1
|
Li W, Xu S, Chen L, Tan W, Deng N, Li Y, Zhang W, Deng C. Astragali Radix-Angelicae Sinensis Radix inhibits the activation of vascular adventitial fibroblasts and vascular intimal proliferation by regulating the TGF-β1/Smad2/3 pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 340:119302. [PMID: 39743186 DOI: 10.1016/j.jep.2024.119302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 12/20/2024] [Accepted: 12/28/2024] [Indexed: 01/04/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Astragali Radix-Angelicae Sinensis Radix is an important traditional Chinese medicine used for the treatment of cardiovascular diseases. Our previous studies have shown that Astragali Radix-Angelicae Sinensis Radix can inhibit vascular intimal hyperplasia and improve the blood vessel wall's ECM deposition, among which six main active components can be absorbed into the blood, suggesting that these components may be the main pharmacodynamic substances of Astragali Radix-Angelicae Sinensis Radix against vascular intimal hyperplasia. AIM OF THE STUDY A mouse model of atherosclerosis was used to study the relationship between the anti-intimal hyperplasia effect of Astragali Radix-Angelicae Sinensis Radix and the inhibition of VAF activation and ECM synthesis. Furthermore, an in vitro rat VAF activation model was used. The effects of the main active ingredients of Astragali Radix-Angelicae Sinensis Radix on the proliferation, migration and ECM synthesis of VAF were observed. The mechanism of its action was investigated by focusing on TGF-β1/Smads signaling pathway. MATERIALS AND METHODS Male ApoE-/- mice were used to establish an AS model. Observe the morphological changes of blood vessels, the expression of Vimentin, α-SMA, ECM-related factors and TGF-β1/Smads signaling pathway-related proteins. Ang Ⅱ was used to induce the VAF activation model. The cell activity, cell proliferation, cell migration, cell phenotypic markers, ECM-related factors, cell cycle regulation-related proteins and TGF-β1/Smads signaling pathway-related proteins were determined. On this basis, TGF-β1/Smads signaling pathway agonists and inhibitors were used to study the effects of the compatibility of six active components on TGF-β1/Smads signaling pathway. RESULTS Astragali Radix-Angelicae Sinensis Radix can reduce aortic intimal hyperplasia, inhibit the expression of aortic α-SMA, Vimentin, ECM components, TGF-β1, p-Samd2 and p-Samd3. Cell experiments showed that the six active ingredients could inhibit the proliferation and migration of VAF to varying degrees, inhibit the expression of α-SMA, cell cycle promoters, ECM components, up-regulate the expression of Vimentin, P21, MMP2 and MMP9. The above effects were enhanced after the combination of the six components. The 6 components and their combinations could inhibit the expression of TGF-β1/Smads signaling pathway-related proteins and up-regulate the expression of Samd7 to varying degrees. The above effects were enhanced after the combination of the 6 components. TGF-β1/Smads signaling pathway inhibitor LY2157299 showed similar effects with the six components. The inhibitory effects of the six active ingredients on TGF-β1/Smads signaling pathway-related proteins and the promotion of Smad7 expression were attenuated when agonists were added into the six active ingredient combinations. However, adding TGFβ1/Smads signaling pathway inhibitor EGF to the six active ingredient combinations had no effect on the above effects. CONCLUSION Astragali Radix-Angelicae Sinensis Radix can inhibit intimal hyperplasia, VAF activation, and ECM synthesis in atherosclerosis. The six active ingredients may be the main pharmacological substances of Astragali Radix-Angelicae Sinensis Radix to inhibit the activation of VAF, and the combination of six ingredients can enhance their effects, which may be mediated by inhibiting the activation of the TGF-β1/Smad2/3 signaling pathway.
Collapse
Affiliation(s)
- Wanyu Li
- School of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, 300 Bachelor Road, Hanpu Science and Education Park, Yuelu District, 410208Changsha City, Hunan Province, China; Hunan Key Laboratory of Integrated Chinese and Western Medicine for Prevention and Treatment of Heart and Brain Diseases, 410208, Changsha, China.
| | - Shunzhou Xu
- The First Affiliated Hospital of Hunan Junior College of Traditional Chinese Medicine, No.571, Renmin Middle Road, Lusong District, 412008, Zhuzhou City, Hunan Province, China.
| | - Lingbo Chen
- Hunan Academy of Chinese Medicine, 142 Yuehua Road, Yuelu District, 410013, Changsha City, Hunan Province, China.
| | - Wei Tan
- School of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, 300 Bachelor Road, Hanpu Science and Education Park, Yuelu District, 410208Changsha City, Hunan Province, China; Hunan Key Laboratory of Integrated Chinese and Western Medicine for Prevention and Treatment of Heart and Brain Diseases, 410208, Changsha, China.
| | - Nujiao Deng
- The First Affiliated Hospital of Hunan University of Chinese Medicine, No. 95 Shaoshan Middle Road, Yuhua District, 410208, Changsha City, Hunan Province, China.
| | - Yanling Li
- School of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, 300 Bachelor Road, Hanpu Science and Education Park, Yuelu District, 410208Changsha City, Hunan Province, China; Hunan Key Laboratory of Integrated Chinese and Western Medicine for Prevention and Treatment of Heart and Brain Diseases, 410208, Changsha, China.
| | - Wei Zhang
- School of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, 300 Bachelor Road, Hanpu Science and Education Park, Yuelu District, 410208Changsha City, Hunan Province, China; Hunan Key Laboratory of Integrated Chinese and Western Medicine for Prevention and Treatment of Heart and Brain Diseases, 410208, Changsha, China.
| | - Changqing Deng
- School of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, 300 Bachelor Road, Hanpu Science and Education Park, Yuelu District, 410208Changsha City, Hunan Province, China; Hunan Key Laboratory of Integrated Chinese and Western Medicine for Prevention and Treatment of Heart and Brain Diseases, 410208, Changsha, China.
| |
Collapse
|
2
|
Waldrep KM, Rodgers JI, Garrett SM, Wolf BJ, Feghali-Bostwick CA. The Role of SOX9 in IGF-II-Mediated Pulmonary Fibrosis. Int J Mol Sci 2023; 24:11234. [PMID: 37510994 PMCID: PMC10378869 DOI: 10.3390/ijms241411234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 06/29/2023] [Accepted: 07/02/2023] [Indexed: 07/30/2023] Open
Abstract
Pulmonary fibrosis (PF) associated with systemic sclerosis (SSc) results in significant morbidity and mortality. We previously reported that insulin-like growth factor-II (IGF-II) is overexpressed in lung tissues and fibroblasts from SSc patients, and IGF-II fosters fibrosis by upregulating collagen type I, fibronectin, and TGFβ. We now show that IGF-II augments mRNA levels of profibrotic signaling molecules TGFβ2 (p ≤ 0.01) and TGFβ3 (p ≤ 0.05), collagen type III (p ≤ 0.01), and the collagen posttranslational modification enzymes P4HA2 (p ≤ 0.05), P3H2 (p ≤ 0.05), LOX (p = 0.065), LOXL2 (p ≤ 0.05), LOXL4 (p ≤ 0.05) in primary human lung fibroblasts. IGF-II increases protein levels of TGFβ2 (p ≤ 0.01), as well as COL3A1, P4HA2, P4Hβ, and LOXL4 (p ≤ 0.05). In contrast, IGF-II decreases mRNA levels of the collagen degradation enzymes cathepsin (CTS) K, CTSB, and CTSL and protein levels of CTSK (p ≤ 0.05). The SRY-box transcription factor 9 (SOX9) is overexpressed in SSc lung tissues at the mRNA (p ≤ 0.05) and protein (p ≤ 0.01) levels compared to healthy controls. IGF-II induces SOX9 in lung fibroblasts (p ≤ 0.05) via the IGF1R/IR hybrid receptor, and SOX9 regulates TGFβ2 (p ≤ 0.05), TGFβ3 (p ≤ 0.05), COL3A1 (p ≤ 0.01), and P4HA2 (p ≤ 0.001) downstream of IGF-II. Our results identify a novel IGF-II signaling axis and downstream targets that are regulated in a SOX9-dependent and -independent manner. Our findings provide novel insights on the role of IGF-II in promoting pulmonary fibrosis.
Collapse
Affiliation(s)
- Kristy M. Waldrep
- Department of Medicine, Rheumatology, Medical University of South Carolina, Charleston, SC 29425, USA; (K.M.W.); (J.I.R.); (S.M.G.)
| | - Jessalyn I. Rodgers
- Department of Medicine, Rheumatology, Medical University of South Carolina, Charleston, SC 29425, USA; (K.M.W.); (J.I.R.); (S.M.G.)
| | - Sara M. Garrett
- Department of Medicine, Rheumatology, Medical University of South Carolina, Charleston, SC 29425, USA; (K.M.W.); (J.I.R.); (S.M.G.)
| | - Bethany J. Wolf
- Department of Public Health Sciences, Biostatistics and Bioinformatics, Medical University of South Carolina, Charleston, SC 29425, USA;
| | - Carol A. Feghali-Bostwick
- Department of Medicine, Rheumatology, Medical University of South Carolina, Charleston, SC 29425, USA; (K.M.W.); (J.I.R.); (S.M.G.)
| |
Collapse
|
3
|
Miotto DS, Duchatsch F, Dionizio A, Buzalaf MAR, Amaral SL. Physical Training vs. Perindopril Treatment on Arterial Stiffening of Spontaneously Hypertensive Rats: A Proteomic Analysis and Possible Mechanisms. Biomedicines 2023; 11:biomedicines11051381. [PMID: 37239052 DOI: 10.3390/biomedicines11051381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/21/2023] [Accepted: 05/02/2023] [Indexed: 05/28/2023] Open
Abstract
(1) Background: Arterial stiffness is an important predictor of cardiovascular events. Perindopril and physical exercise are important in controlling hypertension and arterial stiffness, but the mechanisms are unclear. (2) Methods: Thirty-two spontaneously hypertensive rats (SHR) were evaluated for eight weeks: SHRC (sedentary); SHRP (sedentary treated with perindopril-3 mg/kg) and SHRT (trained). Pulse wave velocity (PWV) analysis was performed, and the aorta was collected for proteomic analysis. (3) Results: Both treatments determined a similar reduction in PWV (-33% for SHRP and -23% for SHRT) vs. SHRC, as well as in BP. Among the altered proteins, the proteomic analysis identified an upregulation of the EH domain-containing 2 (EHD2) protein in the SHRP group, required for nitric oxide-dependent vessel relaxation. The SHRT group showed downregulation of collagen-1 (COL1). Accordingly, SHRP showed an increase (+69%) in the e-NOS protein level and SHRT showed a lower COL1 protein level (-46%) compared with SHRC. (4) Conclusions: Both perindopril and aerobic training reduced arterial stiffness in SHR; however, the results suggest that the mechanisms can be distinct. While treatment with perindopril increased EHD2, a protein involved in vessel relaxation, aerobic training decreased COL1 protein level, an important protein of the extracellular matrix (ECM) that normally enhances vessel rigidity.
Collapse
Affiliation(s)
- Danyelle Siqueira Miotto
- Joint Graduate Program in Physiological Sciences (PIPGCF), Federal University of Sao Carlos and São Paulo State University, UFSCar/UNESP, São Carlos 14801-903, Brazil
| | - Francine Duchatsch
- Joint Graduate Program in Physiological Sciences (PIPGCF), Federal University of Sao Carlos and São Paulo State University, UFSCar/UNESP, São Carlos 14801-903, Brazil
| | - Aline Dionizio
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo-USP, Bauru 17012-901, Brazil
| | | | - Sandra Lia Amaral
- Joint Graduate Program in Physiological Sciences (PIPGCF), Federal University of Sao Carlos and São Paulo State University, UFSCar/UNESP, São Carlos 14801-903, Brazil
- Department of Physical Education, School of Sciences, São Paulo State University-UNESP, Bauru 17033-360, Brazil
| |
Collapse
|
4
|
Wittig C, Szulcek R. Extracellular Matrix Protein Ratios in the Human Heart and Vessels: How to Distinguish Pathological From Physiological Changes? Front Physiol 2021; 12:708656. [PMID: 34421650 PMCID: PMC8371527 DOI: 10.3389/fphys.2021.708656] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 07/14/2021] [Indexed: 12/03/2022] Open
Abstract
Cardiovascular pathology is often accompanied by changes in relative content and/or ratios of structural extracellular matrix (ECM) proteins within the heart and elastic vessels. Three of these proteins, collagen-I, collagen-III, and elastin, make up the bulk of the ECM proteins in these tissues, forming a microenvironment that strongly dictates the tissue biomechanical properties and effectiveness of cardiac and vascular function. In this review, we aim to elucidate how the ratios of collagen-I to collagen-III and elastin to collagen are altered in cardiovascular diseases and the aged individuum. We elaborate on these major cardiovascular ECM proteins in terms of structure, tissue localization, turnover, and physiological function and address how their ratios change in aging, dilated cardiomyopathy, coronary artery disease with myocardial infarction, atrial fibrillation, aortic aneurysms, atherosclerosis, and hypertension. To the end of guiding in vitro modeling approaches, we focus our review on the human heart and aorta, discuss limitations in ECM protein quantification methodology, examine comparability between studies, and highlight potential in vitro applications. In summary, we found collagen-I relative concentration to increase or stay the same in cardiovascular disease, resulting in a tendency for increased collagen-I/collagen-III and decreased elastin/collagen ratios. These ratios were found to fall on a continuous scale with ranges defining distinct pathological states as well as a significant difference between the human heart and aortic ECM protein ratios.
Collapse
Affiliation(s)
- Corey Wittig
- Laboratory of in vitro Modeling Systems of Pulmonary Diseases, Institute of Physiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Robert Szulcek
- Laboratory of in vitro Modeling Systems of Pulmonary Diseases, Institute of Physiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
5
|
Creamer TJ, Bramel EE, MacFarlane EG. Insights on the Pathogenesis of Aneurysm through the Study of Hereditary Aortopathies. Genes (Basel) 2021; 12:183. [PMID: 33514025 PMCID: PMC7912671 DOI: 10.3390/genes12020183] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 12/15/2022] Open
Abstract
Thoracic aortic aneurysms (TAA) are permanent and localized dilations of the aorta that predispose patients to a life-threatening risk of aortic dissection or rupture. The identification of pathogenic variants that cause hereditary forms of TAA has delineated fundamental molecular processes required to maintain aortic homeostasis. Vascular smooth muscle cells (VSMCs) elaborate and remodel the extracellular matrix (ECM) in response to mechanical and biochemical cues from their environment. Causal variants for hereditary forms of aneurysm compromise the function of gene products involved in the transmission or interpretation of these signals, initiating processes that eventually lead to degeneration and mechanical failure of the vessel. These include mutations that interfere with transduction of stimuli from the matrix to the actin-myosin cytoskeleton through integrins, and those that impair signaling pathways activated by transforming growth factor-β (TGF-β). In this review, we summarize the features of the healthy aortic wall, the major pathways involved in the modulation of VSMC phenotypes, and the basic molecular functions impaired by TAA-associated mutations. We also discuss how the heterogeneity and balance of adaptive and maladaptive responses to the initial genetic insult might contribute to disease.
Collapse
Affiliation(s)
- Tyler J. Creamer
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (T.J.C.); (E.E.B.)
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Emily E. Bramel
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (T.J.C.); (E.E.B.)
- Predoctoral Training in Human Genetics and Molecular Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Elena Gallo MacFarlane
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (T.J.C.); (E.E.B.)
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
6
|
Wang M, Monticone RE, McGraw KR. Proinflammation, profibrosis, and arterial aging. Aging Med (Milton) 2020; 3:159-168. [PMID: 33103036 PMCID: PMC7574637 DOI: 10.1002/agm2.12099] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 02/10/2020] [Accepted: 02/11/2020] [Indexed: 12/18/2022] Open
Abstract
Aging is a major risk factor for quintessential cardiovascular diseases, which are closely related to arterial proinflammation. The age-related alterations of the amount, distribution, and properties of the collagen fibers, such as cross-links and degradation in the arterial wall, are the major sequelae of proinflammation. In the aging arterial wall, collagen types I, II, and III are predominant, and are mainly produced by stiffened vascular smooth muscle cells (VSMCs) governed by proinflammatory signaling, leading to profibrosis. Profibrosis is regulated by an increase in the proinflammatory molecules angiotensin II, milk fat globule-EGF-VIII, and transforming growth factor-beta 1 (TGF-β1) signaling and a decrease in the vasorin signaling cascade. The release of these proinflammatory factors triggers the activation of matrix metalloproteinase type II (MMP-2) and activates profibrogenic TGF-β1 signaling, contributing to profibrosis. The age-associated increase in activated MMP-2 cleaves latent TGF-β and subsequently increases TGF-β1 activity leading to collagen deposition in the arterial wall. Furthermore, a blockade of the proinflammatory signaling pathway alleviates the fibrogenic signaling, reduces profibrosis, and prevents arterial stiffening with aging. Thus, age-associated proinflammatory-profibrosis coupling is the underlying molecular mechanism of arterial stiffening with advancing age.
Collapse
Affiliation(s)
- Mingyi Wang
- Laboratory of Cardiovascular Science National Institute on Aging National Institutes of Health Baltimore Maryland
| | - Robert E Monticone
- Laboratory of Cardiovascular Science National Institute on Aging National Institutes of Health Baltimore Maryland
| | - Kimberly R McGraw
- Laboratory of Cardiovascular Science National Institute on Aging National Institutes of Health Baltimore Maryland
| |
Collapse
|
7
|
Drynda A, Drynda S, Kekow J, Lohmann CH, Bertrand J. Differential Effect of Cobalt and Chromium Ions as Well as CoCr Particles on the Expression of Osteogenic Markers and Osteoblast Function. Int J Mol Sci 2018; 19:ijms19103034. [PMID: 30301134 PMCID: PMC6213485 DOI: 10.3390/ijms19103034] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 09/30/2018] [Accepted: 10/01/2018] [Indexed: 01/18/2023] Open
Abstract
The balance of bone formation and resorption is the result of a regulated crosstalk between osteoblasts, osteoclasts, and osteocytes. Inflammation, mechanical load, and external stimuli modulate this system. Exposure of bone cells to metal ions or wear particles are thought to cause osteolysis via activation of osteoclasts and inhibition of osteoblast activity. Co2+ ions have been shown to impair osteoblast function and the expression of the three transforming growth factor (TGF)-β isoforms. The current study was performed to analyze how Co2+ and Cr3+ influence the expression, proliferation, and migration profile of osteoblast-like cells. The influence of Co2+, Cr3+, and CoCr particles on gene expression was analyzed using an osteogenesis PCR Array. The expression of different members of the TGF-β signaling cascade were down-regulated by Co2+, as well as several TGF-β regulated collagens, however, Cr3+ had no effect. CoCr particles partially affected similar genes as the Co2+treatment. Total collagen production of Co2+ treated osteoblasts was reduced, which can be explained by the reduced expression levels of various collagens. While proliferation of MG63 cells appears unaffected by Co2+, the migration capacity was impaired. Our data may improve the knowledge of changes in gene expression patterns, and the proliferation and migration effects caused by artificial materials.
Collapse
Affiliation(s)
- Andreas Drynda
- Department of Orthopaedic Surgery, Otto-von-Guericke University, Leipziger Straße 44, D-39120 Magdeburg, Germany.
| | - Susanne Drynda
- Department of Orthopaedic Surgery, Otto-von-Guericke University, Leipziger Straße 44, D-39120 Magdeburg, Germany.
- Clinic for Rheumatology, Otto-von-Guericke University, Leipziger Straße 44, D-39120 Magdeburg, Germany.
| | - Jörn Kekow
- Clinic for Rheumatology, Otto-von-Guericke University, Leipziger Straße 44, D-39120 Magdeburg, Germany.
| | - Christoph Hubertus Lohmann
- Department of Orthopaedic Surgery, Otto-von-Guericke University, Leipziger Straße 44, D-39120 Magdeburg, Germany.
| | - Jessica Bertrand
- Department of Orthopaedic Surgery, Otto-von-Guericke University, Leipziger Straße 44, D-39120 Magdeburg, Germany.
| |
Collapse
|
8
|
Mi Y, Wang W, Lu J, Zhang C, Wang Y, Ying H, Sun K. Proteasome-mediated degradation of collagen III by cortisol in amnion fibroblasts. J Mol Endocrinol 2018; 60:45-54. [PMID: 29191827 DOI: 10.1530/jme-17-0215] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 11/30/2017] [Indexed: 12/19/2022]
Abstract
Rupture of fetal membranes (ROM) can initiate parturition at both term and preterm. Collagen III in the compact layer of the amnion contributes to the tensile strength of fetal membranes. However, the upstream signals triggering collagen III degradation remain mostly elusive. In this study, we investigated the role of cortisol regenerated by 11β-hydroxysteroid dehydrogenase 1 (11β-HSD1) in collagen III degradation in human amnion fibroblasts with an aim to seek novel targets for the prevention of preterm premature ROM (pPROM)-elicited preterm birth. Human amnion tissue and cultured amnion tissue explants and amnion fibroblasts were used to study the regulation of collagen III, which is composed of three identical 3α 1 chains (COL3A1), by cortisol. Cortisol decreased COL3A1 protein but not mRNA abundance in a concentration-dependent manner. Cortisone also decreased COL3A1 protein, which was blocked by 11β-HSD1 inhibition. The reduction in COL3A1 protein by cortisol was not affected by a transcription inhibitor but was further enhanced by a translation inhibitor. Autophagic pathway inhibitor chloroquine or siRNA-mediated knock-down of ATG7, an essential protein for autophagy, failed to block cortisol-induced reduction in COL3A1 protein abundance, whereas proteasome pathway inhibitors MG132 and bortezomib significantly attenuated cortisol-induced reduction in COL3A1 protein abundance. Moreover, cortisol increased COL3A1 ubiquitination and the reduction of COL3A1 protein by cortisol was blocked by PYR-41, a ubiquitin-activating enzyme inhibitor. Conclusively, cortisol regenerated in amnion fibroblasts may be associated with ROM at parturition by reducing collagen III protein abundance through a ubiquitin-proteasome pathway.
Collapse
Affiliation(s)
- Yabing Mi
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, People's Republic of China
| | - Wangsheng Wang
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, People's Republic of China
| | - Jiangwen Lu
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, People's Republic of China
| | - Chuyue Zhang
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, People's Republic of China
| | - Yawei Wang
- Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Hao Ying
- Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Kang Sun
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, People's Republic of China
| |
Collapse
|
9
|
Li J, Zhang K, Ma W, Wu F, Yang P, He Z, Huang N. Investigation of enhanced hemocompatibility and tissue compatibility associated with multi-functional coating based on hyaluronic acid and Type IV collagen. Regen Biomater 2016; 3:149-57. [PMID: 27252884 PMCID: PMC4881613 DOI: 10.1093/rb/rbv030] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 12/22/2015] [Accepted: 12/30/2015] [Indexed: 12/29/2022] Open
Abstract
The biocompatibility of cardiovascular devices has always been considered crucial for their clinical efficacy. Therefore, a biofunctional coating composed of Type IV collagen (CoIV) and hyaluronan (HA) was previously fabricated onto the titanium (Ti) substrate for the application of promoting vascular smooth muscle cell contractile phenotype and improving surface endothelialization. However, the anti-inflammation property, blood compatibility and in vivo tissue compatibility of the HA/CoIV coating, as paramount consideration of cardiovascular materials surface coating, have not been investigated. Thus, in this study, the three crucial properties of the HA/CoIV coating were tested. The platelet adhesion/activation test and the dynamic whole blood experiment implied that the HA/CoIV coating had better blood compatibility compared with Ti substrate and pure CoIV coating. The macrophage adhesion/activation and inflammatory cytokine release (tumor necrosis factor-alpha and interleukin-1) results indicated that the HA/CoIV coating could significantly improve the anti-inflammation property of the Ti substrate. The in vivo implantation of SD rats for 3 weeks' results demonstrated that the HA/CoIV coating caused milder tissue response. All these results suggested that the multi-functional HA/CoIV coating possessed good biocompatibility. This research is anticipated to be potentially applied for the surface modification of cardiovascular stents.
Collapse
Affiliation(s)
- Jingan Li
- Key Laboratory for Advanced Technologies of Materials, Ministry of Education, School of Material Science and Engineering, Southwest Jiaotong University, Chengdu 610031, People’s Republic of China
| | - Kun Zhang
- Key Laboratory for Advanced Technologies of Materials, Ministry of Education, School of Material Science and Engineering, Southwest Jiaotong University, Chengdu 610031, People’s Republic of China
- School of Life Science, Zhengzhou University, 100 Science Road, Zhengzhou 450001, People’s Republic of China
- Center of Stem Cell and Regenerative Medicine, First Affiliated Hospital of Zhengzhou University, 40 University Road, Zhengzhou 450052, People’s Republic of China
| | - Wenyong Ma
- Key Laboratory for Advanced Technologies of Materials, Ministry of Education, School of Material Science and Engineering, Southwest Jiaotong University, Chengdu 610031, People’s Republic of China
| | - Feng Wu
- Key Laboratory for Advanced Technologies of Materials, Ministry of Education, School of Material Science and Engineering, Southwest Jiaotong University, Chengdu 610031, People’s Republic of China
| | - Ping Yang
- Key Laboratory for Advanced Technologies of Materials, Ministry of Education, School of Material Science and Engineering, Southwest Jiaotong University, Chengdu 610031, People’s Republic of China
| | - Zikun He
- Key Laboratory for Advanced Technologies of Materials, Ministry of Education, School of Material Science and Engineering, Southwest Jiaotong University, Chengdu 610031, People’s Republic of China
| | - Nan Huang
- Key Laboratory for Advanced Technologies of Materials, Ministry of Education, School of Material Science and Engineering, Southwest Jiaotong University, Chengdu 610031, People’s Republic of China
| |
Collapse
|
10
|
RAGE and TGF-β1 Cross-Talk Regulate Extracellular Matrix Turnover and Cytokine Synthesis in AGEs Exposed Fibroblast Cells. PLoS One 2016; 11:e0152376. [PMID: 27015414 PMCID: PMC4807770 DOI: 10.1371/journal.pone.0152376] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 03/14/2016] [Indexed: 01/11/2023] Open
Abstract
AGEs accumulation in the skin affects extracellular matrix (ECM) turnover and triggers diabetes associated skin conditions and accelerated skin aging. The receptor of AGEs (RAGE) has an essential contribution to cellular dysfunction driven by chronic inflammatory responses while TGF-β1 is critical in both dermal homeostasis and inflammation. We investigated the contribution of RAGE and TGF-β1 to the modulation of inflammatory response and ECM turnover in AGEs milieu, using a normal fibroblast cell line. RAGE, TGF-β1, collagen I and III gene and protein expression were upregulated after exposure to AGEs-BSA, and MMP-2 was activated. AGEs-RAGE was pivotal in NF-κB dependent collagen I expression and joined with TGF-β1 to stimulate collagen III expression, probably via ERK1/2 signaling. AGEs-RAGE axis induced upregulation of TGF-β1, TNF-α and IL-8 cytokines. TNF-α and IL-8 were subjected to TGF-β1 negative regulation. RAGE’s proinflammatory signaling also antagonized AGEs-TGF-β1 induced fibroblast contraction, suggesting the existence of an inhibitory cross-talk mechanism between TGF-β1 and RAGE signaling. RAGE and TGF-β1 stimulated anti-inflammatory cytokines IL-2 and IL-4 expression. GM-CSF and IL-6 expression appeared to be dependent only on TGF-β1 signaling. Our data also indicated that IFN-γ upregulated in AGEs-BSA milieu in a RAGE and TGF-β1 independent mechanism. Our findings raise the possibility that RAGE and TGF-β1 are both involved in fibrosis development in a complex cross-talk mechanism, while also acting on their own individual targets. This study contributes to the understanding of impaired wound healing associated with diabetes complications.
Collapse
|
11
|
Bersi MR, Bellini C, Wu J, Montaniel KRC, Harrison DG, Humphrey JD. Excessive Adventitial Remodeling Leads to Early Aortic Maladaptation in Angiotensin-Induced Hypertension. Hypertension 2016; 67:890-896. [PMID: 27001298 DOI: 10.1161/hypertensionaha.115.06262] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 01/22/2016] [Indexed: 01/27/2023]
Abstract
The primary function of central arteries is to store elastic energy during systole and to use it to sustain blood flow during diastole. Arterial stiffening compromises this normal mechanical function and adversely affects end organs, such as the brain, heart, and kidneys. Using an angiotensin II infusion model of hypertension in wild-type mice, we show that the thoracic aorta exhibits a dramatic loss of energy storage within 2 weeks that persists for at least 4 weeks. This diminished mechanical functionality results from increased structural stiffening as a result of an excessive accumulation of adventitial collagen, not a change in the intrinsic stiffness of the wall. A detailed analysis of the transmural biaxial wall stress suggests that the exuberant production of collagen results more from an inflammatory response than from a mechano-adaptation, hence reinforcing the need to control inflammation, not just blood pressure. Although most clinical assessments of arterial stiffening focus on intimal-medial thickening, these results suggest a need to measure and control the highly active and important adventitia.
Collapse
Affiliation(s)
- Mathew R Bersi
- Department of Biomedical Engineering, Yale University, New Haven, CT
| | - Chiara Bellini
- Department of Biomedical Engineering, Yale University, New Haven, CT
| | - Jing Wu
- Departments of Medicine and Pharmacology, Vanderbilt University, Nashville, TN
| | - Kim R C Montaniel
- Departments of Medicine and Pharmacology, Vanderbilt University, Nashville, TN
| | - David G Harrison
- Departments of Medicine and Pharmacology, Vanderbilt University, Nashville, TN
| | - Jay D Humphrey
- Department of Biomedical Engineering, Yale University, New Haven, CT.,Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT
| |
Collapse
|
12
|
Luan YY, Yin CF, Qin QH, Dong N, Zhu XM, Sheng ZY, Zhang QH, Yao YM. Effect of Regulatory T Cells on Promoting Apoptosis of T Lymphocyte and Its Regulatory Mechanism in Sepsis. J Interferon Cytokine Res 2015; 35:969-80. [PMID: 26309018 DOI: 10.1089/jir.2014.0235] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
With both in vivo and in vitro experiments, the present study was conducted to investigate the effect of regulatory T cell (Treg) on promoting T-lymphocyte apoptosis and its regulatory mechanism through transforming growth factor-beta (TGF-β1) signaling in mice. A murine model of polymicrobial sepsis was reproduced by cecal ligation and puncture (CLP); PC61 and anti-TGF-β antibodies were used to decrease counts of CD4(+)CD25(+) Tregs and inhibit TGF-β activity, respectively. Splenic CD4(+)CD25(+) Tregs and CD4(+)CD25(-) T cells were isolated. Phenotypes, including cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4), forkhead/winged helix transcription factor p3 (Foxp3), and TGFβ1(m+), as well as the apoptotic rate of CD4(+)CD25(-) T cell, were analyzed by flow cytometry. Real-time reverse transcription-polymerase chain reaction was performed to determine mRNA expression of TGF-β1, and the expressions of Smad2/Smad3, Bcl-2 superfamily members of Bcl-2/Bim, cytochrome C, the mitochondrial membrane potential, and caspases in CD4(+)CD25(-) T cells were simultaneously determined. After treatment with PC61 or anti-TGF-β antibody, CTLA-4, Foxp3, and TGFβ1(m+) expressions of CD4(+)CD25(+) Tregs were markedly decreased in comparison to that of the CLP group and the apoptosis rate of CD4(+)CD25(-) T cells was significantly positively correlated with the expression of TGF-β1. Meanwhile, levels of P-Smad2/P-Smad3, proapoptotic protein Bim, cytochrome C, and activity of caspase-3, -8, -9 were downregulated, whereas the mitochondrial membrane potential and antiapoptotic protein Bcl-2 expression were restored. Taken together, our data indicated that the TGF-β1 signal could be partly involved in the apoptosis of CD4(+)CD25(-) T cells promoted by CD4(+)CD25(+) Tregs, therefore inhibition of TGF-β1 expression may provide a novel strategy for the improvement of host immunosuppression following sepsis.
Collapse
Affiliation(s)
- Ying-yi Luan
- Department of Microbiology and Immunology, Trauma Research Center , First Hospital Affiliated to the Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Cheng-fen Yin
- Department of Microbiology and Immunology, Trauma Research Center , First Hospital Affiliated to the Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Qing-hua Qin
- Department of Microbiology and Immunology, Trauma Research Center , First Hospital Affiliated to the Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Ning Dong
- Department of Microbiology and Immunology, Trauma Research Center , First Hospital Affiliated to the Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Xiao-mei Zhu
- Department of Microbiology and Immunology, Trauma Research Center , First Hospital Affiliated to the Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Zhi-yong Sheng
- Department of Microbiology and Immunology, Trauma Research Center , First Hospital Affiliated to the Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Qing-hong Zhang
- Department of Microbiology and Immunology, Trauma Research Center , First Hospital Affiliated to the Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Yong-ming Yao
- Department of Microbiology and Immunology, Trauma Research Center , First Hospital Affiliated to the Chinese PLA General Hospital, Beijing, People's Republic of China
| |
Collapse
|
13
|
Bender SB, Castorena-Gonzalez JA, Garro M, Reyes-Aldasoro CC, Sowers JR, DeMarco VG, Martinez-Lemus LA. Regional variation in arterial stiffening and dysfunction in Western diet-induced obesity. Am J Physiol Heart Circ Physiol 2015; 309:H574-82. [PMID: 26092984 DOI: 10.1152/ajpheart.00155.2015] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 06/18/2015] [Indexed: 12/11/2022]
Abstract
Increased central vascular stiffening, assessed in vivo by determination of pulse wave velocity (PWV), is an independent predictor of cardiovascular event risk. Recent evidence demonstrates that accelerated aortic stiffening occurs in obesity; however, little is known regarding stiffening of other disease-relevant arteries or whether regional variation in arterial stiffening occurs in this setting. We addressed this gap in knowledge by assessing femoral PWV in vivo in conjunction with ex vivo analyses of femoral and coronary structure and function in a mouse model of Western diet (WD; high-fat/high-sugar)-induced obesity and insulin resistance. WD feeding resulted in increased femoral PWV in vivo. Ex vivo analysis of femoral arteries revealed a leftward shift in the strain-stress relationship, increased modulus of elasticity, and decreased compliance indicative of increased stiffness following WD feeding. Confocal and multiphoton fluorescence microscopy revealed increased femoral stiffness involving decreased elastin/collagen ratio in conjunction with increased femoral transforming growth factor-β (TGF-β) content in WD-fed mice. Further analysis of the femoral internal elastic lamina (IEL) revealed a significant reduction in the number and size of fenestrae with WD feeding. Coronary artery stiffness and structure was unchanged by WD feeding. Functionally, femoral, but not coronary, arteries exhibited endothelial dysfunction, whereas coronary arteries exhibited increased vasoconstrictor responsiveness not present in femoral arteries. Taken together, our data highlight important regional variations in the development of arterial stiffness and dysfunction associated with WD feeding. Furthermore, our results suggest TGF-β signaling and IEL fenestrae remodeling as potential contributors to femoral artery stiffening in obesity.
Collapse
Affiliation(s)
- Shawn B Bender
- Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri; Department of Biomedical Sciences, University of Missouri School of Medicine, Columbia, Missouri; Dalton Cardiovascular Research Center, University of Missouri School of Medicine, Columbia, Missouri
| | - Jorge A Castorena-Gonzalez
- Dalton Cardiovascular Research Center, University of Missouri School of Medicine, Columbia, Missouri; Department of Biological Engineering, University of Missouri, Columbia, Missouri
| | - Mona Garro
- Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri; Department of Medicine-Endocrinology, Diabetes and Metabolism University of Missouri School of Medicine, Columbia, Missouri
| | | | - James R Sowers
- Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri; Dalton Cardiovascular Research Center, University of Missouri School of Medicine, Columbia, Missouri; Department of Medicine-Endocrinology, Diabetes and Metabolism University of Missouri School of Medicine, Columbia, Missouri, Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri; and
| | - Vincent G DeMarco
- Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri; Department of Medicine-Endocrinology, Diabetes and Metabolism University of Missouri School of Medicine, Columbia, Missouri, Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri; and
| | - Luis A Martinez-Lemus
- Dalton Cardiovascular Research Center, University of Missouri School of Medicine, Columbia, Missouri; Department of Biological Engineering, University of Missouri, Columbia, Missouri; Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri; and
| |
Collapse
|
14
|
Li N, Kong M, Ma T, Gao W, Ma S. Uighur medicine abnormal savda munzip (ASMq) suppresses expression of collagen and TGF-β1 with concomitant induce Smad7 in human hypertrophic scar fibroblasts. Int J Clin Exp Med 2015; 8:8551-8560. [PMID: 26309506 PMCID: PMC4538185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 06/07/2015] [Indexed: 06/04/2023]
Abstract
BACKGROUND Hypertrophic scar (HS) is a common dermal disease, for which numerous treatments are currently available but they do not always yield excellent therapeutic results. Hence, alternative strategy are needed. Recent basic and clinic research has shown that Uighur medicine abnormal savda munzip (ASMq) has anti-hypertrophic scar properties but its molecular mechanism is unknown. The aim of this study was to explore the effect of ASMq on TGF-β/Smads signaling in fibroblasts derived from hypertrophic scar. PURPOSE To investigate the effect of ASMq on the TGF-β/Smads signaling pathway in hypertrophic scar fibroblasts (HSFs). METHODS Hypertrophic scar fibroblasts (HSFs) were isolated from human of hypertrophic scar and passaged to the 3~4 generation, which were treated with the different concentrations of ASMq. Cells treated with 5-Fu served as the positive control group. After treatment for 48 hours, expressions of Smad7, TGF-β1, type I and III collagen, were examined by immunocytochemistry, reverse transcription PCR and Western blotting, respectively. RESULTS ASMq markedly enhanced the expression of inhibitory Smad7, with suppression of type I and III collagen and TGF-β1. We observed that treatment of ASMq induced Smad7 to enter the cytoplasm from the nucleus of hypertrophic fibroblasts. CONCLUSIONS ASMq inhibits scarring probably by enhancing the expression of inhibitory Smad7, and inhibiting TGF-β1, collagen expression, and is a potential treatment for scarring.
Collapse
Affiliation(s)
- Nan Li
- Department of Plastic Surgery, 1st Affiliated Hospital of Xinjiang Medical University Urumqi 830054, China
| | - Menglong Kong
- Department of Plastic Surgery, 1st Affiliated Hospital of Xinjiang Medical University Urumqi 830054, China
| | - Tao Ma
- Department of Plastic Surgery, 1st Affiliated Hospital of Xinjiang Medical University Urumqi 830054, China
| | - Weicheng Gao
- Department of Plastic Surgery, 1st Affiliated Hospital of Xinjiang Medical University Urumqi 830054, China
| | - Shaolin Ma
- Department of Plastic Surgery, 1st Affiliated Hospital of Xinjiang Medical University Urumqi 830054, China
| |
Collapse
|
15
|
Preeclampsia serum-induced collagen I expression and intracellular calcium levels in arterial smooth muscle cells are mediated by the PLC-γ1 pathway. Exp Mol Med 2014; 46:e115. [PMID: 25257609 PMCID: PMC4183944 DOI: 10.1038/emm.2014.59] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 07/28/2014] [Accepted: 07/31/2014] [Indexed: 01/08/2023] Open
Abstract
In women with preeclampsia (PE), endothelial cell (EC) dysfunction can lead to altered secretion of paracrine factors that induce peripheral vasoconstriction and proteinuria. This study examined the hypothesis that PE sera may directly or indirectly, through human umbilical vein ECs (HUVECs), stimulate phospholipase C-γ1-1,4,5-trisphosphate (PLC-γ1-IP3) signaling, thereby increasing protein kinase C-α (PKC-α) activity, collagen I expression and intracellular Ca2+ concentrations ([Ca2+]i) in human umbilical artery smooth muscle cells (HUASMCs). HUASMCs and HUVECs were cocultured with normal or PE sera before PLC-γ1 silencing. Increased PLC-γ1 and IP3 receptor (IP3R) phosphorylation was observed in cocultured HUASMCs stimulated with PE sera (P<0.05). In addition, PE serum significantly increased HUASMC viability and reduced their apoptosis (P<0.05); these effects were abrogated with PLC-γ1 silencing. Compared with normal sera, PE sera increased [Ca2+]i in cocultured HUASMCs (P<0.05), which was inhibited by PLC-γ1 and IP3R silencing. Finally, PE sera-induced PKC-α activity and collagen I expression was inhibited by PLC-γ1 small interfering RNA (siRNA) (P<0.05). These results suggest that vasoactive substances in the PE serum may induce deposition in the extracellular matrix through the activation of PLC-γ1, which may in turn result in thickening and hardening of the placental vascular wall, placental blood supply shortage, fetal hypoxia–ischemia and intrauterine growth retardation or intrauterine fetal death. PE sera increased [Ca2+]i and induced PKC-α activation and collagen I expression in cocultured HUASMCs via the PLC-γ1 pathway.
Collapse
|
16
|
Guo LW, Wang B, Goel SA, Little C, Takayama T, Shi XD, Roenneburg D, DiRenzo D, Kent KC. Halofuginone stimulates adaptive remodeling and preserves re-endothelialization in balloon-injured rat carotid arteries. Circ Cardiovasc Interv 2014; 7:594-601. [PMID: 25074254 DOI: 10.1161/circinterventions.113.001181] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND Three major processes, constrictive vessel remodeling, intimal hyperplasia (IH), and retarded re-endothelialization, contribute to restenosis after vascular reconstructions. Clinically used drugs inhibit IH but delay re-endothelialization and also cause constrictive remodeling. Here we have examined halofuginone, an herbal derivative, for its beneficial effects on vessel remodeling and differential inhibition of IH versus re-endothelialization. METHODS AND RESULTS Two weeks after perivascular application to balloon-injured rat common carotid arteries, halofuginone versus vehicle (n=6 animals) enlarged luminal area 2.14-fold by increasing vessel size (adaptive remodeling; 123%), reducing IH (74.3%) without inhibiting re-endothelialization. Consistent with its positive effect on vessel expansion, halofuginone reduced collagen type 1 (but not type 3) production in injured arteries as well as that from adventitial fibroblasts in vitro. In support of its differential effects on IH versus re-endothelialization, halofuginone produced greater inhibition of vascular smooth muscle cell versus endothelial cell proliferation at concentrations ≈50 nmol/L. Furthermore, halofuginone at 50 nmol/L effectively blocked Smad3 phosphorylation in smooth muscle cells, which is known to promote smooth muscle cell proliferation, migration, and IH, but halofuginone had no effect on phospho-Smad3 in endothelial cells. CONCLUSIONS Periadventitial delivery of halofuginone dramatically increased lumen patency via adaptive remodeling and selective inhibition of IH without affecting endothelium recovery. Halofuginone is the first reported small molecule that has favorable effects on all 3 major processes involved in restenosis.
Collapse
Affiliation(s)
- Lian-Wang Guo
- From the Department of Surgery, University of Wisconsin, Madison.
| | - Bowen Wang
- From the Department of Surgery, University of Wisconsin, Madison
| | - Shakti A Goel
- From the Department of Surgery, University of Wisconsin, Madison
| | | | - Toshio Takayama
- From the Department of Surgery, University of Wisconsin, Madison
| | - Xu Dong Shi
- From the Department of Surgery, University of Wisconsin, Madison
| | - Drew Roenneburg
- From the Department of Surgery, University of Wisconsin, Madison
| | - Daniel DiRenzo
- From the Department of Surgery, University of Wisconsin, Madison
| | - K Craig Kent
- From the Department of Surgery, University of Wisconsin, Madison
| |
Collapse
|
17
|
Golpour M, Fattahi S, Niaki HA, Hadipoor A, Abedian Z, Ahangarian GR, Parsian H, Mosapour A, Khorasani HR, Vaziri HR, Bijani A, Mostafazadeh A. Starved human fibroblasts secrete acidic proteins inducing post re-feeding proliferation and in vitro cell migration: a potential tool for wound healing. Biol Cell 2014; 106:139-50. [PMID: 24612410 DOI: 10.1111/boc.201300063] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 02/18/2014] [Indexed: 12/27/2022]
Abstract
BACKGROUND INFORMATION There are several reports indicating that starved fibroblasts show higher proliferation rates when re-fed with foetal bovine serum. We have evidence demonstrating that this phenomenon is related to secretory proteins which may be beneficial to wound healing. RESULTS After re-feeding, 16 and 72 h serum-starved fibroblasts showed the highest and lowest proliferation rates, 1.59 and 0.51-fold difference compared to the non-starved control, respectively (P < 0.05). However, the latest value could be normalised by incubating cells with 16 h-starved fibroblast cell culture supernatant (16-SFS), prior to re-feeding. A strong correlation was found between total protein level in starved fibroblast culture supernatants and post re-feeding proliferation rates (r(2) = 0.90, P < 0.001). Two-dimensional gel electrophoresis analysis of 16-SFS confirmed the presence of proteins with relative molecular weights of 10-120 kDa and pI ranging from 4 to 6. A significant difference in calcium influx course was found between 16-SFS and the negative control (Dulbecco's Modified Eagle Medium) (P < 0.05). There was no significant difference in Ca(2+) concentrations after 1 h between non-starved controls and 16-SFS-treated fibroblasts. The scratch test demonstrated that the 16-SFS is able to induce fibroblast migration. CONCLUSIONS We concluded that human starved fibroblasts secrete proteins that are able to induce post re-feeding cell proliferation and fibroblasts migration, probably through the induction of a sustained calcium influx. This is worth being considered as a potential tool for wound healing.
Collapse
Affiliation(s)
- Monireh Golpour
- Cellular and Molecular Biology Research Center, Babol University of Medical Sciences, Babol, Iran; University of Guilan, Rasht, Iran
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Chen C, Lei W, Chen W, Zhong J, Gao X, Li B, Wang H, Huang C. Serum TGF-β1 and SMAD3 levels are closely associated with coronary artery disease. BMC Cardiovasc Disord 2014; 14:18. [PMID: 24533640 PMCID: PMC3936998 DOI: 10.1186/1471-2261-14-18] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 02/11/2014] [Indexed: 12/21/2022] Open
Abstract
Background Coronary artery disease (CAD) is one of the most common diseases leading to mortality and morbidity worldwide. There is considerable debate on whether serum transforming growth factor β1 (TGF-β1) levels are associated with long-term major adverse cardiovascular events in patients with CAD, and to date, no study has specifically addressed levels in patients with different degrees of CAD severity. Methods Serum TGF-β1 and mothers against decapentaplegic homolog 3 (SMAD3) concentrations were evaluated in 279 patients with CAD and 268 controls without CAD. The clinical and biochemical characteristics of all subjects were also determined and analyzed. Results TGF-β1 and SMAD3 concentrations in CAD patients were significantly higher than those in the controls. The serum TGF-β1 level in acute myocardial infarction (AMI) was significantly higher than that in both stable angina pectoris (SAP) and unstable angina pectoris (UAP) (p < 0.05), while there was no marked difference between levels in SAP and UAP (p > 0.05). SMAD3 levels showed no obvious difference among AMI, SAP, and UAP. TGF-β1 and SMAD3 are potential biomarkers for CAD, and may be more accurate than Lpa, ApoA1, uric acid, BUN, or triglycerides (TG). Conclusions Serum TGF-β1 and SMAD3 levels are closely associated with CAD, and may become useful biomarkers for diagnosis and risk stratification.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Congxin Huang
- Department of Cardiovascular Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| |
Collapse
|
19
|
Ieronimakis N, Hays AL, Janebodin K, Mahoney WM, Duffield JS, Majesky MW, Reyes M. Coronary adventitial cells are linked to perivascular cardiac fibrosis via TGFβ1 signaling in the mdx mouse model of Duchenne muscular dystrophy. J Mol Cell Cardiol 2013; 63:122-34. [PMID: 23911435 PMCID: PMC3834000 DOI: 10.1016/j.yjmcc.2013.07.014] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2013] [Revised: 06/20/2013] [Accepted: 07/23/2013] [Indexed: 01/12/2023]
Abstract
In Duchenne muscular dystrophy (DMD), progressive accumulation of cardiac fibrosis promotes heart failure. While the cellular origins of fibrosis in DMD hearts remain enigmatic, fibrotic tissue conspicuously forms near the coronary adventitia. Therefore, we sought to characterize the role of coronary adventitial cells in the formation of perivascular fibrosis. Utilizing the mdx model of DMD, we have identified a population of Sca1+, PDGFRα+, CD31-, and CD45- coronary adventitial cells responsible for perivascular fibrosis. Histopathology of dystrophic hearts revealed that Sca1+ cells extend from the adventitia and occupy regions of perivascular fibrosis. The number of Sca1+ adventitial cells increased two-fold in fibrotic mdx hearts vs. age matched wild-type hearts. Moreover, relative to Sca1-, PDGFRα+, CD31-, and CD45- cells and endothelial cells, Sca1+ adventitial cells FACS-sorted from mdx hearts expressed the highest level of Collagen1α1 and 3α1, Connective tissue growth factor, and Tgfβr1 transcripts. Surprisingly, mdx endothelial cells expressed the greatest level of the Tgfβ1 ligand. Utilizing Collagen1α1-GFP reporter mice, we confirmed that the majority of Sca1+ adventitial cells expressed type I collagen, an abundant component of cardiac fibrosis, in both wt (71%±4.1) and mdx (77%±3.5) hearts. In contrast, GFP+ interstitial fibroblasts were PDGFRα+ but negative for Sca1. Treatment of cultured Collagen1α1-GFP+ adventitial cells with TGFβ1 resulted in increased collagen synthesis, whereas pharmacological inhibition of TGFβR1 signaling reduced the fibrotic response. Therefore, perivascular cardiac fibrosis by coronary adventitial cells may be mediated by TGFβ1 signaling. Our results implicate coronary endothelial cells in mediating cardiac fibrosis via transmural TGFβ signaling, and suggest that the coronary adventitia is a promising target for developing novel anti-fibrotic therapies.
Collapse
|
20
|
Ruddy JM, Jones JA, Ikonomidis JS. Pathophysiology of thoracic aortic aneurysm (TAA): is it not one uniform aorta? Role of embryologic origin. Prog Cardiovasc Dis 2013; 56:68-73. [PMID: 23993239 DOI: 10.1016/j.pcad.2013.04.002] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Thoracic aortic aneurysm (TAA) is a clinically silent and potentially fatal disease whose pathophysiology is poorly understood. Application of data derived from animal models and human tissue analysis of abdominal aortic aneurysms may prove misleading given current evidence of structural and biochemical aortic heterogeneity above and below the diaphragm. Genetic predisposition is more common in TAA and includes multi-faceted syndromes such as Marfan, Loeys-Dietz, and type IV Ehlers-Danlos as well as autosomal-dominant familial patterns of inheritance. Investigation into the consequences of these known mutations has provided insight into the cell signaling cascades leading to degenerative remodeling of the aortic medial extracellular matrix (ECM) with TGF-β playing a major role. Targeted research into modifying the upstream regulation or downstream effects of the TGF-β1 pathway may provide opportunities for intervention to attenuate TAA progression.
Collapse
Affiliation(s)
- Jean Marie Ruddy
- Division of Cardiothoracic Surgery, Department of Surgery, Medical University of South Carolina, Charlston, SC, USA
| | | | | |
Collapse
|