1
|
Volpini X, Natali L, Brugo MB, de la Cruz-Thea B, Baigorri RE, Cerbán FM, Fozzatti L, Motran CC, Musri MM. Trypanosoma cruzi Infection Promotes Vascular Remodeling and Coexpression of α-Smooth Muscle Actin and Macrophage Markers in Cells of the Aorta. ACS Infect Dis 2022; 8:2271-2290. [PMID: 36083791 DOI: 10.1021/acsinfecdis.2c00318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Chagas disease is an emerging global health problem; however, it remains neglected. Increased aortic stiffness (IAS), a predictor of cardiovascular events, has recently been reported in asymptomatic chronic Chagas patients. After vascular injury, smooth muscle cells (SMCs) can undergo alterations associated with phenotypic switch and transdifferentiation, promoting vascular remodeling and IAS. By studying different mouse aortic segments, we tested the hypothesis that Trypanosoma cruzi infection promotes vascular remodeling. Interestingly, the thoracic aorta was the most affected by the infection. Decreased expression of SMC markers and increased expression of proliferative markers were observed in the arteries of acutely infected mice. In acutely and chronically infected mice, we observed cells coexpressing SMC and macrophage (Mo) markers in the media and adventitia layers of the aorta, indicating that T. cruzi might induce cellular processes associated with SMC transdifferentiation into Mo-like cells or vice versa. In the adventitia, the Mo cell functional polarization was associated with an M2-like CD206+arginase-1+ phenotype despite the T. cruzi presence in the tissue. Only Mo-like cells in inflammatory foci were CD206+iNOS+. In addition to the disorganization of elastic fibers, we found thickening of the aortic layers during the acute and chronic phases of the disease. Our findings indicate that T. cruzi infection induces a vascular remodeling with SMC dedifferentiation and increased cell populations coexpressing α-SMA and Mo markers that could be associated with IAS promotion. These data highlight the importance of studying large vessel homeostasis in Chagas disease.
Collapse
Affiliation(s)
- Ximena Volpini
- Instituto de Investigaciones Médicas Mercedes y Martín Ferreyra. Consejo Nacional de Investigaciones Científicas y Tecnicas. Universidad Nacional de Córdoba (INIMEC-CONICET-UNC), Friuli 2434. Colinas de Velez Sarfield, Córdoba, PC X5016NST, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología. Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Haya de la Torre y Medina Allende. Ciudad Universitaria, Córdoba, PC X5000HUA, Argentina.,Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba (FCQ-UNC). Ciudad Universitaria, Córdoba, PC X5000HUA, Argentina
| | - Lautaro Natali
- Instituto de Investigaciones Médicas Mercedes y Martín Ferreyra. Consejo Nacional de Investigaciones Científicas y Tecnicas. Universidad Nacional de Córdoba (INIMEC-CONICET-UNC), Friuli 2434. Colinas de Velez Sarfield, Córdoba, PC X5016NST, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología. Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Haya de la Torre y Medina Allende. Ciudad Universitaria, Córdoba, PC X5000HUA, Argentina
| | - Maria Belén Brugo
- Centro de Investigaciones en Bioquímica Clínica e Inmunología. Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Haya de la Torre y Medina Allende. Ciudad Universitaria, Córdoba, PC X5000HUA, Argentina.,Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba (FCQ-UNC). Ciudad Universitaria, Córdoba, PC X5000HUA, Argentina
| | - Benjamin de la Cruz-Thea
- Instituto de Investigaciones Médicas Mercedes y Martín Ferreyra. Consejo Nacional de Investigaciones Científicas y Tecnicas. Universidad Nacional de Córdoba (INIMEC-CONICET-UNC), Friuli 2434. Colinas de Velez Sarfield, Córdoba, PC X5016NST, Argentina
| | - Ruth Eliana Baigorri
- Centro de Investigaciones en Bioquímica Clínica e Inmunología. Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Haya de la Torre y Medina Allende. Ciudad Universitaria, Córdoba, PC X5000HUA, Argentina.,Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba (FCQ-UNC). Ciudad Universitaria, Córdoba, PC X5000HUA, Argentina
| | - Fabio Marcelo Cerbán
- Centro de Investigaciones en Bioquímica Clínica e Inmunología. Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Haya de la Torre y Medina Allende. Ciudad Universitaria, Córdoba, PC X5000HUA, Argentina.,Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba (FCQ-UNC). Ciudad Universitaria, Córdoba, PC X5000HUA, Argentina
| | - Laura Fozzatti
- Centro de Investigaciones en Bioquímica Clínica e Inmunología. Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Haya de la Torre y Medina Allende. Ciudad Universitaria, Córdoba, PC X5000HUA, Argentina.,Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba (FCQ-UNC). Ciudad Universitaria, Córdoba, PC X5000HUA, Argentina
| | - Claudia Cristina Motran
- Centro de Investigaciones en Bioquímica Clínica e Inmunología. Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Haya de la Torre y Medina Allende. Ciudad Universitaria, Córdoba, PC X5000HUA, Argentina.,Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba (FCQ-UNC). Ciudad Universitaria, Córdoba, PC X5000HUA, Argentina
| | - Melina Mara Musri
- Instituto de Investigaciones Médicas Mercedes y Martín Ferreyra. Consejo Nacional de Investigaciones Científicas y Tecnicas. Universidad Nacional de Córdoba (INIMEC-CONICET-UNC), Friuli 2434. Colinas de Velez Sarfield, Córdoba, PC X5016NST, Argentina.,Departamento de Fisiología, Facultad de Ciencias Exactas Físicas y Naturales. Universidad Nacional de Córdoba (FCEFyN-UNC). Av. Velez Sarfield 299, Centro, Córdoba, PC X5000JJC, Argentina
| |
Collapse
|
2
|
Muta K, Nakazawa Y, Obata Y, Inoue H, Torigoe K, Nakazawa M, Abe K, Furusu A, Miyazaki M, Yamamoto K, Koji T, Nishino T. An inhibitor of Krüppel-like factor 5 suppresses peritoneal fibrosis in mice. Perit Dial Int 2021; 41:394-403. [PMID: 33522431 DOI: 10.1177/0896860820981322] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACK GROUND Krüppel-like transcription factor 5 (KLF5) is a transcription factor regulating cell proliferation, angiogenesis and differentiation. It has been recently reported that Am80, a synthetic retinoic acid receptor α-specific agonist, inhibits the expression of KLF5. In the present study, we have examined the expression of KLF5 in fibrotic peritoneum induced by chlorhexidine gluconate (CG) in mouse and evaluated that Am80, as an inhibitor of KLF5, can reduce peritoneal fibrosis. METHODS Peritoneal fibrosis was induced by intraperitoneal injection of CG into peritoneal cavity of ICR mice. Am80 was administered orally for every day from the start of CG injection. Control mice received only a vehicle (0.5% carboxymethylcellulose solution). After 3 weeks of treatment, peritoneal equilibration test (PET) was performed and peritoneal tissues were examined by immunohistochemistry. RESULTS The expression of KLF5 was less found in the peritoneal tissue of control mice, while KLF5 was expressed in the thickened submesothelial area of CG-injected mice receiving the vehicle. Am80 treatment reduced KLF5 expression and remarkably attenuated peritoneal thickening, accompanied with the reduction of type III collagen expression. The numbers of transforming growth factor β-positive cells, α-smooth muscle actin-positive cells and infiltrating macrophages were significantly decreased in Am80-treated group. PET revealed the increased peritoneal permeability in CG mice, whereas Am80 administration significantly improved the peritoneal high permeability state. CONCLUSIONS These results indicate the involvement of KLF5 in the progression of experimental peritoneal fibrosis and suggest that Am80 may be potentially useful for the prevention of peritoneal fibrosis through inhibition of KLF5 expression.
Collapse
Affiliation(s)
- Kumiko Muta
- Department of Nephrology, 88380Nagasaki University Hospital, Japan
| | - Yuka Nakazawa
- Department of Nephrology, Hokusyo Central Hospital, Nagasaki, Japan
| | - Yoko Obata
- Department of Nephrology, 88380Nagasaki University Hospital, Japan.,Medical Education Development Center, 88380Nagasaki University Hospital, Japan
| | - Hiro Inoue
- Department of Nephrology, 88380Nagasaki University Hospital, Japan
| | - Kenta Torigoe
- Department of Nephrology, 88380Nagasaki University Hospital, Japan
| | - Masayuki Nakazawa
- Department of Nephrology, Sasebo City Central Hospital, Nagasaki, Japan
| | | | - Akira Furusu
- Department of Nephrology, Wajinkai Hospital, Nagasaki, Japan
| | | | - Kazuo Yamamoto
- Biomedical Research Support Center, Nagasaki University School of Medicine, Japan
| | - Takehiko Koji
- Department of Histology and Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Japan
| | - Tomoya Nishino
- Department of Nephrology, 88380Nagasaki University Hospital, Japan
| |
Collapse
|
3
|
Chen F, Shao F, Hinds A, Yao S, Ram-Mohan S, Norman TA, Krishnan R, Fine A. Retinoic acid signaling is essential for airway smooth muscle homeostasis. JCI Insight 2018; 3:120398. [PMID: 30135301 DOI: 10.1172/jci.insight.120398] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 07/11/2018] [Indexed: 12/18/2022] Open
Abstract
Airway smooth muscle (ASM) is a dynamic and complex tissue involved in regulation of bronchomotor tone, but the molecular events essential for the maintenance of ASM homeostasis are not well understood. Observational and genome-wide association studies in humans have linked airway function to the nutritional status of vitamin A and its bioactive metabolite retinoic acid (RA). Here, we provide evidence that ongoing RA signaling is critical for the regulation of adult ASM phenotype. By using dietary, pharmacologic, and genetic models in mice and humans, we show that (a) RA signaling is active in adult ASM in the normal lung, (b) RA-deficient ASM cells are hypertrophic, hypercontractile, profibrotic, but not hyperproliferative, (c) TGF-β signaling, known to cause ASM hypertrophy and airway fibrosis in human obstructive lung diseases, is hyperactivated in RA-deficient ASM, (d) pharmacologic and genetic inhibition of the TGF-β activity in ASM prevents the development of the aberrant phenotype induced by RA deficiency, and (e) the consequences of transient RA deficiency in ASM are long-lasting. These results indicate that RA signaling actively maintains adult ASM homeostasis, and disruption of RA signaling leads to aberrant ASM phenotypes similar to those seen in human chronic airway diseases such as asthma.
Collapse
Affiliation(s)
- Felicia Chen
- The Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Fengzhi Shao
- The Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Anne Hinds
- The Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Sean Yao
- Center for Vascular Biology Research, Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Sumati Ram-Mohan
- Center for Vascular Biology Research, Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Timothy A Norman
- The Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Ramaswamy Krishnan
- Center for Vascular Biology Research, Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Alan Fine
- The Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA.,Division of Pulmonary, Critical Care, and Allergy, West Roxbury Veterans Hospital, West Roxbury, Massachusetts, USA
| |
Collapse
|
4
|
Mozos I, Stoian D, Caraba A, Malainer C, Horbańczuk JO, Atanasov AG. Lycopene and Vascular Health. Front Pharmacol 2018; 9:521. [PMID: 29875663 PMCID: PMC5974099 DOI: 10.3389/fphar.2018.00521] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 04/30/2018] [Indexed: 01/20/2023] Open
Abstract
Lycopene is a lipophilic, unsaturated carotenoid, found in red-colored fruits and vegetables, including tomatoes, watermelon, papaya, red grapefruits, and guava. The present work provides an up to date overview of mechanisms linking lycopene in the human diet and vascular changes, considering epidemiological data, clinical studies, and experimental data. Lycopene may improve vascular function and contributes to the primary and secondary prevention of cardiovascular disorders. The main activity profile of lycopene includes antiatherosclerotic, antioxidant, anti-inflammatory, antihypertensive, antiplatelet, anti-apoptotic, and protective endothelial effects, the ability to improve the metabolic profile, and reduce arterial stiffness. In this context, lycopene has been shown in numerous studies to exert a favorable effect in patients with subclinical atherosclerosis, metabolic syndrome, hypertension, peripheral vascular disease, stroke and several other cardiovascular disorders, although the obtained results are sometimes inconsistent, which warrants further studies focusing on its bioactivity.
Collapse
Affiliation(s)
- Ioana Mozos
- Department of Functional Sciences, “Victor Babes” University of Medicine and Pharmacy, Timiṣoara, Romania
- Center for Translational Research and Systems Medicine, “Victor Babes” University of Medicine and Pharmacy, Timiṣoara, Romania
| | - Dana Stoian
- 2nd Department of Internal Medicine, “Victor Babes” University of Medicine and Pharmacy, Timiṣoara, Romania
| | - Alexandru Caraba
- 1st Department of Internal Medicine, “Victor Babes” University of Medicine and Pharmacy, Timiṣoara, Romania
| | | | - Jarosław O. Horbańczuk
- Institute of Genetics and Animal Breeding, Polish Academy of Sciences, Magdalenka, Poland
| | - Atanas G. Atanasov
- Institute of Genetics and Animal Breeding, Polish Academy of Sciences, Magdalenka, Poland
- Department of Pharmacognosy, Faculty of Life Sciences, University of Vienna, Vienna, Austria
| |
Collapse
|
5
|
Ogata F, Fujiu K, Koshima I, Nagai R, Manabe I. Phenotypic modulation of smooth muscle cells in lymphoedema. Br J Dermatol 2015; 172:1286-93. [DOI: 10.1111/bjd.13482] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2014] [Indexed: 01/22/2023]
Affiliation(s)
- F. Ogata
- Department of Cardiovascular Medicine; Graduate School of Medicine; University of Tokyo; 7-3-1 Hongo Bunkyo Tokyo 113-8655 Japan
- Department of Plastic, Reconstructive and Aesthetic Surgery; Graduate School of Medicine; University of Tokyo; 7-3-1 Hongo Bunkyo Tokyo 113-8655 Japan
| | - K. Fujiu
- Department of Cardiovascular Medicine; Graduate School of Medicine; University of Tokyo; 7-3-1 Hongo Bunkyo Tokyo 113-8655 Japan
- Translational Systems Biology and Medicine Initiative; Graduate School of Medicine; University of Tokyo; 7-3-1 Hongo Bunkyo Tokyo 113-8655 Japan
| | - I. Koshima
- Department of Plastic, Reconstructive and Aesthetic Surgery; Graduate School of Medicine; University of Tokyo; 7-3-1 Hongo Bunkyo Tokyo 113-8655 Japan
| | - R. Nagai
- Jichi Medical University; 3311-1 Yakushiji Shimotsuke-shi Tochigi-ken 329-0498 Japan
| | - I. Manabe
- Department of Cardiovascular Medicine; Graduate School of Medicine; University of Tokyo; 7-3-1 Hongo Bunkyo Tokyo 113-8655 Japan
| |
Collapse
|
6
|
Okada H, Takemura G, Kanamori H, Tsujimoto A, Goto K, Kawamura I, Watanabe T, Morishita K, Miyazaki N, Tanaka T, Ushikoshi H, Kawasaki M, Miyazaki T, Suzui N, Nishigaki K, Mikami A, Ogura S, Minatoguchi S. Phenotype and physiological significance of the endocardial smooth muscle cells in human failing hearts. Circ Heart Fail 2014; 8:149-55. [PMID: 25466765 DOI: 10.1161/circheartfailure.114.001746] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Extravascular smooth muscle cells are often observed in the endocardium of human failing hearts. Here, we characterized the phenotype of those cells and investigated their physiological significance. METHODS AND RESULTS We examined left ventricular biopsy specimens obtained from 44 patients with dilated cardiomyopathy and 6 nonfailing hearts. In Masson trichrome-stained histological preparations, bundles of smooth muscle cells were seen localized in the endocardium in 23 of the 44 specimens (none of the 6 controls). These cells were immunopositive for α-smooth muscle actin, type 2 smooth muscle myosin, desmin, and calponin, but were negative for embryonic smooth muscle myosin, vimentin, fibronectin, and periostin. This profile is indicative of a late differentiation (contractile) smooth muscle phenotype. Electron microscopy confirmed that phenotype, revealing the cells to contain abundant myofilaments with dense bodies but little rough endoplasmic reticulum or Golgi apparatus. In the endocardial smooth muscle-positive group, the left ventricular end-systolic volume index (73±34 versus 105±50 mL/m(2); P=0.021), left ventricular peak wall stress (164±47 versus 196±43 dynes 10(3)/cm(2); P=0.023), and left ventricular end-systolic meridional wall stress (97±38 versus 121±37 dynes 10(3)/cm(2); P=0.036) were all significantly smaller, and the ejection fraction was larger (41±8.8 versus 33±9.3%; P=0.005) than in the endocardial smooth muscle-negative group. However, no histological parameters differed between the 2 groups. CONCLUSIONS Endocardial smooth muscle cell bundles in hearts with dilated cardiomyopathy exhibit a mature contractile phenotype and may play a compensatory role mitigating heart failure by reducing left ventricular wall stress and systolic dysfunction.
Collapse
Affiliation(s)
- Hideshi Okada
- From the Departments of Emergency and Disaster Medicine (H.O., K.M., H.U., S.O.) and Cardiology (H.K., A.T., K.G., I.K., T.W., N.M., T.T., M.K., K.N., A.M., S.M.), Gifu University Graduate School of Medicine, Gifu, Japan; Department of Internal Medicine, Asahi University, Mizuho, Japan (G.T.); and Division of Pathology, Gifu University Hospital, Gifu, Japan (T.M., N.S.)
| | - Genzou Takemura
- From the Departments of Emergency and Disaster Medicine (H.O., K.M., H.U., S.O.) and Cardiology (H.K., A.T., K.G., I.K., T.W., N.M., T.T., M.K., K.N., A.M., S.M.), Gifu University Graduate School of Medicine, Gifu, Japan; Department of Internal Medicine, Asahi University, Mizuho, Japan (G.T.); and Division of Pathology, Gifu University Hospital, Gifu, Japan (T.M., N.S.).
| | - Hiromitsu Kanamori
- From the Departments of Emergency and Disaster Medicine (H.O., K.M., H.U., S.O.) and Cardiology (H.K., A.T., K.G., I.K., T.W., N.M., T.T., M.K., K.N., A.M., S.M.), Gifu University Graduate School of Medicine, Gifu, Japan; Department of Internal Medicine, Asahi University, Mizuho, Japan (G.T.); and Division of Pathology, Gifu University Hospital, Gifu, Japan (T.M., N.S.)
| | - Akiko Tsujimoto
- From the Departments of Emergency and Disaster Medicine (H.O., K.M., H.U., S.O.) and Cardiology (H.K., A.T., K.G., I.K., T.W., N.M., T.T., M.K., K.N., A.M., S.M.), Gifu University Graduate School of Medicine, Gifu, Japan; Department of Internal Medicine, Asahi University, Mizuho, Japan (G.T.); and Division of Pathology, Gifu University Hospital, Gifu, Japan (T.M., N.S.)
| | - Kazuko Goto
- From the Departments of Emergency and Disaster Medicine (H.O., K.M., H.U., S.O.) and Cardiology (H.K., A.T., K.G., I.K., T.W., N.M., T.T., M.K., K.N., A.M., S.M.), Gifu University Graduate School of Medicine, Gifu, Japan; Department of Internal Medicine, Asahi University, Mizuho, Japan (G.T.); and Division of Pathology, Gifu University Hospital, Gifu, Japan (T.M., N.S.)
| | - Itta Kawamura
- From the Departments of Emergency and Disaster Medicine (H.O., K.M., H.U., S.O.) and Cardiology (H.K., A.T., K.G., I.K., T.W., N.M., T.T., M.K., K.N., A.M., S.M.), Gifu University Graduate School of Medicine, Gifu, Japan; Department of Internal Medicine, Asahi University, Mizuho, Japan (G.T.); and Division of Pathology, Gifu University Hospital, Gifu, Japan (T.M., N.S.)
| | - Takatomo Watanabe
- From the Departments of Emergency and Disaster Medicine (H.O., K.M., H.U., S.O.) and Cardiology (H.K., A.T., K.G., I.K., T.W., N.M., T.T., M.K., K.N., A.M., S.M.), Gifu University Graduate School of Medicine, Gifu, Japan; Department of Internal Medicine, Asahi University, Mizuho, Japan (G.T.); and Division of Pathology, Gifu University Hospital, Gifu, Japan (T.M., N.S.)
| | - Kentaro Morishita
- From the Departments of Emergency and Disaster Medicine (H.O., K.M., H.U., S.O.) and Cardiology (H.K., A.T., K.G., I.K., T.W., N.M., T.T., M.K., K.N., A.M., S.M.), Gifu University Graduate School of Medicine, Gifu, Japan; Department of Internal Medicine, Asahi University, Mizuho, Japan (G.T.); and Division of Pathology, Gifu University Hospital, Gifu, Japan (T.M., N.S.)
| | - Nagisa Miyazaki
- From the Departments of Emergency and Disaster Medicine (H.O., K.M., H.U., S.O.) and Cardiology (H.K., A.T., K.G., I.K., T.W., N.M., T.T., M.K., K.N., A.M., S.M.), Gifu University Graduate School of Medicine, Gifu, Japan; Department of Internal Medicine, Asahi University, Mizuho, Japan (G.T.); and Division of Pathology, Gifu University Hospital, Gifu, Japan (T.M., N.S.)
| | - Toshiki Tanaka
- From the Departments of Emergency and Disaster Medicine (H.O., K.M., H.U., S.O.) and Cardiology (H.K., A.T., K.G., I.K., T.W., N.M., T.T., M.K., K.N., A.M., S.M.), Gifu University Graduate School of Medicine, Gifu, Japan; Department of Internal Medicine, Asahi University, Mizuho, Japan (G.T.); and Division of Pathology, Gifu University Hospital, Gifu, Japan (T.M., N.S.)
| | - Hiroaki Ushikoshi
- From the Departments of Emergency and Disaster Medicine (H.O., K.M., H.U., S.O.) and Cardiology (H.K., A.T., K.G., I.K., T.W., N.M., T.T., M.K., K.N., A.M., S.M.), Gifu University Graduate School of Medicine, Gifu, Japan; Department of Internal Medicine, Asahi University, Mizuho, Japan (G.T.); and Division of Pathology, Gifu University Hospital, Gifu, Japan (T.M., N.S.)
| | - Masanori Kawasaki
- From the Departments of Emergency and Disaster Medicine (H.O., K.M., H.U., S.O.) and Cardiology (H.K., A.T., K.G., I.K., T.W., N.M., T.T., M.K., K.N., A.M., S.M.), Gifu University Graduate School of Medicine, Gifu, Japan; Department of Internal Medicine, Asahi University, Mizuho, Japan (G.T.); and Division of Pathology, Gifu University Hospital, Gifu, Japan (T.M., N.S.)
| | - Tatsuhiko Miyazaki
- From the Departments of Emergency and Disaster Medicine (H.O., K.M., H.U., S.O.) and Cardiology (H.K., A.T., K.G., I.K., T.W., N.M., T.T., M.K., K.N., A.M., S.M.), Gifu University Graduate School of Medicine, Gifu, Japan; Department of Internal Medicine, Asahi University, Mizuho, Japan (G.T.); and Division of Pathology, Gifu University Hospital, Gifu, Japan (T.M., N.S.)
| | - Natsuko Suzui
- From the Departments of Emergency and Disaster Medicine (H.O., K.M., H.U., S.O.) and Cardiology (H.K., A.T., K.G., I.K., T.W., N.M., T.T., M.K., K.N., A.M., S.M.), Gifu University Graduate School of Medicine, Gifu, Japan; Department of Internal Medicine, Asahi University, Mizuho, Japan (G.T.); and Division of Pathology, Gifu University Hospital, Gifu, Japan (T.M., N.S.)
| | - Kazuhiko Nishigaki
- From the Departments of Emergency and Disaster Medicine (H.O., K.M., H.U., S.O.) and Cardiology (H.K., A.T., K.G., I.K., T.W., N.M., T.T., M.K., K.N., A.M., S.M.), Gifu University Graduate School of Medicine, Gifu, Japan; Department of Internal Medicine, Asahi University, Mizuho, Japan (G.T.); and Division of Pathology, Gifu University Hospital, Gifu, Japan (T.M., N.S.)
| | - Atsushi Mikami
- From the Departments of Emergency and Disaster Medicine (H.O., K.M., H.U., S.O.) and Cardiology (H.K., A.T., K.G., I.K., T.W., N.M., T.T., M.K., K.N., A.M., S.M.), Gifu University Graduate School of Medicine, Gifu, Japan; Department of Internal Medicine, Asahi University, Mizuho, Japan (G.T.); and Division of Pathology, Gifu University Hospital, Gifu, Japan (T.M., N.S.)
| | - Shinji Ogura
- From the Departments of Emergency and Disaster Medicine (H.O., K.M., H.U., S.O.) and Cardiology (H.K., A.T., K.G., I.K., T.W., N.M., T.T., M.K., K.N., A.M., S.M.), Gifu University Graduate School of Medicine, Gifu, Japan; Department of Internal Medicine, Asahi University, Mizuho, Japan (G.T.); and Division of Pathology, Gifu University Hospital, Gifu, Japan (T.M., N.S.)
| | - Shinya Minatoguchi
- From the Departments of Emergency and Disaster Medicine (H.O., K.M., H.U., S.O.) and Cardiology (H.K., A.T., K.G., I.K., T.W., N.M., T.T., M.K., K.N., A.M., S.M.), Gifu University Graduate School of Medicine, Gifu, Japan; Department of Internal Medicine, Asahi University, Mizuho, Japan (G.T.); and Division of Pathology, Gifu University Hospital, Gifu, Japan (T.M., N.S.)
| |
Collapse
|
7
|
Zhang J, Chen J, Xu C, Yang J, Guo Q, Hu Q, Jiang H. Resveratrol inhibits phenotypic switching of neointimal vascular smooth muscle cells after balloon injury through blockade of Notch pathway. J Cardiovasc Pharmacol 2014; 63:233-239. [PMID: 24603118 DOI: 10.1097/fjc.0000000000000040] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Phenotypic switching of vascular smooth muscle cells (VSMCs) plays an initial role in neointimal hyperplasia, the main cause of many occlusive vascular diseases. The aim of this study was to measure the effects of resveratrol (RSV) on the phenotypic transformation of VSMCs and to investigate its mechanism of action. METHODS Cultured VSMCs isolated from rat thoracic aorta were prepared with serum starvation for 72 hours followed by RSV treatment (50-200 μmol/L) and 10% serum stimulation. Male Sprague-Dawley rats, subjected to carotid arteries injury from a balloon catheter, were exposed to intraperitoneal injection of RSV (1 mg/kg) or saline and were killed after 7 or 28 days. RESULTS Compared with cells in the serum-induced group, VSMCs in the RSV or N-[N-(3, 5-Difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester (DAPT) treatment group exhibited significant decreases of proliferation and migration. The total and cytoplasmic Notch-1 levels were declined by RSV, accompanied by a significant increase in smooth muscle α-actin and smooth muscle myosin heavy chain protein. The expression of Notch-1, Jagged-1, Hey-1, and Hey-2 mRNA in balloon-injured arteries at 7 days was decreased by RSV treatment. Arteries from RSV-treated rats showed less neointimal hyperplasia, lower collagen content, and a lower rate of cells positive for proliferating cell nuclear antigen 28 days after injury, compared with saline controls. CONCLUSIONS The results indicate that RSV can attenuate phenotypic switching of VSMCs after arterial injury through inhibition of the Notch pathway.
Collapse
MESH Headings
- Animals
- Antioxidants/pharmacology
- Aorta, Thoracic/cytology
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/pathology
- Carotid Arteries/drug effects
- Carotid Arteries/pathology
- Carotid Artery Injuries/metabolism
- Carotid Artery Injuries/pathology
- Cells, Cultured
- Disease Models, Animal
- Hyperplasia/prevention & control
- Male
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Neointima/prevention & control
- Rats
- Rats, Sprague-Dawley
- Receptor, Notch1/genetics
- Resveratrol
- Stilbenes/pharmacology
- Time Factors
Collapse
Affiliation(s)
- Jing Zhang
- *Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; †Departments of Cardiology; and ‡Ophthalmology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, China
| | | | | | | | | | | | | |
Collapse
|
8
|
Chen F, Marquez H, Kim YK, Qian J, Shao F, Fine A, Cruikshank WW, Quadro L, Cardoso WV. Prenatal retinoid deficiency leads to airway hyperresponsiveness in adult mice. J Clin Invest 2014; 124:801-11. [PMID: 24401276 DOI: 10.1172/jci70291] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Accepted: 10/30/2013] [Indexed: 01/02/2023] Open
Abstract
There is increasing evidence that vitamin A deficiency in utero correlates with abnormal airway smooth muscle (SM) function in postnatal life. The bioactive vitamin A metabolite retinoic acid (RA) is essential for formation of the lung primordium; however, little is known about the impact of early fetal RA deficiency on postnatal lung structure and function. Here, we provide evidence that during murine lung development, endogenous RA has a key role in restricting the airway SM differentiation program during airway formation. Using murine models of pharmacological, genetic, and dietary vitamin A/RA deficiency, we found that disruption of RA signaling during embryonic development consistently resulted in an altered airway SM phenotype with markedly increased expression of SM markers. The aberrant phenotype persisted postnatally regardless of the adult vitamin A status and manifested as structural changes in the bronchial SM and hyperresponsiveness of the airway without evidence of inflammation. Our data reveal a role for endogenous RA signaling in restricting SM differentiation and preventing precocious and excessive SM differentiation when airways are forming.
Collapse
|
9
|
Shen H, Eguchi K, Kono N, Fujiu K, Matsumoto S, Shibata M, Oishi-Tanaka Y, Komuro I, Arai H, Nagai R, Manabe I. Saturated fatty acid palmitate aggravates neointima formation by promoting smooth muscle phenotypic modulation. Arterioscler Thromb Vasc Biol 2013; 33:2596-607. [PMID: 23968977 DOI: 10.1161/atvbaha.113.302099] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Obesity is a major risk factor of atherosclerotic cardiovascular disease. Circulating free fatty acid levels are known to be elevated in obese individuals and, along with dietary saturated fatty acids, are known to associate with cardiovascular events. However, little is known about the molecular mechanisms by which free fatty acids are linked to cardiovascular disease. APPROACH AND RESULTS We found that administration of palmitate, a major saturated free fatty acid, to mice markedly aggravated neointima formation induced by carotid artery ligation and that the neointima primarily consisted of phenotypically modulated smooth muscle cells (SMCs). In cultured SMCs, palmitate-induced phenotypic modulation was characterized by downregulation of SMC differentiation markers, such as SM α-actin and SM-myosin heavy chain, and upregulation of mediators involved in inflammation and remodeling of the vessel wall, such as platelet-derived growth factor B and matrix metalloproteinases. We also found that palmitate induced the expression of proinflammatory genes via a novel toll-like receptor 4/myeloid differentiation primary response 88/nuclear factor-κB/NADPH oxidase 1/reactive oxygen species signaling pathway: nuclear factor-κB was activated by palmitate via toll-like receptor 4 and its adapter, MyD88, and once active, it transactivated Nox1, encoding NADPH oxidase 1, a major reactive oxygen species generator in SMCs. Pharmacological inhibition and small interfering RNA-mediated knockdown of the components of this signaling pathway mitigated the palmitate-induced upregulation of proinflammatory genes. More importantly, Myd88 knockout mice were resistant to palmitate-induced exacerbation of neointima formation. CONCLUSIONS Palmitate seems to promote neointima formation by inducing inflammatory phenotypes in SMCs.
Collapse
Affiliation(s)
- Hua Shen
- From the Department of Cardiovascular Medicine (H.S., K.E., K.F., S.M., M.S., I.K., I.M.), Translational Systems Biology and Medicine Initiative (K.F.), Graduate School of Medicine, and Graduate School of Pharmaceutical Sciences (N.K., H.A.), The University of Tokyo, Tokyo, Japan; Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan (Y.O.-T.); and Jichi Medical University, Tochigi, Japan (R.N.)
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Joghetaei N, Stein A, Byrne RA, Schulz C, King L, May AE, Schmidt R. The Extracellular Matrix Metalloproteinase Inducer (EMMPRIN, CD147) - a potential novel target in atherothrombosis prevention? Thromb Res 2013; 131:474-80. [DOI: 10.1016/j.thromres.2013.04.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Revised: 04/10/2013] [Accepted: 04/16/2013] [Indexed: 11/17/2022]
|
11
|
Chen G, Wu L, Deng CQ. The effects of BuYang HuanWu Decoction and its effective components on proliferation-related factors and ERK1/2 signal transduction pathway in cultured vascular smooth muscle cells. JOURNAL OF ETHNOPHARMACOLOGY 2011; 135:7-14. [PMID: 21385603 DOI: 10.1016/j.jep.2011.02.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2010] [Revised: 01/19/2011] [Accepted: 02/11/2011] [Indexed: 05/30/2023]
Abstract
Buyang Huanwu Decoction (BYHWD) was a commonly used traditional Chinese medicine for the treatment and prevention of ischemic cardiovascular and cerebral disease. Previous studies had shown that BYHWD alkaloids and glycosides could inhibit intimal hyperplasia and vascular smooth muscle cell (VSMC) proliferation after injury caused by balloon catheter. The present study aims to explore the mechanisms by which cell cycle was affected by BYHWD and its components. Primary rat VSMC was treated with platelet-derived growth factor (PDGF) and cell cycle phase and extracellular-signal regulated protein kinase (ERK) transduction pathway factors were measured. PDGF-treated cells were associated with a significant increase in the number of cells in the G(2)/M phase and S phase, and in the expression of P-ERK1/2, proliferating cell nuclear antigen (PCNA), c-fos, cyclinD(1) and cyclin-dependent kinase-4, as well as a decrease in the number of cells in the G(0)/G(1) phase, and in the expression of cyclin-dependent kinase inhibitor P21 protein and mitogen-activated protein kinase phosphatase-1 (MKP-1). Treatment with plasma of rats fed seven doses of BYHWD crude extract (22.2g/kg), BYHWD alkaloids (1.66g/kg), BYHWD glycosides (14.2g/kg) or the negative control atorvastatin (20mg/kg) inhibited these changes. All drug-containing plasma had similar activity to the mitogen activated protein kinase (MAPK)/ERK antagonist PD098059 which inhibited PDGF-induced expression of P-ERK1/2 and enhanced MKP-1. These suggest that BYHWD and its components may prevent VSMC proliferation by interfering with the ERK transduction pathway.
Collapse
MESH Headings
- Alkaloids/pharmacology
- Animals
- Atorvastatin
- Cardiovascular Agents/pharmacology
- Cell Cycle/drug effects
- Cell Proliferation/drug effects
- Cells, Cultured
- Cyclin-Dependent Kinase Inhibitor p21/metabolism
- Drugs, Chinese Herbal/chemistry
- Drugs, Chinese Herbal/pharmacology
- Dual Specificity Phosphatase 1/metabolism
- Enzyme Inhibitors/pharmacology
- Extracellular Signal-Regulated MAP Kinases/metabolism
- Flavonoids/pharmacology
- Glycosides/pharmacology
- Heptanoic Acids/pharmacology
- Male
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Platelet-Derived Growth Factor/metabolism
- Pyrroles/pharmacology
- Rats
- Rats, Sprague-Dawley
- Signal Transduction/drug effects
Collapse
Affiliation(s)
- Gang Chen
- Pathophysiology Laboratory, Hunan University of Traditional Chinese Medicine, Shaoshan Road No. 113, Changsha, Hunan, China
| | | | | |
Collapse
|
12
|
Wang T, Zhang M, Han Y, Zhang X, Yan C, Liang Z, Sun Y, Kang J. Lack of association between cellular repressor of E1A-stimulated genes (GREG) polymorphisms and coronary artery disease in the Han population of North China. Clin Chim Acta 2011; 412:249-52. [DOI: 10.1016/j.cca.2010.10.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2010] [Revised: 10/08/2010] [Accepted: 10/09/2010] [Indexed: 12/01/2022]
|
13
|
Iwata H, Manabe I, Fujiu K, Yamamoto T, Takeda N, Eguchi K, Furuya A, Kuro-o M, Sata M, Nagai R. Bone marrow-derived cells contribute to vascular inflammation but do not differentiate into smooth muscle cell lineages. Circulation 2010; 122:2048-57. [PMID: 21041690 DOI: 10.1161/circulationaha.110.965202] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND It has been proposed that bone marrow-derived cells infiltrate the neointima, where they differentiate into smooth muscle (SM) cells; however, technical limitations have hindered clear identification of the lineages of bone marrow-derived "SM cell-like" cells. METHODS AND RESULTS Using a specific antibody against the definitive SM cell lineage marker SM myosin heavy chain (SM-MHC) and mouse lines in which reporter genes were driven by regulatory programs for either SM-MHC or SM α-actin, we demonstrated that although some bone marrow-derived cells express SM α-actin in the wire injury-induced neointima, those cells did not express SM-MHC, even 30 weeks after injury. Likewise, no SM-MHC(+) bone marrow-derived cells were found in vascular lesions in apolipoprotein E(-/-)mice or in a heart transplantation vasculopathy model. Instead, the majority of bone marrow-derived SM α-actin(+) cells were also CD115(+)CD11b(+)F4/80(+)Ly-6C(+), which is the surface phenotype of inflammatory monocytes. Moreover, adoptively transferred CD11b(+)Ly-6C(+) bone marrow cells expressed SM α-actin in the injured artery. Expression of inflammation-related genes was significantly higher in neointimal subregions rich in bone marrow-derived SM α-actin(+) cells than in other regions. CONCLUSIONS It appears that bone marrow-derived SM α-actin(+) cells are of monocyte/macrophage lineage and are involved in vascular remodeling. It is very unlikely that these cells acquire the definitive SM cell lineage.
Collapse
Affiliation(s)
- Hiroshi Iwata
- Department of Cardiovascular Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Wu L, Zhang W, Li H, Zhang GM, Chen BY, Tang YH, Deng CQ. [Effects of Buyang Huanwu Decoction and its alkaloids and glycosides on aortic intimal hyperplasia and expression of proliferating cell nuclear antigen in rats with aortic intimal injuries]. ACTA ACUST UNITED AC 2010; 6:836-42. [PMID: 18664354 DOI: 10.3736/jcim20080813] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
OBJECTIVE To explore the effects of alkaloids and glycosides extracted from Buyang Huanwu Decoction (BYHWD), a compound of traditional Chinese herbal medicine, on aortic intimal hyperplasia and the expression of the proliferating cell nuclear antigen (PCNA) in rats with aortic intimal injuries. METHODS The vessel restenosis model was established by denuding aortic endothelium with domestic balloon catheter in rats. Drugs were administered intragastrically on the first day after operation. The injured segments of aorta were taken on the fifteenth day after operation to determine the degree of intimal hyperplasia and observe the expression of PCNA. RESULTS Aortic intimal hyperplasias were very obvious on the fifteenth day after operation. The media hyperplasias in the drug-treated groups were not significant (P>0.05), but the intimal hyperplasia were remarkable as compared with that in the sham-operated group (P<0.01). The degrees of intimal hyperplasia in BYHWD, alkaloids, glycosides and atorvastatin groups were less than that in the untreated group (P<0.01). There was significant difference of PCNA expression between the untreated group and the sham-operated group (P<0.01). The expressions of PCNA in alkaloids, glycosides, and atorvastatin groups were higher than that in the sham-operated group (P<0.01, P<0.05), but still lower than that in the untreated group (P<0.01). The expression of PCNA in BYHWD group was higher than that in the sham-operated group (P<0.01), and no significant difference was found between the BYHWD group and the untreated group (P>0.05). CONCLUSION Alkaloids and glycosides extracted from BYHWD can inhibit intimal hyperplasia induced by denuding arterial endothelium with domestic balloon catheter in rats. Alkaloids and glycoside may be among basal substances in BYHWD inhibiting intimal hyperplasia of blood vessel, the effect of which may relate to down-regulating the expression of PCNA.
Collapse
Affiliation(s)
- Lu Wu
- Laboratory of Pathophysiology, Hunan University of Traditional Chinese Medicine, Changsha, Hunan Province 410007, China
| | | | | | | | | | | | | |
Collapse
|
15
|
Gratzke C, Angulo J, Chitaley K, Dai YT, Kim NN, Paick JS, Simonsen U, Uckert S, Wespes E, Andersson KE, Lue TF, Stief CG. Anatomy, physiology, and pathophysiology of erectile dysfunction. J Sex Med 2010; 7:445-75. [PMID: 20092448 DOI: 10.1111/j.1743-6109.2009.01624.x] [Citation(s) in RCA: 247] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
INTRODUCTION Significant scientific advances during the past 3 decades have deepened our understanding of the physiology and pathophysiology of penile erection. A critical evaluation of the current state of knowledge is essential to provide perspective for future research and development of new therapies. AIM To develop an evidence-based, state-of-the-art consensus report on the anatomy, physiology, and pathophysiology of erectile dysfunction (ED). METHODS Consensus process over a period of 16 months, representing the opinions of 12 experts from seven countries. MAIN OUTCOME MEASURE Expert opinion was based on the grading of scientific and evidence-based medical literature, internal committee discussion, public presentation, and debate. RESULTS ED occurs from multifaceted, complex mechanisms that can involve disruptions in neural, vascular, and hormonal signaling. Research on central neural regulation of penile erection is progressing rapidly with the identification of key neurotransmitters and the association of neural structures with both spinal and supraspinal pathways that regulate sexual function. In parallel to advances in cardiovascular physiology, the most extensive efforts in the physiology of penile erection have focused on elucidating mechanisms that regulate the functions of the endothelium and vascular smooth muscle of the corpus cavernosum. Major health concerns such as atherosclerosis, hyperlipidemia, hypertension, diabetes, and metabolic syndrome (MetS) have become well integrated into the investigation of ED. CONCLUSIONS Despite the efficacy of current therapies, they remain insufficient to address growing patient populations, such as those with diabetes and MetS. In addition, increasing awareness of the adverse side effects of commonly prescribed medications on sexual function provides a rationale for developing new treatment strategies that minimize the likelihood of causing sexual dysfunction. Many basic questions with regard to erectile function remain unanswered and further laboratory and clinical studies are necessary.
Collapse
Affiliation(s)
- Christian Gratzke
- Department of Urology, Ludwig-Maximilians-Universität, München, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Oishi Y, Manabe I, Imai Y, Hara K, Horikoshi M, Fujiu K, Tanaka T, Aizawa T, Kadowaki T, Nagai R. Regulatory polymorphism in transcription factor KLF5 at the MEF2 element alters the response to angiotensin II and is associated with human hypertension. FASEB J 2010; 24:1780-8. [PMID: 20086047 DOI: 10.1096/fj.09-146589] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Krüppel-like factor 5 (KLF5) is a zinc-finger-type transcription factor that mediates the tissue remodeling in cardiovascular diseases, such as atherosclerosis, restenosis, and cardiac hypertrophy. Our previous studies have shown that KLF5 is induced by angiotensin II (AII), although the precise molecular mechanism is not yet known. Here we analyzed regulatory single nucleotide polymorphisms (SNPs) within the KLF5 locus to identify clinically relevant signaling pathways linking AII and KLF5. One SNP was located at -1282 bp and was associated with an increased risk of hypertension: subjects with the A/A and A/G genotypes at -1282 were at significantly higher risk for hypertension than those with the G/G genotype. Interestingly, a reporter construct corresponding to the -1282G genotype showed much weaker responses to AII than a construct corresponding to -1282A. Electrophoretic mobility shift, chromatin immunoprecipitation, and reporter assays collectively showed that the -1282 SNP is located within a functional myocyte enhancer factor 2 (MEF2) binding site, and that the -1282G genotype disrupts the site and reduces the AII responsiveness of the promoter. Moreover, MEF2 activation via reactive oxygen species and p38 mitogen-activated protein kinase induced KLF5 expression in response to AII, and KLF5 and MEF2 were coexpressed in coronary atherosclerotic plaques. These results suggest that a novel signaling and transcription network involving MEF2A and KLF5 plays an important role in the pathogenesis of cardiovascular diseases such as hypertension.
Collapse
Affiliation(s)
- Yumiko Oishi
- Department of Cardiovascular Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Grenier S, Sandig M, Mequanint K. Smooth muscle alpha-actin and calponin expression and extracellular matrix production of human coronary artery smooth muscle cells in 3D scaffolds. Tissue Eng Part A 2009; 15:3001-11. [PMID: 19323608 DOI: 10.1089/ten.tea.2009.0057] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
For a tissue-engineered coronary artery substitute to be a viable clinical option in the treatment of vascular diseases, it is necessary to use tissue-specific human cells. Coronary artery smooth muscle cells are the main resident cells in the tunica media of arteries. In this work, we examined the behavior and differentiation state of human coronary artery smooth muscle cells (HCASMCs) when cultured on 3D polyurethane scaffolds to fabricate hybrid vascular tissues. As the mechanical strength of the scaffold is an important element in engineered hybrid vascular substitutes, porous 3D polyurethane scaffolds fabricated using paraffin spheres and ammonium chloride particles were tested for their mechanical properties both in tension and in compression. The use of ammonium chloride particles as porogen generated scaffolds with superior mechanical properties, which are suitable for vascular tissue engineering. When seeded on uncoated, fibronectin-coated, and Matrigel-coated scaffolds, HCASMCs were well spread and started producing collagen as judged by histochemical analysis but appeared to lack elastin production. Fibronectin coating appeared to promote the infiltration of HCASMCs into the scaffold better than Matrigel coating but did not appear to affect the expression of collagen and elastin. Western blot analyses after successful cell recovery from the scaffolds indicated that HCASMCs, after culturing for 4 and 7 days, expressed similar amounts of smooth muscle alpha-actin and calponin regardless of extracellular matrix coating. Taken together, our data showed that the behavior and differentiation phenotype of HCASMCs can be analyzed after culture in 3D polyurethane scaffolds to establish appropriate conditions that will favor the fabrication of hybrid-engineered vascular substitutes.
Collapse
Affiliation(s)
- Stephanie Grenier
- The Graduate Program of Biomedical Engineering, The University of Western Ontario, London, Ontario, Canada
| | | | | |
Collapse
|
18
|
Zhang XH, Zheng B, Han M, Miao SB, Wen JK. Synthetic retinoid Am80 inhibits interaction of KLF5 with RARα through inducing KLF5 dephosphorylation mediated by the PI3K/Akt signaling in vascular smooth muscle cells. FEBS Lett 2009; 583:1231-6. [DOI: 10.1016/j.febslet.2009.03.016] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2008] [Revised: 02/22/2009] [Accepted: 03/10/2009] [Indexed: 11/30/2022]
|
19
|
Jiang M, Bujo H, Ohwaki K, Unoki H, Yamazaki H, Kanaki T, Shibasaki M, Azuma K, Harigaya K, Schneider WJ, Saito Y. Ang II-stimulated migration of vascular smooth muscle cells is dependent on LR11 in mice. J Clin Invest 2008; 118:2733-46. [PMID: 18618022 DOI: 10.1172/jci32381] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2007] [Accepted: 05/21/2008] [Indexed: 11/17/2022] Open
Abstract
Medial-to-intimal migration of SMCs is critical to atherosclerotic plaque formation and remodeling of injured arteries. Considerable amounts of the shed soluble form of the LDL receptor relative LR11 (sLR11) produced by intimal SMCs enhance SMC migration in vitro via upregulation of urokinase-type plasminogen activator receptor (uPAR) expression. Here, we show that circulating sLR11 is a novel marker of carotid intima-media thickness (IMT) and that targeted disruption of the LR11 gene greatly reduces intimal thickening of arteries through attenuation of Ang II-induced migration of SMCs. Serum concentrations of sLR11 were positively correlated with IMT in dyslipidemic subjects, and multivariable regression analysis suggested sLR11 levels as an index of IMT, independent of classical atherosclerosis risk factors. In Lr11-/- mice, femoral artery intimal thickness after cuff placement was decreased, and Ang II-stimulated migration and attachment of SMCs from these mice were largely abolished. In isolated murine SMCs, sLR11 caused membrane ruffle formation via activation of focal adhesion kinase/ERK/Rac1 accompanied by complex formation between uPAR and integrin alphavbeta3, a process accelerated by Ang II. Overproduction of sLR11 decreased the sensitivity of Ang II-induced activation pathways to inhibition by an Ang II type 1 receptor blocker in mice. Thus, we demonstrate a requirement for sLR11 in Ang II-induced SMC migration and propose what we believe is a novel role for sLR11 as a biomarker of carotid IMT.
Collapse
Affiliation(s)
- Meizi Jiang
- Department of Genome Research and Clinical Application, Chiba University Graduate School of Medicine, Chiba, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Ohara F, Nii A, Sakiyama Y, Tsuchiya M, Ogawa S. Pathophysiological characteristics of dimethylnitrosamine-induced liver fibrosis in acute and chronic injury models: a possible contribution of KLF5 to fibrogenic responses. Dig Dis Sci 2008; 53:2222-32. [PMID: 18095165 DOI: 10.1007/s10620-007-0112-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2007] [Accepted: 10/27/2007] [Indexed: 12/11/2022]
Abstract
Dimethylnitrosamine administration induces a rapid increase in collagen deposition with concomitant proliferation of hepatic stellate cells in rats. Here, we investigated the pathophysiological profiles of acute and chronic hepatic fibrosis states and attempted to determine the possible role of Kruppel-like factor-5 (KLF5) in this model. In acute study using a single drug injection, we observed a rapid transient increase of ALT and mRNA levels of KLF5 followed by increases in fibrosis-related genes. Repeated administration of dimethylnitrosamine once a week caused early damage with severe fibrosis and sustained hepatocyte injury, while intermittent injections at 2-week intervals induced only modest fibrosis from 3 weeks. Weekly administration also induced profound upregulation of collagen I, alpha-smooth muscle actin, and KLF5 mRNA. In contrast, such continued augmentation was not observed after intermittent injections; KLF5 increased only after 3 weeks. These results suggested that dimethylnitrosamine induced a rapid hepatic fibrogenic response with a possible participation of KLF5.
Collapse
Affiliation(s)
- Fumihiro Ohara
- St Louis Laboratories, Pfizer Global Research and Development, Chesterfield, MO 63017, USA.
| | | | | | | | | |
Collapse
|
21
|
Abraham S, Kogata N, Fässler R, Adams RH. Integrin β1 Subunit Controls Mural Cell Adhesion, Spreading, and Blood Vessel Wall Stability. Circ Res 2008; 102:562-70. [DOI: 10.1161/circresaha.107.167908] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Sabu Abraham
- From the Cancer Research UK London Research Institute (S.A., N.K., R.H.A.), Vascular Development Laboratory, London, UK; Max-Planck-Institute of Biochemistry (R.F.), Department of Molecular Medicine, Martinsried, Germany; Max-Planck-Institute for Molecular Biomedicine (R.H.A.), Department of Tissue Morphogenesis, and University of Münster, Faculty of Medicine, Münster, Germany
| | - Naoko Kogata
- From the Cancer Research UK London Research Institute (S.A., N.K., R.H.A.), Vascular Development Laboratory, London, UK; Max-Planck-Institute of Biochemistry (R.F.), Department of Molecular Medicine, Martinsried, Germany; Max-Planck-Institute for Molecular Biomedicine (R.H.A.), Department of Tissue Morphogenesis, and University of Münster, Faculty of Medicine, Münster, Germany
| | - Reinhard Fässler
- From the Cancer Research UK London Research Institute (S.A., N.K., R.H.A.), Vascular Development Laboratory, London, UK; Max-Planck-Institute of Biochemistry (R.F.), Department of Molecular Medicine, Martinsried, Germany; Max-Planck-Institute for Molecular Biomedicine (R.H.A.), Department of Tissue Morphogenesis, and University of Münster, Faculty of Medicine, Münster, Germany
| | - Ralf H. Adams
- From the Cancer Research UK London Research Institute (S.A., N.K., R.H.A.), Vascular Development Laboratory, London, UK; Max-Planck-Institute of Biochemistry (R.F.), Department of Molecular Medicine, Martinsried, Germany; Max-Planck-Institute for Molecular Biomedicine (R.H.A.), Department of Tissue Morphogenesis, and University of Münster, Faculty of Medicine, Münster, Germany
| |
Collapse
|
22
|
Abstract
Arterial reconstruction procedures, including balloon angioplasty, stenting and coronary artery bypass, are used to restore blood flow in atherosclerotic arteries. Restenosis of these arteries has remained a major limitation of the application of these procedures, especially in the case of balloon angioplasty. Post-angioplasty restenosis results from two major processes: neointimal formation and constrictive remodelling. Neointimal formation is initiated by arterial injury with a resultant loss of contractile phenotype in tunica media, leading to VSMC [vascular SM (smooth muscle) cell] migration from the tunica media to the intima. Migrated VSMCs contribute to the intimal thickening by the excessive synthesis of ECM (extracellular matrix) and proliferation. However, increased neointimal mass is not solely responsible for luminal narrowing. Inward constrictive remodelling is also considered as a major cause of delayed failure of angioplasty. At later stages after angioplasty, the increase in contractile forces leads to lumen narrowing. Recent studies show that SM contractile proteins are re-expressed in the neointima, concomitant with late lumen loss. Therefore one important question is whether the restoration of contractile phenotype, which can suppress VSMC migration, is favourable or detrimental. In this review, the importance of viewing restenosis as a multistage process is discussed. Different stages of restenosis occur in a sequential manner and are related to each other, but in each stage a different strategy should be taken into consideration to reduce restenosis. Defining the role of each process not only reshapes the current concept, but also helps us to target restenosis with more efficacy.
Collapse
|
23
|
Zargham R, Pepin J, Thibault G. alpha8beta1 Integrin is up-regulated in the neointima concomitant with late luminal loss after balloon injury. Cardiovasc Pathol 2007; 16:212-20. [PMID: 17637429 DOI: 10.1016/j.carpath.2007.01.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2006] [Revised: 01/26/2007] [Accepted: 01/30/2007] [Indexed: 10/23/2022] Open
Abstract
INTRODUCTION Constrictive remodeling of the neointima results in the late lumen loss and restenosis after balloon angioplasty. Intense expression of alpha8beta1 integrin in the contractile state of vascular smooth muscle cells (VSMCs) and in myofibroblasts led us to hypothesize that it might be involved in the process of late constrictive remodeling. METHODS AND RESULTS Balloon injury was used to induce neointima formation in the rat carotid artery. Immunohistochemical analysis and immunoconfocal studies showed that late lumen narrowing was concomitant with the up-regulation of smooth muscle alpha-actin and alpha8 integrin in the neointima. The transforming growth factor-beta (TGF-beta)-induced contractile properties of fibroblasts and VSMCs populated in a three-dimensional collagen matrix was associated with up-regulation of alpha8 integrin. TGF-beta-induced myofibroblastic features in Rat1 fibroblasts were impaired in cells pretreated with a small interference RNA silencing the alpha8 integrin gene. CONCLUSION The close correlation between alpha8 integrin up-regulation in the neointima and late luminal loss and alpha8 integrin being required for contractile properties induced by TGF-beta highlight a possible role for alpha8 integrin in postangioplasty restenosis.
Collapse
Affiliation(s)
- Ramin Zargham
- Institut de recherches cliniques de Montréal, Université de Montréal, Montréal, Québec, Canada
| | | | | |
Collapse
|
24
|
Byun Y, Lee G. Changes in the Expression of Smooth Muscle Myosin Heavy Chain mRNA following Partial Bladder Obstruction or Spinal Cord Injury in Rat: A Preliminary Study. Korean J Urol 2007. [DOI: 10.4111/kju.2007.48.5.522] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Youngmin Byun
- Department of Urology, Dankook University College of Medicine, Cheonan, Korea
| | - Gilho Lee
- Department of Urology, Dankook University College of Medicine, Cheonan, Korea
| |
Collapse
|
25
|
Morrow D, Sweeney C, Birney YA, Guha S, Collins N, Cummins PM, Murphy R, Walls D, Redmond EM, Cahill PA. Biomechanical regulation of hedgehog signaling in vascular smooth muscle cells in vitro and in vivo. Am J Physiol Cell Physiol 2007; 292:C488-96. [PMID: 16943241 DOI: 10.1152/ajpcell.00337.2005] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Hedgehog (Hh) signaling has recently been shown to be both responsive to mechanical loading in vitro and to control vascular development in vivo. We investigated the role of cyclic strain and pulsatile flow in modulating Hh signaling and growth of adult rat vascular smooth muscle cells (SMC) in culture. Exposure of SMC to defined equibiaxial cyclic strain (0% and 10% stretch, 60 cycles/min, for 24 h) significantly decreased sonic hedgehog (Shh) and patched 1 (Ptc1) expression while concurrently inhibiting Gli2-dependent promoter activity and mRNA expression, respectively. Cyclic strain significantly decreased SMC proliferation (cell counts and proliferating cell nuclear antigen expression) concomitant with a marked increase in SMC apoptosis (fluorescence-activated cell sorter analysis, acridine orange staining of apoptotic nuclei and Bax/Bcl-xL ratio). These strain-induced changes in proliferation and apoptosis were significantly attenuated following addition of either recombinant Shh (3.5 μg/ml) or overexpression of the Notch 3 intracellular domain (Notch IC). Further studies using a perfused transcapillary culture system demonstrated a significant decrease in Hh signaling in SMC following exposure of cells to increased pulsatile flow concomitant with a decrease in proliferation and an increase in apoptosis. Finally, the pulsatile flow-induced decreases in Hh signaling were validated in vivo following flow-induced rat carotid arterial remodeling after 28 days. These data suggest that Hh expression is diminished by biomechanical stimulation in vitro and in vivo and thus may play a fundamental role in arterial remodeling and atherogenesis in vivo.
Collapse
MESH Headings
- Animals
- Apoptosis
- Cell Line
- Cell Proliferation
- Hedgehog Proteins/metabolism
- Hedgehog Proteins/pharmacology
- Kruppel-Like Transcription Factors/genetics
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/physiology
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/physiology
- Patched Receptors
- Patched-1 Receptor
- Promoter Regions, Genetic
- RNA, Messenger/metabolism
- Rats
- Receptor, Notch3
- Receptors, Cell Surface/metabolism
- Receptors, Notch/metabolism
- Recombinant Proteins/pharmacology
- Signal Transduction/physiology
- Stress, Mechanical
- Zinc Finger Protein Gli2
Collapse
Affiliation(s)
- David Morrow
- Faculty of Science and Health, Vascular Health Research Centre, Dublin City University, Glasnevin, Dublin 9, Ireland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Nishimura G, Manabe I, Tsushima K, Fujiu K, Oishi Y, Imai Y, Maemura K, Miyagishi M, Higashi Y, Kondoh H, Nagai R. DeltaEF1 mediates TGF-beta signaling in vascular smooth muscle cell differentiation. Dev Cell 2006; 11:93-104. [PMID: 16824956 DOI: 10.1016/j.devcel.2006.05.011] [Citation(s) in RCA: 111] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2005] [Revised: 03/21/2006] [Accepted: 05/10/2006] [Indexed: 10/24/2022]
Abstract
Alteration in the differentiated state of smooth muscle cells (SMCs) is known to be integral to vascular development and the pathogenesis of vascular disease. However, it is still largely unknown how environmental cues translate into transcriptional control of SMC genes. We found that deltaEF1 is upregulated during SMC differentiation and selectively transactivates the promoters of SMC differentiation marker genes, SM alpha-actin and SM myosin heavy chain (SM-MHC). DeltaEF1 physically interacts with SRF and Smad3, resulting in a synergistic activation of SM alpha-actin promoter. Chromatin immunoprecipitation assays and knockdown experiments showed that deltaEF1 is involved in the control of the SMC differentiation programs induced by TGF-beta signaling. Overexpression of deltaEF1 inhibited neointima formation and promoted SMC differentiation, whereas heterozygous deltaEF1 knockout mice exhibited exaggerated neointima formation. It thus appears deltaEF1 mediates SMC differentiation via interaction with SRF and Smad3 during development and in vascular disease.
Collapse
Affiliation(s)
- Go Nishimura
- Department of Cardiovascular Medicine, School of Engineering, The University of Tokyo, Bunkyo, Tokyo 113-8655, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Jiang M, Bujo H, Zhu Y, Yamazaki H, Hirayama S, Kanaki T, Shibasaki M, Takahashi K, Schneider WJ, Saito Y. Pitavastatin attenuates the PDGF-induced LR11/uPA receptor-mediated migration of smooth muscle cells. Biochem Biophys Res Commun 2006; 348:1367-77. [PMID: 16919601 DOI: 10.1016/j.bbrc.2006.07.204] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2006] [Accepted: 07/31/2006] [Indexed: 11/26/2022]
Abstract
Statins, inhibitors of HMG-CoA reductase, elicit various actions on vascular cells including the modulation of proliferation and migration of smooth muscle cells (SMCs). Here, we have elucidated the mechanism by which statins, in particular pitavastatin, attenuate the migration activity of SMCs. The expression of LR11, a member of the LDL receptor family and an enhancer of cell surface localization of urokinase-type plasminogen activator receptor (uPAR), is increased in cultured SMCs by treatment with PDGF-BB. Pitavastatin attenuates the PDGF-BB -induced surface expression of LR11 and uPAR. The increased migration of SMCs observed both upon overexpression of LR11 and via stimulation of secretion of soluble LR11 is not reversed by pitavastatin. In vivo studies showed that the SMCs expressing LR11 in plaques are almost congruent with intimal cells expressing nonmuscle myosin heavy chain (SMemb). Pitavastatin reduced the expression of LR11 and SMemb, and the levels of LR11, uPAR, and SMemb in cultured intimal SMCs were reduced to those seen in medial SMCs. We propose that this statin reduces PDGF-induced migration through the attenuation of the LR11/uPAR system in SMCs. Modulation of the LR11/uPAR system with statins suggests a novel treatment strategy for atherogenesis based on suppression of intimal SMC migration.
Collapse
MESH Headings
- Animals
- Becaplermin
- Cell Movement/drug effects
- Cells, Cultured
- Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology
- Male
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/physiology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/physiology
- Myosin Heavy Chains/metabolism
- Platelet-Derived Growth Factor/antagonists & inhibitors
- Platelet-Derived Growth Factor/pharmacology
- Proto-Oncogene Proteins c-sis
- Quinolines/pharmacology
- Rabbits
- Receptors, Cell Surface/antagonists & inhibitors
- Receptors, Cell Surface/metabolism
- Receptors, LDL/antagonists & inhibitors
- Receptors, LDL/metabolism
- Receptors, Urokinase Plasminogen Activator
Collapse
Affiliation(s)
- Meizi Jiang
- Department of Genome Research and Clinical Application, Chiba University Graduate School of Medicine, Chiba, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Bujo H, Saito Y. Modulation of Smooth Muscle Cell Migration by Members of the Low-Density Lipoprotein Receptor Family. Arterioscler Thromb Vasc Biol 2006; 26:1246-52. [PMID: 16574889 DOI: 10.1161/01.atv.0000219692.78477.17] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Low-density lipoprotein receptor family members (LRs) play a key role in the catabolism of many membrane-associated proteins, such as complexes between proteinases and their receptors, in addition to being involved in lipoprotein metabolism as suspected by the hitherto well-established functions of low-density lipoprotein receptor, in a variety of tissues. Recent studies using receptor-deficient or -overexpressing animals and cells have suggested that certain LRs are important regulators of the migration (and proliferation) of vascular smooth muscle cells (SMCs). LR expression is markedly induced in intimal or medial SMCs during the formation of atherosclerotic lesions. Because LRs can modulate the activity of the urokinase-type plasminogen activator (uPA) receptor and possibly of the platelet-derived growth factor (PDGF) receptor, LRs may influence the migration of SMCs through functional modulation of these membrane receptors. Therefore, SMC migration may be regulated by time-restricted expression of LRs. In agreement with the concept of functional interaction between LRs and membrane signaling receptors, a negative regulator of uPA receptor protein catabolism, LR11, has been identified. Statins modulate the PDGF-induced migration of intimal SMCs via the LR11/uPA receptor cascade. Selective modification of the LRs/uPA receptor/PDGF receptor systems in SMCs may be important for suppression of atherosclerotic plaque formation as well as for preventing intimal thickening after angioplasty.
Collapse
Affiliation(s)
- Hideaki Bujo
- Department of Genome Research and Clinical Application, Chiba University Graduate School of Medicine, Japan.
| | | |
Collapse
|
29
|
Fujiu K, Manabe I, Ishihara A, Oishi Y, Iwata H, Nishimura G, Shindo T, Maemura K, Kagechika H, Shudo K, Nagai R. Synthetic retinoid Am80 suppresses smooth muscle phenotypic modulation and in-stent neointima formation by inhibiting KLF5. Circ Res 2005; 97:1132-41. [PMID: 16224062 DOI: 10.1161/01.res.0000190613.22565.13] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Modulation of smooth muscle cell (SMC) phenotype plays a central role in neointima formation. We recently demonstrated that Am80, a synthetic retinoic acid receptor alpha-specific agonist, inhibits the activity of the transcription factor KLF5, which is essential for neointima formation after vascular injury. In the present study, we aimed to further analyze the mechanism by which Am80 inhibits KLF5 and the effects of inhibiting KLF5 on SMCs and vascular lesion formation, as well as to evaluate potential of Am80 for use in the prevention of in-stent neointima formation. We found that Am80 inhibited both the expression and transcriptional function of KLF5. Of particular interest was our finding that KLF5 forms a transcriptionally active complex with unliganded RAR/RXR heterodimer on the PDGF-A promoter; Am80 disrupts this complex, thereby inhibiting KLF5-dependent transcriptional activation. Knocking down KLF5 using small interfering RNA suppressed serum-induced downregulation of SMC differentiation marker gene expression in cultured SMCs, and haploinsufficiency of KLF5 in mice attenuated phenotypic modulation of SMCs after vascular injury, indicating that KLF5 plays a key role in the control of SMC phenotype. Am80 augmented expression of the SMC differentiation marker genes in culture and within the vessel walls, and oral administration of Am80 significantly inhibited in-stent neointima formation in a rabbit stent-placement model. Taken together, these results demonstrate that KLF5 plays an important role in the control of SMC phenotype after vascular injury and suggest the feasibility of using Am80, delivered systemically and/or with a drug eluting stent, to prevent in-stent neointima formation.
Collapse
MESH Headings
- Actins/genetics
- Animals
- Benzoates/pharmacology
- Benzoates/therapeutic use
- Cell Differentiation/drug effects
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Kruppel-Like Transcription Factors/antagonists & inhibitors
- Male
- Mice
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- NIH 3T3 Cells
- Phenotype
- Platelet-Derived Growth Factor/genetics
- Promoter Regions, Genetic
- Rabbits
- Receptors, Retinoic Acid/chemistry
- Receptors, Retinoic Acid/genetics
- Retinoic Acid Receptor alpha
- Retinoid X Receptor alpha/chemistry
- Retinoid X Receptor alpha/genetics
- Stents/adverse effects
- Tetrahydronaphthalenes/pharmacology
- Tetrahydronaphthalenes/therapeutic use
- Transcription, Genetic/drug effects
- Tunica Intima/drug effects
- Tunica Intima/pathology
Collapse
Affiliation(s)
- Katsuhito Fujiu
- Department of Cardiovascular Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Seki N, Bujo H, Jiang M, Tanaga K, Takahashi K, Yagui K, Hashimoto N, Schneider WJ, Saito Y. LRP1B is a negative modulator of increased migration activity of intimal smooth muscle cells from rabbit aortic plaques. Biochem Biophys Res Commun 2005; 331:964-70. [PMID: 15882972 DOI: 10.1016/j.bbrc.2005.04.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2005] [Indexed: 10/25/2022]
Abstract
The migration of cultured cultured smooth muscle cells (SMCs) is regulated by the time-specific expression of members of the LDL receptor family (LRs). LRP1B, a member of LRs, modulates the catabolism of PDGF beta-receptor, affecting the migration of SMCs. An involvement of PDGF beta-receptor in atherosclerosis is focused because of its abundant expression in intimal SMCs. Here, in order to know a functional significance of LRP1B in the increased migration of intimal SMCs, the functions of three groups of cultured SMCs with different origins in atherosclerotic arteries were studied. Each group of SMCs (central, marginal or medial SMCs) was isolated from explanted pieces of central or marginal area of thickened intima, or media prepared from rabbit aortic plaques. The LRP1B expression levels were significantly decreased in intimal SMCs, particularly in marginal SMCs, compared to medial SMCs. The expression levels of LRP1B in SMCs were negatively correlated with those of PDGF beta-receptor. The level of PDGF beta-receptor-mediated phosphorylation of ERK 1/2 in central SMCs was increased to 5.2-fold with the functional inhibition of LRP1B using anti-LRP1B IgY. The antibody increased the PDGF-BB-stimulated migration and invasion activities in SMCs. The increase in the PDGF beta-receptor-mediated outgrowth activity of SMCs from the explants was also inhibited by the functional inhibition of LRP1B. These results indicate that LRP1B regulated the migration activity of SMCs through the modulation of PDGF beta-receptor-mediated pathway. The regulation of LRP1B expression is possibly involved in the activated migration of intimal SMCs in the course of atherosclerosis.
Collapse
Affiliation(s)
- Naoto Seki
- Department of Clinical Cell Biology (F5), Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Morrow D, Sweeney C, Birney YA, Cummins PM, Walls D, Redmond EM, Cahill PA. Cyclic Strain Inhibits Notch Receptor Signaling in Vascular Smooth Muscle Cells In Vitro. Circ Res 2005; 96:567-75. [PMID: 15705961 DOI: 10.1161/01.res.0000159182.98874.43] [Citation(s) in RCA: 117] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Notch signaling has been shown recently to regulate vascular cell fate in adult cells. By applying a uniform equibiaxial cyclic strain to vascular smooth muscle cells (SMCs), we investigated the role of strain in modulating Notch-mediated growth of SMCs in vitro. Rat SMCs cultured under conditions of defined equibiaxial cyclic strain (0% to 15% stretch; 60 cycles/min; 0 to 24 hours) exhibited a significant temporal and force-dependent reduction in Notch 3 receptor expression, concomitant with a significant reduction in Epstein Barr virus latency C promoter-binding factor-1/recombination signal-binding protein of the Jkappa immunoglobulin gene-dependent Notch target gene promoter activity and mRNA levels when compared with unstrained controls. The decrease in Notch signaling was Gi-protein- and mitogen-activated protein kinase-dependent. In parallel cultures, cyclic strain inhibited SMC proliferation (cell number and proliferating cell nuclear antigen expression) while significantly promoting SMC apoptosis (annexin V binding, caspase-3 activity and bax/bcl-x(L) ratio). Notch 3 receptor overexpression significantly reversed the strain-induced changes in SMC proliferation and apoptosis to levels comparable to unstrained control cells, whereas Notch inhibition further potentiated the changes in SMC apoptosis and proliferation. These findings suggest that cyclic strain inhibits SMC growth while enhancing SMC apoptosis, in part, through regulation of Notch receptor and downstream target gene expression.
Collapse
MESH Headings
- Animals
- Apoptosis/physiology
- Cell Division/physiology
- Cells, Cultured/physiology
- DNA-Binding Proteins/physiology
- Gene Expression Regulation
- Immunoglobulin J Recombination Signal Sequence-Binding Protein
- Muscle, Smooth, Vascular/cytology
- Myocytes, Smooth Muscle/physiology
- Nuclear Proteins/physiology
- Proto-Oncogene Proteins/antagonists & inhibitors
- Proto-Oncogene Proteins/biosynthesis
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/physiology
- Proto-Oncogene Proteins c-bcl-2/biosynthesis
- Proto-Oncogene Proteins c-bcl-2/genetics
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- RNA, Small Interfering/pharmacology
- Rats
- Receptor, Notch1
- Receptors, Cell Surface/antagonists & inhibitors
- Receptors, Cell Surface/biosynthesis
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/physiology
- Recombinant Fusion Proteins/antagonists & inhibitors
- Recombinant Fusion Proteins/biosynthesis
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/physiology
- Signal Transduction/physiology
- Stress, Mechanical
- Transcription Factors/genetics
- Transcription Factors/physiology
- Transfection
- bcl-2-Associated X Protein
- bcl-X Protein
Collapse
Affiliation(s)
- David Morrow
- Vascular Health Research Centre, Faculty of Science and Health, Dublin City University, Dublin, Ireland
| | | | | | | | | | | | | |
Collapse
|
32
|
Urbanek T, Skop B, Wiaderkiewicz R, Wilczok T, Ziaja K, Lebda-Wyborny T, Pawlicki K. Smooth Muscle Cell Apoptosis in Primary Varicose Veins. Eur J Vasc Endovasc Surg 2004; 28:600-11. [PMID: 15531194 DOI: 10.1016/j.ejvs.2004.09.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2004] [Indexed: 01/31/2023]
Abstract
OBJECTIVES One of the important factors responsible for vessel wall remodelling is programmed cell death. In the paper the role of smooth muscle cell (SMC) apoptosis in primary varicose veins (PVV) is investigated. MATERIAL AND METHODS Vein specimens were obtained from 40 patients with PVV. In each case proximal and distal (upper crural) great saphenous veins (GSV) were harvested. Morphometric computer assessed quantitative evaluation of SMCs, collagen and elastin content was carried out. Apoptotic cells were detected by TUNEL assay. The levels of p53, BAX, BCLl-2 and p21 mRNA expression were assessed by real time RT-QPCR and the presence of respective proteins in the vessel wall was confirmed by immunohistochemistry. RESULTS In the proximal GSV segments a significant increase of p53, p21 and BCL-2 mRNA levels was found in PVV patients. In the distal segments BAX and BCL-2 expression levels were higher. Taking into account the patient age, elevated p53 mRNA expression level was noticed in the distal incompetent GSVs of young PVV patients. In this group a statistically significant increase in the apoptotic index (APIx) within the vein media was found which correlated positively with p53 mRNA expression level. There was no increase of the apoptotic activity in elderly patients that led to the structural changes increase. In proximal GSV segments, despite SMC amount reduction or presence of structural changes in perivalvular wall region, no increase of the APIx with was noticed. CONCLUSIONS P53-related apoptosis is one of the regulatory mechanisms of vein wall homeostasis maintenance. During varicose vein development its activation is related to the early stages of the disease. In the further course, the down-regulation of the SMC apoptosis within the vein media leads to the structural changes increase. The reduction of the SMC population corresponding to an increase of p21 expression in proximal saphenous vein segments suggests that the cell cycle disturbances may lead to the 'weakness' of the proximal GSV wall. Valve injury is not the only factor leading to the varicose veins occurrence.
Collapse
Affiliation(s)
- T Urbanek
- Department of General and Vascular Surgery, Medical University of Silesia, ul. Ziołowa 45/47, Katowice 40-635, Poland.
| | | | | | | | | | | | | |
Collapse
|
33
|
Sweeney C, Morrow D, Birney YA, Coyle S, Hennessy C, Scheller A, Cummins PM, Walls D, Redmond EM, Cahill PA. Notch 1 and 3 receptors modulate vascular smooth muscle cell growth, apoptosis and migration via a CBF‐1/RBP‐Jk dependent pathway. FASEB J 2004; 18:1421-3. [PMID: 15247148 DOI: 10.1096/fj.04-1700fje] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Vascular smooth muscle cell (SMC) fate decisions (cell growth, migration, and apoptosis) are fundamental features in the pathogenesis of vascular disease. We investigated the role of Notch 1 and 3 receptor signaling in controlling adult SMC fate in vitro by establishing that hairy enhancer of split (hes-1 and -5) and related hrt's (hrt-1, -2, and -3) are direct downstream target genes of Notch 1 and 3 receptors in SMC and identified an essential role for nuclear protein CBF-1/RBP-Jk in their regulation. Constitutive expression of active Notch 1 and 3 receptors (Notch IC) resulted in a significant up-regulation of CBF-1/RBP-Jk-dependent promoter activity and Notch target gene expression concomitant with significant increases in SMC growth while concurrently inhibiting SMC apoptosis and migration. Moreover, inhibition of endogenous Notch mediated CBF-1/RBP-Jk regulated gene expression with a non-DNA binding mutant of CBF-1, a Notch IC deleted of its delta RAM domain and the Epstein-Barr virus encoded RPMS-1, in conjunction with pharmacological inhibitors of Notch IC receptor trafficking (brefeldin A and monensin), resulted in a significant decrease in cell growth while concomitantly increasing SMC apoptosis and migration. These findings suggest that endogenous Notch receptors and downstream target genes control vascular cell fate in vitro. Notch signaling, therefore, represents a novel therapeutic target for disease states in which changes in vascular cell fate occur in vivo.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Brefeldin A/pharmacology
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Gene Expression Regulation
- Immunoglobulin J Recombination Signal Sequence-Binding Protein
- Intracellular Signaling Peptides and Proteins/metabolism
- Membrane Proteins/metabolism
- Models, Biological
- Monensin/pharmacology
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Promoter Regions, Genetic/genetics
- Rats
- Receptor, Notch1
- Receptor, Notch3
- Receptors, Cell Surface/metabolism
- Receptors, Notch
- Signal Transduction/drug effects
- Transcription Factors/metabolism
Collapse
Affiliation(s)
- Catherine Sweeney
- Vascular Health Research Centre, Faculty of Science and Health, Dublin City University, Dublin 9, Ireland
| | | | | | | | | | | | | | | | | | | |
Collapse
|