1
|
Tang T, Zhong W, Tang P, Dai R, Guo J, Gao Z. Linalool combats Saprolegnia parasitica infections through direct killing of microbes and modulation of host immune system. eLife 2025; 13:RP100393. [PMID: 40183210 PMCID: PMC11970904 DOI: 10.7554/elife.100393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025] Open
Abstract
Saprolegnia parasitica is one of the most virulent oomycete species in freshwater aquatic environments, causing severe saprolegniasis and leading to significant economic losses in the aquaculture industry. Thus far, the prevention and control of saprolegniasis face a shortage of medications. Linalool, a natural antibiotic alternative found in various essential oils, exhibits promising antimicrobial activity against a wide range of pathogens. In this study, the specific role of linalool in protecting S. parasitica infection at both in vitro and in vivo levels was investigated. Linalool showed multifaceted anti-oomycetes potential by both of antimicrobial efficacy and immunomodulatory efficacy. For in vitro test, linalool exhibited strong anti-oomycetes activity and mode of action included: (1) Linalool disrupted the cell membrane of the mycelium, causing the intracellular components leak out; (2) Linalool prohibited ribosome function, thereby inhibiting protein synthesis and ultimately affecting mycelium growth. Surprisingly, meanwhile we found the potential immune protective mechanism of linalool in the in vivo test: (1) Linalool enhanced the complement and coagulation system which in turn activated host immune defense and lysate S. parasitica cells; (2) Linalool promoted wound healing, tissue repair, and phagocytosis to cope with S. parasitica infection; (3) Linalool positively modulated the immune response by increasing the abundance of beneficial Actinobacteriota; (4) Linalool stimulated the production of inflammatory cytokines and chemokines to lyse S. parasitica cells. In all, our findings showed that linalool possessed multifaceted anti-oomycetes potential which would be a promising natural antibiotic alternative to cope with S. parasitica infection in the aquaculture industry.
Collapse
Affiliation(s)
- Tao Tang
- Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Fisheries College, Hunan Agricultural UniversityChangshaChina
| | - Weiming Zhong
- Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Fisheries College, Hunan Agricultural UniversityChangshaChina
| | - Puyu Tang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of SciencesWuhanChina
| | - Rongsi Dai
- Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Fisheries College, Hunan Agricultural UniversityChangshaChina
| | - Jiajing Guo
- Hunan Agriculture Product Processing Institute, Hunan Academy of Agricultural SciencesChangshaChina
| | - Zhipeng Gao
- Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Fisheries College, Hunan Agricultural UniversityChangshaChina
| |
Collapse
|
2
|
Courvan EMC, Parker RR. Hypoxia and inflammation induce synergistic transcriptome turnover in macrophages. Cell Rep 2024; 43:114452. [PMID: 38968068 DOI: 10.1016/j.celrep.2024.114452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 04/24/2024] [Accepted: 06/21/2024] [Indexed: 07/07/2024] Open
Abstract
Macrophages are effector immune cells that experience substantial changes to oxygenation when transiting through tissues, especially when entering tumors or infected wounds. How hypoxia alters gene expression and macrophage effector function at the post-transcriptional level remains poorly understood. Here, we use TimeLapse-seq to measure how inflammatory activation modifies the hypoxic response in primary macrophages. Nucleoside recoding sequencing allows the derivation of steady-state transcript levels, degradation rates, and transcriptional synthesis rates from the same dataset. We find that hypoxia produces distinct responses from resting and inflammatory macrophages. Hypoxia induces destabilization of mRNA transcripts, though inflammatory macrophages substantially increase mRNA degradation compared to resting macrophages. Increased RNA turnover results in the upregulation of ribosomal protein genes and downregulation of extracellular matrix components in inflammatory macrophages. Pathways regulated by mRNA decay in vitro are differentially regulated in tumor-associated macrophages implying that mixed stimuli could induce post-transcriptional regulation of macrophage function in solid tumors.
Collapse
Affiliation(s)
- Edward M C Courvan
- Department of Biochemistry, University of Colorado, Boulder, CO 80303, USA; Howard Hughes Medical Institute, University of Colorado, Boulder, CO 80303, USA.
| | - Roy R Parker
- Department of Biochemistry, University of Colorado, Boulder, CO 80303, USA; Howard Hughes Medical Institute, University of Colorado, Boulder, CO 80303, USA; BioFrontiers Institute, University of Colorado, Boulder, CO 80303, USA.
| |
Collapse
|
3
|
The uPA/uPAR System Orchestrates the Inflammatory Response, Vascular Homeostasis, and Immune System in Fibrosis Progression. Int J Mol Sci 2023; 24:ijms24021796. [PMID: 36675310 PMCID: PMC9866279 DOI: 10.3390/ijms24021796] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 01/18/2023] Open
Abstract
Fibrotic diseases, such as systemic sclerosis (SSc), idiopathic pulmonary fibrosis, renal fibrosis and liver cirrhosis are characterized by tissue overgrowth due to excessive extracellular matrix (ECM) deposition. Fibrosis progression is caused by ECM overproduction and the inhibition of ECM degradation due to several events, including inflammation, vascular endothelial dysfunction, and immune abnormalities. Recently, it has been reported that urokinase plasminogen activator (uPA) and its receptor (uPAR), known to be fibrinolytic factors, orchestrate the inflammatory response, vascular homeostasis, and immune homeostasis system. The uPA/uPAR system may show promise as a potential therapeutic target for fibrotic diseases. This review considers the role of the uPA/uPAR system in the progression of fibrotic diseases.
Collapse
|
4
|
Lu H, Xiao L, Wang W, Li X, Ma Y, Zhang Y, Wang X. Fibrinolysis Regulation: A Promising Approach to Promote Osteogenesis. TISSUE ENGINEERING. PART B, REVIEWS 2022; 28:1192-1208. [PMID: 35442086 DOI: 10.1089/ten.teb.2021.0222] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Soon after bone fracture, the initiation of the coagulation cascade results in the formation of a blood clot, which acts as a natural material to facilitate cell migration and osteogenic differentiation at the fracture site. The existence of hematoma is important in early stage of bone healing, but the persistence of hematoma is considered harmful for bone regeneration. Fibrinolysis is recently regarded as a period of critical transition in angiogenic-osteogenic coupling, it thereby is vital for the complete healing of the bone. Moreover, the enhanced fibrinolysis is proposed to boost bone regeneration through promoting the formation of blood vessels, and fibrinolysis system as well as the products of fibrinolysis also play crucial roles in the bone healing process. Therefore, the purpose of this review is to elucidate the fibrinolysis-derived effects on osteogenesis and summarize the potential approaches-improving bone healing by regulating fibrinolysis, with the purpose to further understand the integral roles of fibrinolysis in bone regeneration and to provide theoretical knowledge for potential fibrinolysis-related osteogenesis strategies. Impact statement Fibrinolysis emerging as a new and viable therapeutic intervention to be contained within osteogenesis strategies, however to now, there have been no review articles which collates the information between fibrinolysis and osteogenesis. This review, therefore, focusses on the effects that fibrinolysis exerts on bone healing, with a purpose to provide theoretical reference to develop new strategies to modulate fibrinolysis to accelerate fibrinolysis thus enhancing bone healing.
Collapse
Affiliation(s)
- Haiping Lu
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Lan Xiao
- School of Mechanical, Medical and Process Engineering, Center for Biomedical Technologies, Queensland University of Technology, Brisbane, Queensland, Australia.,The Australia-China Center for Tissue Engineering and Regenerative Medicine, Kelvin Grove, Brisbane, Queensland, Australia
| | - Weiqun Wang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Xuyan Li
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Yaping Ma
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Yi Zhang
- Department of Hygiene Toxicology, School of Public Health, Zunyi Medical University, Zunyi, Guizhou, China
| | - Xin Wang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China.,School of Mechanical, Medical and Process Engineering, Center for Biomedical Technologies, Queensland University of Technology, Brisbane, Queensland, Australia.,The Australia-China Center for Tissue Engineering and Regenerative Medicine, Kelvin Grove, Brisbane, Queensland, Australia
| |
Collapse
|
5
|
Bernard C, Zavoriti A, Pucelle Q, Chazaud B, Gondin J. Role of macrophages during skeletal muscle regeneration and hypertrophy-Implications for immunomodulatory strategies. Physiol Rep 2022; 10:e15480. [PMID: 36200266 PMCID: PMC9535344 DOI: 10.14814/phy2.15480] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 09/12/2022] [Indexed: 06/16/2023] Open
Abstract
Skeletal muscle is a plastic tissue that regenerates ad integrum after injury and adapts to raise mechanical loading/contractile activity by increasing its mass and/or myofiber size, a phenomenon commonly refers to as skeletal muscle hypertrophy. Both muscle regeneration and hypertrophy rely on the interactions between muscle stem cells and their neighborhood, which include inflammatory cells, and particularly macrophages. This review first summarizes the role of macrophages in muscle regeneration in various animal models of injury and in response to exercise-induced muscle damage in humans. Then, the potential contribution of macrophages to skeletal muscle hypertrophy is discussed on the basis of both animal and human experiments. We also present a brief comparative analysis of the role of macrophages during muscle regeneration versus hypertrophy. Finally, we summarize the current knowledge on the impact of different immunomodulatory strategies, such as heat therapy, cooling, massage, nonsteroidal anti-inflammatory drugs and resolvins, on skeletal muscle regeneration and their potential impact on muscle hypertrophy.
Collapse
Affiliation(s)
- Clara Bernard
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du MuscleUniversité Claude Bernard Lyon 1, CNRS UMR 5261, INSERM U1315, Université LyonLyonFrance
| | - Aliki Zavoriti
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du MuscleUniversité Claude Bernard Lyon 1, CNRS UMR 5261, INSERM U1315, Université LyonLyonFrance
| | - Quentin Pucelle
- Université de Versailles Saint‐Quentin‐En‐YvelinesVersaillesFrance
| | - Bénédicte Chazaud
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du MuscleUniversité Claude Bernard Lyon 1, CNRS UMR 5261, INSERM U1315, Université LyonLyonFrance
| | - Julien Gondin
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du MuscleUniversité Claude Bernard Lyon 1, CNRS UMR 5261, INSERM U1315, Université LyonLyonFrance
| |
Collapse
|
6
|
Xie Y, Zhang H, Huang T. Quantitative proteomics reveal three potential biomarkers for risk assessment of acute myocardial infarction. Bioengineered 2022; 13:4939-4950. [PMID: 35156527 PMCID: PMC8973584 DOI: 10.1080/21655979.2022.2037365] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Acute myocardial infarction (AMI) is the one of the main cause of death worldwide. Exosomes carry important information about intercellular communication and could be diagnostic marker for many diseases. Here, we aimed to find potential key proteins for the early diagnosis of AMI. A label free proteomics strategy was used to identify the differentially expressed proteins (DEPs) of AMI patients’ plasma exosome. By bioinformatics analysis and enzyme-linked immunosorbent assay to validate the candidate proteins. Compared to healthy control plasma exosome, we totally identified 72 differentially expressed proteins (DEPs) in AMI patients. Also, we found that complement and coagulation cascades was activated by KEGG analysis and GSEA. PLG, C8B and F2 were selected as candidate molecules for further study, and then validated another 40 plasma samples using enzyme-linked immunosorbent assay. Finally, we found that the expression levels of these three proteins (PLG, C8B and F2) were significantly higher than those of healthy controls (P < 0.05). ROC analysis revealed that PLG, C8B and F2 had potential value for AMI early diagnosis. In conclusion, our study identified three potential biomarkers for AMI diagnosis. But there remains a need to further study the mechanism of the biomarkers.
Collapse
Affiliation(s)
| | | | - Tieqiu Huang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
7
|
Zhang Y, Zheng Y, Shu F, Zhou R, Bao B, Xiao S, Li K, Lin Q, Zhu L, Xia Z. In situ-formed adhesive hyaluronic acid hydrogel with prolonged amnion-derived conditioned medium release for diabetic wound repair. Carbohydr Polym 2022; 276:118752. [PMID: 34823781 DOI: 10.1016/j.carbpol.2021.118752] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 10/06/2021] [Accepted: 10/08/2021] [Indexed: 12/20/2022]
Abstract
Hydrogels have long been used for encapsulating stem cell-derived conditioned mediums to achieve skin regeneration after wounding. However, inappropriate mechanical strength, low adhesion and low elasticity limit their clinical application. To address these challenges, we engineered a hyaluronic acid-based hydrogel grafted with methacrylic anhydride and N-(2-aminoethyl)-4-[4-(hydroxymethyl)-2-methoxy-5-nitrophenoxy]-butanamide (NB) groups to encapsulate a lyophilized amnion-derived conditioned medium (AM-CM). This hydrogel can photopolymerize in situ within 3 s by photo-initiated free-radical crosslinking between methacrylate moieties. Meanwhile, the formed o-nitrosobenzaldehyde groups by photo-irradiation could covalently bond with the amino groups of tissue surface, which allowed strong tissue adhesion. Furthermore, the hydrogel possessed excellent mechanical properties, high elasticity, favorable biocompatibility and prolonged AM-CM release. Our further vitro and in vivo studies showed that the hydrogel significantly accelerated diabetic wound healing by regulating macrophage polarization and promoting angiogenesis. The engineered hydrogel with AM-CM release has high potential to treat chronic wounds in clinics.
Collapse
Affiliation(s)
- Yiqing Zhang
- School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Yongjun Zheng
- School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, China; Department of Burn Surgery, Changhai Hospital, Naval Medical University, Shanghai 200433, China.
| | - Futing Shu
- Department of Burn Surgery, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Renjie Zhou
- School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Bingkun Bao
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Shichu Xiao
- Department of Burn Surgery, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Kai Li
- School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Qiuning Lin
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Linyong Zhu
- School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, China; School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Zhaofan Xia
- Department of Burn Surgery, Changhai Hospital, Naval Medical University, Shanghai 200433, China.
| |
Collapse
|
8
|
Contingent intramuscular boosting of P2XR7 axis improves motor function in transgenic ALS mice. Cell Mol Life Sci 2021; 79:7. [PMID: 34936028 PMCID: PMC8695421 DOI: 10.1007/s00018-021-04070-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 11/30/2021] [Accepted: 12/01/2021] [Indexed: 11/06/2022]
Abstract
Amyotrophic lateral sclerosis is a fatal neurodegenerative disorder that leads to progressive degeneration of motor neurons and severe muscle atrophy without effective treatment. Most research on the disease has been focused on studying motor neurons and supporting cells of the central nervous system. Strikingly, the recent observations have suggested that morpho-functional alterations in skeletal muscle precede motor neuron degeneration, bolstering the interest in studying muscle tissue as a potential target for the delivery of therapies. We previously showed that the systemic administration of the P2XR7 agonist, 2′(3′)-O‐(4-benzoylbenzoyl) adenosine 5-triphosphate (BzATP), enhanced the metabolism and promoted the myogenesis of new fibres in the skeletal muscles of SOD1G93A mice. Here we further corroborated this evidence showing that intramuscular administration of BzATP improved the motor performance of ALS mice by enhancing satellite cells and the muscle pro-regenerative activity of infiltrating macrophages. The preservation of the skeletal muscle retrogradely propagated along with the motor unit, suggesting that backward signalling from the muscle could impinge on motor neuron death. In addition to providing the basis for a suitable adjunct multisystem therapeutic approach in ALS, these data point out that the muscle should be at the centre of ALS research as a target tissue to address novel therapies in combination with those oriented to the CNS.
Collapse
|
9
|
Luque-Campos N, Bustamante-Barrientos FA, Pradenas C, García C, Araya MJ, Bohaud C, Contreras-López R, Elizondo-Vega R, Djouad F, Luz-Crawford P, Vega-Letter AM. The Macrophage Response Is Driven by Mesenchymal Stem Cell-Mediated Metabolic Reprogramming. Front Immunol 2021; 12:624746. [PMID: 34149687 PMCID: PMC8213396 DOI: 10.3389/fimmu.2021.624746] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 05/13/2021] [Indexed: 12/17/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent adult stromal cells widely studied for their regenerative and immunomodulatory properties. They are capable of modulating macrophage plasticity depending on various microenvironmental signals. Current studies have shown that metabolic changes can also affect macrophage fate and function. Indeed, changes in the environment prompt phenotype change. Therefore, in this review, we will discuss how MSCs orchestrate macrophage’s metabolic plasticity and the impact on their function. An improved understanding of the crosstalk between macrophages and MSCs will improve our knowledge of MSC’s therapeutic potential in the context of inflammatory diseases, cancer, and tissue repair processes in which macrophages are pivotal.
Collapse
Affiliation(s)
- Noymar Luque-Campos
- Laboratorio de Inmunología Celular y Molecular, Facultad de Medicina, Universidad de los Andes, Santiago, Chile.,Centro de Investigación e Innovación Biomédica, Universidad de los Andes, Santiago, Chile.,Programa de Doctorado en Biomedicina, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Felipe A Bustamante-Barrientos
- Laboratorio de Inmunología Celular y Molecular, Facultad de Medicina, Universidad de los Andes, Santiago, Chile.,Centro de Investigación e Innovación Biomédica, Universidad de los Andes, Santiago, Chile
| | - Carolina Pradenas
- Laboratorio de Inmunología Celular y Molecular, Facultad de Medicina, Universidad de los Andes, Santiago, Chile.,Centro de Investigación e Innovación Biomédica, Universidad de los Andes, Santiago, Chile.,Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Cynthia García
- Laboratorio de Inmunología Celular y Molecular, Facultad de Medicina, Universidad de los Andes, Santiago, Chile.,Centro de Investigación e Innovación Biomédica, Universidad de los Andes, Santiago, Chile.,Escuela de Biotecnología, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | - María Jesús Araya
- Laboratorio de Inmunología Celular y Molecular, Facultad de Medicina, Universidad de los Andes, Santiago, Chile.,Centro de Investigación e Innovación Biomédica, Universidad de los Andes, Santiago, Chile.,Escuela de Biotecnología, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | | | | | - Roberto Elizondo-Vega
- Laboratorio de Biología Celular, Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | | | - Patricia Luz-Crawford
- Laboratorio de Inmunología Celular y Molecular, Facultad de Medicina, Universidad de los Andes, Santiago, Chile.,Centro de Investigación e Innovación Biomédica, Universidad de los Andes, Santiago, Chile
| | - Ana María Vega-Letter
- Centro de Investigación e Innovación Biomédica, Universidad de los Andes, Santiago, Chile.,Cells for Cells, Regenero, Las Condes, Santiago, Chile.,Laboratory of Nano-Regenerative Medicine, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| |
Collapse
|
10
|
Tidball JG, Flores I, Welc SS, Wehling-Henricks M, Ochi E. Aging of the immune system and impaired muscle regeneration: A failure of immunomodulation of adult myogenesis. Exp Gerontol 2020; 145:111200. [PMID: 33359378 DOI: 10.1016/j.exger.2020.111200] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/17/2020] [Accepted: 12/08/2020] [Indexed: 12/16/2022]
Abstract
Skeletal muscle regeneration that follows acute injury is strongly influenced by interactions with immune cells that invade and proliferate in the damaged tissue. Discoveries over the past 20 years have identified many of the key mechanisms through which myeloid cells, especially macrophages, regulate muscle regeneration. In addition, lymphoid cells that include CD8+ T-cells and regulatory T-cells also significantly affect the course of muscle regeneration. During aging, the regenerative capacity of skeletal muscle declines, which can contribute to progressive loss of muscle mass and function. Those age-related reductions in muscle regeneration are accompanied by systemic, age-related changes in the immune system, that affect many of the myeloid and lymphoid cell populations that can influence muscle regeneration. In this review, we present recent discoveries that indicate that aging of the immune system contributes to the diminished regenerative capacity of aging muscle. Intrinsic, age-related changes in immune cells modify their expression of factors that affect the function of a population of muscle stem cells, called satellite cells, that are necessary for normal muscle regeneration. For example, age-related reductions in the expression of growth differentiation factor-3 (GDF3) or CXCL10 by macrophages negatively affect adult myogenesis, by disrupting regulatory interactions between macrophages and satellite cells. Those changes contribute to a reduction in the numbers and myogenic capacity of satellite cells in old muscle, which reduces their ability to restore damaged muscle. In addition, aging produces changes in the expression of molecules that regulate the inflammatory response to injured muscle, which also contributes to age-related defects in muscle regeneration. For example, age-related increases in the production of osteopontin by macrophages disrupts the normal inflammatory response to muscle injury, resulting in regenerative defects. These nascent findings represent the beginning of a newly-developing field of investigation into mechanisms through which aging of the immune system affects muscle regeneration.
Collapse
Affiliation(s)
- James G Tidball
- Molecular, Cellular & Integrative Physiology Program, University of California, Los Angeles, CA, United States of America; Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, United States of America; Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA, United States of America.
| | - Ivan Flores
- Molecular, Cellular & Integrative Physiology Program, University of California, Los Angeles, CA, United States of America
| | - Steven S Welc
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, United States of America; Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN 46202, United States of America
| | - Michelle Wehling-Henricks
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, United States of America
| | - Eisuke Ochi
- Hosei University, Faculty of Bioscience and Applied Chemistry, 3-7-2, Kajino, Koganei, Tokyo 184-8584, Japan
| |
Collapse
|
11
|
Cruz AF, Rohban R, Esni F. Macrophages in the pancreas: Villains by circumstances, not necessarily by actions. IMMUNITY INFLAMMATION AND DISEASE 2020; 8:807-824. [PMID: 32885589 PMCID: PMC7654401 DOI: 10.1002/iid3.345] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/13/2020] [Accepted: 08/19/2020] [Indexed: 12/17/2022]
Abstract
Introduction Mounting evidence suggest that macrophages play crucial roles in disease and tissue regeneration. However, despite much efforts during the past decade, our knowledge about the extent of macrophages' contribution to adult pancreatic regeneration after injury or during pancreatic disease progression is still limited. Nevertheless, it is generally accepted that some macrophage features that normally would contribute to healing and regeneration may be detrimental in pancreatic cancer. Altogether, the current literature contains conflicting reports on whether macrophages act as friends or foe in these conditions. Methods and Results In this review, we briefly review the origins of tissue resident and infiltrating macrophages and the importance of cellular crosstalking between macrophages and other resident cells in tissue regeneration. The primary objective of this review is to summarize our knowledge of the distinct roles of tissue resident and infiltrating macrophages, the impact of M1 and M2 macrophage phenotypes, and emerging evidence on macrophage crosstalking in pancreatic injury, regeneration, and disease. Conclusion Macrophages are involved with various stages of pancreatic cancer development, pancreatitis, and diabetes. Elucidating their role in these conditions will aid the development of targeted therapeutic treatments.
Collapse
Affiliation(s)
- Andrea F Cruz
- Division of Pediatric General and Thoracic Surgery, Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Rokhsareh Rohban
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Farzad Esni
- Division of Pediatric General and Thoracic Surgery, Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania.,Department of Developmental Biology, University of Pittsburgh, Pittsburgh, Pennsylvania.,University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania
| |
Collapse
|
12
|
Martin RA, Buckley KH, Mankowski DC, Riley BM, Sidwell AN, Douglas SL, Worth RG, Pizza FX. Myogenic Cell Expression of Intercellular Adhesion Molecule-1 Contributes to Muscle Regeneration after Injury. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:2039-2055. [PMID: 32650005 DOI: 10.1016/j.ajpath.2020.06.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 06/10/2020] [Accepted: 06/15/2020] [Indexed: 10/23/2022]
Abstract
This study investigated intercellular adhesion molecule-1 (ICAM-1), a membrane protein that mediates cell-to-cell adhesion and communication, as a mechanism through which the inflammatory response facilitates muscle regeneration after injury. Toxin-induced muscle injury to tibialis anterior muscles of wild-type mice caused ICAM-1 to be expressed by a population of satellite cells/myoblasts and myofibers. Myogenic cell expression of ICAM-1 contributed to the restoration of muscle structure after injury, as regenerating myofibers were more abundant and myofiber size was larger for wild-type compared with Icam1-/- mice during 28 days of recovery. Contrastingly, restoration of muscle function after injury was similar between the genotypes. ICAM-1 facilitated the restoration of muscle structure after injury through mechanisms involving the regulation of myofiber branching, protein synthesis, and the organization of nuclei within myofibers after myogenic cell fusion. These findings provide support for a paradigm in which ICAM-1 expressed by myogenic cells after muscle injury augments their adhesive and fusogenic properties, which, in turn, facilitates regenerative and hypertrophic processes that restore structure to injured muscle.
Collapse
Affiliation(s)
- Ryan A Martin
- School of Exercise and Rehabilitation Sciences, The University of Toledo, Toledo, Ohio
| | - Kole H Buckley
- School of Exercise and Rehabilitation Sciences, The University of Toledo, Toledo, Ohio
| | - Drew C Mankowski
- School of Exercise and Rehabilitation Sciences, The University of Toledo, Toledo, Ohio
| | - Benjamin M Riley
- School of Exercise and Rehabilitation Sciences, The University of Toledo, Toledo, Ohio
| | - Alena N Sidwell
- School of Exercise and Rehabilitation Sciences, The University of Toledo, Toledo, Ohio
| | - Stephanie L Douglas
- School of Exercise and Rehabilitation Sciences, The University of Toledo, Toledo, Ohio
| | - Randall G Worth
- Department of Medical Microbiology and Immunology, The University of Toledo, Toledo, Ohio
| | - Francis X Pizza
- School of Exercise and Rehabilitation Sciences, The University of Toledo, Toledo, Ohio.
| |
Collapse
|
13
|
Barrett CD, Yaffe MB. Influence of tranexamic acid on the complement system in trauma. ANZ J Surg 2020; 90:418-420. [DOI: 10.1111/ans.15538] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 10/02/2019] [Accepted: 10/03/2019] [Indexed: 11/30/2022]
Affiliation(s)
- Christopher D. Barrett
- Koch InstituteMassachusetts Institute of Technology Cambridge Massachusetts USA
- Department of Surgery, Beth Israel Deaconess Medical CenterHarvard Medical School Boston Massachusetts USA
| | - Michael B. Yaffe
- Koch InstituteMassachusetts Institute of Technology Cambridge Massachusetts USA
- Department of Surgery, Beth Israel Deaconess Medical CenterHarvard Medical School Boston Massachusetts USA
| |
Collapse
|
14
|
Wu CZ, Ou SH, Chang LC, Lin YF, Pei D, Chen JS. Deficiency of Urokinase Plasminogen Activator May Impair β Cells Regeneration and Insulin Secretion in Type 2 Diabetes Mellitus. Molecules 2019; 24:molecules24234208. [PMID: 31756973 PMCID: PMC6930534 DOI: 10.3390/molecules24234208] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 11/05/2019] [Accepted: 11/16/2019] [Indexed: 01/09/2023] Open
Abstract
Background: The relationship between urokinase-type plasminogen activator (uPA) and the development of type 2 diabetes mellitus (T2DM) was investigated in the study by using mice and cell models, as well as patients with T2DM. Methods: In mice models, wild-type and uPA knockout (uPA-/-) BALB/c mice were used for induction of T2DM. In cell models, insulin secretion rate and β cell proliferation were assessed in normal and high glucose after treating uPA siRNA, uPA, or anti-uPA antibody. In our clinical study, patients with T2DM received an oral glucose-tolerance test, and the relationship between uPA and insulin secretion was assessed. Results: Insulin particles and insulin secretion were mildly restored one month after induction in wild-type mice, but not in uPA-/- mice. In cell models, insulin secretion rate and cell proliferation declined in high glucose after uPA silencing either by siRNA or by anti-uPA antibody. After treatment with uPA, β cell proliferation increased in normal glucose. In clinical study, patients with T2DM and higher uPA levels had better ability of insulin secretion than those with lower uPA levels. Conclusion: uPA may play a substantial role in insulin secretion, β cell regeneration, and progressive development of T2DM. Supplementation of uPA might be a novel approach for prevention and treatment of T2DM in the future.
Collapse
Affiliation(s)
- Chung-Ze Wu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
| | - Shih-Hsiang Ou
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 81362, Taiwan;
| | - Li-Chien Chang
- School of Pharmacy, National Defense Medical Center, Taipei 11490, Taiwan;
| | - Yuh-Feng Lin
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
- Deputy Superintendent, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
| | - Dee Pei
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Fu Jen Catholic University Hospital, New Taipei City 24352, Taiwan;
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City 24205, Taiwan
| | - Jin-Shuen Chen
- Department of Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 81362, Taiwan
- Correspondence: ; Tel.: +886-7-3468057; Fax: +886-7-3468056
| |
Collapse
|
15
|
Oishi Y, Manabe I. Macrophages in inflammation, repair and regeneration. Int Immunol 2019; 30:511-528. [PMID: 30165385 DOI: 10.1093/intimm/dxy054] [Citation(s) in RCA: 401] [Impact Index Per Article: 66.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 08/22/2018] [Indexed: 12/12/2022] Open
Abstract
Tissue injury triggers a complex series of cellular responses, starting from inflammation activated by tissue and cell damage and proceeding to healing. By clearing cell debris, activating and resolving inflammation and promoting fibrosis, macrophages play key roles in most, if not all, phases of the response to injury. Recent studies of the mechanisms underlying the initial inflammation and later tissue regeneration and repair revealed that macrophages bridge these processes in part by supporting and activating stem/progenitor cells, clearing damaged tissue, remodeling extracellular matrix to prepare scaffolding for regeneration and promoting angiogenesis. However, macrophages also have a central role in the development of pathology induced by failed resolution (e.g. chronic inflammation) and excessive scarring. In this review, we summarize the activities of macrophages in inflammation and healing in response to acute injury in tissues with differing regenerative capacities. While macrophages lead similar processes in response to tissue injury in these tissues, their priorities and the consequences of their activities differ among tissues. Moreover, the magnitude, nature and duration of injury also greatly affect cellular responses and healing processes. In particular, continuous injury and/or failed resolution of inflammation leads to chronic ailments in which macrophage activities may become detrimental.
Collapse
Affiliation(s)
- Yumiko Oishi
- Department of Biochemistry & Molecular Biology, Nippon Medical School, Bunkyo-ku, Tokyo, Japan
| | - Ichiro Manabe
- Department of Disease Biology and Molecular Medicine, Chiba University Graduate School of Medicine, Chuo-ku, Chiba, Japan
| |
Collapse
|
16
|
Tranexamic acid mediates proinflammatory and anti-inflammatory signaling via complement C5a regulation in a plasminogen activator–dependent manner. J Trauma Acute Care Surg 2019; 86:101-107. [DOI: 10.1097/ta.0000000000002092] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
17
|
Functional muscle recovery with nanoparticle-directed M2 macrophage polarization in mice. Proc Natl Acad Sci U S A 2018; 115:10648-10653. [PMID: 30275293 DOI: 10.1073/pnas.1806908115] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Persistence of inflammation, and associated limits in tissue regeneration, are believed to be due in part to the imbalance of M1 over M2 macrophages. Here, we hypothesized that providing a sustained source of an antiinflammatory polarizing cytokine would shift the balance of macrophages at a site of tissue damage to improve functional regeneration. Specifically, IL-4-conjugated gold nanoparticles (PA4) were injected into injured murine skeletal muscle, resulting in improved histology and an ∼40% increase in muscle force compared with mice treated with vehicle only. Macrophages were the predominant infiltrating immune cell, and treatment with PA4 resulted in an approximately twofold increase in the percentage of macrophages expressing the M2a phenotype and an approximately twofold decrease in M1 macrophages, compared with mice treated with vehicle only. Intramuscular injection of soluble IL-4 did not shift macrophage polarization or result in functional muscle improvements. Depletion of monocytes/macrophages eliminated the therapeutic effects of PA4, suggesting that improvement in muscle function was the result of M2-shifted macrophage polarization. The ability of PA4 to direct macrophage polarization in vivo may be beneficial in the treatment of many injuries and inflammatory diseases.
Collapse
|
18
|
Fang MM, Barman PK, Thiruppathi M, Mirza RE, McKinney RD, Deng J, Christman JW, Du X, Fukai T, Ennis WJ, Koh TJ, Ushio-Fukai M, Urao N. Oxidant Signaling Mediated by Nox2 in Neutrophils Promotes Regenerative Myelopoiesis and Tissue Recovery following Ischemic Damage. THE JOURNAL OF IMMUNOLOGY 2018; 201:2414-2426. [PMID: 30201810 DOI: 10.4049/jimmunol.1800252] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 08/06/2018] [Indexed: 01/09/2023]
Abstract
Ischemic tissue damage activates hematopoietic stem and progenitor cells (HSPCs) in the bone marrow (BM)-generating myeloid cells, and persistent HSPC activity may drive chronic inflammation and impair tissue recovery. Although increased reactive oxygen species in the BM regulate HSPC functions, their roles in myelopoiesis of activated HSPCs and subsequent tissue recovery during ischemic damage are not well understood. In this paper, we report that deletion of Nox2 NADPH oxidase in mice results in persistent elevations in BM HSPC activity and levels of inflammatory monocytes/macrophages in BM and ischemic tissue in a model of hindlimb ischemia. Ischemic tissue damage induces oxidants in BM such as elevations of hydrogen peroxide and oxidized phospholipids, which activate redox-sensitive Lyn kinase in a Nox2-dependent manner. Moreover, during tissue recovery after ischemic injury, this Nox2-ROS-Lyn kinase axis is induced by Nox2 in neutrophils that home to the BM, which inhibits HSPC activity and inflammatory monocyte generation and promotes tissue regeneration after ischemic damage. Thus, oxidant signaling in the BM mediated by Nox2 in neutrophils regulates myelopoiesis of HSPCs to promote regeneration of damaged tissue.
Collapse
Affiliation(s)
- Milie M Fang
- Center for Wound Healing and Tissue Regeneration, Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612
| | - Pijus K Barman
- Center for Wound Healing and Tissue Regeneration, Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612
| | - Muthusamy Thiruppathi
- Center for Wound Healing and Tissue Regeneration, Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612
| | - Rita E Mirza
- Center for Wound Healing and Tissue Regeneration, Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612
| | - Ronald D McKinney
- Department of Pharmacology, Center for Cardiovascular Research, University of Illinois at Chicago College of Medicine, Chicago, IL 60612
| | - Jing Deng
- Department of Medicine, Ohio State University School of Medicine, Columbus, OH 43210.,Department of Medicine, University of Illinois at Chicago College of Medicine, Chicago, IL 60612
| | - John W Christman
- Department of Medicine, Ohio State University School of Medicine, Columbus, OH 43210.,Department of Medicine, University of Illinois at Chicago College of Medicine, Chicago, IL 60612
| | - Xiaoping Du
- Department of Pharmacology, Center for Cardiovascular Research, University of Illinois at Chicago College of Medicine, Chicago, IL 60612
| | - Tohru Fukai
- Vascular Biology Center, Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, GA 30912.,Charlie Norwood Veterans Affairs Medical Center, Augusta, GA 30904
| | - William J Ennis
- Department of Surgery, University of Illinois at Chicago College of Medicine, Chicago, IL 60612; and
| | - Timothy J Koh
- Center for Wound Healing and Tissue Regeneration, Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612
| | - Masuko Ushio-Fukai
- Vascular Biology Center, Department of Medicine (Cardiology), Medical College of Georgia at Augusta University, Augusta, GA 30912
| | - Norifumi Urao
- Center for Wound Healing and Tissue Regeneration, Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612; .,Department of Pharmacology, Center for Cardiovascular Research, University of Illinois at Chicago College of Medicine, Chicago, IL 60612
| |
Collapse
|
19
|
Amniotic Epithelial Cells Accelerate Diabetic Wound Healing by Modulating Inflammation and Promoting Neovascularization. Stem Cells Int 2018; 2018:1082076. [PMID: 30210547 PMCID: PMC6120261 DOI: 10.1155/2018/1082076] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Revised: 05/17/2018] [Accepted: 06/10/2018] [Indexed: 12/26/2022] Open
Abstract
Human amniotic epithelial cells (hAECs) are nontumorigenic, highly abundant, and low immunogenic and possess multipotent differentiation ability, which make them become ideal alternative stem cell source for regenerative medicine. Previous studies have demonstrated the therapeutic potential of hAECs in many tissue repairs. However, the therapeutic effect of hAECs on diabetic wound healing is still unknown. In this study, we injected hAECs intradermally around the full-thickness excisional skin wounds of db/db mice and found that hAECs significantly accelerated diabetic wound healing and granulation tissue formation. To explore the underlying mechanisms, we measured inflammation and neovascularization in diabetic wounds. hAECs could modulate macrophage phenotype toward M2 macrophage, promote switch from proinflammatory status to prohealing status of wounds, and increase capillary density in diabetic wounds. Furthermore, we found that the hAEC-conditioned medium promoted macrophage polarization toward M2 phenotype and facilitated migration, proliferation, and tube formation of endothelial cells through in vitro experiments. Taken together, we first reported that hAECs could promote diabetic wound healing, at least partially, through paracrine effects to regulate inflammation and promote neovascularization.
Collapse
|
20
|
Response of macrophages in rat skeletal muscle after eccentric exercise. Chin J Traumatol 2018; 21:88-95. [PMID: 29550244 PMCID: PMC5911737 DOI: 10.1016/j.cjtee.2017.12.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 12/24/2017] [Accepted: 01/05/2018] [Indexed: 02/04/2023] Open
Abstract
PURPOSE Macrophages are known to be important for healing numerous injured tissues depending on their functional phenotypes in response to different stimuli. The objective of this study was to reveal macrophage phenotypic changes involved in exercise-induced skeletal muscle injury and regeneration. METHODS Adult male Sprague-Dawley rats experienced one session of downhill running (16° decline, 16 m/min) for 90 min. After exercise the blood and soleus muscles were collected at 0 h, 6 h, 12 h, 1 d, 2 d, 3 d, 1 w and 2 w after exercise, separately. RESULTS It was showed that CD68+ M1 macrophages mainly infiltrated into muscle necrotic sites at 1-3 d, while CD163+ M2 macrophages were present in muscles from 0 h to 2 weeks after exercise. Using transmission electron microscopy, we observed activated satellite cells 1 d after exercise. Th1-associated transcripts of iNOS and Ccl2 were inhibited post exercise, while COX-2 mRNA was dramatically increased 12 h after running (p < 0.01). M2 phenotype marker Arg-1 increased 12 h and 3 d (p < 0.05, p < 0.01) after exercise, and Clec10a and Mrc2 were up-regulated in muscles 12 h following exercise (p < 0.05, p < 0.05). CONCLUSION The data demonstrate the dynamic patterns of macrophage phenotype in skeletal muscle upon eccentric exercise stimuli, and M1 and M2 phenotypes perform different functions during exercise-induced skeletal muscle injury and recovery.
Collapse
|
21
|
Abstract
Recovery from traumatic muscle injuries is typically prolonged and incomplete, leading to impaired muscle and joint function. We sought to determine whether mechanical stimulation via whole-body low-intensity vibration (LIV) could (1) improve muscle regeneration and (2) reduce muscle fibrosis following traumatic injury. C57BL/6J mice were subjected to a laceration of the gastrocnemius muscle and were treated with LIV (0.2 g at 90 Hz or 0.4 g at 45 Hz for 30 min/day) or non-LIV sham treatment (controls) for seven or 14 days. Muscle regeneration and fibrosis were assessed in hematoxylin and eosin or Masson's trichrome stained muscle cryosections, respectively. Compared to non-LIV control mice, the myofiber cross-sectional area was larger in mice treated with each LIV protocol after 14 days of treatment. Minimum fiber diameter was also larger in mice treated with LIV of 90 Hz/0.2 g after 14 days of treatment. There was also a trend toward a reduction in collagen deposition after 14 days of treatment with 45 Hz/0.4 g (p = 0.059). These findings suggest that LIV may improve muscle healing by enhancing myofiber growth and reducing fibrosis. The LIV-induced improvements in muscle healing suggest that LIV may represent a novel therapeutic approach for improving the healing of traumatic muscle injuries.
Collapse
|
22
|
Liu X, Liu Y, Zhao L, Zeng Z, Xiao W, Chen P. Macrophage depletion impairs skeletal muscle regeneration: The roles of regulatory factors for muscle regeneration. Cell Biol Int 2017; 41:228-238. [PMID: 27888539 DOI: 10.1002/cbin.10705] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 11/18/2016] [Indexed: 01/28/2023]
Abstract
Though macrophages are essential for skeletal muscle regeneration, which is a complex process, the roles and mechanisms of the macrophages in the process of muscle regeneration are still not fully understood. The objective of this study is to explore the roles of macrophages and the mechanisms involved in the regeneration of injured skeletal muscle. One hundred and twelve C57BL/6 mice were randomly divided into muscle contusion and macrophages depleted groups. Their gastrocnemius muscles were harvested at the time points of 12 h, 1, 3, 5, 7, 14 d post-injury. The changes in skeletal muscle morphology were assessed by hematoxylin and eosin (HE) stain. The gene expression was analyzed by real-time polymerase chain reaction. The data showed that CL-liposomes treatment did affect the expression of myogenic regulatory factors (MyoD, myogenin) after injury. In addition, CL-liposomes treatment decreased the expression of regulatory factors of muscle regeneration (HGF, uPA, COX-2, IGF-1, MGF, FGF6) and increased the expression of inflammatory cytokines (TGF-β1, TNF-α, IL-1β, RANTES) in the late stage of regeneration. Moreover, there were significant correlations between macrophages and some regulatory factors (such as HGF, uPA) for muscle regeneration. These results suggested that macrophages depletion impairs skeletal muscle regeneration and that the regulatory factors for muscle regeneration may play important roles in this process.
Collapse
Affiliation(s)
- Xiaoguang Liu
- School of Kinesiology, Shanghai University of Sport, Shanghai, 200438, China
| | - Yu Liu
- School of Kinesiology, Shanghai University of Sport, Shanghai, 200438, China.,Department of Exercise Science, Shenyang Sport University, Shenyang, 110001, China
| | - Linlin Zhao
- School of Kinesiology, Shanghai University of Sport, Shanghai, 200438, China
| | - Zhigang Zeng
- School of Kinesiology, Shanghai University of Sport, Shanghai, 200438, China
| | - Weihua Xiao
- School of Kinesiology, Shanghai University of Sport, Shanghai, 200438, China
| | - Peijie Chen
- School of Kinesiology, Shanghai University of Sport, Shanghai, 200438, China
| |
Collapse
|
23
|
Zheng Y, Ji S, Wu H, Tian S, Zhang Y, Wang L, Fang H, Luo P, Wang X, Hu X, Xiao S, Xia Z. Topical administration of cryopreserved living micronized amnion accelerates wound healing in diabetic mice by modulating local microenvironment. Biomaterials 2016; 113:56-67. [PMID: 27810642 DOI: 10.1016/j.biomaterials.2016.10.031] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 10/20/2016] [Accepted: 10/20/2016] [Indexed: 01/09/2023]
Abstract
Approximately 25% of diabetic patients suffer from diabetic lower-extremity ulcer throughout their lives and 7%-20% of patients will eventually need an amputation despite standard care treatment. The development of new therapies to treat diabetic wounds is urgent. In this study, we used cryopreserved living micronized amnion (300-600 μm) to treat wounds in diabetic mice. Post-thaw micronized amnion retained high cell viability, as well as intact cell morphology and membrane structure. When transplanted onto the wounds of db/db mice, the cryopreserved living micronized amnion greatly promoted wound healing in diabetic mice mainly by secreting growth, inflammation, and chemotaxis-related factors that regulated macrophage migration and phenotype switch, recruited CD34+ progenitor cells, and increased neovascularization. In addition, the micronized amnion matrix can exist in the dermis and serve as a long-term dermal scaffold. These results demonstrated the potential of the cryopreserved living micronized amnion as a ready-to-use living dermal substitute that addresses multiple defective physiological processes of impaired wounds to treat diabetic ulcers and other chronic wounds in clinics.
Collapse
Affiliation(s)
- Yongjun Zheng
- Burns Center of Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Shizhao Ji
- Burns Center of Changhai Hospital, The Second Military Medical University, Shanghai, China.
| | - Haibin Wu
- Burns Center of Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Song Tian
- Burns Center of Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Yunqing Zhang
- Burns Center of Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Li Wang
- Burns Center of Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - He Fang
- Burns Center of Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Pengfei Luo
- Burns Center of Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Xingtong Wang
- Burns Center of Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Xiaoyan Hu
- Burns Center of Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Shichu Xiao
- Burns Center of Changhai Hospital, The Second Military Medical University, Shanghai, China.
| | - Zhaofan Xia
- Burns Center of Changhai Hospital, The Second Military Medical University, Shanghai, China.
| |
Collapse
|
24
|
Capote J, Kramerova I, Martinez L, Vetrone S, Barton ER, Sweeney HL, Miceli MC, Spencer MJ. Osteopontin ablation ameliorates muscular dystrophy by shifting macrophages to a pro-regenerative phenotype. J Cell Biol 2016; 213:275-88. [PMID: 27091452 PMCID: PMC5084275 DOI: 10.1083/jcb.201510086] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 03/15/2016] [Indexed: 11/22/2022] Open
Abstract
In the degenerative disease Duchenne muscular dystrophy, inflammatory cells enter muscles in response to repetitive muscle damage. Immune factors are required for muscle regeneration, but chronic inflammation creates a profibrotic milieu that exacerbates disease progression. Osteopontin (OPN) is an immunomodulator highly expressed in dystrophic muscles. Ablation of OPN correlates with reduced fibrosis and improved muscle strength as well as reduced natural killer T (NKT) cell counts. Here, we demonstrate that the improved dystrophic phenotype observed with OPN ablation does not result from reductions in NKT cells. OPN ablation skews macrophage polarization toward a pro-regenerative phenotype by reducing M1 and M2a and increasing M2c subsets. These changes are associated with increased expression of pro-regenerative factors insulin-like growth factor 1, leukemia inhibitory factor, and urokinase-type plasminogen activator. Furthermore, altered macrophage polarization correlated with increases in muscle weight and muscle fiber diameter, resulting in long-term improvements in muscle strength and function in mdx mice. These findings suggest that OPN ablation promotes muscle repair via macrophage secretion of pro-myogenic growth factors.
Collapse
Affiliation(s)
- Joana Capote
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095 Molecular, Cellular, and Integrative Physiology Interdepartmental PhD Program, University of California, Los Angeles, Los Angeles, CA 90095
| | - Irina Kramerova
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095
| | - Leonel Martinez
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095
| | - Sylvia Vetrone
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095
| | - Elisabeth R Barton
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL 32611 Wellstone Muscular Dystrophy Center, University of Florida, Gainesville, FL 32610
| | - H Lee Sweeney
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32610 Wellstone Muscular Dystrophy Center, University of Florida, Gainesville, FL 32610
| | - M Carrie Miceli
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095 Center for Duchenne Muscular Dystrophy at UCLA, Los Angeles, CA 90095 Wellstone Muscular Dystrophy Center, University of Florida, Gainesville, FL 32610
| | - Melissa J Spencer
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095 Center for Duchenne Muscular Dystrophy at UCLA, Los Angeles, CA 90095 Wellstone Muscular Dystrophy Center, University of Florida, Gainesville, FL 32610
| |
Collapse
|
25
|
Piccoli M, Urbani L, Alvarez-Fallas ME, Franzin C, Dedja A, Bertin E, Zuccolotto G, Rosato A, Pavan P, Elvassore N, De Coppi P, Pozzobon M. Improvement of diaphragmatic performance through orthotopic application of decellularized extracellular matrix patch. Biomaterials 2015; 74:245-55. [PMID: 26461117 DOI: 10.1016/j.biomaterials.2015.10.005] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 09/29/2015] [Accepted: 10/01/2015] [Indexed: 12/15/2022]
Abstract
Muscle tissue engineering can provide support to large congenital skeletal muscle defects using scaffolds able to allow cell migration, proliferation and differentiation. Acellular extracellular matrix (ECM) scaffold can generate a positive inflammatory response through the activation of anti-inflammatory T-cell populations and M2 polarized macrophages that together lead to a local pro-regenerative environment. This immunoregulatory effect is maintained when acellular matrices are transplanted in a xenogeneic setting, but it remains unclear whether it can be therapeutic in a model of muscle diseases. We demonstrated here for the first time that orthotopic transplantation of a decellularized diaphragmatic muscle from wild animals promoted tissue functional recovery in an established atrophic mouse model. In particular, ECM supported a local immunoresponse activating a pro-regenerative environment and stimulating host muscle progenitor cell activation and migration. These results indicate that acellular scaffolds may represent a suitable regenerative medicine option for improving performance of diseased muscles.
Collapse
Affiliation(s)
- M Piccoli
- Stem Cells and Regenerative Medicine Lab, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy.
| | - L Urbani
- Stem Cells & Regenerative Medicine Section, Developmental Biology & Cancer Programme, UCL Institute of Child Health and Great Ormond Street Hospital, London, United Kingdom.
| | - M E Alvarez-Fallas
- Stem Cells and Regenerative Medicine Lab, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy
| | - C Franzin
- Stem Cells and Regenerative Medicine Lab, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy
| | - A Dedja
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padua, Padua, Italy
| | - E Bertin
- Stem Cells and Regenerative Medicine Lab, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy
| | - G Zuccolotto
- Department of Medicine, University of Padua, Padua, Italy
| | - A Rosato
- Veneto Institute of Oncology IOV - IRCCS, Padua, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - P Pavan
- Department of Industrial Engineering, University of Padua, Padua, Italy; Centre for Mechanics of Biological Materials, University of Padua, Padua, Italy
| | - N Elvassore
- Department of Industrial Engineering, University of Padua, and Venetian Institute of Molecular Medicine, Padua, Italy
| | - P De Coppi
- Stem Cells & Regenerative Medicine Section, Developmental Biology & Cancer Programme, UCL Institute of Child Health and Great Ormond Street Hospital, London, United Kingdom.
| | - M Pozzobon
- Stem Cells and Regenerative Medicine Lab, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy.
| |
Collapse
|
26
|
Torres-Palsa MJ, Koziol MV, Goh Q, Cicinelli PA, Peterson JM, Pizza FX. Expression of intercellular adhesion molecule-1 by myofibers in mdx mice. Muscle Nerve 2015; 52:795-802. [PMID: 25728314 DOI: 10.1002/mus.24626] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/24/2015] [Indexed: 01/13/2023]
Abstract
INTRODUCTION We investigated the extent to which intercellular adhesion molecule-1 (ICAM-1), a critical protein of the inflammatory response, is expressed in skeletal muscles of mdx mice (a murine model of Duchenne muscular dystrophy). METHODS Muscles were collected from control and mdx mice at 2-24 weeks of age and analyzed for ICAM-1 expression by means of Western blot and immunofluorescence. RESULTS Western blot revealed higher expression of ICAM-1 in mdx compared with control muscles through 24 weeks of age. In contrast to control muscles, ICAM-1 was expressed on the membrane of damaged, regenerating, and normal myofibers of mdx mice. CD11b+ myeloid cells also expressed ICAM-1 in mdx muscles, and CD11b+ cells were closely associated with the membrane of myofibers expressing ICAM-1. CONCLUSIONS These findings support a paradigm in which ICAM-1 and its localization to myofibers in muscles of mdx mice contributes to the dystrophic pathology.
Collapse
Affiliation(s)
- Maria J Torres-Palsa
- Department of Kinesiology, The University of Toledo, 2801 W. Bancroft Street, Toledo, Ohio, 43606, USA
| | - Matthew V Koziol
- Department of Kinesiology, The University of Toledo, 2801 W. Bancroft Street, Toledo, Ohio, 43606, USA
| | - Qingnian Goh
- Department of Kinesiology, The University of Toledo, 2801 W. Bancroft Street, Toledo, Ohio, 43606, USA
| | - Peter A Cicinelli
- Department of Kinesiology, The University of Toledo, 2801 W. Bancroft Street, Toledo, Ohio, 43606, USA
| | - Jennifer M Peterson
- Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University, Columbus, Ohio, USA
| | - Francis X Pizza
- Department of Kinesiology, The University of Toledo, 2801 W. Bancroft Street, Toledo, Ohio, 43606, USA
| |
Collapse
|
27
|
Hammers DW, Rybalko V, Merscham-Banda M, Hsieh PL, Suggs LJ, Farrar RP. Anti-inflammatory macrophages improve skeletal muscle recovery from ischemia-reperfusion. J Appl Physiol (1985) 2015; 118:1067-74. [PMID: 25678696 DOI: 10.1152/japplphysiol.00313.2014] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 02/10/2015] [Indexed: 11/22/2022] Open
Abstract
The presence of macrophages (MPs) is essential for skeletal muscle to properly regenerate following injury. The aim of this study was the evaluation of MP profiles and their importance in skeletal muscle recovering from tourniquet-induced ischemia-reperfusion (I/R). Using flow cytometry, we identified two distinct CD11b(+) MP populations that differ in expression of the surface markers Ly-6C and F4/80. These populations are prominent at 3 and 5 days of reperfusion and molecularly correspond to inflammatory and anti-inflammatory MP phenotypes. Sorted MP populations demonstrated high levels of IGF-I expression, and whole muscle post-I/R IGF-I expression strongly correlates with F4/80 expression. This suggests MPs largely influence postinjury IGF-I upregulation. We additionally demonstrate that direct intramuscular injection of FACS-isolated CD11b(+)Ly-6C(lo)F4/80(hi) MPs improves the functional and histological recovery of I/R-affected muscle. Taken together, these data further support the substantial influence of the innate immune system on muscle regeneration and suggest MP-focused therapeutic approaches may greatly facilitate skeletal muscle recovery from substantial injury.
Collapse
Affiliation(s)
- David W Hammers
- Department of Kinesiology, The University of Texas at Austin, Austin, Texas; and
| | - Viktoriya Rybalko
- Department of Kinesiology, The University of Texas at Austin, Austin, Texas; and
| | | | - Pei-Ling Hsieh
- Department of Kinesiology, The University of Texas at Austin, Austin, Texas; and
| | - Laura J Suggs
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas
| | - Roger P Farrar
- Department of Kinesiology, The University of Texas at Austin, Austin, Texas; and
| |
Collapse
|
28
|
Mechanisms of antitumor and immune-enhancing activities of MUC1/sec, a secreted form of mucin-1. Immunol Res 2014; 57:70-80. [PMID: 24222275 DOI: 10.1007/s12026-013-8451-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mucin 1 (MUC1) is a polymorphic type 1 transmembrane protein found on the apical surface of normal cells lining the lumen of ducts and glands. Mucins are thought to provide mucosal protection from environmental exposures and carcinogens. An altered form of the MUC1 glycoprotein, which is hypoglycosylated, is expressed in several types of human cancers. In our laboratory, we have found that transfection of a murine mammary tumor cell line with a human secreted isoform of MUC1 rendered these DA-3 cells (DA-3/sec) incapable of growing in intact BALB/c mice. In contrast, implantation of DA-3 cells transfected with the human transmembrane isoform of MUC1 (DA-3/TM), resulted in tumor formation and ultimately death of the animals, similar to the DA-3 parental line. Importantly, inoculation of the DA-3/sec cells in immunodeficient nude mice resulted in tumor formation, indicating that the MUC1/sec molecule's antitumor activity is immunologically controlled. In this review, we summarize the studies we have performed to elucidate possible mechanisms for the immune-mediated antitumor effect of MUC1/sec and/or a unique peptide present in this mucin. Understanding these mechanisms may provide new immunotherapeutic approaches that could be used to target different types of cancer.
Collapse
|
29
|
Acute resistance exercise increases the expression of chemotactic factors within skeletal muscle. Eur J Appl Physiol 2014; 114:2157-67. [PMID: 24968868 DOI: 10.1007/s00421-014-2936-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 06/10/2014] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Intense resistance exercise causes mechanical loading of skeletal muscle, followed by muscle adaptation. Chemotactic factors likely play an important role in these processes. PURPOSE We investigated the time course of changes in the expression and tissue localization of several key chemotactic factors in skeletal muscle during the early phase of recovery following resistance exercise. METHODS Muscle biopsy samples were obtained from vastus lateralis of eight untrained men (22 ± 0.5 years) before and 2, 4 and 24 h after three sets of leg press, squat and leg extension at 80 % 1-RM. RESULTS Monocyte chemotactic protein-1 (95×), interleukin-8 (2,300×), IL-6 (317×), urokinase-type plasminogen activator (15×), vascular endothelial growth factor (2×) and fractalkine (2.5×) mRNA was significantly elevated 2 h post-exercise. Interleukin-8 (38×) and interleukin-6 (58×) protein was also significantly elevated 2 h post-exercise, while monocyte chemotactic protein-1 protein was significantly elevated at 2 h (22×) and 4 h (21×) post-exercise. Monocyte chemotactic protein-1 and interleukin-8 were expressed by cells residing in the interstitial space between muscle fibers and, in some cases, were co-localized with CD68 + macrophages, PAX7 + satellite cells and blood vessels. However, the patterns of staining were inconclusive and not consistent. CONCLUSION In conclusion, resistance exercise stimulated a marked increase in the mRNA and protein expression of various chemotactic factors in skeletal muscle. Myofibers were not the dominant source of these factors. These findings suggest that chemotactic factors regulate remodeling/adaptation of skeletal muscle during the early phase of recovery following resistance exercise.
Collapse
|
30
|
Fleetwood AJ, Achuthan A, Schultz H, Nansen A, Almholt K, Usher P, Hamilton JA. Urokinase plasminogen activator is a central regulator of macrophage three-dimensional invasion, matrix degradation, and adhesion. THE JOURNAL OF IMMUNOLOGY 2014; 192:3540-7. [PMID: 24616477 DOI: 10.4049/jimmunol.1302864] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Urokinase plasminogen activator (uPA) and its receptor (uPAR) coordinate a plasmin-mediated proteolytic cascade that has been implicated in cell adhesion, cell motility, and matrix breakdown, for example, during inflammation. As part of their function during inflammatory responses, macrophages move through tissues and encounter both two-dimensional (2D) surfaces and more complex three-dimensional (3D) interstitial matrices. Based on approaches employing uPA gene-deficient macrophages, plasminogen supplementation, and neutralization with specific protease inhibitors, it is reported in this study that uPA activity is a central component of the invasion of macrophages through a 3D Matrigel barrier; it also has a nonredundant role in macrophage-mediated matrix degradation. For murine macrophages, matrix metalloproteinase-9 activity was found to be required for these uPA-mediated effects. Evidence for a unique role for uPA in the inverse relationship between macrophage adhesion and 2D migration was also noted: macrophage adhesion to vitronectin was enhanced by uPA and blocked by plasminogen activator inhibitor-1, the latter approach also able to enhance in turn the 2D migration on this matrix protein. It is therefore proposed that uPA can have a key role in the inflammatory response at several levels as a central regulator of macrophage 3D invasion, matrix remodeling, and adhesion.
Collapse
Affiliation(s)
- Andrew J Fleetwood
- Department of Medicine, University of Melbourne, The Royal Melbourne Hospital, Parkville, Victoria 3050, Australia
| | | | | | | | | | | | | |
Collapse
|
31
|
Novak ML, Weinheimer-Haus EM, Koh TJ. Macrophage activation and skeletal muscle healing following traumatic injury. J Pathol 2014; 232:344-55. [PMID: 24255005 DOI: 10.1002/path.4301] [Citation(s) in RCA: 160] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2013] [Revised: 10/29/2013] [Accepted: 11/06/2013] [Indexed: 12/19/2022]
Abstract
Following injury to different tissues, macrophages can contribute to both regenerative and fibrotic healing. These seemingly contradictory roles of macrophages may be related to the markedly different phenotypes that macrophages can assume upon exposure to different stimuli. We hypothesized that fibrotic healing after traumatic muscle injury would be dominated by a pro-fibrotic M2a macrophage phenotype, with M1 activation limited to the very early stages of repair. We found that macrophages accumulated in lacerated mouse muscle for at least 21 days, accompanied by limited myofibre regeneration and persistent collagen deposition. However, muscle macrophages did not exhibit either of the canonical M1 or M2a phenotypes, but instead up-regulated both M1- and M2a-associated genes early after injury, followed by down-regulation of most markers examined. Particularly, IL-10 mRNA and protein were markedly elevated in macrophages from 3-day injured muscle. Additionally, though flow cytometry identified distinct subpopulations of macrophages based on high or low expression of TNFα, these subpopulations did not clearly correspond to M1 or M2a phenotypes. Importantly, cell therapy with exogenous M1 macrophages but not non-activated macrophages reduced fibrosis and enhanced muscle fibre regeneration in lacerated muscles. These data indicate that manipulation of macrophage function has potential to improve healing following traumatic injury.
Collapse
Affiliation(s)
- Margaret L Novak
- Department of Kinesiology and Nutrition, University of Illinois at Chicago
| | | | | |
Collapse
|
32
|
Mirza RE, Fang MM, Weinheimer-Haus EM, Ennis WJ, Koh TJ. Sustained inflammasome activity in macrophages impairs wound healing in type 2 diabetic humans and mice. Diabetes 2014; 63:1103-14. [PMID: 24194505 PMCID: PMC3931398 DOI: 10.2337/db13-0927] [Citation(s) in RCA: 230] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The hypothesis of this study was that sustained activity of the Nod-like receptor protein (NLRP)-3 inflammasome in wounds of diabetic humans and mice contributes to the persistent inflammatory response and impaired healing characteristic of these wounds. Macrophages (Mp) isolated from wounds on diabetic humans and db/db mice exhibited sustained inflammasome activity associated with low level of expression of endogenous inflammasome inhibitors. Soluble factors in the biochemical milieu of these wounds are sufficient to activate the inflammasome, as wound-conditioned medium activates caspase-1 and induces release of interleukin (IL)-1β and IL-18 in cultured Mp via a reactive oxygen species-mediated pathway. Importantly, inhibiting inflammasome activity in wounds of db/db mice using topical application of pharmacological inhibitors improved healing of these wounds, induced a switch from proinflammatory to healing-associated Mp phenotypes, and increased levels of prohealing growth factors. Furthermore, data generated from bone marrow-transfer experiments from NLRP-3 or caspase-1 knockout to db/db mice indicated that blocking inflammasome activity in bone marrow cells is sufficient to improve healing. Our findings indicate that sustained inflammasome activity in wound Mp contributes to impaired early healing responses of diabetic wounds and that the inflammasome may represent a new therapeutic target for improving healing in diabetic individuals.
Collapse
Affiliation(s)
- Rita E. Mirza
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL
| | - Milie M. Fang
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL
| | - Eileen M. Weinheimer-Haus
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL
- Center for Tissue Repair and Regeneration, University of Illinois at Chicago, Chicago, IL
| | - William J. Ennis
- Center for Tissue Repair and Regeneration, University of Illinois at Chicago, Chicago, IL
- Department of Surgery, University of Illinois at Chicago, Chicago, IL
| | - Timothy J. Koh
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL
- Center for Tissue Repair and Regeneration, University of Illinois at Chicago, Chicago, IL
- Corresponding author: Timothy J. Koh,
| |
Collapse
|
33
|
Côté CH, Bouchard P, van Rooijen N, Marsolais D, Duchesne E. Monocyte depletion increases local proliferation of macrophage subsets after skeletal muscle injury. BMC Musculoskelet Disord 2013; 14:359. [PMID: 24354415 PMCID: PMC3878260 DOI: 10.1186/1471-2474-14-359] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 12/08/2013] [Indexed: 11/10/2022] Open
Abstract
Background Sequential accumulation of M1 and M2 macrophages is critical for skeletal muscle recovery after an acute injury. While M1 accumulation is believed to rely on monocyte infiltration, the mechanisms of M2 accumulation remain controversial, but could involve an infiltrating precursor. Yet, strong depletion of monocytes only partially impairs skeletal muscle healing, supporting the existence of alternative mechanisms to palliate the loss of infiltrating macrophage progenitors. The aims of this study are thus to investigate if proliferation occurs in macrophage subsets within injured skeletal muscles; and to determine if monocyte depletion leads to increased proliferation of macrophages after injury. Methods Injury was induced by bupivacaine injection in the tibialis anterior muscle of rats. Blood monocytes were depleted by daily intravenous injections of liposome-encapsulated clodronate, starting 24 h prior to injury. In separate experiments, irradiation of hind limb was also performed to prevent resident cell proliferation. Upon euthanasia, blood and muscles were collected for flow cytometric analyses of macrophage/monocyte subsets. Results Clodronate induced a 80%-90% depletion of monocyte but only led to 57% and 41% decrease of M1 and M2 macrophage accumulation, respectively, 2 d following injury. Conversely, the number of M1 macrophages in monocyte-depleted rats was 2.4-fold higher than in non-depleted rats 4 d after injury. This was associated with a 16-fold increase in the number of proliferative M1 macrophages, which was reduced by 46% in irradiated animals. Proliferation of M2 macrophages was increased tenfold by clodronate treatment 4 d post injury. The accumulation of M2 macrophages was partially impaired by irradiation, regardless of monocyte depletion. Conclusions M1 and M2 subsets proliferate after skeletal muscle injury and their proliferation is enhanced under condition of monocyte depletion. Our study supports the conclusion that both infiltrating and resident precursors could contribute to M1 or M2 macrophage accumulation in muscle injury.
Collapse
Affiliation(s)
| | | | | | | | - Elise Duchesne
- Centre de Recherche du CHUL (CHUQ), 2705 Boulevard Laurier, RC-9800 Québec, Québec, Canada.
| |
Collapse
|
34
|
Novak ML, Koh TJ. Phenotypic transitions of macrophages orchestrate tissue repair. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:1352-1363. [PMID: 24091222 DOI: 10.1016/j.ajpath.2013.06.034] [Citation(s) in RCA: 259] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 06/14/2013] [Accepted: 06/18/2013] [Indexed: 12/16/2022]
Abstract
Macrophages are essential for the efficient healing of numerous tissues, and they contribute to impaired healing and fibrosis. Tissue repair proceeds through overlapping phases of inflammation, proliferation, and remodeling, and macrophages are present throughout this progression. Macrophages exhibit transitions in phenotype and function as tissue repair progresses, although the precise factors regulating these transitions remain poorly defined. In efficiently healing injuries, macrophages present during a given stage of repair appear to orchestrate transition into the next phase and, in turn, can promote debridement of the injury site, cell proliferation and angiogenesis, collagen deposition, and matrix remodeling. However, dysregulated macrophage function can contribute to failure to heal or fibrosis in several pathological situations. This review will address current knowledge of the origins and functions of macrophages during the progression of tissue repair, with emphasis on skin and skeletal muscle. Dysregulation of macrophages in disease states and therapies targeting macrophage activation to promote tissue repair are also discussed.
Collapse
Affiliation(s)
- Margaret L Novak
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, Illinois
| | - Timothy J Koh
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, Illinois.
| |
Collapse
|
35
|
Chazaud B. Macrophages: supportive cells for tissue repair and regeneration. Immunobiology 2013; 219:172-8. [PMID: 24080029 DOI: 10.1016/j.imbio.2013.09.001] [Citation(s) in RCA: 223] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Revised: 08/21/2013] [Accepted: 09/01/2013] [Indexed: 12/12/2022]
Abstract
Macrophages, and more broadly inflammation, have been considered for a long time as bad markers of tissue homeostasis. However, if it is indisputable that macrophages are associated with many diseases in a deleterious way, new roles have emerged, showing beneficial properties of macrophages during tissue repair and regeneration. This discrepancy is likely due to the high plasticity of macrophages, which may exhibit a wide range of phenotypes and functions depending on their environment. Therefore, regardless of their role in immunity, macrophages play a myriad of roles in the maintenance and recovery of tissue homeostasis. They take a major part in the resolution of inflammation. They also exert various effects of parenchymal cells, including stem and progenitor cell, of which they regulate the fate. In the present review, few examples from various tissues are presented to illustrate that, beyond their specific properties in a given tissue, common features have been described that sustain a role of macrophages in the recovery and maintenance of tissue homeostasis.
Collapse
Affiliation(s)
- Bénédicte Chazaud
- Institut Cochin, INSERM U1016, Paris, France; CNRS 8104, Paris, France; Université Paris Descartes, Paris, France.
| |
Collapse
|
36
|
Krause MP, Al-Sajee D, D’Souza DM, Rebalka IA, Moradi J, Riddell MC, Hawke TJ. Impaired macrophage and satellite cell infiltration occurs in a muscle-specific fashion following injury in diabetic skeletal muscle. PLoS One 2013; 8:e70971. [PMID: 23951058 PMCID: PMC3741394 DOI: 10.1371/journal.pone.0070971] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Accepted: 06/26/2013] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Systemic elevations in PAI-1 suppress the fibrinolytic pathway leading to poor collagen remodelling and delayed regeneration of tibialis anterior (TA) muscles in type-1 diabetic Akita mice. However, how impaired collagen remodelling was specifically attenuating regeneration in Akita mice remained unknown. Furthermore, given intrinsic differences between muscle groups, it was unclear if the reparative responses between muscle groups were different. PRINCIPAL FINDINGS Here we reveal that diabetic Akita muscles display differential regenerative responses with the TA and gastrocnemius muscles exhibiting reduced regenerating myofiber area compared to wild-type mice, while soleus muscles displayed no difference between animal groups following injury. Collagen levels in TA and gastrocnemius, but not soleus, were significantly increased post-injury versus controls. At 5 days post-injury, when degenerating/necrotic regions were present in both animal groups, Akita TA and gastrocnemius muscles displayed reduced macrophage and satellite cell infiltration and poor myofiber formation. By 10 days post-injury, necrotic regions were absent in wild-type TA but persisted in Akita TA. In contrast, Akita soleus exhibited no impairment in any of these measures compared to wild-type soleus. In an effort to define how impaired collagen turnover was attenuating regeneration in Akita TA, a PAI-1 inhibitor (PAI-039) was orally administered to Akita mice following cardiotoxin injury. PAI-039 administration promoted macrophage and satellite cell infiltration into necrotic areas of the TA and gastrocnemius. Importantly, soleus muscles exhibit the highest inducible expression of MMP-9 following injury, providing a mechanism for normative collagen degradation and injury recovery in this muscle despite systemically elevated PAI-1. CONCLUSIONS Our findings suggest the mechanism underlying how impaired collagen remodelling in type-1 diabetes results in delayed regeneration is an impairment in macrophage infiltration and satellite cell recruitment to degenerating areas; a phenomena that occurs differentially between muscle groups.
Collapse
Affiliation(s)
- Matthew P. Krause
- Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Dhuha Al-Sajee
- Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Donna M. D’Souza
- Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Irena A. Rebalka
- Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Jasmin Moradi
- Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Michael C. Riddell
- Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | - Thomas J. Hawke
- Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
- Muscle Health Research Centre, York University, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
37
|
Alikhan MA, Ricardo SD. Mononuclear phagocyte system in kidney disease and repair. Nephrology (Carlton) 2013. [PMID: 23194390 DOI: 10.1111/nep.12014] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The mononuclear phagocyte system is comprised of circulating monocytes, tissue macrophages and dendritic cells (DCs) that play key roles in tissue homeostasis, immune surveillance, and immune and non-immune-mediated tissue injury and repair. This review summarizes the various subsets within this system that exhibit significant functional and phenotypic diversity that can adapt to their surrounding microenvironments during inflammation and in response to colony-stimulating factor (CSF)-1. The current understanding of the co-ordination of monocyte infiltration into the homeostatic and diseased kidney through adhesion molecules, chemokines and chemokine receptors, and cytokines are described. Furthermore, the significant confusion and controversy associated with monocyte differentiation into renal macrophages and DCs following infiltration into the kidney, the considerable functional and phenotypic overlap between both tissue populations and their respective roles in immune and non-immune-mediated renal is also discussed. Understanding the factors that control the activation and recruitment of cells from the mononuclear phagocyte system during renal injury may offer an avenue for the development of new cellular and growth factor-based therapies in combination with existing therapies as an alternative treatment option for patients with renal disease.
Collapse
Affiliation(s)
- Maliha A Alikhan
- Monash Immunology and Stem Cell Laboratories (MISCL), Monash University, Melbourne, Victoria, Australia
| | | |
Collapse
|
38
|
Mirza RE, Fang MM, Ennis WJ, Koh TJ. Blocking interleukin-1β induces a healing-associated wound macrophage phenotype and improves healing in type 2 diabetes. Diabetes 2013; 62:2579-87. [PMID: 23493576 PMCID: PMC3712034 DOI: 10.2337/db12-1450] [Citation(s) in RCA: 317] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Diabetes is associated with persistent inflammation and defective tissue repair responses. The hypothesis of this study was that interleukin (IL)-1β is part of a proinflammatory positive feedback loop that sustains a persistent proinflammatory wound macrophage phenotype that contributes to impaired healing in diabetes. Macrophages isolated from wounds in diabetic humans and mice exhibited a proinflammatory phenotype, including expression and secretion of IL-1β. The diabetic wound environment appears to be sufficient to induce these inflammatory phenomena because in vitro studies demonstrated that conditioned medium of both mouse and human wounds upregulates expression of proinflammatory genes and downregulates expression of prohealing factors in cultured macrophages. Furthermore, inhibiting the IL-1β pathway using a neutralizing antibody and macrophages from IL-1 receptor knockout mice blocked the conditioned medium-induced upregulation of proinflammatory genes and downregulation of prohealing factors. Importantly, inhibiting the IL-1β pathway in wounds of diabetic mice using a neutralizing antibody induced a switch from proinflammatory to healing-associated macrophage phenotypes, increased levels of wound growth factors, and improved healing of these wounds. Our findings indicate that targeting the IL-1β pathway represents a new therapeutic approach for improving the healing of diabetic wounds.
Collapse
Affiliation(s)
- Rita E. Mirza
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, Illinois
| | - Milie M. Fang
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, Illinois
| | - William J. Ennis
- Department of Surgery, University of Illinois at Chicago, Chicago, Illinois
- Center for Tissue Repair and Regeneration, University of Illinois at Chicago, Chicago, Illinois
| | - Timothy J. Koh
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, Illinois
- Center for Tissue Repair and Regeneration, University of Illinois at Chicago, Chicago, Illinois
- Corresponding author: Timothy J. Koh,
| |
Collapse
|
39
|
Plasma tissue-type plasminogen activator increases fibrinolytic activity of exogenous urokinase-type plasminogen activator. Blood Coagul Fibrinolysis 2013; 23:729-33. [PMID: 22918041 DOI: 10.1097/mbc.0b013e32835897d5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
The relationship between tissue-type plasminogen activator (tPA) and urokinase-type plasminogen activator (uPA) function is not fully understood. The aim of this study was to compare in vitro the fibrinolytic activity of tPA and uPA and evaluate their possible interaction. Blood coagulation and fibrinolysis were conducted by rotation thromboelastometry, whereas blood clotting was induced by CaCl2 and tissue factor and fibrinolysis additively by tPA and uPA. Effective concentration 50% of tPA and uPA fibrinolytic activity in blood was found to be 90 and 33 IU/ml relating to the units of activity established by manufacturers in the absence of blood. uPA-induced fibrinolysis in blood was faster compared with tPA used at the same units of activity. In contrast, in a blood-free system containing fibrinogen, plasminogen, and thrombin, fibrinolysis induced by uPA was weaker than by tPA. Treating of blood with tranexamic acid (60 mmol/l) was followed by decreased fibrinolytic potential of both exogenous tPA and uPA, despite uPA by itself is known to be not sensitive to aminocaproic acids. Thus, uPA exerted stronger activity in blood but weaker activity in blood-free system, compared with tPA. Taking into account the intermolecular binding of uPA to tPA, it could be suggested that interaction of exogenous uPA with plasma-containing tPA provided amplification of fibrinolysis due to formation of uPA/tPA complex possessing high affinity to fibrin.
Collapse
|
40
|
Pillon NJ, Bilan PJ, Fink LN, Klip A. Cross-talk between skeletal muscle and immune cells: muscle-derived mediators and metabolic implications. Am J Physiol Endocrinol Metab 2013; 304:E453-65. [PMID: 23277185 DOI: 10.1152/ajpendo.00553.2012] [Citation(s) in RCA: 211] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Skeletal muscles contain resident immune cell populations and their abundance and type is altered in inflammatory myopathies, endotoxemia or different types of muscle injury/insult. Within tissues, monocytes differentiate into macrophages and polarize to acquire pro- or anti-inflammatory phenotypes. Skeletal muscle macrophages play a fundamental role in repair and pathogen clearance. These events require a precisely regulated cross-talk between myofibers and immune cells, involving paracrine/autocrine and contact interactions. Skeletal muscle also undergoes continuous repair as a result of contractile activity that involves participation of myokines and anti-inflammatory input. Finally, skeletal muscle is the major site of dietary glucose disposal; therefore, muscle insulin resistance is essential to the development of whole body insulin resistance. Notably, muscle inflammation is emerging as a potential contributor to insulin resistance. Recent reports show that inflammatory macrophage numbers within muscle are elevated during obesity and that muscle cells in vitro can mount autonomous inflammatory responses under metabolic challenge. Here, we review the nature of skeletal muscle inflammation associated with muscle exercise, damage, and regeneration, endotoxin presence, and myopathies, as well as the new evidence of local inflammation arising with obesity that potentially contributes to insulin resistance.
Collapse
Affiliation(s)
- Nicolas J Pillon
- Program in Cell Biology, Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | | | | | | |
Collapse
|
41
|
Saclier M, Cuvellier S, Magnan M, Mounier R, Chazaud B. Monocyte/macrophage interactions with myogenic precursor cells during skeletal muscle regeneration. FEBS J 2013; 280:4118-30. [PMID: 23384231 DOI: 10.1111/febs.12166] [Citation(s) in RCA: 185] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Revised: 01/27/2013] [Accepted: 01/31/2013] [Indexed: 12/14/2022]
Abstract
Adult skeletal muscle has the remarkable property of regenerating after damage, owing to satellite cells and myogenic precursor cells becoming committed to adult myogenesis to rebuild the muscle. This process is accompanied by the continuing presence of macrophages, from the phagocytosis of damaged myofibres to the full re-formation of new myofibres. In recent years, there has been huge progress in our understanding of the roles of macrophages during skeletal muscle regeneration, notably concerning their effects on myogenic precursor cells. Here, we review the most recent knowledge acquired on monocyte entry into damaged muscle, the various macrophage subpopulations, and their respective roles during the sequential phases of muscle repair. We also discuss the role of macrophages after exercise-induced muscle damage, notably in humans.
Collapse
|
42
|
Macrophage Colony-Stimulating Factor–Induced Macrophage Differentiation Promotes Regrowth in Atrophied Skeletal Muscles and C2C12 Myotubes. THE AMERICAN JOURNAL OF PATHOLOGY 2013. [DOI: 10.1016/j.ajpath.2012.10.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
43
|
Almholt K, Juncker-Jensen A, Lærum OD, Johnsen M, Rømer J, Lund LR. Spontaneous metastasis in congenic mice with transgenic breast cancer is unaffected by plasminogen gene ablation. Clin Exp Metastasis 2012; 30:277-88. [PMID: 22996753 DOI: 10.1007/s10585-012-9534-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Accepted: 09/12/2012] [Indexed: 11/25/2022]
Abstract
Plasminogen (Plg) plays a central role in tissue remodeling during ontogeny, development, and in pathological tissue remodeling following physical injury, inflammation and cancer. Plg/plasmin is, however, not critical for these processes, as they all occur to a varying extent in its absence, suggesting that there is a functional redundancy with other proteases. To explore this functional overlap in the transgenic MMTV-PyMT breast cancer metastasis model, we have combined Plg deficiency and a pharmacological metalloprotease inhibitor, which is known to reduce metastasis in this model, and has been shown to synergistically inhibit other tissue remodeling events in Plg-deficient mice. While metalloprotease inhibition dramatically reduced metastasis, we found no effect of Plg deficiency on metastasis, either independently or in combination with metalloprotease inhibition. We further show that Plg gene deficiency is of no significant consequence in this metastasis model, when analyzed in two different congenic strains: the FVB strain, and a F1 hybrid of the FVB and C57BL/6J strains. We suggest that the extensive backcrossing performed prior to our studies has eliminated the confounding effect of a known polymorphic metastasis modifier gene region located adjacent to the Plg gene.
Collapse
Affiliation(s)
- Kasper Almholt
- Finsen Laboratory, Rigshospitalet, Copenhagen Biocenter, Copenhagen, Denmark.
| | | | | | | | | | | |
Collapse
|
44
|
Kawao N, Nagai N, Tamura Y, Horiuchi Y, Okumoto K, Okada K, Suzuki Y, Umemura K, Yano M, Ueshima S, Kaji H, Matsuo O. Urokinase-type plasminogen activator and plasminogen mediate activation of macrophage phagocytosis during liver repair in vivo. Thromb Haemost 2012; 107:749-59. [PMID: 22318286 DOI: 10.1160/th11-08-0567] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2011] [Accepted: 01/09/2012] [Indexed: 12/17/2022]
Abstract
Urokinase-type plasminogen activator (u-PA) and plasminogen play a primary role in liver repair through the accumulation of macrophages and alteration of their phenotype. However, it is still unclear whether u-PA and plasminogen mediate the activation of macrophage phagocytosis during liver repair. Herein, we investigated the morphological changes in macrophages that accumulated at the edge of damaged tissue induced by a photochemical reaction or hepatic ischaemia-reperfusion in mice with u-PA ( u-PA-/- ) or plasminogen ( Plg-/- ) gene deficiency by using transmission electron and fluorescence microscopy. In wild-type mice, the macrophages aligned at the edge of the damaged tissue and extended a large number of long pseudopodia. These macrophages clearly engulfed cellular debris and showed well-developed organelles, including lysosome-like vacuoles, nuclei, and Golgi complexes. In wild-type mice, the distribution of the Golgi complex in these macrophages was biased towards the direction of the damaged tissue, indicating the extension of their pseudopodia in this direction. Conversely, in u-PA-/- and Plg-/- mice, the macrophages located at the edge of the damaged tissue had few pseudopodia and less developed organelles. The Golgi complex was randomly distributed in these macrophages in u-PA-/- mice. Furthermore, interferon γ and IL-4 were expressed at a low level at the border region of the damaged tissue in u-PA-/- mice. Our data provide novel evidence that u-PA and plasminogen are essential for the phagocytosis of cellular debris by macrophages during liver repair. Furthermore, u-PA plays a critical role in the induction of macrophage polarity by affecting the microenvironment at the edge of damaged tissue.
Collapse
Affiliation(s)
- N Kawao
- Department of Physiology and Regenerative Medicine, Kinki University Faculty of Medicine, 377-2 Ohnohigashi, Osakasayama 589-8511, Osaka, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Alikhan MA, Jones CV, Williams TM, Beckhouse AG, Fletcher AL, Kett MM, Sakkal S, Samuel CS, Ramsay RG, Deane JA, Wells CA, Little MH, Hume DA, Ricardo SD. Colony-stimulating factor-1 promotes kidney growth and repair via alteration of macrophage responses. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 179:1243-56. [PMID: 21762674 PMCID: PMC3157188 DOI: 10.1016/j.ajpath.2011.05.037] [Citation(s) in RCA: 124] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/27/2010] [Revised: 05/05/2011] [Accepted: 05/23/2011] [Indexed: 11/18/2022]
Abstract
Colony-stimulating factor (CSF)-1 controls the survival, proliferation, and differentiation of macrophages, which are recognized as scavengers and agents of the innate and the acquired immune systems. Because of their plasticity, macrophages are endowed with many other essential roles during development and tissue homeostasis. We present evidence that CSF-1 plays an important trophic role in postnatal organ growth and kidney repair. Notably, the injection of CSF-1 postnatally enhanced kidney weight and volume and was associated with increased numbers of tissue macrophages. Moreover, CSF-1 promotes postnatal renal repair in mice after ischemia-reperfusion injury by recruiting and influencing macrophages toward a reparative state. CSF-1 treatment rapidly accelerated renal repair with tubular epithelial cell replacement, attenuation of interstitial fibrosis, and functional recovery. Analysis of macrophages from CSF-1-treated kidneys showed increased expression of insulin-like growth factor-1 and anti-inflammatory genes that are known CSF-1 targets. Taken together, these data suggest that CSF-1 is important in kidney growth and the promotion of endogenous repair and resolution of inflammatory injury.
Collapse
Affiliation(s)
- Maliha A. Alikhan
- Monash Immunology and Stem Cell Laboratories (MISCL), Monash University, Melbourne, Australia
| | - Christina V. Jones
- Monash Immunology and Stem Cell Laboratories (MISCL), Monash University, Melbourne, Australia
| | - Timothy M. Williams
- Monash Immunology and Stem Cell Laboratories (MISCL), Monash University, Melbourne, Australia
| | - Anthony G. Beckhouse
- Australian Institute for Bioengineering and Nanotechnology (AIBN), University of Queensland, Brisbane, Australia
| | - Anne L. Fletcher
- Monash Immunology and Stem Cell Laboratories (MISCL), Monash University, Melbourne, Australia
| | - Michelle M. Kett
- Department of Physiology, Monash University, Melbourne, Australia
| | - Samy Sakkal
- Monash Immunology and Stem Cell Laboratories (MISCL), Monash University, Melbourne, Australia
| | - Chrishan S. Samuel
- Howard Florey Institute, University of Melbourne, Melbourne, Australia
- Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, Australia
| | | | - James A. Deane
- Monash Immunology and Stem Cell Laboratories (MISCL), Monash University, Melbourne, Australia
| | - Christine A. Wells
- Australian Institute for Bioengineering and Nanotechnology (AIBN), University of Queensland, Brisbane, Australia
| | - Melissa H. Little
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
| | - David A. Hume
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Scotland, United Kingdom
| | - Sharon D. Ricardo
- Monash Immunology and Stem Cell Laboratories (MISCL), Monash University, Melbourne, Australia
| |
Collapse
|
46
|
Dragu A, Schnürer S, Surmann-Schmitt C, von der Mark K, Stürzl M, Unglaub F, Wolf MB, Leffler M, Beier JP, Kneser U, Horch RE. Gene expression analysis of ischaemia and reperfusion in human microsurgical free muscle tissue transfer. J Cell Mol Med 2011; 15:983-93. [PMID: 20345846 PMCID: PMC3922682 DOI: 10.1111/j.1582-4934.2010.01061.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The aim of this study was to analyse various gene expression profiles of muscle tissue during normoxia, ischaemia and after reperfusion in human muscle free flaps, to gain an understanding of the occurring regulatory, inflammatory and apoptotic processes on a cellular and molecular basis. Eleven Caucasian patients with soft tissue defects needing coverage with microsurgical free muscle flaps were included in this study. In all patients, the muscle samples were taken from free myocutaneous flaps. The first sample was taken before induction of ischaemia in normoxia (I), another one after ischaemia (II), and the last one was taken after reperfusion (III). The samples were analysed using DNA-microarray, real-time-quantitative-PCR and immunohistochemistry. DNA-microarray analysis detected multiple, differentially regulated genes when comparing the different groups (I–III) with statistical significance. Comparing ischaemia (II) versus normoxia (I) educed 13 genes and comparing reperfusion (III) versus ischaemia (II) educed 19 genes. The comparison of reperfusion (III) versus normoxia (I) yielded 100 differentially regulated genes. Real-time-quantitative-PCR confirmed the results of the DNA-microarrays for a subset of four genes (CASP8, IL8, PLAUR and S100A8). This study shows that ischaemia and reperfusion induces alterations on the gene expression level in human muscle free flaps. Data may suggest that the four genes CASP8, IL8, PLAUR and S100A8 are of great importance in this context. We could not confirm the DNA-microarry and real-time-quantitative-PCR results on the protein level. Finally, these findings correspond with the surgeon’s clinical experience that the accepted times of ischaemia, generally up to 90 min., are not sufficient to induce pathophysiological processes, which can ultimately lead to flap loss. When inflammatory and apoptotic proteins are expressed at high levels, flap damage might occur and flap loss is likely. The sole expression on mRNA level might explain why flap loss is unlikely.
Collapse
Affiliation(s)
- Adrian Dragu
- Department of Plastic and Hand Surgery, Friedrich-Alexander-University of Erlangen-Nürnberg, University Hospital, Erlangen, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Nalla AK, Gogineni VR, Gupta R, Dinh DH, Rao JS. Suppression of uPA and uPAR blocks radiation-induced MCP-1 mediated recruitment of endothelial cells in meningioma. Cell Signal 2011; 23:1299-310. [PMID: 21426933 PMCID: PMC3095686 DOI: 10.1016/j.cellsig.2011.03.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Revised: 03/10/2011] [Accepted: 03/10/2011] [Indexed: 02/02/2023]
Abstract
Chemokines play a vital role in recruiting various cell types in the process of tissue repair. Radiation, a major therapeutic modality in cancer treatment, has been described to induce inflammatory response that might lead to the expression of several chemokines. In the present study, we investigated the mechanism of monocyte chemoattractant protein-1 (MCP-1) induction by radiation in meningioma cell lines and the paracrine effect on human microvascular endothelial cells (HMEC). After radiation, meningioma cell lines (IOMM Lee and SF-3061) showed an increased expression of MCP-1. In addition, irradiated meningioma cancer cell conditioned medium (CM) showed an increased ability to attract HMEC and to stimulate MCP-1-induced protein (MCPIP), VEGF and angiogenin expression in HMEC. This chemotactic activity and angiogenic stimulator effect on HMEC were almost abrogated by depleting MCP-1 from the irradiated cancer cell CM. Further, inhibition of either ERK activation/expression or NF-κB nuclear translocation hindered radiation-induced MCP-1 expression in both meningioma cell lines. Further, supplementing cancer cells with exogenous ATF-uPA (with and without radiation) activated ERK phosphorylation, nuclear translocation of the NF-κB p65 sub-unit (Rel-A), and MCP-1 expression. Downregulation of uPA and uPAR, simultaneously by transfecting the cancer cells with bi-cistronic siRNA-expressing plasmid (pU) inhibited radiation-induced ERK activation, nuclear translocation of Rel-A, NF-κB DNA binding activity, and MCP-1 expression. In addition, pU-transfected cancer cells (with or without radiation) reduced radiation-induced MCP-1 and blocked the recruitment of other cell types during the inflammatory process induced by radiation both in in vitro and in vivo conditions.
Collapse
Affiliation(s)
- Arun Kumar Nalla
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, One Illini Drive, Peoria, Illinois
| | - Venkateswara Rao Gogineni
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, One Illini Drive, Peoria, Illinois
| | - Reshu Gupta
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, One Illini Drive, Peoria, Illinois
| | - Dzung H. Dinh
- Department of Neurosurgery, University of Illinois College of Medicine at Peoria, One Illini Drive, Peoria, Illinois
| | - Jasti S. Rao
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, One Illini Drive, Peoria, Illinois
- Department of Neurosurgery, University of Illinois College of Medicine at Peoria, One Illini Drive, Peoria, Illinois
| |
Collapse
|
48
|
Novak ML, Bryer SC, Cheng M, Nguyen MH, Conley KL, Cunningham AK, Xue B, Sisson TH, You JS, Hornberger TA, Koh TJ. Macrophage-specific expression of urokinase-type plasminogen activator promotes skeletal muscle regeneration. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2011; 187:1448-57. [PMID: 21709151 PMCID: PMC3140545 DOI: 10.4049/jimmunol.1004091] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Macrophages (Mp) and the plasminogen system play important roles in tissue repair following injury. We hypothesized that Mp-specific expression of urokinase-type plasminogen activator (uPA) is sufficient for Mp to migrate into damaged muscle and for efficient muscle regeneration. We generated transgenic mice expressing uPA only in Mp, and we assessed the ability of these mice to repair muscle injury. Mp-only uPA expression was sufficient to induce wild-type levels of Mp accumulation, angiogenesis, and new muscle fiber formation. In mice with wild-type uPA expression, Mp-specific overexpression further increased Mp accumulation and enhanced muscle fiber regeneration. Furthermore, Mp expression of uPA regulated the level of active hepatocyte growth factor, which is required for muscle fiber regeneration, in damaged muscle. In vitro studies demonstrated that uPA promotes Mp migration through proteolytic and nonproteolytic mechanisms, including proteolytic activation of hepatocyte growth factor. In summary, Mp-derived uPA promotes muscle regeneration by inducing Mp migration, angiogenesis, and myogenesis.
Collapse
Affiliation(s)
- Margaret L. Novak
- Department of Kinesiology and Nutrition, University of Illinois at Chicago
| | - Scott C. Bryer
- Department of Kinesiology and Nutrition, University of Illinois at Chicago
| | - Ming Cheng
- Department of Kinesiology and Nutrition, University of Illinois at Chicago
| | - Mai-Huong Nguyen
- Department of Kinesiology and Nutrition, University of Illinois at Chicago
| | - Kevin L. Conley
- Department of Kinesiology and Nutrition, University of Illinois at Chicago
| | | | - Bing Xue
- Department of Medicine, University of Michigan, Ann Arbor
| | | | - Jae-Sung You
- Department of Comparative Biosciences, University of Wisconsin, Madison
| | | | - Timothy J. Koh
- Department of Kinesiology and Nutrition, University of Illinois at Chicago
| |
Collapse
|
49
|
Activation of urokinase plasminogen activator and its receptor axis is essential for macrophage infiltration in a prostate cancer mouse model. Neoplasia 2011; 13:23-30. [PMID: 21245937 DOI: 10.1593/neo.10728] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2010] [Revised: 09/29/2010] [Accepted: 10/05/2010] [Indexed: 12/21/2022] Open
Abstract
Macrophages within the tumor microenvironment promote angiogenesis, extracellular matrix breakdown, and tumor cell migration, invasion, and metastasis. Activation of the urokinase plasminogen activator (uPA) and its receptor (uPAR) axis promotes prostate cancer tumorigenicity, invasion, metastasis, and survival within the tumor microenvironment. The link between macrophage infiltration and the uPA/uPAR axis in prostate cancer development has not been established, although it has been reported that uPA plays a critical role inmonocyte and macrophage chemotaxis. In this study, murine prostate cancer RM-1 cells were subcutaneously inoculated into wild-type (WT), uPA(-/-), and uPAR(-/-) mice. Tumor volume was significantly diminished in both uPA(-/-) and uPAR(-/-) mice compared with WT controls. Greater inhibition of tumor volume was also observed in uPA(-/-) mice compared with uPAR(-/-) mice, suggesting the important contribution of stromal-derived uPA to sustain the tumor growth. Immunohistochemical staining revealed that tumors in uPA(-/-) and uPAR(-/-) mice displayed significantly lower proliferative indices, higher apoptotic indices, and less neovascularity compared with the tumors in WT mice. Tumors in uPA(-/-) and uPAR(-/-) mice displayed significantly less macrophage infiltration as demonstrated by F4/80 staining and MAC3(+) cell numbers by flow cytometry compared with the tumors from WT mice. These findings suggest that the uPA/uPAR axis acts in both autocrine and paracrine manners in the tumor microenvironment, and activation of uPA/uPAR axis is essential for macrophage infiltration into prostate tumors.
Collapse
|
50
|
Kawao N, Nagai N, Tamura Y, Okada K, Yano M, Suzuki Y, Umemura K, Ueshima S, Matsuo O. Urokinase-type plasminogen activator contributes to heterogeneity of macrophages at the border of damaged site during liver repair in mice. Thromb Haemost 2011; 105:892-900. [PMID: 21301782 DOI: 10.1160/th10-08-0516] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2010] [Accepted: 01/24/2011] [Indexed: 02/06/2023]
Abstract
Urokinase-type plasminogen activator (u-PA) plays an important role in tissue remodelling through the activation of plasminogen in the liver, but its mechanisms are less well known. Here, we investigated the involvement of u-PA in the accumulation and phenotypic heterogeneity of macrophages at the damaged site during liver repair. After induction of liver injury by photochemical reaction in mice, the subsequent pathological responses and expression of phenotypic markers in activated macrophages were analysed histologically. Fibrinolytic activity at the damaged site was also examined by fibrin zymography. In wild-type mice, the extent of damage decreased gradually until day 14 and was associated with an accumulation of macrophages at the border of the damaged site. In addition, the macrophages that accumulated near the damaged tissue expressed CD206, a marker of highly phagocytic macrophages, on day 7. Further, macrophages that were adjacent to CD206-positive cells expressed inducible nitric oxide synthase (iNOS), a pro-inflammatory marker. u-PA activity increased at the damaged site on days 4 and 7, which distributed primarily at the border region. In contrast, in u-PA-deficient mice, the decrease in damage size and the accumulation of macrophages were impaired. Further, neither CD206 nor iNOS was expressed in the macrophages that accumulated at the border region in u-PA-deficient mice. Mice deficient for the gene encoding either u-PA receptor (u-PAR) or tissue-type plasminogen activator experienced normal recovery during liver repair. These data indicate that u-PA mediates the accumulation of macrophages and their phenotypic heterogeneity at the border of damaged sites through u-PAR-independent mechanisms.
Collapse
Affiliation(s)
- N Kawao
- Department of Physiology, Kinki University Faculty of Medicine, Osakasayama, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|