR of male-specific Smcy gene calculated that the total (surviving plus proliferated) number of donor-derived cells was 18.3 +/- 1.6 and 23.3 +/- 1.3% at 24 and 72 h, respectively, indicating that proliferation of the surviving cells began after 24 h. Acute inflammation became prominent by 24 h and was reduced by 72 h as indicated by myeloperoxidase activity and histological findings. Multiplex RT-PCR revealed corresponding changes in IL-1beta, TGF-beta, IL-6, and TNF-alpha expression. Treatment with CuZn-superoxide dismutase attenuated the initial rapid death and resulted in enhanced cell numbers afterward, giving a twofold increased total number at 72 h compared with the nontreatment. This effect was associated with reduced inflammatory response, suggesting a causative role for superoxide in the initial rapid graft death and subsequent inflammation. These data describe the early dynamics of myoblasts implanted into the myocardium and suggest that initial oxidative stress and following inflammatory response may be important mechanisms contributing to acute graft attrition, both of which could be potential therapeutic targets to improve the efficiency of cell transplantation to the heart.
Collapse
Affiliation(s)
- Ken Suzuki
- Harefield Heart Science Centre, National Heart and Lung Institute, Harefield, Middlesex, UK.
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Chan YS, Li Y, Foster W, Horaguchi T, Somogyi G, Fu FH, Huard J. Antifibrotic effects of suramin in injured skeletal muscle after laceration. J Appl Physiol (1985) 2003; 95:771-80. [PMID: 12730151 DOI: 10.1152/japplphysiol.00915.2002] [Citation(s) in RCA: 111] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Muscle injuries are very common in traumatology and sports medicine. Although muscle tissue can regenerate postinjury, the healing process is slow and often incomplete; complete recovery after skeletal muscle injury is hindered by fibrosis. Our studies have shown that decreased fibrosis could improve muscle healing. Suramin has been found to inhibit transforming growth factor (TGF)-beta1 expression by competitively binding to the growth factor receptor. We conducted a series of tests to determine the antifibrotic effects of suramin on muscle laceration injuries. Our results demonstrate that suramin (50 microg/ml) can effectively decrease fibroblast proliferation and fibrotic-protein expression (alpha-smooth muscle actin) in vitro. In vivo, direct injection of suramin (2.5 mg) into injured murine muscle resulted in effective inhibition of muscle fibrosis and enhanced muscle regeneration, which led to efficient functional muscle recovery. These results support our hypothesis that prevention of fibrosis could enhance muscle regeneration, thereby facilitating more efficient muscle healing. This study could significantly contribute to the development of strategies to promote efficient muscle healing and functional recovery.
Collapse
Affiliation(s)
- Yi-Sheng Chan
- Department of Orthopaedic Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA 15213-2583, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
López-Conejo MT, Olmo N, Turnay J, López De Silanes I, Lizarbe MA. Interaction of fibronectin with human colon adenocarcinoma cells: effect on the in vivo tumorigenic capacity. Oncology 2002; 62:371-80. [PMID: 12138246 DOI: 10.1159/000065070] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Fibronectin (FN) modulates the behavior of the poorly differentiated, human colon adenocarcinoma-derived BCS-TC2 cells by promoting adhesion through the alpha(5)beta(1)-integrin, as this effect is blocked by anti-alpha(5) and beta(1 )chain antibodies. BCS-TC2 cells are not tumorigenic in vivo, but are able to form tumors when coinjected with FN in nude mice. From these tumors, a tumorigenic cell subline (BCS-TC2.FN) was established. In vivo passaging of BCS-TC2.FN cells in the absence of FN allowed the selection of another tumorigenic subline (BCS-TC2.FN2). The new sublines are characterized by: (1) increased differentiation, (2) slightly higher adhesion to FN, and (3) a higher uptake of [(3)H]thymidine, less dependent on the presence of serum or FN. No significant modifications in alpha(5)-chain surface levels were observed in the tumor-derived sublines, suggesting that the amount of alpha(5)beta(1)-integrin is not related to tumorigenicity. Within the heterogeneous parental cells, FN seems to favor the selection of a cell subpopulation that presents phenotypic and genotypic alterations that are stably maintained throughout in vitro culture and in vivo passaging. These cell lines constitute a model system that may help to extend our knowledge on the events underlying tumor progression and malignancy of colorectal cancer, and the influence of extracellular matrix components and their receptors in these processes.
Collapse
Affiliation(s)
- M Teresa López-Conejo
- Departamento de Bioquímica y Biología Molecular, Facultad de Química, Universidad Complutense, Madrid, Spain
| | | | | | | | | |
Collapse
|
37
|
|