1
|
Veshkini A, Kühn C, Dengler F, Bachmann L, Liermann W, Helm C, Ulrich R, Delling C, Hammon HM. Cryptosporidium parvum infection alters the intestinal mucosa transcriptome in neonatal calves: impacts on epithelial barriers and transcellular transport systems. Front Cell Infect Microbiol 2024; 14:1495309. [PMID: 39703373 PMCID: PMC11656319 DOI: 10.3389/fcimb.2024.1495309] [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: 09/12/2024] [Accepted: 11/18/2024] [Indexed: 12/21/2024] Open
Abstract
Introduction Cryptosporidium parvum (C. parvum) is the most prevalent enteric protozoan parasite causing infectious diarrhea in neonatal calves worldwide with a direct negative impact on their health and welfare. This study utilized next-generation sequencing (NGS) to deepen our understanding of intestinal epithelial barriers and transport mechanisms in the pathophysiology of infectious diarrhea in neonatal calves, which could potentially unveil novel solutions for treatment. Methods At day 1 of life, male Holstein-Friesian calves were either orally infected (n = 5) or not (control group, n = 5) with C. parvum oocysts (in-house strain LE-01-Cp-15). On day 8 after infection, calves were slaughtered and jejunum mucosa samples were taken. The RNA was extracted from collected samples and subjected to sequencing. Differentially expressed genes (DEG) between the infected and CTRL groups were assessed using DESeq2 at a false discovery rate < 0.05 and used for gene ontology (GO) and pathway enrichment analysis in Cytoscape (v3.9.1). Results and discussion To study the pathophysiology of infectious diarrhea on intestinal permeability, 459 genes related to epithelial cell barrier integrity and paracellular and transmembrane transport systems were selected from 12,908 identified genes in mucus. Among, there were 61 increased and 109 decreased gene transcripts belonged to adhesion molecules (e.g. ADGRD1 and VCAM1), ATP-binding cassette (ABC, e.g. ABCC2 and ABCD1) and solute carrier (SLC, e.g. SLC28A2 and SLC38A3) transporters, and ion channels (e.g. KCNJ15). Our results suggest deregulation of cellular junctions and thus a possibly increased intestinal permeability, whereas deregulation of ABC and SLC transporters and ion channels may influence the absorption/secretion of amino acids, carbohydrates, fats, and organic compounds, as well as acid-based balance and osmotic hemostasis. Besides pathogen-induced gene expression alterations, part of the DEG may have been triggered or consequently affected by inflammatory mechanisms. The study provided a deeper understanding of the pathophysiology of infectious diarrhea in neonatal calves and the host-pathogen interactions at the transcript level. For further studies with a particular focus on the transport system, these results could lead to a new approach to elucidating pathophysiological regulatory mechanisms.
Collapse
Affiliation(s)
- Arash Veshkini
- Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Christa Kühn
- Friedrich-Loeffler-Institute, Greifswald-Insel Riems, Germany
- Agricultural and Environmental Faculty, University Rostock, Rostock, Germany
| | - Franziska Dengler
- Institute of Animal Sciences, University of Hohenheim, Hohenheim, Germany
| | - Lisa Bachmann
- Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
- Faculty of Agriculture and Food Science, University of Applied Science Neubrandenburg, Neubrandenburg, Germany
| | - Wendy Liermann
- Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Christiane Helm
- Institute for Veterinary Pathology, Leipzig University, Leipzig, Germany
| | - Reiner Ulrich
- Institute for Veterinary Pathology, Leipzig University, Leipzig, Germany
| | - Cora Delling
- Institute of Veterinary Parasitology, Leipzig University, Leipzig, Germany
| | - Harald M. Hammon
- Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| |
Collapse
|
2
|
Zuo YB, Wen ZJ, Cheng MD, Jia DD, Zhang YF, Yang HY, Xu HM, Xin H, Zhang YF. The pro-atherogenic effects and the underlying mechanisms of chronic bisphenol S (BPS) exposure in apolipoprotein E-deficient mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 285:117133. [PMID: 39342757 DOI: 10.1016/j.ecoenv.2024.117133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/23/2024] [Accepted: 09/26/2024] [Indexed: 10/01/2024]
Abstract
Atherosclerosis (AS) and its related cardiovascular diseases (CVDs) remain the most frequent cause of morbidity and mortality worldwide. Researches showed that bisphenol A (BPA) exposure might exacerbate AS progression. However, as an analogue of BPA, little is known about the cardiovascular toxicity of bisphenol S (BPS), especially whether BPS exposure has the pro-atherogenic effects in mammals is still unknown. Here, we firstly constructed an apolipoprotein E knockout (ApoE-/-) mouse model and cultured cells to investigate the risk of BPS on AS and explore the underlying mechanisms. Results showed that prolonged exposure to 50 μg/kg body weight (bw)/day BPS indeed aggravated AS lesions both in the en face aortas and aortic sinuses of ApoE-/- mice. Moreover, BPS were found to be implicated in the AS pathological process: 1) stimulates adhesion molecule expression to promote monocyte-endothelial cells (ECs) adhesion with 3.6 times more than the control group in vivo; 2) increases the distribution of vascular smooth muscle cells (VSMCs) with 9.3 times more than the control group in vivo, possibly through the migration of VSMCs; and 3) induces an inflammatory response by increasing the number of macrophages (MACs), with 3.7 times more than the control group in vivo, and the release of inflammatory mediators. Furthermore, we have identified eight significant AS-related genes induced by BPS, including angiopoietin-like protein 7 (Angptl17) and lipocalin-2 (Lcn2) in ECs; matrix metalloproteinase 9 (Mmp13), secreted phosphoprotein 1 (Spp1), and collagen type II alpha 1 (Col2a1) in VSMCs; and kininogen 1 (Kng1), integrin alpha X (Itgax), and MAC-expressed gene 1 (Mpeg1) in MACs. Overall, this study firstly found BPS exposure could exacerbate mammalian AS and might also provide a theoretical basis for elucidating BPS and its analogues induced AS and related CVDs.
Collapse
Affiliation(s)
- Ying-Bing Zuo
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China; Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao University, 16 Jiangsu Road, Qingdao 266000, China
| | - Zeng-Jin Wen
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China
| | - Meng-Die Cheng
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China; Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao University, 16 Jiangsu Road, Qingdao 266000, China
| | - Dong-Dong Jia
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China
| | - Yi-Fei Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China
| | - Hong-Yu Yang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China
| | - Hai-Ming Xu
- School of Public Health, Ningxia Medical University, Yinchuan, Ningxia 750004, China.
| | - Hui Xin
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao University, 16 Jiangsu Road, Qingdao 266000, China.
| | - Yin-Feng Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China.
| |
Collapse
|
3
|
Li J, Lu Z, Xu L, Wang J, Qian S, Hu Q, Ge Y. Poly(ethylenimine)-Cyclodextrin-Based Cationic Polymer Mediated HIF-1α Gene Delivery for Hindlimb Ischemia Treatment. ACS APPLIED BIO MATERIALS 2024; 7:1081-1094. [PMID: 38294873 DOI: 10.1021/acsabm.3c01020] [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] [Indexed: 02/01/2024]
Abstract
Hindlimb ischemia is a common disease worldwide featured by the sudden decrease in limb perfusion, which usually causes a potential threat to limb viability and even amputation or death. Revascularization has been defined as the gold-standard therapy for hindlimb ischemia. Considering that vascular injury recovery requires cellular adaptation to the hypoxia, hypoxia-inducible factor 1 α (HIF-1α) is a potential gene for tissue restoration and angiogenesis. In this manuscript, effective gene delivery vector PEI-β-CD (PC) was reported for the first application in the hindlimb ischemia treatment to deliver HIF-1α plasmid in vitro and in vivo. Our in vitro finding demonstrated that PC/HIF-1α-pDNA could be successfully entered into the cells and mediated efficient gene transfection with good biocompatibility. More importantly, under hypoxic conditions, PC/HIF-1α-pDNA could up-regulate the HUEVC cell viability. In addition, the mRNA levels of VEGF, Ang-1, and PDGF were upregulated, and transcriptome results also demonstrated that the cell-related function of response to hypoxia was enhanced. The therapeutic effect of PC/HIF-1α-pDNA was further estimated in a murine acute hindlimb ischemia model, which demonstrated that intramuscular injection of PC/HIF-1α-pDNA resulted in significantly increased blood perfusion and alleviation in tissue damage, such as tissue fibrosis and inflammation. The results provide a rationale that HIF-1α-mediated gene therapy might be a practical strategy for the treatment of limb ischemia.
Collapse
Affiliation(s)
- Jingyu Li
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Zhuoting Lu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Liwang Xu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Jing Wang
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 314408, China
| | - Shaojie Qian
- Center for Rehabilitation Medicine, Department of Anesthesiology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 314408, China
| | - Qinglian Hu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Yunfen Ge
- Center for Rehabilitation Medicine, Department of Anesthesiology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 314408, China
| |
Collapse
|
4
|
Curry CW, Sturgeon SM, O'Grady BJ, Yates A, Kjar A, Paige H, Mowery LS, Katdare KA, Patel R, Mlouk K, Stiefbold MR, Vafaie-Partin S, Kawabata A, McKee R, Moore-Lotridge S, Hawkes A, Kusunose J, Gibson-Corley KN, Schmeckpeper J, Schoenecker JG, Caskey CF, Lippmann ES. Growth factor free, peptide-functionalized gelatin hydrogel promotes arteriogenesis and attenuates tissue damage in a murine model of critical limb ischemia. Biomaterials 2023; 303:122397. [PMID: 37979513 PMCID: PMC10843678 DOI: 10.1016/j.biomaterials.2023.122397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 11/02/2023] [Accepted: 11/10/2023] [Indexed: 11/20/2023]
Abstract
Critical limb ischemia (CLI) occurs when blood flow is restricted through the arteries, resulting in ulcers, necrosis, and chronic wounds in the downstream extremities. The development of collateral arterioles (i.e. arteriogenesis), either by remodeling of pre-existing vascular networks or de novo growth of new vessels, can prevent or reverse ischemic damage, but it remains challenging to stimulate collateral arteriole development in a therapeutic context. Here, we show that a gelatin-based hydrogel, devoid of growth factors or encapsulated cells, promotes arteriogenesis and attenuates tissue damage in a murine CLI model. The gelatin hydrogel is functionalized with a peptide derived from the extracellular epitope of Type 1 cadherins. Mechanistically, these "GelCad" hydrogels promote arteriogenesis by recruiting smooth muscle cells to vessel structures in both ex vivo and in vivo assays. In a murine femoral artery ligation model of CLI, delivery of in situ crosslinking GelCad hydrogels was sufficient to restore limb perfusion and maintain tissue health for 14 days, whereas mice treated with gelatin hydrogels had extensive necrosis and autoamputated within 7 days. A small cohort of mice receiving the GelCad hydrogels were aged out to 5 months and exhibited no decline in tissue quality, indicating durability of the collateral arteriole networks. Overall, given the simplicity and off-the-shelf format of the GelCad hydrogel platform, we suggest it could have utility for CLI treatment and potentially other indications that would benefit from arteriole development.
Collapse
Affiliation(s)
- Corinne W Curry
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Sarah M Sturgeon
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Brian J O'Grady
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Alexis Yates
- Interdisciplinary Materials Science Program, Vanderbilt University, Nashville, TN, USA
| | - Andrew Kjar
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Hayden Paige
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Lucas S Mowery
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Ketaki A Katdare
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
| | - Riya Patel
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Kate Mlouk
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
| | - Madison R Stiefbold
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Sidney Vafaie-Partin
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Atsuyuki Kawabata
- Department of Orthopedics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Rachel McKee
- Department of Orthopedics, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Adrienne Hawkes
- Department of Radiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jiro Kusunose
- Department of Radiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Katherine N Gibson-Corley
- Department of Pathology, Microbiology and Immunology, Division of Comparative Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jeffrey Schmeckpeper
- Department of Cardiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Charles F Caskey
- Department of Radiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ethan S Lippmann
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA; Interdisciplinary Materials Science Program, Vanderbilt University, Nashville, TN, USA; Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
5
|
Liman N, Sağsöz H. The immunolocalization of cadherins and beta-catenin in the cervix and vagina of cycling cows. Vet Res Commun 2023; 47:1155-1175. [PMID: 36729278 DOI: 10.1007/s11259-023-10075-4] [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: 03/02/2022] [Accepted: 01/14/2023] [Indexed: 02/03/2023]
Abstract
The adherens junctions (AJs) maintain the epithelial cell layers' structural integrity and barrier function. AJs also play a vital role in various biological and pathological processes. AJs perform these functions through the cadherin-catenin adhesion complex. This study investigated the presence, cell-specific localization, and temporal distribution of AJ components such as classical type I cadherins and beta-catenin in the cow cervix and vagina during the estrous cycle. Immunohistochemistry and Western blot analysis results demonstrated that beta-catenin and epithelial (E)-, neural (N)-, and placental (P)-cadherins are expressed in the cow cervix and vagina during the estrous cycle. These adhesion molecules were localized in the membrane and cytoplasm of the ciliated and non-ciliated cervical cells and the stratified vaginal epithelial cells. Positive immunostaining for P-, N-cadherin, and beta-catenin was also observed in the vascular endothelial cells of the cervical and vaginal stroma. Quantitative immunohistochemistry examinations revealed that in the cervical and vaginal epithelia, P-cadherin's optical density values (ODv) were the highest; in contrast, the N-cadherin ODv were the lowest. The ODv of P-cadherin and beta-catenin in the cervical epithelium and E-cadherin in the vagina were significantly higher in the luteal phase versus the follicular phase of the estrous cycle. Furthermore, the ODv of P-cadherin, N-cadherin, and beta-catenin in the cervix's central and peripheral epithelial regions were different during the estrous cycle. These findings indicate that classical cadherins and beta-catenin in the cervix and vagina exhibit cell- and tissue-specific expression patterns under the influence of estrogen and progesterone hormones during the estrous cycle.
Collapse
Affiliation(s)
- Narin Liman
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Erciyes University, 38039, Kayseri, Turkey.
| | - Hakan Sağsöz
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Dicle University, Diyarbakır, Turkey
| |
Collapse
|
6
|
Curry CW, Sturgeon SM, O’Grady BJ, Yates AK, Kjar A, Paige HA, Mowery LS, Katdare KA, Patel RV, Mlouk K, Stiefbold MR, Vafaie-Partin S, Kawabata A, McKee RM, Moore-Lotridge S, Hawkes A, Kusunose J, Gibson-Corley KN, Schmeckpeper J, Schoenecker JG, Caskey CF, Lippmann ES. Growth factor-free, peptide-functionalized gelatin hydrogel promotes arteriogenesis and attenuates tissue damage in a murine model of critical limb ischemia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.24.542150. [PMID: 37292898 PMCID: PMC10245920 DOI: 10.1101/2023.05.24.542150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Critical limb ischemia (CLI) occurs when blood flow is restricted through the arteries, resulting in ulcers, necrosis, and chronic wounds in the downstream extremities. The development of collateral arterioles (i.e. arteriogenesis), either by remodeling of pre-existing vascular networks or de novo growth of new vessels, can prevent or reverse ischemic damage, but it remains challenging to stimulate collateral arteriole development in a therapeutic context. Here, we show that a gelatin-based hydrogel, devoid of growth factors or encapsulated cells, promotes arteriogenesis and attenuates tissue damage in a murine CLI model. The gelatin hydrogel is functionalized with a peptide derived from the extracellular epitope of Type 1 cadherins. Mechanistically, these "GelCad" hydrogels promote arteriogenesis by recruiting smooth muscle cells to vessel structures in both ex vivo and in vivo assays. In a murine femoral artery ligation model of CLI, delivery of in situ crosslinking GelCad hydrogels was sufficient to restore limb perfusion and maintain tissue health for 14 days, whereas mice treated with gelatin hydrogels had extensive necrosis and autoamputated within 7 days. A small cohort of mice receiving the GelCad hydrogels were aged out to 5 months and exhibited no decline in tissue quality, indicating durability of the collateral arteriole networks. Overall, given the simplicity and off-the-shelf format of the GelCad hydrogel platform, we suggest it could have utility for CLI treatment and potentially other indications that would benefit from arteriole development.
Collapse
Affiliation(s)
- Corinne W. Curry
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Sarah M. Sturgeon
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Brian J. O’Grady
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Alexis K. Yates
- Interdisciplinary Materials Science Program, Vanderbilt University, Nashville, TN, USA
| | - Andrew Kjar
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Hayden A. Paige
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Lucas S. Mowery
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Ketaki A. Katdare
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
| | - Riya V. Patel
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Kate Mlouk
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
| | - Madison R. Stiefbold
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Sidney Vafaie-Partin
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Atsuyuki Kawabata
- Department of Orthopedics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Rachel M. McKee
- Department of Orthopedics, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Adrienne Hawkes
- Department of Radiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jiro Kusunose
- Department of Radiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Katherine N. Gibson-Corley
- Department of Pathology, Microbiology and Immunology, Division of Comparative Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jeffrey Schmeckpeper
- Department of Cardiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Charles F. Caskey
- Department of Radiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ethan S. Lippmann
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
- Interdisciplinary Materials Science Program, Vanderbilt University, Nashville, TN, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
7
|
Chang JW, Kim S, Lee EY, Leem CH, Kim SH, Park CS. Cell-cell contacts via N-cadherin induce a regulatory renin secretory phenotype in As4.1 cells. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2022; 26:479-499. [PMID: 36302623 PMCID: PMC9614399 DOI: 10.4196/kjpp.2022.26.6.479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 09/05/2022] [Accepted: 09/19/2022] [Indexed: 11/07/2022]
Abstract
The lack of a clonal renin-secreting cell line has greatly hindered the investigation of the regulatory mechanisms of renin secretion at the cellular, biochemical, and molecular levels. In the present study, we investigated whether it was possible to induce phenotypic switching of the renin-expressing clonal cell line As4.1 from constitutive inactive renin secretion to regulated active renin secretion. When grown to postconfluence for at least two days in media containing fetal bovine serum or insulin-like growth factor-1, the formation of cell-cell contacts via N-cadherin triggered downstream cellular signaling cascades and activated smooth muscle-specific genes, culminating in phenotypic switching to a regulated active renin secretion phenotype, including responding to the key stimuli of active renin secretion. With the use of phenotype-switched As4.1 cells, we provide the first evidence that active renin secretion via exocytosis is regulated by phosphorylation/dephosphorylation of the 20 kDa myosin light chain. The molecular mechanism of phenotypic switching in As4.1 cells described here could serve as a working model for full phenotypic modulation of other secretory cell lines with incomplete phenotypes.
Collapse
Affiliation(s)
- Jai Won Chang
- Department of Physiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea.,Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Soohyun Kim
- Department of Physiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Eun Young Lee
- Department of Internal Medicine, The Catholic University of Korea, Seoul St. Mary's Hospital, Seoul 06591, Korea
| | - Chae Hun Leem
- Department of Physiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Suhn Hee Kim
- Department of Physiology, Jeonbuk National University Medical School, Jeonju 54907, Korea
| | - Chun Sik Park
- Department of Physiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| |
Collapse
|
8
|
Rubina KA, Semina EV, Kalinina NI, Sysoeva VY, Balatskiy AV, Tkachuk VA. Revisiting the multiple roles of T-cadherin in health and disease. Eur J Cell Biol 2021; 100:151183. [PMID: 34798557 DOI: 10.1016/j.ejcb.2021.151183] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 11/08/2021] [Accepted: 11/08/2021] [Indexed: 01/02/2023] Open
Abstract
As a non-canonical member of cadherin superfamily, T-cadherin was initially described as a molecule involved in homophilic recognition in the nervous and vascular systems. The ensuing decades clearly demonstrated that T-cadherin is a remarkably multifunctional molecule. It was validated as a bona fide receptor for both: LDL exerting adverse atherogenic action and adiponectin mediating many protective metabolic and cardiovascular effects. Motivated by the latest progress and accumulated data unmasking important roles of T-cadherin in blood vessel function and tissue regeneration, here we revisit the original function of T-cadherin as a guidance receptor for the growing axons and blood vessels, consider the recent data on T-cadherin-induced exosomes' biogenesis and their role in myocardial regeneration and revascularization. The review expands upon T-cadherin contribution to mesenchymal stem/stromal cell compartment in adipose tissue. We also dwell upon T-cadherin polymorphisms (SNP) and their possible therapeutic applications. Furthermore, we scrutinize the molecular hub of insulin and adiponectin receptors (AdipoR1 and AdipoR2) conveying signals to their downstream targets in quest for defining a putative place of T-cadherin in this molecular circuitry.
Collapse
Affiliation(s)
- K A Rubina
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia.
| | - E V Semina
- Institute of Experimental Cardiology, National Cardiology Research Center of the Ministry of Health of the Russian Federation, 121552 Moscow, Russia
| | - N I Kalinina
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia
| | - V Yu Sysoeva
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia
| | - A V Balatskiy
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia
| | - V A Tkachuk
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia; Institute of Experimental Cardiology, National Cardiology Research Center of the Ministry of Health of the Russian Federation, 121552 Moscow, Russia
| |
Collapse
|
9
|
Malecki C, Hambly BD, Jeremy RW, Robertson EN. The RNA-binding fragile-X mental retardation protein and its role beyond the brain. Biophys Rev 2020; 12:903-916. [PMID: 32654068 DOI: 10.1007/s12551-020-00730-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 07/06/2020] [Indexed: 12/17/2022] Open
Abstract
It is well-established that variations of a CGG repeat expansion in the gene FMR1, which encodes the fragile-X mental retardation protein (FMRP), cause the neurocognitive disorder, fragile-X syndrome (FXS). However, multiple observations suggest a general and complex regulatory role of FMRP in processes outside the brain: (1) FMRP is ubiquitously expressed in the body, suggesting it functions in multiple organ systems; (2) patients with FXS can exhibit a physical phenotype that is consistent with an underlying abnormality in connective tissue; (3) different CGG repeat expansion lengths in FMR1 result in different clinical outcomes due to different pathogenic mechanisms; (4) the function of FMRP as an RNA-binding protein suggests it has a general regulatory role. This review details the complex nature of FMRP and the different CGG repeat expansion lengths and the evidence supporting the essential role of the protein in a variety of biological and pathological processes.
Collapse
Affiliation(s)
- Cassandra Malecki
- Discipline of Pathology and Bosch Institute, The University of Sydney, Level 4 West, Charles Perkins Centre D17, Sydney, NSW, 2006, Australia.
| | - Brett D Hambly
- Discipline of Pathology and Bosch Institute, The University of Sydney, Level 4 West, Charles Perkins Centre D17, Sydney, NSW, 2006, Australia
| | - Richmond W Jeremy
- Discipline of Pathology and Bosch Institute, The University of Sydney, Level 4 West, Charles Perkins Centre D17, Sydney, NSW, 2006, Australia.,Cardiology Department, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Elizabeth N Robertson
- Discipline of Pathology and Bosch Institute, The University of Sydney, Level 4 West, Charles Perkins Centre D17, Sydney, NSW, 2006, Australia.,Cardiology Department, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| |
Collapse
|
10
|
Hong KS, Kim K, Hill MA. Regulation of blood flow in small arteries: mechanosensory events underlying myogenic vasoconstriction. J Exerc Rehabil 2020; 16:207-215. [PMID: 32724777 PMCID: PMC7365734 DOI: 10.12965/jer.2040432.216] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 06/10/2020] [Indexed: 01/10/2023] Open
Abstract
As blood flow is proportional to the fourth power of the vascular radius small changes in the diameter of resistance arteries/arterioles following an increase in intraluminal pressure would be expected to substantially increase blood flow. However, arteriolar myocytes display an intrinsic ability to locally regulate blood flow according to metabolic demands by tuning the diameter of small arteries in response to local changes in he-modynamics. Critical to this, observations were made more than 100 years ago that mechanosensitive small arteries exhibit the "myogenic response" or pressure-induced vasoconstriction or vasodilation in re-sponse to increased or decreased intravascular pressure, respectively. Although cellular mechanisms underlying the myogenic response have now been studied extensively, the precise cellular mechanisms under-lying this intriguing phenomenon still remain uncertain. In particular, the biological machinery that senses changes in intravascular pressure in vascular smooth muscle cells have not been unquestionably identified and remain a significant issue in vascular biology to be fully elucidated. As such, this brief review focuses on putative mechanosensors that have been proposed to contribute to myogenic vasoreactivity. Specific attention is paid to the roles of integrins, G protein-coupled receptors, and cadherins.
Collapse
Affiliation(s)
- Kwang-Seok Hong
- Department of Physical Education, College of Education, Chung-Ang University, Seoul, Korea
| | - Kijeong Kim
- School of Exercise & Sport Science, College of Natural Sciences, University of Ulsan, Ulsan, Korea
| | - Michael A Hill
- Dalton Cardiovascular Research Center, University of Missouri-School of Medicine, Columbia, MO, USA.,Department of Medical Pharmacology and Physiology, University of Missouri-School of Medicine, Columbia, MO, USA
| |
Collapse
|
11
|
Ehyai S, Miyake T, Williams D, Vinayak J, Bayfield MA, McDermott JC. FMRP recruitment of β-catenin to the translation pre-initiation complex represses translation. EMBO Rep 2018; 19:embr.201745536. [PMID: 30361391 DOI: 10.15252/embr.201745536] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 09/12/2018] [Accepted: 09/19/2018] [Indexed: 01/07/2023] Open
Abstract
Canonical Wnt/β-catenin signaling is an essential regulator of various cellular functions throughout development and adulthood. Aberrant Wnt/β-catenin signaling also contributes to various pathologies including cancer, necessitating an understanding of cell context-dependent mechanisms regulating this pathway. Since protein-protein interactions underpin β-catenin function and localization, we sought to identify novel β-catenin interacting partners by affinity purification coupled with tandem mass spectrometry in vascular smooth muscle cells (VSMCs), where β-catenin is involved in both physiological and pathological control of cell proliferation. Here, we report novel components of the VSMC β-catenin interactome. Bioinformatic analysis of the protein networks implies potentially novel functions for β-catenin, particularly in mRNA translation, and we confirm a direct interaction between β-catenin and the fragile X mental retardation protein (FMRP). Biochemical studies reveal a basal recruitment of β-catenin to the messenger ribonucleoprotein and translational pre-initiation complex, fulfilling a translational repressor function. Wnt stimulation antagonizes this function, in part, by sequestering β-catenin away from the pre-initiation complex. In conclusion, we present evidence that β-catenin fulfills a previously unrecognized function in translational repression.
Collapse
Affiliation(s)
- Saviz Ehyai
- Department of Biology, York University, Toronto, ON, Canada.,Muscle Health Research Centre (MHRC), York University, Toronto, ON, Canada.,Centre for Research in Biomolecular Interactions (CRBI), York University, Toronto, ON, Canada
| | - Tetsuaki Miyake
- Department of Biology, York University, Toronto, ON, Canada.,Muscle Health Research Centre (MHRC), York University, Toronto, ON, Canada.,Centre for Research in Biomolecular Interactions (CRBI), York University, Toronto, ON, Canada
| | - Declan Williams
- Department of Chemistry, York University, Toronto, ON, Canada.,Centre for Research in Mass Spectrometry (CRMS), York University, Toronto, ON, Canada
| | - Jyotsna Vinayak
- Department of Biology, York University, Toronto, ON, Canada.,Centre for Research in Biomolecular Interactions (CRBI), York University, Toronto, ON, Canada
| | - Mark A Bayfield
- Department of Biology, York University, Toronto, ON, Canada.,Centre for Research in Biomolecular Interactions (CRBI), York University, Toronto, ON, Canada
| | - John C McDermott
- Department of Biology, York University, Toronto, ON, Canada .,Muscle Health Research Centre (MHRC), York University, Toronto, ON, Canada.,Centre for Research in Biomolecular Interactions (CRBI), York University, Toronto, ON, Canada.,Department of Chemistry, York University, Toronto, ON, Canada.,Centre for Research in Mass Spectrometry (CRMS), York University, Toronto, ON, Canada
| |
Collapse
|
12
|
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: 268] [Impact Index Per Article: 38.3] [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
|
13
|
The potential applications of mushrooms against some facets of atherosclerosis: A review. Food Res Int 2018; 105:517-536. [DOI: 10.1016/j.foodres.2017.11.023] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 11/08/2017] [Accepted: 11/19/2017] [Indexed: 12/16/2022]
|
14
|
Hansmeier N, Buttigieg J, Kumar P, Pelle S, Choi KY, Kopriva D, Chao TC. Identification of Mature Atherosclerotic Plaque Proteome Signatures Using Data-Independent Acquisition Mass Spectrometry. J Proteome Res 2017; 17:164-176. [DOI: 10.1021/acs.jproteome.7b00487] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Nicole Hansmeier
- Department
of Biology/Chemistry, Division of Microbiology, University of Osnabrück, Barbarastrasse 11, 49076 Osnabrück, Germany
| | - Josef Buttigieg
- Department
of Biology, University of Regina, 3737 Wascana Parkway, Regina, Saskatchewan S4S 0A2, Canada
| | - Pankaj Kumar
- Department
of Biology, University of Regina, 3737 Wascana Parkway, Regina, Saskatchewan S4S 0A2, Canada
| | - Shaneen Pelle
- Department
of Biology, University of Regina, 3737 Wascana Parkway, Regina, Saskatchewan S4S 0A2, Canada
| | - Kyoo Yoon Choi
- Department
of Biology, University of Regina, 3737 Wascana Parkway, Regina, Saskatchewan S4S 0A2, Canada
| | - David Kopriva
- Regina Qu’Appelle Health Region and University of Saskatchewan, 1440-14th Avenue, Regina, Saskatchewan S4P 0W5, Canada
| | - Tzu-Chiao Chao
- Department
of Biology, University of Regina, 3737 Wascana Parkway, Regina, Saskatchewan S4S 0A2, Canada
| |
Collapse
|
15
|
Sun Z, Li M, Li Z, Hill MA, Meininger GA. N-Cadherin, a novel and rapidly remodelling site involved in vasoregulation of small cerebral arteries. J Physiol 2017; 595:1987-2000. [PMID: 28008617 DOI: 10.1113/jp272995] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 12/18/2016] [Indexed: 12/29/2022] Open
Abstract
KEY POINTS N-cadherin formed punctate adherens junctions (AJ) along the borders between vascular smooth muscle cells (VSMCs) in the pressurized rat superior cerebellar artery. The formation of N-cadherin AJs in the vessel wall depends on the intraluminal pressure and was responsive to treatment with phenylephrine (PE) (10-5 m) and ACh (10-5 m). N-cadherin-coated beads were able to induce clustering of N-cadherin-enhanced green fluorescent protein (EGFP) on the plasma membrane of isolated VSMCs, whereas treatment with PE (10-5 m) or sodium nitroprusside (10-5 m) induced a significant increase or decrease in the N-cadherin-EGFP clustering, respectively. Application of pulling force (∼1 nN) to the N-cadherin-coated beads via an atomic force microscope induced a localized mechanical response from the VSMCs that opposed the pulling. ABSTRACT N-cadherin is the major cell-cell adhesion molecule in vascular smooth muscle cells (VSMCs). We tested the hypothesis that N-cadherin is part of a novel mechanosensory mechanism in VSMCs and plays an active role in both the arteriolar myogenic response and during changes in vascular tone induced by vasomotor agonists. Intact and pressurized rat superior cerebellar arteries were labelled for confocal immunofluorescence imaging. N-cadherin formed punctate adherens junctions (AJ) along the borders between VSMCs. When the lumen pressure was raised from 50 to 90 mmHg, both the density and the average size of N-cadherin AJs increased significantly. Similarly, arteriolar constriction with phenylephrine (PE) (10-5 m) induced a significant increase of N-cadherin AJ density at 50 mmHg, whereas vasodilatation induced by ACh (10-5 m) was accompanied by a significant decrease in density and size of N-cadherin AJs. An atomic force microscope (AFM) was employed to further examine the mechano-responsive properties of N-cadherin adhesion sites in isolated VSMCs. AFM probes with an attached N-cadherin-coated microbead (5 μm) induced a progressive clustering of N-cadherin-enhanced green fluorescent protein (EGFP) on the VSMC surface. Application of pulling force (∼1 nN) to the N-cadherin-coated-beads with the AFM induced a localized mechanical response from the VSMCs that opposed the pulling. Treatment with PE (10-5 m) or sodium nitroprusside (10-5 m) induced a significant increase or decrease of the N-cadherin-EGFP clustering, respectively. These observations provide compelling evidence that N-cadherin AJs are sensitive to pressure and vasomotor agonists in VSMCs and support a functional role of N-cadherin AJs in vasomotor regulation.
Collapse
Affiliation(s)
- Zhe Sun
- Dalton Cardiovascular Research Center, Columbia, MO, USA.,Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
| | - Min Li
- Dalton Cardiovascular Research Center, Columbia, MO, USA
| | - Zhaohui Li
- Dalton Cardiovascular Research Center, Columbia, MO, USA
| | - Michael A Hill
- Dalton Cardiovascular Research Center, Columbia, MO, USA.,Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
| | - Gerald A Meininger
- Dalton Cardiovascular Research Center, Columbia, MO, USA.,Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
| |
Collapse
|
16
|
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
|
17
|
Balint B, Yin H, Chakrabarti S, Chu MW, Sims SM, Pickering JG. Collectivization of Vascular Smooth Muscle Cells via TGF-β–Cadherin-11–Dependent Adhesive Switching. Arterioscler Thromb Vasc Biol 2015; 35:1254-64. [DOI: 10.1161/atvbaha.115.305310] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 02/27/2015] [Indexed: 01/26/2023]
Abstract
Objective—
Smooth muscle cells (SMCs) in healthy arteries are arranged as a collective. However, in diseased arteries, SMCs commonly exist as individual cells, unconnected to each other. The purpose of this study was to elucidate the events that enable individualized SMCs to enter into a stable and interacting cell collective.
Approach and Results—
Human SMCs stimulated to undergo programmed collectivization were tracked by time-lapse microscopy. We uncovered a switch in the behavior of contacting SMCs from semiautonomous motility to cell–cell adherence. Central to the cell-adherent phenotype was the formation of uniquely elongated adherens junctions, up to 60 μm in length, which appeared to strap adjacent SMCs to each other. Remarkably, these junctions contained both N-cadherin and cadherin-11. Ground-state depletion super-resolution microscopy revealed that these hybrid assemblies were comprised of 2 parallel nanotracks of each cadherin, separated by 50 nm. Blocking either N-cadherin or cadherin-11 inhibited collectivization. Cell–cell adhesion and adherens junction elongation were associated with reduced transforming growth factor-β signaling, and exogenous transforming growth factor-β1 suppressed junction elongation via the noncanonical p38 pathway. Imaging of fura-2–loaded SMCs revealed that SMC assemblies displayed coordinated calcium oscillations and cell–cell transmission of calcium waves which, together with increased connexin 43–containing junctions, depended on cadherin-11 and N-cadherin function.
Conclusions—
SMCs can self-organize, structurally and functionally, via transforming growth factor-β–p38–dependent adhesive switching and a novel adherens junction architecture comprised of hybrid nanotracks of cadherin-11 and N-cadherin. The findings define a mechanism for the assembly of SMCs into networks, a process that may be relevant to the stability and function of blood vessels.
Collapse
Affiliation(s)
- Brittany Balint
- From the Robarts Research Institute (B.B., H.Y., J.G.P.), Departments of Medicine (Cardiology) (J.G.P.), Biochemistry (J.G.P.), Medical Biophysics (B.B., J.G.P.), Pathology and Laboratory Medicine (S.C.), Surgery (M.W.A.C.), and Physiology and Pharmacology (S.M.S.), University of Western Ontario, London Health Sciences Centre (S.C., J.G.P.), London, Ontario, Canada
| | - Hao Yin
- From the Robarts Research Institute (B.B., H.Y., J.G.P.), Departments of Medicine (Cardiology) (J.G.P.), Biochemistry (J.G.P.), Medical Biophysics (B.B., J.G.P.), Pathology and Laboratory Medicine (S.C.), Surgery (M.W.A.C.), and Physiology and Pharmacology (S.M.S.), University of Western Ontario, London Health Sciences Centre (S.C., J.G.P.), London, Ontario, Canada
| | - Subrata Chakrabarti
- From the Robarts Research Institute (B.B., H.Y., J.G.P.), Departments of Medicine (Cardiology) (J.G.P.), Biochemistry (J.G.P.), Medical Biophysics (B.B., J.G.P.), Pathology and Laboratory Medicine (S.C.), Surgery (M.W.A.C.), and Physiology and Pharmacology (S.M.S.), University of Western Ontario, London Health Sciences Centre (S.C., J.G.P.), London, Ontario, Canada
| | - Michael W.A. Chu
- From the Robarts Research Institute (B.B., H.Y., J.G.P.), Departments of Medicine (Cardiology) (J.G.P.), Biochemistry (J.G.P.), Medical Biophysics (B.B., J.G.P.), Pathology and Laboratory Medicine (S.C.), Surgery (M.W.A.C.), and Physiology and Pharmacology (S.M.S.), University of Western Ontario, London Health Sciences Centre (S.C., J.G.P.), London, Ontario, Canada
| | - Stephen M. Sims
- From the Robarts Research Institute (B.B., H.Y., J.G.P.), Departments of Medicine (Cardiology) (J.G.P.), Biochemistry (J.G.P.), Medical Biophysics (B.B., J.G.P.), Pathology and Laboratory Medicine (S.C.), Surgery (M.W.A.C.), and Physiology and Pharmacology (S.M.S.), University of Western Ontario, London Health Sciences Centre (S.C., J.G.P.), London, Ontario, Canada
| | - J. Geoffrey Pickering
- From the Robarts Research Institute (B.B., H.Y., J.G.P.), Departments of Medicine (Cardiology) (J.G.P.), Biochemistry (J.G.P.), Medical Biophysics (B.B., J.G.P.), Pathology and Laboratory Medicine (S.C.), Surgery (M.W.A.C.), and Physiology and Pharmacology (S.M.S.), University of Western Ontario, London Health Sciences Centre (S.C., J.G.P.), London, Ontario, Canada
| |
Collapse
|
18
|
Gancheva K, Postadjian A, Brazma D, Grace C, Chanalaris A, Nacheva E, Apostolova M. Copy Number Variants: Distribution in Patients with Coronary Atherosclerosis. BIOTECHNOL BIOTEC EQ 2014. [DOI: 10.1080/13102818.2009.10817620] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
|
19
|
Sun Z, Parrish AR, Hill MA, Meininger GA. N-cadherin, A Vascular Smooth Muscle Cell-Cell Adhesion Molecule: Function and Signaling for Vasomotor Control. Microcirculation 2014; 21:208-18. [DOI: 10.1111/micc.12123] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 02/05/2014] [Indexed: 12/22/2022]
Affiliation(s)
- Zhe Sun
- Dalton Cardiovascular Research Center; University of Missouri; Columbia Missouri USA
| | - Alan R. Parrish
- Department of Medical Pharmacology and Physiology; University of Missouri; Columbia Missouri USA
| | - Michael A. Hill
- Dalton Cardiovascular Research Center; University of Missouri; Columbia Missouri USA
- Department of Medical Pharmacology and Physiology; University of Missouri; Columbia Missouri USA
| | - Gerald A. Meininger
- Dalton Cardiovascular Research Center; University of Missouri; Columbia Missouri USA
- Department of Medical Pharmacology and Physiology; University of Missouri; Columbia Missouri USA
| |
Collapse
|
20
|
Cai X. Regulation of smooth muscle cells in development and vascular disease: current therapeutic strategies. Expert Rev Cardiovasc Ther 2014; 4:789-800. [PMID: 17173496 DOI: 10.1586/14779072.4.6.789] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Vascular smooth muscle cells (SMCs) exhibit extensive phenotypic diversity and rapid growth during embryonic development, but maintain a quiescent, differentiated state in adult. The pathogenesis of vascular proliferative diseases involves the proliferation and migration of medial vascular SMCs into the vessel intima, possibly reinstating their embryonic gene expression programs. Multiple mitogenic stimuli induce vascular SMC proliferation through cell cycle progression. Therapeutic strategies targeting cell cycle progression and mitogenic stimuli have been developed and evaluated in animal models of atherosclerosis and vascular injury, and several clinical studies. Recent discoveries on the recruitment of vascular progenitor cells to the sites of vascular injury suggest new therapeutic potentials of progenitor cell-based therapies to accelerate re-endothelialization and prevent engraftment of SMC-lineage progenitor cells. Owing to the complex and multifactorial nature of SMC regulation, combinatorial antiproliferative approaches are likely to be used in the future in order to achieve maximal efficacy and reduce toxicity.
Collapse
MESH Headings
- Animals
- Cell Differentiation
- Cellular Senescence
- Clinical Trials as Topic
- Disease Progression
- Drug Delivery Systems
- Gene Expression
- Genetic Therapy
- Humans
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/embryology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Phenotype
- Stents
- Vascular Diseases/drug therapy
- Vascular Diseases/genetics
- Vascular Diseases/metabolism
- Vascular Diseases/pathology
Collapse
Affiliation(s)
- Xinjiang Cai
- Duke University Medical Center, Departments of Medicine (Cardiology) & Cell Biology, Durham, North Carolina 27710, USA.
| |
Collapse
|
21
|
Karagiannis GS, Weile J, Bader GD, Minta J. Integrative pathway dissection of molecular mechanisms of moxLDL-induced vascular smooth muscle phenotype transformation. BMC Cardiovasc Disord 2013; 13:4. [PMID: 23324130 PMCID: PMC3556327 DOI: 10.1186/1471-2261-13-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Accepted: 12/29/2012] [Indexed: 01/08/2023] Open
Abstract
Background Atherosclerosis (AT) is a chronic inflammatory disease characterized by the accumulation of inflammatory cells, lipoproteins and fibrous tissue in the walls of arteries. AT is the primary cause of heart attacks and stroke and is the leading cause of death in Western countries. To date, the pathogenesis of AT is not well-defined. Studies have shown that the dedifferentiation of contractile and quiescent vascular smooth muscle cells (SMC) to the proliferative, migratory and synthetic phenotype in the intima is pivotal for the onset and progression of AT. To further delineate the mechanisms underlying the pathogenesis of AT, we analyzed the early molecular pathways and networks involved in the SMC phenotype transformation. Methods Quiescent human coronary artery SMCs were treated with minimally-oxidized LDL (moxLDL), for 3 hours and 21 hours, respectively. Transcriptomic data was generated for both time-points using microarrays and was subjected to pathway analysis using Gene Set Enrichment Analysis, GeneMANIA and Ingenuity software tools. Gene expression heat maps and pathways enriched in differentially expressed genes were compared to identify functional biological themes to elucidate early and late molecular mechanisms of moxLDL-induced SMC dedifferentiation. Results Differentially expressed genes were found to be enriched in cholesterol biosynthesis, inflammatory cytokines, chemokines, growth factors, cell cycle control and myogenic contraction themes. These pathways are consistent with inflammatory responses, cell proliferation, migration and ECM production, which are characteristic of SMC dedifferentiation. Furthermore, up-regulation of cholesterol synthesis and dysregulation of cholesterol metabolism was observed in moxLDL-induced SMC. These observations are consistent with the accumulation of cholesterol and oxidized cholesterol esters, which induce proinflammatory reactions during atherogenesis. Our data implicate for the first time IL12, IFN-α, HGF, CSF3, and VEGF signaling in SMC phenotype transformation. GPCR signaling, HBP1 (repressor of cyclin D1 and CDKN1B), and ID2 and ZEB1 transcriptional regulators were also found to have important roles in SMC dedifferentiation. Several microRNAs were observed to regulate the SMC phenotype transformation via an interaction with IFN-γ pathway. Also, several “nexus” genes in complex networks, including components of the multi-subunit enzyme complex involved in the terminal stages of cholesterol synthesis, microRNAs (miR-203, miR-511, miR-590-3p, miR-346*/miR- 1207-5p/miR-4763-3p), GPCR proteins (GPR1, GPR64, GPRC5A, GPR171, GPR176, GPR32, GPR25, GPR124) and signal transduction pathways, were found to be regulated. Conclusions The systems biology analysis of the in vitro model of moxLDL-induced VSMC phenotype transformation was associated with the regulation of several genes not previously implicated in SMC phenotype transformation. The identification of these potential candidate genes enable hypothesis generation and in vivo functional experimentation (such as gain and loss-of-function studies) to establish causality with the process of SMC phenotype transformation and atherogenesis.
Collapse
Affiliation(s)
- George S Karagiannis
- Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, and Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON, M5S 1A8, Canada
| | | | | | | |
Collapse
|
22
|
Yousif LF, Di Russo J, Sorokin L. Laminin isoforms in endothelial and perivascular basement membranes. Cell Adh Migr 2012; 7:101-10. [PMID: 23263631 DOI: 10.4161/cam.22680] [Citation(s) in RCA: 181] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Laminins, one of the major functional components of basement membranes, are found underlying endothelium, and encasing pericytes and smooth muscle cells in the vessel wall. Depending on the type of blood vessel (capillary, venule, postcapillary venule, vein or artery) and their maturation state, both the endothelial and mural cell phenotype vary, with associated changes in laminin isoform expression. Laminins containing the α4 and α5 chains are the major isoforms found in the vessel wall, with the added contribution of laminin α2 in larger vessels. We here summarize current data on the precise localization of these laminin isoforms and their receptors in the different layers of the vessel wall, and their potential contribution to vascular homeostasis.
Collapse
Affiliation(s)
- Lema F Yousif
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany
| | | | | |
Collapse
|
23
|
Marinou K, Christodoulides C, Antoniades C, Koutsilieris M. Wnt signaling in cardiovascular physiology. Trends Endocrinol Metab 2012; 23:628-36. [PMID: 22902904 DOI: 10.1016/j.tem.2012.06.001] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Revised: 06/01/2012] [Accepted: 06/04/2012] [Indexed: 01/17/2023]
Abstract
Wnt signaling pathways play a key role in cardiac development, angiogenesis, and cardiac hypertrophy; emerging evidence suggests that they are also involved in the pathophysiology of atherosclerosis. Specifically, an important role for Wnts has been described in the regulation of endothelial inflammation, vascular calcification, and mesenchymal stem cell differentiation. Wnt signaling also induces monocyte adhesion to endothelial cells and is crucial for the regulation of vascular smooth-muscle cell (VSMC) behavior. We discuss how the Wnt pathways are implicated in vascular biology and outline the role of Wnt signaling in atherosclerosis. Dissecting Wnt pathways involved in atherogenesis and cardiovascular disease may provide crucial insights into novel mechanisms with therapeutic potential for atherosclerosis.
Collapse
Affiliation(s)
- K Marinou
- Department of Physiology, Athens University Medical School, Athens, Greece.
| | | | | | | |
Collapse
|
24
|
Overlapping and divergent signaling pathways of N-cadherin and VE-cadherin in endothelial cells. Blood 2012; 119:2159-70. [DOI: 10.1182/blood-2011-09-381012] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Abstract
Endothelial cells (ECs) express 2 members of the cadherin family, VE and N-cadherin. Although VE-cadherin induces EC homotypic adhesion, N-cadherin function in ECs remains largely unknown. EC-specific inactivation of either VE or N-cadherin leads to early fetal lethality suggesting that these cadherins play a nonredundant role in vascular development. We report here that VE-cadherin negatively controls junctional localization and expression of N-cadherin by limiting p120-catenin availability and reducing β-catenin transcriptional activity. Using EC lines expressing either VE or N-cadherin we found that both cadherins inhibit cell proliferation and apoptosis. Both trigger the phosphatidylinositol-3-OH-kinase (PI3K)–AKT-Forkhead-box protein-O1 (FoxO1) pathway and reduce β-catenin transcriptional activity. The extent of signaling correlates with the total level of cadherins regardless of the type of cadherin expressed. In contrast, basal and fibroblast growth factor (FGF)–induced cell motility is promoted by N-cadherin and strongly inhibited by VE-cadherin. This opposite effect is partly because of the ability of VE-cadherin to associate with FGF receptor and the density-enhanced phosphatase-1 (Dep-1) which, in turn, inhibits receptor signaling. We conclude that VE and N-cadherin have both additive and divergent effects on ECs. Differences in signaling are due, in part, to cadherin association with growth factor receptors and modulation of their downstream signaling.
Collapse
|
25
|
Redmond EM, Cahill PA. The NOX-ROS connection: targeting Nox1 control of N-cadherin shedding in vascular smooth muscle cells. Cardiovasc Res 2012; 93:386-7. [PMID: 22266749 DOI: 10.1093/cvr/cvs020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
|
26
|
Jagadeesha DK, Takapoo M, Banfi B, Bhalla RC, Miller FJ. Nox1 transactivation of epidermal growth factor receptor promotes N-cadherin shedding and smooth muscle cell migration. Cardiovasc Res 2011; 93:406-13. [PMID: 22102727 DOI: 10.1093/cvr/cvr308] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
AIMS In atherosclerosis and restenosis, vascular smooth muscle cells (SMCs) migrate into the subendothelial space and proliferate, contributing to neointimal formation. The goal of this study was to define the signalling pathway by which Nox1 NAPDH oxidase mediates SMC migration. METHODS AND RESULTS SMCs were cultured from thoracic aorta from Nox1(-/y) (Nox1 knockout, KO) and wild-type (WT) mice. In response to thrombin, WT but not Nox1 KO SMCs generated increased levels of reactive oxygen species (ROS). Deficiency of Nox1 prevented thrombin-induced phosphorylation of Src and the subsequent transactivation of the epidermal growth factor receptor (EGFR) at multiple tyrosine residues. Next, activation of extracellular signal-regulated kinase 1/2 (ERK1/2) and matrix metalloproteinase-9 (MMP-9) by thrombin was inhibited by the EGFR inhibitor AG1478 and in Nox1 KO SMCs. Thrombin-induced shedding of N-cadherin from the plasma membrane was dependent on the presence of Nox1 and was blocked by AG1478 and an inhibitor of metalloproteinases. Migration of SMCs to thrombin was impaired in the Nox1 KO SMCs and was restored by expression of Nox1. Finally, treatment of WT SMCs with AG1478 abrogated Nox1-dependent SMC migration. CONCLUSIONS The Nox1 NADPH oxidase signals through EGFR to activate MMP-9 and promote the shedding of N-cadherin, thereby contributing to SMC migration.
Collapse
|
27
|
Sklepkiewicz P, Schermuly RT, Tian X, Ghofrani HA, Weissmann N, Sedding D, Kashour T, Seeger W, Grimminger F, Pullamsetti SS. Glycogen synthase kinase 3beta contributes to proliferation of arterial smooth muscle cells in pulmonary hypertension. PLoS One 2011; 6:e18883. [PMID: 21533110 PMCID: PMC3078925 DOI: 10.1371/journal.pone.0018883] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2010] [Accepted: 03/24/2011] [Indexed: 01/17/2023] Open
Abstract
Rationale Pulmonary arterial hypertension (PAH) is a rare progressive pulmonary vascular disorder associated with vascular remodeling and right heart failure. Vascular remodeling involves numerous signaling cascades governing pulmonary arterial smooth muscle cell (PASMC) proliferation, migration and differentiation. Glycogen synthase kinase 3beta (GSK3ß) is a serine/threonine kinase and can act as a downstream regulatory switch for numerous signaling pathways. Hence, we hypothesized that GSK3ß plays a crucial role in pulmonary vascular remodeling. Methods All experiments were done with lung tissue or isolated PASMCs in a well-established monocrotaline (MCT)-induced PAH rat model. The mRNA expression of Wnt ligands (Wnt1, Wnt3a, Wnt5a), upstream Wnt signaling regulator genes (Frizzled Receptors 1, 2 and secreted Frizzled related protein sFRP-1) and canonical Wnt intracellular effectors (GSK3ß, Axin1) were assessed by real-time polymerase chain reaction and protein levels of GSK3ß, phospho-GSK3ß (ser 9) by western blotting and localization by immunohistochemistry. The role of GSK3ß in PASMCs proliferation was assessed by overexpression of wild-type GSK3ß (WT) and constitutively active GSK3ß S9A by [3H]-thymidine incorporation assay. Results Increased levels of total and phosphorylated GSK3ß (inhibitory phosphorylation) were observed in lungs and PASMCs isolated from MCT-induced PAH rats compared to controls. Further, stimulation of MCT-PASMCs with growth factors induced GSK3ß inactivation. Most importantly, treatment with the PDGFR inhibitor, Imatinib, attenuated PDGF-BB and FCS induced GSK3ß phosphorylation. Increased expression of GSK3ß observed in lungs and PASMC isolated from MCT-induced PAH rats was confirmed to be clinically relevant as the same observation was identified in human iPAH lung explants. Overexpression of GSK3ß significantly increased MCT-PASMCs proliferation by regulating ERK phosphorylation. Constitutive activation of GSK3ß (GSK3ß S9A, 9th serine replaced to alanine) inhibited MCT-PASMCs proliferation by decreasing ERK phosphorylation. Conclusion This study supports a central role for GSK3ß in vascular remodeling processes and suggests a novel therapeutic opportunity for the treatment of PAH.
Collapse
MESH Headings
- Animals
- Arteries/enzymology
- Arteries/pathology
- Base Sequence
- Blotting, Western
- Cell Proliferation
- Cells, Cultured
- Cloning, Molecular
- DNA Primers
- Disease Models, Animal
- Glycogen Synthase Kinase 3/genetics
- Glycogen Synthase Kinase 3/metabolism
- Glycogen Synthase Kinase 3 beta
- Humans
- Hypertension, Pulmonary/enzymology
- Hypertension, Pulmonary/pathology
- Immunohistochemistry
- Male
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/pathology
- Mutagenesis
- Phosphorylation
- Polymerase Chain Reaction
- Rats
- Wnt Proteins/metabolism
Collapse
Affiliation(s)
| | - Ralph Theo Schermuly
- Medical Clinic II/V, University Hospital, Giessen, Germany
- Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Xia Tian
- Medical Clinic II/V, University Hospital, Giessen, Germany
| | | | | | | | | | - Werner Seeger
- Medical Clinic II/V, University Hospital, Giessen, Germany
- Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | | | - Soni Savai Pullamsetti
- Medical Clinic II/V, University Hospital, Giessen, Germany
- Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
- * E-mail:
| |
Collapse
|
28
|
Nuessle JM, Giehl K, Herzog R, Stracke S, Menke A. TGFβ1 suppresses vascular smooth muscle cell motility by expression of N-cadherin. Biol Chem 2011; 392:461-74. [PMID: 21375457 DOI: 10.1515/bc.2011.053] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neointimal formation in atheromatous blood vessels is associated with both growth factor-induced differentiation of smooth muscle cells and endothelial-to-mesenchymal transition. Transforming growth factor beta (TGFβ)-signaling is well known to play a critical role in the regulation of vessel remodeling as well as in atherosclerosis and restenosis. Here, we investigated the role of TGFβ1 and N-cadherin on the differentiation and migration of human vascular smooth muscle cells (VSMC). TGFβ1-treatment of cultured VSMC reduced their migratory activity as determined in cell migration assays. This reduced migration correlated with increased concentration of N-cadherin on mRNA and protein level. The TGFβ1-induced increase of N-cadherin was sensitive against pharmacological inhibition of the ALK5 TGFβ receptor and was accompanied by TGFβ1-induced expression of the transcription factor snail1. Activation of N-cadherin by using a HAV-containing peptide of N-cadherin also decreased the migration of VSMC. N-cadherin-mediated suppression of VSMC migration was associated with an increased activity of RhoA, which is activated by binding of the HAV peptide to N-cadherin. Our results demonstrate that TGFβ1 induces the differentiation of primary VSMC cells by Smad2/3-dependent up-regulation of the transcription factor snail1 and subsequently of N-cadherin, leading to inhibition of VSMC migration by RhoA-dependent modulation of the actin cytoskeleton.
Collapse
Affiliation(s)
- Johannes M Nuessle
- Internal Medicine I, Nephrology, University of Ulm, Albert-Einstein-Allee, Germany
| | | | | | | | | |
Collapse
|
29
|
Jackson TY, Sun Z, Martinez-Lemus LA, Hill MA, Meininger GA. N-cadherin and integrin blockade inhibit arteriolar myogenic reactivity but not pressure-induced increases in intracellular Ca. Front Physiol 2010; 1:165. [PMID: 21423400 PMCID: PMC3059933 DOI: 10.3389/fphys.2010.00165] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2010] [Accepted: 12/09/2010] [Indexed: 11/13/2022] Open
Abstract
The vascular myogenic response is characterized by arterial constriction in response to an increase in intraluminal pressure and dilatation to a decrease in pressure. This mechanism is important for the regulation of blood flow, capillary pressure, and arterial pressure. The identity of the mechanosensory mechanism(s) for this response is incompletely understood but has been shown to include the integrins as cell-extracellular matrix receptors. The possibility that a cell-cell adhesion receptor is involved has not been studied. Thus, we tested the hypothesis that N-cadherin, a cell-cell adhesion molecule in vascular smooth muscle cells (VSMCs), was important for myogenic responsiveness. The purpose of this study was to investigate: (1) whether cadherin inhibition blocks myogenic responses to increases in intraluminal pressure and (2) the effect of the cadherin or integrin blockade on pressure-induced changes in [Ca(2+)]i. Cadherin blockade was tested in isolated rat cremaster arterioles on myogenic responses to acute pressure steps from 60 to 100 mmHg and changes in VSMC Ca(2+) were measured using fura-2. In the presence of a synthetic cadherin inhibitory peptide or a function-blocking antibody, myogenic responses were inhibited. In contrast, during N-cadherin blockade, pressure-induced changes in [Ca(2+)]i were not altered. Similarly, vessels treated with function-blocking β1- or β3-integrin antibodies maintained pressure-induced [Ca(2+)]i responses despite inhibition of myogenic constriction. Collectively, these data suggest that both cadherins and integrins play a fundamental role in mediating myogenic constriction but argue against their direct involvement in mediating pressure-induced [Ca(2+)]i increases.
Collapse
Affiliation(s)
- Teresa Y Jackson
- Department of Medical Pharmacology and Physiology, Dalton Cardiovascular Research Center, University of Missouri Columbia, MO, USA
| | | | | | | | | |
Collapse
|
30
|
Han F, Yu H, Tian C, Chen HE, Benedict-Alderfer C, Zheng Y, Wang Q, Han X, Zheng QY. A new mouse mutant of the Cdh23 gene with early-onset hearing loss facilitates evaluation of otoprotection drugs. THE PHARMACOGENOMICS JOURNAL 2010; 12:30-44. [PMID: 20644563 PMCID: PMC3000876 DOI: 10.1038/tpj.2010.60] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
We report a novel mutation (erlong, erl) of the cadherin 23 (Cdh23) gene in a mouse model for DFNB12 characterized by progressive hearing loss beginning from postnatal day 27 (P27). Genetic and sequencing analysis revealed a 208 T >C transition causing an amino-acid substitution (70S-P). Caspase expression was upregulated in mutant inner ears. Hearing was preserved (up to 35-dB improvement) in pan-caspase inhibitor Z-VAD-FMK-treated mutants compared with untreated mutants (P<0.05). Outer hair cell (OHC) loss in the cochleae of Z-VAD-FMK-treated mutants was significantly reduced compared with those of untreated mice. Thus, the erl mutation can lead to hearing loss through apoptosis. This is the first genetic mouse model of hearing loss shown to respond to otoprotective drug therapy. The short interval from initial hearing loss to deafness (P27-P90) makes this model ideal for screening and validating otoprotective drugs.
Collapse
Affiliation(s)
- F Han
- Department of Otolaryngology-HNS, Case Western Reserve University, Cleveland, OH 44106, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Gosens R, Baarsma HA, Heijink IH, Oenema TA, Halayko AJ, Meurs H, Schmidt M. De novo synthesis of {beta}-catenin via H-Ras and MEK regulates airway smooth muscle growth. FASEB J 2009; 24:757-68. [PMID: 19906679 DOI: 10.1096/fj.09-136325] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
beta-Catenin is a component of adherens junctions that also acts as a transcriptional coactivator when expressed in the nucleus. Growth factors are believed to regulate the nuclear expression of beta-catenin via inactivation of glycogen synthase kinase 3 (GSK-3) by phosphorylation, resulting in increased beta-catenin protein stability. Here, we report on a novel pathway that regulates the expression and nuclear presence of beta-catenin. In proliferating human airway smooth muscle cells, we observed increased expression of beta-catenin, which was required for proliferation. Interestingly, increased beta-catenin expression was accompanied by an increase in beta-catenin mRNA and was independent of beta-catenin liberation from the plasma membrane, suggesting a role for de novo synthesis. This was confirmed using actinomycin D and cycloheximide, which abrogated the induction and nuclear localization of beta-catenin protein. GSK-3 inhibition using SB216763 failed to regulate beta-catenin mRNA. However, expression of dominant negative H-Ras or pharmacological inhibition of MEK reduced serum and TGF-beta-induced beta-catenin mRNA and protein. Collectively, these data indicate that beta-catenin is an important signaling intermediate in airway smooth muscle growth and that its cellular accumulation and nuclear localization require de novo protein synthesis effected, in part, via H-Ras and MEK.-Gosens, R., Baarsma, H. A., Heijink, I. H., Oenema, T. A., Halayko, A. J., Meurs, H., Schmidt, M. De novo synthesis of beta-catenin via H-Ras and MEK regulates airway smooth muscle growth.
Collapse
Affiliation(s)
- Reinoud Gosens
- Department of Molecular Pharmacology, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
32
|
Cell-cell bond modulates vascular smooth muscle cell responsiveness to Angiotensin II. Biochem Biophys Res Commun 2009; 388:523-8. [PMID: 19665992 DOI: 10.1016/j.bbrc.2009.08.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2009] [Accepted: 08/05/2009] [Indexed: 11/21/2022]
Abstract
Cell attachment is provided by cell-matrix and cell-cell bonds, and acts as a regulator of vascular smooth muscle cell (VSMC) survival, activity and homeostasis, as well as of VSMCs response to pathogenic stimuli. In this work we elicited an exclusive cell-cell contact by culturing A7r5 VSMCs on agarose-coated wells to form floating cell clusters, and we demonstrated that a steady state with a reduced response to the vasoactive peptide Angiotensin II (ATII) was induced. We found that clustered VSMCs showed subcortical stabilization of beta-catenin and Caveolin 1 (Cav1), unlike adherent confluent counterparts. We demonstrated that beta-catenin and Cav1 stabilization at the membrane level hampers the molecular cross-talk induced by ATII-activated AT1 receptor (AT1R), thereby impeding the phosphorylation of Cav1 and IGF1R, the NADPH oxidase activity, and counteracting ATII-dependent hypertrophy. Thus, elective cell-cell bond might modulate the proatherogenic activity of ATII, reducing the adverse vascular remodelling associated with AT1R activation.
Collapse
|
33
|
Stintzing S, Ocker M, Hartner A, Amann K, Barbera L, Neureiter D. Differentiation patterning of vascular smooth muscle cells (VSMC) in atherosclerosis. Virchows Arch 2009; 455:171-185. [PMID: 19557430 DOI: 10.1007/s00428-009-0800-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2009] [Revised: 05/11/2009] [Accepted: 06/06/2009] [Indexed: 01/17/2023]
Abstract
To investigate the involvement of transdifferentiation and dedifferentiation phenomena inside atherosclerotic plaques, we analyzed the differentiation status of vascular smooth muscle cells (VSMC) in vitro and in vivo. Forty normal autoptic and 20 atherosclerotic carotid endarterectomy specimens as well as 20 specimens of infrarenal and suprarenal aortae were analyzed for the expression of cytokeratins 7 and 18 and beta-catenin as markers (epithelial transdifferentiation) as well as CD31 and CD34 (embryonic dedifferentiation) by conventional and double fluorescence immunohistochemistry and reverse transcription polymerase chain reaction. Looking at these markers, additional cell culture experiments with human aortic (HA)-VSMC were done under stimulation with IL-1beta, IL-6, and TNF-alpha. Cytokeratins and beta-catenin were expressed significantly higher in atherosclerotic than in normal carotids primarily localized in VSMC of the shoulder/cap region of atherosclerotic lesions. Additionally, heterogeneous cellular coexpression of CD31 and/or CD34 was observed in subregions of progressive atherosclerotic lesions by VSMC. The expression of those differentiation markers by stimulated HA-VSMC showed a time and cytokine dependency in vitro. Our findings show that (1) VSMC of progressive atheromas have the ability of differentiation, (2) that transdifferentiation and dedifferentiation phenomena are topographically diverse localized in the subregions of advanced atherosclerotic lesions, and (3) are influenced by inflammatory cytokines like IL-1beta, IL-6, and TNF-alpha.
Collapse
Affiliation(s)
- Sebastian Stintzing
- Department of Medicine III, University Hospital Grosshadern, LMU Munich, Marchioninistrasse 15, 81377 Munich, Germany.
| | | | | | | | | | | |
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
|
Chan-Park MB, Shen JY, Cao Y, Xiong Y, Liu Y, Rayatpisheh S, Kang GCW, Greisler HP. Biomimetic control of vascular smooth muscle cell morphology and phenotype for functional tissue-engineered small-diameter blood vessels. J Biomed Mater Res A 2009; 88:1104-21. [PMID: 19097157 DOI: 10.1002/jbm.a.32318] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Small-diameter blood vessel substitutes are urgently needed for patients requiring replacements of their coronary and below-the-knee vessels and for better arteriovenous dialysis shunts. Circulatory diseases, especially those arising from atherosclerosis, are the predominant cause of mortality and morbidity in the developed world. Current therapies include the use of autologous vessels or synthetic materials as vessel replacements. The limited availability of healthy vessels for use as bypass grafts and the failure of purely synthetic materials in small-diameter sites necessitate the development of a biological substitute. Tissue engineering is such an approach and has achieved promising results, but reconstruction of a functional vascular tunica media, with circumferentially oriented contractile smooth muscle cells (SMCs) and extracellular matrix, appropriate mechanical properties, and vasoactivity has yet to be demonstrated. This review focuses on strategies to effect the switch of SMC phenotype from synthetic to contractile, which is regarded as crucial for the engineering of a functional vascular media. The synthetic SMC phenotype is desired initially for cell proliferation and tissue remodeling, but the contractile phenotype is then necessary for sufficient vasoactivity and inhibition of neointima formation. The factors governing the switch to a more contractile phenotype with in vitro culture are reviewed.
Collapse
Affiliation(s)
- Mary B Chan-Park
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 50 Nanyang Ave, Singapore 639798, Singapore.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Bedel A, Nègre-Salvayre A, Heeneman S, Grazide MH, Thiers JC, Salvayre R, Maupas-Schwalm F. E-Cadherin/β-Catenin/T-Cell Factor Pathway Is Involved in Smooth Muscle Cell Proliferation Elicited by Oxidized Low-Density Lipoprotein. Circ Res 2008; 103:694-701. [DOI: 10.1161/circresaha.107.166405] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The E-cadherin/β-catenin/T-cell factor (Tcf) signaling pathway plays a crucial role in embryogenesis and carcinogenesis and has recently emerged in atherosclerosis. The aim of this work was to investigate whether this signaling pathway is involved in smooth muscle cell proliferation induced by oxidized low-density lipoprotein (LDL). In human aortic smooth muscle cells, mitogenic concentration of mildly oxidized LDL induced the activation of β-catenin, as assessed by the dissociation of the β-catenin/cadherin complex, and the concomitant rise of active β-catenin in the cytosol. The oxidized LDL–induced rise of active β-catenin required metalloproteinase activation, as well as epidermal growth factor receptor and Src signaling, as assessed by the use of pharmacological inhibitors and cells overexpressing a SrcK-inactive form. The concomitant phosphatidylinositol 3-kinase/Akt activation and glycogen synthase kinase 3-β phosphorylation induced the inhibition of the proteasomal degradation of β-catenin. Then active β-catenin associated with Tcf4 and translocated into the nucleus. This enhanced the expression of the cell cycle activator cyclin D1. This crucial role of β-catenin in the mitogenic effect of oxidized LDL was confirmed by silencing β-catenin by specific small interfering RNA that blocked DNA synthesis. Immunohistochemistry staining of stable and disrupted plaques from carotid endarterectomy sections showed a correlation between active β-catenin and Ki67, a proliferation marker, and a more intense staining in the smooth muscle cell layer surrounding the lipid core of disrupted plaques. In conclusion, the β-catenin pathway is required for the mitogenic effect of oxidized LDL on human aortic smooth muscle cells. This study highlights the putative important role of the E-cadherin/β-catenin/Tcf signaling pathway in atherosclerosis.
Collapse
Affiliation(s)
- Aurélie Bedel
- From the Institut National de la Santé et de la Recherche Médicale U858 (A.B., A.N.-S., M.-H.G., J.-C.T., R.S., F.M.-S.), Toulouse, France; Faculty of Medicine-Rangueil (A.B., A.N.-S., M.-H.G., J.-C.T., R.S., F.M.-S.), University Paul-Sabatier Toulouse III, Toulouse, France; and Department of Pathology (S.H.), Cardiovascular Research Institute Maastricht, University of Maastricht, The Netherlands
| | - Anne Nègre-Salvayre
- From the Institut National de la Santé et de la Recherche Médicale U858 (A.B., A.N.-S., M.-H.G., J.-C.T., R.S., F.M.-S.), Toulouse, France; Faculty of Medicine-Rangueil (A.B., A.N.-S., M.-H.G., J.-C.T., R.S., F.M.-S.), University Paul-Sabatier Toulouse III, Toulouse, France; and Department of Pathology (S.H.), Cardiovascular Research Institute Maastricht, University of Maastricht, The Netherlands
| | - Sylvia Heeneman
- From the Institut National de la Santé et de la Recherche Médicale U858 (A.B., A.N.-S., M.-H.G., J.-C.T., R.S., F.M.-S.), Toulouse, France; Faculty of Medicine-Rangueil (A.B., A.N.-S., M.-H.G., J.-C.T., R.S., F.M.-S.), University Paul-Sabatier Toulouse III, Toulouse, France; and Department of Pathology (S.H.), Cardiovascular Research Institute Maastricht, University of Maastricht, The Netherlands
| | - Marie-Hélène Grazide
- From the Institut National de la Santé et de la Recherche Médicale U858 (A.B., A.N.-S., M.-H.G., J.-C.T., R.S., F.M.-S.), Toulouse, France; Faculty of Medicine-Rangueil (A.B., A.N.-S., M.-H.G., J.-C.T., R.S., F.M.-S.), University Paul-Sabatier Toulouse III, Toulouse, France; and Department of Pathology (S.H.), Cardiovascular Research Institute Maastricht, University of Maastricht, The Netherlands
| | - Jean-Claude Thiers
- From the Institut National de la Santé et de la Recherche Médicale U858 (A.B., A.N.-S., M.-H.G., J.-C.T., R.S., F.M.-S.), Toulouse, France; Faculty of Medicine-Rangueil (A.B., A.N.-S., M.-H.G., J.-C.T., R.S., F.M.-S.), University Paul-Sabatier Toulouse III, Toulouse, France; and Department of Pathology (S.H.), Cardiovascular Research Institute Maastricht, University of Maastricht, The Netherlands
| | - Robert Salvayre
- From the Institut National de la Santé et de la Recherche Médicale U858 (A.B., A.N.-S., M.-H.G., J.-C.T., R.S., F.M.-S.), Toulouse, France; Faculty of Medicine-Rangueil (A.B., A.N.-S., M.-H.G., J.-C.T., R.S., F.M.-S.), University Paul-Sabatier Toulouse III, Toulouse, France; and Department of Pathology (S.H.), Cardiovascular Research Institute Maastricht, University of Maastricht, The Netherlands
| | - Françoise Maupas-Schwalm
- From the Institut National de la Santé et de la Recherche Médicale U858 (A.B., A.N.-S., M.-H.G., J.-C.T., R.S., F.M.-S.), Toulouse, France; Faculty of Medicine-Rangueil (A.B., A.N.-S., M.-H.G., J.-C.T., R.S., F.M.-S.), University Paul-Sabatier Toulouse III, Toulouse, France; and Department of Pathology (S.H.), Cardiovascular Research Institute Maastricht, University of Maastricht, The Netherlands
| |
Collapse
|
37
|
Gosens R, Meurs H, Schmidt M. The GSK-3/beta-catenin-signalling axis in smooth muscle and its relationship with remodelling. Naunyn Schmiedebergs Arch Pharmacol 2008; 378:185-91. [PMID: 18612673 PMCID: PMC2493600 DOI: 10.1007/s00210-008-0269-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2007] [Accepted: 01/30/2008] [Indexed: 12/11/2022]
Abstract
beta-Catenin is a plasma membrane-associated protein that plays a dual role in cellular signalling by stabilizing cadherin mediated cell-cell contact and by regulating TCF-/LEF-mediated gene transcription. Traditionally, the role of beta-catenin in health and disease has mainly been studied in the context of development and uncontrolled cell growth in diseases such as cancer. Recent findings indicate, however, that beta-catenin also plays a significant role in fibro-proliferative diseases of several organ systems and that beta-catenin regulates mitogenic responses of smooth muscle cells. As several diseases of the internal organs are characterized by structural and phenotypic abnormalities of smooth muscle, including increased fibro-proliferative responses, these findings implicate that beta-catenin could play a broad pathophysiological role. This article will review this potential novel role for beta-catenin and associated intracellular signalling in smooth muscle and discuss the hypothesis that it plays a central role in smooth muscle remodelling.
Collapse
Affiliation(s)
- Reinoud Gosens
- Department of Molecular Pharmacology, University of Groningen, Antonius Deusinglaan 1, 9713, AV, Groningen, The Netherlands.
| | | | | |
Collapse
|
38
|
Ikonomou L, Geras-Raaka E, Raaka BM, Gershengorn MC. Beta-catenin signalling in mesenchymal islet-derived precursor cells. Cell Prolif 2008; 41:474-91. [PMID: 18422699 DOI: 10.1111/j.1365-2184.2008.00527.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVES Previously, we characterized human islet-derived precursor cells (hIPCs) as mesenchymal stem cells that migrate out from islets in vitro and can differentiate into functional islet-like structures following proliferative expansion. Here, we investigate the role of beta-catenin signalling in derivation and proliferation of hIPCs. MATERIALS AND METHODS Localization of beta-catenin was performed using confocal microscopy. Expression levels of beta-catenin target genes were measured by quantitative real-time polymerase chain reaction. Loss-of-function studies were performed using specific short interfering RNAs. RESULTS Immunostaining of islet outgrowths revealed translocation of beta-catenin from plasma membranes in intact islets to the nucleus in cells migrating out. There were no nuclear beta-catenin-positive cells in intact islets whereas between 35% and 70% of cells in established hIPC cultures exhibited nuclear beta-catenin. Transcripts for beta-catenin target genes were increased in hIPCs compared to those in islets. Beta-catenin translocated to the cell membrane when hIPCs formed epithelial cell clusters. In proliferating hIPCs, there was a strong correlation between markers of proliferation and nuclear beta-catenin. Treatment of hIPCs with the glycogen synthase kinase-3beta inhibitor (2'Z,3'E)-6-Bromoindirubin-3'-oxime increased intracellular beta-catenin but reduced nuclear beta-catenin, and was associated with reduced cell proliferation. Finally, knockdown of beta-catenin decreased beta-catenin target gene expression and hIPC proliferation. CONCLUSIONS These results support a functional role for beta-catenin during proliferation of hIPCs and suggest that activated beta-catenin signalling may also be important during hIPC derivation from islets.
Collapse
Affiliation(s)
- L Ikonomou
- Clinical Endocrinology Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892-8029, USA
| | | | | | | |
Collapse
|
39
|
Wang T, Holt CM, Xu C, Ridley C, P O Jones R, Baron M, Trump D. Notch3 activation modulates cell growth behaviour and cross-talk to Wnt/TCF signalling pathway. Cell Signal 2007; 19:2458-67. [PMID: 17822871 DOI: 10.1016/j.cellsig.2007.07.019] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2007] [Accepted: 07/23/2007] [Indexed: 01/07/2023]
Abstract
Notch3 is one of the four Notch receptors identified in mammal and expressed mainly in the arterial smooth muscle cells of human adult. Signalling via Notch3 is thought to be important in maintaining the phenotypic stability of the cells, but the nature of the signalling and its regulation to other signalling pathways are largely unknown. To understand further of the cellular function of Notch3 signalling, we generated cell lines stably expressing a constitutively active form of human Notch3 comprising of its soluble intracellular domain (N3IC). The N3IC expressing cells showed accelerated proliferation, decreased migration, increased cell surface N-cadherin, and growth in a colonised fashion that was reversible by N-cadherin blockade. N3IC expressing cells were also protected significantly against staurosporine-induced apoptosis and exhibited lower caspase 3/7 activity, accompanied by up-regulation of pAKT compared to control cells. We also found a complex cross-talk between Notch3 signalling and the Wnt pathway. N3IC stimulated Wnt-independent T-cell factor (TCF, the target transcription factor in the Wnt pathway) activation which was associated with increased Tyr-142 phosphorylation of beta-catenin. In contrast N3IC suppressed TCF activation in response to LiCl, which mimics the Wnt-dependent TCF activation mechanism. We conclude that Notch3 promotes cell growth and survival by activating PI3-kinase/AKT pathway; N-cadherin participates in the change of cell growth caused by Notch3 activation; and Notch3 signalling has dual-effects on the Wnt/TCF pathway suggesting a buffering role that Notch3 signalling may play in balancing these two important signalling pathways in regulating cell function.
Collapse
Affiliation(s)
- Tao Wang
- Medical Genetics Research Group and Centre for Molecular Medicine, Faculty of Medicine and Human Sciences, The University of Manchester, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|
40
|
Razuvaev A, Henderson B, Girnita L, Larsson O, Axelson M, Hedin U, Roy J. The cyclolignan picropodophyllin attenuates intimal hyperplasia after rat carotid balloon injury by blocking insulin-like growth factor-1 receptor signaling. J Vasc Surg 2007; 46:108-15. [PMID: 17606126 DOI: 10.1016/j.jvs.2007.02.066] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2006] [Accepted: 02/27/2007] [Indexed: 10/23/2022]
Abstract
OBJECTIVE Smooth muscle cell proliferation (SMC) is a pivotal factor in the development of intimal hyperplasia after vascular injury. A number of growth factors, including insulin-like growth factor-1 (IGF-1), have been shown to be involved in SMC proliferation. We evaluated the effect of picropodophyllin (PPP), a new IGF-1 receptor inhibitor, in the prevention of SMC proliferation and development of intimal hyperplasia after vascular injury. METHODS The effects of systemic administration of PPP on intimal hyperplasia were studied in a balloon rat carotid injury model. Lesions were quantified by morphometry and SMC proliferation and apoptosis was studied by immunohistochemical staining for proliferating cell nuclear antigen (PCNA) and activated caspase 3, respectively. The effect of PPP on rat aortic SMC proliferation and apoptosis was studied in vitro by using cell counting, 3[H]-thymidine incorporation, and a flow cytometry assay for annexin V. Phosphorylation of the IGF-1 receptor, protein kinase B (Akt), and extracellular signal-regulated kinase 1/2 (ERK1/2) in vitro and in vivo were analyzed by using Western blotting. RESULTS PPP inhibited IGF-1-mediated SMC proliferation in vitro but no significant increase in apoptosis was detected. In rats treated with PPP, a more than a twofold reduction in carotid intima area was observed 2 weeks after balloon injury, a significant decrease in PCNA staining was demonstrated in early lesions, but activated caspase 3 was not detected. In addition, PPP attenuated phosphorylation of the IGF-1 receptor, Akt, and ERK1/2 in IGF-1-stimulated SMCs in vitro, and a reduced phosphorylation of the IGF-1 receptor and Akt was found in balloon-injured carotid arteries in rats treated with PPP. CONCLUSION These results show that PPP potently blocks IGF-1-mediated phosphorylation of the IGF-1 receptor in SMCs, decreases downstream Akt and ERK1/2 activation, inhibits SMC replication, and subsequently attenuates intimal hyperplasia after balloon injury of rat carotid arteries.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Cardiovascular Agents/pharmacology
- Cardiovascular Agents/therapeutic use
- Carotid Artery Injuries/drug therapy
- Carotid Artery Injuries/metabolism
- Carotid Artery Injuries/pathology
- Catheterization
- Cell Proliferation/drug effects
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Hyperplasia
- Insulin-Like Growth Factor I/metabolism
- Male
- Mitogen-Activated Protein Kinase 1/metabolism
- Mitogen-Activated Protein Kinase 3/metabolism
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Phosphorylation/drug effects
- Podophyllotoxin/analogs & derivatives
- Podophyllotoxin/pharmacology
- Podophyllotoxin/therapeutic use
- Proto-Oncogene Proteins c-akt/metabolism
- Rats
- Rats, Sprague-Dawley
- Receptor, IGF Type 1/antagonists & inhibitors
- Receptor, IGF Type 1/metabolism
- Signal Transduction/drug effects
- Time Factors
- Tunica Intima/drug effects
- Tunica Intima/metabolism
- Tunica Intima/pathology
Collapse
Affiliation(s)
- Anton Razuvaev
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Karolinska University Hospital, SE-17176 Stockholm, Sweden.
| | | | | | | | | | | | | |
Collapse
|
41
|
Proctor BM, Ren J, Chen Z, Schneider JG, Coleman T, Lupu TS, Semenkovich CF, Muslin AJ. Grb2 Is Required for Atherosclerotic Lesion Formation. Arterioscler Thromb Vasc Biol 2007; 27:1361-7. [PMID: 17363695 DOI: 10.1161/atvbaha.106.134007] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
OBJECTIVE Grb2 is a ubiquitously expressed linker protein that couples growth factor receptor activation to downstream mitogen-activated protein kinase (MAPK) cascades. Macrophage proliferation and uptake of modified lipoproteins are critical components of atherogenesis which require MAPK activation. However, the precise role of upstream signaling factors and the interrelationship of various MAPK cascades in the pathogenesis of atherosclerosis remains uncertain. Complete deletion of Grb2 in mice results in early embryonic lethality. However, Grb2 heterozygous mice appear normal at birth. To test the role of the Grb2 adapter protein in atherosclerotic lesion formation, we generated Grb2+/- mice in the apoE-/- genetic background. METHODS AND RESULTS Grb2+/- apoE-/- and apoE-/- mice exhibited similar body weight and serum lipid profiles. However, Grb2+/- apoE-/- mice on a Western diet had reduced lesion formation compared with apoE-/- mice by aortic sinus and en face assays. Transplantation of apoE-/- mice with Grb2+/- apoE-/- or apoE-/- bone marrow indicated that Grb2 haploinsufficiency in blood-borne cells confers resistance to Western diet-induced atherosclerosis. Cell culture experiments with bone marrow-derived macrophages showed that Grb2 is required for oxidized low density lipoprotein (oxLDL)-induced MAPK activation and foam cell formation. CONCLUSIONS Grb2 is required for atherosclerotic lesion formation and uptake of oxidized LDL by macrophages.
Collapse
Affiliation(s)
- Brandon M Proctor
- Center for Cardiovascular Research, Washington University School of Medicine, 660 South Euclid Avenue, Box 8086, St. Louis, MO 63110, USA
| | | | | | | | | | | | | | | |
Collapse
|
42
|
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
|