151
|
de Melo CAV, Alves AN, Terena SML, Fernandes KPS, Nunes FD, da Silva DDFT, Bussadori SK, Deana AM, Mesquita-Ferrari RA. Light-emitting diode therapy increases collagen deposition during the repair process of skeletal muscle. Lasers Med Sci 2016; 31:531-8. [PMID: 26873500 DOI: 10.1007/s10103-016-1888-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 01/19/2016] [Indexed: 01/08/2023]
Abstract
This study analyzed the effects of light-emitting diode (LED) therapy on the morphology of muscle tissue as well as collagen remodeling and matrix metalloproteinase 2 (MMP-2) activity in the skeletal muscle of rats following acute injury. Wistar rats were divided into four groups: (1) control, (2) sham, (3) untreated cryoinjury, and (4) cryoinjury treated with LED. Cryoinjury was induced by two applications of a metal probe cooled in liquid nitrogen directly onto the belly of the tibialis anterior muscle. For treatment, the LED equipment (wavelength 850 nm, output power 30 mW, and total energy 3.2 J) was used daily. The study periods were 1, 3, and 7 days after cryoinjury. Morphological aspects were evaluated through hematoxylin-eosin staining. The amount of collagen fibers was evaluated using Picro Sirius Red staining under polarized light. The gelatinase activity of MMP-2 was evaluated using zymography. The results showed significant reductions in inflammatory infiltrate after 3 days and an increased number of immature muscle fibers after 7 days. Furthermore, treatment induced a reduction in the gelatinolytic activity of MMP-2 after 1, 3, and 7 days in comparison to the untreated injury groups and increased the collagen deposition after 3 and 7 days in the treated groups. LED therapy at 850 nm induced a significant reduction in inflammation, decreased MMP-2 activity, and increased the amount of immature muscle and collagen fibers during the muscle repair process following acute injury.
Collapse
Affiliation(s)
- Claudia Aparecida Viana de Melo
- Postgraduate Program in Rehabilitation Sciences, Universidade Nove de Julho (UNINOVE), Rua Vergueiro, 235/249, Liberdade, CEP 01504-001, São Paulo, SP, Brazil
| | - Agnelo Neves Alves
- Postgraduate Program in Rehabilitation Sciences, Universidade Nove de Julho (UNINOVE), Rua Vergueiro, 235/249, Liberdade, CEP 01504-001, São Paulo, SP, Brazil
| | - Stella Maris Lins Terena
- Postgraduate Program in Biophotonics Applied to Health Sciences, Universidade Nove de Julho (UNINOVE), São Paulo, SP, Brazil
| | - Kristianne Porta Santos Fernandes
- Postgraduate Program in Rehabilitation Sciences, Universidade Nove de Julho (UNINOVE), Rua Vergueiro, 235/249, Liberdade, CEP 01504-001, São Paulo, SP, Brazil
- Postgraduate Program in Biophotonics Applied to Health Sciences, Universidade Nove de Julho (UNINOVE), São Paulo, SP, Brazil
| | - Fábio Daumas Nunes
- Department of Oral Pathology, School of Dentistry, University of São Paulo, São Paulo, SP, Brazil
| | | | - Sandra Kalil Bussadori
- Postgraduate Program in Rehabilitation Sciences, Universidade Nove de Julho (UNINOVE), Rua Vergueiro, 235/249, Liberdade, CEP 01504-001, São Paulo, SP, Brazil
- Postgraduate Program in Biophotonics Applied to Health Sciences, Universidade Nove de Julho (UNINOVE), São Paulo, SP, Brazil
| | - Alessandro Melo Deana
- Postgraduate Program in Biophotonics Applied to Health Sciences, Universidade Nove de Julho (UNINOVE), São Paulo, SP, Brazil
| | - Raquel Agnelli Mesquita-Ferrari
- Postgraduate Program in Rehabilitation Sciences, Universidade Nove de Julho (UNINOVE), Rua Vergueiro, 235/249, Liberdade, CEP 01504-001, São Paulo, SP, Brazil.
- Postgraduate Program in Biophotonics Applied to Health Sciences, Universidade Nove de Julho (UNINOVE), São Paulo, SP, Brazil.
| |
Collapse
|
152
|
Domingues-Faria C, Vasson MP, Goncalves-Mendes N, Boirie Y, Walrand S. Skeletal muscle regeneration and impact of aging and nutrition. Ageing Res Rev 2016; 26:22-36. [PMID: 26690801 DOI: 10.1016/j.arr.2015.12.004] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 12/01/2015] [Accepted: 12/07/2015] [Indexed: 01/08/2023]
Abstract
After skeletal muscle injury a regeneration process takes place to repair muscle. Skeletal muscle recovery is a highly coordinated process involving cross-talk between immune and muscle cells. It is well known that the physiological activities of both immune cells and muscle stem cells decline with advancing age, thereby blunting the capacity of skeletal muscle to regenerate. The age-related reduction in muscle repair efficiency contributes to the development of sarcopenia, one of the most important factors of disability in elderly people. Preserving muscle regeneration capacity may slow the development of this syndrome. In this context, nutrition has drawn much attention: studies have demonstrated that nutrients such as amino acids, n-3 polyunsaturated fatty acids, polyphenols and vitamin D can improve skeletal muscle regeneration by targeting key functions of immune cells, muscle cells or both. Here we review the process of skeletal muscle regeneration with a special focus on the cross-talk between immune and muscle cells. We address the effect of aging on immune and skeletal muscle cells involved in muscle regeneration. Finally, the mechanisms of nutrient action on muscle regeneration are described, showing that quality of nutrition may help to preserve the capacity for skeletal muscle regeneration with age.
Collapse
|
153
|
Impaired Ribosome Biogenesis and Skeletal Muscle Growth in a Murine Model of Inflammatory Bowel Disease. Inflamm Bowel Dis 2016; 22:268-78. [PMID: 26588088 DOI: 10.1097/mib.0000000000000616] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
BACKGROUND Inflammation is a factor potentially underpinning skeletal muscle mass. Intestinal-derived inflammation in inflammatory bowel disease (IBD) results in loss of muscle mass; however, the underlying mechanism is unclear. The interleukin 10 gene-deficient (Il10-/-) mouse is a genetically modified animal model of IBD that can be used to study the effect of intestinal-derived inflammation on muscles. METHODS Il10-/- and C57BL/6 wild-type (WT) mice were inoculated with intestinal bacteria to induce colon inflammation at the fifth week of age. Skeletal muscles were collected between 7 and 14 weeks of age for analysis of muscle weight, myofiber cross-sectional area (CSA), and molecular markers of inflammation and anabolism pathways, with a focus on ribosome biogenesis. RESULTS Il10-/- animals that developed colon inflammation had a marked increase in muscle immunoglobulin G (IgG) compared with WT. Inflamed Il10-/- animals had impaired muscle mass gain and smaller myofiber CSA. Intramuscular IgG deposition negatively correlated with muscle mass. After the onset of muscle inflammation, Il10-/- mice had decreased levels of total and ribosomal RNAs (45S, 28S, 18S, and 5.8S rRNAs). Inflammation inversely correlated with muscle levels of total RNA and 28S rRNA which in turn positively correlated with muscle mass. The abundance of growth-related proteins (p70S6K and upstream binding factor, UBF) was decreased in Il10-/- mice. CONCLUSIONS Muscle inflammation and associated decline of ribosome biogenesis lead to muscle growth impairment in Il10-/- mice. This may have implications for maintenance of muscle mass in conditions associated with chronic intestinal-derived inflammation.
Collapse
|
154
|
Dearth CL, Slivka PF, Stewart SA, Keane TJ, Tay JK, Londono R, Goh Q, Pizza FX, Badylak SF. Inhibition of COX1/2 alters the host response and reduces ECM scaffold mediated constructive tissue remodeling in a rodent model of skeletal muscle injury. Acta Biomater 2016; 31:50-60. [PMID: 26612417 DOI: 10.1016/j.actbio.2015.11.043] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 11/19/2015] [Accepted: 11/19/2015] [Indexed: 12/26/2022]
Abstract
Extracellular matrix (ECM) has been used as a biologic scaffold material to both reinforce the surgical repair of soft tissue and serve as an inductive template to promote a constructive tissue remodeling response. Success of such an approach is dependent on macrophage-mediated degradation and remodeling of the biologic scaffold. Macrophage phenotype during these processes is a predictive factor of the eventual remodeling outcome. ECM scaffolds have been shown to promote an anti-inflammatory or M2-like macrophage phenotype in vitro that includes secretion of downstream products of cycolooxygenases 1 and 2 (COX1/2). The present study investigated the effect of a common COX1/2 inhibitor (Aspirin) on macrophage phenotype and tissue remodeling in a rodent model of ECM scaffold treated skeletal muscle injury. Inhibition of COX1/2 reduced the constructive remodeling response by hindering myogenesis and collagen deposition in the defect area. The inhibited response was correlated with a reduction in M2-like macrophages in the defect area. The effects of Aspirin on macrophage phenotype were corroborated using an established in vitro macrophage model which showed a reduction in both ECM induced prostaglandin secretion and expression of a marker of M2-like macrophages (CD206). These results raise questions regarding the common peri-surgical administration of COX1/2 inhibitors when biologic scaffold materials are used to facilitate muscle repair/regeneration. STATEMENT OF SIGNIFICANCE COX1/2 inhibitors such as nonsteroidal anti-inflammatory drugs (NSAIDs) are routinely administered post-surgically for analgesic purposes. While COX1/2 inhibitors are important in pain management, they have also been shown to delay or diminish the healing process, which calls to question their clinical use for treating musculotendinous injuries. The present study aimed to investigate the influence of a common NSAID, Aspirin, on the constructive remodeling response mediated by an ECM scaffold (UBM) in a rat skeletal muscle injury model. The COX1/2 inhibitor, Aspirin, was found to mitigate the ECM scaffold-mediated constructive remodeling response both in an in vitro co-culture system and an in vivo rat model of skeletal muscle injury. The results presented herein provide data showing that NSAIDs may significantly alter tissue remodeling outcomes when a biomaterial is used in a regenerative medicine/tissue engineering application. Thus, the decision to prescribe NSAIDs to manage the symptoms of inflammation post-ECM scaffold implantation should be carefully considered.
Collapse
|
155
|
Cardoso ESB, Santana TA, Diniz PBF, Montalvão MM, Bani CC, Thomazzi SM. Thymol accelerates the recovery of the skeletal muscle of mice injured with cardiotoxin. ACTA ACUST UNITED AC 2016; 68:352-60. [PMID: 26817998 DOI: 10.1111/jphp.12520] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 12/13/2015] [Indexed: 11/30/2022]
Abstract
OBJECTIVES The aim of this study was to investigate the preventive effect of thymol in in vivo muscle inflammation and regeneration on cardiotoxin-induced injury. METHODS Mice were pretreated (p.o.) with thymol (10-100 mg/kg), and after 1 h, cardiotoxin (25 μM, 40 μl) was administrated into the gastrocnemius muscle. The quantification of the areas of inflammation and regeneration of muscle tissue (3, 7 and 10 days) in HE-stained slides as well as the count of total mast cells and different phenotypes of mast cells were made. Sirius red staining was used to analyse total collagen expression. KEY FINDINGS The pretreatment with thymol significantly reduced the area of inflammation (30 and 100 mg/kg) and increased the area of regeneration (100 mg/kg) 3 days after the cardiotoxin injection. Thymol at 30 and 100 mg/kg increased the area of collagen in 3 days and also decreased this area in 7 and 10 days, compared to the injured group. The pretreatment with thymol did not affect the number of total mast cells; however, it was able to change the number of mucosal mast cells within 10 days. CONCLUSIONS This study suggests that thymol ameliorates inflammatory response and accelerates regeneration in cardiotoxin-induced muscle injury.
Collapse
Affiliation(s)
- Eroneide S B Cardoso
- Department of Physiology, Federal University of Sergipe, São Cristóvão, Sergipe, Brazil
| | - Tayse A Santana
- Department of Physiology, Federal University of Sergipe, São Cristóvão, Sergipe, Brazil
| | | | - Monalisa M Montalvão
- Department of Morphology, Federal University of Sergipe, São Cristóvão, Sergipe, Brazil
| | - Cristiane C Bani
- Department of Morphology, Federal University of Sergipe, São Cristóvão, Sergipe, Brazil
| | - Sara M Thomazzi
- Department of Physiology, Federal University of Sergipe, São Cristóvão, Sergipe, Brazil
| |
Collapse
|
156
|
Chen T, Moore TM, Ebbert MTW, McVey NL, Madsen SR, Hallowell DM, Harris AM, Char RE, Mackay RP, Hancock CR, Hansen JM, Kauwe JS, Thomson DM. Liver kinase B1 inhibits the expression of inflammation-related genes postcontraction in skeletal muscle. J Appl Physiol (1985) 2016; 120:876-88. [PMID: 26796753 DOI: 10.1152/japplphysiol.00727.2015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 01/20/2016] [Indexed: 01/06/2023] Open
Abstract
Skeletal muscle-specific liver kinase B1 (LKB1) knockout mice (skmLKB1-KO) exhibit elevated mitogen-activated protein kinase (MAPK) signaling after treadmill running. MAPK activation is also associated with inflammation-related signaling in skeletal muscle. Since exercise can induce muscle damage, and inflammation is a response triggered by damaged tissue, we therefore hypothesized that LKB1 plays an important role in dampening the inflammatory response to muscle contraction, and that this may be due in part to increased susceptibility to muscle damage with contractions in LKB1-deficient muscle. Here we studied the inflammatory response and muscle damage with in situ muscle contraction or downhill running. After in situ muscle contractions, the phosphorylation of both NF-κB and STAT3 was increased more in skmLKB1-KO vs. wild-type (WT) muscles. Analysis of gene expression via microarray and RT-PCR shows that expression of many inflammation-related genes increased after contraction only in skmLKB1-KO muscles. This was associated with mild skeletal muscle fiber membrane damage in skmLKB1-KO muscles. Gene markers of oxidative stress were also elevated in skmLKB1-KO muscles after contraction. Using the downhill running model, we observed significantly more muscle damage after running in skmLKB1-KO mice, and this was associated with greater phosphorylation of both Jnk and STAT3 and increased expression of SOCS3 and Fos. In conclusion, we have shown that the lack of LKB1 in skeletal muscle leads to an increased inflammatory state in skeletal muscle that is exacerbated by muscle contraction. Increased susceptibility of the muscle to damage may underlie part of this response.
Collapse
Affiliation(s)
- Ting Chen
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah
| | - Timothy M Moore
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah
| | - Mark T W Ebbert
- Department of Biology, Brigham Young University, Provo, Utah
| | - Natalie L McVey
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah
| | - Steven R Madsen
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah
| | - David M Hallowell
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah
| | - Alexander M Harris
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah
| | - Robin E Char
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah
| | - Ryan P Mackay
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah
| | - Chad R Hancock
- Department of Nutrition, Dietetics and Food Science, Brigham Young University, Provo, Utah; and
| | - Jason M Hansen
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah
| | - John S Kauwe
- Department of Biology, Brigham Young University, Provo, Utah
| | - David M Thomson
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah;
| |
Collapse
|
157
|
Kozakowska M, Pietraszek-Gremplewicz K, Jozkowicz A, Dulak J. The role of oxidative stress in skeletal muscle injury and regeneration: focus on antioxidant enzymes. J Muscle Res Cell Motil 2016; 36:377-93. [PMID: 26728750 PMCID: PMC4762917 DOI: 10.1007/s10974-015-9438-9] [Citation(s) in RCA: 209] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 12/07/2015] [Indexed: 12/20/2022]
Abstract
Reactive oxygen species (ROS) are generated in skeletal muscle both during the rest and contractile activity. Myogenic cells are equipped with antioxidant enzymes, like superoxide dismutase, catalase, glutathione peroxidase, γ-glutamylcysteine synthetase and heme oxygenase-1. These enzymes not only neutralise excessive ROS, but also affect myogenic regeneration at several stages: influence post-injury inflammatory reaction, enhance viability and proliferation of muscle satellite cells and myoblasts and affect their differentiation. Finally, antioxidant enzymes regulate also processes accompanying muscle regeneration-induce angiogenesis and reduce fibrosis. Elevated ROS production was also observed in Duchenne muscular dystrophy (DMD), a disease characterised by degeneration of muscle tissue and therefore-increased rate of myogenic regeneration. Antioxidant enzymes are consequently considered as target for therapies counteracting dystrophic symptoms. In this review we present current knowledge regarding the role of oxidative stress and systems of enzymatic antioxidant defence in muscular regeneration after both acute injury and persistent muscular degeneration.
Collapse
Affiliation(s)
- Magdalena Kozakowska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland.
| | - Katarzyna Pietraszek-Gremplewicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland.
| | - Alicja Jozkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland.
| | - Jozef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland. .,Malopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland.
| |
Collapse
|
158
|
Localization and quantification of intramuscular damage using statistical parametric mapping and skeletal muscle parcellation. Sci Rep 2015; 5:18580. [PMID: 26689827 PMCID: PMC4686971 DOI: 10.1038/srep18580] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 11/23/2015] [Indexed: 01/27/2023] Open
Abstract
In the present study, we proposed an original and robust methodology which combines the spatial normalization of skeletal muscle images, the statistical parametric mapping (SPM) analysis and the use of a specific parcellation in order to accurately localize and quantify the extent of skeletal muscle damage within the four heads of the quadriceps femoris. T2 maps of thigh muscles were characterized before, two (D2) and four (D4) days after 40 maximal isometric electrically-evoked contractions in 25 healthy young males. On the basis of SPM analysis of coregistrated T2 maps, the alterations were similarly detected at D2 and D4 in the superficial and distal regions of the vastus medialis (VM) whereas the proportion of altered muscle was higher in deep muscle regions of the vastus lateralis at D4 (deep: 35 ± 25%, superficial: 23 ± 15%) as compared to D2 (deep: 18 ± 13%, superficial: 17 ± 13%). The present methodology used for the first time on skeletal muscle would be of utmost interest to detect subtle intramuscular alterations not only for the diagnosis of muscular diseases but also for assessing the efficacy of potential therapeutic interventions and clinical treatment strategies.
Collapse
|
159
|
Jönsson L, Dellenmark Blom M, Friberg L, Gatzinsky V, Holmquist O, Jennische E, Sandin A, Abrahamsson K. Macrophage Phenotype Is Associated With the Regenerative Response in Experimental Replacement of the Porcine Esophagus. Artif Organs 2015; 40:950-958. [PMID: 26667982 DOI: 10.1111/aor.12652] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
A porcine model for bridging circumferential defects in the intrathoracic esophagus has been developed in order to improve the treatment of children born with long-gap esophageal atresia. The aim of this study was to identify factors beneficial for tissue regeneration in the bridging area in this model and to describe the histological progression 20 days after replacement with a silicone-stented Biodesign mesh. Resection of 3 cm of intrathoracic esophagus and replacement with a bridging graft was performed in six newly weaned piglets. They were fed through a gastrostomy for 10 days, and then had probe formula orally for another 10 days prior to sacrifice. Two out of six piglets had stent loss prior to sacrifice. In the four piglets with the stent in place, a tissue tube, with visible muscle in the wall, was seen at sacrifice. Histology showed that the wall of the healing area was well organized with layers of inflammatory cells, in-growing vessels, and smooth muscle cells. CD163+ macrophages was seen toward the esophageal lumen. In the animals where the stent was lost, the bridging area was narrow, and histology showed a less organized structure in the bridging area without the presence of CD163+ macrophages. This study indicates that regenerative healing was seen in the porcine esophagus 20 days after replacement of a part of the intrathoracic esophagus with a silicone-stented Biodesign mesh, if the bridging graft is retained. If the graft is lost, the inflammatory pattern changes with invasion of proinflammatory, M1 macrophages in the entire wall, which seems to redirect the healing process toward scar formation.
Collapse
Affiliation(s)
- Linus Jönsson
- Institute of Clinical Sciences, Department of Pediatric Surgery, University of Gothenburg, Gothenburg, Sweden.
| | - Michaela Dellenmark Blom
- Institute of Clinical Sciences, Department of Pediatric Surgery, University of Gothenburg, Gothenburg, Sweden
| | - Lars Friberg
- Institute of Clinical Sciences, Department of Pediatric Surgery, University of Gothenburg, Gothenburg, Sweden
| | - Vladimir Gatzinsky
- Institute of Clinical Sciences, Department of Pediatric Surgery, University of Gothenburg, Gothenburg, Sweden
| | - Olof Holmquist
- Institute of Clinical Sciences, Department of Pediatric Surgery, University of Gothenburg, Gothenburg, Sweden
| | - Eva Jennische
- Institute of Biomedicine, Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Gothenburg, Sweden
| | - Anders Sandin
- Institute of Clinical Sciences, Department of Pediatric Surgery, University of Gothenburg, Gothenburg, Sweden
| | - Kate Abrahamsson
- Institute of Clinical Sciences, Department of Pediatric Surgery, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
160
|
Crupi A, Costa A, Tarnok A, Melzer S, Teodori L. Inflammation in tissue engineering: The Janus between engraftment and rejection. Eur J Immunol 2015; 45:3222-36. [DOI: 10.1002/eji.201545818] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 10/07/2015] [Accepted: 11/05/2015] [Indexed: 01/10/2023]
Affiliation(s)
- Annunziata Crupi
- Department of Fusion and Technologies for Nuclear Safety and Security; Diagnostic and Metrology (FSN-TECFIS-DIM), ENEA; Frascati-Rome Italy
- Fondazione San Raffaele; Ceglie Messapica Italy
| | - Alessandra Costa
- Department of Surgery; McGowan Institute; University of Pittsburgh Medical Center; Pittsburgh PA USA
| | - Attila Tarnok
- Department of Pediatric Cardiology; Heart Center GmbH Leipzig; and Translational Center for Regenerative Medicine; University Leipzig; Leipzig Germany
| | - Susanne Melzer
- Department of Pediatric Cardiology; Heart Center GmbH Leipzig; and Translational Center for Regenerative Medicine; University Leipzig; Leipzig Germany
| | - Laura Teodori
- Department of Fusion and Technologies for Nuclear Safety and Security; Diagnostic and Metrology (FSN-TECFIS-DIM), ENEA; Frascati-Rome Italy
- Fondazione San Raffaele; Ceglie Messapica Italy
| |
Collapse
|
161
|
von Pfeil DJF, Cummings BP, Loftus JP, Levine CB, Mann S, Downey RL, Griffitts C, Wakshlag JJ. Evaluation of plasma inflammatory cytokine concentrations in racing sled dogs. THE CANADIAN VETERINARY JOURNAL = LA REVUE VETERINAIRE CANADIENNE 2015; 56:1252-1256. [PMID: 26663920 PMCID: PMC4668826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
In human athletes significant changes in cytokine concentrations secondary to exercise have been observed. This prospective study evaluated the effect of a multi-day stage sled dog race on plasma concentrations of monocyte chemoattractant protein-1 (MCP-1), tumor necrosis factor-alpha (TNF-α), interleukin-2 (IL-2), interleukin-6 (IL-6), interleukin-8 (IL-8), and interleukin-10 (IL-10). Samples from 20 dogs were harvested prior to and on days 2 and 8 of an 8-day race. Exercise resulted in significantly decreased TNF-α and IL-8 as well as increases of MCP-1, IL-6, and IL-10 concentrations (P-value between 0.01 and < 0.0001 for all parameters). The proportion of values for IL-2 that were below the detection limit increased from 40% on day 0 to 75% on day 2 and decreased on day 8 to 40% (P = 0.04). Racing sled dogs show cytokine-concentration changes that are different from those in humans.
Collapse
|
162
|
Cheema N, Herbst A, McKenzie D, Aiken JM. Apoptosis and necrosis mediate skeletal muscle fiber loss in age-induced mitochondrial enzymatic abnormalities. Aging Cell 2015; 14:1085-93. [PMID: 26365892 PMCID: PMC4693455 DOI: 10.1111/acel.12399] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2015] [Indexed: 01/07/2023] Open
Abstract
Sarcopenia, the age‐induced loss of skeletal muscle mass and function, results from the contributions of both fiber atrophy and loss of myofibers. We have previously characterized sarcopenia in FBN rats, documenting age‐dependent declines in muscle mass and fiber number along with increased fiber atrophy and fibrosis in vastus lateralis and rectus femoris muscles. Concomitant with these sarcopenic changes is an increased abundance of mitochondrial DNA deletion mutations and electron transport chain (ETC) abnormalities. In this study, we used immunohistological and histochemical approaches to define cell death pathways involved in sarcopenia. Activation of muscle cell death pathways was age‐dependent with most apoptotic and necrotic muscle fibers exhibiting ETC abnormalities. Although activation of apoptosis was a prominent feature of electron transport abnormal muscle fibers, necrosis was predominant in atrophic and broken ETC‐abnormal fibers. These data suggest that mitochondrial dysfunction is a major contributor to the activation of cell death processes in aged muscle fibers. The link between ETC abnormalities, apoptosis, fiber atrophy, and necrosis supports the hypothesis that mitochondrial DNA deletion mutations are causal in myofiber loss. These studies suggest a progression of events beginning with the generation and accumulation of a mtDNA deletion mutation, the concomitant development of ETC abnormalities, a subsequent triggering of apoptotic and, ultimately, necrotic events resulting in muscle fiber atrophy, breakage, and fiber loss.
Collapse
Affiliation(s)
- Nashwa Cheema
- Department of Biological Sciences Centre for Prions and Protein Folding Diseases University of Alberta Edmonton AB Canada
| | - Allen Herbst
- Department of Agricultural Food and Nutritional Sciences Centre for Prions and Protein Folding Diseases University of Alberta Edmonton AB Canada
| | - Debbie McKenzie
- Department of Biological Sciences Centre for Prions and Protein Folding Diseases University of Alberta Edmonton AB Canada
| | - Judd M. Aiken
- Department of Agricultural Food and Nutritional Sciences Centre for Prions and Protein Folding Diseases University of Alberta Edmonton AB Canada
| |
Collapse
|
163
|
Grabowska I, Mazur MA, Kowalski K, Helinska A, Moraczewski J, Stremińska W, Hoser G, Kawiak J, Ciemerych MA, Brzoska E. Progression of inflammation during immunodeficient mouse skeletal muscle regeneration. J Muscle Res Cell Motil 2015; 36:395-404. [PMID: 26613733 PMCID: PMC4762921 DOI: 10.1007/s10974-015-9433-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 11/18/2015] [Indexed: 12/31/2022]
Abstract
The skeletal muscle injury triggers the inflammatory response which is crucial for damaged muscle fiber degradation and satellite cell activation. Immunodeficient mice are often used as a model to study the myogenic potential of transplanted human stem cells. Therefore, it is crucial to elucidate whether such model truly reflects processes occurring under physiological conditions. To answer this question we compared skeletal muscle regeneration of BALB/c, i.e. animals producing all types of inflammatory cells, and SCID mice. Results of our study documented that initial stages of muscles regeneration in both strains of mice were comparable. However, lower number of mononucleated cells was noticed in regenerating SCID mouse muscles. Significant differences in the number of CD14-/CD45+ and CD14+/CD45+ cells between BALB/c and SCID muscles were also observed. In addition, we found important differences in M1 and M2 macrophage levels of BALB/c and SCID mouse muscles identified by CD68 and CD163 markers. Thus, our data show that differences in inflammatory response during muscle regeneration, were not translated into significant modifications in muscle regeneration.
Collapse
Affiliation(s)
- Iwona Grabowska
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1 St., 02-096, Warsaw, Poland
| | - Magdalena A Mazur
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1 St., 02-096, Warsaw, Poland
| | - K Kowalski
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1 St., 02-096, Warsaw, Poland
| | - A Helinska
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1 St., 02-096, Warsaw, Poland
| | - Jerzy Moraczewski
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1 St., 02-096, Warsaw, Poland
| | - Władysława Stremińska
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1 St., 02-096, Warsaw, Poland
| | - Grażyna Hoser
- Laboratory of Flow Cytometry, Medical Center of Postgraduate Education, Marymoncka 99/103 St., 01-813, Warsaw, Poland
| | - Jerzy Kawiak
- Laboratory of Flow Cytometry, Medical Center of Postgraduate Education, Marymoncka 99/103 St., 01-813, Warsaw, Poland
| | - 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
|
164
|
Mahajan V, Gaymalov Z, Alakhova D, Gupta R, Zucker IH, Kabanov AV. Horizontal gene transfer from macrophages to ischemic muscles upon delivery of naked DNA with Pluronic block copolymers. Biomaterials 2015; 75:58-70. [PMID: 26480472 DOI: 10.1016/j.biomaterials.2015.10.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 09/30/2015] [Accepted: 10/01/2015] [Indexed: 12/31/2022]
Abstract
Intramuscular administration of plasmid DNA (pDNA) with non-ionic Pluronic block copolymers increases gene expression in injected muscles and lymphoid organs. We studied the role of immune cells in muscle transfection upon inflammation. Local inflammation in murine hind limb ischemia model (MHLIM) drastically increased DNA, RNA and expressed protein levels in ischemic muscles injected with pDNA/Pluronic. The systemic inflammation (MHLIM or peritonitis) also increased expression of pDNA/Pluronic in the muscles. When pDNA/Pluronic was injected in ischemic muscles the reporter gene, Green Fluorescent Protein (GFP) co-localized with desmin(+) muscle fibers and CD11b(+) macrophages (MØs), suggesting transfection of MØs along with the muscle cells. P85 enhanced (∼ 4 orders) transfection of MØs with pDNA in vitro. Moreover, adoptively transferred MØs were shown to pass the transgene to inflamed muscle cells in MHLIM. Using a co-culture of myotubes (MTs) and transfected MØs expressing a reporter gene under constitutive (cmv-luciferase) or muscle specific (desmin-luciferase) promoter we demonstrated that P85 enhances horizontal gene transfer from MØ to MTs. Therefore, MØs can play an important role in muscle transfection with pDNA/Pluronic during inflammation, with both inflammation and Pluronic contributing to the increased gene expression. pDNA/Pluronic has potential for therapeutic gene delivery in muscle pathologies that involve inflammation.
Collapse
Affiliation(s)
- Vivek Mahajan
- Division of Molecular Pharmaceutics, Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA; Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-5850, USA
| | - Zagit Gaymalov
- Division of Molecular Pharmaceutics, Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA; Department of Pharmaceutical Sciences and Center for Drug Delivery and Nanomedicine, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198-5850, USA
| | - Daria Alakhova
- Division of Molecular Pharmaceutics, Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Richa Gupta
- Division of Molecular Pharmaceutics, Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Irving H Zucker
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198-5850, USA
| | - Alexander V Kabanov
- Division of Molecular Pharmaceutics, Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, USA; Department of Pharmaceutical Sciences and Center for Drug Delivery and Nanomedicine, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198-5850, USA; Laboratory of Chemical Design of Bionanomaterials, Faculty of Chemistry, M.V. Lomonosov Moscow State University, 119899 Moscow, Russia.
| |
Collapse
|
165
|
White MJV, Gomer RH. Trypsin, Tryptase, and Thrombin Polarize Macrophages towards a Pro-Fibrotic M2a Phenotype. PLoS One 2015; 10:e0138748. [PMID: 26407067 PMCID: PMC4583378 DOI: 10.1371/journal.pone.0138748] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 09/03/2015] [Indexed: 02/06/2023] Open
Abstract
For both wound healing and the formation of a fibrotic lesion, circulating monocytes enter the tissue and differentiate into fibroblast-like cells called fibrocytes and pro-fibrotic M2a macrophages, which together with fibroblasts form scar tissue. Monocytes can also differentiate into classically activated M1 macrophages and alternatively activated M2 macrophages. The proteases thrombin, which is activated during blood clotting, and tryptase, which is released by activated mast cells, potentiate fibroblast proliferation and fibrocyte differentiation, but their effect on macrophages is unknown. Here we report that thrombin, tryptase, and the protease trypsin bias human macrophage differentiation towards a pro-fibrotic M2a phenotype expressing high levels of galectin-3 from unpolarized monocytes, or from M1 and M2 macrophages, and that these effects appear to operate through protease-activated receptors. These results suggest that proteases can initiate scar tissue formation by affecting fibroblasts, fibrocytes, and macrophages.
Collapse
Affiliation(s)
- Michael J. V. White
- Department of Biology, Texas A&M University, College Station, Texas, United States of America
| | - Richard H. Gomer
- Department of Biology, Texas A&M University, College Station, Texas, United States of America
| |
Collapse
|
166
|
Brown LA, Lee DE, Patton JF, Perry RA, Brown JL, Baum JI, Smith-Blair N, Greene NP, Washington TA. Diet-induced obesity alters anabolic signalling in mice at the onset of skeletal muscle regeneration. Acta Physiol (Oxf) 2015; 215:46-57. [PMID: 26052759 DOI: 10.1111/apha.12537] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 04/03/2015] [Accepted: 05/31/2015] [Indexed: 12/15/2022]
Abstract
AIM Obesity is classified as a metabolic disorder that is associated with delayed muscle regeneration following damage. For optimal skeletal muscle regeneration, inflammation along with extracellular matrix remodelling and muscle growth must be tightly regulated. Moreover, the regenerative process is dependent on the activation of myogenic regulatory factors (MRFs) for myoblast proliferation and differentiation. The purpose of this study was to determine how obesity alters inflammatory and protein synthetic signalling and MRF expression at the onset of muscle regeneration in mice. METHODS Forty-eight male C57BL/6J mice (3 weeks old) were randomly assigned to either a high-fat diet (HFD, 60% fat) or a lean diet (10% fat) for 12 weeks. At 15 weeks, bupivacaine was injected into the tibialis anterior (TA) of the injured group (n = 5-8/group) and PBS was injected into the control (n = 5-6). The TA was excised 3 or 28 days after injection. RESULTS We demonstrated impaired muscle regeneration in obese mice. The obese mice had reduced IL-6, MyoD and IGF-1 mRNA abundance compared to the lean mice (P < 0.05). Three days following muscle damage, TNF-α mRNA and protein levels of P-STAT3 and P-Akt were 14-fold, fourfold and fivefold greater in the lean mice respectively. However, there were no differences observed in the obese injured group compared to the uninjured group. Moreover, p70S6K1 was threefold greater in lean injured mice compared to uninjured but was reduced by 28% in the obese injured mice. CONCLUSION Obese mice have impaired inflammatory and protein synthetic signalling that may negatively influence muscle regeneration.
Collapse
Affiliation(s)
- L. A. Brown
- Exercise Muscle Biology Laboratory; Department of Health, Human Performance and Recreation; University of Arkansas; Fayetteville AR USA
| | - D. E. Lee
- Integrative Muscle Metabolism Laboratory; Department of Health, Human Performance and Recreation; University of Arkansas; Fayetteville AR USA
| | - J. F. Patton
- Exercise Muscle Biology Laboratory; Department of Health, Human Performance and Recreation; University of Arkansas; Fayetteville AR USA
| | - R. A. Perry
- Exercise Muscle Biology Laboratory; Department of Health, Human Performance and Recreation; University of Arkansas; Fayetteville AR USA
| | - J. L. Brown
- Integrative Muscle Metabolism Laboratory; Department of Health, Human Performance and Recreation; University of Arkansas; Fayetteville AR USA
| | - J. I. Baum
- Department of Food Science; University of Arkansas; Fayetteville AR USA
| | - N. Smith-Blair
- Eleanor Mann School of Nursing; University of Arkansas; Fayetteville AR USA
| | - N. P. Greene
- Integrative Muscle Metabolism Laboratory; Department of Health, Human Performance and Recreation; University of Arkansas; Fayetteville AR USA
| | - T. A. Washington
- Exercise Muscle Biology Laboratory; Department of Health, Human Performance and Recreation; University of Arkansas; Fayetteville AR USA
| |
Collapse
|
167
|
Lindsay A, Carr S, Othman MI, Marks E, Davies S, Petersen C, Draper N, Gieseg SP. The physiological and mononuclear cell activation response to cryotherapy following a mixed martial arts contest: a pilot study. Pteridines 2015. [DOI: 10.1515/pterid-2015-0010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Abstract
Cold water immersion is thought to reduce the inflammatory response to injury. Using cultured mononuclear cells and human subjects in a mixed martial arts (MMA) contest, we examined the effect of cryotherapy on 7,8-dihydroneopterin and neopterin generation. Urine was collected from 10 elite male mixed martial artists before, immediately post and 1, 2, 24 and 48 h following a contest. Myoglobin was analysed by reverse-phase high performance liquid chromatography, and urinary neopterin and total neopterin (neopterin+7,8-dihydroneopterin) were measured by strong cation exchange high-performance liquid chromatography. Cold water immersion and passive recovery were compared using changes in these markers, while cryotherapy tested total neopterin production in γ-interferon and phorbol myristate acetate (PMA)-stimulated blood-derived mononuclear cells (monocytes/T cells). Myoglobin significantly increased (p<0.05) at 1 h post-contest, neopterin significantly increased at 1 and 24 h (p<0.05), total neopterin significantly increased (p<0.05) at 1 h post for the passive group only, and significant individual variation was observed for all markers (p<0.01). Cold water immersion attenuated total neopterin production (p<0.05), while cryotherapy significantly reduced total neopterin production in PMA-stimulated mononuclear cells (p<0.01). Cryotherapy attenuates the post-exercise inflammatory response following an MMA contest. The evidence also suggests that the mechanisms responsible for this may be related to direct immune cell suppression.
Collapse
Affiliation(s)
- Angus Lindsay
- Free Radical Biochemistry Laboratory, School of Biological Sciences, University of Canterbury, Private Bag 4800, Christchurch, New Zealand
| | - Sam Carr
- Free Radical Biochemistry Laboratory, School of Biological Sciences, University of Canterbury, Private Bag 4800, Christchurch, New Zealand
| | | | - Edward Marks
- Free Radical Biochemistry Laboratory, School of Biological Sciences, University of Canterbury, Private Bag 4800, Christchurch, New Zealand
| | - Sian Davies
- Free Radical Biochemistry Laboratory, School of Biological Sciences, University of Canterbury, Private Bag 4800, Christchurch, New Zealand
| | - Carl Petersen
- School of Sport and Physical Education, University of Canterbury, Private Bag 4800, Christchurch, New Zealand
| | - Nick Draper
- College of Life and Natural Sciences, University of Derby, Kedleston Road, Derby, UK
| | | |
Collapse
|
168
|
Fearing CM, Melton DW, Lei X, Hancock H, Wang H, Sarwar ZU, Porter L, McHale M, McManus LM, Shireman PK. Increased Adipocyte Area in Injured Muscle With Aging and Impaired Remodeling in Female Mice. J Gerontol A Biol Sci Med Sci 2015; 71:992-1004. [PMID: 26273023 DOI: 10.1093/gerona/glv104] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 06/15/2015] [Indexed: 12/30/2022] Open
Abstract
We demonstrated that young male and female mice similarly regenerated injured skeletal muscle; however, female mice transiently increased adipocyte area within regenerated muscle in a sex hormone-dependent manner. We extended these observations to investigate the effect of aging and sex on sarcopenia and muscle regeneration. Cardiotoxin injury to the tibialis anterior muscle of young, middle, and old-aged C57Bl/6J male and female mice was used to measure regenerated myofiber cross-sectional area (CSA), adipocyte area, residual necrosis, and inflammatory cell recruitment. Baseline (uninjured) myofiber CSA was decreased in old mice of both sexes compared to young and middle-aged mice. Regenerated CSA was similar in male mice in all age groups until baseline CSA was attained but decreased in middle and old age female mice compared to young females. Furthermore, adipocyte area within regenerated muscle was transiently increased in young females compared to young males and these sex-dependent increases persisted in middle and old age female mice and were associated with increased Pparg Young female mice had more pro-inflammatory monocytes/macrophages in regenerating muscle than young male mice and increased Sca-1(+)CD45(-)cells. In conclusion, sex and age influence pro-inflammatory cell recruitment, muscle regeneration, and adipocyte area following skeletal muscle injury.
Collapse
Affiliation(s)
| | - David W Melton
- Department of Surgery, Department of Cellular and Structural Biology, Sam and Ann Barshop Institute for Longevity and Aging Studies
| | | | | | | | | | | | | | - Linda M McManus
- Sam and Ann Barshop Institute for Longevity and Aging Studies, Department of Pathology, and Department of Periodontics, University of Texas Health Science Center, San Antonio
| | - Paula K Shireman
- Department of Surgery, Sam and Ann Barshop Institute for Longevity and Aging Studies, The South Texas Veterans Health Care System, San Antonio.
| |
Collapse
|
169
|
Cohen TV, Many GM, Fleming BD, Gnocchi VF, Ghimbovschi S, Mosser DM, Hoffman EP, Partridge TA. Upregulated IL-1β in dysferlin-deficient muscle attenuates regeneration by blunting the response to pro-inflammatory macrophages. Skelet Muscle 2015; 5:24. [PMID: 26251696 PMCID: PMC4527226 DOI: 10.1186/s13395-015-0048-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 06/16/2015] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Loss-of-function mutations in the dysferlin gene (DYSF) result in a family of muscle disorders known collectively as the dysferlinopathies. Dysferlin-deficient muscle is characterized by inflammatory foci and macrophage infiltration with subsequent decline in muscle function. Whereas macrophages function to remove necrotic tissue in acute injury, their prevalence in chronic myopathy is thought to inhibit resolution of muscle regeneration. Two major classes of macrophages, classical (M1) and alternative (M2a), play distinct roles during the acute injury process. However, their individual roles in chronic myopathy remain unclear and were explored in this study. METHODS To test the roles of the two macrophage phenotypes on regeneration in dysferlin-deficient muscle, we developed an in vitro co-culture model of macrophages and muscle cells. We assayed the co-cultures using ELISA and cytokine arrays to identify secreted factors and performed transcriptome analysis of molecular networks induced in the myoblasts. RESULTS Dysferlin-deficient muscle contained an excess of M1 macrophage markers, compared with WT, and regenerated poorly in response to toxin injury. Co-culturing macrophages with muscle cells showed that M1 macrophages inhibit muscle regeneration whereas M2a macrophages promote it, especially in dysferlin-deficient muscle cells. Examination of soluble factors released in the co-cultures and transcriptome analysis implicated two soluble factors in mediating the effects: IL-1β and IL-4, which during acute injury are secreted from M1 and M2a macrophages, respectively. To test the roles of these two factors in dysferlin-deficient muscle, myoblasts were treated with IL-4, which improved muscle differentiation, or IL-1β, which inhibited it. Importantly, blockade of IL-1β signaling significantly improved differentiation of dysferlin-deficient cells. CONCLUSIONS We propose that the inhibitory effects of M1 macrophages on myogenesis are mediated by IL-1β signals and suppression of the M1-mediated immune response may improve muscle regeneration in dysferlin deficiency. Our studies identify a potential therapeutic approach to promote muscle regeneration in dystrophic muscle.
Collapse
Affiliation(s)
- Tatiana V. Cohen
- />Center for Genetic Medicine Research, Children’s National Medical Center, 111 Michigan Avenue NW, Washington, DC 20010 USA
- />Center for Genetic Muscle Disorders, Kennedy Krieger Institute, 707 N. Broadway, Baltimore, MD 21205 USA
- />Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Gina M. Many
- />Center for Genetic Medicine Research, Children’s National Medical Center, 111 Michigan Avenue NW, Washington, DC 20010 USA
| | - Bryan D. Fleming
- />Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742 USA
| | - Viola F. Gnocchi
- />Center for Genetic Medicine Research, Children’s National Medical Center, 111 Michigan Avenue NW, Washington, DC 20010 USA
| | - Svetlana Ghimbovschi
- />Center for Genetic Medicine Research, Children’s National Medical Center, 111 Michigan Avenue NW, Washington, DC 20010 USA
| | - David M. Mosser
- />Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742 USA
| | - Eric P. Hoffman
- />Center for Genetic Medicine Research, Children’s National Medical Center, 111 Michigan Avenue NW, Washington, DC 20010 USA
| | - Terence A. Partridge
- />Center for Genetic Medicine Research, Children’s National Medical Center, 111 Michigan Avenue NW, Washington, DC 20010 USA
| |
Collapse
|
170
|
Exercise and the Regulation of Inflammatory Responses. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 135:337-54. [PMID: 26477921 DOI: 10.1016/bs.pmbts.2015.07.003] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Exercise initiates a cascade of inflammatory events, which ultimately lead to long-term effects on human health. During and after acute exercise in skeletal muscle, interactions between immune cells, cytokines, and other intracellular components, create an inflammatory milieu responsible for the recovery and adaption from an exercise bout. In the systemic circulation, cytokines released from muscle (myokines) mediate metabolic and inflammatory processes. Moderate exercise training results in improvements in systemic inflammation, evident by reductions in acute phase proteins. The anti-inflammatory effects of regular exercise include actions dependent and independent of changes in adipose tissue mass. Future research should encompass approaches, which attempt to integrate other, less-recognized physiological processes with acute and long-term inflammatory changes. This will include investigation into metabolic, endocrine, and immune components of various tissues and organs.
Collapse
|
171
|
Nilotinib reduces muscle fibrosis in chronic muscle injury by promoting TNF-mediated apoptosis of fibro/adipogenic progenitors. Nat Med 2015; 21:786-94. [PMID: 26053624 DOI: 10.1038/nm.3869] [Citation(s) in RCA: 512] [Impact Index Per Article: 51.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 04/29/2015] [Indexed: 12/14/2022]
Abstract
Depending on the inflammatory milieu, injury can result either in a tissue's complete regeneration or in its degeneration and fibrosis, the latter of which could potentially lead to permanent organ failure. Yet how inflammatory cells regulate matrix-producing cells involved in the reparative process is unknown. Here we show that in acutely damaged skeletal muscle, sequential interactions between multipotent mesenchymal progenitors and infiltrating inflammatory cells determine the outcome of the reparative process. We found that infiltrating inflammatory macrophages, through their expression of tumor necrosis factor (TNF), directly induce apoptosis of fibro/adipogenic progenitors (FAPs). In states of chronic damage, however, such as those in mdx mice, macrophages express high levels of transforming growth factor β1 (TGF-β1), which prevents the apoptosis of FAPs and induces their differentiation into matrix-producing cells. Treatment with nilotinib, a kinase inhibitor with proposed anti-fibrotic activity, can block the effect of TGF-β1 and reduce muscle fibrosis in mdx mice. Our findings reveal an unexpected anti-fibrotic role of TNF and suggest that disruption of the precisely timed progression from a TNF-rich to a TGF-β-rich environment favors fibrotic degeneration of the muscle during chronic injury.
Collapse
|
172
|
FOXP3+ T Cells Recruited to Sites of Sterile Skeletal Muscle Injury Regulate the Fate of Satellite Cells and Guide Effective Tissue Regeneration. PLoS One 2015; 10:e0128094. [PMID: 26039259 PMCID: PMC4454513 DOI: 10.1371/journal.pone.0128094] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2015] [Accepted: 04/23/2015] [Indexed: 01/07/2023] Open
Abstract
Muscle injury induces a classical inflammatory response in which cells of the innate immune system rapidly invade the tissue. Macrophages are prominently involved in this response and required for proper healing, as they are known to be important for clearing cellular debris and supporting satellite cell differentiation. Here, we sought to assess the role of the adaptive immune system in muscle regeneration after acute damage. We show that T lymphocytes are transiently recruited into the muscle after damage and appear to exert a pro-myogenic effect on muscle repair. We observed a decrease in the cross-sectional area of regenerating myofibers after injury in Rag2-/- γ-chain-/- mice, as compared to WT controls, suggesting that T cell recruitment promotes muscle regeneration. Skeletal muscle infiltrating T lymphocytes were enriched in CD4+CD25+FOXP3+ cells. Direct exposure of muscle satellite cells to in vitro induced Treg cells effectively enhanced their expansion, and concurrently inhibited their myogenic differentiation. In vivo, the recruitment of Tregs to acutely injured muscle was limited to the time period of satellite expansion, with possibly important implications for situations in which inflammatory conditions persist, such as muscular dystrophies and inflammatory myopathies. We conclude that the adaptive immune system, in particular T regulatory cells, is critically involved in effective skeletal muscle regeneration. Thus, in addition to their well-established role as regulators of the immune/inflammatory response, T regulatory cells also regulate the activity of skeletal muscle precursor cells, and are instrumental for the proper regeneration of this tissue.
Collapse
|
173
|
Prezioso C, Iaconis S, Andolfi G, Zentilin L, Iavarone F, Guardiola O, Minchiotti G. Conditional Cripto overexpression in satellite cells promotes myogenic commitment and enhances early regeneration. Front Cell Dev Biol 2015; 3:31. [PMID: 26052513 PMCID: PMC4439575 DOI: 10.3389/fcell.2015.00031] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 05/02/2015] [Indexed: 01/06/2023] Open
Abstract
Skeletal muscle regeneration mainly depends on satellite cells, a population of resident muscle stem cells. Despite extensive studies, knowledge of the molecular mechanisms underlying the early events associated with satellite cell activation and myogenic commitment in muscle regeneration remains still incomplete. Cripto is a novel regulator of postnatal skeletal muscle regeneration and a promising target for future therapy. Indeed, Cripto is expressed both in myogenic and inflammatory cells in skeletal muscle after acute injury and it is required in the satellite cell compartment to achieve effective muscle regeneration. A critical requirement to further explore the in vivo cellular contribution of Cripto in regulating skeletal muscle regeneration is the possibility to overexpress Cripto in its endogenous configuration and in a cell and time-specific manner. Here we report the generation and the functional characterization of a novel mouse model for conditional expression of Cripto, i.e., the Tg:DsRedloxP/loxPCripto-eGFP mice. Moreover, by using a satellite cell specific Cre-driver line we investigated the biological effect of Cripto overexpression in vivo, and provided evidence that overexpression of Cripto in the adult satellite cell compartment promotes myogenic commitment and differentiation, and enhances early regeneration in a mouse model of acute injury.
Collapse
Affiliation(s)
- Carolina Prezioso
- Stem Cell Fate Laboratory, Institute of Genetics and Biophysics "A. Buzzati-Traverso," CNR Naples, Italy
| | - Salvatore Iaconis
- Stem Cell Fate Laboratory, Institute of Genetics and Biophysics "A. Buzzati-Traverso," CNR Naples, Italy
| | - Gennaro Andolfi
- Stem Cell Fate Laboratory, Institute of Genetics and Biophysics "A. Buzzati-Traverso," CNR Naples, Italy
| | - Lorena Zentilin
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology Trieste, Italy
| | - Francescopaolo Iavarone
- Stem Cell Fate Laboratory, Institute of Genetics and Biophysics "A. Buzzati-Traverso," CNR Naples, Italy
| | - Ombretta Guardiola
- Stem Cell Fate Laboratory, Institute of Genetics and Biophysics "A. Buzzati-Traverso," CNR Naples, Italy
| | - Gabriella Minchiotti
- Stem Cell Fate Laboratory, Institute of Genetics and Biophysics "A. Buzzati-Traverso," CNR Naples, Italy
| |
Collapse
|
174
|
Chung T, Christopher-Stine L, Paik JJ, Corse A, Mammen AL. The composition of cellular infiltrates in anti-HMG-CoA reductase-associated myopathy. Muscle Nerve 2015; 52:189-95. [PMID: 25737145 DOI: 10.1002/mus.24642] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 02/23/2015] [Accepted: 02/28/2015] [Indexed: 11/05/2022]
Abstract
INTRODUCTION To characterize cellular infiltrates in muscle biopsies from patients with anti-3-hydroxy-3-methyl-gulatryl-CoA reductase (HMGCR)-associated myopathy. METHODS Biopsies from 18 anti-HMGCR myopathy and 7 control dermatomyositis patients were analyzed. RESULTS CD4+ and CD8+ T-cells were scattered within the endomysium in 50% of anti-HMGCR biopsies. All anti-HMGCR biopsies included increased endomysial and/or perivascular CD163+ M2 macrophages; CD11c+ M1 macrophages were present in 18.8%. CD123+ plasmacytoid dendritic (PD) cells were observed within the endomysium and perivascular spaces in 62.5% of anti-HMGCR biopsies. Membrane attack complex was deposited on endothelial cells in 50% and on the sarcolemma of nonnecrotic muscle fibers in 85.7% of anti-HMGCR cases. Major histocompatibility complex class I antigen was up-regulated in 87.5% of the anti-HMGCR cases. CONCLUSIONS In addition to necrosis, scattered CD4+, CD8+, and PD cells are characteristic of anti-HMGCR myopathy. Predominant M2 polarization suggests infiltrating macrophages are more likely to be involved with tissue repair than destruction.
Collapse
Affiliation(s)
- Tae Chung
- Department of Physical Medicine and Rehabilitation, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Lisa Christopher-Stine
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Julie J Paik
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andrea Corse
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andrew L Mammen
- Muscle Disease Unit, Laboratory of Muscle Stem Cells and Gene Expression, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, 50 South Drive, Room 1146, Building 50, MSC 8024, Bethesda, Maryland, 20892, USA
| |
Collapse
|
175
|
Abstract
INTRODUCTION Every year recreational and professional sports cause thousands of lesions to muscle, ligament and tendon. Critical progress in biological interventions could meet a pressing health need to help athletes resume their activity levels. AREAS COVERED We perform a narrative review on platelet-rich plasma (PRP) therapies and muscle injuries. The field is eminently translational, thus besides clinical data we summarize experimental studies that bring meaningful biological insights on PRP effects. Some concepts regarding healing mechanisms are reviewed including innate immune response, myogenesis and fibrosis. It is commonly thought that PRPs are not uniform and cannot be assessed against each other, thus current PRP classifications are addressed. PRP effects also depend on the characteristics of the host tissue; therefore we focus on clinical muscle injury classifications. Controversial clinical findings are attributed to both the variability in PRP formulations and variability in the application protocols, so we discuss current clinical data in this basis. EXPERT OPINION Currently, there is little clinical evidence to support the use of PRP in skeletal muscle injuries. The future of PRP therapies relies not only in finding the best products, most appropriate indications and application protocols, but also in conceiving combination products. Moreover, as our understanding of healing mechanisms progresses, off-the-shelf allogenic PRP products could be part of the solution for sport injuries.
Collapse
Affiliation(s)
- Isabel Andia
- Cruces University Hospital, BioCruces Health Research Institute, Regenerative Medicine Laboratory , Barakaldo 48903 , Spain
| | | |
Collapse
|
176
|
Dezfuli BS, Manera M, Lorenzoni M, Pironi F, Shinn AP, Giari L. Histopathology and the inflammatory response of European perch, Perca fluviatilis muscle infected with Eustrongylides sp. (Nematoda). Parasit Vectors 2015; 8:227. [PMID: 25889096 PMCID: PMC4404125 DOI: 10.1186/s13071-015-0838-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 03/31/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The European perch, Perca fluviatilis L. is a common paratenic host of dioctophymatid nematodes belonging to the genus Eustrongylides. In this host, once infected oligochaetes, which serve as the first intermediate host, are ingested, Eustrongylides migrates through the intestine and is frequently encountered within the musculature, free within the body cavity, or encapsulated on the viscera. The current study details the first Italian record of Eustrongylides sp. with larvae reported in the muscle of P. fluviatilis. METHODS Uninfected and nematode-infected muscle tissues of perch were fixed and prepared for histological evaluation and electron microscopy. Some sections were subjected to an indirect immunohistochemical method using anti-PCNA, anti-piscidin 3 and anti-piscidin 4 antibodies. RESULTS A total of 510 P. fluviatilis (TL range 15-25 cm) from Lake Trasimeno, Perugia were post-mortemed; 31 individuals had encysted nematode larvae within their musculature (1-2 worms fish(-1)). Histologically, larvae were surrounded by a capsule with an evident acute inflammatory reaction. Muscle degeneration and necrosis extending throughout the sarcoplasm, sarcolemmal basal lamina, endomysial connective tissue cells and capillaries was frequently observed. Within the encapsulating reaction, macrophage aggregates (MAs) were seen. Immunohistochemical staining with the proliferating cell nuclear antigen (PCNA) revealed numerous PCNA-positive cells within the thickness of the capsule and in the immediate vicinity surrounding Eustrongylides sp. larvae (i.e. fibroblasts and satellite cells), suggesting a host response had been initiated to repair the nematode-damaged muscle. Mast cells (MCs) staining positively for piscidin 3, were demonstrated for the first time in response to a muscle-infecting nematode. The piscidin 3 positive MC's were seen principally in the periphery of the capsule surrounding the Eustrongylides sp. larva. CONCLUSIONS A host tissue response to Eustrongylides sp. larvae infecting the musculature of P. fluviatilis was observed. Numerous fibroblasts, MAs and MCs were seen throughout the thick fibroconnectival layer of the capsule enclosing larvae. PCNA positive cells within the capsule suggest that host repair of nematode damaged muscle does occur, while the presence of the antimicrobial peptide piscidin 3 is shown for the first time. This is first report of Eustrongylides sp. in an Italian population of P. fluviatilis.
Collapse
Affiliation(s)
- Bahram S Dezfuli
- Department of Life Sciences and Biotechnology, University of Ferrara, St. Borsari 46, 44121, Ferrara, Italy.
| | - Maurizio Manera
- Faculty of Biosciences, Agro-Alimentary and Environmental Technologies, University of Teramo, St. Crispi 212, I-64100, Teramo, Italy.
| | - Massimo Lorenzoni
- Department of Cellular and Environmental Biology, University of Perugia, St. Elce di sotto 5, 06123, Perugia, Italy.
| | - Flavio Pironi
- Department of Life Sciences and Biotechnology, University of Ferrara, St. Borsari 46, 44121, Ferrara, Italy.
| | - Andrew P Shinn
- Fish Vet Group Asia Limited, 99/386, Chaengwattana Building, Chaengwattana Rd., Kwaeng Toongsonghong, Khet Laksi, Bangkok, 10210, Thailand.
| | - Luisa Giari
- Department of Life Sciences and Biotechnology, University of Ferrara, St. Borsari 46, 44121, Ferrara, Italy.
| |
Collapse
|
177
|
MILLS DEANE, JOHNSON MICHAELA, BARNETT YVONNEA, SMITH WILLIAMHT, SHARPE GRAHAMR. The Effects of Inspiratory Muscle Training in Older Adults. Med Sci Sports Exerc 2015; 47:691-7. [DOI: 10.1249/mss.0000000000000474] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
178
|
Oyster N, Witt M, Gharaibeh B, Poddar M, Schneppendahl J, Huard J. Characterization of a compartment syndrome-like injury model. Muscle Nerve 2015; 51:750-8. [PMID: 25242666 DOI: 10.1002/mus.24461] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Revised: 08/23/2014] [Accepted: 09/17/2014] [Indexed: 01/16/2023]
Abstract
INTRODUCTION Acute compartment syndrome (CS) is caused by an elevation of pressure within a muscular compartment that can be caused by numerous factors, including blunt trauma. In this study, we characterized a rodent model of CS-like injury. METHODS Forty male athymic rats received a standardized injury of ischemia and compression to their hindlimbs, while the intracompartmental pressure (ICP) was measured using an implantable transmitter. Tetanic muscle function was evaluated, and histology was performed on the tibialis anterior (TA) muscle. RESULTS ICPs were held at 260.70 ± 2.70 mm Hg during injury. Injured muscles recovered 59% of their total function 4 weeks after injury, and histology showed high levels of edema, inflammation (CD68(+) ), angiogenesis (CD31(+) ), and fibrosis within 72 hours after injury. CONCLUSIONS We describe a novel CS-like injury model and a novel method to measure ICP, which could potentially be used to develop innovative therapies to manage CS injury in patients.
Collapse
Affiliation(s)
- Nick Oyster
- Stem Cell Research Center, University of Pittsburgh, Suite 206, Bridgeside Point II, 450 Technology Drive, Pittsburgh, Pennsylvania, 15219, USA
| | | | | | | | | | | |
Collapse
|
179
|
Macrophage gene expression associated with remodeling of the prepartum rat cervix: microarray and pathway analyses. PLoS One 2015; 10:e0119782. [PMID: 25811906 PMCID: PMC4374766 DOI: 10.1371/journal.pone.0119782] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 01/16/2015] [Indexed: 11/19/2022] Open
Abstract
As the critical gatekeeper for birth, prepartum remodeling of the cervix is associated with increased resident macrophages (Mφ), proinflammatory processes, and extracellular matrix degradation. This study tested the hypothesis that expression of genes unique to Mφs characterizes the prepartum from unremodeled nonpregnant cervix. Perfused cervix from prepartum day 21 postbreeding (D21) or nonpregnant (NP) rats, with or without Mφs, had RNA extracted and whole genome microarray analysis performed. By subtractive analyses, expression of 194 and 120 genes related to Mφs in the cervix from D21 rats were increased and decreased, respectively. In both D21 and NP groups, 158 and 57 Mφ genes were also more or less up- or down-regulated, respectively. Mφ gene expression patterns were most strongly correlated within groups and in 5 major clustering patterns. In the cervix from D21 rats, functional categories and canonical pathways of increased expression by Mφ gene related to extracellular matrix, cell proliferation, differentiation, as well as cell signaling. Pathways were characteristic of inflammation and wound healing, e.g., CD163, CD206, and CCR2. Signatures of only inflammation pathways, e.g., CSF1R, EMR1, and MMP12 were common to both D21 and NP groups. Thus, a novel and complex balance of Mφ genes and clusters differentiated the degraded extracellular matrix and cellular genomic activities in the cervix before birth from the unremodeled state. Predicted Mφ activities, pathways, and networks raise the possibility that expression patterns of specific genes characterize and promote prepartum remodeling of the cervix for parturition at term and with preterm labor.
Collapse
|
180
|
Qazi TH, Mooney DJ, Pumberger M, Geissler S, Duda GN. Biomaterials based strategies for skeletal muscle tissue engineering: existing technologies and future trends. Biomaterials 2015; 53:502-21. [PMID: 25890747 DOI: 10.1016/j.biomaterials.2015.02.110] [Citation(s) in RCA: 281] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 02/18/2015] [Accepted: 02/24/2015] [Indexed: 12/20/2022]
Abstract
Skeletal muscles have a robust capacity to regenerate, but under compromised conditions, such as severe trauma, the loss of muscle functionality is inevitable. Research carried out in the field of skeletal muscle tissue engineering has elucidated multiple intrinsic mechanisms of skeletal muscle repair, and has thus sought to identify various types of cells and bioactive factors which play an important role during regeneration. In order to maximize the potential therapeutic effects of cells and growth factors, several biomaterial based strategies have been developed and successfully implemented in animal muscle injury models. A suitable biomaterial can be utilized as a template to guide tissue reorganization, as a matrix that provides optimum micro-environmental conditions to cells, as a delivery vehicle to carry bioactive factors which can be released in a controlled manner, and as local niches to orchestrate in situ tissue regeneration. A myriad of biomaterials, varying in geometrical structure, physical form, chemical properties, and biofunctionality have been investigated for skeletal muscle tissue engineering applications. In the current review, we present a detailed summary of studies where the use of biomaterials favorably influenced muscle repair. Biomaterials in the form of porous three-dimensional scaffolds, hydrogels, fibrous meshes, and patterned substrates with defined topographies, have each displayed unique benefits, and are discussed herein. Additionally, several biomaterial based approaches aimed specifically at stimulating vascularization, innervation, and inducing contractility in regenerating muscle tissues are also discussed. Finally, we outline promising future trends in the field of muscle regeneration involving a deeper understanding of the endogenous healing cascades and utilization of this knowledge for the development of multifunctional, hybrid, biomaterials which support and enable muscle regeneration under compromised conditions.
Collapse
Affiliation(s)
- Taimoor H Qazi
- Julius Wolff Institute, Charité - Universitätsmedizin Berlin, Germany; Berlin-Brandenburg School for Regenerative Therapies, Berlin, Germany.
| | - David J Mooney
- School of Engineering and Applied Sciences, Harvard University, Cambridge, USA.
| | - Matthias Pumberger
- Berlin-Brandenburg School for Regenerative Therapies, Berlin, Germany; Center for Musculoskeletal Surgery, Charitè - Universitätsmedizin Berlin, Germany.
| | - Sven Geissler
- Julius Wolff Institute, Charité - Universitätsmedizin Berlin, Germany; Berlin-Brandenburg School for Regenerative Therapies, Berlin, Germany; Berlin-Brandenburg Center for Regenerative Therapies, Berlin, Germany.
| | - Georg N Duda
- Julius Wolff Institute, Charité - Universitätsmedizin Berlin, Germany; Berlin-Brandenburg School for Regenerative Therapies, Berlin, Germany; Berlin-Brandenburg Center for Regenerative Therapies, Berlin, Germany.
| |
Collapse
|
181
|
Rattray B, Argus C, Martin K, Northey J, Driller M. Is it time to turn our attention toward central mechanisms for post-exertional recovery strategies and performance? Front Physiol 2015; 6:79. [PMID: 25852568 PMCID: PMC4362407 DOI: 10.3389/fphys.2015.00079] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 02/27/2015] [Indexed: 11/23/2022] Open
Abstract
Key PointsCentral fatigue is accepted as a contributor to overall athletic performance, yet little research directly investigates post-exercise recovery strategies targeting the brain Current post-exercise recovery strategies likely impact on the brain through a range of mechanisms, but improvements to these strategies is needed Research is required to optimize post-exercise recovery with a focus on the brain
Post-exercise recovery has largely focused on peripheral mechanisms of fatigue, but there is growing acceptance that fatigue is also contributed to through central mechanisms which demands that attention should be paid to optimizing recovery of the brain. In this narrative review we assemble evidence for the role that many currently utilized recovery strategies may have on the brain, as well as potential mechanisms for their action. The review provides discussion of how common nutritional strategies as well as physical modalities and methods to reduce mental fatigue are likely to interact with the brain, and offer an opportunity for subsequent improved performance. We aim to highlight the fact that many recovery strategies have been designed with the periphery in mind, and that refinement of current methods are likely to provide improvements in minimizing brain fatigue. Whilst we offer a number of recommendations, it is evident that there are many opportunities for improving the research, and practical guidelines in this area.
Collapse
Affiliation(s)
- Ben Rattray
- Discipline of Sport and Exercise Science, Faculty of Health, University of Canberra Canberra, ACT, Australia ; University of Canberra Research Institute for Sport and Exercise, University of Canberra Canberra, ACT, Australia
| | - Christos Argus
- University of Canberra Research Institute for Sport and Exercise, University of Canberra Canberra, ACT, Australia
| | - Kristy Martin
- Discipline of Sport and Exercise Science, Faculty of Health, University of Canberra Canberra, ACT, Australia ; University of Canberra Research Institute for Sport and Exercise, University of Canberra Canberra, ACT, Australia
| | - Joseph Northey
- Discipline of Sport and Exercise Science, Faculty of Health, University of Canberra Canberra, ACT, Australia ; University of Canberra Research Institute for Sport and Exercise, University of Canberra Canberra, ACT, Australia
| | - Matthew Driller
- Department of Sport and Leisure Studies, The University of Waikato Hamilton, New Zealand
| |
Collapse
|
182
|
Kizil C, Kyritsis N, Brand M. Effects of inflammation on stem cells: together they strive? EMBO Rep 2015; 16:416-26. [PMID: 25739812 DOI: 10.15252/embr.201439702] [Citation(s) in RCA: 152] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 02/19/2015] [Indexed: 12/12/2022] Open
Abstract
Inflammation entails a complex set of defense mechanisms acting in concert to restore the homeostatic balance in organisms after damage or pathogen invasion. This immune response consists of the activity of various immune cells in a highly complex manner. Inflammation is a double-edged sword as it is reported to have both detrimental and beneficial consequences. In this review, we discuss the effects of inflammation on stem cell activity, focusing primarily on neural stem/progenitor cells in mammals and zebrafish. We also give a brief overview of the effects of inflammation on other stem cell compartments, exemplifying the positive and negative role of inflammation on stemness. The majority of the chronic diseases involve an unremitting phase of inflammation due to improper resolution of the initial pro-inflammatory response that impinges on the stem cell behavior. Thus, understanding the mechanisms of crosstalk between the inflammatory milieu and tissue-resident stem cells is an important basis for clinical efforts. Not only is it important to understand the effect of inflammation on stem cell activity for further defining the etiology of the diseases, but also better mechanistic understanding is essential to design regenerative therapies that aim at micromanipulating the inflammatory milieu to offset the negative effects and maximize the beneficial outcomes.
Collapse
Affiliation(s)
- Caghan Kizil
- German Centre for Neurodegenerative Diseases (DZNE) Dresden within the Helmholtz Association, Dresden, Germany DFG-Center for Regenerative Therapies Dresden, Cluster of Excellence (CRTD) of the Technische Universität Dresden, Dresden, Germany
| | - Nikos Kyritsis
- DFG-Center for Regenerative Therapies Dresden, Cluster of Excellence (CRTD) of the Technische Universität Dresden, Dresden, Germany
| | - Michael Brand
- DFG-Center for Regenerative Therapies Dresden, Cluster of Excellence (CRTD) of the Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
183
|
Kumar VA, Taylor NL, Shi S, Wickremasinghe NC, D'Souza RN, Hartgerink JD. Self-assembling multidomain peptides tailor biological responses through biphasic release. Biomaterials 2015; 52:71-8. [PMID: 25818414 DOI: 10.1016/j.biomaterials.2015.01.079] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 01/25/2015] [Accepted: 01/25/2015] [Indexed: 01/08/2023]
Abstract
Delivery of small molecules and drugs to tissues is a mainstay of several tissue engineering strategies. Next generation treatments focused on localized drug delivery offer a more effective means in dealing with refractory healing when compared to systemic approaches. Here we describe a novel multidomain peptide hydrogel that capitalizes on synthetic peptide chemistry, supramolecular self-assembly and cytokine delivery to tailor biological responses. This material is biomimetic, shows shear stress recovery and offers a nanofibrous matrix that sequesters cytokines. The biphasic pattern of cytokine release results in the spatio-temporal activation of THP-1 monocytes and macrophages. Furthermore, macrophage-material interactions are promoted without generation of a proinflammatory environment. Subcutaneous implantation of injectable scaffolds showed a marked increase in macrophage infiltration and polarization dictated by cytokine loading as early as 3 days, with complete scaffold resorption by day 14. Macrophage interaction and response to the peptide composite facilitated the (i) recruitment of monocytes/macrophages, (ii) sustained residence of immune cells until degradation, and (iii) promotion of a pro-resolution M2 environment. Our results suggest the potential use of this injectable cytokine loaded hydrogel scaffold in a variety of tissue engineering applications.
Collapse
Affiliation(s)
- Vivek A Kumar
- Department of Chemistry, Rice University, Houston, TX 77030, USA; Department of Bioengineering, Rice University, Houston, TX 77030, USA
| | - Nichole L Taylor
- Department of Chemistry, Rice University, Houston, TX 77030, USA; Department of Bioengineering, Rice University, Houston, TX 77030, USA
| | - Siyu Shi
- Department of Chemistry, Rice University, Houston, TX 77030, USA; Department of Bioengineering, Rice University, Houston, TX 77030, USA
| | - Navindee C Wickremasinghe
- Department of Chemistry, Rice University, Houston, TX 77030, USA; Department of Bioengineering, Rice University, Houston, TX 77030, USA
| | - Rena N D'Souza
- School of Dentistry, University of Utah, Salt Lake City, UT 84112, USA
| | - Jeffrey D Hartgerink
- Department of Chemistry, Rice University, Houston, TX 77030, USA; Department of Bioengineering, Rice University, Houston, TX 77030, USA.
| |
Collapse
|
184
|
Kornegay JN, Spurney CF, Nghiem PP, Brinkmeyer-Langford CL, Hoffman EP, Nagaraju K. Pharmacologic management of Duchenne muscular dystrophy: target identification and preclinical trials. ILAR J 2015; 55:119-49. [PMID: 24936034 DOI: 10.1093/ilar/ilu011] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked human disorder in which absence of the protein dystrophin causes degeneration of skeletal and cardiac muscle. For the sake of treatment development, over and above definitive genetic and cell-based therapies, there is considerable interest in drugs that target downstream disease mechanisms. Drug candidates have typically been chosen based on the nature of pathologic lesions and presumed underlying mechanisms and then tested in animal models. Mammalian dystrophinopathies have been characterized in mice (mdx mouse) and dogs (golden retriever muscular dystrophy [GRMD]). Despite promising results in the mdx mouse, some therapies have not shown efficacy in DMD. Although the GRMD model offers a higher hurdle for translation, dogs have primarily been used to test genetic and cellular therapies where there is greater risk. Failed translation of animal studies to DMD raises questions about the propriety of methods and models used to identify drug targets and test efficacy of pharmacologic intervention. The mdx mouse and GRMD dog are genetically homologous to DMD but not necessarily analogous. Subcellular species differences are undoubtedly magnified at the whole-body level in clinical trials. This problem is compounded by disparate cultures in clinical trials and preclinical studies, pointing to a need for greater rigor and transparency in animal experiments. Molecular assays such as mRNA arrays and genome-wide association studies allow identification of genetic drug targets more closely tied to disease pathogenesis. Genes in which polymorphisms have been directly linked to DMD disease progression, as with osteopontin, are particularly attractive targets.
Collapse
|
185
|
Bosurgi L, Brunelli S, Rigamonti E, Monno A, Manfredi AA, Rovere-Querini P. Vessel-associated myogenic precursors control macrophage activation and clearance of apoptotic cells. Clin Exp Immunol 2015; 179:62-7. [PMID: 24749786 DOI: 10.1111/cei.12356] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2014] [Indexed: 12/11/2022] Open
Abstract
Swift and regulated clearance of apoptotic cells prevents the accumulation of cell remnants in injured tissues and contributes to the shift of macrophages towards alternatively activated reparatory cells that sustain wound healing. Environmental signals, most of which are unknown, in turn control the efficiency of the clearance of apoptotic cells and as such determine whether tissues eventually heal. In this study we show that vessel-associated stem cells (mesoangioblasts) specifically modulate the expression of genes involved in the clearance of apoptotic cells and in macrophage alternative activation, including those of scavenger receptors and of molecules that bridge dying cells and phagocytes. Mesoangioblasts, but not immortalized myoblasts or neural precursor cells, enhance CD163 membrane expression in vitro as assessed by flow cytometry, indicating that the effect is specific. Mesoangioblasts transplanted in acutely or chronically injured skeletal muscles determine the expansion of the population of CD163(+) infiltrating macrophages and increase the extent of CD163 expression. Conversely, macrophages challenged with mesoangioblasts engulf significantly better apoptotic cells in vitro. Collectively, the data reveal a feed-forward loop between macrophages and vessel-associated stem cells, which has implications for the skeletal muscle homeostatic response to sterile injury and for diseases in which homeostasis is jeopardized, including muscle dystrophies and inflammatory myopathies.
Collapse
Affiliation(s)
- L Bosurgi
- Division of Regenerative Medicine, Stem Cells and Gene Therapy, IRCCS Ospedale San Raffaele, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | | | | | | | | | | |
Collapse
|
186
|
Fu X, Wang H, Hu P. Stem cell activation in skeletal muscle regeneration. Cell Mol Life Sci 2015; 72:1663-77. [PMID: 25572293 PMCID: PMC4412728 DOI: 10.1007/s00018-014-1819-5] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 12/21/2014] [Accepted: 12/22/2014] [Indexed: 12/31/2022]
Abstract
Muscle stem cell (satellite cell) activation post muscle injury is a transient and critical step in muscle regeneration. It is regulated by physiological cues, signaling molecules, and epigenetic regulatory factors. The mechanisms that coherently turn on the complex activation process shortly after trauma are just beginning to be illuminated. In this review, we will discuss the current knowledge of satellite cell activation regulation.
Collapse
Affiliation(s)
- Xin Fu
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | | | | |
Collapse
|
187
|
Fujita N, Ono M, Tomioka T, Deie M. Effects of hyperbaric oxygen at 1.25 atmospheres absolute with normal air on macrophage number and infiltration during rat skeletal muscle regeneration. PLoS One 2014; 9:e115685. [PMID: 25531909 PMCID: PMC4274106 DOI: 10.1371/journal.pone.0115685] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 12/01/2014] [Indexed: 12/17/2022] Open
Abstract
Use of mild hyperbaric oxygen less than 2 atmospheres absolute (2026.54 hPa) with normal air is emerging as a common complementary treatment for severe muscle injury. Although hyperbaric oxygen at over 2 atmospheres absolute with 100% O2 promotes healing of skeletal muscle injury, it is not clear whether mild hyperbaric oxygen is equally effective. The purpose of the present study was to investigate the impact of hyperbaric oxygen at 1.25 atmospheres absolute (1266.59 hPa) with normal air on muscle regeneration. The tibialis anterior muscle of male Wistar rats was injured by injection of bupivacaine hydrochloride, and rats were randomly assigned to a hyperbaric oxygen experimental group or to a non-hyperbaric oxygen control group. Immediately after the injection, rats were exposed to hyperbaric oxygen, and the treatment was continued for 28 days. The cross-sectional area of centrally nucleated muscle fibers was significantly larger in rats exposed to hyperbaric oxygen than in controls 5 and 7 days after injury. The number of CD68- or CD68- and CD206-positive cells was significantly higher in rats exposed to hyperbaric oxygen than in controls 24 h after injury. Additionally, tumor necrosis factor-α and interleukin-10 mRNA expression levels were significantly higher in rats exposed to hyperbaric oxygen than in controls 24 h after injury. The number of Pax7- and MyoD- or MyoD- and myogenin-positive nuclei per mm2 and the expression levels of these proteins were significantly higher in rats exposed to hyperbaric oxygen than in controls 5 days after injury. These results suggest that mild hyperbaric oxygen promotes skeletal muscle regeneration in the early phase after injury, possibly due to reduced hypoxic conditions leading to accelerated macrophage infiltration and phenotype transition. In conclusion, mild hyperbaric oxygen less than 2 atmospheres absolute with normal air is an appropriate support therapy for severe muscle injuries.
Collapse
Affiliation(s)
- Naoto Fujita
- Graduate School of Biomedicine and Health Sciences, Hiroshima University, Hiroshima City, Hiroshima, Japan
- Faculty of Medicine, Hiroshima University, Hiroshima City, Hiroshima, Japan
- * E-mail:
| | - Miharu Ono
- Graduate School of Biomedicine and Health Sciences, Hiroshima University, Hiroshima City, Hiroshima, Japan
| | - Tomoka Tomioka
- Graduate School of Biomedicine and Health Sciences, Hiroshima University, Hiroshima City, Hiroshima, Japan
| | - Masataka Deie
- Graduate School of Biomedicine and Health Sciences, Hiroshima University, Hiroshima City, Hiroshima, Japan
- Faculty of Medicine, Hiroshima University, Hiroshima City, Hiroshima, Japan
| |
Collapse
|
188
|
Gomez-Rodriguez V, Orbe J, Martinez-Aguilar E, Rodriguez JA, Fernandez-Alonso L, Serneels J, Bobadilla M, Perez-Ruiz A, Collantes M, Mazzone M, Paramo JA, Roncal C. Functional MMP-10 is required for efficient tissue repair after experimental hind limb ischemia. FASEB J 2014; 29:960-72. [PMID: 25414484 DOI: 10.1096/fj.14-259689] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
We studied the role of matrix metalloproteinase-10 (MMP-10) during skeletal muscle repair after ischemia using a model of femoral artery excision in wild-type (WT) and MMP-10 deficient (Mmp10(-/-)) mice. Functional changes were analyzed by small animal positron emission tomography and tissue morphology by immunohistochemistry. Gene expression and protein analysis were used to study the molecular mechanisms governed by MMP-10 in hypoxia. Early after ischemia, MMP-10 deficiency resulted in delayed tissue reperfusion (10%, P < 0.01) and in increased necrosis (2-fold, P < 0.01), neutrophil (4-fold, P < 0.01), and macrophage (1.5-fold, P < 0.01) infiltration. These differences at early time points resulted in delayed myotube regeneration in Mmp10(-/-) soleus at later stages (regenerating myofibers: 30 ± 9% WT vs. 68 ± 10% Mmp10(-/-), P < 0.01). The injection of MMP-10 into Mmp10(-/-) mice rescued the observed phenotype. A molecular analysis revealed higher levels of Cxcl1 mRNA (10-fold, P < 0.05) and protein (30%) in the ischemic Mmp10(-/-) muscle resulting from a lack of transcriptional inhibition by MMP-10. This was further confirmed using siRNA against MMP-10 in vivo. Our results demonstrate an important role of MMP-10 for proper muscle repair after ischemia, and suggest that chemokine regulation such as Cxcl1 by MMP-10 is involved in muscle regeneration.
Collapse
Affiliation(s)
- Violeta Gomez-Rodriguez
- *Laboratory of Atherothrombosis, Division of Cardiovascular Sciences, and Cell Therapy Area, Division of Cancer, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain; Department of Vascular Surgery, Complejo Hopitalario de Navarra, Pamplona, Spain; Laboratory of Molecular Oncology and Angiogenesis, Vesalius Research Center, VIB, Leuven, Belgium; Laboratory of Molecular Oncology and Angiogenesis, Vesalius Research Center, Department of Oncology, KU Leuven, Leuven, Belgium; and Small Animal Imaging Research Unit, CIMA and Clínica Universidad de Navarra, Pamplona, Spain
| | - Josune Orbe
- *Laboratory of Atherothrombosis, Division of Cardiovascular Sciences, and Cell Therapy Area, Division of Cancer, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain; Department of Vascular Surgery, Complejo Hopitalario de Navarra, Pamplona, Spain; Laboratory of Molecular Oncology and Angiogenesis, Vesalius Research Center, VIB, Leuven, Belgium; Laboratory of Molecular Oncology and Angiogenesis, Vesalius Research Center, Department of Oncology, KU Leuven, Leuven, Belgium; and Small Animal Imaging Research Unit, CIMA and Clínica Universidad de Navarra, Pamplona, Spain
| | - Esther Martinez-Aguilar
- *Laboratory of Atherothrombosis, Division of Cardiovascular Sciences, and Cell Therapy Area, Division of Cancer, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain; Department of Vascular Surgery, Complejo Hopitalario de Navarra, Pamplona, Spain; Laboratory of Molecular Oncology and Angiogenesis, Vesalius Research Center, VIB, Leuven, Belgium; Laboratory of Molecular Oncology and Angiogenesis, Vesalius Research Center, Department of Oncology, KU Leuven, Leuven, Belgium; and Small Animal Imaging Research Unit, CIMA and Clínica Universidad de Navarra, Pamplona, Spain
| | - Jose A Rodriguez
- *Laboratory of Atherothrombosis, Division of Cardiovascular Sciences, and Cell Therapy Area, Division of Cancer, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain; Department of Vascular Surgery, Complejo Hopitalario de Navarra, Pamplona, Spain; Laboratory of Molecular Oncology and Angiogenesis, Vesalius Research Center, VIB, Leuven, Belgium; Laboratory of Molecular Oncology and Angiogenesis, Vesalius Research Center, Department of Oncology, KU Leuven, Leuven, Belgium; and Small Animal Imaging Research Unit, CIMA and Clínica Universidad de Navarra, Pamplona, Spain
| | - Leopoldo Fernandez-Alonso
- *Laboratory of Atherothrombosis, Division of Cardiovascular Sciences, and Cell Therapy Area, Division of Cancer, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain; Department of Vascular Surgery, Complejo Hopitalario de Navarra, Pamplona, Spain; Laboratory of Molecular Oncology and Angiogenesis, Vesalius Research Center, VIB, Leuven, Belgium; Laboratory of Molecular Oncology and Angiogenesis, Vesalius Research Center, Department of Oncology, KU Leuven, Leuven, Belgium; and Small Animal Imaging Research Unit, CIMA and Clínica Universidad de Navarra, Pamplona, Spain
| | - Jens Serneels
- *Laboratory of Atherothrombosis, Division of Cardiovascular Sciences, and Cell Therapy Area, Division of Cancer, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain; Department of Vascular Surgery, Complejo Hopitalario de Navarra, Pamplona, Spain; Laboratory of Molecular Oncology and Angiogenesis, Vesalius Research Center, VIB, Leuven, Belgium; Laboratory of Molecular Oncology and Angiogenesis, Vesalius Research Center, Department of Oncology, KU Leuven, Leuven, Belgium; and Small Animal Imaging Research Unit, CIMA and Clínica Universidad de Navarra, Pamplona, Spain
| | - Miriam Bobadilla
- *Laboratory of Atherothrombosis, Division of Cardiovascular Sciences, and Cell Therapy Area, Division of Cancer, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain; Department of Vascular Surgery, Complejo Hopitalario de Navarra, Pamplona, Spain; Laboratory of Molecular Oncology and Angiogenesis, Vesalius Research Center, VIB, Leuven, Belgium; Laboratory of Molecular Oncology and Angiogenesis, Vesalius Research Center, Department of Oncology, KU Leuven, Leuven, Belgium; and Small Animal Imaging Research Unit, CIMA and Clínica Universidad de Navarra, Pamplona, Spain
| | - Ana Perez-Ruiz
- *Laboratory of Atherothrombosis, Division of Cardiovascular Sciences, and Cell Therapy Area, Division of Cancer, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain; Department of Vascular Surgery, Complejo Hopitalario de Navarra, Pamplona, Spain; Laboratory of Molecular Oncology and Angiogenesis, Vesalius Research Center, VIB, Leuven, Belgium; Laboratory of Molecular Oncology and Angiogenesis, Vesalius Research Center, Department of Oncology, KU Leuven, Leuven, Belgium; and Small Animal Imaging Research Unit, CIMA and Clínica Universidad de Navarra, Pamplona, Spain
| | - Maria Collantes
- *Laboratory of Atherothrombosis, Division of Cardiovascular Sciences, and Cell Therapy Area, Division of Cancer, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain; Department of Vascular Surgery, Complejo Hopitalario de Navarra, Pamplona, Spain; Laboratory of Molecular Oncology and Angiogenesis, Vesalius Research Center, VIB, Leuven, Belgium; Laboratory of Molecular Oncology and Angiogenesis, Vesalius Research Center, Department of Oncology, KU Leuven, Leuven, Belgium; and Small Animal Imaging Research Unit, CIMA and Clínica Universidad de Navarra, Pamplona, Spain
| | - Massimiliano Mazzone
- *Laboratory of Atherothrombosis, Division of Cardiovascular Sciences, and Cell Therapy Area, Division of Cancer, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain; Department of Vascular Surgery, Complejo Hopitalario de Navarra, Pamplona, Spain; Laboratory of Molecular Oncology and Angiogenesis, Vesalius Research Center, VIB, Leuven, Belgium; Laboratory of Molecular Oncology and Angiogenesis, Vesalius Research Center, Department of Oncology, KU Leuven, Leuven, Belgium; and Small Animal Imaging Research Unit, CIMA and Clínica Universidad de Navarra, Pamplona, Spain
| | - Jose A Paramo
- *Laboratory of Atherothrombosis, Division of Cardiovascular Sciences, and Cell Therapy Area, Division of Cancer, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain; Department of Vascular Surgery, Complejo Hopitalario de Navarra, Pamplona, Spain; Laboratory of Molecular Oncology and Angiogenesis, Vesalius Research Center, VIB, Leuven, Belgium; Laboratory of Molecular Oncology and Angiogenesis, Vesalius Research Center, Department of Oncology, KU Leuven, Leuven, Belgium; and Small Animal Imaging Research Unit, CIMA and Clínica Universidad de Navarra, Pamplona, Spain
| | - Carmen Roncal
- *Laboratory of Atherothrombosis, Division of Cardiovascular Sciences, and Cell Therapy Area, Division of Cancer, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain; Department of Vascular Surgery, Complejo Hopitalario de Navarra, Pamplona, Spain; Laboratory of Molecular Oncology and Angiogenesis, Vesalius Research Center, VIB, Leuven, Belgium; Laboratory of Molecular Oncology and Angiogenesis, Vesalius Research Center, Department of Oncology, KU Leuven, Leuven, Belgium; and Small Animal Imaging Research Unit, CIMA and Clínica Universidad de Navarra, Pamplona, Spain
| |
Collapse
|
189
|
Yang HL, Lin SW, Lee CC, Lin KY, Liao CH, Yang TY, Wang HM, Huang HC, Wu CR, Hseu YC. Induction of Nrf2-mediated genes by Antrodia salmonea inhibits ROS generation and inflammatory effects in lipopolysaccharide-stimulated RAW264.7 macrophages. Food Funct 2014; 6:230-41. [PMID: 25380370 DOI: 10.1039/c4fo00869c] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Antrodia salmonea (AS), a well-known medicinal mushroom in Taiwan, has been reported to exhibit anti-oxidant, anti-angiogenic, anti-atherogenic, and anti-inflammatory effects. In the present study, we investigated the activation of Nrf2-mediated antioxidant genes in RAW264.7 macrophages by the fermented culture broth of AS, studied the resulting protection against lipopolysaccharide (LPS)-stimulated inflammation, and revealed the molecular mechanisms underlying these protective effects. We found that non-cytotoxic concentrations of AS (25-100 μg mL⁻¹) protected macrophages from LPS-induced cell death and ROS generation in a dose-dependent manner. The antioxidant potential of AS was directly correlated with the increased expression of the antioxidant genes HO-1, NQO-1, and γ-GCLC, as well as the level of intracellular GSH followed by an increase in the nuclear translocation and transcriptional activation of the Nrf2-ARE pathway. Furthermore, Nrf2 knockdown diminished the protective effects of AS, as evidenced by the increased production of pro-inflammatory cytokines and chemokines, including PGE₂, NO, TNF-α, and IL-1β, in LPS-stimulated macrophages. Notably, AS treatment significantly inhibited LPS-induced ICAM-1 expression in macrophages. Our data suggest that the anti-inflammatory potential of Antrodia salmonea is mediated by the activation of Nrf2-dependent antioxidant defense mechanisms. Results support the traditional usage of this beneficial mushroom for the treatment of free radical-related diseases and inflammation.
Collapse
Affiliation(s)
- Hsin-Ling Yang
- Institute of Nutrition, China Medical University, Taichung 40402, Taiwan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
190
|
Sánchez M, Anitua E, Delgado D, Sánchez P, Orive G, Padilla S. Muscle repair: platelet-rich plasma derivates as a bridge from spontaneity to intervention. Injury 2014; 45 Suppl 4:S7-14. [PMID: 25384475 DOI: 10.1016/s0020-1383(14)70004-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Muscle injuries account for between 10% and 55% of all sporting injuries. Although the skeletal muscle is a plastic organ capable of responding efficiently to environmental changes, the appropriate treatment of muscle injuries remains a daunting clinical challenge in sports medicine. There is considerable evidence to indicate that growth factors, such as transforming growth factor-β (TGFβ), hepatocyte growth factor (HGF) or insulin-like growth factor (IGF), and fibrin matrix are key in cellular events required for muscle repair and regeneration, namely myogenesis, angiogenesis and fibrogenesis. An innovative biological approach to the treatment of muscle injuries is the application of Plasma Rich in Growth Factors (PRGF) in intramuscular infiltrations. PRGF delivers growth factors, cytokines and adhesive proteins present in platelets and plasma, as well as other biologically-active proteins conveyed by the plasma, such as fibrinogen, prothrombin and fibronectin. This autologous, mimetic biomaterial embedded with a pool of growth factors acts as a smart dynamic scaffold, and should be applied taking into account a biological approach. A clinical trial is required to assess the functional repair outcome of PRGF infiltrations in muscle injuries.
Collapse
Affiliation(s)
- Mikel Sánchez
- Arthroscopic Surgery Unit (ASU). Hospital Vithas San Jose. C/Beato Tomás de Zumárraga 10, 01008 Vitoria-Gasteiz, Spain; ASU Research AIE. Hospital Vithas San Jose. C/Beato Tomás de Zumárraga 10, 01008 Vitoria-Gasteiz, Spain
| | - Eduardo Anitua
- Biotechnology Institute (BTI) Vitoria, Vitoria-Gasteiz, Spain; Foundation Eduardo Anitua. C/José María Cagigal 19, 01007 Vitoria-Gasteiz, Spain
| | - Diego Delgado
- ASU Research AIE. Hospital Vithas San Jose. C/Beato Tomás de Zumárraga 10, 01008 Vitoria-Gasteiz, Spain
| | - Pello Sánchez
- ASU Research AIE. Hospital Vithas San Jose. C/Beato Tomás de Zumárraga 10, 01008 Vitoria-Gasteiz, Spain
| | - Gorka Orive
- Foundation Eduardo Anitua. C/José María Cagigal 19, 01007 Vitoria-Gasteiz, Spain
| | - Sabino Padilla
- Biotechnology Institute (BTI) Vitoria, Vitoria-Gasteiz, Spain; Foundation Eduardo Anitua. C/José María Cagigal 19, 01007 Vitoria-Gasteiz, Spain.
| |
Collapse
|
191
|
Makita N, Hizukuri Y, Yamashiro K, Murakawa M, Hayashi Y. IL-10 enhances the phenotype of M2 macrophages induced by IL-4 and confers the ability to increase eosinophil migration. Int Immunol 2014; 27:131-41. [DOI: 10.1093/intimm/dxu090] [Citation(s) in RCA: 121] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
192
|
The need to more precisely define aspects of skeletal muscle regeneration. Int J Biochem Cell Biol 2014; 56:56-65. [PMID: 25242742 DOI: 10.1016/j.biocel.2014.09.010] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 09/04/2014] [Accepted: 09/08/2014] [Indexed: 12/11/2022]
Abstract
A more precise definition of the term 'skeletal muscle regeneration' is required to reduce confusion and misconceptions. In this paper the term is used only for events that follow myofibre necrosis, to result in myogenesis and new muscle formation: other key events include early inflammation and revascularisation, and later fibrosis and re-innervation. The term 'muscle regeneration' is sometimes used casually for situations that do not involve myonecrosis; such as restoration of muscle mass by hypertrophy after atrophy, and other forms of damage to muscle tissue components. These situations are excluded from the definition in this paper which is focussed on mammalian muscles with the long-term aim of clinical translation to enhance new muscle formation after acute or chronic injury or during surgery to replace whole muscles. The paper briefly outlines the cellular events involved in myogenesis during development and post-natal muscle growth, discusses the role of satellite cells in mature normal muscles, and the likely incidence of myofibre necrosis/regeneration in healthy ageing mammals (even when subjected to exercise). The importance of the various components of regeneration is outlined to emphasise that problems in each of these aspects can influence overall new muscle formation; thus care is needed for correct interpretation of altered kinetics. Various markers used to identify regenerating myofibres are critically discussed and, since these can all occur in other conditions, caution is required for accurate interpretation of these cellular events. Finally, clinical situations are outlined where there is a need to enhance skeletal muscle regeneration: these include acute and chronic injuries or transplantation with bioengineering to form new muscles, therapeutic approaches to muscular dystrophies, and comment on proposed stem cell therapies to reduce age-related loss of muscle mass and function. This article is part of a directed issue entitled: Regenerative Medicine: the challenge of translation.
Collapse
|
193
|
Hu L, Klein JD, Hassounah F, Cai H, Zhang C, Xu P, Wang XH. Low-frequency electrical stimulation attenuates muscle atrophy in CKD--a potential treatment strategy. J Am Soc Nephrol 2014; 26:626-35. [PMID: 25228359 DOI: 10.1681/asn.2014020144] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Effective therapeutic strategies to treat CKD-induced muscle atrophy are urgently needed. Low-frequency electrical stimulation (LFES) may be effective in preventing muscle atrophy, because LFES is an acupuncture technique that mimics resistance exercise by inducing muscle contraction. To test this hypothesis, we treated 5/6-nephrectomized mice (CKD mice) and control mice with LFES for 15 days. LFES prevented soleus and extensor digitorum longus muscle weight loss and loss of hind-limb muscle grip in CKD mice. LFES countered the CKD-induced decline in the IGF-1 signaling pathway and led to increases in markers of protein synthesis and myogenesis and improvement in muscle protein metabolism. In control mice, we observed an acute response phase immediately after LFES, during which the expression of inflammatory cytokines (IFN-γ and IL-6) increased. Expression of the M1 macrophage marker IL-1β also increased acutely, but expression of the M2 marker arginase-1 increased 2 days after initiation of LFES, paralleling the change in IGF-1. In muscle cross-sections of LFES-treated mice, arginase-1 colocalized with IGF-1. Additionally, expression of microRNA-1 and -206, which inhibits IGF-1 translation, decreased in the acute response phase after LFES and increased at a later phase. We conclude that LFES ameliorates CKD-induced skeletal muscle atrophy by upregulation of the IGF-1 signaling pathway, which improves protein metabolism and promotes myogenesis. The upregulation of IGF-1 may be mediated by decreased expression of microRNA-1 and -206 and/or activation of M2 macrophages.
Collapse
Affiliation(s)
- Li Hu
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia; Acumox and Tuina Research Section, College of Acumox and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, China; and
| | - Janet D Klein
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia
| | - Faten Hassounah
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia
| | - Hui Cai
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia; Section of Nephrology, Atlanta Veterans Administration Medical Center, Decatur, Georgia
| | - Cong Zhang
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia
| | - Ping Xu
- Acumox and Tuina Research Section, College of Acumox and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, China; and
| | - Xiaonan H Wang
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia;
| |
Collapse
|
194
|
Sawano S, Suzuki T, Do MKQ, Ohtsubo H, Mizunoya W, Ikeuchi Y, Tatsumi R. Supplementary immunocytochemistry of hepatocyte growth factor production in activated macrophages early in muscle regeneration. Anim Sci J 2014; 85:994-1000. [DOI: 10.1111/asj.12264] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 06/06/2014] [Indexed: 12/31/2022]
Affiliation(s)
- Shoko Sawano
- Department of Animal and Marine Bioresource Sciences; Graduate School of Agriculture; Kyushu University; Fukuoka Japan
| | - Takahiro Suzuki
- Department of Animal and Marine Bioresource Sciences; Graduate School of Agriculture; Kyushu University; Fukuoka Japan
| | - Mai-Khoi Q. Do
- Department of Animal and Marine Bioresource Sciences; Graduate School of Agriculture; Kyushu University; Fukuoka Japan
| | - Hideaki Ohtsubo
- Department of Animal and Marine Bioresource Sciences; Graduate School of Agriculture; Kyushu University; Fukuoka Japan
| | - Wataru Mizunoya
- Department of Animal and Marine Bioresource Sciences; Graduate School of Agriculture; Kyushu University; Fukuoka Japan
| | - Yoshihide Ikeuchi
- Department of Animal and Marine Bioresource Sciences; Graduate School of Agriculture; Kyushu University; Fukuoka Japan
| | - Ryuichi Tatsumi
- Department of Animal and Marine Bioresource Sciences; Graduate School of Agriculture; Kyushu University; Fukuoka Japan
| |
Collapse
|
195
|
Pessina P, Cabrera D, Morales MG, Riquelme CA, Gutiérrez J, Serrano AL, Brandan E, Muñoz-Cánoves P. Novel and optimized strategies for inducing fibrosis in vivo: focus on Duchenne Muscular Dystrophy. Skelet Muscle 2014; 4:7. [PMID: 25157321 PMCID: PMC4142391 DOI: 10.1186/2044-5040-4-7] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 01/20/2014] [Indexed: 11/13/2022] Open
Abstract
Background Fibrosis, an excessive collagen accumulation, results in scar formation, impairing function of vital organs and tissues. Fibrosis is a hallmark of muscular dystrophies, including the lethal Duchenne muscular dystrophy (DMD), which remains incurable. Substitution of muscle by fibrotic tissue also complicates gene/cell therapies for DMD. Yet, no optimal models to study muscle fibrosis are available. In the widely used mdx mouse model for DMD, extensive fibrosis develops in the diaphragm only at advanced adulthood, and at about two years of age in the ‘easy-to-access’ limb muscles, thus precluding fibrosis research and the testing of novel therapies. Methods We developed distinct experimental strategies, ranging from chronic exercise to increasing muscle damage on limb muscles of young mdx mice, by myotoxin injection, surgically induced trauma (laceration or denervation) or intramuscular delivery of profibrotic growth factors (such as TGFβ). We also extended these approaches to muscle of normal non-dystrophic mice. Results These strategies resulted in advanced and enhanced muscle fibrosis in young mdx mice, which persisted over time, and correlated with reduced muscle force, thus mimicking the severe DMD phenotype. Furthermore, increased fibrosis was also obtained by combining these procedures in muscles of normal mice, mirroring aberrant repair after severe trauma. Conclusions We have developed new and improved experimental strategies to accelerate and enhance muscle fibrosis in vivo. These strategies will allow rapidly assessing fibrosis in the easily accessible limb muscles of young mdx mice, without necessarily having to use old animals. The extension of these fibrogenic regimes to the muscle of non-dystrophic wild-type mice will allow fibrosis assessment in a wide array of pre-existing transgenic mouse lines, which in turn will facilitate understanding the mechanisms of fibrogenesis. These strategies should improve our ability to combat fibrosis-driven dystrophy progression and aberrant regeneration.
Collapse
Affiliation(s)
- Patrizia Pessina
- Cell Biology Group, Department of Experimental and Health Sciences, CIBER on Neurodegenerative Diseases (CIBERNED), Pompeu Fabra University (UPF), Dr. Aiguader, 88, 08003 Barcelona, Spain
| | - Daniel Cabrera
- Department of Cell and Molecular Biology, Catholic University of Chile, Avenida Libertador Bernardo O'Higgins, 340, Santiago, Chile
| | - María Gabriela Morales
- Department of Cell and Molecular Biology, Catholic University of Chile, Avenida Libertador Bernardo O'Higgins, 340, Santiago, Chile
| | - Cecilia A Riquelme
- Department of Cell and Molecular Biology, Catholic University of Chile, Avenida Libertador Bernardo O'Higgins, 340, Santiago, Chile
| | - Jaime Gutiérrez
- Department of Cell and Molecular Biology, Catholic University of Chile, Avenida Libertador Bernardo O'Higgins, 340, Santiago, Chile
| | - Antonio L Serrano
- Cell Biology Group, Department of Experimental and Health Sciences, CIBER on Neurodegenerative Diseases (CIBERNED), Pompeu Fabra University (UPF), Dr. Aiguader, 88, 08003 Barcelona, Spain
| | - Enrique Brandan
- Department of Cell and Molecular Biology, Catholic University of Chile, Avenida Libertador Bernardo O'Higgins, 340, Santiago, Chile
| | - Pura Muñoz-Cánoves
- Cell Biology Group, Department of Experimental and Health Sciences, CIBER on Neurodegenerative Diseases (CIBERNED), Pompeu Fabra University (UPF), Dr. Aiguader, 88, 08003 Barcelona, Spain ; Institució Catalana de Recerca i Estudis Avançats (ICREA), Dr. Aiguader, 88, 08003 Barcelona, Spain
| |
Collapse
|
196
|
Huang RY, Lin JH, He XH, Li X, Lu CL, Zhou YY, Cai J, He YT. Anti-inflammatory activity of extracts of Bushen-Qiangdu-Zhilv decoction, a Chinese medicinal formula, in M1-polarized RAW264.7. Altern Ther Health Med 2014; 14:268. [PMID: 25070190 PMCID: PMC4132911 DOI: 10.1186/1472-6882-14-268] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 07/17/2014] [Indexed: 12/03/2022]
Abstract
Background Bushen-Qiangdu-Zhilv Decoction (BQZ) is one of famous traditional Chinese medical formula for treating ankylosing spondylitis (AS). However, the mechanisms underlying effects of BQZ remains unknown. Pro-inflammatory cytokines, tumor necrosis factor (TNF)-α and interleukin (IL)-1, play an important role in AS. We therefore evaluated if BQZ could affect the expression of these cytokines. Methods Crude extracts were prepared and fractioned with petroleum ether (PE), ethyl acetate (EA), n-butanol (BU) and finally water (ACE). The stability of the extracts was confirmed by high-pressure liquid chromatography (HPLC) analysis. M1-polarized RAW264.7 was induced and subsequently treated with BQZ extracts. Quantitative real-time PCR experiments were performed to measure mRNA expression of TNF-α and IL-1. Results It was found that TNF-α could be significantly suppressed by ACE extracts, whereas IL-1 was dramatically inhibited by BU extracts, which was further confirmed by dose-dependent experiments. Importantly, MTS assays showed that both ACE and BU extracts had a low cytotoxicity. Conclusion Altogether, our study indicates that BQZ decoction exerts anti-AS effects via its anti-inflammatory activity and may have a low side-effect. Further analysis of the extracts of BQZ decoction could lead to a discovery of some novel drugs adding to therapeutic strategy for AS patients.
Collapse
|
197
|
Ben-Mordechai T, Palevski D, Glucksam-Galnoy Y, Elron-Gross I, Margalit R, Leor J. Targeting macrophage subsets for infarct repair. J Cardiovasc Pharmacol Ther 2014; 20:36-51. [PMID: 24938456 DOI: 10.1177/1074248414534916] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Macrophages are involved in every cardiovascular disease and are an attractive therapeutic target. Macrophage activation is complex and can be either beneficial or deleterious, depending upon its mode of action, its timing, and its duration. An important macrophage characteristic is its plasticity, which enables it to switch from one subset to another. Macrophages, which regulate healing and repair after myocardial infarction, have become a major target for both treatment and diagnosis (theranostic). The aim of the present review is to describe the recent discoveries related to targeting and modulating of macrophage function to improve infarct repair. We will briefly review macrophage polarization, plasticity, heterogeneity, their role in infarct repair, regeneration, and cross talk with mesenchymal cells. Particularly, we will focus on the potential of macrophage targeting in situ by liposomes. The ability to modulate macrophage function could delineate pathways to reactivate the endogenous programs of myocardial regeneration. This will eventually lead to development of small molecules or biologics to enhance the endogenous programs of regeneration and repair.
Collapse
Affiliation(s)
- Tamar Ben-Mordechai
- Sackler Faculty of Medicine, Neufeld Cardiac Research Institute, Tel Aviv University, Tel Aviv, Israel Tamman Cardiovascular Research Institute, Leviev Heart Center, Sheba Medical Center, Tel-hashomer, Israel Sheba Center for Regenerative Medicine, Stem Cell, and Tissue Engineering, Tel-Hashomer, Israel
| | - Dahlia Palevski
- Sackler Faculty of Medicine, Neufeld Cardiac Research Institute, Tel Aviv University, Tel Aviv, Israel Tamman Cardiovascular Research Institute, Leviev Heart Center, Sheba Medical Center, Tel-hashomer, Israel Sheba Center for Regenerative Medicine, Stem Cell, and Tissue Engineering, Tel-Hashomer, Israel
| | - Yifat Glucksam-Galnoy
- Department of Biochemistry and Molecular Biology, the George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Inbar Elron-Gross
- Department of Biochemistry and Molecular Biology, the George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Rimona Margalit
- Department of Biochemistry and Molecular Biology, the George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Jonathan Leor
- Sackler Faculty of Medicine, Neufeld Cardiac Research Institute, Tel Aviv University, Tel Aviv, Israel Tamman Cardiovascular Research Institute, Leviev Heart Center, Sheba Medical Center, Tel-hashomer, Israel Sheba Center for Regenerative Medicine, Stem Cell, and Tissue Engineering, Tel-Hashomer, Israel
| |
Collapse
|
198
|
From innate to adaptive immune response in muscular dystrophies and skeletal muscle regeneration: the role of lymphocytes. BIOMED RESEARCH INTERNATIONAL 2014; 2014:438675. [PMID: 25028653 PMCID: PMC4083765 DOI: 10.1155/2014/438675] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2014] [Accepted: 05/02/2014] [Indexed: 12/04/2022]
Abstract
Skeletal muscle is able to restore contractile functionality after injury thanks to its ability to regenerate. Following muscle necrosis, debris is removed by macrophages, and muscle satellite cells (MuSCs), the muscle stem cells, are activated and subsequently proliferate, migrate, and form muscle fibers restoring muscle functionality. In most muscle dystrophies (MDs), MuSCs fail to properly proliferate, differentiate, or replenish the stem cell compartment, leading to fibrotic deposition. However, besides MuSCs, interstitial nonmyogenic cells and inflammatory cells also play a key role in orchestrating muscle repair. A complete understanding of the complexity of these mechanisms should allow the design of interventions to attenuate MDs pathology without disrupting regenerative processes. In this review we will focus on the contribution of immune cells in the onset and progression of MDs, with particular emphasis on Duchenne muscular dystrophy (DMD). We will briefly summarize the current knowledge and recent advances made in our understanding of the involvement of different innate immune cells in MDs and will move on to critically evaluate the possible role of cell populations within the acquired immune response. Revisiting previous observations in the light of recent evidence will likely change our current view of the onset and progression of the disease.
Collapse
|
199
|
Teodori L, Costa A, Marzio R, Perniconi B, Coletti D, Adamo S, Gupta B, Tarnok A. Native extracellular matrix: a new scaffolding platform for repair of damaged muscle. Front Physiol 2014; 5:218. [PMID: 24982637 PMCID: PMC4058757 DOI: 10.3389/fphys.2014.00218] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 05/22/2014] [Indexed: 11/17/2022] Open
Abstract
Effective clinical treatments for volumetric muscle loss resulting from traumatic injury or resection of a large amount of muscle mass are not available to date. Tissue engineering may represent an alternative treatment approach. Decellularization of tissues and whole organs is a recently introduced platform technology for creating scaffolding materials for tissue engineering and regenerative medicine. The muscle stem cell niche is composed of a three-dimensional architecture of fibrous proteins, proteoglycans, and glycosaminoglycans, synthesized by the resident cells that form an intricate extracellular matrix (ECM) network in equilibrium with the surrounding cells and growth factors. A consistent body of evidence indicates that ECM proteins regulate stem cell differentiation and renewal and are highly relevant to tissue engineering applications. The ECM also provides a supportive medium for blood or lymphatic vessels and for nerves. Thus, the ECM is the nature's ideal biological scaffold material. ECM-based bioscaffolds can be recellularized to create potentially functional constructs as a regenerative medicine strategy for organ replacement or tissue repopulation. This article reviews current strategies for the repair of damaged muscle using bioscaffolds obtained from animal ECM by decellularization of small intestinal submucosa (SIS), urinary bladder mucosa (UB), and skeletal muscle, and proposes some innovative approaches for the application of such strategies in the clinical setting.
Collapse
Affiliation(s)
- Laura Teodori
- UTAPRAD-DIM, ENEA Frascati Rome, Italy ; Fondazione San Raffaele Ceglie Messapica, Italy
| | - Alessandra Costa
- Fondazione San Raffaele Ceglie Messapica, Italy ; Department of Surgery, McGowan Institute, University of Pittsburgh Medical Center Pittsburgh, PA, USA
| | - Rosa Marzio
- Fondazione San Raffaele Ceglie Messapica, Italy
| | - Barbara Perniconi
- UMR 8256 CNRS Biology of Adaptation and Aging, University Pierre et Marie Curie Paris 06 Paris, France
| | - Dario Coletti
- UMR 8256 CNRS Biology of Adaptation and Aging, University Pierre et Marie Curie Paris 06 Paris, France ; Section of Histology and Medical Embryology, Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome Rome, Italy
| | - Sergio Adamo
- Section of Histology and Medical Embryology, Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome Rome, Italy
| | - Bhuvanesh Gupta
- Department of Textile Technology, Indian Institute of Technology New Delhi, India
| | - Attila Tarnok
- Department of Pediatric Cardiology, Heart Centre Leipzig, and Translational Centre for Regenerative Medicine, University of Leipzig Leipzig, Germany
| |
Collapse
|
200
|
Maves L. Recent advances using zebrafish animal models for muscle disease drug discovery. Expert Opin Drug Discov 2014; 9:1033-45. [PMID: 24931439 DOI: 10.1517/17460441.2014.927435] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
INTRODUCTION Animal models have enabled great progress in the discovery and understanding of pharmacological approaches for treating muscle diseases like Duchenne muscular dystrophy. AREAS COVERED With this article, the author provides the reader with a description of the zebrafish animal model, which has been employed to identify and study pharmacological approaches to muscle disease. In particular, the author focuses on how both large-scale chemical screens and targeted drug treatment studies have established zebrafish as an important model for muscle disease drug discovery. EXPERT OPINION There are a number of opportunities arising for the use of zebrafish models for further developing pharmacological approaches to muscle diseases, including studying drug combination therapies and utilizing genome editing to engineer zebrafish muscle disease models. It is the author's particular belief that the availability of a wide range of zebrafish transgenic strains for labeling immune cell types, combined with live imaging and drug treatment of muscle disease models, should allow for new elegant studies demonstrating how pharmacological approaches might influence inflammation and the immune response in muscle disease.
Collapse
Affiliation(s)
- Lisa Maves
- University of Washington School of Medicine, Department of Pediatrics, Division of Cardiology , Seattle, WA , USA
| |
Collapse
|