351
|
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]
|
352
|
Capobianco A, Rovere-Querini P. Endometriosis, a disease of the macrophage. Front Immunol 2013; 4:9. [PMID: 23372570 PMCID: PMC3556586 DOI: 10.3389/fimmu.2013.00009] [Citation(s) in RCA: 194] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Accepted: 01/07/2013] [Indexed: 12/14/2022] Open
Abstract
Endometriosis, a common cause of pelvic pain and female infertility, depends on the growth of vascularized endometrial tissue at ectopic sites. Endometrial fragments reach the peritoneal cavity during the fertile years: local cues decide whether they yield endometriotic lesions. Macrophages are recruited at sites of hypoxia and tissue stress, where they clear cell debris and heme-iron and generate pro-life and pro-angiogenesis signals. Macrophages are abundant in endometriotic lesions, where are recruited and undergo alternative activation. In rodents macrophages are required for lesions to establish and to grow; bone marrow-derived Tie-2 expressing macrophages specifically contribute to lesions neovasculature, possibly because they concur to the recruitment of circulating endothelial progenitors, and sustain their survival and the integrity of the vessel wall. Macrophages sense cues (hypoxia, cell death, iron overload) in the lesions and react delivering signals to restore the local homeostasis: their action represents a necessary, non-redundant step in the natural history of the disease. Endometriosis may be due to a misperception of macrophages about ectopic endometrial tissue. They perceive it as a wound, they activate programs leading to ectopic cell survival and tissue vascularization. Clearing this misperception is a critical area for the development of novel medical treatments of endometriosis, an urgent and unmet medical need.
Collapse
Affiliation(s)
- Annalisa Capobianco
- Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Scientific Institute Milan, Italy
| | | |
Collapse
|
353
|
Tissues use resident dendritic cells and macrophages to maintain homeostasis and to regain homeostasis upon tissue injury: the immunoregulatory role of changing tissue environments. Mediators Inflamm 2012; 2012:951390. [PMID: 23251037 PMCID: PMC3518145 DOI: 10.1155/2012/951390] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Accepted: 10/25/2012] [Indexed: 01/07/2023] Open
Abstract
Most tissues harbor resident mononuclear phagocytes, that is, dendritic cells and macrophages. A classification that sufficiently covers their phenotypic heterogeneity and plasticity during homeostasis and disease does not yet exist because cell culture-based phenotypes often do not match those found in vivo. The plasticity of mononuclear phagocytes becomes obvious during dynamic or complex disease processes. Different data interpretation also originates from different conceptual perspectives. An immune-centric view assumes that a particular priming of phagocytes then causes a particular type of pathology in target tissues, conceptually similar to antigen-specific T-cell priming. A tissue-centric view assumes that changing tissue microenvironments shape the phenotypes of their resident and infiltrating mononuclear phagocytes to fulfill the tissue's need to maintain or regain homeostasis. Here we discuss the latter concept, for example, why different organs host different types of mononuclear phagocytes during homeostasis. We further discuss how injuries alter tissue environments and how this primes mononuclear phagocytes to enforce this particular environment, for example, to support host defense and pathogen clearance, to support the resolution of inflammation, to support epithelial and mesenchymal healing, and to support the resolution of fibrosis to the smallest possible scar. Thus, organ- and disease phase-specific microenvironments determine macrophage and dendritic cell heterogeneity in a temporal and spatial manner, which assures their support to maintain and regain homeostasis in whatever condition. Mononuclear phagocytes contributions to tissue pathologies relate to their central roles in orchestrating all stages of host defense and wound healing, which often become maladaptive processes, especially in sterile and/or diffuse tissue injuries.
Collapse
|
354
|
Abstract
The ovarian steroid hormones progesterone and estradiol are well established regulators of human endometrial function. However, more recent evidence suggests that androgens and locally generated steroids, such as the glucocorticoids, also have a significant impact on endometrial breakdown and repair. The temporal and spatial pattern of steroid receptor presence in endometrial cells has a significant impact on the endometrial response to steroids. Furthermore, regulation of steroid receptor function by modulatory proteins further refines local responses. This review focuses on steroid regulation of endometrial function during the luteo-follicular transition with a focus on menstruation and endometrial repair.
Collapse
Affiliation(s)
- Jacqueline A Maybin
- MRC Centre for Reproductive Health, The University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK
| | | |
Collapse
|
355
|
Frantz S, Hofmann U, Fraccarollo D, Schäfer A, Kranepuhl S, Hagedorn I, Nieswandt B, Nahrendorf M, Wagner H, Bayer B, Pachel C, Schön MP, Kneitz S, Bobinger T, Weidemann F, Ertl G, Bauersachs J. Monocytes/macrophages prevent healing defects and left ventricular thrombus formation after myocardial infarction. FASEB J 2012; 27:871-81. [PMID: 23159933 DOI: 10.1096/fj.12-214049] [Citation(s) in RCA: 151] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Myocardial infarction (MI) leads to rapid necrosis of cardiac myocytes. To achieve tissue integrity and function, inflammatory cells are activated, including monocytes/macrophages. However, the effect of monocyte/macrophage recruitment after MI remains poorly defined. After experimental MI, monocytes and macrophages were depleted through serial injections of clodronate-containing liposomes. Monocyte/macrophage infiltration was reduced in the myocardium after MI by active treatment. Mortality was increased due to thromboembolic events in monocyte- and macrophage-depleted animals (92 vs. 33%; P<0.01). Left ventricular thrombi were detectable as early as 24 h after MI; this was reproduced in a genetic model of monocyte/macrophage ablation. A general prothrombotic state, increased infarct expansion, and deficient neovascularization were not observed. Severely compromised extracellular matrix remodeling (collagen I, placebo liposome vs. clodronate liposome, 2.4 ± 0.2 vs. 0.8 ± 0.2 arbitrary units; P<0.001) and locally lost integrity of the endocardium after MI are potential mechanisms. Patients with a left ventricular thrombus had a relative decrease of CD14CD16 monocyte/macrophage subsets in the peripheral blood after MI (no thrombus vs. thrombus, 14.2 ± 0.9 vs. 7.80 ± 0.4%; P<0.05). In summary, monocytes/macrophages are of central importance for healing after MI. Impaired monocyte/macrophage function appears to be an unrecognized new pathophysiological mechanism for left ventricular thrombus development after MI.
Collapse
Affiliation(s)
- Stefan Frantz
- Department of Internal Medicine I, University Hospital Würzburg, Comprehensive Heart Failure Center, University of Würzburg, Oberdürrbacher Straße 6, 97080 Würzburg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
356
|
Gliem M, Hermsen D, van Rooijen N, Hartung HP, Jander S. Secondary intracerebral hemorrhage due to early initiation of oral anticoagulation after ischemic stroke: an experimental study in mice. Stroke 2012; 43:3352-7. [PMID: 23117725 DOI: 10.1161/strokeaha.112.666818] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND PURPOSE The uncertain risk of secondary intracerebral hemorrhage (sICH) frequently keeps clinicians from initiating oral anticoagulation (OAC) early after ischemic cardioembolic stroke. The goal of this experimental study was to determine the risk of sICH depending on the timing of OAC initiation relative to stroke onset and to address the role of hematogenous macrophages for repair processes preventing OAC-associated sICH. METHODS C57BL/6 mice were subjected to transient middle cerebral artery occlusion. Subgroups were treated with either the vitamin K antagonist (VKA) phenprocoumon or the direct thrombin inhibitor dabigatran etexilate. Hematogenous macrophages were depleted using intraperitoneal injections of clodronate-filled liposomes. RESULTS Time to therapeutic OAC was 48 hours with VKA and 0.5 hours with dabigatran etexilate treatment. In VKA-treated mice, the risk of sICH was high if effective OAC was already present at stroke onset or achieved within 48 hours after ischemia. With more delayed OAC, the risk of sICH rapidly decreased. Compared with VKA treatment, effective anticoagulation with dabigatran etexilate was associated with a significantly reduced extent of sICH, either if present at stroke onset or if achieved 48 hours later. Partial depletion of macrophages greatly increased the extent of OAC-associated sICH in the subacute stage of 3 to 4 days after ischemia. CONCLUSIONS Our findings suggest that repair mechanisms involving hematogenous macrophages rapidly decrease the risk of OAC-associated sICH in the first days after ischemic stroke. The lower risk of sICH under dabigatran etexilate compared with VKA treatment may facilitate early initiation of OAC after cardioembolic stroke.
Collapse
Affiliation(s)
- Michael Gliem
- Department of Neurology, Heinrich-Heine-University, Medical Faculty, Moorenstr. 5, 40225 Düsseldorf, Germany
| | | | | | | | | |
Collapse
|
357
|
Ren D, Wang X, Ha T, Liu L, Kalbfleisch J, Gao X, Williams D, Li C. SR-A deficiency reduces myocardial ischemia/reperfusion injury; involvement of increased microRNA-125b expression in macrophages. Biochim Biophys Acta Mol Basis Dis 2012; 1832:336-46. [PMID: 23123599 DOI: 10.1016/j.bbadis.2012.10.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Revised: 10/19/2012] [Accepted: 10/22/2012] [Indexed: 01/04/2023]
Abstract
The macrophage scavenger receptor class A (SR-A) participates in the innate immune and inflammatory responses. This study examined the role of macrophage SR-A in myocardial ischemia/reperfusion (I/R) injury and hypoxia/reoxygenation (H/R)-induced cell damage. SR-A(-/-) and WT mice were subjected to ischemia (45min) followed by reperfusion for up to 7days. SR-A(-/-) mice showed smaller myocardial infarct size and better cardiac function than did WT I/R mice. SR-A deficiency attenuated I/R-induced myocardial apoptosis by preventing p53-mediated Bak-1 apoptotic signaling. The levels of microRNA-125b in SR-A(-/-) heart were significantly greater than in WT myocardium. SR-A is predominantly expressed on macrophages. To investigate the role of SR-A macrophages in H/R-induced injury, we isolated peritoneal macrophages from SR-A deficient (SR-A(-/-)) and wild type (WT) mice. Macrophages were subjected to hypoxia followed by reoxygenation. H/R markedly increased NF-κB binding activity as well as KC and MCP-1 production in WT macrophages but not in SR-A(-/-) macrophages. H/R induced caspase-3/7 and -8 activities and cell death in WT macrophages, but not in SR-A(-/-) macrophages. The levels of miR-125b in SR-A(-/-) macrophages were significantly higher than in WT macrophages. Transfection of WT macrophages with miR-125b mimics attenuated H/R-induced caspase-3/7 and -8 activities and H/R-decreased viability, and prevented H/R-increased p-53, Bak-1 and Bax expression. The data suggest that SR-A deficiency attenuates myocardial I/R injury by targeting p53-mediated apoptotic signaling. SR-A(-/-) macrophages contain high levels of miR-125b which may play a role in the protective effect of SR-A deficiency on myocardial I/R injury and H/R-induced cell damage.
Collapse
Affiliation(s)
- Danyang Ren
- Department of Surgery, East Tennessee State University, Johnson City, TN 37614, USA
| | | | | | | | | | | | | | | |
Collapse
|
358
|
Communication in the heart: the role of the innate immune system in coordinating cellular responses to ischemic injury. J Cardiovasc Transl Res 2012; 5:827-36. [PMID: 23054658 DOI: 10.1007/s12265-012-9410-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Accepted: 09/05/2012] [Indexed: 12/14/2022]
Abstract
Ischemic cardiac injury is the leading cause of heart failure and mortality in the USA and is a major expense to health-care systems. Once the heart is injured, a highly dynamic and coordinated immune response is initiated, which is dependent on both resident and recruited leukocytes. The goal of the inflammatory response is to remove ischemic and necrotic material and to promote infarct healing. If this system is perturbed, the myocardium heals poorly, leading to significant left ventricular dysfunction. Understanding how inflammatory cells coordinate and interact with each other is required prior to designing therapeutic interventions that target pathological processes at play and leave untouched those processes that are protective. This review will discuss the intercellular cross talk between cells of the innate immune system following myocardial ischemic injury and how that response is coordinated over time.
Collapse
|
359
|
Santini MP, Rosenthal N. Myocardial regenerative properties of macrophage populations and stem cells. J Cardiovasc Transl Res 2012; 5:700-12. [PMID: 22684511 PMCID: PMC3447141 DOI: 10.1007/s12265-012-9383-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Accepted: 05/24/2012] [Indexed: 01/02/2023]
Abstract
The capacity to regenerate damaged tissue and appendages is lost to some extent in higher vertebrates such as mammals, which form a scar tissue at the expenses of tissue reconstitution and functionality. Whereas this process can protect from further damage and elicit fast healing, it can lead to functional deterioration in organs such as the heart. Based on the analyses performed in the last years, stem cell therapies may not be sufficient to induce cardiac regeneration and additional approaches are required to overcome scar formation. Among these, the immune cells and their humoral response have become a key parameter in regenerative processes. In this review, we will describe the recent findings on the possible therapeutical use of progenitor and immune cells to rescue a damaged heart.
Collapse
|
360
|
Cardiac intercellular communication: are myocytes and fibroblasts fair-weather friends? J Cardiovasc Transl Res 2012; 5:768-82. [PMID: 23015462 DOI: 10.1007/s12265-012-9404-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2012] [Accepted: 08/20/2012] [Indexed: 10/27/2022]
Abstract
The cardiac fibroblast (CF) has historically been thought of as a quiescent cell of the heart, passively maintaining the extracellular environment for the cardiomyocytes (CM), the functional cardiac cell type. The increasingly appreciated role of the CF, however, extends well beyond matrix production, governing many aspects of cardiac function including cardiac electrophysiology and contractility. Importantly, its contributions to cardiac pathophysiology and pathologic remodeling have created a shift in the field's focus from the CM to the CF as a therapeutic target in the treatment of cardiac diseases. In response to cardiac injury, the CF undergoes a pathologic phenotypic transition into a myofibroblast, characterized by contractile smooth muscle proteins and upregulation of collagens, matrix proteins, and adhesion molecules. Further, the myofibroblast upregulates expression and secretion of a variety of pro-inflammatory, profibrotic mediators, including cytokines, chemokines, and growth factors. These mediators act in both an autocrine fashion to further activate CFs, as well as in a paracrine manner on both CMs and circulating inflammatory cells to induce myocyte dysfunction and chronic inflammation, respectively. Together, cell-specific cytokine-induced effects exacerbate pathologic remodeling and progression to HF. A better understanding of this dynamic intercellular communication will lead to novel targets for the attenuation of cardiac remodeling. Current strategies aimed at targeting cytokines have been largely unsuccessful in clinical trials, lending insights into ways that such intercellular cross talk can be more effectively attenuated. This review will summarize the current knowledge regarding CF functions in the heart and will discuss the regulation and signaling behind CF-mediated cytokine production and function. We will then highlight clinical trials that have exploited cytokine cross talk in the treatment of heart failure and provide novel strategies currently under investigation that may more effectively target pathologic CF-CM communication for the treatment of cardiac disease. This review explores novel mechanisms to directly attenuate heart failure progression through inhibition of signaling downstream of pro-inflammatory cytokines that are elevated after cardiac injury.
Collapse
|
361
|
Fan D, Takawale A, Lee J, Kassiri Z. Cardiac fibroblasts, fibrosis and extracellular matrix remodeling in heart disease. FIBROGENESIS & TISSUE REPAIR 2012; 5:15. [PMID: 22943504 PMCID: PMC3464725 DOI: 10.1186/1755-1536-5-15] [Citation(s) in RCA: 603] [Impact Index Per Article: 46.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Accepted: 08/16/2012] [Indexed: 12/30/2022]
Abstract
Fibroblasts comprise the largest cell population in the myocardium. In heart disease, the number of fibroblasts is increased either by replication of the resident myocardial fibroblasts, migration and transformation of circulating bone marrow cells, or by transformation of endothelial/epithelial cells into fibroblasts and myofibroblasts. The primary function of fibroblasts is to produce structural proteins that comprise the extracellular matrix (ECM). This can be a constructive process; however, hyperactivity of cardiac fibroblasts can result in excess production and deposition of ECM proteins in the myocardium, known as fibrosis, with adverse effects on cardiac structure and function. In addition to being the primary source of ECM proteins, fibroblasts produce a number of cytokines, peptides, and enzymes among which matrix metalloproteinases (MMPs) and their inhibitors, tissue inhibitor of metalloproteinases (TIMPs), directly impact the ECM turnover and homeostasis. Function of fibroblasts can also in turn be regulated by MMPs and TIMPs. In this review article, we will focus on the function of cardiac fibroblasts in the context of ECM formation, homeostasis and remodeling in the heart. We will discuss the origins and multiple roles of cardiac fibroblasts in myocardial remodeling in different types of heart disease in patients and in animal models. We will further provide an overview of what we have learned from experimental animal models and genetically modified mice with altered expression of ECM regulatory proteins, MMPs and TIMPs.
Collapse
Affiliation(s)
- Dong Fan
- Department of Physiology, University of Alberta, Edmonton, AB, T6G 2S2, Canada.
| | | | | | | |
Collapse
|
362
|
Gliem M, Mausberg AK, Lee JI, Simiantonakis I, van Rooijen N, Hartung HP, Jander S. Macrophages prevent hemorrhagic infarct transformation in murine stroke models. Ann Neurol 2012; 71:743-52. [PMID: 22718543 DOI: 10.1002/ana.23529] [Citation(s) in RCA: 223] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Inflammation is increasingly viewed as a new therapeutic target in subacute stages of brain infarction. However, apart from causing secondary damage, inflammation could equally promote beneficial lesion remodeling and repair. Distinct subpopulations of monocytes/macrophages (MOs/MPs) may critically determine the outcome of lesion-associated inflammation. METHODS We addressed the role of bone marrow-derived MOs/MPs in 2 different mouse models of ischemic stroke using a combined cell-specific depletion, chemokine receptor knockout, bone marrow chimeric, and pharmacological approach. RESULTS Starting within 24 hours of stroke onset, immature Ly6c(hi) monocytes infiltrated into the infarct border zone and differentiated into mature Ly6c(lo) phagocytes within the lesion compartment. MO/MP infiltration was CCR2-dependent, whereas we did not obtain evidence for additional recruitment via CX3CR1. Depletion of circulating MOs/MPs or selective targeting of CCR2 in bone marrow-derived cells caused delayed clinical deterioration and hemorrhagic conversion of the infarctions. Bleeding frequently occurred around thin-walled, dilated neovessels in the infarct border zone and was accompanied by decreased expression of transforming growth factor (TGF)-β1 and collagen-4, along with diminished activation of Smad2. Injection of TGF-β1 into the lesion border zone greatly reduced infarct bleeding in MO/MP-depleted mice. INTERPRETATION Bone marrow-derived MOs/MPs recruited via CCR2 and acting via TGF-β1 are essential for maintaining integrity of the neurovascular unit following brain ischemia. Future therapies should be aimed at enhancing physiological repair functions of CCR2(+) MOs/MPs rather than blocking their hematogenous recruitment.
Collapse
Affiliation(s)
- Michael Gliem
- Departments of Neurology, Heinrich Heine University, Medical Faculty, Moorenstrasse 5, Düsseldorf, Germany
| | | | | | | | | | | | | |
Collapse
|
363
|
Zamilpa R, Ibarra J, de Castro Brás LE, Ramirez TA, Nguyen N, Halade GV, Zhang J, Dai Q, Dayah T, Chiao YA, Lowell W, Ahuja SS, D'Armiento J, Jin YF, Lindsey ML. Transgenic overexpression of matrix metalloproteinase-9 in macrophages attenuates the inflammatory response and improves left ventricular function post-myocardial infarction. J Mol Cell Cardiol 2012; 53:599-608. [PMID: 22884843 DOI: 10.1016/j.yjmcc.2012.07.017] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Accepted: 07/24/2012] [Indexed: 12/29/2022]
Abstract
Following myocardial infarction (MI), activated macrophages infiltrate into the necrotic myocardium as part of a robust pro-inflammatory response and secrete matrix metalloproteinase-9 (MMP-9). Macrophage activation, in turn, modulates the fibrotic response, in part by stimulating fibroblast extracellular matrix (ECM) synthesis. We hypothesized that overexpression of human MMP-9 in mouse macrophages would amplify the inflammatory and fibrotic responses to exacerbate left ventricular dysfunction. Unexpectedly, at day 5 post-MI, ejection fraction was improved in transgenic (TG) mice (25±2%) compared to the wild type (WT) mice (18±2%; p<0.05). By gene expression profiling, 23 of 84 inflammatory genes were decreased in the left ventricle infarct (LVI) region from the TG compared to WT mice (all p<0.05). Concomitantly, TG macrophages isolated from the LVI, as well as TG peritoneal macrophages stimulated with LPS, showed decreased inflammatory marker expression compared to WT macrophages. In agreement with attenuated inflammation, only 7 of 84 cell adhesion and ECM genes were increased in the TG LVI compared to WT LVI, while 43 genes were decreased (all p<0.05). These results reveal a novel role for macrophage-derived MMP-9 in blunting the inflammatory response and limiting ECM synthesis to improve left ventricular function post-MI.
Collapse
Affiliation(s)
- Rogelio Zamilpa
- San Antonio Cardiovascular Proteomics Center, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78245, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
364
|
Ravi S, Caves JM, Martinez AW, Xiao J, Wen J, Haller CA, Davis ME, Chaikof EL. Effect of bone marrow-derived extracellular matrix on cardiac function after ischemic injury. Biomaterials 2012; 33:7736-45. [PMID: 22819498 DOI: 10.1016/j.biomaterials.2012.07.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Accepted: 07/05/2012] [Indexed: 12/29/2022]
Abstract
Ischemic heart disease is a leading cause of death, with few options to retain ventricular function following myocardial infarction. Hematopoietic-derived progenitor cells contribute to angiogenesis and tissue repair following ischemia reperfusion injury. Motivated by the role of bone marrow extracellular matrix (BM-ECM) in supporting the proliferation and regulation of these cell populations, we investigated BM-ECM injection in myocardial repair. In BM-ECM isolated from porcine sternum, we identified several factors important for myocardial healing, including vascular endothelial growth factor, basic fibroblast growth factor-2, and platelet-derived growth factor-BB. We further determined that BM-ECM serves as an adhesive substrate for endothelial cell proliferation. Bone marrow ECM was injected in a rat model of myocardial infarction, with and without a methylcellulose carrier gel. After one day, reduced infarct area was noted in rats receiving BM-ECM injection. After seven days we observed improved fractional shortening, decreased apoptosis, and significantly lower macrophage counts in the infarct border. Improvements in fractional shortening, sustained through 21 days, as well as decreased fibrotic area, enhanced angiogenesis, and greater c-kit-positive cell presence were associated with BM-ECM injection. Notably, the concentrations of BM-ECM growth factors were 10(3)-10(8) fold lower than typically required to achieve a beneficial effect, as reported in pre-clinical studies that have administered single growth factors alone.
Collapse
Affiliation(s)
- Swathi Ravi
- Department of Biomedical Engineering, Georgia Institute of Technology/Emory University, Atlanta, GA 30332, USA
| | | | | | | | | | | | | | | |
Collapse
|
365
|
Yang M, Zheng J, Miao Y, Wang Y, Cui W, Guo J, Qiu S, Han Y, Jia L, Li H, Cheng J, Du J. Serum-Glucocorticoid Regulated Kinase 1 Regulates Alternatively Activated Macrophage Polarization Contributing to Angiotensin II–Induced Inflammation and Cardiac Fibrosis. Arterioscler Thromb Vasc Biol 2012; 32:1675-86. [DOI: 10.1161/atvbaha.112.248732] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Objective—
Inflammatory responses play a pivotal role in the pathogenesis of hypertensive cardiac remodeling. Macrophage recruitment and polarization contribute to the development of cardiac fibrosis. Although serum-glucocorticoid regulated kinase 1 (SGK1) is a key mediator of fibrosis, its role in regulating macrophage function leading to cardiac fibrosis has not been investigated. We aimed to determine the mechanism by which SGK1 regulates the cardiac inflammatory process, thus contributing to hypertensive cardiac fibrosis.
Methods and Results—
After angiotensin II infusion in mice, cardiac hypertrophy and fibrosis developed in wild-type but not SGK1 knockout mice, with equal levels of hypertension in both groups. Compared with wild-type hearts, SGK1 knockout hearts showed less infiltration of leukocytes and macrophages. Importantly, SGK1 deficiency led to decreased proportion of alternatively activated (M2) macrophages and increased levels of profibrotic cytokines. Angiotensin II infusion induced phosphorylation and nuclear localization of signal transducer and activator of transcription 3 (STAT3) whereas SGK1 knockout hearts showed this effect attenuated. In a 3-dimensional peptide gel culture, inhibition of STAT3 suppressed differentiation into M2 macrophages. Coculture of macrophages with cardiac fibroblasts in 3-dimensional peptide gel stimulated the expression of α-smooth muscle actin and collagen in cardiac fibroblasts. However, SGK1 knockout mice with macrophage deficiency showed reduced fibroblast-to-myofibroblast transition.
Conclusion—
SGK1 may play an important role in macrophage recruitment and M2 macrophage activation by activating the STAT3 pathway, which leads to angiotensin II–induced cardiac fibrosis.
Collapse
Affiliation(s)
- Min Yang
- From the Beijing Anzhen Hospital Affiliated to the Capital Medical University (M.Y., J.Z., Y.M., Y.W., W.C., J.G., S.Q., Y.H., L.J., J.C., J.D.); The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Capital Medical University, Ministry of Education, Beijing Institute of Heart, Lung, and Blood Vessel Diseases (M.Y., J.Z., Y.M., Y.W., W.C., J.G., S.Q., Y.H., L.J., J.C., J.D.); and Department of Pathology, School of Basic Medical Sciences, Capital Medical University, Beijing, China (H.L.)
| | - Jiao Zheng
- From the Beijing Anzhen Hospital Affiliated to the Capital Medical University (M.Y., J.Z., Y.M., Y.W., W.C., J.G., S.Q., Y.H., L.J., J.C., J.D.); The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Capital Medical University, Ministry of Education, Beijing Institute of Heart, Lung, and Blood Vessel Diseases (M.Y., J.Z., Y.M., Y.W., W.C., J.G., S.Q., Y.H., L.J., J.C., J.D.); and Department of Pathology, School of Basic Medical Sciences, Capital Medical University, Beijing, China (H.L.)
| | - Yanjv Miao
- From the Beijing Anzhen Hospital Affiliated to the Capital Medical University (M.Y., J.Z., Y.M., Y.W., W.C., J.G., S.Q., Y.H., L.J., J.C., J.D.); The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Capital Medical University, Ministry of Education, Beijing Institute of Heart, Lung, and Blood Vessel Diseases (M.Y., J.Z., Y.M., Y.W., W.C., J.G., S.Q., Y.H., L.J., J.C., J.D.); and Department of Pathology, School of Basic Medical Sciences, Capital Medical University, Beijing, China (H.L.)
| | - Ying Wang
- From the Beijing Anzhen Hospital Affiliated to the Capital Medical University (M.Y., J.Z., Y.M., Y.W., W.C., J.G., S.Q., Y.H., L.J., J.C., J.D.); The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Capital Medical University, Ministry of Education, Beijing Institute of Heart, Lung, and Blood Vessel Diseases (M.Y., J.Z., Y.M., Y.W., W.C., J.G., S.Q., Y.H., L.J., J.C., J.D.); and Department of Pathology, School of Basic Medical Sciences, Capital Medical University, Beijing, China (H.L.)
| | - Wei Cui
- From the Beijing Anzhen Hospital Affiliated to the Capital Medical University (M.Y., J.Z., Y.M., Y.W., W.C., J.G., S.Q., Y.H., L.J., J.C., J.D.); The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Capital Medical University, Ministry of Education, Beijing Institute of Heart, Lung, and Blood Vessel Diseases (M.Y., J.Z., Y.M., Y.W., W.C., J.G., S.Q., Y.H., L.J., J.C., J.D.); and Department of Pathology, School of Basic Medical Sciences, Capital Medical University, Beijing, China (H.L.)
| | - Jun Guo
- From the Beijing Anzhen Hospital Affiliated to the Capital Medical University (M.Y., J.Z., Y.M., Y.W., W.C., J.G., S.Q., Y.H., L.J., J.C., J.D.); The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Capital Medical University, Ministry of Education, Beijing Institute of Heart, Lung, and Blood Vessel Diseases (M.Y., J.Z., Y.M., Y.W., W.C., J.G., S.Q., Y.H., L.J., J.C., J.D.); and Department of Pathology, School of Basic Medical Sciences, Capital Medical University, Beijing, China (H.L.)
| | - Shulan Qiu
- From the Beijing Anzhen Hospital Affiliated to the Capital Medical University (M.Y., J.Z., Y.M., Y.W., W.C., J.G., S.Q., Y.H., L.J., J.C., J.D.); The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Capital Medical University, Ministry of Education, Beijing Institute of Heart, Lung, and Blood Vessel Diseases (M.Y., J.Z., Y.M., Y.W., W.C., J.G., S.Q., Y.H., L.J., J.C., J.D.); and Department of Pathology, School of Basic Medical Sciences, Capital Medical University, Beijing, China (H.L.)
| | - Yalei Han
- From the Beijing Anzhen Hospital Affiliated to the Capital Medical University (M.Y., J.Z., Y.M., Y.W., W.C., J.G., S.Q., Y.H., L.J., J.C., J.D.); The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Capital Medical University, Ministry of Education, Beijing Institute of Heart, Lung, and Blood Vessel Diseases (M.Y., J.Z., Y.M., Y.W., W.C., J.G., S.Q., Y.H., L.J., J.C., J.D.); and Department of Pathology, School of Basic Medical Sciences, Capital Medical University, Beijing, China (H.L.)
| | - Lixin Jia
- From the Beijing Anzhen Hospital Affiliated to the Capital Medical University (M.Y., J.Z., Y.M., Y.W., W.C., J.G., S.Q., Y.H., L.J., J.C., J.D.); The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Capital Medical University, Ministry of Education, Beijing Institute of Heart, Lung, and Blood Vessel Diseases (M.Y., J.Z., Y.M., Y.W., W.C., J.G., S.Q., Y.H., L.J., J.C., J.D.); and Department of Pathology, School of Basic Medical Sciences, Capital Medical University, Beijing, China (H.L.)
| | - Huihua Li
- From the Beijing Anzhen Hospital Affiliated to the Capital Medical University (M.Y., J.Z., Y.M., Y.W., W.C., J.G., S.Q., Y.H., L.J., J.C., J.D.); The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Capital Medical University, Ministry of Education, Beijing Institute of Heart, Lung, and Blood Vessel Diseases (M.Y., J.Z., Y.M., Y.W., W.C., J.G., S.Q., Y.H., L.J., J.C., J.D.); and Department of Pathology, School of Basic Medical Sciences, Capital Medical University, Beijing, China (H.L.)
| | - Jizhong Cheng
- From the Beijing Anzhen Hospital Affiliated to the Capital Medical University (M.Y., J.Z., Y.M., Y.W., W.C., J.G., S.Q., Y.H., L.J., J.C., J.D.); The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Capital Medical University, Ministry of Education, Beijing Institute of Heart, Lung, and Blood Vessel Diseases (M.Y., J.Z., Y.M., Y.W., W.C., J.G., S.Q., Y.H., L.J., J.C., J.D.); and Department of Pathology, School of Basic Medical Sciences, Capital Medical University, Beijing, China (H.L.)
| | - Jie Du
- From the Beijing Anzhen Hospital Affiliated to the Capital Medical University (M.Y., J.Z., Y.M., Y.W., W.C., J.G., S.Q., Y.H., L.J., J.C., J.D.); The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Capital Medical University, Ministry of Education, Beijing Institute of Heart, Lung, and Blood Vessel Diseases (M.Y., J.Z., Y.M., Y.W., W.C., J.G., S.Q., Y.H., L.J., J.C., J.D.); and Department of Pathology, School of Basic Medical Sciences, Capital Medical University, Beijing, China (H.L.)
| |
Collapse
|
366
|
Naresh NK, Xu Y, Klibanov AL, Vandsburger MH, Meyer CH, Leor J, Kramer CM, French BA, Epstein FH. Monocyte and/or macrophage infiltration of heart after myocardial infarction: MR imaging by using T1-shortening liposomes. Radiology 2012; 264:428-35. [PMID: 22723500 DOI: 10.1148/radiol.12111863] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE To test the hypothesis that magnetic resonance (MR) imaging R1 (R1 = 1/T1) mapping after selectively labeling monocytes with a T1-shortening contrast agent in vivo would enable the quantitative measurement of their spatiotemporal kinetics in the setting of infarct healing. MATERIALS AND METHODS All procedures were performed in mice and were approved by the institutional committee on animal research. One hundred microliters of dual-labeled liposomes (DLLs) containing gadolinium (Gd)-diethylenetriaminepentaacetic acid (DTPA)-bis(stearylamide) and DiI dye were used to label monocytes 2 days before myocardial infarction (MI). MI was induced by occlusion of the left anterior descending coronary artery for 1 hour, followed by reperfusion. MR imaging R1 mapping of mouse hearts was performed at baseline on day -3, on day 0 before MI, and on days 1, 4, and 7 after MI. Mice without labeling were used as controls. ΔR1 was calculated as the difference in R1 between mice with labeling and those without labeling. CD68 immunohistochemistry and DiI fluorescence microscopy were used to confirm that labeled monocytes and/or macrophages infiltrated the postinfarct myocardium. Statistical analysis was performed by using two-way analysis of variance and the unpaired two-sample t test. RESULTS Infarct zone ΔR1 was slightly but nonsignificantly increased on day 1, maximum on day 4 (P < .05 vs all other days), and started to decrease by day 7 (P < .05 vs days -3, 0, and 1) after MI, closely reflecting the time course of monocyte and/or macrophage infiltration of the infarcted myocardium shown by prior histologic studies. Histologic results confirmed the presence and location of DLL-labeled monocytes and/or macrophages in the infarct zone on day 4 after MI. CONCLUSION R1 mapping after labeling monocytes with T1-shortening DLLs enables the measurement of post-MI monocyte and/or macrophage spatiotemporal kinetics.
Collapse
Affiliation(s)
- Nivedita K Naresh
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
367
|
Doyle KP, Buckwalter MS. The double-edged sword of inflammation after stroke: What sharpens each edge? Ann Neurol 2012; 71:729-31. [DOI: 10.1002/ana.23579] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
368
|
Toll-like receptor 3 ligand polyinosinic:polycytidylic acid promotes wound healing in human and murine skin. J Invest Dermatol 2012; 132:2085-92. [PMID: 22572822 DOI: 10.1038/jid.2012.120] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Toll-like receptors (TLRs) are pattern-recognition receptors and have a critical role in both innate and adaptive responses to tissue injury. Our previous study showed that wound healing was impaired in TLR3-deficient mice. In this study, we investigated the capacity of the TLR3 agonist polyriboinosinic-polyribocytidylic acid (poly(I:C)) to promote the healing of skin wounds in humans and mice. We found that topical application with poly(I:C) accelerated the closure of wounds in patients with laser plastic surgery. In a mouse model, topical application of poly(I:C) markedly enhanced re-epithelialization, granulation, and neovascularization required for wound closure. Further studies revealed that poly(I:C) treatment resulted in enhanced recruitment of neutrophils and macrophages in association with upregulation of a chemokine, macrophage inflammatory protein-2 (MIP-2/CXCL2), in the wounds. The effect of poly(I:C) was abolished in TLR3-deficient mice or by treatment with MIP-2/CXCL2-neutralizing antibodies. These results suggest a potential therapeutic value of the TLR3 activator poly(I:C) for wound healing.
Collapse
|
369
|
Gao XM, White DA, Dart AM, Du XJ. Post-infarct cardiac rupture: Recent insights on pathogenesis and therapeutic interventions. Pharmacol Ther 2012; 134:156-79. [DOI: 10.1016/j.pharmthera.2011.12.010] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Accepted: 12/20/2011] [Indexed: 01/15/2023]
|
370
|
Weidenbusch M, Anders HJ. Tissue microenvironments define and get reinforced by macrophage phenotypes in homeostasis or during inflammation, repair and fibrosis. J Innate Immun 2012; 4:463-77. [PMID: 22507825 DOI: 10.1159/000336717] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2011] [Accepted: 01/23/2012] [Indexed: 12/17/2022] Open
Abstract
Current macrophage phenotype classifications are based on distinct in vitro culture conditions that do not adequately mirror complex tissue environments. In vivo monocyte progenitors populate all tissues for immune surveillance which supports the maintenance of homeostasis as well as regaining homeostasis after injury. Here we propose to classify macrophage phenotypes according to prototypical tissue environments, e.g. as they occur during homeostasis as well as during the different phases of (dermal) wound healing. In tissue necrosis and/or infection, damage- and/or pathogen-associated molecular patterns induce proinflammatory macrophages by Toll-like receptors or inflammasomes. Such classically activated macrophages contribute to further tissue inflammation and damage. Apoptotic cells and an-tiinflammatory cytokines dominate in postinflammatory tissues which induce macrophages to produce more anti-inflammatory mediators. Similarly, tumor-associated macrophages also confer immunosuppression in tumor stroma. Insufficient parenchymal healing despite abundant growth factors pushes macrophages to gain a profibrotic phenotype and promote fibrocyte recruitment which both enforce tissue scarring. Ischemic scars are largely devoid of cytokines and growth factors so that fibrolytic macrophages that predominantly secrete proteases digest the excess extracellular matrix. Together, macrophages stabilize their surrounding tissue microenvironments by adapting different phenotypes as feed-forward mechanisms to maintain tissue homeostasis or regain it following injury. Furthermore, macrophage heterogeneity in healthy or injured tissues mirrors spatial and temporal differences in microenvironments during the various stages of tissue injury and repair.
Collapse
Affiliation(s)
- Marc Weidenbusch
- Medizinische Klinik IV, Klinikum der Universität München-LMU, München, Deutschland
| | | |
Collapse
|
371
|
Fraccarollo D, Galuppo P, Bauersachs J. Novel therapeutic approaches to post-infarction remodelling. Cardiovasc Res 2012; 94:293-303. [PMID: 22387461 DOI: 10.1093/cvr/cvs109] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Adverse cardiac remodelling is a major cause of morbidity and mortality following acute myocardial infarction (MI). Mechanical and neurohumoral factors involved in structural and molecular post-infarction remodelling were important targets in research and treatment for years. More recently, therapeutic strategies that address myocardial regeneration and pathophysiological mechanisms of infarct wound healing appear to be useful novel tools to prevent progressive ventricular dilation, functional deterioration, life-threatening arrhythmia, and heart failure. This review provides an overview of future and emerging therapies for cardiac wound healing and remodelling after MI.
Collapse
Affiliation(s)
- Daniela Fraccarollo
- Klinik fuer Kardiologie und Angiologie, Medizinische Hochschule Hannover, Hannover, Germany
| | | | | |
Collapse
|
372
|
Moonen JRA, Harmsen MC, Krenning G. Cellular plasticity: the good, the bad, and the ugly? Microenvironmental influences on progenitor cell therapy. Can J Physiol Pharmacol 2012; 90:275-85. [DOI: 10.1139/y11-107] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Progenitor cell based therapies have emerged for the treatment of ischemic cardiovascular diseases where there is insufficient endogenous repair. However, clinical success has been limited, which challenges the original premise that transplanted progenitor cells would orchestrate repair. In this review, we discuss the basics of endothelial progenitor cell therapy and describe how microenvironmental changes (i.e., trophic and mechano-structural factors) in the damaged myocardium influence progenitor cell plasticity and hamper beneficial therapeutic outcome. Further understanding of these microenvironmental clues will enable optimization of cell therapy at all levels. We discuss current concepts and provide future perspectives for the enhancement of progenitor cell therapy, and merge these advances into a combined approach for ischemic tissue repair.
Collapse
Affiliation(s)
- Jan-Renier A.J. Moonen
- Cardiovascular Regenerative Medicine Research Group (CAVAREM), Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1 (EA11), 9713GZ Groningen, the Netherlands
| | - Martin C. Harmsen
- Cardiovascular Regenerative Medicine Research Group (CAVAREM), Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1 (EA11), 9713GZ Groningen, the Netherlands
| | - Guido Krenning
- Cardiovascular Regenerative Medicine Research Group (CAVAREM), Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1 (EA11), 9713GZ Groningen, the Netherlands
| |
Collapse
|
373
|
Peña VBA, Bonini IC, Antollini SS, Kobayashi T, Barrantes FJ. alpha 7-type acetylcholine receptor localization and its modulation by nicotine and cholesterol in vascular endothelial cells. J Cell Biochem 2012; 112:3276-88. [PMID: 21748784 DOI: 10.1002/jcb.23254] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The neuronal-type α7 nicotinic acetylcholine receptor (α7AChR) is also found in various non-neural tissues, including vascular endothelium, where its peculiar ionotropic properties (high Ca(2+) permeability) and its supervening Ca(2+) -mediated intracellular cascades may play important roles in physiology (angiogenesis) and pathology (inflammation and atherogenesis). Changes in molecular (up-regulation, affinity, and conformational states) and cellular (distribution, association with membranes) properties of the α7AChR related to angiogenesis (wound-repair cell migration) and atherogenesis (alterations in cholesterol content) were studied in living endothelial cells, with the aim of determining whether such changes constitute early markers of inflammatory response. The combination of pharmacological, biochemical, and fluorescence microscopy tools showed that α7AChRs in rat arterial endothelial (RAEC) and human venous endothelial (HUVEC) cells occur at extremely low expression levels (∼50 fmol/mg protein) but undergo agonist-induced up-regulation at relatively high nicotine concentrations (∼300-fold with 50 µM ligand), increasing their cell-surface exposure. When analyzed in terms of cold Triton X-100 solubility and subcellular distribution, α7AChRs occur in the "non-raft" subcellular membrane fractions. Acute cholesterol depletion reduced not only cholesterol levels but also the number of cell-surface α7AChRs. Nicotine exposure markedly stimulated cell migration and accelerated wound repair, which drastically diminished in cells deprived of the sterol. The angiogenic effect of nicotine appears to be synergistic with cholesterol content. Finally, the apparent K(D) of α7AChRs for the open-channel blocker crystal violet was found to be ∼600-fold lower in receptor-enriched membranes obtained from up-regulated HUVEC.
Collapse
Affiliation(s)
- Victoria B Ayala Peña
- Instituto de Investigaciones Bioquímicas and UNESCO Chair of Biophysics & Molecular Neurobiology, 8000 Bahía Blanca, Argentina
| | | | | | | | | |
Collapse
|
374
|
Dakin SG, Werling D, Hibbert A, Abayasekara DRE, Young NJ, Smith RKW, Dudhia J. Macrophage sub-populations and the lipoxin A4 receptor implicate active inflammation during equine tendon repair. PLoS One 2012; 7:e32333. [PMID: 22384219 PMCID: PMC3284560 DOI: 10.1371/journal.pone.0032333] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2011] [Accepted: 01/25/2012] [Indexed: 01/15/2023] Open
Abstract
Macrophages (Mϕ) orchestrate inflammatory and reparatory processes in injured connective tissues but their role during different phases of tendon healing is not known. We investigated the contribution of different Mϕ subsets in an equine model of naturally occurring tendon injury. Post mortem tissues were harvested from normal (uninjured), sub-acute (3–6 weeks post injury) and chronically injured (>3 months post injury) superficial digital flexor tendons. To determine if inflammation was present in injured tendons, Mϕ sub-populations were quantified based on surface antigen expression of CD172a (pan Mϕ), CD14highCD206low (pro-inflammatory M1Mϕ), and CD206high (anti-inflammatory M2Mϕ) to assess potential polarised phenotypes. In addition, the Lipoxin A4 receptor (FPR2/ALX) was used as marker for resolving inflammation. Normal tendons were negative for both Mϕ and FPR2/ALX. In contrast, M1Mϕ predominated in sub-acute injury, whereas a potential phenotype-switch to M2Mϕ polarity was seen in chronic injury. Furthermore, FPR2/ALX expression by tenocytes was significantly upregulated in sub-acute but not chronic injury. Expression of the FPR2/ALX ligand Annexin A1 was also significantly increased in sub-acute and chronic injuries in contrast to low level expression in normal tendons. The combination of reduced FPR2/ALX expression and persistence of the M2Mϕ phenotype in chronic injury suggests a potential mechanism for incomplete resolution of inflammation after tendon injury. To investigate the effect of pro-inflammatory mediators on lipoxin A4 (LXA4) production and FPR2/ALX expression in vitro, normal tendon explants were stimulated with interleukin-1 beta and prostaglandin E2. Stimulation with either mediator induced LXA4 release and maximal upregulation of FPR2/ALX expression after 72 hours. Taken together, our data suggests that although tenocytes are capable of mounting a protective mechanism to counteract inflammatory stimuli, this appears to be of insufficient duration and magnitude in natural tendon injury, which may potentiate chronic inflammation and fibrotic repair, as indicated by the presence of M2Mϕ.
Collapse
Affiliation(s)
- Stephanie Georgina Dakin
- Department of Veterinary Clinical Sciences, Royal Veterinary College, University of London, Hatfield, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|
375
|
Abstract
Zoledronate (Zol), one of the class of bisphophonate drugs, is commonly used to treat postmenopausal osteoporosis. Treatment of liposomal bisphosphonates has been shown to worsen myocardial infarct repair in an experimental model. The purpose of this study was to investigate the effect of Zol in the repair of chronically infarcted myocardium without liposomal encapsulation to mimic the clinical setting. Zol (20 μg/kg, a dose known to treat experimental osteoporosis in rats, n = 15) was administered subcutaneously to female Sprague-Dawley rats 1 day before coronary artery ligation. Rats receiving phosphate-buffered saline (n = 12) were used as controls. Left ventricular function, infarct size, and remodeling were studied at 4 weeks postinfarction. Zol pretreatment did not affect left ventricular ejection fraction in hearts with myocardial infarction (49.5 ± 1.4% in Zol; 50.6 ± 2.1% in phosphate-buffered saline). Infarct size was similar in Zol versus untreated hearts (34.2% ± 2.9% in Zol; 33.4% ± 2.9% in phosphate-buffered saline). Left ventricular cavity volume and circumference, infarct thickness, and expansion index were comparable between the groups. To investigate a potential effect of Zol on tissue macrophage infiltration after myocardial infarction, heart specimens were harvested 48 hours postinfarction and sections were immunostained with CD68 antibody, a macrophage-specific marker. Results of macrophage immunostaining revealed that the level of tissue macrophage infiltration was similar between groups. In conclusion, administration of Zol before myocardial infarction had no adverse effects on cardiac contractile function, infarct size, or remodeling. These results suggest that treatment of Zol given before the onset of myocardial infarction does not cause worsening of infarct repair.
Collapse
|
376
|
Anzai A, Anzai T, Nagai S, Maekawa Y, Naito K, Kaneko H, Sugano Y, Takahashi T, Abe H, Mochizuki S, Sano M, Yoshikawa T, Okada Y, Koyasu S, Ogawa S, Fukuda K. Regulatory role of dendritic cells in postinfarction healing and left ventricular remodeling. Circulation 2012; 125:1234-45. [PMID: 22308302 DOI: 10.1161/circulationaha.111.052126] [Citation(s) in RCA: 238] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Inflammation and immune responses are integral components in the healing process after myocardial infarction. We previously reported dendritic cell (DC) infiltration in the infarcted heart; however, the precise contribution of DC in postinfarction healing is unclear. METHODS AND RESULTS Bone marrow cells from CD11c-diphtheria toxin receptor/green fluorescent protein transgenic mice were transplanted into lethally irradiated wild-type recipient mice. After reconstitution of bone marrow-derived cells, the recipient mice were treated with either diphtheria toxin (DC ablation) or vehicle (control), and myocardial infarction was created by left coronary ligation. CD11c(+) green fluorescent protein-positive DCs expressing CD11b and major histocompatibility complex class II were recruited into the heart, peaking on day 7 after myocardial infarction in the control group. Mice with DC ablation for 7 days showed deteriorated left ventricular function and remodeling. The DC-ablated group demonstrated enhanced and sustained expression of inflammatory cytokines such as interleukin-1β, interleukin-18, and tumor necrosis factor-α, prolonged extracellular matrix degradation associated with a high level of matrix metalloproteinase-9 activity, and diminished expression level of interleukin-10 and endothelial cell proliferation after myocardial infarction compared with the control group. In vivo analyses revealed that DC-ablated infarcts had enhanced monocyte/macrophage recruitment. Among these cells, marked infiltration of proinflammatory Ly6C(high) monocytes and F4/80(+) CD206(-) M1 macrophages and, conversely, impaired recruitment of anti-inflammatory Ly6C(low) monocytes and F4/80(+) CD206(+) M2 macrophages in the infarcted myocardium were identified in the DC-ablated group compared with the control group. CONCLUSIONS These results suggest that the DC is a potent immunoprotective regulator during the postinfarction healing process via its control of monocyte/macrophage homeostasis.
Collapse
Affiliation(s)
- Atsushi Anzai
- Division of Cardiology, Department of Medicine, Keio University School of Medicine, Tokyo, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
377
|
Pfister R, Sharp SJ, Luben R, Wareham NJ, Khaw KT. Differential white blood cell count and incident heart failure in men and women in the EPIC-Norfolk study. Eur Heart J 2012; 33:523-530. [PMID: 22173908 DOI: 10.1093/eurheartj/ehr457] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
Abstract
AIMS Markers of inflammation are associated with increased risk of heart failure, but data on differential white blood cell (WBC) count are lacking. We examined the prospective association between differential WBC count and incident heart failure events. METHODS AND RESULTS Hazard ratios (HRs) (per increase of 1000 cells/μL, 95% confidence interval) of total WBC count and individual components on heart failure were calculated in apparently healthy 7195 men and 8816 women aged 39-79 participating in the 'European Prospective Investigation into Cancer and Nutrition' (EPIC) study in Norfolk. During a mean follow-up of 12.4 years, 935 incident cases of heart failure occurred. In women, neither total WBC count (1.02, 0.96-1.09) nor individual components were associated with HR of heart failure after accounting for known risk factors. In men, HR of heart failure increased with increasing levels of total WBC count (1.09, 1.04-1.15) after accounting for established risk factors; analysis of WBC components showed increased hazard with increasing levels of granulocyte count (1.16, 1.09-1.24) and, independently of this, decreased hazard with increasing levels of monocyte count (0.71, 0.53-0.93); lymphocyte count was not significantly associated with heart failure (0.97, 0.83-1.13). Results did not change materially after excluding smokers, adjusting for intermediate myocardial infarction and coronary heart disease and C-reactive protein. CONCLUSION Inflammation as measured by WBC count was independently associated with incident heart failure in apparently healthy men but not women. The association observed in men was driven by granulocyte count, but there was an independent inverse association between monocyte count and incident heart failure.
Collapse
Affiliation(s)
- Roman Pfister
- Medical Research Council Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Addenbrooke's Hospital, Hills Road, Cambridge, UK.
| | | | | | | | | |
Collapse
|
378
|
Chen C, Uludag H, Wang Z, Rezansoff A, Jiang H. Macrophages Inhibit Migration, Metabolic Activity and Osteogenic Differentiation of Human Mesenchymal Stem Cells in vitro. Cells Tissues Organs 2012; 195:473-83. [DOI: 10.1159/000330686] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/22/2011] [Indexed: 12/15/2022] Open
|
379
|
Krenning G, van der Strate BWA, Schipper M, Brouwer LA, Fernandes BCA, van Luyn MJA, Harmsen MC. Combined implantation of CD34+ and CD14+ cells increases neovascularization through amplified paracrine signalling. J Tissue Eng Regen Med 2011; 7:118-28. [PMID: 22125235 DOI: 10.1002/term.503] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2010] [Revised: 02/09/2011] [Accepted: 07/13/2011] [Indexed: 12/20/2022]
Abstract
Cell therapy strategies that use adult peripheral blood-derived CD34⁺ progenitor cells are hampered by low cell numbers and the infrequent cellular incorporation into the neovasculature. Hence, the use of CD34⁺ cells to treat ischaemic diseases is under debate. Interaction between CD34⁺ cells and CD14⁺ cells results in superior endothelial differentiation of CD14⁺ cells in vitro, indicating that cell therapy approaches utilizing both CD34⁺ and CD14⁺ cells may be advantageous in therapeutic neovascularization. Here, human CD34⁺ and CD14⁺ cells were isolated from adult peripheral blood and implanted subcutaneously into nude mice, using matrigel as the carrier. Combined implantation of human CD34⁺ and CD14⁺ cells resulted in superior neovascularization, compared to either cell type alone, albeit incorporation of human cells into the murine vasculature was not observed. Human CD34⁺ and CD14⁺ cells produced and secreted a pentad of pro-angiogenic mediators, such as HGF, MCP-1 and IL-8, bFGF and VEGFa in monoculture. The production and secretion of pro-angiogenic mediators by CD14⁺ cells was highly amplified upon incubation with conditioned medium from CD34⁺ cells. In vivo, neovascularization of matrigel implants did not rely on the endothelial differentiation and incorporation of CD34⁺ or CD14⁺ cells, but depended on the paracrine effects of IL-8, MCP-1, HGF, bFGF and VEGFa secreted by implanted cells. Administration of this growth factor/cytokine pentad using matrigel as a carrier results in cell recruitment and microvessel formation equal to progenitor cell-induced neovascularization. These data provide new insights on neovascularization by cell therapy and may contribute to new strategies for the treatment of ischaemic diseases.
Collapse
Affiliation(s)
- G Krenning
- Cardiovascular Regenerative Medicine Research Group-CAVAREM, Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
380
|
Zhao T, Zhao W, Chen Y, Ahokas RA, Sun Y. Acidic and basic fibroblast growth factors involved in cardiac angiogenesis following infarction. Int J Cardiol 2011; 152:307-13. [PMID: 20674996 PMCID: PMC3061206 DOI: 10.1016/j.ijcard.2010.07.024] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2010] [Revised: 05/25/2010] [Accepted: 07/04/2010] [Indexed: 12/15/2022]
Abstract
Acidic and basic fibroblast growth factors (FGF-1/FGF-2) promote angiogenesis in cancer. Angiogenesis is integral to cardiac repair following myocardial infarction (MI). The potential regulation of FGF-1/FGF-2 in cardiac angiogenesis postMI remains unexplored. Herein, we examined the temporal and spatial expression of FGF-1/FGF-2 and FGF receptors (FGFR) in the infarcted rat heart at days 1, 3, 7, and 14 postMI. FGF-1/-2 gene and protein expression, cells expressing FGF-1/-2 and FGFR expression were examined by quantitative in situ hybridization, RT-PCR; western blot, immunohistochemistry and quantitative in vitro autoradiography. Compared to the normal heart, we found that in the border zone and infarcted myocardium 1) FGF-1 gene expression was increased in the first week postMI and returned to control levels at week 2; FGF-1 protein levels were, however, largely reduced at day 1, then elevated at day 3 peaked at day 7 and declined at day 14; and cells expressing FGF-1 were primarily inflammatory cells; 2) FGF-2 gene expression was significantly elevated from day 1 to day 14; the increase in FGF-2 protein level was most evident at day 7 and cells expressing FGF-2 were primarily endothelial cells; 3) FGFR expression started to increase at day 3 and remained elevated thereafter; and 4) FGF-1/FGF-2 and FGFR expression remained unchanged in the noninfarcted myocardium. Thus, FGF-1/FGF-2 and FGFR expression are enhanced in the infarcted myocardium in the early stage after MI, which is spatially and temporally coincident with angiogenesis, suggesting that FGF-1/FGF-2 are involved in regulating cardiac angiogenesis and repair.
Collapse
Affiliation(s)
- Tieqiang Zhao
- Division of Cardiovascular Diseases, Department of Medicine, University of Tennessee Health Science Center, 956 Court Ave., Rm B324, Memphis, TN 38163, United States
| | | | | | | | | |
Collapse
|
381
|
Ploeger DTA, van Putten SM, Koerts JA, van Luyn MJA, Harmsen MC. Human macrophages primed with angiogenic factors show dynamic plasticity, irrespective of extracellular matrix components. Immunobiology 2011; 217:299-306. [PMID: 22093249 DOI: 10.1016/j.imbio.2011.10.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2011] [Revised: 10/16/2011] [Accepted: 10/18/2011] [Indexed: 12/26/2022]
Abstract
Macrophages are important in inflammation as well as in tissue repair processes. They can be activated by various stimuli and classified into two major groups: M1 (classically activated) or M2 (alternatively activated). Inflammation, angiogenesis and matrix remodeling play a major role in tissue repair. Here, we investigate the combined influence of a pro-angiogenic microenvironment and specific extracellular matrix (ECM) components or tissue culture polystyrene (TCPS) on the dynamics of human macrophage polarization. We established that human angiogenically primed macrophages cultured on different ECM components exhibit an M2-like polarization. These M2-like macrophages polarized to M1 and M2 macrophages with classical (LPS and IFNγ) stimuli and alternative (IL-4 and IL-13) stimuli respectively. Moreover, these M1 and M2 (primary) polarized macrophages rapidly underwent a secondary (re)polarization to M2 and M1 with conditioned media from M2 and M1 primary polarized macrophages respectively. In these initial priming and later (re)polarization processes the soluble factors had a dominant and orchestrating role, while the type of ECM (collagen I, fibronectin, versus tissue culture polystyrene) did not play a crucial role on the polarization of macrophages.
Collapse
Affiliation(s)
- Diana T A Ploeger
- Department of Pathology and Medical Biology, University Medical Centre Groningen, University of Groningen, Hanzeplein 1 (HPC EA11), 9713 GZ Groningen, The Netherlands
| | | | | | | | | |
Collapse
|
382
|
Freytes DO, Santambrogio L, Vunjak-Novakovic G. Optimizing dynamic interactions between a cardiac patch and inflammatory host cells. Cells Tissues Organs 2011; 195:171-82. [PMID: 21996612 DOI: 10.1159/000331392] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Damaged heart muscle has only a minimal ability for regeneration following myocardial infarction in which cardiomyocytes are lost to ischemia. The most clinically promising approach to regeneration of cardiac muscle currently under investigation is that of injecting cardiogenic repair cells or implanting a preformed tissue-engineered patch. While major advances are being made in the derivation of functional human cardiomyocytes and the development of tissue-engineering modalities for cardiac repair, the host environment into which the repair cells are placed is largely overlooked. Within seconds of myocardial ischemia, hypoxia sets in in the myocardium and the inflammatory response starts, characterized by rapid deployment of circulating cells and the release of paracrine and autocrine signals. Therefore, the inflammatory conditions under which these interactions take place, the design of the scaffold material used, and the maturity of the implanted cells will determine the outcomes of any stem cell-based therapy. We discuss here the interactions between implanted and inflammatory cells of the host, which are critical for the design of effective heart repair therapies.
Collapse
Affiliation(s)
- Donald O Freytes
- Department of Biomedical Engineering, Columbia University, New York, N.Y., USA
| | | | | |
Collapse
|
383
|
Wrigley BJ, Lip GYH, Shantsila E. The role of monocytes and inflammation in the pathophysiology of heart failure. Eur J Heart Fail 2011; 13:1161-71. [PMID: 21952932 DOI: 10.1093/eurjhf/hfr122] [Citation(s) in RCA: 147] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
There is growing evidence to support an important role of inflammation in the underlying pathophysiology of heart failure (HF). Indeed, inflammatory cytokine levels are well recognized to be increased in patients with left ventricular dysfunction and appear to have prognostic implications. Monocytes play a pivotal role in the inflammatory cascade and are a major source of both pro- and anti-inflammatory cytokines. They are intimately involved in tissue damage and repair and an imbalance of these processes may have detrimental consequences for the failing myocardium. Importantly, monocytes comprise of distinct subsets with different cell surface markers and functional characteristics and this heterogeneity may be important in understanding their specific role in HF. In HF, monocyte activation involves interplay between pattern recognition molecules, endotoxins, cytokines, and acute phase proteins. Activated monocytes migrate to the myocardium in response to powerful chemokines, where they must then attach to the endothelial wall before infiltrating into the myocardium itself. This review article aims to discuss the role of monocytes and inflammation in HF, focusing on monocyte activation, mobilisation, recruitment and endothelial adherence, as well as the effects they may have on myocardial performance. The therapeutic modulation of inflammation and monocyte activation in HF treatment will also be reviewed.
Collapse
Affiliation(s)
- Benjamin J Wrigley
- University of Birmingham Centre for Cardiovascular Sciences, City Hospital, Birmingham B18 7QH, UK
| | | | | |
Collapse
|
384
|
Boulberdaa M, Urayama K, Nebigil CG. Prokineticin receptor 1 (PKR1) signalling in cardiovascular and kidney functions. Cardiovasc Res 2011; 92:191-8. [DOI: 10.1093/cvr/cvr228] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
|
385
|
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
|
386
|
Hu Y, Zhang H, Lu Y, Bai H, Xu Y, Zhu X, Zhou R, Ben J, Xu Y, Chen Q. Class A scavenger receptor attenuates myocardial infarction-induced cardiomyocyte necrosis through suppressing M1 macrophage subset polarization. Basic Res Cardiol 2011; 106:1311-28. [PMID: 21769674 DOI: 10.1007/s00395-011-0204-x] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Revised: 06/27/2011] [Accepted: 07/07/2011] [Indexed: 12/30/2022]
Abstract
Classically (M1) and alternatively (M2) activated macrophage subsets play differential roles in left ventricular remodeling after myocardial infarction (MI). The precise mechanism underlying the regulation of M1/M2 polarization during MI is unknown. We hypothesized that class A scavenger receptor (SR-A), a key modulator of inflammation, may steer macrophage polarization, which in turn influences cardiomyocytes necrosis after MI. MI was induced in wild type (WT) and SR-A deficient (SR-A(-/-)) mice by left anterior descending coronary artery ligation. Cardiac function deterioration, ventricular dilatation and fibrosis were all exacerbated in SR-A(-/-) mice following MI compared to WT littermates. Meanwhile, enhanced M1 macrophage polarization was observed in SR-A(-/-) mice, along with increased production of M1 signature cytokines including interleukin-1β (IL-1β), IL-6, and tumor necrosis factor-α (TNF-α) as demonstrated by immunohistochemistry, flow cytometry, quantitative real-time PCR, and ELISA assays. Moreover, activation of the activated apoptosis signal regulating kinase 1 (ASK1)/p38 mitogen-activated protein kinase (MAPK)/nuclear factor-κB (NF-κB) signaling pathway was markedly elevated in SR-A(-/-) animals post-MI. Most importantly, transplantation using bone marrow from SR-A(+/+) mice partially restored M1 macrophages and significantly augmented left ventricular fractional shortening in SR-A(-/-) mice. SR-A attenuated MI-induced cardiac remodeling by suppressing macrophage polarization toward a skewed M1 phenotype, reducing secretion of IL-1β, IL-6, and TNF-α, and dampening the ASK1/p38/NF-κB signaling pathway. Therefore, SR-A may exert a protective effect against MI, which may represent a new interventional target for treatment of post-infarct remodeling and subsequent heart failure.
Collapse
Affiliation(s)
- Yulong Hu
- Atherosclerosis Research Center, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing 210029, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
387
|
Chamberlain CS, Leiferman EM, Frisch KE, Wang S, Yang X, van Rooijen N, Baer GS, Brickson SL, Vanderby R. The influence of macrophage depletion on ligament healing. Connect Tissue Res 2011; 52:203-11. [PMID: 21117894 PMCID: PMC3110150 DOI: 10.3109/03008207.2010.511355] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Despite a complex cascade of cellular events to reconstruct damaged extracellular matrix (ECM), ligament healing results in a mechanically inferior, scar-like tissue. During normal healing, the number of macrophages significantly increases within the wound site. Then, granulation tissue expands into any residual, normal ligamentous tissue (creeping substitution), resulting in a larger region of healing, greater mechanical compromise, and an inefficient repair process. To study the effects of macrophages on the repair process, bilateral, surgical rupture of their medial collateral ligaments (MCLs) was done on rats. Treatment animals received liposome-encapsulated clodronate, 2 days before rupture to ablate phagocytosing macrophages. Ligaments were then collected at days 5, 11, and 28 for immunohistochemistry (IHC) and/or mechanical testing. Clodronate treatment reduced both the M1 and M2 macrophages at day 5 and altered early healing. However, the macrophages effectively returned to control levels after day 5 and reinitiated a wound-healing response. Our results suggest that an early macrophage response, which is necessary for debridement of damaged tissue in the wound, is also important for cytokine release to mediate normal repair processes. Additionally, nonspecific inhibition of macrophages (without regard to specific macrophage populations) can control excessive granulation tissue formation but is detrimental to early matrix formation and ligament strength.
Collapse
Affiliation(s)
- Connie S Chamberlain
- Department of Orthopedics and Rehabilitation, University of Wisconsin, Madison, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
388
|
Miyasato SK, Loeffler J, Shohet R, Zhang J, Lindsey M, Le Saux CJ. Caveolin-1 modulates TGF-β1 signaling in cardiac remodeling. Matrix Biol 2011; 30:318-29. [PMID: 21641995 DOI: 10.1016/j.matbio.2011.05.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2010] [Revised: 04/27/2011] [Accepted: 05/20/2011] [Indexed: 02/06/2023]
Abstract
The cardiac response to myocardial injury includes fibrotic and hypertrophic processes and a key mediator in this response is transforming growth factor-β1 (TGF-β1). Caveolin-1 (cav1), the main structural protein of caveolae, is an inhibitor of the TGF-β1 signaling pathway. To examine the role of cav1 in cardiac repair, cav1 deficient (Cav1(-/-)) and wild type (WT) mice were subjected to cryoinjury of the left ventricle (LV). At baseline the two groups exhibited no inflammation, similar collagen content, and similar cardiac function. After injury, Cav1(-/-) animals displayed enhanced TGF-β1 signaling, as reflected by a 3-fold increase in the activation of the Smad2-dependent pathway and more widespread collagen deposition in the heart. Qualitative and quantitative analyses indicated that collagen deposition peaked in the WT LV 14days after injury, accompanied by increased mRNA abundance for procol1a2 (2-fold) and procol3a1 (3-fold). Collagen deposition was further enhanced in Cav1(-/-) mice, which was accompanied by reduced expression of matrix metalloproteinases MMP-8 (3-fold) and -13 mRNA (2-fold). The levels of expression of inflammatory markers of acute phase were similar between the strains However, macrophage clearance in the damaged region was delayed in Cav1(-/-) mice. We observed a 4-fold decrease in collagen deposition in Cav1(-/-) mice injected with a cav1 scaffolding domain peptide (CSD) and a 2-fold decrease in WT mice treated with the CSD. We conclude that cav1 has a direct role in reducing TGF-β1 signaling and as such might be an appropriate target for therapies to influence cardiac remodeling.
Collapse
Affiliation(s)
- Shelley K Miyasato
- Dept. of Cell and Molecular Biology, University of Hawaii, John A. Burns School of Medicine, Honolulu, HI, USA.
| | | | | | | | | | | |
Collapse
|
389
|
Thenappan T, Goel A, Marsboom G, Fang YH, Toth PT, Zhang HJ, Kajimoto H, Hong Z, Paul J, Wietholt C, Pogoriler J, Piao L, Rehman J, Archer SL. A central role for CD68(+) macrophages in hepatopulmonary syndrome. Reversal by macrophage depletion. Am J Respir Crit Care Med 2011; 183:1080-91. [PMID: 21148721 PMCID: PMC3086745 DOI: 10.1164/rccm.201008-1303oc] [Citation(s) in RCA: 143] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2010] [Accepted: 11/21/2010] [Indexed: 12/16/2022] Open
Abstract
RATIONALE The etiology of hepatopulmonary syndrome (HPS), a common complication of cirrhosis, is unknown. Inflammation and macrophage accumulation occur in HPS; however, their importance is unclear. Common bile duct ligation (CBDL) creates an accepted model of HPS, allowing us to investigate the cause of HPS. OBJECTIVES We hypothesized that macrophages are central to HPS and investigated the therapeutic potential of macrophage depletion. METHODS Hemodynamics, alveolar-arterial gradient, vascular reactivity, and histology were assessed in CBDL versus sham rats (n = 21 per group). The effects of plasma on smooth muscle cell proliferation and endothelial tube formation were measured. Macrophage depletion was used to prevent (gadolinium) or regress (clodronate) HPS. CD68(+) macrophages and capillary density were measured in the lungs of patients with cirrhosis versus control patients (n = 10 per group). MEASUREMENTS AND MAIN RESULTS CBDL increased cardiac output and alveolar-arterial gradient by causing capillary dilatation and arteriovenous malformations. Activated CD68(+)macrophages (nuclear factor-κB+) accumulated in HPS pulmonary arteries, drawn by elevated levels of plasma endotoxin and lung monocyte chemoattractant protein-1. These macrophages expressed inducible nitric oxide synthase, vascular endothelial growth factor, and platelet-derived growth factor. HPS plasma increased endothelial tube formation and pulmonary artery smooth muscle cell proliferation. Macrophage depletion prevented and reversed the histological and hemodynamic features of HPS. CBDL lungs demonstrated increased medial thickness and obstruction of small pulmonary arteries. Nitric oxide synthase inhibition unmasked exaggerated pulmonary vasoconstrictor responses in HPS. Patients with cirrhosis had increased pulmonary intravascular macrophage accumulation and capillary density. CONCLUSIONS HPS results from intravascular accumulation of CD68(+)macrophages. An occult proliferative vasculopathy may explain the occasional transition to portopulmonary hypertension. Macrophage depletion may have therapeutic potential in HPS.
Collapse
MESH Headings
- Animals
- Antigens, CD/immunology
- Antigens, CD/physiology
- Antigens, Differentiation, Myelomonocytic/immunology
- Antigens, Differentiation, Myelomonocytic/physiology
- Arteriovenous Malformations/etiology
- Arteriovenous Malformations/physiopathology
- Disease Models, Animal
- Extracellular Signal-Regulated MAP Kinases/metabolism
- Hepatopulmonary Syndrome/etiology
- Hepatopulmonary Syndrome/immunology
- Humans
- Lung/blood supply
- Lung/cytology
- Lung/immunology
- Macrophages/immunology
- Macrophages/physiology
- Male
- Muscle, Smooth, Vascular/physiopathology
- Nitric Oxide Synthase Type II/antagonists & inhibitors
- Nitric Oxide Synthase Type II/physiology
- Platelet-Derived Growth Factor/antagonists & inhibitors
- Platelet-Derived Growth Factor/physiology
- Rats
- Rats, Sprague-Dawley
- Signal Transduction/physiology
- Vascular Endothelial Growth Factor A/antagonists & inhibitors
- Vascular Endothelial Growth Factor A/physiology
Collapse
Affiliation(s)
- Thenappan Thenappan
- Section of Cardiology, Department of Medicine, The University of Chicago, Chicago, Illinois; and The Cardiovascular Research Institute, Kurume University, Kurume, Fukuoka, Japan
| | - Ankush Goel
- Section of Cardiology, Department of Medicine, The University of Chicago, Chicago, Illinois; and The Cardiovascular Research Institute, Kurume University, Kurume, Fukuoka, Japan
| | - Glenn Marsboom
- Section of Cardiology, Department of Medicine, The University of Chicago, Chicago, Illinois; and The Cardiovascular Research Institute, Kurume University, Kurume, Fukuoka, Japan
| | - Yong-Hu Fang
- Section of Cardiology, Department of Medicine, The University of Chicago, Chicago, Illinois; and The Cardiovascular Research Institute, Kurume University, Kurume, Fukuoka, Japan
| | - Peter T. Toth
- Section of Cardiology, Department of Medicine, The University of Chicago, Chicago, Illinois; and The Cardiovascular Research Institute, Kurume University, Kurume, Fukuoka, Japan
| | - Hannah J. Zhang
- Section of Cardiology, Department of Medicine, The University of Chicago, Chicago, Illinois; and The Cardiovascular Research Institute, Kurume University, Kurume, Fukuoka, Japan
| | - Hidemi Kajimoto
- Section of Cardiology, Department of Medicine, The University of Chicago, Chicago, Illinois; and The Cardiovascular Research Institute, Kurume University, Kurume, Fukuoka, Japan
| | - Zhigang Hong
- Section of Cardiology, Department of Medicine, The University of Chicago, Chicago, Illinois; and The Cardiovascular Research Institute, Kurume University, Kurume, Fukuoka, Japan
| | - Jonathan Paul
- Section of Cardiology, Department of Medicine, The University of Chicago, Chicago, Illinois; and The Cardiovascular Research Institute, Kurume University, Kurume, Fukuoka, Japan
| | - Christian Wietholt
- Section of Cardiology, Department of Medicine, The University of Chicago, Chicago, Illinois; and The Cardiovascular Research Institute, Kurume University, Kurume, Fukuoka, Japan
| | - Jennifer Pogoriler
- Section of Cardiology, Department of Medicine, The University of Chicago, Chicago, Illinois; and The Cardiovascular Research Institute, Kurume University, Kurume, Fukuoka, Japan
| | - Lin Piao
- Section of Cardiology, Department of Medicine, The University of Chicago, Chicago, Illinois; and The Cardiovascular Research Institute, Kurume University, Kurume, Fukuoka, Japan
| | - Jalees Rehman
- Section of Cardiology, Department of Medicine, The University of Chicago, Chicago, Illinois; and The Cardiovascular Research Institute, Kurume University, Kurume, Fukuoka, Japan
| | - Stephen L. Archer
- Section of Cardiology, Department of Medicine, The University of Chicago, Chicago, Illinois; and The Cardiovascular Research Institute, Kurume University, Kurume, Fukuoka, Japan
| |
Collapse
|
390
|
Zamilpa R, Kanakia R, Cigarroa J, Dai Q, Escobar GP, Martinez H, Jimenez F, Ahuja SS, Lindsey ML. CC chemokine receptor 5 deletion impairs macrophage activation and induces adverse remodeling following myocardial infarction. Am J Physiol Heart Circ Physiol 2011; 300:H1418-26. [PMID: 21297029 DOI: 10.1152/ajpheart.01002.2010] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Post-myocardial infarction (MI), chemokine homing of inflammatory cells into the injured left ventricle (LV) regulates ventricular remodeling, in part by stimulating the extracellular matrix response. The CC chemokine receptor 5 (CCR5) is a key chemokine receptor expressed on macrophages, and CCR5 ligands are highly upregulated post-MI. We hypothesized that deletion of CCR5 would attenuate adverse remodeling by decreasing inflammatory cell recruitment. Accordingly, we examined LV function, macrophage recruitment and activation, and collagen content in wild-type (WT, n = 25) and CCR5 null (n = 33) mice at 7 days post-MI. Both groups had similar infarct sizes (44 ± 2% in WT and 42 ± 2% in CCR5 null; P = 0.37). However, the LV remodeling index (end diastolic volume/LV mass) increased to a larger extent in CCR5 null (1.28 ± 0.08 μl/mg for CCR5 null and 1.02 ± 0.06 μl/mg for WT; P < 0.05). Although numbers of infiltrated macrophages were similar in WT and CCR5 null mice, CCR5-deficient macrophages isolated from the infarct zone displayed >50% decrease in gene expression levels of proinflammatory activation markers (interleukin-1β, interleukin-6, and tumor necrosis factor-α), as well as anti-inflammatory activation markers (arginase 1, CD163, mannose receptor, and transforming growth factor-β1) compared with WT (all P < 0.05). Concomitant with the reduced macrophage activation, heat shock protein-47 and collagen type I precursor levels in the infarct region decreased in the CCR5 null (1.2 ± 0.3 units in the CCR5 null and 2.3 ± 0.4 units in the WT; P < 0.05), while collagen fragments increased (88.3 ± 5.9 units in the CCR5 null and 32.7 ± 8.5 units in the WT; P < 0.05). We conclude that CCR5 deletion impairs LV remodeling by hindering macrophage activation, which stimulates an imbalance in collagen metabolism and increases the remodeling index.
Collapse
Affiliation(s)
- Rogelio Zamilpa
- Division of Geriatrics, Gerontology and Palliative Medicine, Department of Medicine, The University of Texas Health Science Center at San Antonio, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
391
|
Kwon JS, Kim YS, Cho AS, Cho HH, Kim JS, Hong MH, Jeong SY, Jeong MH, Cho JG, Park JC, Kang JC, Ahn Y. The novel role of mast cells in the microenvironment of acute myocardial infarction. J Mol Cell Cardiol 2011; 50:814-25. [PMID: 21295578 DOI: 10.1016/j.yjmcc.2011.01.019] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2010] [Revised: 01/25/2011] [Accepted: 01/27/2011] [Indexed: 10/18/2022]
Abstract
Mast cells are multifunctional cells containing various mediators, such as cytokines, tryptase, and histamine, and they have been identified in infarct myocardium. Here, we elucidated the roles of mast cells in a myocardial infarction (MI) rat model. We studied the physiological and functional roles of mast cell granules (MCGs), isolated from rat peritoneal fluid, on endothelial cells, neonatal cardiomyocytes, and infarct heart (1-hour occlusion of left coronary artery followed by reperfusion). The number of mast cells had two peak time points of appearance in the infarct region at 1day and 21days after MI induction in rats (p<0.05 in each compared with sham-operated heart). Simultaneous injection of an optimal dose of MCGs modulated the microenvironment and resulted in the increased infiltration of macrophages and decreased apoptosis of cardiomyocytes without change in the mast cell number in infarct myocardium. Moreover, MCG injection attenuated the progression of MI through angiogenesis and preserved left ventricular function after MI. MCG-treated cardiomyocytes were more resistant to hypoxic injury through phosphorylation of Akt, and MCG-treated endothelial cells showed enhanced migration and tube formation. We have shown that MCGs have novel cardioprotective roles in MI via the prolonged survival of cardiomyocytes and the induction of angiogenesis.
Collapse
Affiliation(s)
- Jin Sook Kwon
- Stem Cell Research Center of Chonnam National University Hospital, Gwangju, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
392
|
Vilahur G, Juan-Babot O, Peña E, Oñate B, Casaní L, Badimon L. Molecular and cellular mechanisms involved in cardiac remodeling after acute myocardial infarction. J Mol Cell Cardiol 2011; 50:522-33. [PMID: 21219908 DOI: 10.1016/j.yjmcc.2010.12.021] [Citation(s) in RCA: 165] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2010] [Revised: 12/27/2010] [Accepted: 12/28/2010] [Indexed: 01/04/2023]
Abstract
The extent of cardiac remodeling determines survival after acute MI. However, the mechanisms driving cardiac remodeling remain unknown. We examined the effect of ischemia and reperfusion (R) on myocardial changes up to 6 days post-MI. Pigs underwent 1.5h or 4h mid-LAD balloon occlusion and sacrificed or 1.5h occlusion followed by R and sacrificed at 2.5h, 1 day, 3 days, and 6 days. Ischemic- (IM) and non-ischemic myocardium (NIM) was obtained for molecular analysis of: 1) apoptosis (P-Bcl2, Bax, P-p53, active-caspase-3); 2) the TLR-4-MyD88-dependent and independent pathways; 3) Akt/mTOR/P70(S6K) axis activation; and, 4) fibrosis (TGF-β, collagen1-A1/A3). Histopathology for inflammation, collagen, and fibroblast content, TUNEL staining, and metalloproteinase activity was performed. Apoptosis is only detected upon R in IM cardiomyocytes and progresses up to 6 days post-R mainly associated with infiltrated macrophages. The Akt/mTOR/P70(s6K) pathway is also activated upon R (IM) and remains elevated up to 6 days-R (P<0.05). Ischemia activates the TLR-4-MyD88-dependent (cytokines/chemokines) and -independent (IRF-3) pathways in IM and NIM and remains high up to 6 days post-R (P<0.05). Accordingly, leukocytes and macrophages are progressively recruited to the IM (P<0.05). Ischemia up-regulates pro-fibrotic TGF-β that gradually rises collagen1-A1/-A3 mRNA with subsequent increase in total collagen fibrils and fibroblasts from 3 days-R onwards (P<0.005). MMP-2 activity increases from ischemia to 3 days post-R (P<0.05). We report that there is a timely coordinated cellular and molecular response to myocardial ischemia and R within the first 6 days after MI. In-depth understanding of the mechanisms involved in tissue repair is warranted to timely intervene and better define novel cardioprotective strategies.
Collapse
Affiliation(s)
- Gemma Vilahur
- Cardiovascular Research Center, CSIC-ICCC, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
393
|
Evans CE, Humphries J, Mattock K, Waltham M, Wadoodi A, Saha P, Modarai B, Maxwell PJ, Smith A. Hypoxia and Upregulation of Hypoxia-Inducible Factor 1α Stimulate Venous Thrombus Recanalization. Arterioscler Thromb Vasc Biol 2010; 30:2443-51. [DOI: 10.1161/atvbaha.110.215038] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Colin Edward Evans
- From Kings College London (C.E.E., J.H., K.M., M.W., A.W., P.S., B.M., and A.S.), BHF Centre of Research Excellence & NIHR Biomedical Research Centre at Kings Health Partners, Academic Department of Surgery, London, United Kingdom; and Centre for Cell Signalling and Molecular Genetics (P.H.M.), Rayne Institute, University College London, United Kingdom
| | - Julia Humphries
- From Kings College London (C.E.E., J.H., K.M., M.W., A.W., P.S., B.M., and A.S.), BHF Centre of Research Excellence & NIHR Biomedical Research Centre at Kings Health Partners, Academic Department of Surgery, London, United Kingdom; and Centre for Cell Signalling and Molecular Genetics (P.H.M.), Rayne Institute, University College London, United Kingdom
| | - Katherine Mattock
- From Kings College London (C.E.E., J.H., K.M., M.W., A.W., P.S., B.M., and A.S.), BHF Centre of Research Excellence & NIHR Biomedical Research Centre at Kings Health Partners, Academic Department of Surgery, London, United Kingdom; and Centre for Cell Signalling and Molecular Genetics (P.H.M.), Rayne Institute, University College London, United Kingdom
| | - Matthew Waltham
- From Kings College London (C.E.E., J.H., K.M., M.W., A.W., P.S., B.M., and A.S.), BHF Centre of Research Excellence & NIHR Biomedical Research Centre at Kings Health Partners, Academic Department of Surgery, London, United Kingdom; and Centre for Cell Signalling and Molecular Genetics (P.H.M.), Rayne Institute, University College London, United Kingdom
| | - Ashar Wadoodi
- From Kings College London (C.E.E., J.H., K.M., M.W., A.W., P.S., B.M., and A.S.), BHF Centre of Research Excellence & NIHR Biomedical Research Centre at Kings Health Partners, Academic Department of Surgery, London, United Kingdom; and Centre for Cell Signalling and Molecular Genetics (P.H.M.), Rayne Institute, University College London, United Kingdom
| | - Prakash Saha
- From Kings College London (C.E.E., J.H., K.M., M.W., A.W., P.S., B.M., and A.S.), BHF Centre of Research Excellence & NIHR Biomedical Research Centre at Kings Health Partners, Academic Department of Surgery, London, United Kingdom; and Centre for Cell Signalling and Molecular Genetics (P.H.M.), Rayne Institute, University College London, United Kingdom
| | - Bijan Modarai
- From Kings College London (C.E.E., J.H., K.M., M.W., A.W., P.S., B.M., and A.S.), BHF Centre of Research Excellence & NIHR Biomedical Research Centre at Kings Health Partners, Academic Department of Surgery, London, United Kingdom; and Centre for Cell Signalling and Molecular Genetics (P.H.M.), Rayne Institute, University College London, United Kingdom
| | - Patrick J. Maxwell
- From Kings College London (C.E.E., J.H., K.M., M.W., A.W., P.S., B.M., and A.S.), BHF Centre of Research Excellence & NIHR Biomedical Research Centre at Kings Health Partners, Academic Department of Surgery, London, United Kingdom; and Centre for Cell Signalling and Molecular Genetics (P.H.M.), Rayne Institute, University College London, United Kingdom
| | - Alberto Smith
- From Kings College London (C.E.E., J.H., K.M., M.W., A.W., P.S., B.M., and A.S.), BHF Centre of Research Excellence & NIHR Biomedical Research Centre at Kings Health Partners, Academic Department of Surgery, London, United Kingdom; and Centre for Cell Signalling and Molecular Genetics (P.H.M.), Rayne Institute, University College London, United Kingdom
| |
Collapse
|
394
|
Abstract
Cardiac fibroblasts play a critical role in maintenance of normal cardiac function. They are indispensable for damage control and tissue remodeling on myocardial injury and principal mediators of pathological cardiac remodeling and fibrosis. Despite their manyfold functions, cardiac fibroblasts remain poorly characterized in molecular terms. Evidence is evolving that cardiac fibroblasts are a heterogeneous population and likely derive from various distinct tissue niches in health and disease. Here, we review our emerging understanding of where cardiac fibroblasts come from, as well as how we can possibly use this knowledge to develop novel therapies for cardiac fibrosis.
Collapse
Affiliation(s)
- Elisabeth M Zeisberg
- Division of Matrix Biology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | | |
Collapse
|
395
|
Abstract
Fibroblasts are at the heart of cardiac function and are the principal determinants of cardiac fibrosis. Nevertheless, cardiac fibroblasts remain poorly characterized in molecular terms. Evidence is evolving that the cardiac fibroblast is a highly heterogenic cell population, and that such heterogeneity is caused by the distinct origins of fibroblasts in the heart. Cardiac fibroblasts can derive either from resident fibroblasts, from endothelial cells via an endothelial-mesenchynmal transition or from bone marrow-derived circulating progenitor cells, monocytes and fibrocytes. Here, we review the function and origin of fibroblasts in cardiac fibrosis.NB. The information given is correct.
Collapse
Affiliation(s)
- Guido Krenning
- Division of Matrix Biology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Elisabeth M. Zeisberg
- Division of Matrix Biology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Raghu Kalluri
- Division of Matrix Biology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
- Harvard-MIT Division of Health Sciences and Technology, Boston, Massachusetts
- Department of Biological Chemistry and Molecular Pharmacology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
396
|
Minami E, Castellani C, Malchodi L, Deem J, Bertko K, Meznarich J, Dishmon M, Murry CE, Stempien-Otero A. The role of macrophage-derived urokinase plasminogen activator in myocardial infarct repair: urokinase attenuates ventricular remodeling. J Mol Cell Cardiol 2010; 49:516-24. [PMID: 20380835 PMCID: PMC3041515 DOI: 10.1016/j.yjmcc.2010.03.022] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2010] [Revised: 03/29/2010] [Accepted: 03/29/2010] [Indexed: 02/05/2023]
Abstract
Cardiac plasmin activity is increased following myocardial ischemia. To test the hypothesis that macrophage-derived uPA is a key mediator of repair following myocardial infarction, we performed myocardial infarction on mice with macrophage-specific over-expression of uPA (SR-uPA mice). SR-uPA(+/0) mice and wild-type littermates were sacrificed at 5 days or 4 weeks after infarction and cardiac content of macrophages, collagen, and myofibroblasts was quantified. Cardiac function and dimensions were assessed by echocardiography at baseline and at 4 weeks post-infarction. At 4 weeks after myocardial infarction, macrophage counts were increased in SR-uPA(+/0) mice in the infarct (13.1 vs. 4.9%, P<0.001) and distant uninfarcted regions (5.9 vs. 2.4%, P<0.001). Infarct scar was thicker in SR-uPA(+/0) mice (0.54+/-0.03 mm vs. 0.45+/-0.03 mm, P<0.05) and infarct cardiac collagen content was increased (72.4+/-3.3% vs. 63.0+/-3.6%, P<0.06). Functionally, these changes resulted in mildly improved fractional shortening in SR-uPA(+/0) mice compared to controls (24.6+/-1.68 vs. 19.8+/-1.3%, P=0.03). At 5 days after infarction there was increased collagen content in the scar without increases in macrophages or myofibroblasts. To understand the mechanisms by which macrophage-derived uPA increases collagen, cardiac fibroblasts were treated with macrophage-conditioned medium or plasmin and expression of ColIalpha1 measured by qPCR. Conditioned media from SR-uPA(+/0) or plasmin-treated non-transgenic macrophages but not plasmin alone increased collagen expression in isolated cardiac fibroblasts. We hypothesize that plasmin generation in the heart in response to injury may induce activation of macrophages to a profibrotic phenotype to allow rapid formation of collagenous scar.
Collapse
Affiliation(s)
- Elina Minami
- University of Washington School of Medicine, Division of Cardiology, Seattle, WA
| | - Chiara Castellani
- University of Padua, Division of Cardiovascular Pathology, Padua, Italy
| | - Laura Malchodi
- University of Washington School of Medicine, Division of Cardiology, Seattle, WA
| | - Jennifer Deem
- University of Washington, Department of Pathology, Seattle, WA
| | - Kate Bertko
- University of Washington School of Medicine, Division of Cardiology, Seattle, WA
| | - Jessica Meznarich
- University of Washington School of Medicine, Division of Cardiology, Seattle, WA
| | - Monja Dishmon
- University of Washington, Department of Pathology, Seattle, WA
| | | | - April Stempien-Otero
- University of Washington School of Medicine, Division of Cardiology, Seattle, WA
| |
Collapse
|
397
|
Zorzi P, Aplin AC, Smith KD, Nicosia RF. Technical Advance: The rat aorta contains resident mononuclear phagocytes with proliferative capacity and proangiogenic properties. J Leukoc Biol 2010; 88:1051-9. [PMID: 20628067 DOI: 10.1189/jlb.0310178] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Angiogenesis in the aortic ring model is preceded by activation of the immune system and impaired by ablation of adventitial macrophages. Treatment of aortic cultures with M-CSF induced extensive periaortic outgrowth of CD45(+) CD68(+) mononuclear cells with ultrastructural features of macrophages and DCs. Periaortic lysis of collagen caused many CD45(+) CD68(+) cells to attach to the bottom of the culture dish. Lifting the collagen gels left behind patches of CD45(+) CD68(+) cells, which focally organized into branching cords. These cells also expressed CD14, CD169, F4/80, and α-SMA but not CD31, vWF, desmin, or CD163. DNA synthesis studies showed that M-CSF-stimulated cells were actively proliferating. Aortic patch cells showed phagocytic properties and responded to IL-4 and GM-CSF by expressing MHC II, differentiating into DCs, and forming multinucleated giant cells. They also stimulated angiogenesis and VEGF production in aortic ring cultures. This study demonstrates that the rat aorta contains a distinct subset of immature immunocytes capable of proliferating, differentiating into macrophages and DCs, and stimulating angiogenesis. Isolation of these cells in patches from M-CSF-stimulated aortic rings provides a reproducible system to study the biology and angiogenic role of the resident immune system of the aortic wall.
Collapse
Affiliation(s)
- Penelope Zorzi
- VA Puget Sound Health Care System, 1660 South Columbian Way, Seattle, WA 98108, USA
| | | | | | | |
Collapse
|
398
|
Song G, Nguyen DT, Pietramaggiori G, Scherer S, Chen B, Zhan Q, Ogawa R, Yannas I, Wagers AJ, Orgill DP, Murphy GF. Use of the parabiotic model in studies of cutaneous wound healing to define the participation of circulating cells. Wound Repair Regen 2010; 18:426-32. [PMID: 20546556 PMCID: PMC2935287 DOI: 10.1111/j.1524-475x.2010.00595.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Previous experimental studies to assess the contribution of blood-borne circulating (BBC) cells to cutaneous wound healing have relied on discontinuous pulsing of labeled BBC elements or bone marrow transplant protocols. Such approaches do not allow the examination of stable BBC cells that have matured in a physiologically normal host. We have used a parabiotic murine model for cutaneous wound healing to evaluate the relative contribution of stable populations of peripheral blood cells expressing the green fluorescent protein (GFP) transgene in otherwise normal animals. Circulating cells (mature and immature) expressing the GFP transgene were easily detected and quantified in wounds of GFP- parabiotic twins during all evaluated stages of the healing response. Using multiple antibody probes, the relative contribution of various subsets of BBC cells could be comparatively assessed. In early wounds, some cells expressing mesenchymal epitopes were documented to be of hematopoietic origin, indicating the utility of this model in assessing cell plasticity in the context of tissue regeneration and repair. Application of this approach enables further investigation into the contribution of peripheral blood in normal and abnormal healing responses.
Collapse
Affiliation(s)
- Guodong Song
- Division of Plastic Surgery, Brigham & Women's Hospital, Harvard Medical School, Boston, USA
- Program in Dermatopathology, Brigham & Women's Hospital, Harvard Medical School, Boston, USA
| | - Dinh T. Nguyen
- Division of Plastic Surgery, Brigham & Women's Hospital, Harvard Medical School, Boston, USA
- Program in Dermatopathology, Brigham & Women's Hospital, Harvard Medical School, Boston, USA
| | - Giorgio Pietramaggiori
- Division of Plastic Surgery, Brigham & Women's Hospital, Harvard Medical School, Boston, USA
- Department of Pathology, Joslin Diabetes Center, Harvard Medical School, Boston, USA
| | - Saja Scherer
- Division of Plastic Surgery, Brigham & Women's Hospital, Harvard Medical School, Boston, USA
- Department of Pathology, Joslin Diabetes Center, Harvard Medical School, Boston, USA
| | - Bin Chen
- Division of Plastic Surgery, Brigham & Women's Hospital, Harvard Medical School, Boston, USA
- Program in Dermatopathology, Brigham & Women's Hospital, Harvard Medical School, Boston, USA
| | - Qian Zhan
- Program in Dermatopathology, Brigham & Women's Hospital, Harvard Medical School, Boston, USA
| | - Rei Ogawa
- Division of Plastic Surgery, Brigham & Women's Hospital, Harvard Medical School, Boston, USA
- Program in Dermatopathology, Brigham & Women's Hospital, Harvard Medical School, Boston, USA
| | - I.V. Yannas
- Department of Mechanical Engineering, Materials Science Engineering, and Biological Engineering, Massachusetts Institute of Technology, Cambridge, USA
| | - Amy J. Wagers
- Department of Pathology, Joslin Diabetes Center, Harvard Medical School, Boston, USA
| | - Dennis P. Orgill
- Division of Plastic Surgery, Brigham & Women's Hospital, Harvard Medical School, Boston, USA
| | - George F. Murphy
- Program in Dermatopathology, Brigham & Women's Hospital, Harvard Medical School, Boston, USA
| |
Collapse
|
399
|
Affiliation(s)
- Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, USA.
| | | | | |
Collapse
|
400
|
|