1
|
Takamura N, Yamaguchi Y. Involvement of caveolin-1 in skin diseases. Front Immunol 2022; 13:1035451. [PMID: 36532050 PMCID: PMC9748611 DOI: 10.3389/fimmu.2022.1035451] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 10/17/2022] [Indexed: 12/02/2022] Open
Abstract
The skin is the outermost layer and largest organ in the human body. Since the skin interfaces with the environment, it has a variety of roles, including providing a protective barrier against external factors, regulating body temperature, and retaining water in the body. It is also involved in the immune system, interacting with immune cells residing in the dermis. Caveolin-1 (CAV-1) is essential for caveolae formation and has multiple functions including endocytosis, lipid homeostasis, and signal transduction. CAV-1 is known to interact with a variety of signaling molecules and receptors and may influence cell proliferation and migration. Several skin-related disorders, especially those of the inflammatory or hyperproliferative type such as skin cancers, psoriasis, fibrosis, and wound healing, are reported to be associated with aberrant CAV-1 expression. In this review, we have explored CAV-1 involvement in skin physiology and skin diseases.
Collapse
|
2
|
Caveolin-1 scaffolding domain peptide abrogates autophagy dysregulation in pulmonary fibrosis. Sci Rep 2022; 12:11086. [PMID: 35773303 PMCID: PMC9246916 DOI: 10.1038/s41598-022-14832-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 06/13/2022] [Indexed: 11/09/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is the most common and fatal form of interstitial lung disease. IPF is characterized by irreversible scarring of the lungs leading to lung function decline. Although the etiology remains poorly understood, dysregulated autophagy in alveolar-epithelial cells (AECs) together with interplay between apoptotic-AECs and proliferative-myofibroblasts have been strongly implicated in IPF pathogenesis. Recent studies have revealed that a caveolin-1-derived 7-mer peptide, CSP7, mitigates established PF at least in part by improving AEC viability. In the present study, we aimed to determine whether and how CSP7 regulates autophagy in fibrotic-lung AECs. We found that p53 and autophagic proteins were markedly upregulated in AECs from mice with single/multi-doses of bleomycin—or silica-induced PF. This was abolished following treatment of PF-mice with CSP7. Further, CSP7 abrogated silica- or bleomycin-induced p53 and autophagy proteins in AECs. Immunoprecipitation further revealed that CSP7 abolishes the interaction of caveolin-1 with LC3BII and p62 in AECs. AEC-specific p53-knockout mice resisted silica- or bleomycin-induced changes in autophagy proteins, or CSP7 treatment. Our findings provide a novel mechanism by which CSP7 inhibits dysregulated autophagy in injured AECs and mitigates existing PF. These results affirm the potential of CSP7 for treating established PF, including IPF and silicosis.
Collapse
|
3
|
Yang H, Shi Y, Liu H, Lin F, Qiu B, Feng Q, Wang Y, Yang B. Pyroptosis executor gasdermin D plays a key role in scleroderma and bleomycin-induced skin fibrosis. Cell Death Dis 2022; 8:183. [PMID: 35396386 PMCID: PMC8993883 DOI: 10.1038/s41420-022-00970-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 03/13/2022] [Accepted: 03/23/2022] [Indexed: 11/09/2022]
Abstract
The NLRP3 inflammasome and IL-1β are essential for scleroderma pathogenesis. Nevertheless, the role of pyroptosis executor gasdermin D(GSDMD), which is a downstream molecule of NLRP3 and is required for IL-1β release in some situations, has not yet been well elucidated in scleroderma. Here, we found that GSDMD was significantly up-regulated and activated in the skin of scleroderma patients and bleomycin-induced mouse model. What’s more, the ablation of GSDMD ameliorates bleomycin-induced skin fibrosis according to HE staining, Masson staining and the detection of hydroxyproline contents. GSDMD deficiency also impaired macrophages infiltration and reduced inflammation response. Furthermore, the loss of GSDMD reduced Th17 differentiation in vivo and in vitro. Collectively, these findings provide the first demonstration that GSDMD related pyroptosis plays an important role in scleroderma pathogenesis.
Collapse
Affiliation(s)
- Huan Yang
- Dermatology Hospital, Southern Medical University, Guangzhou, 510091, China
| | - Yanqiang Shi
- Dermatology Hospital, Southern Medical University, Guangzhou, 510091, China
| | - Huiting Liu
- Dermatology Hospital, Southern Medical University, Guangzhou, 510091, China
| | - Feiyan Lin
- Dermatology Hospital, Southern Medical University, Guangzhou, 510091, China
| | - Biying Qiu
- Dermatology Hospital, Southern Medical University, Guangzhou, 510091, China
| | - Qinglan Feng
- Dermatology Hospital, Southern Medical University, Guangzhou, 510091, China
| | - Yu Wang
- Dermatology Hospital, Southern Medical University, Guangzhou, 510091, China.
| | - Bin Yang
- Dermatology Hospital, Southern Medical University, Guangzhou, 510091, China.
| |
Collapse
|
4
|
Reese CF, Chinnakkannu P, Tourkina E, Hoffman S, Kuppuswamy D. Multiple subregions within the caveolin-1 scaffolding domain inhibit fibrosis, microvascular leakage, and monocyte migration. PLoS One 2022; 17:e0264413. [PMID: 35213624 PMCID: PMC8880820 DOI: 10.1371/journal.pone.0264413] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 02/10/2022] [Indexed: 12/27/2022] Open
Abstract
The caveolin-1 scaffolding domain (CSD, amino acids 82-101 of caveolin-1) has been shown to suppress bleomycin-induced lung and skin fibrosis and angiotensin II (AngII)-induced myocardial fibrosis. To identify active subregions within CSD, we split its sequence into three slightly overlapping 8-amino acid subregions (82-89, 88-95, and 94-101). Interestingly, all three peptides showed activity. In bleomycin-treated mice, all three subregions suppressed the pathological effects on lung and skin tissue morphology. In addition, while bone marrow monocytes isolated from bleomycin-treated mice showed greatly enhanced migration in vitro toward CXCL12, treatment in vivo with CSD and its subregions almost completely suppressed this enhanced migration. In AngII-induced heart failure, both 82-89 and 88-95 significantly suppressed fibrosis (both Col I and HSP47 levels), microvascular leakage, and heart weight/ body weight ratio (HW/BW) while improving ventricular function. In contrast, while 94-101 suppressed the increase in Col I, it did not improve the other parameters. The idea that all three subregions can be active depending on the assay was further supported by experiments studying the in vitro migration of human monocytes in which all three subregions were extremely active. These studies are very novel in that it has been suggested that there is only one active region within CSD that is centered on amino acids 90-92. In contrast, we demonstrate here the presence of other active regions within CSD.
Collapse
Affiliation(s)
- Charles F. Reese
- Division of Rheumatology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, Unites States of America
| | - Panneerselvam Chinnakkannu
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, Unites States of America
| | - Elena Tourkina
- Division of Rheumatology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, Unites States of America
| | - Stanley Hoffman
- Division of Rheumatology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, Unites States of America
| | - Dhandapani Kuppuswamy
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, Unites States of America
| |
Collapse
|
5
|
Kuppuswamy D, Chinnakkannu P, Reese C, Hoffman S. The Caveolin-1 Scaffolding Domain Peptide Reverses Aging-Associated Deleterious Changes in Multiple Organs. J Pharmacol Exp Ther 2021; 378:1-9. [PMID: 33879542 DOI: 10.1124/jpet.120.000424] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 04/13/2021] [Indexed: 11/22/2022] Open
Abstract
Aging is a progressive, multifactorial, degenerative process in which deleterious changes occur in the biochemistry and function of organs. We showed that angiotensin II (AngII)-induced pathologies in the heart and kidney of young (3-month-old) mice are suppressed by the caveolin-1 scaffolding domain (CSD) peptide. Because AngII mediates many aging-associated changes, we explored whether CSD could reverse pre-existing pathologies and improve organ function in aged mice. Using 18-month-old mice (similar to 60-year-old humans), we found that >5-fold increases in leakage of serum proteins and >2-fold increases in fibrosis are associated with aging in the heart, kidney, and brain. Because tyrosine phosphorylation of cell junction proteins leads to the loss of microvascular barrier function, we analyzed the activation of the receptor tyrosine kinase PDGFR and the nonreceptor tyrosine kinases c-Src and Pyk2. We observed 5-fold activation of PDGFR and 2- to 3-fold activation of c-Src and Pyk2 in aged mice. Treatment with CSD for 4 weeks reversed these pathologic changes (microvascular leakage, fibrosis, kinase activation) in all organs almost down to the levels in healthy, young mice. In studies of heart function, CSD reduced the aging-associated increase in cardiomyocyte cross-sectional area and enhanced ventricular compliance in that echocardiographic studies demonstrated improved ejection fraction and fractional shortening and reduced isovolumic relation time. These results suggest that versions of CSD may be developed as treatments for aging-associated diseases in human patients based on the concept that CSD inhibits tyrosine kinases, leading to the inhibition of microvascular leakage and associated fibrosis, thereby improving organ function. SIGNIFICANCE STATEMENT: The caveolin-1 scaffolding domain (CSD) peptide reverses aging-associated fibrosis, microvascular leakage, and organ dysfunction in the heart, kidneys, and brain via a mechanism that involves the suppression of the activity of multiple tyrosine kinases, suggesting that CSD can be developed as a treatment for a wide range of diseases found primarily in the aged.
Collapse
Affiliation(s)
- Dhandapani Kuppuswamy
- Divisions of Cardiology (D.K., P.C.) and Rheumatology (C.R., S.H.), Department of Medicine, Medical University of South Carolina, Charleston, Charleston, South Carolina
| | - Panneerselvam Chinnakkannu
- Divisions of Cardiology (D.K., P.C.) and Rheumatology (C.R., S.H.), Department of Medicine, Medical University of South Carolina, Charleston, Charleston, South Carolina
| | - Charles Reese
- Divisions of Cardiology (D.K., P.C.) and Rheumatology (C.R., S.H.), Department of Medicine, Medical University of South Carolina, Charleston, Charleston, South Carolina
| | - Stanley Hoffman
- Divisions of Cardiology (D.K., P.C.) and Rheumatology (C.R., S.H.), Department of Medicine, Medical University of South Carolina, Charleston, Charleston, South Carolina
| |
Collapse
|
6
|
Huang M, Cai G, Baugh LM, Liu Z, Smith A, Watson M, Popovich D, Zhang T, Stawski LS, Trojanowska M, Georgakoudi I, Black LD, Pioli PA, Whitfield ML, Garlick J. Systemic Sclerosis Dermal Fibroblasts Induce Cutaneous Fibrosis Through Lysyl Oxidase-like 4: New Evidence From Three-Dimensional Skin-like Tissues. Arthritis Rheumatol 2020; 72:791-801. [PMID: 31705627 DOI: 10.1002/art.41163] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 11/05/2019] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Systemic sclerosis (SSc) is a clinically heterogeneous disease characterized by increased collagen accumulation and skin stiffness. Our previous work has demonstrated that transforming growth factor β (TGFβ) induces extracellular matrix (ECM) modifications through lysyl oxidase-like 4 (LOXL-4), a collagen crosslinking enzyme, in bioengineered human skin equivalents (HSEs) and self-assembled stromal tissues (SAS). We undertook this study to investigate cutaneous fibrosis and the role of LOXL-4 in SSc pathogenesis using HSEs and SAS. METHODS SSc-derived dermal fibroblasts (SScDFs; n = 8) and normal dermal fibroblasts (NDFs; n = 6) were incorporated into HSEs and SAS. These 3-dimensional skin-like microenvironments were used to study the effects of dysregulated LOXL-4 on ECM remodeling, fibroblast activation, and response to TGFβ stimulation. RESULTS SScDF-containing SAS showed increased stromal thickness, collagen deposition, and interleukin-6 secretion compared to NDF-containing SAS (P < 0.05). In HSE, SScDFs altered collagen as seen by a more mature and aligned fibrillar structure (P < 0.05). With SScDFs, enhanced stromal rigidity with increased collagen crosslinking (P < 0.05), up-regulation of LOXL4 expression (P < 0.01), and innate immune signaling genes were observed in both tissue models. Conversely, knockdown of LOXL4 suppressed rigidity, contraction, and α-smooth muscle actin expression in SScDFs in HSE, and TGFβ-induced ECM aggregation and collagen crosslinking in SAS. CONCLUSION A limitation to the development of effective therapeutics in SSc is the lack of in vitro human model systems that replicate human skin. Our findings demonstrate that SAS and HSE can serve as complementary in vitro skin-like models for investigation of the mechanisms and mediators that drive fibrosis in SSc and implicate a pivotal role for LOXL-4 in SSc pathogenesis.
Collapse
Affiliation(s)
- Mengqi Huang
- Tufts University School of Dental Medicine and Boston University School of Medicine, Boston, Massachusetts, and Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Guoshuai Cai
- Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, and University of South Carolina Arnold School of Public Health, Columbia
| | | | - Zhiyi Liu
- Tufts University, Medford, Massachusetts, and Zhejiang University College of Optical Science and Engineering, Hangzhou, China
| | - Avi Smith
- Tufts University School of Dental Medicine, Boston, Massachusetts
| | | | - Dillon Popovich
- Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - Tianyue Zhang
- Tufts University School of Dental Medicine, Boston, Massachusetts
| | | | | | | | - Lauren D Black
- Tufts University School of Medicine Sackler School for Graduate Biomedical Sciences, Boston, Massachusetts
| | | | | | - Jonathan Garlick
- Tufts University School of Dental Medicine, Boston, Massachusetts
| |
Collapse
|
7
|
Egger AN, Rajabi‐Estarabadi A, Williams NM, Resnik SR, Fox JD, Wong LL, Jozic I. The importance of caveolins and caveolae to dermatology: Lessons from the caves and beyond. Exp Dermatol 2020; 29:136-148. [PMID: 31845391 PMCID: PMC7028117 DOI: 10.1111/exd.14068] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 11/25/2019] [Accepted: 11/28/2019] [Indexed: 12/15/2022]
Abstract
Caveolae are flask-shaped invaginations of the cell membrane rich in cholesterol and sphingomyelin, with caveolin proteins acting as their primary structural components that allow compartmentalization and orchestration of various signalling molecules. In this review, we discuss how pleiotropic functions of caveolin-1 (Cav1) and its intricate roles in numerous cellular functions including lipid trafficking, signalling, cell migration and proliferation, as well as cellular senescence, infection and inflammation, are integral for normal development and functioning of skin and its appendages. We then examine how disruption of the homeostatic levels of Cav1 can lead to development of various cutaneous pathophysiologies including skin cancers, cutaneous fibroses, psoriasis, alopecia, age-related changes in skin and aberrant wound healing and propose how levels of Cav1 may have theragnostic value in skin physiology/pathophysiology.
Collapse
Affiliation(s)
- Andjela N. Egger
- Wound Healing and Regenerative Medicine Research ProgramDr. Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFLUSA
| | - Ali Rajabi‐Estarabadi
- Wound Healing and Regenerative Medicine Research ProgramDr. Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFLUSA
| | - Natalie M. Williams
- Wound Healing and Regenerative Medicine Research ProgramDr. Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFLUSA
| | - Sydney R. Resnik
- Wound Healing and Regenerative Medicine Research ProgramDr. Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFLUSA
| | - Joshua D. Fox
- Wound Healing and Regenerative Medicine Research ProgramDr. Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFLUSA
| | - Lulu L. Wong
- Wound Healing and Regenerative Medicine Research ProgramDr. Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFLUSA
| | - Ivan Jozic
- Wound Healing and Regenerative Medicine Research ProgramDr. Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFLUSA
| |
Collapse
|
8
|
Lee R, Del Papa N, Introna M, Reese CF, Zemskova M, Bonner M, Carmen-Lopez G, Helke K, Hoffman S, Tourkina E. Adipose-derived mesenchymal stromal/stem cells in systemic sclerosis: Alterations in function and beneficial effect on lung fibrosis are regulated by caveolin-1. JOURNAL OF SCLERODERMA AND RELATED DISORDERS 2019; 4:127-136. [PMID: 35382388 PMCID: PMC8922642 DOI: 10.1177/2397198318821510] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 12/02/2018] [Indexed: 08/10/2023]
Abstract
The potential value of mesenchymal stromal/stem cell therapy in treating skin fibrosis in scleroderma (systemic sclerosis) and of the caveolin-1 scaffolding domain peptide in treating lung, skin, and heart fibrosis is known. To understand how these observations may relate to differences between mesenchymal stromal/stem cells from healthy subjects and subjects with fibrosis, we have characterized the fibrogenic and adipogenic potential of adipose-derived mesenchymal stromal/stem cells from systemic sclerosis patients, from mice with fibrotic lung and skin disease induced by systemic bleomycin treatment, and from healthy controls. Early passage systemic sclerosis adipose-derived mesenchymal stromal/stem cells have a profibrotic/anti-adipogenic phenotype compared to healthy adipose-derived mesenchymal stromal/stem cells (low caveolin-1, high α-smooth muscle actin, high HSP47, low pAKT, low capacity for adipogenic differentiation). This phenotype is mimicked by treating healthy adipose-derived mesenchymal stromal/stem cells with transforming growth factor beta or caveolin-1 small interfering RNA and is reversed in systemic sclerosis adipose-derived mesenchymal stromal/stem cells by treatment with caveolin-1 scaffolding domain peptide, but not scrambled caveolin-1 scaffolding domain peptide. Similar results were obtained with adipose-derived mesenchymal stromal/stem cells from systemic sclerosis patients and from bleomycin-treated mice, indicating the central role of caveolin-1 in mesenchymal stromal/stem cell differentiation in fibrotic disease.
Collapse
Affiliation(s)
- Rebecca Lee
- Division of Rheumatology & Immunology, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Nicoletta Del Papa
- U.O.C. Day Hospital di Reumatologia, Dipartimento di Reumatologia e Scienze Mediche, ASST Gaetano Pini-CTO, Milano, Italy
| | - Martin Introna
- USS Centro di Terapia Cellulare “G. Lanzani,” USC Ematologia, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Charles F Reese
- Division of Rheumatology & Immunology, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Marina Zemskova
- Division of Rheumatology & Immunology, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Michael Bonner
- Division of Rheumatology & Immunology, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Gustavo Carmen-Lopez
- Division of Rheumatology & Immunology, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Kristi Helke
- Department of Comparative Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Stanley Hoffman
- Division of Rheumatology & Immunology, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Elena Tourkina
- Division of Rheumatology & Immunology, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
9
|
Lysyl oxidase enzymes mediate TGF-β1-induced fibrotic phenotypes in human skin-like tissues. J Transl Med 2019; 99:514-527. [PMID: 30568176 DOI: 10.1038/s41374-018-0159-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 10/17/2018] [Accepted: 10/29/2018] [Indexed: 01/09/2023] Open
Abstract
Cutaneous fibrosis is a common complication seen in mixed connective tissue diseases. It often occurs as a result of TGF-β-induced deposition of excessive amounts of collagen in the skin. Lysyl oxidases (LOXs), a family of extracellular matrix (ECM)-modifying enzymes responsible for collagen cross-linking, are known to be increased in dermal fibroblasts from patients with fibrotic diseases, denoting a possible role of LOXs in fibrosis. To directly study this, we have developed two bioengineered, in vitro skin-like models: human skin equivalents (hSEs), and self-assembled stromal tissues (SASs) that contain either normal or systemic sclerosis (SSc; scleroderma) patient-derived fibroblasts. These tissues provide an organ-level structure that could be combined with non-invasive, label-free, multiphoton microscopy (SHG/TPEF) to reveal alterations in the organization and cross-linking levels of collagen fibers during the development of cutaneous fibrosis, which demonstrated increased stromal rigidity and activation of dermal fibroblasts in response to TGF-β1. Specifically, inhibition of specific LOXs isoforms, LOX and LOXL4, in foreskin fibroblasts (HFFs) resulted in antagonistic effects on TGF-β1-induced fibrogenic hallmarks in both hSEs and SASs. In addition, a translational relevance of these models was seen as similar antifibrogenic phenotypes were achieved upon knocking down LOXL4 in tissues containing SSc patient-derived-dermal fibroblasts (SScDFs). These findings point to a pivotal role of LOXs in TGF-β1-induced cutaneous fibrosis through impaired ECM homeostasis in skin-like tissues, and show the value of these tissue platforms in accelerating the discovery of antifibrosis therapeutics.
Collapse
|
10
|
Chinnakkannu P, Reese C, Gaspar JA, Panneerselvam S, Pleasant-Jenkins D, Mukherjee R, Baicu C, Tourkina E, Hoffman S, Kuppuswamy D. Suppression of angiotensin II-induced pathological changes in heart and kidney by the caveolin-1 scaffolding domain peptide. PLoS One 2018; 13:e0207844. [PMID: 30576317 PMCID: PMC6303044 DOI: 10.1371/journal.pone.0207844] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 11/07/2018] [Indexed: 01/15/2023] Open
Abstract
Dysregulation of the renin-angiotensin system leads to systemic hypertension and maladaptive fibrosis in various organs. We showed recently that myocardial fibrosis and the loss of cardiac function in mice with transverse aortic constriction (TAC) could be averted by treatment with the caveolin-1 scaffolding domain (CSD) peptide. Here, we used angiotensin II (AngII) infusion (2.1 mg/kg/day for 2 wk) in mice as a second model to confirm and extend our observations on the beneficial effects of CSD on heart and kidney disease. AngII caused cardiac hypertrophy (increased heart weight to body weight ratio (HW/BW) and cardiomyocyte cross-sectional area); fibrosis in heart and kidney (increased levels of collagen I and heat shock protein-47 (HSP47)); and vascular leakage (increased levels of IgG in heart and kidney). Echocardiograms of AngII-infused mice showed increased left ventricular posterior wall thickness (pWTh) and isovolumic relaxation time (IVRT), and decreased ejection fraction (EF), stroke volume (SV), and cardiac output (CO). CSD treatment (i.p. injections, 50 μg/mouse/day) of AngII-infused mice significantly suppressed all of these pathological changes in fibrosis, hypertrophy, vascular leakage, and ventricular function. AngII infusion increased β1 and β3 integrin levels and activated Pyk2 in both heart and kidney. These changes were also suppressed by CSD. Finally, bone marrow cell (BMC) isolated from AngII-infused mice showed hyper-migration toward SDF1. When AngII-infused mice were treated with CSD, BMC migration was reduced to the basal level observed in cells from control mice. Importantly, CSD did not affect the AngII-induced increase in blood pressure (BP), indicating that the beneficial effects of CSD were not mediated via normalization of BP. These results strongly indicate that CSD suppresses AngII-induced pathological changes in mice, suggesting that CSD can be developed as a treatment for patients with hypertension and pressure overload-induced heart failure.
Collapse
Affiliation(s)
- Panneerselvam Chinnakkannu
- Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Charles Reese
- Division of Rheumatology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | | | - Saraswathi Panneerselvam
- Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Dorea Pleasant-Jenkins
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Rupak Mukherjee
- Division of Cardiothoracic Surgery, Department of Surgery, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Catalin Baicu
- Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Elena Tourkina
- Division of Rheumatology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Stanley Hoffman
- Division of Rheumatology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Dhandapani Kuppuswamy
- Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute, Medical University of South Carolina, Charleston, South Carolina, United States of America
| |
Collapse
|
11
|
Jin LG, Zeng S, Sun XQ, Wu C, Chen JL, Cui M, Pang QF. Deletion 101 residue at caveolin-1 scaffolding domain peptides impairs the ability of increasing heme oxygenase-1 activity. Int Immunopharmacol 2018; 63:137-144. [PMID: 30092496 DOI: 10.1016/j.intimp.2018.07.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 07/02/2018] [Accepted: 07/23/2018] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Resident alveolar macrophages (AMs) are activated and release proinflammatory mediators and chemokines during acute lung injury. We have previous reported that caveolin-1 (Cav-1) scaffolding domain (CSD) peptide inhibited the proinflammatory cytokines expression by up-regulating heme oxygenase-1 (HO-1) activity. In this study, we aimed to investigate the effect of residue R101 in CSD peptide on the activity of HO-1 in AMs. METHODS The binding mode between HO-1 and CSD peptides (WT CSD and Δ101 CSD truncation peptides) was analyzed and the free energy was calculated. The inflammatory genes and M1/M2macrophage polarization-associated genes expression were measured by real-time PCR. The activities of HO-1 were determined by the spectrophotometical method. Western blot analyzed the content of Cav-1, HO-1, IκB and MAPK signals (phosphorylated ERK, JNK and p38 MAPK). RESULTS Δ101CSD peptide could bind to HO-1 protein and to disrupt the interaction of HO-1 and Cav-1. However, Δ101CSD peptide had lower activity of HO-1 in LPS-treated AMs compared with WT CSD. The expression of IL-1β and MCP-1 and NO content were decreased by WT CSD peptide in LPS treated AMs. However, only MCP-1 expression and NO content were downregulated byΔ101CSD peptide. Meanwhile, compared with those in LPS + hemin + WT CSD group, the mRNA expression of TNF-α, Cd86, IL-12b and NOS2 significantly increased while expression of IL10, Arg1 and CD163 significantly decreased in LPS + hemin + Δ101CSD group. The effect of WT CSD peptide on the inhibition of MAPK signaling pathway were stronger than Δ101 CSD peptide evidenced by the level of phosphorylated ERK, JNK and p38 MAPK. CONCLUSION Deletion of residue R101 impairs the ability of CSD peptide to increase HO-1 activity and to dampen inflammatory response in LPS-challenged rat AMs.
Collapse
Affiliation(s)
- Liu-Gen Jin
- Wuxi Medical School, Jiangnan University, Wuxi, Jiangsu 214122, China; The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, China
| | - Si Zeng
- Department of Anesthesiology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, China
| | - Xue-Qian Sun
- Wuxi Medical School, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Chen Wu
- Wuxi Medical School, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Jun-Liang Chen
- Wuxi Medical School, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Meng Cui
- Wuxi Medical School, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Qing-Feng Pang
- Wuxi Medical School, Jiangnan University, Wuxi, Jiangsu 214122, China.
| |
Collapse
|
12
|
Marangoni RG, Masui Y, Fang F, Korman B, Lord G, Lee J, Lakota K, Wei J, Scherer PE, Otvos L, Yamauchi T, Kubota N, Kadowaki T, Asano Y, Sato S, Tourtellotte WG, Varga J. Adiponectin is an endogenous anti-fibrotic mediator and therapeutic target. Sci Rep 2017; 7:4397. [PMID: 28667272 PMCID: PMC5493638 DOI: 10.1038/s41598-017-04162-1] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 05/22/2017] [Indexed: 12/21/2022] Open
Abstract
Skin fibrosis in systemic sclerosis (SSc) is accompanied by attrition of dermal white adipose tissue (dWAT) and reduced levels of circulating adiponectin. Since adiponectin has potent regulatory effects on fibroblasts, we sought to assess adiponectin signaling in SSc skin biopsies, and evaluate fibrosis in mice with adiponectin gain- and loss-of-function mutations. Furthermore, we investigated the effects and mechanism of action of agonist peptides targeting adiponectin receptors in vitro and in vivo. We found that adiponectin pathway activity was significantly reduced in a subset of SSc skin biopsies. Mice lacking adiponectin mounted an exaggerated dermal fibrotic response, while transgenic mice with constitutively elevated adiponectin showed selective dWAT expansion and protection from skin and peritoneal fibrosis. Adiponectin receptor agonists abrogated ex vivo fibrotic responses in explanted normal and SSc fibroblasts and in 3D human skin equivalents, in part by attenuating focal adhesion complex assembly, and prevented and reversed experimentally-induced organ fibrosis in mice. These results implicate aberrant adiponectin pathway activity in skin fibrosis, identifying a novel function for this pleiotropic adipokine in regulation of tissue remodeling. Restoring adiponectin signaling in SSc patients therefore might represent an innovative pharmacological strategy for intractable organ fibrosis.
Collapse
Affiliation(s)
- Roberta G Marangoni
- Northwestern Scleroderma Program, Feinberg School of Medicine, Chicago, IL, 60611, USA.
| | - Yuri Masui
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Feng Fang
- Northwestern Scleroderma Program, Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Benjamin Korman
- Northwestern Scleroderma Program, Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Gabriel Lord
- Northwestern Scleroderma Program, Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Junghwa Lee
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Katja Lakota
- Department of Rheumatology, University Medical Centre Ljubljana, 1000, Ljubljana, Slovenia
| | - Jun Wei
- Northwestern Scleroderma Program, Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Philipp E Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Laszlo Otvos
- Department of Biology, Temple University, Philadelphia, PA, 19140, USA
| | - Toshimasa Yamauchi
- Department of Diabetes and Metabolic Diseases, University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Naoto Kubota
- Department of Diabetes and Metabolic Diseases, University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Takashi Kadowaki
- Department of Diabetes and Metabolic Diseases, University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Yoshihide Asano
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Shinichi Sato
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Warren G Tourtellotte
- Department of Pathology and Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - John Varga
- Northwestern Scleroderma Program, Feinberg School of Medicine, Chicago, IL, 60611, USA.
| |
Collapse
|
13
|
Optimization of a murine and human tissue model to recapitulate dermal and pulmonary features of systemic sclerosis. PLoS One 2017. [PMID: 28651005 PMCID: PMC5484495 DOI: 10.1371/journal.pone.0179917] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The murine bleomycin (BLM)-induced fibrosis model is the most widely used in systemic sclerosis (SSc) studies. It has been reported that systemic delivery of BLM via continuous diffusion from subcutaneously implanted osmotic minipumps can cause fibrosis of the skin, lungs, and other internal organs. However, the mouse strain, dosage of BLM, administration period, and additional important features differ from one report to the next. In this study, by employing the pump model in C57BL/6J mice, we show a dose-dependent increase in lung fibrosis by day 28 and a transient increase in dermal thickness. Dermal thickness and the level of collagen in skin treated with high-dose BLM was significantly higher than in skin treated with low dose BLM or vehicle. A reduction in the thickness of the adipose layer was noted in both high and low dose groups at earlier time points suggesting that the loss of the fat layer precedes the onset of fibrosis. High-dose BLM also induced dermal fibrosis and increased expression of fibrosis-associated genes ex vivo in human skin, thus confirming and extending the in vivo findings, and demonstrating that a human organ culture model can be used to assess the effect of BLM on skin. In summary, our findings suggest that the BLM pump model is an attractive model to analyze the underlying mechanisms of fibrosis and test the efficacy of potential therapies. However, the choice of mouse strain, duration of BLM administration and dose must be carefully considered when using this model.
Collapse
|
14
|
Lee R, Reese C, Carmen-Lopez G, Perry B, Bonner M, Zemskova M, Wilson CL, Helke KL, Silver RM, Hoffman S, Tourkina E. Deficient Adipogenesis of Scleroderma Patient and Healthy African American Monocytes. Front Pharmacol 2017; 8:174. [PMID: 28420992 PMCID: PMC5376579 DOI: 10.3389/fphar.2017.00174] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 03/15/2017] [Indexed: 12/01/2022] Open
Abstract
Monocytes from systemic sclerosis (SSc, scleroderma) patients and healthy African Americans (AA) are deficient in the regulatory protein caveolin-1 leading to enhanced migration toward chemokines and fibrogenic differentiation. While dermal fibrosis is the hallmark of SSc, loss of subcutaneous adipose tissue is a lesser-known feature. To better understand the etiology of SSc and the predisposition of AA to SSc, we studied the adipogenic potential of SSc and healthy AA monocytes. The ability of SSc and healthy AA monocytes to differentiate into adipocyte-like cells (ALC) is inhibited compared to healthy Caucasian (C) monocytes. We validated that monocyte-derived ALCs are distinct from macrophages by flow cytometry and immunocytochemistry. Like their enhanced fibrogenic differentiation, their inhibited adipogenic differentiation is reversed by the caveolin-1 scaffolding domain peptide (CSD, a surrogate for caveolin-1). The altered differentiation of SSc and healthy AA monocytes is additionally regulated by peroxisome proliferator-activated receptor γ (PPARγ) which is also present at reduced levels in these cells. In vivo studies further support the importance of caveolin-1 and PPARγ in fibrogenesis and adipogenesis. In SSc patients, healthy AA, and mice treated systemically with bleomycin, adipocytes lose caveolin-1 and PPARγ and the subcutaneous adipose layer is diminished. CSD treatment of these mice leads to a reappearance of the caveolin-1+/PPARγ+/FABP4+ subcutaneous adipose layer. Moreover, many of these adipocytes are CD45+, suggesting they are monocyte derived. Tracing experiments with injected EGFP+ monocytes confirm that monocytes contribute to the repair of the adipose layer when it is damaged by bleomycin treatment. Our observations strongly suggest that caveolin-1 and PPARγ work together to maintain a balance between the fibrogenic and adipogenic differentiation of monocytes, that this balance is altered in SSc and in healthy AA, and that monocytes make a major contribution to the repair of the adipose layer.
Collapse
Affiliation(s)
- Rebecca Lee
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South CarolinaCharleston, SC, USA
| | - Charles Reese
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South CarolinaCharleston, SC, USA
| | - Gustavo Carmen-Lopez
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South CarolinaCharleston, SC, USA
| | - Beth Perry
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South CarolinaCharleston, SC, USA
| | - Michael Bonner
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South CarolinaCharleston, SC, USA
| | - Marina Zemskova
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South CarolinaCharleston, SC, USA
| | - Carole L Wilson
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Medical University of South CarolinaCharleston, SC, USA
| | - Kristi L Helke
- Department of Comparative Medicine, Medical University of South CarolinaCharleston, SC, USA
| | - Richard M Silver
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South CarolinaCharleston, SC, USA
| | - Stanley Hoffman
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South CarolinaCharleston, SC, USA.,Department of Regenerative Medicine and Cell Biology, Medical University of South CarolinaCharleston, SC, USA
| | - Elena Tourkina
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South CarolinaCharleston, SC, USA.,Department of Regenerative Medicine and Cell Biology, Medical University of South CarolinaCharleston, SC, USA
| |
Collapse
|
15
|
Pleasant-Jenkins D, Reese C, Chinnakkannu P, Kasiganesan H, Tourkina E, Hoffman S, Kuppuswamy D. Reversal of maladaptive fibrosis and compromised ventricular function in the pressure overloaded heart by a caveolin-1 surrogate peptide. J Transl Med 2017; 97:370-382. [PMID: 28112757 PMCID: PMC5909408 DOI: 10.1038/labinvest.2016.153] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 11/21/2016] [Accepted: 12/01/2016] [Indexed: 12/19/2022] Open
Abstract
Chronic ventricular pressure overload (PO) results in congestive heart failure (CHF) in which myocardial fibrosis develops in concert with ventricular dysfunction. Caveolin-1 is important in fibrosis in various tissues due to its decreased expression in fibroblasts and monocytes. The profibrotic effects of low caveolin-1 can be blocked with the caveolin-1 scaffolding domain peptide (CSD, a caveolin-1 surrogate) using both mouse models and human cells. We have studied the beneficial effects of CSD on mice in which PO was induced by trans-aortic constriction (TAC). Beneficial effects observed in TAC mice receiving CSD injections daily included: improved ventricular function (increased ejection fraction, stroke volume, and cardiac output; reduced wall thickness); decreased collagen I, collagen chaperone HSP47, fibronectin, and CTGF levels; decreased activation of non-receptor tyrosine kinases Pyk2 and Src; and decreased activation of eNOS. To determine the source of cells that contribute to fibrosis in CHF, flow cytometric studies were performed that suggested that myofibroblasts in the heart are in large part bone marrow-derived. Two CD45+ cell populations were observed. One (Zone 1) contained CD45+/HSP47-/macrophage marker+ cells (macrophages). The second (Zone 2) contained CD45moderate/HSP47+/macrophage marker- cells often defined as fibrocytes. TAC increased the number of cells in Zones 1 and 2 and the level of HSP47 in Zone 2. These studies are a first step in elucidating the mechanism of action of CSD in heart fibrosis and promoting the development of CSD as a novel treatment to reduce fibrosis and improve ventricular function in CHF patients.
Collapse
Affiliation(s)
- Dorea Pleasant-Jenkins
- Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute, Charleston, SC, USA
| | - Charles Reese
- Division of Rheumatology, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | | | - Harinath Kasiganesan
- Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute, Charleston, SC, USA
| | - Elena Tourkina
- Division of Rheumatology, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Stanley Hoffman
- Division of Rheumatology, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Dhandapani Kuppuswamy
- Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute, Charleston, SC, USA
| |
Collapse
|
16
|
Qin Z, Wan JJ, Sun Y, Wang PY, Su DF, Lei H, Liu X. ORM Promotes Skeletal Muscle Glycogen Accumulation via CCR5-Activated AMPK Pathway in Mice. Front Pharmacol 2016; 7:302. [PMID: 27679573 PMCID: PMC5020064 DOI: 10.3389/fphar.2016.00302] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 08/26/2016] [Indexed: 01/08/2023] Open
Abstract
We found previously that acute phase protein orosomucoid reacts to fatigue and activates C-C chemokine receptor type 5 to increase muscle glycogen storage and enhance muscle endurance (Lei et al., 2016). To explore the underlying molecular mechanisms, we investigated the role of AMP-activated protein kinase, a critical fuel sensor in skeletal muscle, in C-C chemokine receptor type 5-mediated orosomucoid action. It was found orosomucoid increased skeletal muscle AMP-activated protein kinase activation in a time- and dose- dependent manner, which was largely prevented by pharmacological blocking or knockout of C-C chemokine receptor type 5. Administration of orosomucoid also significantly increased the de-phosphorylation and activity of muscle glycogen synthase, the rate-limiting enzyme for glycogen synthesis. The effect was largely absent in mice deficient in C-C chemokine receptor type 5−/− or AMP-activated protein kinase α2−/−, the predominant isoform in skeletal muscle. Moreover, deletion of AMP-activated protein kinase α2 abolished the effect of orosomucoid on fatigue and muscle glycogen. These findings indicate that orosomucoid may promote glycogen storage and enhance muscle function through C-C chemokine receptor type 5-mdiated activation of AMP-activated protein kinase, which in turn activates glycogen synthase and increases muscle glycogen.
Collapse
Affiliation(s)
- Zhen Qin
- Department of Pharmacology, School of Pharmacy, Second Military Medical University Shanghai, China
| | - Jing-Jing Wan
- Department of Pharmacology, School of Pharmacy, Second Military Medical University Shanghai, China
| | - Yang Sun
- Department of Pharmacology, School of Pharmacy, Second Military Medical University Shanghai, China
| | - Peng-Yuan Wang
- Department of Pharmacology, School of Pharmacy, Second Military Medical University Shanghai, China
| | - Ding-Feng Su
- Department of Pharmacology, School of Pharmacy, Second Military Medical University Shanghai, China
| | - Hong Lei
- Department of Pharmacology, School of Pharmacy, Second Military Medical University Shanghai, China
| | - Xia Liu
- Department of Pharmacology, School of Pharmacy, Second Military Medical University Shanghai, China
| |
Collapse
|
17
|
Rog-Zielinska EA, Norris RA, Kohl P, Markwald R. The Living Scar – Cardiac Fibroblasts and the Injured Heart. Trends Mol Med 2016. [DOI: 10.1016/j.molmed.2015.12.006 order by 1-- -] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
18
|
Rog-Zielinska EA, Norris RA, Kohl P, Markwald R. The Living Scar – Cardiac Fibroblasts and the Injured Heart. Trends Mol Med 2016. [DOI: 10.1016/j.molmed.2015.12.006 and 1880=1880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
19
|
Rog-Zielinska EA, Norris RA, Kohl P, Markwald R. The Living Scar – Cardiac Fibroblasts and the Injured Heart. Trends Mol Med 2016. [DOI: 10.1016/j.molmed.2015.12.006 order by 8029-- -] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
20
|
Rog-Zielinska EA, Norris RA, Kohl P, Markwald R. The Living Scar – Cardiac Fibroblasts and the Injured Heart. Trends Mol Med 2016. [DOI: 10.1016/j.molmed.2015.12.006 order by 8029-- awyx] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
21
|
Rog-Zielinska EA, Norris RA, Kohl P, Markwald R. The Living Scar – Cardiac Fibroblasts and the Injured Heart. Trends Mol Med 2016. [DOI: 10.1016/j.molmed.2015.12.006 order by 1-- gadu] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
22
|
Rog-Zielinska EA, Norris RA, Kohl P, Markwald R. The Living Scar – Cardiac Fibroblasts and the Injured Heart. Trends Mol Med 2016. [DOI: 10.1016/j.molmed.2015.12.006 order by 1-- #] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
23
|
Rog-Zielinska EA, Norris RA, Kohl P, Markwald R. The Living Scar – Cardiac Fibroblasts and the Injured Heart. Trends Mol Med 2016. [DOI: 10.1016/j.molmed.2015.12.006 order by 8029-- #] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
24
|
Rog-Zielinska EA, Norris RA, Kohl P, Markwald R. The Living Scar--Cardiac Fibroblasts and the Injured Heart. Trends Mol Med 2016; 22:99-114. [PMID: 26776094 DOI: 10.1016/j.molmed.2015.12.006] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 12/16/2015] [Accepted: 12/18/2015] [Indexed: 12/22/2022]
Abstract
Cardiac scars, often dubbed 'dead tissue', are very much alive, with heterocellular activity contributing to the maintenance of structural and mechanical integrity following heart injury. To form a scar, non-myocytes such as fibroblasts are recruited from intra- and extra-cardiac sources. Fibroblasts perform important autocrine and paracrine signaling functions. They also establish mechanical and, as is increasingly evident, electrical junctions with other cells. While fibroblasts were previously thought to act simply as electrical insulators, they may be electrically connected among themselves and, under some circumstances, to other cells including cardiomyocytes. A better understanding of these biophysical interactions will help to target scar structure and function, and will facilitate the development of novel therapies aimed at modifying scar properties for patient benefit.
Collapse
Affiliation(s)
- Eva A Rog-Zielinska
- Institute for Experimental Cardiovascular Medicine, University of Freiburg, Freiburg, Germany; National Heart and Lung Institute, Imperial College London, London, UK
| | - Russell A Norris
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Peter Kohl
- Institute for Experimental Cardiovascular Medicine, University of Freiburg, Freiburg, Germany; National Heart and Lung Institute, Imperial College London, London, UK.
| | - Roger Markwald
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
25
|
Enhanced chemokine-receptor expression, function, and signaling in healthy African American and scleroderma-patient monocytes are regulated by caveolin-1. FIBROGENESIS & TISSUE REPAIR 2015; 8:11. [PMID: 26322128 PMCID: PMC4551709 DOI: 10.1186/s13069-015-0028-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 06/08/2015] [Indexed: 01/13/2023]
Abstract
Background A major health disparity suffered by African Americans (AA) is a predisposition toward fibrotic diseases of the skin, lung, and other organs. We previously showed that healthy AA and scleroderma (systemic sclerosis (SSc)) patient monocytes share biochemical and functional differences from control Caucasian (C) monocytes that may predispose AA to SSc. The central difference is a decrease in caveolin-1. Low caveolin-1 levels promote monocyte migration, their differentiation into fibrocytes, and fibrocyte recruitment into fibrotic tissues. Here we have greatly expanded our studies on the mechanism of action in fibrosis of caveolin-1 in AA and SSc monocytes. Results Expression of chemokine receptors (CCR1, CCR2, CCR3) is enhanced in healthy AA monocytes compared to healthy C monocytes and further increased in SSc monocytes. A parallel increase in function occurs assessed by migration toward chemokines MCP-1 and MCP-3. Chemokine-receptor expression and function are inhibited by the caveolin-1 scaffolding domain peptide (CSD) via its action as a surrogate for caveolin-1. Cells bearing chemokine receptors accumulate to high levels in fibrotic lung and skin tissue from SSc patients and from mice treated with bleomycin. This accumulation is almost completely blocked in mice treated with CSD. In signaling studies, Src activation is enhanced in AA monocytes compared to C monocytes and further increased in SSc monocytes. Lyn is also highly activated in SSc monocytes. Src and Lyn activation are inhibited by CSD. Src and Lyn’s roles in monocyte migration were demonstrated using specific inhibitors. Conclusions To the best of our knowledge, this is the first report that the expression and function of CCR1, CCR2, and CCR3 are upregulated in monocytes from healthy AA and from SSc patients via molecular mechanisms involving caveolin-1, Src/Lyn, and MEK/ERK. The results suggest that the migration/recruitment of monocytes and fibrocytes into fibrotic tissues, mediated at least in part by CCR1, CCR2, and CCR3, plays a major role in the progression of lung and skin fibrosis and in the predisposition of AA to fibrotic diseases. Our findings further suggest that chemokine receptors and signaling molecules, particularly caveolin-1, that control their expression/function are promising targets for treating fibrotic diseases. Electronic supplementary material The online version of this article (doi:10.1186/s13069-015-0028-7) contains supplementary material, which is available to authorized users.
Collapse
|
26
|
Sanon VP, Sawaki D, Mjaatvedt CH, Jourdan‐Le Saux C. Myocardial Tissue Caveolae. Compr Physiol 2015; 5:871-86. [DOI: 10.1002/cphy.c140050] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
27
|
Qi XJ, Ning W, Xu F, Dang HX, Fang F, Li J. Fasudil, an inhibitor of Rho-associated coiled-coil kinase, attenuates hyperoxia-induced pulmonary fibrosis in neonatal rats. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:12140-50. [PMID: 26722398 PMCID: PMC4680343 DOI: pmid/26722398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 09/24/2015] [Indexed: 11/18/2022]
Abstract
BACKGROUND Oxygen therapy is important during the management of high-risk neonatal infants, such as those with preterm birth, low birth weight, and asphyxia. However, prolonged exposure to high oxygen concentrations can readily lead to diffuse nonspecific inflammation, which promotes airway remodeling and pulmonary fibrosis. The Rho/Rho-associated coiled-coil kinase (Rho/ROCK) signaling pathway plays an important role in numerous developmental and proliferative diseases. This study was performed to determine the efficacy of ROCK inhibitor fasudil in blocking the development of hyperoxia-induced lung injury and fibrosis in neonatal rats. METHODS Neonatal rats were randomly divided into four groups: air + saline group, air + fasudil group, hyperoxia + saline group, and hyperoxia + fasudil group. The hyperoxia + saline and Hyp + fasudil groups were exposed to 95% oxygen for 21 days and administered intraperitoneal saline or fasudil once daily. The air + saline and air + fasudil group were exposed to 21% oxygen (room air) and administered the same volume of intraperitoneal saline or fasudil. RESULTS Fasudil-treated rats exhibited improved histopathological changes and decreased lung hydroxyproline content. Fasudil attenuated the protein level of alpha-smooth muscle actin, transforming growth factor-β1, and connective tissue growth factor. Additionally, fasudil reduced the activation of ROCK1 and myosin phosphatase targeting subunit 1 protein in the Rho/ROCK signaling pathway. CONCLUSIONS Fasudil may be a potentially effective therapeutic drug for hyperoxia-induced pulmonary fibrosis.
Collapse
Affiliation(s)
- Xiu-Jie Qi
- Ministry of Education Key Laboratory of Child Development and Disorders, Key Laboratory of Pediatrics in Chongqing, Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Children's Hospital of Chongqing Medical University Chongqing 400014, China ; Department of Pediatric Intensive Care Unit, Children's Hospital of Chongqing Medical University Chongqing 400014, China
| | - Wei Ning
- Daping Hospital and The Research Institute of Surgery of The Third Military Medical University Chongqing 400042, China
| | - Feng Xu
- Department of Pediatric Intensive Care Unit, Children's Hospital of Chongqing Medical University Chongqing 400014, China
| | - Hong-Xing Dang
- Department of Pediatric Intensive Care Unit, Children's Hospital of Chongqing Medical University Chongqing 400014, China
| | - Fang Fang
- Ministry of Education Key Laboratory of Child Development and Disorders, Key Laboratory of Pediatrics in Chongqing, Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Children's Hospital of Chongqing Medical University Chongqing 400014, China
| | - Jing Li
- Department of Pediatric Intensive Care Unit, Children's Hospital of Chongqing Medical University Chongqing 400014, China
| |
Collapse
|
28
|
Cianci E, Recchiuti A. Targeting caveolin-1 deficiency in bone marrow derived cells: a new therapeutic window for fibrotic diseases? Front Pharmacol 2014; 5:193. [PMID: 25202273 PMCID: PMC4141328 DOI: 10.3389/fphar.2014.00193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 08/01/2014] [Indexed: 12/03/2022] Open
Affiliation(s)
- Eleonora Cianci
- Department of Medicine and Aging Science, G. d'Annunzio University Chieti, Italy ; Center of Excellence on Aging, G. d'Annunzio University Chieti, Italy
| | - Antonio Recchiuti
- Center of Excellence on Aging, G. d'Annunzio University Chieti, Italy ; Department of Experimental and Clinical Science, G. d'Annunzio University Chieti, Italy
| |
Collapse
|