1
|
Wen Y, Ma L, Liu Y, Xiong H, Shi D. Decoding the enigmatic role of T-cadherin in tumor angiogenesis. Front Immunol 2025; 16:1564130. [PMID: 40230838 PMCID: PMC11994602 DOI: 10.3389/fimmu.2025.1564130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 03/14/2025] [Indexed: 04/16/2025] Open
Abstract
The cadherin family, which includes T-cadherin, plays a significant role in angiogenesis, a critical process involved in tumor growth, metastasis, and recurrence. T-cadherin is extensively expressed in both normal and tumor vascular tissues and has been shown to facilitate the proliferation and migration of vascular cells in some studies. However, T-cadherin also exerts inhibitory effects on angiogenesis in various tumor tissues. The functional role of T-cadherin may vary depending on the tumor type and the interaction between tumor cells and vascular cells, suggesting that it acts as a modulator rather than a primary driver of angiogenesis. Additionally, T-cadherin exhibits distinct characteristics depending on the tumor microenvironment. This review provides an overview of recent research on the role of T-cadherin in tumor angiogenesis and discusses its potential as a diagnostic or therapeutic marker in the field of tumor biology.
Collapse
Affiliation(s)
- Yiyang Wen
- The Laboratory of Medical Mycology, Jining No.1 People’s Hospital, Jining, Shandong, China
- Department of Pathology, Jining No.1 People’s Hospital, Jining, Shandong, China
| | - Li Ma
- The Laboratory of Medical Mycology, Jining No.1 People’s Hospital, Jining, Shandong, China
| | - Yuanyuan Liu
- The Second Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Huabao Xiong
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, Shandong, China
| | - Dongmei Shi
- Department of Pathology, Jining No.1 People’s Hospital, Jining, Shandong, China
- Department of Dermatology, Jining No.1 People’s Hospital, Jining, Shandong, China
| |
Collapse
|
2
|
Semina E, Popov V, Khabibullin N, Klimovich P, Sysoeva V, Kurilina E, Tsokolaeva Z, Tkachuk V, Rubina K. New evidence for T-cadherin in COVID-19 pathogenesis, endothelial dysfunction, and lung fibrosis. Front Cell Dev Biol 2025; 13:1476329. [PMID: 40109358 PMCID: PMC11920143 DOI: 10.3389/fcell.2025.1476329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 02/04/2025] [Indexed: 03/22/2025] Open
Abstract
The COVID-19 pandemic had an unprecedented impact on all aspects of human activity worldwide, frequently resulting in post-acute sequelae and affecting multiple organ systems. The underlying mechanisms driving both acute and post-acute manifestations of COVID-19 are still poorly understood, warranting further investigation for new targets. The study represents the first attempt to explore the role of T-cadherin in COVID-19 pathogenesis as well as its implications in pulmonary fibrosis and endothelial dysfunction. First, we revealed a significant decrease in T-cadherin expression in post-mortem lung samples from COVID-19 patients. This downregulated T-cadherin expression correlated with the elevated levels of VE-cadherin and reduced levels of β-catenin, suggesting a disruption in endothelial cell-cell contact integrity and function. Second, the reciprocal relation of T-cadherin and VE-cadherin expression was further confirmed using cultured human endothelial Ea.hy926 cells. T-cadherin overexpression caused a decrease in VE-cadherin mRNA expression in cultured endothelial cells providing additional evidence in favor of their interplay. Third, employing Cdh13 -/- mice, we unveiled the protective role of T-cadherin deficiency against bleomycin-induced lung fibrosis. Fourth, we demonstrated the mice lacking T-cadherin to have downregulated reactive oxygen species production and Nox2 mRNA expression in an angiotensin II-mediated endothelial dysfunction model. Our findings provide rationale for further studies into T-cadherin-mediated mechanisms in these processes.
Collapse
Affiliation(s)
- Ekaterina Semina
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Vladimir Popov
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | | | - Polina Klimovich
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
- Institute of Experimental Cardiology, National Cardiology Research Center of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Veronika Sysoeva
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Ella Kurilina
- Institute of Experimental Cardiology, National Cardiology Research Center of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Zoya Tsokolaeva
- Institute of Experimental Cardiology, National Cardiology Research Center of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Vsevolod Tkachuk
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
- Institute of Experimental Cardiology, National Cardiology Research Center of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Kseniya Rubina
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
3
|
Guo Q, Li J, Wang Z, Wu X, Jin Z, Zhu S, Li H, Zhang D, Hu W, Xu H, Yang L, Shi L, Wang Y. Potassium dehydroandrographolide succinate regulates the MyD88/CDH13 signaling pathway to enhance vascular injury-induced pathological vascular remodeling. Chin J Nat Med 2024; 22:62-74. [PMID: 38278560 DOI: 10.1016/s1875-5364(24)60562-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Indexed: 01/28/2024]
Abstract
Pathological vascular remodeling is a hallmark of various vascular diseases. Previous research has established the significance of andrographolide in maintaining gastric vascular homeostasis and its pivotal role in modulating endothelial barrier dysfunction, which leads to pathological vascular remodeling. Potassium dehydroandrographolide succinate (PDA), a derivative of andrographolide, has been clinically utilized in the treatment of inflammatory diseases precipitated by viral infections. This study investigates the potential of PDA in regulating pathological vascular remodeling. The effect of PDA on vascular remodeling was assessed through the complete ligation of the carotid artery in C57BL/6 mice. Experimental approaches, including rat aortic primary smooth muscle cell culture, flow cytometry, bromodeoxyuridine (BrdU) incorporation assay, Boyden chamber cell migration assay, spheroid sprouting assay, and Matrigel-based tube formation assay, were employed to evaluate the influence of PDA on the proliferation and motility of smooth muscle cells (SMCs). Molecular docking simulations and co-immunoprecipitation assays were conducted to examine protein interactions. The results revealed that PDA exacerbates vascular injury-induced pathological remodeling, as evidenced by enhanced neointima formation. PDA treatment significantly increased the proliferation and migration of SMCs. Further mechanistic studies disclosed that PDA upregulated myeloid differentiation factor 88 (MyD88) expression in SMCs and interacted with T-cadherin (CDH13). This interaction augmented proliferation, migration, and extracellular matrix deposition, culminating in pathological vascular remodeling. Our findings underscore the critical role of PDA in the regulation of pathological vascular remodeling, mediated through the MyD88/CDH13 signaling pathway.
Collapse
Affiliation(s)
- Qiru Guo
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610000, China
| | - Jiali Li
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610000, China
| | - Zheng Wang
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610000, China
| | - Xiao Wu
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610000, China
| | - Zhong Jin
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610000, China
| | - Song Zhu
- Chengdu University of Traditional Chinese Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610000, China
| | - Hongfei Li
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610000, China
| | - Delai Zhang
- Chengdu University of Traditional Chinese Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610000, China
| | - Wangming Hu
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610000, China
| | - Huan Xu
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610000, China
| | - Lan Yang
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610000, China
| | - Liangqin Shi
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610000, China
| | - Yong Wang
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610000, China.
| |
Collapse
|
4
|
Dasen B, Pigeot S, Born GM, Verrier S, Rivero O, Dittrich PS, Martin I, Filippova M. T-cadherin is a novel regulator of pericyte function during angiogenesis. Am J Physiol Cell Physiol 2023; 324:C821-C836. [PMID: 36802732 DOI: 10.1152/ajpcell.00326.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
Pericytes are mural cells that play an important role in regulation of angiogenesis and endothelial function. Cadherins are a superfamily of adhesion molecules mediating Ca2+-dependent homophilic cell-cell interactions that control morphogenesis and tissue remodeling. To date, classical N-cadherin is the only cadherin described on pericytes. Here, we demonstrate that pericytes also express T-cadherin (H-cadherin, CDH13), an atypical glycosyl-phosphatidylinositol (GPI)-anchored member of the superfamily that has previously been implicated in regulation of neurite guidance, endothelial angiogenic behavior, and smooth muscle cell differentiation and progression of cardiovascular disease. The aim of the study was to investigate T-cadherin function in pericytes. Expression of T-cadherin in pericytes from different tissues was performed by immunofluorescence analysis. Using lentivirus-mediated gain-of-function and loss-of-function in cultured human pericytes, we demonstrate that T-cadherin regulates pericyte proliferation, migration, invasion, and interactions with endothelial cells during angiogenesis in vitro and in vivo. T-cadherin effects are associated with the reorganization of the cytoskeleton, modulation of cyclin D1, α-smooth muscle actin (αSMA), integrin β3, metalloprotease MMP1, and collagen expression levels, and involve Akt/GSK3β and ROCK intracellular signaling pathways. We also report the development of a novel multiwell 3-D microchannel slide for easy analysis of sprouting angiogenesis from a bioengineered microvessel in vitro. In conclusion, our data identify T-cadherin as a novel regulator of pericyte function and support that it is required for pericyte proliferation and invasion during active phase of angiogenesis, while T-cadherin loss shifts pericytes toward the myofibroblast state rendering them unable to control endothelial angiogenic behavior.
Collapse
Affiliation(s)
- Boris Dasen
- Tissue Engineering Lab, Department of Biomedicine and Department of Surgery, Basel University Hospital, Basel, Switzerland
| | - Sebastien Pigeot
- Tissue Engineering Lab, Department of Biomedicine and Department of Surgery, Basel University Hospital, Basel, Switzerland
| | - Gordian Manfred Born
- Tissue Engineering Lab, Department of Biomedicine and Department of Surgery, Basel University Hospital, Basel, Switzerland
| | | | - Olga Rivero
- Research Group on Psychiatry and Neurodegenerative Disorders, Biomedical Network Research Centre on Mental Health (CIBERSAM), Valencia, Spain
| | - Petra S Dittrich
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Ivan Martin
- Tissue Engineering Lab, Department of Biomedicine and Department of Surgery, Basel University Hospital, Basel, Switzerland
| | - Maria Filippova
- Tissue Engineering Lab, Department of Biomedicine and Department of Surgery, Basel University Hospital, Basel, Switzerland
| |
Collapse
|
5
|
Guerrero J, Dasen B, Frismantiene A, Pigeot S, Ismail T, Schaefer DJ, Philippova M, Resink TJ, Martin I, Scherberich A. OUP accepted manuscript. Stem Cells Transl Med 2022; 11:213-229. [PMID: 35259280 PMCID: PMC8929526 DOI: 10.1093/stcltm/szab021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 10/31/2021] [Indexed: 11/24/2022] Open
Abstract
Cells of the stromal vascular fraction (SVF) of human adipose tissue have the capacity to generate osteogenic grafts with intrinsic vasculogenic properties. However, cultured adipose-derived stromal cells (ASCs), even after minimal monolayer expansion, lose osteogenic capacity in vivo. Communication between endothelial and stromal/mesenchymal cell lineages has been suggested to improve bone formation and vascularization by engineered tissues. Here, we investigated the specific role of a subpopulation of SVF cells positive for T-cadherin (T-cad), a putative endothelial marker. We found that maintenance during monolayer expansion of a T-cad-positive cell population, composed of endothelial lineage cells (ECs), is mandatory to preserve the osteogenic capacity of SVF cells in vivo and strongly supports their vasculogenic properties. Depletion of T-cad-positive cells from the SVF totally impaired bone formation in vivo and strongly reduced vascularization by SVF cells in association with decreased VEGF and Adiponectin expression. The osteogenic potential of T-cad-depleted SVF cells was fully rescued by co-culture with ECs from a human umbilical vein (HUVECs), constitutively expressing T-cad. Ectopic expression of T-cad in ASCs stimulated mineralization in vitro but failed to rescue osteogenic potential in vivo, indicating that the endothelial nature of the T-cad-positive cells is the key factor for induction of osteogenesis in engineered grafts based on SVF cells. This study demonstrates that crosstalk between stromal and T-cad expressing endothelial cells within adipose tissue critically regulates osteogenesis, with VEGF and adiponectin as associated molecular mediators.
Collapse
Affiliation(s)
- Julien Guerrero
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Boris Dasen
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Agne Frismantiene
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Sebastien Pigeot
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Tarek Ismail
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, Basel, Switzerland
| | - Dirk J Schaefer
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, Basel, Switzerland
| | - Maria Philippova
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Therese J Resink
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Ivan Martin
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Arnaud Scherberich
- Corresponding author: Arnaud Scherberich, Department of Biomedicine, Hebelstrasse 20, University Hospital Basel, 4031 Basel, Switzerland. Tel: +41 061 328 73 75;
| |
Collapse
|
6
|
Yuan XD, Wang JW, Fang Y, Qian Y, Gao S, Fan YC, Wang K. Methylation status of the T-cadherin gene promotor in peripheral blood mononuclear cells is associated with HBV-related hepatocellular carcinoma progression. Pathol Res Pract 2020; 216:152914. [PMID: 32147273 DOI: 10.1016/j.prp.2020.152914] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 02/17/2020] [Accepted: 03/01/2020] [Indexed: 12/11/2022]
Abstract
DNA methylation is one of the epigenetic mechanisms to regulate gene expression and frequently occurs in human cancer cells. T-cadherin (CDH13) is a new member of the cadherin superfamily and possesses multiple functions. Our study included 26 normal controls (NCs), 65 chronic hepatitis B patients (CHB), 14 liver cirrhosis patients (LC) and 157 hepatocellular carcinoma patients (HCC). We mainly focused on the mRNA expression and methylation status of CDH13 in peripheral blood mononuclear cells (PBMCs), which were detected by semi-quantitative real-time polymerase chain reaction (RT-qPCR) and methylation-specific polymerase chain reaction (MSP) respectively. The CDH13 mRNA level was lower in HCC, especially in early-stage of HCC than in NCs and CHB groups (p < 0.05). Methylation frequency of the CDH13 promoter was significantly higher in HCC patients than in the NCs and CHB groups (67.52 % vs 0.00 %, p < 0.001, 67.52 % vs 52.31 %, p < 0.05, respectively). CDH13 mRNA level was significantly and relatively lower in methylated groups than in unmethylated groups among the whole participants. The methylation level of CDH13 promoter in HCC might be influenced or partly influenced by some critical factors such as TBil, ALB and AFP (p < 0.05). As an important factor in signaling pathway regulating by CDH13 to promote carcinogenesis, JNK level was significantly higher in HCC which had a higher methylation frequency than in NCs, CHB and LC (p < 0.05). Furthermore, the combination of the methylated CDH13 level and AFP level showed a better score: AUC = 0.796 (SE = 0.031, 95 %CI 0.735-0.857; p < 0.001) in male and AUC = 0.832 (SE = 0.057, 95 %CI 0.721-0.944; p < 0.001) in female compared to AFP alone for diagnosing HCC from NCs, CHB and LC. The methylation of CDH13 promoter was an independent predictor for assessing the prognosis of HCC patients (r=-1.378 p < 0.05). In conclusion, hypermethylation of CDH13 in PBMCs was associated with the underexpression of mRNA and the high risk of HCC. The methylation status of the CDH13 promoter in PBMCs was a potential noninvasive biomarker to predict the prognosis of HCC patients.
Collapse
Affiliation(s)
- Xiao-Dong Yuan
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Jing-Wen Wang
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Yu Fang
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Yu Qian
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Shuai Gao
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Yu-Chen Fan
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan 250012, China; Shenzhen Research Institute of Shandong University, Shenzhen 518000, China; Institute of Hepatology, Shandong University, Jinan 250012, China
| | - Kai Wang
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan 250012, China; Shenzhen Research Institute of Shandong University, Shenzhen 518000, China; Institute of Hepatology, Shandong University, Jinan 250012, China.
| |
Collapse
|
7
|
Rorabaugh BR, Mabe NW, Seeley SL, Stoops TS, Mucher KE, Ney CP, Goodman CS, Hertenstein BJ, Rush AE, Kasler CD, Sargeant AM, Zoladz PR. Myocardial fibrosis, inflammation, and altered cardiac gene expression profiles in rats exposed to a predator-based model of posttraumatic stress disorder. Stress 2020; 23:125-135. [PMID: 31347429 PMCID: PMC6982550 DOI: 10.1080/10253890.2019.1641081] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
People who are exposed to life-threatening trauma are at risk of developing posttraumatic stress disorder (PTSD). In addition to psychological manifestations, PTSD is associated with an increased risk of myocardial infarction, arrhythmias, hypertension, and other cardiovascular problems. We previously reported that rats exposed to a predator-based model of PTSD develop myocardial hypersensitivity to ischemic injury. This study characterized cardiac changes in histology and gene expression in rats exposed this model. Male rats were subjected to two cat exposures (separated by a period of 10 d) and daily cage-mate changes for 31 d. Control rats were not exposed to the cat or cage-mate changes. Ventricular tissue was analyzed by RNA sequencing, western blotting, histology, and immunohistochemistry. Multifocal lesions characterized by necrosis, mononuclear cell infiltration, and collagen deposition were observed in hearts from all stressed rats but none of the control rats. Gene expression analysis identified clusters of upregulated genes associated with endothelial to mesenchymal transition, endothelial migration, mesenchyme differentiation, and extracellular matrix remodeling in hearts from stressed rats. Consistent with endothelial to mesenchymal transition, rats from stressed hearts exhibited increased expression of α-smooth muscle actin (a myofibroblast marker) and a decrease in the number of CD31 positive endothelial cells. These data provide evidence that predator-based stress induces myocardial lesions and reprograming of cardiac gene expression. These changes may underlie the myocardial hypersensitivity to ischemia observed in these animals. This rat model may provide a useful tool for investigating the cardiac impact of PTSD and other forms of chronic psychological stress.Lay summaryChronic predator stress induces the formation of myocardial lesions characterized by necrosis, collagen deposition, and mononuclear cell infiltration. This is accompanied by changes in gene expression and histology that are indicative of cardiac remodeling. These changes may underlie the increased risk of arrhythmias, myocardial infarction, and other cardiac pathologies in people who have PTSD or other forms of chronic stress.
Collapse
Affiliation(s)
- Boyd R. Rorabaugh
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, Ohio Northern University, Ada, Ohio 45810 USA
- Correspondence: Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, Ohio Northern University, 525 South Main Street, Ada, OH, 45810 USA; Telephone: 419-772-1695; Fax:419-772-1917;
| | - Nathaniel W. Mabe
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina, USA
| | - Sarah L. Seeley
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, Ohio Northern University, Ada, Ohio 45810 USA
| | - Thorne S. Stoops
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, Ohio Northern University, Ada, Ohio 45810 USA
| | - Kasey E. Mucher
- Department of Psychology, Sociology, & Criminal Justice, Ohio Northern University, Ada, OH, USA
| | - Connor P. Ney
- Department of Psychology, Sociology, & Criminal Justice, Ohio Northern University, Ada, OH, USA
| | - Cassandra S. Goodman
- Department of Psychology, Sociology, & Criminal Justice, Ohio Northern University, Ada, OH, USA
| | - Brooke J. Hertenstein
- Department of Psychology, Sociology, & Criminal Justice, Ohio Northern University, Ada, OH, USA
| | - Austen E. Rush
- Department of Psychology, Sociology, & Criminal Justice, Ohio Northern University, Ada, OH, USA
| | - Charis D. Kasler
- Department of Psychology, Sociology, & Criminal Justice, Ohio Northern University, Ada, OH, USA
| | | | - Phillip R. Zoladz
- Department of Psychology, Sociology, & Criminal Justice, Ohio Northern University, Ada, OH, USA
| |
Collapse
|
8
|
Coban N, Pirim D, Erkan AF, Dogan B, Ekici B. Hsa-miR-584-5p as a novel candidate biomarker in Turkish men with severe coronary artery disease. Mol Biol Rep 2019; 47:1361-1369. [PMID: 31863331 DOI: 10.1007/s11033-019-05235-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 12/07/2019] [Indexed: 12/16/2022]
Abstract
Coronary artery disease (CAD) is still the preliminary cause of mortality and morbidity in the developed world. Identification of novel predictive and therapeutic biomarkers is crucial for accurate diagnosis, prognosis and treatment of the CAD. The aim of this study was to detect novel candidate miRNA biomarker that may be used in the management of CAD. We performed miRNA profiling in whole blood samples of angiographically confirmed Turkish men with CAD and non-CAD controls with insignificant coronary stenosis. Validation of microarray results was performed by qRT-PCR in a larger cohort of 62 samples. We subsequently assessed the diagnostic value of the miRNA and correlations of miRNA with clinical parameters. miRNA-target identification and network analyses were conducted by Ingenuity Pathway Analysis (IPA) software. Hsa-miR-584-5p was one of the top significantly dysregulated miRNA observed in miRNA microarray. Men-specific down-regulation (p = 0.040) of hsa-miR-584-5p was confirmed by qRT-PCR. ROC curve analysis highlighted the potential diagnostic value of hsa-miR-584-5p with a power area under the curve (AUC) of 0.714 and 0.643 in men and in total sample, respectively. The expression levels of hsa-miR-584-5p showed inverse correlation with stenosis and Gensini scores. IPA revealed CDH13 as the only CAD related predicted target for the miRNA with biological evidence of its involvement in CAD. This study suggests that hsa-miR-584-5p, known to be tumor suppressor miRNA, as a candidate biomarker for CAD and highlighted its putative role in the CAD pathogenesis. The validation of results in larger samples incorporating functional studies warrant further research.
Collapse
Affiliation(s)
- Neslihan Coban
- Department of Genetics, Aziz Sancar Institute for Experimental Medicine, Istanbul University, Istanbul, Turkey.
| | - Dilek Pirim
- Faculty of Arts & Science, Department of Molecular Biology and Genetics, Bursa Uludag University, Bursa, Turkey
| | - Aycan Fahri Erkan
- Faculty of Medicine, Department of Cardiology, Ufuk University, Ankara, Turkey
| | - Berkcan Dogan
- Institute of Graduate Studies in Sciences, Department of Molecular Biology and Genetics, Istanbul University, Istanbul, Turkey
- Department of Medical Genetics, Bursa Uludag University, Bursa, Turkey
| | - Berkay Ekici
- Faculty of Medicine, Department of Cardiology, Ufuk University, Ankara, Turkey
| |
Collapse
|
9
|
Pastushkova LH, Rusanov VB, Goncharova AG, Brzhozovskiy AG, Kononikhin AS, Chernikova AG, Kashirina DN, Nosovsky AM, Baevsky RM, Nikolaev EN, Larina IM. Urine proteome changes associated with autonomic regulation of heart rate in cosmonauts. BMC SYSTEMS BIOLOGY 2019; 13:17. [PMID: 30836973 PMCID: PMC6399814 DOI: 10.1186/s12918-019-0688-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Background The strategy of adaptation of the human body in microgravity is largely associated with the plasticity of cardiovascular system regulatory mechanisms. During long-term space flights the changes in the stroke volume of the heart are observed, the heart rate decreases, the phase structure of cardiac cycle is readjusted The purpose of this work was to clarify urine proteome changes associated with the initial condition of the heart rate autonomic regulation mechanisms in cosmonauts who have participated in long space missions. Urine proteome of each cosmonaut was analyzed before and after space flight, depending on the initial parameters characterizing the regulatory mechanisms of the cardiovascular system. Results The proteins cadherin-13, mucin-1, alpha-1 of collagen subunit type VI (COL6A1), hemisentin-1, semenogelin-2, SH3 domain-binding protein, transthyretin and serine proteases inhibitors realize a homeostatic role in individuals with different initial type of the cardiovascular system regulation. The role of significantly changed urine proteins in the cardiovascular homeostasis maintenance is associated with complex processes of atherogenesis, neoangiogenesis, activation of calcium channels, changes in cell adhesion and transmembrane properties, changes in extracellular matrix, participation in protection from oxidative stress and leveling the effects of hypoxia. Therefore, the concentrations of these proteins significantly differ between groups with dominant parasympathetic and sympathetic influences. Conclusion The space flight induced urine proteome changes are significantly different in the groups identified by heart rate autonomic regulation peculiarities before space flight. All these proteins regulate the associated biological processes which affect the stiffness of the vascular wall, blood pressure level, the severity of atherosclerotic changes, the rate and degree of age-related involution of elastin and fibulin, age-related increase in collagen stiffness, genetically determined features of elastin fibers. The increased vascular rigidity (including the aorta) and of myocardium may be regarded as a universal response to various extreme factors. Significant differences in the semi-quantitative analysis of signal proteins between groups with different types of autonomic regulation are explained by a common goal: to ensure optimal adaptation regardless of age and of the genetically determined type of responses to the extreme environmental factors effects. Electronic supplementary material The online version of this article (10.1186/s12918-019-0688-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lyudmila H Pastushkova
- Institute for Biomedical Problems - Russian Federation State Scientific Research Center Russian Academy of Sciences, Moscow, Russia
| | - Vasily B Rusanov
- Institute for Biomedical Problems - Russian Federation State Scientific Research Center Russian Academy of Sciences, Moscow, Russia
| | - Anna G Goncharova
- Institute for Biomedical Problems - Russian Federation State Scientific Research Center Russian Academy of Sciences, Moscow, Russia
| | - Alexander G Brzhozovskiy
- Institute for Biomedical Problems - Russian Federation State Scientific Research Center Russian Academy of Sciences, Moscow, Russia
| | - Alexey S Kononikhin
- Institute for Biomedical Problems - Russian Federation State Scientific Research Center Russian Academy of Sciences, Moscow, Russia.,V.L. Talrose Institute for Energy Problems of Chemical Physics, Russian Academy of Sciences, Moscow, Russia.,Moscow Institute of Physics and Technology, Dolgoprudny, Moscow region, Russia
| | - Anna G Chernikova
- Institute for Biomedical Problems - Russian Federation State Scientific Research Center Russian Academy of Sciences, Moscow, Russia
| | - Daria N Kashirina
- Institute for Biomedical Problems - Russian Federation State Scientific Research Center Russian Academy of Sciences, Moscow, Russia
| | - Andrey M Nosovsky
- Institute for Biomedical Problems - Russian Federation State Scientific Research Center Russian Academy of Sciences, Moscow, Russia
| | - Roman M Baevsky
- Institute for Biomedical Problems - Russian Federation State Scientific Research Center Russian Academy of Sciences, Moscow, Russia
| | - Evgeny N Nikolaev
- V.L. Talrose Institute for Energy Problems of Chemical Physics, Russian Academy of Sciences, Moscow, Russia. .,Moscow Institute of Physics and Technology, Dolgoprudny, Moscow region, Russia. .,Skolkovo Institute of Science and Technology, Skolkovo, Moscow region, Russia.
| | - Irina M Larina
- Institute for Biomedical Problems - Russian Federation State Scientific Research Center Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
10
|
Frismantiene A, Philippova M, Erne P, Resink TJ. Smooth muscle cell-driven vascular diseases and molecular mechanisms of VSMC plasticity. Cell Signal 2018; 52:48-64. [PMID: 30172025 DOI: 10.1016/j.cellsig.2018.08.019] [Citation(s) in RCA: 282] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 08/28/2018] [Accepted: 08/28/2018] [Indexed: 02/06/2023]
Abstract
Vascular smooth muscle cells (VSMCs) are the major cell type in blood vessels. Unlike many other mature cell types in the adult body, VSMC do not terminally differentiate but retain a remarkable plasticity. Fully differentiated medial VSMCs of mature vessels maintain quiescence and express a range of genes and proteins important for contraction/dilation, which allows them to control systemic and local pressure through the regulation of vascular tone. In response to vascular injury or alterations in local environmental cues, differentiated/contractile VSMCs are capable of switching to a dedifferentiated phenotype characterized by increased proliferation, migration and extracellular matrix synthesis in concert with decreased expression of contractile markers. Imbalanced VSMC plasticity results in maladaptive phenotype alterations that ultimately lead to progression of a variety of VSMC-driven vascular diseases. The nature, extent and consequences of dysregulated VSMC phenotype alterations are diverse, reflecting the numerous environmental cues (e.g. biochemical factors, extracellular matrix components, physical) that prompt VSMC phenotype switching. In spite of decades of efforts to understand cues and processes that normally control VSMC differentiation and their disruption in VSMC-driven disease states, the crucial molecular mechanisms and signalling pathways that shape the VSMC phenotype programme have still not yet been precisely elucidated. In this article we introduce the physiological functions of vascular smooth muscle/VSMCs, outline VSMC-driven cardiovascular diseases and the concept of VSMC phenotype switching, and review molecular mechanisms that play crucial roles in the regulation of VSMC phenotypic plasticity.
Collapse
Affiliation(s)
- Agne Frismantiene
- Department of Biomedicine, Laboratory for Signal Transduction, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Maria Philippova
- Department of Biomedicine, Laboratory for Signal Transduction, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Paul Erne
- Department of Biomedicine, Laboratory for Signal Transduction, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Therese J Resink
- Department of Biomedicine, Laboratory for Signal Transduction, University Hospital Basel and University of Basel, Basel, Switzerland.
| |
Collapse
|
11
|
Frismantiene A, Philippova M, Erne P, Resink TJ. Cadherins in vascular smooth muscle cell (patho)biology: Quid nos scimus? Cell Signal 2018; 45:23-42. [DOI: 10.1016/j.cellsig.2018.01.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 01/23/2018] [Accepted: 01/23/2018] [Indexed: 12/16/2022]
|
12
|
Li TD, Zeng ZH. Adiponectin as a potential therapeutic target for the treatment of restenosis. Biomed Pharmacother 2018; 101:798-804. [PMID: 29525676 DOI: 10.1016/j.biopha.2018.03.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 02/28/2018] [Accepted: 03/02/2018] [Indexed: 12/14/2022] Open
Abstract
Restenosis is a pathologic re-narrowing of a coronary artery lesion after mechanical injury. Its pathophysiological mechanisms have not been fully elucidated at present, but are thought to include inflammation, vascular smooth muscle cell (VSMC) proliferation, and matrix remodeling, beginning with insufficient endothelium healing. Restenosis presents with angina symptoms or acute coronary syndromes and lead to a revascularization, either with coronary artery bypass or repeat percutaneous coronary intervention. Some studies have reported that hypoadiponectinemia has been an independent risk factor for the onset of acute coronary syndromes and restenosis. Accumulating evidence shows that low concentrations of adiponectin may be involved in impairing endothelium functions, inflammation, and VSMC proliferation that lead to restenosis. Preclinical studies have proven that adiponectin promotes endothelium healing, effectively inhibits inflammation, and maintains contractile phenotypes of VSMCs, indicating that it may be developed as a new therapeutic target for the treatment of restenosis.
Collapse
Affiliation(s)
- Tu di Li
- Department of Cardiovascular Diseases, The First Affiliated Hospital of Guangdong Pharmaceutical University, China
| | - Zhi Huan Zeng
- Department of Cardiovascular Diseases, The First Affiliated Hospital of Guangdong Pharmaceutical University, China.
| |
Collapse
|
13
|
Exogenous BMP7 in aortae of rats with chronic uremia ameliorates expression of profibrotic genes, but does not reverse established vascular calcification. PLoS One 2018; 13:e0190820. [PMID: 29304096 PMCID: PMC5755916 DOI: 10.1371/journal.pone.0190820] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 12/20/2017] [Indexed: 12/24/2022] Open
Abstract
Hyperphosphatemia and vascular calcification are frequent complications of chronic renal failure and bone morphogenetic protein 7 (BMP7) has been shown to protect against development of vascular calcification in uremia. The present investigation examined the potential reversibility of established uremic vascular calcification by treatment of uremic rats with BMP7. A control model of isogenic transplantation of a calcified aorta from uremic rats into healthy littermates examined whether normalization of the uremic environment reversed vascular calcification. Uremia and vascular calcification were induced in rats by 5/6 nephrectomy, high phosphate diet and alfacalcidol treatment. After 14 weeks severe vascular calcification was present and rats were allocated to BMP7, vehicle or aorta transplantation. BMP7 treatment caused a significant decrease of plasma phosphate to 1.56 ± 0.17 mmol/L vs 2.06 ± 0.34 mmol/L in the vehicle group even in the setting of uremia and high phosphate diet. Uremia and alfacalcidol resulted in an increase in aortic expression of genes related to fibrosis, osteogenic transformation and extracellular matrix calcification, and the BMP7 treatment resulted in a decrease in the expression of profibrotic genes. The total Ca-content of the aorta was however unchanged both in the abdominal aorta: 1.9 ± 0.6 μg/mg tissue in the vehicle group vs 2.2 ± 0.6 μg/mg tissue in the BMP7 group and in the thoracic aorta: 71 ± 27 μg/mg tissue in the vehicle group vs 54 ± 18 μg/mg tissue in the BMP7 group. Likewise, normalization of the uremic environment by aorta transplantation had no effect on the Ca-content of the calcified aorta: 16.3 ± 0.6 μg/mg tissue pre-transplantation vs 15.9 ± 2.3 μg/mg tissue post-transplantation. Aortic expression of genes directly linked to extracellular matrix calcification was not affected by BMP7 treatment, which hypothetically might explain persistent high Ca-content in established vascular calcification. The present results highlight the importance of preventing the development of vascular calcification in chronic kidney disease. Once established, vascular calcification persists even in the setting when hyperphosphatemia or the uremic milieu is abolished.
Collapse
|
14
|
Rubina KA, Semina EV, Tkachuk VA. Guidance molecules and chemokines in angiogenesis and vascular remodeling. J EVOL BIOCHEM PHYS+ 2017. [DOI: 10.1134/s0022093017050015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
15
|
The T > A (rs11646213) gene polymorphism of cadherin-13 ( CDH13 ) gene is associated with decreased risk of developing hypertension in Mexican population. Immunobiology 2017; 222:973-978. [DOI: 10.1016/j.imbio.2016.09.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 07/26/2016] [Accepted: 09/07/2016] [Indexed: 12/26/2022]
|
16
|
Kyriakakis E, Frismantiene A, Dasen B, Pfaff D, Rivero O, Lesch KP, Erne P, Resink TJ, Philippova M. T-cadherin promotes autophagy and survival in vascular smooth muscle cells through MEK1/2/Erk1/2 axis activation. Cell Signal 2017; 35:163-175. [DOI: 10.1016/j.cellsig.2017.04.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 03/20/2017] [Accepted: 04/05/2017] [Indexed: 10/19/2022]
|
17
|
Rubina KA, Tkachuk VA. Guidance Receptors in the Nervous and Cardiovascular Systems. BIOCHEMISTRY (MOSCOW) 2016; 80:1235-53. [PMID: 26567567 DOI: 10.1134/s0006297915100041] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Blood vessels and nervous fibers grow in parallel, for they express similar receptors for chemokine substances. Recently, much attention is being given to studying guidance receptors and their ligands besides the growth factors, cytokines, and chemokines necessary to form structures in the nervous and vascular systems. Such guidance molecules determine trajectory for growing axons and vessels. Guidance molecules include Ephrins and their receptors, Neuropilins and Plexins as receptors for Semaphorins, Robos as receptors for Slit-proteins, and UNC5B receptors binding Netrins. Apart from these receptors and their ligands, urokinase and its receptor (uPAR) and T-cadherin are also classified as guidance molecules. The urokinase system mediates local proteolysis at the leading edge of cells, thereby providing directed migration. T-cadherin is a repellent molecule that regulates the direction of growing axons and blood vessels. Guidance receptors also play an important role in the diseases of the nervous and cardiovascular systems.
Collapse
Affiliation(s)
- K A Rubina
- Lomonosov Moscow State University, Faculty of Fundamental Medicine, Moscow, 119192, Russia.
| | | |
Collapse
|
18
|
Balatskaya MN, Balatskii AV, Sharonov GV, Tkachuk VA. T-cadherin as a novel receptor regulating metabolism in the blood vessel and heart cells: from structure to function. J EVOL BIOCHEM PHYS+ 2016. [DOI: 10.1134/s0022093016020010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
19
|
Frismantiene A, Dasen B, Pfaff D, Erne P, Resink TJ, Philippova M. T-cadherin promotes vascular smooth muscle cell dedifferentiation via a GSK3β-inactivation dependent mechanism. Cell Signal 2016; 28:516-530. [DOI: 10.1016/j.cellsig.2016.02.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 02/12/2016] [Accepted: 02/18/2016] [Indexed: 11/24/2022]
|
20
|
Schoenenberger AW, Pfaff D, Dasen B, Frismantiene A, Erne P, Resink TJ, Philippova M. Gender-Specific Associations between Circulating T-Cadherin and High Molecular Weight-Adiponectin in Patients with Stable Coronary Artery Disease. PLoS One 2015; 10:e0131140. [PMID: 26083608 PMCID: PMC4470588 DOI: 10.1371/journal.pone.0131140] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 05/27/2015] [Indexed: 12/18/2022] Open
Abstract
Close relationships exist between presence of adiponectin (APN) within vascular tissue and expression of T-cadherin (T-cad) on vascular cells. APN and T-cad are also present in the circulation but here their relationships are unknown. This study investigates associations between circulating levels of high molecular weight APN (HMW-APN) and T-cad in a population comprising 66 women and 181 men with angiographically proven stable coronary artery disease (CAD). Plasma HMW-APN and T-cad were measured by ELISA and analysed for associations with baseline clinical characteristics and with each other. In multivariable analysis BMI and HDL were independently associated with HMW-APN in both genders, while diabetes and extent of coronary stenosis were independently associated with T-cad in males only. Regression analysis showed no significant association between HMW-APN and T-cad in the overall study population. However, there was a negative association between HMW-APN and T-cad (P=0.037) in a subgroup of young men (age <60 years, had no diabetes and no or 1-vessel CAD) which persisted after multivariable analysis with adjustment for all potentially influential variables (P=0.021). In the corresponding subgroup of women there was a positive association between HMW-APN and T-cad (P=0.013) which disappeared after adjustment for HDL. After exclusion of the young men, a positive association (P=0.008) between HMW-APN and T-cad was found for the remaining participants of the overall population which disappeared after adjustment for HDL and BMI. The existence of opposing correlations between circulating HMW-APN and T-cad in male and female patient populations underscores the necessity to consider gender as a confounding variable when evaluating biomarker potentials of APN and T-cad.
Collapse
Affiliation(s)
- Andreas W. Schoenenberger
- Division of Geriatrics, Department of General Internal Medicine, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland
| | - Dennis Pfaff
- Department of Biomedicine, Laboratory for Signal Transduction, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Boris Dasen
- Department of Biomedicine, Laboratory for Signal Transduction, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Agne Frismantiene
- Department of Biomedicine, Laboratory for Signal Transduction, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Paul Erne
- Department of Biomedicine, Laboratory for Signal Transduction, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Therese J. Resink
- Department of Biomedicine, Laboratory for Signal Transduction, University Hospital Basel and University of Basel, Basel, Switzerland
- * E-mail:
| | - Maria Philippova
- Department of Biomedicine, Laboratory for Signal Transduction, University Hospital Basel and University of Basel, Basel, Switzerland
| |
Collapse
|
21
|
Hartwig S, Goeddeke S, Poschmann G, Dicken HD, Jacob S, Nitzgen U, Passlack W, Stühler K, Ouwens DM, Al-Hasani H, Knebel B, Kotzka J, Lehr S. Identification of novel adipokines differential regulated in C57BL/Ks and C57BL/6. Arch Physiol Biochem 2014; 120:208-15. [PMID: 25319219 DOI: 10.3109/13813455.2014.970197] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Visceral adiposity is associated with metabolic disorders, but little is known on the underlying pathophysiological mechanism. One possible link might be the release of various signalling and mediator proteins, named adipokines. Our hypothesis was that dependent on genetic background factors are released which might trigger a primary disease susceptibility. This study characterizes the adipokines released from visceral adipose tissue from two metabolic healthy mouse strains, i.e. C57BL/Ks (BKS) and C57BL/6 (C57), of which the former genetic background is more sensitive to develop diabetes following metabolic challenge. Using liquid chromatography (LC)-electrospray ionization (ESI)-MS/MS, a reference map comprising 597 adipokines was generated (http://www.diabesityprot.org). Thirty-five adipokines, including six not previously described ones, were differentially released between the mouse strains. Most notable is the reduced release of the adiponectin-binding protein T-Cadherin (CAD13) in BKS mice. This observation highlights the importance of secretome profiling in unravelling the complex interplay between genetic diversity and lifestyle.
Collapse
Affiliation(s)
- Sonja Hartwig
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at the Heinrich-Heine-University Duesseldorf, Leibniz Center for Diabetes Research , Duesseldorf , Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Frismantiene A, Pfaff D, Frachet A, Coen M, Joshi MB, Maslova K, Bochaton-Piallat ML, Erne P, Resink TJ, Philippova M. Regulation of contractile signaling and matrix remodeling by T-cadherin in vascular smooth muscle cells: Constitutive and insulin-dependent effects. Cell Signal 2014; 26:1897-908. [DOI: 10.1016/j.cellsig.2014.05.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 04/29/2014] [Accepted: 05/02/2014] [Indexed: 12/29/2022]
|
23
|
Nakatsuji H, Kishida K, Sekimoto R, Komura N, Kihara S, Funahashi T, Shimomura I. Accumulation of adiponectin in inflamed adipose tissues of obese mice. Metabolism 2014; 63:542-53. [PMID: 24467915 DOI: 10.1016/j.metabol.2013.12.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Revised: 11/29/2013] [Accepted: 12/31/2013] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Adipose tissue inflammation plays an important role in the pathogenesis of obesity-associated complications, such as atherosclerosis. Adiponectin secreted from adipocytes has various beneficial effects including anti-inflammatory effect. Obesity often presents with hypoadiponectinemia. However, the mechanism and adiponectin movement in obesity remain uncharacterized. Here we investigated tissue distribution of adiponectin protein in lean and obese mice. METHODS Adiponectin protein levels were evaluated by enzyme-linked immunosorbent assay and western blotting. Adipose tissues were fractionated into mature adipocyte fraction (MAF) and stromal vascular fraction (SVF). RESULTS Adiponectin protein was detected not only in MAF but also in SVF, which lacks adiponectin mRNA expression, of adipose tissue remarkably. SVF adiponectin protein level was higher in obese mice than in lean mice. The mechanism of adiponectin accumulation was investigated in adiponectin-deficient (APN-KO) mice after injection of plasma from wild-type mice. These mice showed accumulation of exogenous adiponectin, which derived from wild type mice, in adipose tissues, and the adiponectin was more observed in SVF of diet induced obese APN-KO mice than lean APN-KO mice. Among the adiponectin binding proteins, T-cadherin mRNA and protein levels in SVF of obese mice were remarkably higher than in lean mice. Oxidative stress levels were also significantly higher in SVF of obese mice than lean mice. Mechanistically, H2O2 up-regulated T-cadherin mRNA level in murine macrophages. CONCLUSIONS The results demonstrated adiponectin targets to adipose SVF of obese mice. These findings should shed a new light on the pathology of adipose tissue inflammation and hypoadiponectinemia of obesity.
Collapse
Affiliation(s)
- Hideaki Nakatsuji
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Ken Kishida
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan.
| | - Ryohei Sekimoto
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Noriyuki Komura
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Shinji Kihara
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Tohru Funahashi
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan; Department of Metabolism and Atherosclerosis, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Iichiro Shimomura
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|
24
|
Nakamura MT, Yudell BE, Loor JJ. Regulation of energy metabolism by long-chain fatty acids. Prog Lipid Res 2013; 53:124-44. [PMID: 24362249 DOI: 10.1016/j.plipres.2013.12.001] [Citation(s) in RCA: 548] [Impact Index Per Article: 45.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Revised: 12/03/2013] [Accepted: 12/04/2013] [Indexed: 12/12/2022]
Abstract
In mammals, excess energy is stored primarily as triglycerides, which are mobilized when energy demands arise. This review mainly focuses on the role of long chain fatty acids (LCFAs) in regulating energy metabolism as ligands of peroxisome proliferator-activated receptors (PPARs). PPAR-alpha expressed primarily in liver is essential for metabolic adaptation to starvation by inducing genes for beta-oxidation and ketogenesis and by downregulating energy expenditure through fibroblast growth factor 21. PPAR-delta is highly expressed in skeletal muscle and induces genes for LCFA oxidation during fasting and endurance exercise. PPAR-delta also regulates glucose metabolism and mitochondrial biogenesis by inducing FOXO1 and PGC1-alpha. Genes targeted by PPAR-gamma in adipocytes suggest that PPAR-gamma senses incoming non-esterified LCFAs and induces the pathways to store LCFAs as triglycerides. Adiponectin, another important target of PPAR-gamma may act as a spacer between adipocytes to maintain their metabolic activity and insulin sensitivity. Another topic of this review is effects of skin LCFAs on energy metabolism. Specific LCFAs are required for the synthesis of skin lipids, which are essential for water barrier and thermal insulation functions of the skin. Disturbance of skin lipid metabolism often causes apparent resistance to developing obesity at the expense of normal skin function.
Collapse
Affiliation(s)
- Manabu T Nakamura
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, 905 South Goodwin Avenue, Urbana, IL 61801, USA.
| | - Barbara E Yudell
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, 905 South Goodwin Avenue, Urbana, IL 61801, USA
| | - Juan J Loor
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, 905 South Goodwin Avenue, Urbana, IL 61801, USA
| |
Collapse
|
25
|
Lee JH, Shin DJ, Park S, Kang SM, Jang Y, Lee SH. Association between CDH13 variants and cardiometabolic and vascular phenotypes in a Korean population. Yonsei Med J 2013; 54:1305-12. [PMID: 24142632 PMCID: PMC3809859 DOI: 10.3349/ymj.2013.54.6.1305] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
PURPOSE Although some CDH13 single nucleotide polymorphisms (SNPs) have been shown to be determinants of blood adiponectin levels, the clinical implications of CDH13 variants are not yet completely understood. The purpose of this study was to evaluate the effects of SNPs of CDH13 on metabolic and vascular phenotypes. MATERIALS AND METHODS We included 238 hypertensive subjects and 260 age- and sex-matched controls. Seven tagging-SNPs were identified in the CDH13 gene by whole gene sequencing. The association between these SNP variants and the risk of hypertension, metabolic traits, and carotid intima-media thickness (IMT) was examined. RESULTS Minor allele carriers of rs12444338 had a lower risk of hypertension, but the association turned out just marginal after adjusting confoudners. Blood glucose levels were higher in the minor allele carriers of c.1407C>T (p=0.01), whereas low-density lipoprotein-cholesterol levels were greater in those of rs6565105 (p=0.02). The minor allele of rs1048612 was associated with a higher body mass index (p=0.01). In addition, the mean carotid IMT was significantly associated with rs12444338 (p=0.02) and rs1048612 (p=0.02). CONCLUSION These results provide evidence that CDH13 variants are associated with metabolic traits and carotid atherosclerosis in Koreans. This study shows the multifaceted effects of CDH13 variants on cardiometabolic risk.
Collapse
Affiliation(s)
- Ji Hyun Lee
- Cardiology Division, Department of Internal Medicine, Severance Cardiovascular Hospital, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 120-752, Korea.
| | | | | | | | | | | |
Collapse
|
26
|
Novel mechanism regulating endothelial permeability via T-cadherin-dependent VE-cadherin phosphorylation and clathrin-mediated endocytosis. Mol Cell Biochem 2013; 387:39-53. [PMID: 24136461 PMCID: PMC3904039 DOI: 10.1007/s11010-013-1867-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 10/09/2013] [Indexed: 12/21/2022]
Abstract
T-cadherin is a unique member of the cadherin superfamily of adhesion molecules. In contrast to “classical” cadherins, T-cadherin lacks transmembrane and cytoplasmic domains and is anchored to the cell membrane via a glycosilphosphoinositol moiety. T-cadherin is predominantly expressed in cardiovascular system. Clinical and biochemical studies evidence that expression of T-cadherin increases in post-angioplasty restenosis and atherosclerotic lesions—conditions associated with endothelial dysfunction and pathological expression of adhesion molecules. Here, we provide data suggesting a new signaling mechanism by which T-cadherin regulates endothelial permeability. T-cadherin overexpression leads to VE-cadherin phosphorylation on Y731 (β-catenin-binding site), VE-cadherin clathrin-dependent endocytosis and its degradation in lysosomes. Moreover, T-cadherin overexpression results in activation of Rho GTPases signaling and actin stress fiber formation. Thus, T-cadherin up-regulation is involved in degradation of a key endothelial adhesion molecule, VE-cadherin, resulting in the disruption of endothelial barrier function. Our results point to the role of T-cadherin in regulation of endothelial permeability and its possible engagement in endothelial dysfunction.
Collapse
|
27
|
Philippova M, Joshi MB, Pfaff D, Kyriakakis E, Maslova K, Erne P, Resink TJ. T-cadherin attenuates insulin-dependent signalling, eNOS activation, and angiogenesis in vascular endothelial cells. Cardiovasc Res 2012; 93:498-507. [PMID: 22235028 DOI: 10.1093/cvr/cvs004] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
AIMS T-cadherin (T-cad) is a glycosylphosphatidylinositol-anchored cadherin family member. Experimental, clinical, and genomic studies suggest a role for T-cad in vascular disorders such as atherosclerosis and hypertension, which are associated with endothelial dysfunction and insulin resistance (InsRes). In endothelial cells (EC), T-cad and insulin activate similar signalling pathways [e.g. PI3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR)] and processes (e.g. angiogenesis). We hypothesize that T-cad is a regulatory component of insulin signalling in EC and therefore a determinant of the development of endothelial InsRes. METHODS AND RESULTS We investigated T-cad-dependent effects on insulin sensitivity using human EC stably transduced with respect to T-cad overexpression or T-cad silencing. Responsiveness to insulin was examined at the level of effectors of the insulin signalling cascade, EC nitric oxide synthase (eNOS) activation, and angiogenic behaviour. Overexpression and ligation of T-cad on EC attenuates insulin-dependent activation of the PI3K/Akt/mTOR signalling axis, eNOS, EC migration, and angiogenesis. Conversely, T-cad silencing enhances these actions of insulin. Attenuation of EC responsiveness to insulin results from T-cad-mediated chronic activation of the Akt/mTOR-dependent negative feedback loop of the insulin cascade and enhanced degradation of the insulin receptor (IR) substrate. Co-immunoprecipitation experiments revealed an association between T-cad and IR. Filipin abrogated inhibitory effects of T-cad on insulin signalling, demonstrating localization of T-cad-insulin cross-talk to lipid raft plasma membrane domains. Hyperinsulinaemia up-regulates T-cad mRNA and protein levels in EC. CONCLUSION T-cad expression modulates signalling and functional responses of EC to insulin. We have identified a novel signalling mechanism regulating insulin function in the endothelium and attribute a role for T-cad up-regulation in the pathogenesis of endothelial InsRes.
Collapse
Affiliation(s)
- Maria Philippova
- Laboratory for Signal Transduction, Department of Biomedicine, Basel University Hospital, Hebelstrasse 20, CH 4031 Basel, Switzerland
| | | | | | | | | | | | | |
Collapse
|
28
|
Chung CM, Lin TH, Chen JW, Leu HB, Yang HC, Ho HY, Ting CT, Sheu SH, Tsai WC, Chen JH, Lin SJ, Chen YT, Pan WH. A genome-wide association study reveals a quantitative trait locus of adiponectin on CDH13 that predicts cardiometabolic outcomes. Diabetes 2011; 60:2417-23. [PMID: 21771975 PMCID: PMC3161336 DOI: 10.2337/db10-1321] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
OBJECTIVE The plasma adiponectin level, a potential upstream and internal facet of metabolic and cardiovascular diseases, has a reasonably high heritability. Whether other novel genes influence the variation in adiponectin level and the roles of these genetic variants on subsequent clinical outcomes has not been thoroughly investigated. Therefore, we aimed not only to identify genetic variants modulating plasma adiponectin levels but also to investigate whether these variants are associated with adiponectin-related metabolic traits and cardiovascular diseases. RESEARCH DESIGN AND METHODS We conducted a genome-wide association study (GWAS) to identify quantitative trait loci (QTL) associated with high molecular weight forms of adiponectin levels by genotyping 382 young-onset hypertensive (YOH) subjects with Illumina HumanHap550 SNP chips. The culpable single nucleotide polymorphism (SNP) variants responsible for lowered adiponectin were then confirmed in another 559 YOH subjects, and the association of these SNP variants with the risk of metabolic syndrome (MS), type 2 diabetes mellitus (T2DM), and ischemic stroke was examined in an independent community-based prospective cohort, the CardioVascular Disease risk FACtors Two-township Study (CVDFACTS, n = 3,350). RESULTS The SNP (rs4783244) most significantly associated with adiponectin levels was located in intron 1 of the T-cadherin (CDH13) gene in the first stage (P = 7.57 × 10(-9)). We replicated and confirmed the association between rs4783244 and plasma adiponectin levels in an additional 559 YOH subjects (P = 5.70 × 10(-17)). This SNP was further associated with the risk of MS (odds ratio [OR] = 1.42, P = 0.027), T2DM in men (OR = 3.25, P = 0.026), and ischemic stroke (OR = 2.13, P = 0.002) in the CVDFACTS. CONCLUSIONS These findings indicated the role of T-cadherin in modulating adiponectin levels and the involvement of CDH13 or adiponectin in the development of cardiometabolic diseases.
Collapse
Affiliation(s)
- Chia-Min Chung
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Tsung-Hsien Lin
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Internal Medicine, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jaw-Wen Chen
- Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan
- Department of Medical Research and Education, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Hsin-Bang Leu
- Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Hsin-Chou Yang
- Institute of Statistical Science, Academia Sinica, Taipei, Taiwan
| | - Hung-Yun Ho
- Taichung Veterans General Hospital, Taichung, Taiwan
| | - Chih-Tai Ting
- Taichung Veterans General Hospital, Taichung, Taiwan
| | - Sheng-Hsiung Sheu
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Internal Medicine, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Wei-Chuan Tsai
- College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jyh-Hong Chen
- College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shing-Jong Lin
- Department of Medical Research and Education, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yuan-Tsong Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Wen-Harn Pan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Division of Preventive Medicine and Health Service Research, National Health Research Institutes, Miaoli, Taiwan
- Corresponding author: Wen-Harn Pan,
| |
Collapse
|
29
|
Yurlova EI, Rubina KA, Sysoeva VY, Sharonov GV, Semina EV, Parfenova EV, Tkachuk VA. T-cadherin suppresses the cell proliferation of mouse melanoma B16F10 and tumor angiogenesis in the model of the chorioallantoic membrane. Russ J Dev Biol 2010. [DOI: 10.1134/s1062360410040028] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
30
|
T-cadherin expression in cardiac allograft vasculopathy: Bench to bedside translational investigation. J Heart Lung Transplant 2010; 29:792-9. [DOI: 10.1016/j.healun.2010.03.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2009] [Revised: 03/02/2010] [Accepted: 03/03/2010] [Indexed: 11/19/2022] Open
|
31
|
Philippova M, Suter Y, Toggweiler S, Schoenenberger AW, Joshi MB, Kyriakakis E, Erne P, Resink TJ. T-cadherin is present on endothelial microparticles and is elevated in plasma in early atherosclerosis. Eur Heart J 2010; 32:760-71. [PMID: 20584775 DOI: 10.1093/eurheartj/ehq206] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
AIMS The presence of endothelial cell (EC)-derived surface molecules in the circulation is among hallmarks of endothelial activation and damage in vivo. Previous investigations suggest that upregulation of T-cadherin (T-cad) on the surface of ECs may be a characteristic marker of EC activation and stress. We investigated whether T-cad might also be shed from ECs and in amounts reflecting the extent of activation or damage. METHODS AND RESULTS Immunoblotting showed the presence of T-cad protein in the culture medium from normal proliferating ECs and higher levels in the medium from stressed/apoptotic ECs. Release of T-cad into the circulation occurs in vivo and in association with endothelial dysfunction. Sandwich ELISA revealed negligible T-cad protein in the plasma of healthy volunteers (0.90 ± 0.90 ng/mL, n = 30), and increased levels in the plasma from patients with non-significant atherosclerosis (9.23 ± 2.61 ng/mL, n = 63) and patients with chronic coronary artery disease (6.93 ± 1.31 ng/mL, n = 162). In both patient groups there was a significant (P = 0.043) dependency of T-cad and degree of endothelial dysfunction as measured by reactive hyperaemia peripheral tonometry. Flow cytometry analysis showed that the major fraction of T-cad was released into the EC culture medium and the plasma as a surface component of EC-derived annexin V- and CD144/CD31-positive microparticles (MPs). Gain-of-function and loss-of-function studies demonstrate that MP-bound T-cad induced Akt phosphorylation and activated angiogenic behaviour in target ECs via homophilic-based interactions. CONCLUSION Our findings reveal a novel mechanism of T-cad-dependent signalling in the vascular endothelium. We identify T-cad as an endothelial MP antigen in vivo and demonstrate that its level in plasma is increased in early atherosclerosis and correlates with endothelial dysfunction.
Collapse
Affiliation(s)
- Maria Philippova
- Department of Biomedicine, Laboratory for Signal Transduction, Basel University Hospital, ZLF 316 Hebelstrasse 20, CH 4031, Basel, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Brochu-Gaudreau K, Rehfeldt C, Blouin R, Bordignon V, Murphy BD, Palin MF. Adiponectin action from head to toe. Endocrine 2010; 37:11-32. [PMID: 20963555 DOI: 10.1007/s12020-009-9278-8] [Citation(s) in RCA: 224] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2009] [Accepted: 10/14/2009] [Indexed: 02/06/2023]
Abstract
Adiponectin, the most abundant protein secreted by white adipose tissue, is known for its involvement in obesity-related disorders such as insulin resistance, type 2 diabetes mellitus and atherosclerosis. Moreover, modulation of the circulating adiponectin concentration is observed in pathologies that are more or less obesity-related, such as cancer and rheumatoid arthritis. The wide distribution of adiponectin receptors in various organs and tissues suggests that adiponectin has pleiotropic effects on numerous physiological processes. Besides its well-known insulin-sensitizing, anti-inflammatory and antiatherosclerotic properties, accumulating evidence suggests that adiponectin may also have anticancer properties and be cardioprotective. A beneficial effect of adiponectin on female reproductive function was also suggested. Since adiponectin has numerous beneficial biological functions, its use as a therapeutic agent has been suggested. However, the use of adiponectin or its receptors as therapeutic targets is complicated by the presence of different adiponectin oligomeric isoforms and production sites, by multiple receptors with differing affinities for adiponectin isoforms, and by cell-type-specific effects in different tissues. In this review, we discuss the known and potential roles of adiponectin in various tissues and pathologies. The therapeutic promise of administration of adiponectin and the use of its circulating levels as a diagnostic biomarker are further discussed based on the latest experimental studies.
Collapse
|
33
|
Joshi MB, Kyriakakis E, Pfaff D, Rupp K, Philippova M, Erne P, Resink TJ. Extracellular cadherin repeat domains EC1 and EC5 of T-cadherin are essential for its ability to stimulate angiogenic behavior of endothelial cells. FASEB J 2009; 23:4011-21. [PMID: 19638398 DOI: 10.1096/fj.09-133611] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
T-cadherin (T-cad) promotes survival, proliferation, and migration of endothelial cells and induces angiogenesis. We aimed to identify domains of T-cad functionally relevant to its effects on endothelial cell behavior. To specifically target the functional properties of the 5 cadherin repeat domains (EC1-EC5) of T-cad, endothelial cells were transduced with lentivectors containing specific T-cad-domain-deletion mutant constructs (DeltaI, DeltaII, DeltaIII, DeltaIV, DeltaV). Empty (E) lentivector-transduced cells served as control. Similarly to overexpression of native T-cad, cells expressing DeltaII, DeltaIII, or DeltaIV displayed elevated levels of p-Akt and p-GSK3beta and increased proliferation rates (for DeltaII, DeltaIII) vs. E. DeltaI- and DeltaV-transduced cells exhibited reduced levels of p-Akt and p-GSK3beta and retarded growth rates vs. E. Stimulatory effects of native T-cad overexpression on Akt and GSK3beta phosphorylation were dose dependently inhibited by coexpression of DeltaI or DeltaV. Subsequent functional analyses compared only DeltaI-, DeltaII-, and DeltaV-mutant constructs with E as a negative control. Unlike DeltaII cells, DeltaI and DeltaV cells failed to exhibit homophilic ligation and deadhesion responses on a substratum of T-cad protein. In the wound assay, migration was increased for DeltaII cells but impaired for DeltaI and DeltaV cells. In endothelial cell-spheroid assay, angiogenic sprouting was augmented for DeltaII cells but inhibited for DeltaI and DeltaV cells. We conclude that EC1 and EC5 domains of T-cad are essential for its proangiogenic effects. DeltaI and DeltaV constructs may serve as dominant-negative mutants and as potential tools targeting excessive angiogenesis.
Collapse
Affiliation(s)
- Manjunath B Joshi
- Basel University Hospital, Department of Biomedicine, Laboratory for Signal Transduction, ZLF 316, Hebelstrasse 20, CH 4031 Basel, Switzerland
| | | | | | | | | | | | | |
Collapse
|
34
|
Semina EV, Rubina KA, Rutkevich PN, Voyno-Yasenetskaya TA, Parfyonova YV, Tkachuk VA. T-cadherin activates Rac1 and Cdc42 and changes endothelial permeability. BIOCHEMISTRY (MOSCOW) 2009; 74:362-70. [PMID: 19463088 DOI: 10.1134/s0006297909040026] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
In the present study, expression of T-cadherin was shown to induce intracellular signaling in NIH3T3 fibroblasts: it activated Rac1 and Cdc42 (p < 0.01) but not RhoA. T-Cadherin overexpression in human umbilical vein endothelial cells (HUVEC) using adenoviral constructs induced disassembly of microtubules and polymerization of actin stress fibers, whereas down-regulation of endogenous T-cadherin expression in HUVEC using lentiviral constructs resulted in microtubule polymerization and a decrease in the number of actin stress fibers. Moreover, suppression of the T-cadherin expression significantly decreased the endothelial monolayer permeability as compared to the control (p < 0.001).
Collapse
Affiliation(s)
- E V Semina
- Institute of Experimental Cardiology, Cardiology Research Center, Moscow, 121552, Russia
| | | | | | | | | | | |
Collapse
|
35
|
|
36
|
Org E, Eyheramendy S, Juhanson P, Gieger C, Lichtner P, Klopp N, Veldre G, Döring A, Viigimaa M, Sõber S, Tomberg K, Eckstein G, KORA, Kelgo P, Rebane T, Shaw-Hawkins S, Howard P, Onipinla A, Dobson RJ, Newhouse SJ, Brown M, Dominiczak A, Connell J, Samani N, Farrall M, BRIGHT, Caulfield MJ, Munroe PB, Illig T, Wichmann HE, Meitinger T, Laan M. Genome-wide scan identifies CDH13 as a novel susceptibility locus contributing to blood pressure determination in two European populations. Hum Mol Genet 2009; 18:2288-96. [PMID: 19304780 PMCID: PMC2685752 DOI: 10.1093/hmg/ddp135] [Citation(s) in RCA: 143] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Collaborators] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2008] [Revised: 02/04/2009] [Accepted: 03/18/2009] [Indexed: 11/13/2022] Open
Abstract
Hypertension is a complex disease that affects a large proportion of adult population. Although approximately half of the inter-individual variance in blood pressure (BP) level is heritable, identification of genes responsible for its regulation has remained challenging. Genome-wide association study (GWAS) is a novel approach to search for genetic variants contributing to complex diseases. We conducted GWAS for three BP traits [systolic and diastolic blood pressure (SBP and DBP); hypertension (HYP)] in the Kooperative Gesundheitsforschung in der Region Augsburg (KORA) S3 cohort (n = 1644) recruited from general population in Southern Germany. GWAS with 395,912 single nucleotide polymorphisms (SNPs) identified an association between BP traits and a common variant rs11646213 (T/A) upstream of the CDH13 gene at 16q23.3. The initial associations with HYP and DBP were confirmed in two other European population-based cohorts: KORA S4 (Germans) and HYPEST (Estonians). The associations between rs11646213 and three BP traits were replicated in combined analyses (dominant model: DBP, P = 5.55 x 10(-5), effect -1.40 mmHg; SBP, P = 0.007, effect -1.56 mmHg; HYP, P = 5.30 x 10(-8), OR = 0.67). Carriers of the minor allele A had a decreased risk of hypertension. A non-significant trend for association was also detected with severe family based hypertension in the BRIGHT sample (British). The novel susceptibility locus, CDH13, encodes for an adhesion glycoprotein T-cadherin, a regulator of vascular wall remodeling and angiogenesis. Its function is compatible with the BP biology and may improve the understanding of the pathogenesis of hypertension.
Collapse
Affiliation(s)
- Elin Org
- Department of Biotechnology, Institute of Molecular and Cell Biology, University of Tartu, Riia 23, 51010 Tartu, Estonia
| | - Susana Eyheramendy
- Institute of Epidemiology
- Department of Statistics, Pontificia Universidad Catolica de Chile, Vicuña Mackena 4860, Santiago, Chile
| | - Peeter Juhanson
- Department of Biotechnology, Institute of Molecular and Cell Biology, University of Tartu, Riia 23, 51010 Tartu, Estonia
| | | | - Peter Lichtner
- Institute of Human Genetics, Helmholtz Zentrum München, German Research Centre for Environmental Health, 85764 Neuherberg, Germany
| | | | - Gudrun Veldre
- Department of Biotechnology, Institute of Molecular and Cell Biology, University of Tartu, Riia 23, 51010 Tartu, Estonia
- Department of Cardiology, University of Tartu, L. Puusepa 1a, 50406 Tartu, Estonia
| | | | - Margus Viigimaa
- Centre of Cardiology, North Estonia Medical Centre, Sütiste tee 19, 13419 Tallinn, Estonia
| | - Siim Sõber
- Department of Biotechnology, Institute of Molecular and Cell Biology, University of Tartu, Riia 23, 51010 Tartu, Estonia
| | - Kärt Tomberg
- Department of Biotechnology, Institute of Molecular and Cell Biology, University of Tartu, Riia 23, 51010 Tartu, Estonia
| | - Gertrud Eckstein
- Institute of Human Genetics, Helmholtz Zentrum München, German Research Centre for Environmental Health, 85764 Neuherberg, Germany
| | | | - Piret Kelgo
- Department of Biotechnology, Institute of Molecular and Cell Biology, University of Tartu, Riia 23, 51010 Tartu, Estonia
| | - Tiina Rebane
- Department of Biotechnology, Institute of Molecular and Cell Biology, University of Tartu, Riia 23, 51010 Tartu, Estonia
| | - Sue Shaw-Hawkins
- Clinical Pharmacology and the Genome Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, London EC1M 6BQ, UK
| | - Philip Howard
- Clinical Pharmacology and the Genome Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, London EC1M 6BQ, UK
| | - Abiodun Onipinla
- Clinical Pharmacology and the Genome Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, London EC1M 6BQ, UK
| | - Richard J. Dobson
- Clinical Pharmacology and the Genome Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, London EC1M 6BQ, UK
| | - Stephen J. Newhouse
- Clinical Pharmacology and the Genome Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, London EC1M 6BQ, UK
| | - Morris Brown
- Clinical Pharmacology Unit, Addenbrookes Hospital, University of Cambridge, Cambridge CB2 2QQ, UK
| | - Anna Dominiczak
- Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow G12 8TA, UK
| | - John Connell
- Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow G12 8TA, UK
| | - Nilesh Samani
- Cardiovascular Sciences, Glenfield Hospital, University of Leicester, Groby Road, Leicester LE3 9QP, UK
| | - Martin Farrall
- Cardiovascular Medicine, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | | | - Mark J. Caulfield
- Clinical Pharmacology and the Genome Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, London EC1M 6BQ, UK
| | - Patricia B. Munroe
- Clinical Pharmacology and the Genome Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, London EC1M 6BQ, UK
| | | | - H.-Erich Wichmann
- Institute of Epidemiology
- Institute of Medical Informatics, Biometry and Epidemiology, Ludwig-Maximilians-Universität, D-81377 Munich, Germany
| | - Thomas Meitinger
- Institute of Human Genetics, Helmholtz Zentrum München, German Research Centre for Environmental Health, 85764 Neuherberg, Germany
- Institute of Human Genetics, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Maris Laan
- Department of Biotechnology, Institute of Molecular and Cell Biology, University of Tartu, Riia 23, 51010 Tartu, Estonia
| |
Collapse
Collaborators
H-E Wichmann, A Peters, C Meisinger, T Illig, R Holle, J John, Chris Wallace, Mark Lathrop, John Webster,
Collapse
|
37
|
Buechner SA, Philippova M, Erne P, Mathys T, Resink TJ. High T-cadherin expression is a feature of basal cell carcinoma. Br J Dermatol 2009; 161:199-202. [PMID: 19438454 DOI: 10.1111/j.1365-2133.2009.09195.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
38
|
Wakayama K, Shimamura M, Sata M, Koibuchi N, Sato N, Ogihara T, Morishita R. A model of cerebrovascular injury in rats. J Neurosci Methods 2008; 175:187-95. [PMID: 18786566 DOI: 10.1016/j.jneumeth.2008.08.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2007] [Revised: 07/27/2008] [Accepted: 08/04/2008] [Indexed: 12/01/2022]
Abstract
Although the pathophysiology of post-angioplasty restenosis has been extensively studied in extracranial arteries using transluminal vascular injury model in rodents, it is still not well known in the intracranial arteries, which have quite different structures from extracranial arteries. Here, we examined whether 1-min placement of modified intraluminal suture could induce an injury in the internal carotid artery (ICA) in rats and observed temporal profile of histological change after the injury. HE staining showed that the injured intracranial ICA was dilated, while the media was markedly thinned at 1 day after injury. The internal elastic lamina was not observed, and the media contained few cells. At 1 week after injury, a thin layer of neointimal hyperplasia was observed on the luminal side of the internal elastic lamina. Neointimal hyperplasia developed until at least 4 weeks after injury. Morphometric analysis demonstrated that the healing process of the injury was related to arterial remodeling. Immunohistochemical staining for alpha-smooth muscle actin and electron microscopic analysis showed that the neointima was composed of smooth muscle cells. Re-endothelialization was observed from 1 to 4 weeks after injury by immunohistochemical staining for von Willebrand's factor and electron microscopic analysis. Vascular endothelial growth factor was expressed in neointima on days 7 and 14. Interestingly, superoxide anion was not increased in injured arteries on day 3, when the infiltration of macrophages was intensive, but increased on day 7, when infiltrating macrophages almost disappeared. These findings might shed new light on pathophysiology of post-angioplasty restenosis in intracranial arteries.
Collapse
Affiliation(s)
- Kouji Wakayama
- Department of Advanced Clinical Science and Therapeutics, Graduate School of Medicine, University of Tokyo, Japan
| | | | | | | | | | | | | |
Collapse
|
39
|
Il’inskaya OP, Antropova YG, Kalinina NI, Mishina VA, Radyukhina NV, Tararak EM. Detection of bone marrow-derived cells in neointimal thickening in the rat carotid artery by nested polymerase chain reaction. Russ J Dev Biol 2008. [DOI: 10.1134/s1062360408040048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
40
|
Chan DW, Lee JMF, Chan PCY, Ng IOL. Genetic and epigenetic inactivation of T-cadherin in human hepatocellular carcinoma cells. Int J Cancer 2008; 123:1043-52. [PMID: 18553387 DOI: 10.1002/ijc.23634] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
T-cadherin is an atypical cadherin and growing evidence has indicated that T-cadherin exerts tumor-suppressive effects on cancers of epithelial cell type and also causes positive effects on tumor angiogenesis. Human hepatocellular carcinoma (HCC) is a hypervascular tumor and T-cadherin has been shown to be overexpressed in intratumoral endothelial cells of HCCs. However, the expression status and functions of T-cadherin in hepatocytes or HCC cells remain unclear. Here, we demonstrated that T-cadherin was underexpressed in HCC cells (26.5%, 13/49 cases), but was frequently (77.6%, 38/49) overexpressed in intratumoral endothelial cells immunohistochemically. Semiquantitative RT-PCR analysis also showed that the T-cadherin gene was underexpressed in 7 of 11 HCC cell lines. Loss of heterozygosity analysis revealed that 32-38% of the 42 human HCC samples had allelic losses at this locus. Upon pharmacological treatment with demethylating agent 5-aza-2'-deoxycytidine or histone deacetylase inhibitor trichostatin A, T-cadherin promoter hypermethylation and/or histone deacetylation was frequently observed in HCC samples and cell lines. Functionally, enforced expression of T-cadherin induced G(2)/M cell cycle arrest, reduced cell proliferation in low serum medium, suppressed anchorage-independent growth in soft agar and increased sensitivity to TNFalpha-mediated apoptosis in HCC cells. Intriguingly, we found that T-cadherin significantly suppressed the activity of c-Jun, a crucial oncoprotein constitutively activated in HCC cells. To conclude, T-cadherin was differentially expressed in human HCCs. The underexpression of T-cadherin in HCC cells suggests it may be another critical event in addition to T-cadherin-mediated angiogenesis during HCC development.
Collapse
Affiliation(s)
- David W Chan
- Liver Cancer and Hepatitis Research Laboratory and SH Ho Foundation Research Laboratories, Department of Pathology, The University of Hong Kong, Hong Kong, China
| | | | | | | |
Collapse
|
41
|
Identification of proteins associating with glycosylphosphatidylinositol- anchored T-cadherin on the surface of vascular endothelial cells: role for Grp78/BiP in T-cadherin-dependent cell survival. Mol Cell Biol 2008; 28:4004-17. [PMID: 18411300 DOI: 10.1128/mcb.00157-08] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
There is scant knowledge regarding how cell surface lipid-anchored T-cadherin (T-cad) transmits signals through the plasma membrane to its intracellular targets. This study aimed to identify membrane proteins colocalizing with atypical glycosylphosphatidylinositol (GPI)-anchored T-cad on the surface of endothelial cells and to evaluate their role as signaling adaptors for T-cad. Application of coimmunoprecipitation from endothelial cells expressing c-myc-tagged T-cad and high-performance liquid chromatography revealed putative association of T-cad with the following proteins: glucose-related protein GRP78, GABA-A receptor alpha1 subunit, integrin beta3, and two hypothetical proteins, LOC124245 and FLJ32070. Association of Grp78 and integrin beta3 with T-cad on the cell surface was confirmed by surface biotinylation and reciprocal immunoprecipitation and by confocal microscopy. Use of anti-Grp78 blocking antibodies, Grp78 small interfering RNA, and coexpression of constitutively active Akt demonstrated an essential role for surface Grp78 in T-cad-dependent survival signal transduction via Akt in endothelial cells. The findings herein are relevant in the context of both the identification of transmembrane signaling partners for GPI-anchored T-cad as well as the demonstration of a novel mechanism whereby Grp78 can influence endothelial cell survival as a cell surface signaling receptor rather than an intracellular chaperone.
Collapse
|
42
|
Shibata R, Ouchi N, Walsh K, Murohara T. Potential of adiponectin as a cardioprotective agent. Future Cardiol 2007; 3:647-56. [DOI: 10.2217/14796678.3.6.647] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
In this review, we focus on the role of adiponectin as a cardioprotective agent in several pathological heart conditions. Obesity is closely associated with Type 2 diabetes, hypertension and heart disease. Adiponectin is an adipose tissue-derived hormone whose concentration is downregulated in subjects with obesity-related diseases. Hypoadiponectinemia has been identified as an independent risk factor for Type 2 diabetes, coronary artery disease, acute coronary syndrome and hypertension. More recent experimental findings have shown that adiponectin directly affects signaling in cardiac myocytes and has beneficial effects on several pathological heart conditions, including cardiac hypertrophy and myocardial infarction. The favorable effects of adiponectin are associated with attenuated inflammatory response, decreased myocyte death, decreased hypertrophic response, maintained ischemia-induced angiogenesis and reduced interstitial fibrosis. Therefore, adiponectin could represent a molecular target for treating obesity-linked cardiac diseases.
Collapse
Affiliation(s)
- Rei Shibata
- Nagoya University Graduate School of Medicine, Department of Cardiology, 65 Tsurumai, Showa, Nagoya, 466–8550, Japan
| | - Noriyuki Ouchi
- Boston University School of Medicine, Molecular Cardiology/Whitaker Cardiovascular Institute, 715 Albany Street, W611, Boston, MA 02118, USA
| | - Kenneth Walsh
- Boston University School of Medicine, Molecular Cardiology/Whitaker Cardiovascular Institute, 715 Albany Street, W611, Boston, MA 02118, USA
| | - Toyoaki Murohara
- Nagoya University Graduate School of Medicine, Department of Cardiology, 65 Tsurumai, Showa, Nagoya, 466–8550, Japan
| |
Collapse
|
43
|
Rubina K, Kalinina N, Potekhina A, Efimenko A, Semina E, Poliakov A, Wilkinson DG, Parfyonova Y, Tkachuk V. T-cadherin suppresses angiogenesis in vivo by inhibiting migration of endothelial cells. Angiogenesis 2007; 10:183-95. [PMID: 17486418 DOI: 10.1007/s10456-007-9072-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2006] [Accepted: 02/28/2007] [Indexed: 01/26/2023]
Abstract
Our previous studies have revealed the abundant expression of T-cadherin--a glycosylphosphatidylinositol (GPI)-anchored member of cadherin superfamily--in endothelial and mural cells in the heart and vasculature. The upregulation of T-cadherin in vascular proliferative disorders such as atherosclerosis and restenosis suggests the involvement of T-cadherin in vascular growth and remodeling. However, the functional significance of this molecule in the vasculature remains unknown. The effect of T-cadherin on angiogenesis in vivo was evaluated using Matrigel implant model. We demonstrate that T-cadherin overexpression in L929 cells injected in Matrigel inhibits neovascularization of the plug. In vitro T-cadherin inhibits the directional migration of endothelial cells, capillary growth, and tube formation but has no effect on endothelial cell proliferation, adhesion, or apoptosis in vitro. These data suggest that T-cadherin expressed in the stroma could act as a negative guidance cue for the ingrowing blood vessels and thus could have an important potential therapeutic application.
Collapse
Affiliation(s)
- Kseniya Rubina
- Department of Biological and Medical Chemistry, Faculty of Fundamental Medicine, Lomonosov Moscow State University, 31-5, Lomonosovsky av., Moscow, 119192, Russia.
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Joshi MB, Ivanov D, Philippova M, Erne P, Resink TJ. Integrin-linked kinase is an essential mediator for T-cadherin-dependent signaling via Akt and GSK3beta in endothelial cells. FASEB J 2007; 21:3083-95. [PMID: 17485554 DOI: 10.1096/fj.06-7723com] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Glycosylphosphatidylinositol-anchored T-cadherin (T-cad) influences several parameters of angiogenesis including endothelial cell (EC) differentiation, migration, proliferation, and survival. This presupposes signal transduction networking via mediatory regulators and molecular adaptors since T-cad lacks transmembrane and cytosolic domains. Here, using pharmacological inhibition of PI3K, adenoviral-mediated T-cad-overexpression, siRNA-mediated T-cad-depletion, and agonistic antibody-mediated ligation, we demonstrate signaling by T-cad through PI3K-Akt-GSK3beta pathways in EC. T-cad-overexpressing EC exhibited increased levels and nuclear accumulation of active beta-catenin, which was transcriptionally active as shown by increased Lef/Tcf reporter activity and cyclin D1 levels. Cotransduction of EC with constitutively active GSK3beta (S9A-GSK3beta) abrogated the stimulatory effects of T-cad on active beta-catenin accumulation, proliferation, and survival. Integrin-linked kinase (ILK), a membrane proximal upstream regulator of Akt and GSK3beta, was considered a candidate signaling mediator for T-cad. T-cad was present in anti-ILK immunoprecipitates, and confocal microscopy revealed colocalization of T-cad and ILK within lamellipodia of migrating cells. ILK-siRNA abolished T-cad-dependent effects on (Ser-473)Akt/(Ser-9)GSK3beta phosphorylation, active beta-catenin accumulation, and survival. We conclude ILK is an essential mediator for T-cad signaling via Akt and GSK3beta in EC. This is the first demonstration that ILK can regulate inward signaling by GPI-anchored proteins. Furthermore, ILK-GSK3beta-dependent modulation of active beta-catenin levels by GPI-anchored T-cad represents a novel mechanism for controlling cellular beta-catenin activity.
Collapse
Affiliation(s)
- Manjunath B Joshi
- Department of Research, Cardiovascular Laboratories, Basel University Hospital, Hebelstrasse 20, CH 4031 Basel, Switzerland
| | | | | | | | | |
Collapse
|
45
|
Bora PS, Kaliappan S, Lyzogubov VV, Tytarenko RG, Thotakura S, Viswanathan T, Bora NS. Expression of adiponectin in choroidal tissue and inhibition of laser induced choroidal neovascularization by adiponectin. FEBS Lett 2007; 581:1977-82. [PMID: 17466298 DOI: 10.1016/j.febslet.2007.04.024] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2007] [Revised: 04/03/2007] [Accepted: 04/05/2007] [Indexed: 02/06/2023]
Abstract
The aim of this study was to investigate the role of adiponectin (APN) in a mouse model of laser induced choroidal neovascularization (CNV). We have shown by immunohistochemistry that the expression of APN, adiponectin receptor 1, adiponectin receptor 2 and T cadherin gradually increased from day 1 to day 7 post-laser in laser treated mice compared to controls. Recombinant APN (rAPN) was injected intraperitoneally (i.p., 25 microg/mouse) or intravitreally (2 microg/eye) in lasered mice. Another set of lasered mice received APN peptide via i.p. (75 microg/mouse) or intravitreal (30 microg/eye) route. Control mice received a similar treatment with PBS, control protein or control peptide after laser treatment. We found that in the i.p. and intravitreal injection of rAPN resulted in 78% and 68% inhibition respectively in the size of CNV complex compared to control mice. Similar results were observed when APN peptide was injected intravitreally or i.p. Treatment with rAPN or the peptide resulted in decreased levels of vascular endothelial growth factor. Thus, APN inhibited choroidal angiogenesis and may have therapeutic implications in the treatment of wet age related macular degeneration.
Collapse
Affiliation(s)
- Puran S Bora
- Department of Ophthalmology, Jones Eye Institute, Pat & Willard Walker Eye Research Center, 4301 West Markham, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | | | | | | | | | | | | |
Collapse
|
46
|
Monahan TS, Andersen ND, Panossian H, Kalish JA, Daniel S, Shrikhande GV, Ferran C, Logerfo FW. A novel function for cadherin 11/osteoblast-cadherin in vascular smooth muscle cells: modulation of cell migration and proliferation. J Vasc Surg 2007; 45:581-9. [PMID: 17321345 DOI: 10.1016/j.jvs.2006.12.016] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2006] [Accepted: 12/03/2006] [Indexed: 01/06/2023]
Abstract
OBJECTIVE Intimal hyperplasia is a common cause of vein graft failure in cardiovascular surgery. The molecular basis for intimal hyperplasia remains poorly defined. We have previously identified, by gene chip analysis of vein grafts, increased messenger (mRNA) for the adhesion molecule cadherin 11/osteoblast-cadherin (CDH11). The function of CDH11 in vascular cells is unknown. The aim of the present study is to confirm CDH11 expression in vein grafts and characterize its role in vascular remodeling. METHODS Cephalic vein interposition grafts were implanted in a canine model and harvested at predetermined time points. CDH11 protein expression was determined by immunohistochemistry. Early passage human coronary artery smooth muscle cells (SMCs) were used for in vitro studies. Real-time polymerase chain reaction was used to assess cellular CDH11 mRNA levels. CDH11 signaling was inhibited by either transfection with silencing RNA targeting CDH11 or with a blocking antibody to CDH11. Cellular migration was evaluated and cellular proliferation was assessed. RESULTS Expression of CDH11 was increased in medial SMCs of vein grafts recovered at 7, 14, and 30 days after surgery compared with control veins from the same animals. In vitro CDH11 mRNA was up-regulated 1.8 +/- 0.2-fold (P = .003) in SMCs after treatment with tumor necrosis factor-alpha. Cellular migration was attenuated by inhibition of CDH11 both with a blocking antibody (0.67 +/- 0.09; P = .063) and gene knockdown mediated by small interfering RNA (0.67 +/- 0.14; P = .036). SMC proliferation decreased by 3.1-fold (P = .006) in the presence of CDH11-blocking antibody. Knockdown of CDH11 mediated by small interfering RNA resulted in a 1.3-fold (P = .018) decrease in proliferation. CONCLUSIONS CDH11 is up-regulated in SMC in vivo and in vitro as part of the response to injury. Inhibition of CDH11 decreases SMC migration and proliferation, two pathogenic effectors of intimal hyperplasia.
Collapse
MESH Headings
- Anastomosis, Surgical
- Animals
- Antibodies/pharmacology
- Cadherins/biosynthesis
- Cadherins/genetics
- Cadherins/immunology
- Cell Cycle/drug effects
- Cell Movement
- Cell Proliferation
- Cells, Cultured
- Coronary Vessels/metabolism
- Coronary Vessels/physiopathology
- Dogs
- Femoral Artery/surgery
- Humans
- Hyperplasia
- Models, Animal
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/physiopathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- RNA Interference
- RNA, Messenger/metabolism
- RNA, Small Interfering/metabolism
- Time Factors
- Tumor Necrosis Factor-alpha/pharmacology
- Up-Regulation
- Veins/metabolism
- Veins/physiopathology
- Veins/transplantation
Collapse
Affiliation(s)
- Thomas S Monahan
- Department of Surgery, Division of Vascular Surgery, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Riou P, Saffroy R, Chenailler C, Franc B, Gentile C, Rubinstein E, Resink T, Debuire B, Piatier-Tonneau D, Lemoine A. Expression of T-cadherin in tumor cells influences invasive potential of human hepatocellular carcinoma. FASEB J 2006; 20:2291-301. [PMID: 17077306 DOI: 10.1096/fj.06-6085com] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Overexpression of T-cadherin (T-cad) transcripts occurs in approximately 50% of human hepatocellular carcinomas (HCCs). To elucidate T-cad functions in HCC, we examined T-cad protein expression in normal and tumoral human livers and hepatoma cell lines and investigated its influence on invasive potential of HCC using RNA interference silencing of T-cad expression in Mahlavu cells. Whereas T-cad expression was restricted to endothelial cells (EC) from large blood vessels in normal livers, it was up-regulated in sinusoidal EC from 8/15 invasive HCCs. Importantly, in three of them (38%) T-cad was detected in tumor cells within regions in which E-cadherin expression was absent. Among six hepatoma cell lines, only Mahlavu expressed T-cad but not E-cadherin. T-cad exhibited a globally punctuate distribution in quiescent Mahlavu and additionally it concentrated at the leading edge of migrating cells. Matrigel invasion assay revealed that Mahlavu possess a high invasive potential that was significantly inhibited by T-cad silencing. Wound healing and random motility assays demonstrated that inhibition of T-cad expression in Mahlavu significantly reduced their motility. We propose that T-cad expression in tumor cells might occur by cadherin-switching during epithelial-mesenchymal transition and may represent an additional mechanism contributing to HCC metastasis.
Collapse
|
48
|
Philippova M, Banfi A, Ivanov D, Gianni-Barrera R, Allenspach R, Erne P, Resink T. Atypical GPI-anchored T-cadherin stimulates angiogenesis in vitro and in vivo. Arterioscler Thromb Vasc Biol 2006; 26:2222-30. [PMID: 16873731 DOI: 10.1161/01.atv.0000238356.20565.92] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
OBJECTIVE T-cadherin (T-cad) is an atypical GPI-anchored member of the cadherin superfamily. In vascular tissue, T-cad expression is increased during atherosclerosis, restenosis, and tumor neovascularization. In vitro, overexpression and/or homophilic ligation of T-cad on endothelial cells (ECs) facilitates migration, proliferation, and survival. This study investigated T-cad effects on angiogenesis. METHODS AND RESULTS In vitro, T-cad homophilic ligation induced arrangement of ECs into a capillary-like network in a 2-dimensional model of EC differentiation and stimulated in-gel endothelial sprout outgrowth in an EC spheroid model and a modified Nicosia tissue assay. Sprouting from spheroids composed of adenoviral-infected T-cad overexpressing ECs or T-cad siRNA transfected ECs were significantly increased or reduced, respectively. In vivo, T-cad potentiated VEGF effects on neovascularization in a model of myoblast-mediated gene transfer to mouse skeletal muscle; vessel caliber after co-delivery of T-cad and VEGF was significantly greater than after delivery of VEGF alone. CONCLUSIONS We unequivocally identify T-cad as a novel modulator of angiogenesis and suggest that this molecule can be exploited as a target for modulation of therapeutic angiogenesis, as well as for prevention of pathological conditions associated with abnormal neovascularization.
Collapse
Affiliation(s)
- Maria Philippova
- Cardiovascular Signalling Group, Basel University Hospital, Hebelstrasse 20, CH-4031 Basel, Switzerland.
| | | | | | | | | | | | | |
Collapse
|
49
|
Kipmen-Korgun D, Osibow K, Zoratti C, Schraml E, Greilberger J, Kostner GM, Jürgens G, Graier WF. T-cadherin mediates low-density lipoprotein-initiated cell proliferation via the Ca(2+)-tyrosine kinase-Erk1/2 pathway. J Cardiovasc Pharmacol 2005; 45:418-30. [PMID: 15821437 DOI: 10.1097/01.fjc.0000157458.91433.86] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The GPI-anchored protein T-cadherin was found to be an atypical LDL binding site that is expressed in various types of cells, including endothelial cells, smooth muscle cells, and neurons. Notably, the expression of T-cadherin was reduced in numerous types of cancers, although it was up-regulated in tumor-penetrating blood vessels, atherosclerotic lesions, and during neointima formation. Despite these intriguing findings, our knowledge of the physiological role and the signal transduction pathways associated with this protein is limited. Therefore, T-cadherin was overexpressed in the human umbilical vein-derived endothelial cell line EA.hy926, the human embryonic kidney cell line HEK293, and LDL-initiated signal transduction, and its consequences were elucidated. Our data revealed that T-cadherin serves as a receptor specifically for LDL. Following LDL binding to T-cadherin, mitogenic signal transduction was initiated that involved activation of PLC and IP3 formation, which subsequently yielded intracellular Ca2+ mobilization. Downstream to these early phenomena, activation of tyrosine kinase(s) Erk 1/2 kinase, and the translocation of NF kappa B toward the nucleus were found. Finally, overexpression of T-cadherin in HEK293 cells resulted in accelerated cell proliferation in an LDL-dependent manner, although cell viability was not influenced. Because LDL uptake was not facilitated by T-cadherin, our data suggest that T-cadherin serves as a signaling receptor for LDL that facilitates an LDL-dependent mitogenic signal in the vasculature.
Collapse
Affiliation(s)
- Dijle Kipmen-Korgun
- Institute of Molecular Biology & Biochemistry, Center of Molecular Medicine, Medical University Graz, Graz, Austria
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Joshi MB, Philippova M, Ivanov D, Allenspach R, Erne P, Resink TJ. T-cadherin protects endothelial cells from oxidative stress-induced apoptosis. FASEB J 2005; 19:1737-9. [PMID: 16099944 DOI: 10.1096/fj.05-3834fje] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
In vascular tissue, T-cadherin (T-cad) is up-regulated in vivo under disease conditions associated with oxidative stress and concomitant cell migration, proliferation and apoptosis/survival. Using cultures of human umbilical vein endothelial cells (HUVEC), we examined whether there is a functional relationship between oxidative stress, T-cad expression, and cell survival status. Culture of HUVEC under conditions of oxidative stress (e.g., serum deprivation, inclusion of H2O2) resulted in increased T-cad expression. Oxidative stress-induced increases in T-cad were inhibited by the free radical-scavenging antioxidant, N-acetylcysteine, and the flavin-containing oxidase inhibitor, diphenyleneiodonium. Thus reactive oxygen species (ROS) contribute to stress-induced elevation of T-cad in HUVEC. Compared with control cells, HUVEC overexpressing T-cad (T-cad+-HUVEC) had higher phosphorylation levels for phosphatidylinositol 3-kinase (PI3K) target Akt and mTOR target p70(S6K) (survival pathway regulators), but lower levels for p38MAPK (death pathway regulator). T-cad+-HUVEC exposed to stress (serum-deprivation, TNF-alpha, actinomycin D, staurosporine) exhibited reduced caspase activation together with increased cell survival. Protection against stress-induced apoptosis in T-cad+-HUVEC was abrogated by either PI3K-inhibitor wortmannin or mTOR-inhibitor rapamycin. We conclude that T-cad overexpression in HUVEC protects against stress-induced apoptosis through activation of the PI3K/Akt/mTOR survival signal pathway and concomitant suppression of the p38 MAPK proapoptotic pathway. ROS-induced changes in T-cad expression may play an important role in controlling tissue cellularity during vascular remodeling.
Collapse
Affiliation(s)
- Manjunath B Joshi
- Department of Research, Cardiovascular Laboratories, Basel University Hospital, Basel, Switzerland
| | | | | | | | | | | |
Collapse
|