1
|
Shu S, Cui H, Liu Z, Zhang H, Yang Y, Chen X, Zeng Z, Du L, Fu M, Yang Z, Wang P, Wang C, Gao H, Yang Q, Lin X, Yang T, Chen Z, Wu S, Wang X, Zhao R, Hu S, Song J. Suppression of RCAN1 alleviated lipid accumulation and mitochondrial fission in diabetic cardiomyopathy. Metabolism 2024; 158:155977. [PMID: 39053690 DOI: 10.1016/j.metabol.2024.155977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/05/2024] [Accepted: 07/14/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND Although metabolic disturbance is a characteristic of diabetic cardiomyopathy (DbCM), the detailed pathogenesis of DbCM remains unknown. METHODS We used a heart transplantation (HTx) cohort to explore the effect of diabetes mellitus on heart failure (HF) progression dependent of myocardium. Microscopic and ultramicroscopic pathology were used to depict the pathological features of human myocardium of DbCM. We performed targeted metabolomics to characterize the metabolic phenotype of human DbCM. Transcriptomics data were analyzed and weighted gene co-expression network analysis was performed to explore the potential upstream regulator for metabolic remodeling of DbCM. In vivo and in vitro experiments were further conducted to demonstrate the therapeutic effects and molecular mechanisms. RESULTS DbCM promoted the progression of HF and increased death or HF-rehospitalization after HTx. Lipid accumulation and mitochondrial fission were the obvious pathological features of DbCM myocardium. The concentrations of C14:0-CoA and C16:1-CoA were significantly increased in the myocardium, and they were positively correlated with the accelerated HF progression and RCAN1 expression in DbCM patients. Knockdown of RCAN1 improved cardiac dysfunction, lipid accumulation, and mitochondrial fission in db/db mice. In vitro studies showed that RCAN1 knockdown improved mitochondrial dysfunction in DbCM cardiomyocytes via the RCAN1-p-Drp1 Ser616 axis. CONCLUSIONS Diabetes is associated with faster progression of HF and causes poor prognosis after HTx, accompanied by metabolic remodeling in the myocardium. Accumulation of long chain acyl-CoA in the myocardium is the metabolic hallmark of human DbCM and is associated with more rapid disease progression for DbCM patients. Upregulation of RCAN1 in the myocardium is associated with the metabolic signatures of DbCM and RCAN1 is a potential therapeutic target for DbCM.
Collapse
Affiliation(s)
- Songren Shu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; The Cardiomyopathy Research Group, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China; Department of Cardiac Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Hao Cui
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Fuwai Hospital, National Centre for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China; The Cardiomyopathy Research Group, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Zirui Liu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; The Cardiomyopathy Research Group, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Hang Zhang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; The Cardiomyopathy Research Group, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Yicheng Yang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiao Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; The Cardiomyopathy Research Group, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Zhiwei Zeng
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Leilei Du
- Laboratory of Cardiovascular Science, Beijing Clinical Research Institute, Beijing Friendship Hospital, Capital Medical University, 95 Yongan Road, Beijing 100050, China
| | - Mengxia Fu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; The Cardiomyopathy Research Group, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China; Galactophore Department, Galactophore Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Ziang Yang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Peizhi Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, CH 8952 Schlieren, Zurich, Switzerland
| | - Chuangshi Wang
- Medical Research and Biometrics Center, National Clinical Research Center for Cardiovascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College &Chinese Academy of Medical Sciences, Beijing, China
| | - Huimin Gao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qiaoxi Yang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaojun Lin
- Department of Big Data in Health Science School of Public Health, and Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Tianshuo Yang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhice Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Sijin Wu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaohu Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ruojin Zhao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shengshou Hu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Fuwai Hospital, National Centre for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China; The Cardiomyopathy Research Group, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China; Department of Cardiac Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China; Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen 518057, China.
| | - Jiangping Song
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Fuwai Hospital, National Centre for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China; The Cardiomyopathy Research Group, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China; Department of Cardiac Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China; Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen 518057, China.
| |
Collapse
|
2
|
Wen SY, Zhi X, Liu HX, Wang X, Chen YY, Wang L. Is the suppression of CD36 a promising way for atherosclerosis therapy? Biochem Pharmacol 2024; 219:115965. [PMID: 38043719 DOI: 10.1016/j.bcp.2023.115965] [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: 10/07/2023] [Revised: 11/29/2023] [Accepted: 11/30/2023] [Indexed: 12/05/2023]
Abstract
Atherosclerosis is the main underlying pathology of many cardiovascular diseases and is marked by plaque formation in the artery wall. It has posed a serious threat to the health of people all over the world. CD36 acts as a significant regulator of lipid homeostasis, which is closely associated with the onset and progression of atherosclerosis and may be a new therapeutic target. The abnormal overexpression of CD36 facilitates lipid accumulation, foam cell formation, inflammation, endothelial apoptosis, and thrombosis. Numerous natural products and lipid-lowering agents are found to target the suppression of CD36 or inhibit the upregulation of CD36 to prevent and treat atherosclerosis. Here, the structure, expression regulation and function of CD36 in atherosclerosis and its related pharmacological therapies are reviewed. This review highlights the importance of drugs targeting CD36 suppression in the treatment and prevention of atherosclerosis, in order to develop new therapeutic strategies and potential anti-atherosclerotic drugs both preclinically and clinically.
Collapse
Affiliation(s)
- Shi-Yuan Wen
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China
| | - Xiaoyan Zhi
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China
| | - Hai-Xin Liu
- School of Traditional Chinese Materia Medica, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Xiaohui Wang
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China
| | - Yan-Yan Chen
- School of Medicine, Jiangsu University, Zhenjiang, China.
| | - Li Wang
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
3
|
Mice with double knockout of Egr-1 and RCAN1 exhibit reduced inflammation during Pseudomonas aeruginosa lung infection. Immunobiology 2023; 228:152377. [PMID: 36933529 DOI: 10.1016/j.imbio.2023.152377] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/26/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023]
Abstract
Pseudomonas aeruginosa represents one of the major opportunistic pathogens, which causes nosocomial infections in immunocompromised individuals. The molecular mechanisms controlling the host immune response to P. aeruginosa infections are not completely understood. In our previous study, early growth response 1 (Egr-1) and regulator of calcineurin 1 (RCAN1) were found to positively and negatively regulate the inflammatory responses, respectively, during P. aeruginosa pulmonary infection, and both of them had an impact on activating NF-κB pathway. Herein, we examined the inflammatory responses of Egr-1/RCAN1 double knockout mice using a mouse model of P. aeruginosa acute pneumonia. As a result, the Egr-1/RCAN1 double knockout mice showed reduced production of proinflammatory cytokines (IL-1β, IL-6, TNF and MIP-2), diminished inflammatory cell infiltration and decreased mortality, which were similar to those of Egr-1-deficienct mice but different from those of RCAN1-deficient mice. In vitro studies demonstrated that Egr-1 mRNA transcription preceded RCAN1 isoform 4 (RCAN1.4) mRNA transcription in macrophages, and the macrophages with Egr-1 deficiency exhibited decreased RCAN1.4 mRNA levels upon P. aeruginosa LPS stimulation. Moreover, Egr-1/RCAN1 double-deficient macrophages had reduced NF-κB activation compared to RCAN1-deficient macrophages. Taken together, Egr-1 predominates over RCAN1 in regulating inflammation during P. aeruginosa acute lung infection, which influences RCAN1.4 gene expression.
Collapse
|
4
|
Yang X, Yun Y, Wang P, Zhao J, Sun X. Upregulation of RCAN1.4 by HIF1α alleviates OGD-induced inflammatory response in astrocytes. Ann Clin Transl Neurol 2022; 9:1224-1240. [PMID: 35836352 PMCID: PMC9380140 DOI: 10.1002/acn3.51624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 06/03/2022] [Accepted: 06/28/2022] [Indexed: 11/11/2022] Open
Abstract
OBJECTIVE Ischemic stroke is a leading cause of human mortality and long-term disability worldwide. As one of the main forms of regulator of calcineurin 1 (RCAN1), the contribution of RCAN1.4 in diverse biological and pathological conditions has been implicated. But the role of RCAN1.4 in ischemic stroke progression remains elusive. This study is to explore the expression changes and roles of RCAN1.4 in ischemic stroke as well as the underlying mechanisms for these changes and effects of RCAN1.4 in ischemic stroke. METHODS Middle cerebral artery occlusion model in C57BL/6J mice and oxygen-glucose deprivation (OGD) model in primary astrocytes were performed to induce the cerebral ischemic stroke. The expression pattern of RCAN1.4 was assessed using real-time quantitative PCR and western blotting in vivo and in vitro. Mechanistically, the underlying mechanism for the elevation of RCAN1.4 in the upstream was investigated. Lentiviruses were administrated, and the effect of RCAN1.4 in postischemic inflammation was clearly clarified. RESULTS Here we uncovered that RCAN1.4 was dramatically increased in mouse ischemic brains and OGD-induced primary astrocytes. HIF1α, activated upon OGD, significantly upregulated RCAN1.4 gene expression through specifically binding to the RCAN1.4 promoter region and activating its promoter activity. The functional hypoxia-responsive element (HRE) was located between -254 and -245 bp in the RCAN1.4 promoter region. Moreover, elevated RCAN1.4 alleviated the release of pro-inflammatory cytokines TNFα, IL1β, IL6 and reduced expression of iNOS, COX2 in primary astrocytes upon OGD, whereas RCAN1.4 silencing has the opposite effect. Of note, RCAN1.4 overexpression inhibited OGD-induced NF-κB activation in primary astrocytes, leading to decreased degradation of IκBα and reduced nuclear translocation of NF-κB/p65. INTERPRETATION Our results reveal a novel mechanism underscoring the upregulation of RCAN1.4 by HIF1α and the protective effect of RCAN1.4 against postischemic inflammation, suggesting its significance as a promising therapeutic target for ischemic stroke treatment.
Collapse
Affiliation(s)
- Xiaxin Yang
- Department of NeurologyQilu Hospital of Shandong UniversityJinanShandong ProvinceChina
| | - Yan Yun
- Department of RadiologyQilu Hospital of Shandong UniversityJinanShandong ProvinceChina
| | - Pin Wang
- NHC Key Laboratory of OtorhinolaryngologyQilu Hospital of Shandong UniversityJinanShandong ProvinceChina
- Department of OtorhinolaryngologyQilu Hospital of Shandong UniversityJinanShandong ProvinceChina
| | - Juan Zhao
- NHC Key Laboratory of OtorhinolaryngologyQilu Hospital of Shandong UniversityJinanShandong ProvinceChina
- Department of OtorhinolaryngologyQilu Hospital of Shandong UniversityJinanShandong ProvinceChina
| | - Xiulian Sun
- NHC Key Laboratory of OtorhinolaryngologyQilu Hospital of Shandong UniversityJinanShandong ProvinceChina
- Brain Research InstituteQilu Hospital of Shandong UniversityJinanShandong ProvinceChina
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health CommissionQilu Hospital of Shandong UniversityJinanShandong ProvinceChina
| |
Collapse
|
5
|
Nolze A, Köhler C, Ruhs S, Quarch K, Strätz N, Gekle M, Grossmann C. Calcineurin (PPP3CB) regulates angiotensin II-dependent vascular remodelling by potentiating EGFR signalling in mice. Acta Physiol (Oxf) 2021; 233:e13715. [PMID: 34228904 DOI: 10.1111/apha.13715] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 06/08/2021] [Accepted: 07/03/2021] [Indexed: 12/14/2022]
Abstract
AIM This study investigates the role of calcineurin for angiotensin II (AngII)-induced vascular remodelling with the help of a mouse model lacking the catalytic beta subunit of calcineurin (PPP3CB KO). METHODS Wildtype (WT) and PPP3CB KO mice were treated for 4 weeks with AngII followed by assessment of blood pressure, histological evaluation of aortas and mRNA analysis of aortic genes PPP3CB-dependently regulated by AngII. Primary murine vascular smooth muscle cells (VSMCs) were used for qPCR, ELISA and Western Blot experiments as well as wound healing and cell proliferation assays. RESULTS Upon AngII treatment, PPP3CB KO mice showed less aortic media thickening, lumen dilation and systolic blood pressure compared to WT mice. Next-generation sequencing data of aortic tissue indicated an increase in extracellular matrix components (EMCs), cell migration and cell proliferation. A PPP3CB-dependent increase in EMC was confirmed by qPCR in aorta and VSMCs. PPP3CB-dependent stimulation of VSMC migration could be verified by wound healing assays but markers of enhanced cell proliferation were only detectable in aortic tissue of WT mice but not in isolated WT or KO VSMCs. We could demonstrate in VSMCs with pharmacological inhibitors that PPP3CB leads to enhanced heparin-binding EGF-like growth factor (HB-EGF) secretion, epidermal growth factor receptor (EGFR) activation and consecutive stimulation of transforming growth factor β(TGFβ) and connective tissue growth factor (CTGF) signalling that enhances collagen expression. CONCLUSION AngII-induced vascular remodelling involves PPP3CB, which leads to enhanced EMC production, VSMC migration and sustained increase in systolic blood pressure via HBEGF/EGFR-TGFβ-CTGF signalling.
Collapse
Affiliation(s)
- Alexander Nolze
- Julius‐Bernstein‐Institute of PhysiologyMartin Luther University Halle‐Wittenberg Halle Germany
| | - Conny Köhler
- Julius‐Bernstein‐Institute of PhysiologyMartin Luther University Halle‐Wittenberg Halle Germany
| | - Stefanie Ruhs
- Julius‐Bernstein‐Institute of PhysiologyMartin Luther University Halle‐Wittenberg Halle Germany
| | - Katja Quarch
- Julius‐Bernstein‐Institute of PhysiologyMartin Luther University Halle‐Wittenberg Halle Germany
| | - Nicole Strätz
- Julius‐Bernstein‐Institute of PhysiologyMartin Luther University Halle‐Wittenberg Halle Germany
| | - Michael Gekle
- Julius‐Bernstein‐Institute of PhysiologyMartin Luther University Halle‐Wittenberg Halle Germany
| | - Claudia Grossmann
- Julius‐Bernstein‐Institute of PhysiologyMartin Luther University Halle‐Wittenberg Halle Germany
| |
Collapse
|
6
|
Ding HX, Xing N, Ma HF, Hou L, Zhou CX, Du YP, Wang FJ. Effect of umbilical cord blood stem cell transplantation on restenosis after endovascular interventional therapy for diabetic hindlimb vascular disease. PLoS One 2021; 16:e0255162. [PMID: 34379650 PMCID: PMC8357084 DOI: 10.1371/journal.pone.0255162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 07/09/2021] [Indexed: 11/18/2022] Open
Abstract
This study aimed to investigate the mechanism of human umbilical cord blood stem cell (HUCBSC) transplantation on restenosis after percutaneous transluminal angioplasty (PTA) for diabetic hindlimb vascular disease in rabbits. After successfully preparing a rabbit model of diabetic hindlimb vascular disease, 16 rabbits were randomly assigned to two groups. Of these, 8 rabbits received PTA surgery alone (PTA group), and the other 8 rabbits received PTA and HUCBSC (PTA+HUCBSC group) treatments. Five more healthy rabbits were set as healthy control (HC group). Samples were collected after 4 weeks of treatment. The expressions of regulator of calcineurin 1 (RCAN1) and calcineurin A (CnA) in the diseased artery were detected by immunofluorescence staining. The distribution of HUCBSCs was observed by pathological examination in transplanted artery, distal artery, and liver. Cytology experiments were applied to assess the levels of JAK and STAT3, and the migration and proliferation of human aortic vascular smooth muscle cells (HA-VSMC). In the rabbit model of diabetic vascular lesions in the hindlimbs, we found the stenosis of the femoral artery became more and more serious with time, and the expression level of PCNA positive cells was also gradually increased. The expression levels of RCAN1 and CnA in the PTA+HUCBSC group were significantly lower than those in PTA group. HUCBSC inhibited the migration and proliferation of HA-VSMC via JAK/STAT3 pathway. After HUCBSC local transplantation, HUCBSC had no distal tissue distribution. HUCBSC transplantation may prevent restenosis after PTA of diabetic hindlimb vascular disease through JAK/STAT3 pathway.
Collapse
Affiliation(s)
- Hai-Xia Ding
- Department of Endocrinology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Na Xing
- Department of Endocrinology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Hong-Fang Ma
- Department of Endocrinology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lin Hou
- Department of Endocrinology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Chao-Xi Zhou
- Department of Gastrointestinal Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ya-Ping Du
- Department of Endocrinology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Fu-Jun Wang
- Department of Endocrinology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- * E-mail:
| |
Collapse
|
7
|
Aortic disease in Marfan syndrome is caused by overactivation of sGC-PRKG signaling by NO. Nat Commun 2021; 12:2628. [PMID: 33976159 PMCID: PMC8113458 DOI: 10.1038/s41467-021-22933-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 04/01/2021] [Indexed: 02/06/2023] Open
Abstract
Thoracic aortic aneurysm, as occurs in Marfan syndrome, is generally asymptomatic until dissection or rupture, requiring surgical intervention as the only available treatment. Here, we show that nitric oxide (NO) signaling dysregulates actin cytoskeleton dynamics in Marfan Syndrome smooth muscle cells and that NO-donors induce Marfan-like aortopathy in wild-type mice, indicating that a marked increase in NO suffices to induce aortopathy. Levels of nitrated proteins are higher in plasma from Marfan patients and mice and in aortic tissue from Marfan mice than in control samples, indicating elevated circulating and tissue NO. Soluble guanylate cyclase and cGMP-dependent protein kinase are both activated in Marfan patients and mice and in wild-type mice treated with NO-donors, as shown by increased plasma cGMP and pVASP-S239 staining in aortic tissue. Marfan aortopathy in mice is reverted by pharmacological inhibition of soluble guanylate cyclase and cGMP-dependent protein kinase and lentiviral-mediated Prkg1 silencing. These findings identify potential biomarkers for monitoring Marfan Syndrome in patients and urge evaluation of cGMP-dependent protein kinase and soluble guanylate cyclase as therapeutic targets. Aortic aneurysm and dissection, the major problem linked to Marfan syndrome (MFS), lacks effective pharmacological treatment. Here, the authors show that the NO pathway is overactivated in MFS and that inhibition of guanylate cyclase and cGMP-dependent protein kinase reverts MFS aortopathy in mice.
Collapse
|
8
|
Loss of Down syndrome critical region-1 leads to cholesterol metabolic dysfunction that exaggerates hypercholesterolemia in ApoE-null background. J Biol Chem 2021; 296:100697. [PMID: 33895138 PMCID: PMC8142255 DOI: 10.1016/j.jbc.2021.100697] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/12/2021] [Accepted: 04/21/2021] [Indexed: 01/07/2023] Open
Abstract
Down syndrome critical region (DSCR)-1 functions as a feedback modulator for calcineurin-nuclear factor for activated T cell (NFAT) signals, which are crucial for cell proliferation and inflammation. Stable expression of DSCR-1 inhibits pathological angiogenesis and septic inflammation. DSCR-1 also plays a critical role in vascular wall remodeling associated with aneurysm development that occurs primarily in smooth muscle cells. Besides, Dscr-1 deficiency promotes the M1-to M2-like phenotypic switch in macrophages, which correlates to the reduction of denatured cholesterol uptakes. However, the distinct roles of DSCR-1 in cholesterol and lipid metabolism are not well understood. Here, we show that loss of apolipoprotein (Apo) E in mice with chronic hypercholesterolemia induced Dscr-1 expression in the liver and aortic atheroma. In Dscr-1-null mice fed a high-fat diet, oxidative- and endoplasmic reticulum (ER) stress was induced, and sterol regulatory element-binding protein (SREBP) 2 production in hepatocytes was stimulated. This exaggerated ApoE-/--mediated nonalcoholic fatty liver disease (NAFLD) and subsequent hypercholesterolemia. Genome-wide screening revealed that loss of both ApoE and Dscr-1 resulted in the induction of immune- and leukocyte activation-related genes in the liver compared with ApoE deficiency alone. However, expressions of inflammation-activated markers and levels of monocyte adhesion were suspended upon induction of the Dscr-1 null background in the aortic endothelium. Collectively, our study shows that the combined loss of Dscr-1 and ApoE causes metabolic dysfunction in the liver but reduces atherosclerotic plaques, thereby leading to a dramatic increase in serum cholesterol and the formation of sporadic vasculopathy.
Collapse
|
9
|
Wang S, Wang Y, Qiu K, Zhu J, Wu Y. RCAN1 in cardiovascular diseases: molecular mechanisms and a potential therapeutic target. Mol Med 2020; 26:118. [PMID: 33267791 PMCID: PMC7709393 DOI: 10.1186/s10020-020-00249-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 11/26/2020] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of mortality worldwide. Considerable efforts are needed to elucidate the underlying mechanisms for the prevention and treatment of CVDs. Regulator of calcineurin 1 (RCAN1) is involved in both development/maintenance of the cardiovascular system and the pathogenesis of CVDs. RCAN1 reduction protects against atherosclerosis by reducing the uptake of oxidized low-density lipoproteins, whereas RCAN1 has a protective effect on myocardial ischemia/reperfusion injury, myocardial hypertrophy and intramural hematoma/aortic rupture mainly mediated by maintaining mitochondrial function and inhibiting calcineurin and Rho kinase activity, respectively. In this review, the regulation and the function of RCAN1 are summarized. Moreover, the dysregulation of RCAN1 in CVDs is reviewed. In addition, the beneficial role of RCAN1 reduction in atherosclerosis and the protective role of RCAN1 in myocardial ischemia/reperfusion injury, myocardial hypertrophy and intramural hematoma /aortic rupture are discussed, as well as underlying mechanisms. Furthermore, the therapeutic potential and challenges of targeting RCAN1 for CVDs treatment are also discussed.
Collapse
Affiliation(s)
- Shuai Wang
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jianshe South Road No. 45, Rencheng District, Jining, 272013, Shandong, China.,Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, Jianshe South Road No. 45, Rencheng District, Jining, 272013, Shandong, China
| | - Yuqing Wang
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jianshe South Road No. 45, Rencheng District, Jining, 272013, Shandong, China.,Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, Jianshe South Road No. 45, Rencheng District, Jining, 272013, Shandong, China.,Cheeloo College of Medicine, Shandong University, Wenhua West Road No. 44, Lixia District, JinanShandong, 250012, China
| | - Kaixin Qiu
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jianshe South Road No. 45, Rencheng District, Jining, 272013, Shandong, China.,Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, Jianshe South Road No. 45, Rencheng District, Jining, 272013, Shandong, China.,Cheeloo College of Medicine, Shandong University, Wenhua West Road No. 44, Lixia District, JinanShandong, 250012, China
| | - Jin Zhu
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jianshe South Road No. 45, Rencheng District, Jining, 272013, Shandong, China.,Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, Jianshe South Road No. 45, Rencheng District, Jining, 272013, Shandong, China
| | - Yili Wu
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jianshe South Road No. 45, Rencheng District, Jining, 272013, Shandong, China. .,Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, Jianshe South Road No. 45, Rencheng District, Jining, 272013, Shandong, China.
| |
Collapse
|
10
|
Abstract
PURPOSE OF REVIEW Atherosclerosis is a chronic disease characterized by lipid retention and inflammation in the artery wall. The retention and oxidation of low-density lipoprotein (LDL) in sub-endothelial space play a critical role in atherosclerotic plaque formation and destabilization. Oxidized LDL (ox-LDL) and other modified LDL particles are avidly taken up by endothelial cells, smooth muscle cells, and macrophages mainly through several scavenger receptors, including CD36 which is a class B scavenger receptor and membrane glycoprotein. RECENT FINDINGS Animal studies performed on CD36-deficient mice suggest that deficiency of CD36 prevents the development of atherosclerosis, though with some debate. CD36 serves as a signaling hub protein at the crossroad of inflammation, lipid metabolism, and fatty acid metabolism. In addition, the level of soluble CD36 (unattached to cells) in the circulating blood was elevated in patients with atherosclerosis and other metabolic disorders. We performed a state-of-the-art review on the structure, ligands, functions, and regulation of CD36 in the context of atherosclerosis by focusing on the pathological role of CD36 in the dysfunction of endothelial cells, smooth muscle cells, monocytes/macrophages, and platelets. Finally, we highlight therapeutic possibilities to target CD36 expression/activity in atherosclerosis.
Collapse
|
11
|
Lloret A, Monllor P, Fuchsberger T, Giraldo E, Perluigi M, Vina J. Increased basal antioxidant levels in RCAN1 - deficient mice lowers oxidative injury after acute paraquat insult. Free Radic Res 2020; 54:442-454. [PMID: 32686528 DOI: 10.1080/10715762.2020.1798002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
RCAN1 is an inhibitor of the phosphatase calcineurin, which is involved in the regulation of oxidative stress and apoptosis, among other important cell processes. Here we have used RCAN1 deficient mice (RCAN1-/-) to elucidate its role after an acute oxidative insult such as paraquat injection. We have observed that RCAN1-/- mice show less oxidative damage than wildtype (WT) mice after treatment. Under basal conditions, RCAN1-/- animals express more calcineurin, heme oxygenase-1, Nrf2, and catalase compared to WT mice (controls). This may explain the less severe effect of paraquat treatment on RCAN1-/- mice compared to WT. We showed that oxidative stress is involved in the early stages of apoptosis, thus we determined the apoptotic effector BAD and found that decreases in RCAN1-/- mice after treatment with paraquat compared with WT in similar experimental conditions. Our results suggest that RCAN1 may be involved in the balance between oxidant and antioxidant species production in vivo.
Collapse
Affiliation(s)
- Ana Lloret
- Freshage Research Group, Department of Physiology, University of Valencia, CIBERFES-ISCIII, INCLIVA, Valencia, Spain
| | - Paloma Monllor
- Freshage Research Group, Department of Physiology, University of Valencia, CIBERFES-ISCIII, INCLIVA, Valencia, Spain
| | - Tanja Fuchsberger
- Freshage Research Group, Department of Physiology, University of Valencia, CIBERFES-ISCIII, INCLIVA, Valencia, Spain
| | - Esther Giraldo
- Freshage Research Group, Department of Physiology, University of Valencia, CIBERFES-ISCIII, INCLIVA, Valencia, Spain.,Department of Biotechnology, Universitat Politècnica de València, Valencia, Spain.,The Principe Felipe Research Center, Valencia, Spain
| | - Marzia Perluigi
- Department of Biochemical Sciences, Sapienza University of Rome, Rome, Italy
| | - Jose Vina
- Freshage Research Group, Department of Physiology, University of Valencia, CIBERFES-ISCIII, INCLIVA, Valencia, Spain
| |
Collapse
|
12
|
Roy-Vallejo E, Galván-Román JM, Moldenhauer F, Real de Asúa D. Adults with Down syndrome challenge another paradigm: When aging no longer entails arterial hypertension. J Clin Hypertens (Greenwich) 2020; 22:1127-1133. [PMID: 32644285 DOI: 10.1111/jch.13930] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 05/09/2020] [Accepted: 05/17/2020] [Indexed: 12/12/2022]
Abstract
The paradigmatic relationship between aging and atherosclerotic cardiovascular events does not apply to all patient populations. Though trisomy 21 (T21) and its phenotypic expression, Down syndrome (DS), are conditions that involve premature aging, the cardiovascular system of adults with DS appears to be particularly spared from this early senescence. Despite a higher prevalence of some classic cardiovascular risk factors in adults with DS than in the general population, such as dyslipidemia, obesity, or sedentarism, these individuals do not develop hypertension or suffer major cardiovascular events as they age. The protective factors that prevent the development of hypertension in T21 are not well established. Genes like RCAN1 and DYRK1A, both on chromosome 21 and over-expressed in adults with DS, appear to play a major role in cardiovascular prevention. Their regulation of the renin-angiotensin-aldosterone system (RAAS) and neprilysin synthesis could underlie the constitutive protection against arterial hypertension in adults with DS and explain the absence of increased arterial stiffness in this population. A better understanding of these molecular pathways could have enormous implications for the clinical management of adults with DS and might foster the development of novel therapeutic targets in cardiovascular prevention for the general population.
Collapse
Affiliation(s)
- Emilia Roy-Vallejo
- Adult Down Syndrome Unit, Department of Internal Medicine, Hospital Universitario de La Princesa, Madrid, Spain
| | - José María Galván-Román
- Adult Down Syndrome Unit, Department of Internal Medicine, Hospital Universitario de La Princesa, Madrid, Spain
| | - Fernando Moldenhauer
- Adult Down Syndrome Unit, Department of Internal Medicine, Hospital Universitario de La Princesa, Madrid, Spain
| | - Diego Real de Asúa
- Adult Down Syndrome Unit, Department of Internal Medicine, Hospital Universitario de La Princesa, Madrid, Spain
| |
Collapse
|
13
|
Dierssen M, Fructuoso M, Martínez de Lagrán M, Perluigi M, Barone E. Down Syndrome Is a Metabolic Disease: Altered Insulin Signaling Mediates Peripheral and Brain Dysfunctions. Front Neurosci 2020; 14:670. [PMID: 32733190 PMCID: PMC7360727 DOI: 10.3389/fnins.2020.00670] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 06/02/2020] [Indexed: 12/19/2022] Open
Abstract
Down syndrome (DS) is the most frequent chromosomal abnormality that causes intellectual disability, resulting from the presence of an extra complete or segment of chromosome 21 (HSA21). In addition, trisomy of HSA21 contributes to altered energy metabolism that appears to be a strong determinant in the development of pathological phenotypes associated with DS. Alterations include, among others, mitochondrial defects, increased oxidative stress levels, impaired glucose, and lipid metabolism, finally resulting in reduced energy production and cellular dysfunctions. These molecular defects seem to account for a high incidence of metabolic disorders, i.e., diabetes and/or obesity, as well as a higher risk of developing Alzheimer’s disease (AD) in DS. A dysregulation of the insulin signaling with reduced downstream pathways represents a common pathophysiological aspect in the development of both peripheral and central alterations leading to diabetes/obesity and AD. This is further strengthened by evidence showing that the molecular mechanisms responsible for such alterations appear to be similar between peripheral organs and brain. Considering that DS subjects are at high risk to develop either peripheral or brain metabolic defects, this review will discuss current knowledge about the link between trisomy of HSA21 and defects of insulin and insulin-related pathways in DS. Drawing the molecular signature underlying these processes in DS is a key challenge to identify novel drug targets and set up new prevention strategies aimed to reduce the impact of metabolic disorders and cognitive decline.
Collapse
Affiliation(s)
- Mara Dierssen
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Barcelona, Spain.,Universitat Pompeu Fabra, Barcelona, Spain.,Human Pharmacology and Clinical Neurosciences Research Group, Neurosciences Research Program, Hospital Del Mar Medical Research Institute (IMIM), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Marta Fructuoso
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Barcelona, Spain
| | - María Martínez de Lagrán
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Marzia Perluigi
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Rome, Italy
| | - Eugenio Barone
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Rome, Italy
| |
Collapse
|
14
|
Abstract
Purpose of Review In recent years, a family of adiponectin paralogs designated as C1q/TNF-related protein (CTRP) has attracted increasing attention. They are inflammatory adipocytokines mostly secreted from epicardial adipose tissue, which modulate the development and prognosis of coronary artery disease (CAD). This review summarizes the pathophysiological roles of individual members of the CTRP superfamily in the development of CAD. Recent Findings Recent studies have revealed how members of the CTRP family, CTRP1, CTRP3, CTRP5, CTRP9, CTRP12, and CTRP13, can influence both development and progression of CAD by modulating metabolic pathways, influencing immuno-inflammatory response, and regulating cardiovascular functions. Summary Research to date has not been sufficient to answer the specific mechanism of the CTRP family in the occurrence and development of CAD. This review explores the evidence of CTRP superfamily regulating different pathophysiology stages of CAD through the immuno-inflammation, glucose and lipid metabolism, and vascular endothelial function.
Collapse
Affiliation(s)
- Yueqiao Si
- Department of Cardiology, The Affiliated Hospital of Chengde Medical University, Chengde, 067000, Hebei, China
| | - Wenjun Fan
- Department of Cardiology, The Affiliated Hospital of Chengde Medical University, Chengde, 067000, Hebei, China
| | - Lixian Sun
- Department of Cardiology, The Affiliated Hospital of Chengde Medical University, Chengde, 067000, Hebei, China.
| |
Collapse
|
15
|
Mendez-Barbero N, Yuste-Montalvo A, Nuñez-Borque E, Jensen BM, Gutiérrez-Muñoz C, Tome-Amat J, Garrido-Arandia M, Díaz-Perales A, Ballesteros-Martinez C, Laguna JJ, Beitia J, Poulsen LK, Cuesta-Herranz J, Blanco-Colio LM, Esteban V. The TNF-like weak inducer of the apoptosis/fibroblast growth factor–inducible molecule 14 axis mediates histamine and platelet-activating factor–induced subcutaneous vascular leakage and anaphylactic shock. J Allergy Clin Immunol 2020; 145:583-596.e6. [DOI: 10.1016/j.jaci.2019.09.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 08/11/2019] [Accepted: 09/04/2019] [Indexed: 02/06/2023]
|
16
|
Conditional deletion of Rcan1 predisposes to hypertension-mediated intramural hematoma and subsequent aneurysm and aortic rupture. Nat Commun 2018; 9:4795. [PMID: 30442942 PMCID: PMC6237779 DOI: 10.1038/s41467-018-07071-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 10/04/2018] [Indexed: 12/20/2022] Open
Abstract
Aortic intramural hematoma (IMH) can evolve toward reabsorption, dissection or aneurysm. Hypertension is the most common predisposing factor in IMH and aneurysm patients, and the hypertensive mediator angiotensin-II induces both in mice. We have previously shown that constitutive deletion of Rcan1 isoforms prevents Angiotensin II-induced aneurysm in mice. Here we generate mice conditionally lacking each isoform or all isoforms in vascular smooth muscle cells, endothelial cells, or ubiquitously, to determine the contribution to aneurysm development of Rcan1 isoforms in vascular cells. Surprisingly, conditional Rcan1 deletion in either vascular cell-type induces a hypercontractile phenotype and aortic medial layer disorganization, predisposing to hypertension-mediated aortic rupture, IMH, and aneurysm. These processes are blocked by ROCK inhibition. We find that Rcan1 associates with GSK-3β, whose inhibition decreases myosin activation. Our results identify potential therapeutic targets for intervention in IMH and aneurysm and call for caution when interpreting phenotypes of constitutively and inducibly deficient mice.
Collapse
|
17
|
Vojinovic D, Kavousi M, Ghanbari M, Brouwer RWW, van Rooij JGJ, van den Hout MCGN, Kraaij R, van Ijcken WFJ, Uitterlinden AG, van Duijn CM, Amin N. Whole-Genome Linkage Scan Combined With Exome Sequencing Identifies Novel Candidate Genes for Carotid Intima-Media Thickness. Front Genet 2018; 9:420. [PMID: 30356672 PMCID: PMC6189289 DOI: 10.3389/fgene.2018.00420] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 09/10/2018] [Indexed: 01/06/2023] Open
Abstract
Carotid intima-media thickness (cIMT) is an established heritable marker for subclinical atherosclerosis. In this study, we aim to identify rare variants with large effects driving differences in cIMT by performing genome-wide linkage analysis of individuals in the extremes of cIMT trait distribution (>90th percentile) in a large family-based study from a genetically isolated population in the Netherlands. Linked regions were subsequently explored by fine-mapping using exome sequencing. We observed significant evidence of linkage on chromosomes 2p16.3 [rs1017418, heterogeneity LOD (HLOD) = 3.35], 19q13.43 (rs3499, HLOD = 9.09), 20p13 (rs1434789, HLOD = 4.10), and 21q22.12 (rs2834949, HLOD = 3.59). Fine-mapping using exome sequencing data identified a non-coding variant (rs62165235) in PNPT1 gene under the linkage peak at chromosome 2 that is likely to have a regulatory function. The variant was associated with quantitative cIMT in the family-based study population (effect = 0.27, p-value = 0.013). Furthermore, we identified several genes under the linkage peak at chromosome 21 highly expressed in tissues relevant for atherosclerosis. To conclude, our linkage analysis identified four genomic regions significantly linked to cIMT. Further analyses are needed to demonstrate involvement of identified candidate genes in development of atherosclerosis.
Collapse
Affiliation(s)
- Dina Vojinovic
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Maryam Kavousi
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Mohsen Ghanbari
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, Netherlands.,Department of Genetics, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Rutger W W Brouwer
- Department of Cell Biology, Center for Biomics, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Jeroen G J van Rooij
- Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Mirjam C G N van den Hout
- Department of Cell Biology, Center for Biomics, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Robert Kraaij
- Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Wilfred F J van Ijcken
- Department of Cell Biology, Center for Biomics, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Andre G Uitterlinden
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, Netherlands.,Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Cornelia M van Duijn
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, Netherlands.,Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Najaf Amin
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
18
|
Li H, Zhang W, Zhong F, Das GC, Xie Y, Li Z, Cai W, Jiang G, Choi J, Sidani M, Hyink DP, Lee K, Klotman PE, He JC. Epigenetic regulation of RCAN1 expression in kidney disease and its role in podocyte injury. Kidney Int 2018; 94:1160-1176. [PMID: 30366682 DOI: 10.1016/j.kint.2018.07.023] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Revised: 06/26/2018] [Accepted: 07/19/2018] [Indexed: 12/24/2022]
Abstract
Mounting evidence suggests that epigenetic modification is important in kidney disease pathogenesis. To determine whether epigenetic regulation is involved in HIV-induced kidney injury, we performed genome-wide methylation profiling and transcriptomic profiling of human primary podocytes infected with HIV-1. Comparison of DNA methylation and RNA sequencing profiles identified several genes that were hypomethylated with corresponding upregulated RNA expression in HIV-infected podocytes. Notably, we found only one hypermethylated gene with corresponding downregulated RNA expression, namely regulator of calcineurin 1 (RCAN1). Further, we found that RCAN1 RNA expression was suppressed in glomeruli in human diabetic nephropathy, IgA nephropathy, and lupus nephritis, and in mouse models of HIV-associated nephropathy and diabetic nephropathy. We confirmed that HIV infection or high glucose conditions suppressed RCAN1 expression in cultured podocytes. This suppression was alleviated upon pretreatment with DNA methyltransferase inhibitor 5-Aza-2'-deoxycytidine, suggesting that RCAN1 expression is epigenetically suppressed in the context of HIV infection and diabetic conditions. Mechanistically, increased expression of RCAN1 decreased HIV- or high glucose-induced nuclear factor of activated T cells (NFAT) transcriptional activity. Increased RCAN1 expression also stabilized actin cytoskeleton organization, consistent with the inhibition of the calcineurin pathway. In vivo, knockout of RCAN1 aggravated albuminuria and podocyte injury in mice with Adriamycin-induced nephropathy. Our findings suggest that epigenetic suppression of RCAN1 aggravates podocyte injury in the setting of HIV infection and diabetic nephropathy.
Collapse
Affiliation(s)
- Huilin Li
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Division of Nephrology, Department of Medicine, Xinhua Hospital affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Weijia Zhang
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Fang Zhong
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Gokul C Das
- Division of Nephrology, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Yifan Xie
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Zhengzhe Li
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Weijing Cai
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Gengru Jiang
- Division of Nephrology, Department of Medicine, Xinhua Hospital affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jae Choi
- Division of Nephrology, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Mohamad Sidani
- Division of Nephrology, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Deborah P Hyink
- Division of Nephrology, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Kyung Lee
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Paul E Klotman
- Division of Nephrology, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA.
| | - John Cijiang He
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Kidney Center at James J. Peters VA Medical Center, Bronx, New York, USA.
| |
Collapse
|
19
|
Norberg KJ, Nania S, Li X, Gao H, Szatmary P, Segersvärd R, Haas S, Wagman A, Arnelo U, Sutton R, Heuchel RL, Löhr JM. RCAN1 is a marker of oxidative stress, induced in acute pancreatitis. Pancreatology 2018; 18:734-741. [PMID: 30139658 DOI: 10.1016/j.pan.2018.08.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Accepted: 08/14/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND To date, there still is a lack of specific acute pancreatitis markers and specifically an early marker that can reliably predict disease severity. The inflammatory response in acute pancreatitis is mediated in part through oxidative stress and calcineurin-NFAT (Nuclear Factor of Activated T-cells) signaling, which is inducing its own negative regulator, regulator of calcineurin 1 (RCAN1). Caerulein induction is a commonly used in vivo model of experimental acute pancreatitis. Caerulein induces CN-NFAT signaling, reactive oxygen species and inflammation. METHODS To screen for potential markers of acute pancreatitis, we used the caerulein model of experimental acute pancreatitis (AP) in C57Bl/6 J mice. Pancreata from treated and control mice were used for expression profiling. Promising gene candidates were validated in cell culture experiments using primary murine acinar cells and rat AR42J cells. These candidates were then further tested for their usefulness as biomarkers in mouse and human plasma. RESULTS We identified a number of novel genes, including Regulator of calcineurin 1 (Rcan1) and Sestrin 2 (Sesn2) and demonstrated that they are induced by oxidative stress, by stimulation with H2O2 and by inhibiting caerulein stimulated expression with the antioxidant N-acetylcysteine. We found Rcan1 protein to be significantly elevated in AP-induced mouse plasma as well as in plasma from AP patients. CONCLUSION We demonstrated that Rcan1 is regulated by oxidative stress and identified RCAN1 as a potential diagnostic marker of AP.
Collapse
Affiliation(s)
- K Jessica Norberg
- Pancreas Cancer Research Lab, Dept. of Clinical Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
| | - Salvatore Nania
- Pancreas Cancer Research Lab, Dept. of Clinical Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
| | - Xuan Li
- Pancreas Cancer Research Lab, Dept. of Clinical Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
| | - Hui Gao
- Dept. of Biosciences and Nutrition (BioNut), Karolinska Institutet, Stockholm, Sweden
| | - Peter Szatmary
- Liverpool Pancreatitis Research Group, Institute of Translational Medicine, University of Liverpool, Liverpool, England, UK
| | - Ralf Segersvärd
- Centre for Digestive Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Stephan Haas
- Centre for Digestive Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Annika Wagman
- Pancreas Cancer Research Lab, Dept. of Clinical Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
| | - Urban Arnelo
- Centre for Digestive Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Robert Sutton
- Liverpool Pancreatitis Research Group, Institute of Translational Medicine, University of Liverpool, Liverpool, England, UK
| | - Rainer L Heuchel
- Pancreas Cancer Research Lab, Dept. of Clinical Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
| | - J Matthias Löhr
- Pancreas Cancer Research Lab, Dept. of Clinical Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden; Centre for Digestive Diseases, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
20
|
Wang C, Xu W, Liang M, Huang D, Huang K. CTRP13 inhibits atherosclerosis
via
autophagy‐lysosome‐dependent degradation of CD36. FASEB J 2018; 33:2290-2300. [DOI: 10.1096/fj.201801267rr] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Cheng Wang
- Clinic Center of Human Gene ResearchUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of CardiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Wenjing Xu
- Clinic Center of Human Gene ResearchUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of CardiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Minglu Liang
- Clinic Center of Human Gene ResearchUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Dan Huang
- Clinic Center of Human Gene ResearchUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of CardiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Kai Huang
- Clinic Center of Human Gene ResearchUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of CardiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
21
|
MT4-MMP deficiency increases patrolling monocyte recruitment to early lesions and accelerates atherosclerosis. Nat Commun 2018; 9:910. [PMID: 29500407 PMCID: PMC5834547 DOI: 10.1038/s41467-018-03351-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 02/07/2018] [Indexed: 12/16/2022] Open
Abstract
Matrix metalloproteinases are involved in vascular remodeling. Little is known about their immune regulatory role in atherosclerosis. Here we show that mice deficient for MT4-MMP have increased adherence of macrophages to inflamed peritonea, and larger lipid deposits and macrophage burden in atherosclerotic plaques. We also demonstrate that MT4-MMP deficiency results in higher numbers of patrolling monocytes crawling and adhered to inflamed endothelia, and the accumulation of Mafb+ apoptosis inhibitor of macrophage (AIM)+ macrophages at incipient atherosclerotic lesions in mice. Functionally, MT4-MMP-null Mafb+AIM+ peritoneal macrophages express higher AIM and scavenger receptor CD36, are more resistant to apoptosis, and bind acLDL avidly, all of which contribute to atherosclerosis. CCR5 inhibition alleviates these effects by hindering the enhanced recruitment of MT4-MMP-null patrolling monocytes to early atherosclerotic lesions, thus blocking Mafb+AIM+ macrophage accumulation and atherosclerosis acceleration. Our results suggest that MT4-MMP targeting may constitute a novel strategy to boost patrolling monocyte activity in early inflammation.
Collapse
|
22
|
García-Redondo AB, Esteban V, Briones AM, Díaz Del Campo LS, González-Amor M, Méndez-Barbero N, Campanero MR, Redondo JM, Salaices M. Regulator of calcineurin 1 modulates vascular contractility and stiffness through the upregulation of COX-2-derived prostanoids. Pharmacol Res 2018; 133:236-249. [PMID: 29309904 DOI: 10.1016/j.phrs.2018.01.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 12/18/2017] [Accepted: 01/03/2018] [Indexed: 12/14/2022]
Abstract
Cyclooxygenase-2 (COX-2) derived-prostanoids participate in the altered vascular function and mechanical properties in cardiovascular diseases. We investigated whether regulator of calcineurin 1 (Rcan1) participates in vascular contractility and stiffness through the regulation of COX-2. For this, wild type (Rcan1+/+) and Rcan1-deficient (Rcan1-/-) mice untreated or treated with the COX-2 inhibitor rofecoxib were used. Vascular function and structure were analysed by myography. COX-2 and phospo-p65 expression were studied by western blotting and immunohistochemistry and TXA2 production by ELISA. We found that Rcan1 deficiency increases COX-2 and IL-6 expression and NF-κB activation in arteries and vascular smooth muscle cells (VSMC). Adenoviral-mediated re-expression of Rcan1.4 in Rcan1-/- VSMC normalized COX-2 expression. Phenylephrine-induced vasoconstrictor responses were greater in aorta from Rcan1-/- compared to Rcan1+/+ mice. This increased response were diminished by etoricoxib, furegrelate, SQ 29548, cyclosporine A and parthenolide, inhibitors of COX-2, TXA2 synthase, TP receptors, calcineurin and NF-κB, respectively. Endothelial removal and NOS inhibition increased phenylephrine responses only in Rcan1+/+ mice. TXA2 levels were greater in Rcan1-/- mice. In small mesenteric arteries, vascular function and structure were similar in both groups of mice; however, vessels from Rcan1-/- mice displayed an increase in vascular stiffness that was diminished by rofecoxib. In conclusion, our results suggest that Rcan1 might act as endogenous negative modulator of COX-2 expression and activity by inhibiting calcineurin and NF-kB pathways to maintain normal contractility and vascular stiffness in aorta and small mesenteric arteries, respectively. Our results uncover a new role for Rcan1 in vascular contractility and mechanical properties.
Collapse
Affiliation(s)
- Ana B García-Redondo
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain; CIBER de Enfermedades Cardiovasculares, Spain
| | - Vanesa Esteban
- Gene Regulation in Cardiovascular Remodeling and Inflammation Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; Departamento de Inmunología, Instituto de Investigación Fundación Jiménez Díaz, Madrid, Spain
| | - Ana M Briones
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain; CIBER de Enfermedades Cardiovasculares, Spain.
| | - Lucía S Díaz Del Campo
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - María González-Amor
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - Nerea Méndez-Barbero
- Gene Regulation in Cardiovascular Remodeling and Inflammation Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Miguel R Campanero
- CIBER de Enfermedades Cardiovasculares, Spain; Departmento de Biología del Cáncer, Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-UAM, Madrid, Spain
| | - Juan M Redondo
- CIBER de Enfermedades Cardiovasculares, Spain; Gene Regulation in Cardiovascular Remodeling and Inflammation Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Mercedes Salaices
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain; CIBER de Enfermedades Cardiovasculares, Spain.
| |
Collapse
|
23
|
Valenti D, Braidy N, De Rasmo D, Signorile A, Rossi L, Atanasov AG, Volpicella M, Henrion-Caude A, Nabavi SM, Vacca RA. Mitochondria as pharmacological targets in Down syndrome. Free Radic Biol Med 2018; 114:69-83. [PMID: 28838841 DOI: 10.1016/j.freeradbiomed.2017.08.014] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 08/16/2017] [Accepted: 08/18/2017] [Indexed: 12/17/2022]
Abstract
Mitochondria play a pivotal role in cellular energy-generating processes and are considered master regulators of cell life and death fate. Mitochondrial function integrates signalling networks in several metabolic pathways controlling neurogenesis and neuroplasticity. Indeed, dysfunctional mitochondria and mitochondrial-dependent activation of intracellular stress cascades are critical initiating events in many human neurodegenerative or neurodevelopmental diseases including Down syndrome (DS). It is well established that trisomy of human chromosome 21 can cause DS. DS is associated with neurodevelopmental delay, intellectual disability and early neurodegeneration. Recently, molecular mechanisms responsible for mitochondrial damage and energy deficits have been identified and characterized in several DS-derived human cells and animal models of DS. Therefore, therapeutic strategies targeting mitochondria could have great potential for new treatment regimens in DS. The purpose of this review is to highlight recent studies concerning mitochondrial impairment in DS, focusing on alterations of the molecular pathways controlling mitochondrial function. We will also discuss the effects and molecular mechanisms of naturally occurring and chemically synthetized drugs that exert neuroprotective effects through modulation of mitochondrial function and attenuation of oxidative stress. These compounds might represent novel therapeutic tools for the modulation of energy deficits in DS.
Collapse
Affiliation(s)
- Daniela Valenti
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Council of Research, Bari, Italy
| | - Nady Braidy
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Australia
| | - Domenico De Rasmo
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Council of Research, Bari, Italy
| | - Anna Signorile
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari, Italy
| | - Leonardo Rossi
- Department of Clinical and Experimental Medicine, University of Pisa, Italy
| | - A G Atanasov
- Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, 05-552 Jastrzebiec, Poland; Department of Pharmacognosy, University of Vienna, 1090 Vienna, Austria; Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Mariateresa Volpicella
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Alexandra Henrion-Caude
- INSERM U1163, Université Paris Descartes, Sorbonne Paris Cité, Institut Imagine, GenAtlas Platform, 24 Boulevard du Montparnasse, 75015 Paris, France
| | - S M Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - R A Vacca
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Council of Research, Bari, Italy.
| |
Collapse
|
24
|
Ballesteros-Martinez C, Mendez-Barbero N, Montalvo-Yuste A, Jensen BM, Gomez-Cardenosa A, Klitfod L, Garrido-Arandia M, Alvarez-Llamas G, Pastor-Vargas C, Vivanco F, Garvey LH, Cuesta-Herranz J, Poulsen LK, Esteban V. Endothelial Regulator of Calcineurin 1 Promotes Barrier Integrity and Modulates Histamine-Induced Barrier Dysfunction in Anaphylaxis. Front Immunol 2017; 8:1323. [PMID: 29104573 PMCID: PMC5655011 DOI: 10.3389/fimmu.2017.01323] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 09/29/2017] [Indexed: 12/21/2022] Open
Abstract
Anaphylaxis, the most serious and life-threatening allergic reaction, produces the release of inflammatory mediators by mast cells and basophils. Regulator of calcineurin 1 (Rcan1) is a negative regulator of mast-cell degranulation. The action of mediators leads to vasodilation and an increase in vascular permeability, causing great loss of intravascular volume in a short time. Nevertheless, the molecular basis remains unexplored on the vascular level. We investigated Rcan1 expression induced by histamine, platelet-activating factor (PAF), and epinephrine in primary human vein (HV)-/artery (HA)-derived endothelial cells (ECs) and human dermal microvascular ECs (HMVEC-D). Vascular permeability was analyzed in vitro in human ECs with forced Rcan1 expression using Transwell migration assays and in vivo using Rcan1 knockout mice. Histamine, but neither PAF nor epinephrine, induced Rcan1-4 mRNA and protein expression in primary HV-ECs, HA-ECs, and HMVEC-D through histamine receptor 1 (H1R). These effects were prevented by pharmacological inhibition of calcineurin with cyclosporine A. Moreover, intravenous histamine administration increased Rcan1 expression in lung tissues of mice undergoing experimental anaphylaxis. Functional in vitro assays showed that overexpression of Rcan1 promotes barrier integrity, suggesting a role played by this molecule in vascular permeability. Consistent with these findings, in vivo models of subcutaneous and intravenous histamine-mediated fluid extravasation showed increased response in skin, aorta, and lungs of Rcan1-deficient mice compared with wild-type animals. These findings reveal that endothelial Rcan1 is synthesized in response to histamine through a calcineurin-sensitive pathway and may reduce barrier breakdown, thus contributing to the strengthening of the endothelium and resistance to anaphylaxis. These new insights underscore its potential role as a regulator of sensitivity to anaphylaxis in humans.
Collapse
Affiliation(s)
| | - Nerea Mendez-Barbero
- Department of Vascular Physiopathology, IIS-Fundación Jiménez Díaz, Autonomous University of Madrid, Madrid, Spain
| | - Alma Montalvo-Yuste
- Department of Immunology, IIS-Fundación Jiménez Díaz, Autonomous University of Madrid, Madrid, Spain
| | - Bettina M Jensen
- Allergy Clinic, Gentofte Hospital, Copenhagen University Hospital, Hellerup, Denmark
| | | | - Lotte Klitfod
- Surgery Department, Gentofte Hospital, Copenhagen University Hospital, Hellerup, Denmark
| | - María Garrido-Arandia
- Center for Plant Biotechnology and Genomics, Technical University of Madrid, Madrid, Spain
| | - Gloria Alvarez-Llamas
- Department of Immunology, IIS-Fundación Jiménez Díaz, Autonomous University of Madrid, Madrid, Spain
| | - Carlos Pastor-Vargas
- Department of Immunology, IIS-Fundación Jiménez Díaz, Autonomous University of Madrid, Madrid, Spain
| | - Fernando Vivanco
- Department of Immunology, IIS-Fundación Jiménez Díaz, Autonomous University of Madrid, Madrid, Spain
| | - Lene Heise Garvey
- Allergy Clinic, Gentofte Hospital, Copenhagen University Hospital, Hellerup, Denmark
| | - Javier Cuesta-Herranz
- Department of Immunology, IIS-Fundación Jiménez Díaz, Autonomous University of Madrid, Madrid, Spain.,Department of Allergy, Fundación Jiménez Díaz, Madrid, Spain
| | - Lars K Poulsen
- Allergy Clinic, Gentofte Hospital, Copenhagen University Hospital, Hellerup, Denmark
| | - Vanesa Esteban
- Department of Immunology, IIS-Fundación Jiménez Díaz, Autonomous University of Madrid, Madrid, Spain.,Allergy Clinic, Gentofte Hospital, Copenhagen University Hospital, Hellerup, Denmark
| |
Collapse
|
25
|
Colvin KL, Yeager ME. What people with Down Syndrome can teach us about cardiopulmonary disease. Eur Respir Rev 2017; 26:26/143/160098. [DOI: 10.1183/16000617.0098-2016] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 11/13/2016] [Indexed: 12/19/2022] Open
Abstract
Down syndrome is the most common chromosomal abnormality among live-born infants. Through full or partial trisomy of chromosome 21, Down syndrome is associated with cognitive impairment, congenital malformations (particularly cardiovascular) and dysmorphic features. Immune disturbances in Down syndrome account for an enormous disease burden ranging from quality-of-life issues (autoimmune alopecia) to more serious health issues (autoimmune thyroiditis) and life-threatening issues (leukaemia, respiratory tract infections and pulmonary hypertension). Cardiovascular and pulmonary diseases account for ∼75% of the mortality seen in persons with Down syndrome. This review summarises the cardiovascular, respiratory and immune challenges faced by individuals with Down syndrome, and the genetic underpinnings of their pathobiology. We strongly advocate increased comparative studies of cardiopulmonary disease in persons with and without Down syndrome, as we believe these will lead to new strategies to prevent and treat diseases affecting millions of people worldwide.
Collapse
|
26
|
Nitric oxide mediates aortic disease in mice deficient in the metalloprotease Adamts1 and in a mouse model of Marfan syndrome. Nat Med 2017; 23:200-212. [PMID: 28067899 DOI: 10.1038/nm.4266] [Citation(s) in RCA: 138] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 12/13/2016] [Indexed: 12/29/2022]
Abstract
Heritable thoracic aortic aneurysms and dissections (TAAD), including Marfan syndrome (MFS), currently lack a cure, and causative mutations have been identified for only a fraction of affected families. Here we identify the metalloproteinase ADAMTS1 and inducible nitric oxide synthase (NOS2) as therapeutic targets in individuals with TAAD. We show that Adamts1 is a major mediator of vascular homeostasis, given that genetic haploinsufficiency of Adamts1 in mice causes TAAD similar to MFS. Aortic nitric oxide and Nos2 levels were higher in Adamts1-deficient mice and in a mouse model of MFS (hereafter referred to as MFS mice), and Nos2 inactivation protected both types of mice from aortic pathology. Pharmacological inhibition of Nos2 rapidly reversed aortic dilation and medial degeneration in young Adamts1-deficient mice and in young or old MFS mice. Patients with MFS showed elevated NOS2 and decreased ADAMTS1 protein levels in the aorta. These findings uncover a possible causative role for the ADAMTS1-NOS2 axis in human TAAD and warrant evaluation of NOS2 inhibitors for therapy.
Collapse
|
27
|
Oh B, Lee CH. Development of Thiolated-Graphene Quantum Dots for Regulation of ROS in macrophages. Pharm Res 2016; 33:2736-47. [DOI: 10.1007/s11095-016-2000-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 07/11/2016] [Indexed: 01/18/2023]
|
28
|
Regulator of Calcineurin 1 in Periodontal Disease. Mediators Inflamm 2016; 2016:5475821. [PMID: 27403036 PMCID: PMC4925939 DOI: 10.1155/2016/5475821] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 04/20/2016] [Accepted: 05/05/2016] [Indexed: 11/18/2022] Open
Abstract
Nuclear factor of activated T-cells (NFAT) and NF-kB pathway associated processes are involved in the pathogenesis of various inflammatory disorders, for example, periodontal disease. The activation of these pathways is controlled by the regulator of calcineurin 1 (RCAN1). The aim of this study was to elucidate the role of RCAN1 in periodontal disease. Healthy and inflamed periodontal tissues were analyzed by immunohistochemistry and immunofluorescence using specific rabbit polyclonal anti-RCAN1 antibodies. For expression analysis human umbilical vein endothelial cells (HUVEC) were used. HUVEC were incubated for 2 h with Vascular Endothelial Growth Factor (VEGF) or with wild type and laboratory strains of Porphyromonas gingivalis (P. gingivalis). Expression analysis of rcan1 and cox2 was done by real time PCR using specific primers for rcan1.4 and cox2. The expression of rcan1 was found to be significantly suppressed in endothelial cells of chronically inflamed periodontal tissues compared to healthy controls. Rcan1 and cox2 were significantly induced by VEGF and wild type and laboratory P. gingivalis strains. Interestingly, the magnitude of the rcan1 and cox2 induction was strain dependent. The results of this study indicate that RCAN1 is suppressed in endothelial cells of chronically inflamed periodontal tissues. During an acute infection, however, rcan1 seems to be upregulated in endothelial cells, indicating a modulating role in immune homeostasis of periodontal tissues.
Collapse
|
29
|
Fearnley GW, Bruns AF, Wheatcroft SB, Ponnambalam S. VEGF-A isoform-specific regulation of calcium ion flux, transcriptional activation and endothelial cell migration. Biol Open 2015; 4:731-42. [PMID: 25910937 PMCID: PMC4467193 DOI: 10.1242/bio.201410884] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Vascular endothelial growth factor A (VEGF-A) regulates many aspects of vascular physiology such as cell migration, proliferation, tubulogenesis and cell-cell interactions. Numerous isoforms of VEGF-A exist but their physiological significance is unclear. Here we evaluated two different VEGF-A isoforms and discovered differential regulation of cytosolic calcium ion flux, transcription factor localisation and endothelial cell response. Analysis of VEGF-A isoform-specific stimulation of VEGFR2-dependent signal transduction revealed differential capabilities for isoform activation of multiple signal transduction pathways. VEGF-A165 treatment promoted increased phospholipase Cγ1 phosphorylation, which was proportional to the subsequent rise in cytosolic calcium ions, in comparison to cells treated with VEGF-A121. A major consequence of this VEGF-A isoform-specific calcium ion flux in endothelial cells is differential dephosphorylation and subsequent nuclear translocation of the transcription factor NFATc2. Using reverse genetics, we discovered that NFATc2 is functionally required for VEGF-A-stimulated endothelial cell migration but not tubulogenesis. This work presents a new mechanism for understanding how VEGF-A isoforms program complex cellular outputs by converting signal transduction pathways into transcription factor redistribution to the nucleus, as well as defining a novel role for NFATc2 in regulating the endothelial cell response.
Collapse
Affiliation(s)
- Gareth W Fearnley
- Endothelial Cell Biology Unit, School of Molecular & Cellular Biology, LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, UK
| | - Alexander F Bruns
- Division of Cardiovascular & Diabetes Research, Faculty of Medicine & Health, LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, UK
| | - Stephen B Wheatcroft
- Division of Cardiovascular & Diabetes Research, Faculty of Medicine & Health, LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, UK
| | - Sreenivasan Ponnambalam
- Endothelial Cell Biology Unit, School of Molecular & Cellular Biology, LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, UK
| |
Collapse
|
30
|
Yang H, Liu HP, Weng D, Ge BX. IL-10 negatively regulates oxLDL-P38 pathway inhibited macrophage emigration. Exp Mol Pathol 2014; 97:590-9. [DOI: 10.1016/j.yexmp.2014.10.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 10/28/2014] [Indexed: 01/10/2023]
|
31
|
Baggott RR, Alfranca A, López-Maderuelo D, Mohamed TMA, Escolano A, Oller J, Ornes BC, Kurusamy S, Rowther FB, Brown JE, Oceandy D, Cartwright EJ, Wang W, Gómez-del Arco P, Martínez-Martínez S, Neyses L, Redondo JM, Armesilla AL. Plasma membrane calcium ATPase isoform 4 inhibits vascular endothelial growth factor-mediated angiogenesis through interaction with calcineurin. Arterioscler Thromb Vasc Biol 2014; 34:2310-20. [PMID: 25147342 DOI: 10.1161/atvbaha.114.304363] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Vascular endothelial growth factor (VEGF) has been identified as a crucial regulator of physiological and pathological angiogenesis. Among the intracellular signaling pathways triggered by VEGF, activation of the calcineurin/nuclear factor of activated T cells (NFAT) signaling axis has emerged as a critical mediator of angiogenic processes. We and others previously reported a novel role for the plasma membrane calcium ATPase (PMCA) as an endogenous inhibitor of the calcineurin/NFAT pathway, via interaction with calcineurin, in cardiomyocytes and breast cancer cells. However, the functional significance of the PMCA/calcineurin interaction in endothelial pathophysiology has not been addressed thus far. APPROACH AND RESULTS Using in vitro and in vivo assays, we here demonstrate that the interaction between PMCA4 and calcineurin in VEGF-stimulated endothelial cells leads to downregulation of the calcineurin/NFAT pathway and to a significant reduction in the subsequent expression of the NFAT-dependent, VEGF-activated, proangiogenic genes RCAN1.4 and Cox-2. PMCA4-dependent inhibition of calcineurin signaling translates into a reduction in endothelial cell motility and blood vessel formation that ultimately impairs in vivo angiogenesis by VEGF. CONCLUSIONS Given the importance of the calcineurin/NFAT pathway in the regulation of pathological angiogenesis, targeted modulation of PMCA4 functionality might open novel therapeutic avenues to promote or attenuate new vessel formation in diseases that occur with angiogenesis.
Collapse
Affiliation(s)
- Rhiannon R Baggott
- From the Molecular Pharmacology Group, School of Pharmacy (R.R.B., S.K., A.L.A.), Brain Tumor UK Neuro-oncology Research Centre (F.B.R.), and Oncology Group (W.W.), Research Institute in Healthcare Science, Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton, United Kingdom; Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (A.A., D.L.-M., A.E., J.O., B.C.O., P.G.-d.A., S.M.-M., J.M.R.); Human Genetics Department, Institute for Rare Diseases Research, Carlos III Health Institute, Madrid, Spain (A.A.); Institute of Cardiovascular Sciences, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, United Kingdom (T.M.A.M., D.O., E.J.C., L.N.); Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt (T.M.A.M.); Aston Research Centre for Healthy Ageing, School of Life and Health Sciences, Aston University, Birmingham, United Kingdom (J.E.B.); Department of Molecular Biology, Universidad Autónoma de Madrid, Madrid, Spain (P.G.-d.A.); and University of Luxembourg, Walferdange, Luxembourg (L.N.)
| | - Arantzazu Alfranca
- From the Molecular Pharmacology Group, School of Pharmacy (R.R.B., S.K., A.L.A.), Brain Tumor UK Neuro-oncology Research Centre (F.B.R.), and Oncology Group (W.W.), Research Institute in Healthcare Science, Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton, United Kingdom; Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (A.A., D.L.-M., A.E., J.O., B.C.O., P.G.-d.A., S.M.-M., J.M.R.); Human Genetics Department, Institute for Rare Diseases Research, Carlos III Health Institute, Madrid, Spain (A.A.); Institute of Cardiovascular Sciences, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, United Kingdom (T.M.A.M., D.O., E.J.C., L.N.); Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt (T.M.A.M.); Aston Research Centre for Healthy Ageing, School of Life and Health Sciences, Aston University, Birmingham, United Kingdom (J.E.B.); Department of Molecular Biology, Universidad Autónoma de Madrid, Madrid, Spain (P.G.-d.A.); and University of Luxembourg, Walferdange, Luxembourg (L.N.)
| | - Dolores López-Maderuelo
- From the Molecular Pharmacology Group, School of Pharmacy (R.R.B., S.K., A.L.A.), Brain Tumor UK Neuro-oncology Research Centre (F.B.R.), and Oncology Group (W.W.), Research Institute in Healthcare Science, Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton, United Kingdom; Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (A.A., D.L.-M., A.E., J.O., B.C.O., P.G.-d.A., S.M.-M., J.M.R.); Human Genetics Department, Institute for Rare Diseases Research, Carlos III Health Institute, Madrid, Spain (A.A.); Institute of Cardiovascular Sciences, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, United Kingdom (T.M.A.M., D.O., E.J.C., L.N.); Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt (T.M.A.M.); Aston Research Centre for Healthy Ageing, School of Life and Health Sciences, Aston University, Birmingham, United Kingdom (J.E.B.); Department of Molecular Biology, Universidad Autónoma de Madrid, Madrid, Spain (P.G.-d.A.); and University of Luxembourg, Walferdange, Luxembourg (L.N.)
| | - Tamer M A Mohamed
- From the Molecular Pharmacology Group, School of Pharmacy (R.R.B., S.K., A.L.A.), Brain Tumor UK Neuro-oncology Research Centre (F.B.R.), and Oncology Group (W.W.), Research Institute in Healthcare Science, Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton, United Kingdom; Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (A.A., D.L.-M., A.E., J.O., B.C.O., P.G.-d.A., S.M.-M., J.M.R.); Human Genetics Department, Institute for Rare Diseases Research, Carlos III Health Institute, Madrid, Spain (A.A.); Institute of Cardiovascular Sciences, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, United Kingdom (T.M.A.M., D.O., E.J.C., L.N.); Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt (T.M.A.M.); Aston Research Centre for Healthy Ageing, School of Life and Health Sciences, Aston University, Birmingham, United Kingdom (J.E.B.); Department of Molecular Biology, Universidad Autónoma de Madrid, Madrid, Spain (P.G.-d.A.); and University of Luxembourg, Walferdange, Luxembourg (L.N.)
| | - Amelia Escolano
- From the Molecular Pharmacology Group, School of Pharmacy (R.R.B., S.K., A.L.A.), Brain Tumor UK Neuro-oncology Research Centre (F.B.R.), and Oncology Group (W.W.), Research Institute in Healthcare Science, Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton, United Kingdom; Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (A.A., D.L.-M., A.E., J.O., B.C.O., P.G.-d.A., S.M.-M., J.M.R.); Human Genetics Department, Institute for Rare Diseases Research, Carlos III Health Institute, Madrid, Spain (A.A.); Institute of Cardiovascular Sciences, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, United Kingdom (T.M.A.M., D.O., E.J.C., L.N.); Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt (T.M.A.M.); Aston Research Centre for Healthy Ageing, School of Life and Health Sciences, Aston University, Birmingham, United Kingdom (J.E.B.); Department of Molecular Biology, Universidad Autónoma de Madrid, Madrid, Spain (P.G.-d.A.); and University of Luxembourg, Walferdange, Luxembourg (L.N.)
| | - Jorge Oller
- From the Molecular Pharmacology Group, School of Pharmacy (R.R.B., S.K., A.L.A.), Brain Tumor UK Neuro-oncology Research Centre (F.B.R.), and Oncology Group (W.W.), Research Institute in Healthcare Science, Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton, United Kingdom; Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (A.A., D.L.-M., A.E., J.O., B.C.O., P.G.-d.A., S.M.-M., J.M.R.); Human Genetics Department, Institute for Rare Diseases Research, Carlos III Health Institute, Madrid, Spain (A.A.); Institute of Cardiovascular Sciences, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, United Kingdom (T.M.A.M., D.O., E.J.C., L.N.); Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt (T.M.A.M.); Aston Research Centre for Healthy Ageing, School of Life and Health Sciences, Aston University, Birmingham, United Kingdom (J.E.B.); Department of Molecular Biology, Universidad Autónoma de Madrid, Madrid, Spain (P.G.-d.A.); and University of Luxembourg, Walferdange, Luxembourg (L.N.)
| | - Beatriz C Ornes
- From the Molecular Pharmacology Group, School of Pharmacy (R.R.B., S.K., A.L.A.), Brain Tumor UK Neuro-oncology Research Centre (F.B.R.), and Oncology Group (W.W.), Research Institute in Healthcare Science, Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton, United Kingdom; Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (A.A., D.L.-M., A.E., J.O., B.C.O., P.G.-d.A., S.M.-M., J.M.R.); Human Genetics Department, Institute for Rare Diseases Research, Carlos III Health Institute, Madrid, Spain (A.A.); Institute of Cardiovascular Sciences, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, United Kingdom (T.M.A.M., D.O., E.J.C., L.N.); Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt (T.M.A.M.); Aston Research Centre for Healthy Ageing, School of Life and Health Sciences, Aston University, Birmingham, United Kingdom (J.E.B.); Department of Molecular Biology, Universidad Autónoma de Madrid, Madrid, Spain (P.G.-d.A.); and University of Luxembourg, Walferdange, Luxembourg (L.N.)
| | - Sathishkumar Kurusamy
- From the Molecular Pharmacology Group, School of Pharmacy (R.R.B., S.K., A.L.A.), Brain Tumor UK Neuro-oncology Research Centre (F.B.R.), and Oncology Group (W.W.), Research Institute in Healthcare Science, Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton, United Kingdom; Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (A.A., D.L.-M., A.E., J.O., B.C.O., P.G.-d.A., S.M.-M., J.M.R.); Human Genetics Department, Institute for Rare Diseases Research, Carlos III Health Institute, Madrid, Spain (A.A.); Institute of Cardiovascular Sciences, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, United Kingdom (T.M.A.M., D.O., E.J.C., L.N.); Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt (T.M.A.M.); Aston Research Centre for Healthy Ageing, School of Life and Health Sciences, Aston University, Birmingham, United Kingdom (J.E.B.); Department of Molecular Biology, Universidad Autónoma de Madrid, Madrid, Spain (P.G.-d.A.); and University of Luxembourg, Walferdange, Luxembourg (L.N.)
| | - Farjana B Rowther
- From the Molecular Pharmacology Group, School of Pharmacy (R.R.B., S.K., A.L.A.), Brain Tumor UK Neuro-oncology Research Centre (F.B.R.), and Oncology Group (W.W.), Research Institute in Healthcare Science, Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton, United Kingdom; Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (A.A., D.L.-M., A.E., J.O., B.C.O., P.G.-d.A., S.M.-M., J.M.R.); Human Genetics Department, Institute for Rare Diseases Research, Carlos III Health Institute, Madrid, Spain (A.A.); Institute of Cardiovascular Sciences, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, United Kingdom (T.M.A.M., D.O., E.J.C., L.N.); Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt (T.M.A.M.); Aston Research Centre for Healthy Ageing, School of Life and Health Sciences, Aston University, Birmingham, United Kingdom (J.E.B.); Department of Molecular Biology, Universidad Autónoma de Madrid, Madrid, Spain (P.G.-d.A.); and University of Luxembourg, Walferdange, Luxembourg (L.N.)
| | - James E Brown
- From the Molecular Pharmacology Group, School of Pharmacy (R.R.B., S.K., A.L.A.), Brain Tumor UK Neuro-oncology Research Centre (F.B.R.), and Oncology Group (W.W.), Research Institute in Healthcare Science, Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton, United Kingdom; Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (A.A., D.L.-M., A.E., J.O., B.C.O., P.G.-d.A., S.M.-M., J.M.R.); Human Genetics Department, Institute for Rare Diseases Research, Carlos III Health Institute, Madrid, Spain (A.A.); Institute of Cardiovascular Sciences, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, United Kingdom (T.M.A.M., D.O., E.J.C., L.N.); Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt (T.M.A.M.); Aston Research Centre for Healthy Ageing, School of Life and Health Sciences, Aston University, Birmingham, United Kingdom (J.E.B.); Department of Molecular Biology, Universidad Autónoma de Madrid, Madrid, Spain (P.G.-d.A.); and University of Luxembourg, Walferdange, Luxembourg (L.N.)
| | - Delvac Oceandy
- From the Molecular Pharmacology Group, School of Pharmacy (R.R.B., S.K., A.L.A.), Brain Tumor UK Neuro-oncology Research Centre (F.B.R.), and Oncology Group (W.W.), Research Institute in Healthcare Science, Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton, United Kingdom; Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (A.A., D.L.-M., A.E., J.O., B.C.O., P.G.-d.A., S.M.-M., J.M.R.); Human Genetics Department, Institute for Rare Diseases Research, Carlos III Health Institute, Madrid, Spain (A.A.); Institute of Cardiovascular Sciences, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, United Kingdom (T.M.A.M., D.O., E.J.C., L.N.); Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt (T.M.A.M.); Aston Research Centre for Healthy Ageing, School of Life and Health Sciences, Aston University, Birmingham, United Kingdom (J.E.B.); Department of Molecular Biology, Universidad Autónoma de Madrid, Madrid, Spain (P.G.-d.A.); and University of Luxembourg, Walferdange, Luxembourg (L.N.)
| | - Elizabeth J Cartwright
- From the Molecular Pharmacology Group, School of Pharmacy (R.R.B., S.K., A.L.A.), Brain Tumor UK Neuro-oncology Research Centre (F.B.R.), and Oncology Group (W.W.), Research Institute in Healthcare Science, Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton, United Kingdom; Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (A.A., D.L.-M., A.E., J.O., B.C.O., P.G.-d.A., S.M.-M., J.M.R.); Human Genetics Department, Institute for Rare Diseases Research, Carlos III Health Institute, Madrid, Spain (A.A.); Institute of Cardiovascular Sciences, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, United Kingdom (T.M.A.M., D.O., E.J.C., L.N.); Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt (T.M.A.M.); Aston Research Centre for Healthy Ageing, School of Life and Health Sciences, Aston University, Birmingham, United Kingdom (J.E.B.); Department of Molecular Biology, Universidad Autónoma de Madrid, Madrid, Spain (P.G.-d.A.); and University of Luxembourg, Walferdange, Luxembourg (L.N.)
| | - Weiguang Wang
- From the Molecular Pharmacology Group, School of Pharmacy (R.R.B., S.K., A.L.A.), Brain Tumor UK Neuro-oncology Research Centre (F.B.R.), and Oncology Group (W.W.), Research Institute in Healthcare Science, Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton, United Kingdom; Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (A.A., D.L.-M., A.E., J.O., B.C.O., P.G.-d.A., S.M.-M., J.M.R.); Human Genetics Department, Institute for Rare Diseases Research, Carlos III Health Institute, Madrid, Spain (A.A.); Institute of Cardiovascular Sciences, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, United Kingdom (T.M.A.M., D.O., E.J.C., L.N.); Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt (T.M.A.M.); Aston Research Centre for Healthy Ageing, School of Life and Health Sciences, Aston University, Birmingham, United Kingdom (J.E.B.); Department of Molecular Biology, Universidad Autónoma de Madrid, Madrid, Spain (P.G.-d.A.); and University of Luxembourg, Walferdange, Luxembourg (L.N.)
| | - Pablo Gómez-del Arco
- From the Molecular Pharmacology Group, School of Pharmacy (R.R.B., S.K., A.L.A.), Brain Tumor UK Neuro-oncology Research Centre (F.B.R.), and Oncology Group (W.W.), Research Institute in Healthcare Science, Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton, United Kingdom; Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (A.A., D.L.-M., A.E., J.O., B.C.O., P.G.-d.A., S.M.-M., J.M.R.); Human Genetics Department, Institute for Rare Diseases Research, Carlos III Health Institute, Madrid, Spain (A.A.); Institute of Cardiovascular Sciences, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, United Kingdom (T.M.A.M., D.O., E.J.C., L.N.); Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt (T.M.A.M.); Aston Research Centre for Healthy Ageing, School of Life and Health Sciences, Aston University, Birmingham, United Kingdom (J.E.B.); Department of Molecular Biology, Universidad Autónoma de Madrid, Madrid, Spain (P.G.-d.A.); and University of Luxembourg, Walferdange, Luxembourg (L.N.)
| | - Sara Martínez-Martínez
- From the Molecular Pharmacology Group, School of Pharmacy (R.R.B., S.K., A.L.A.), Brain Tumor UK Neuro-oncology Research Centre (F.B.R.), and Oncology Group (W.W.), Research Institute in Healthcare Science, Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton, United Kingdom; Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (A.A., D.L.-M., A.E., J.O., B.C.O., P.G.-d.A., S.M.-M., J.M.R.); Human Genetics Department, Institute for Rare Diseases Research, Carlos III Health Institute, Madrid, Spain (A.A.); Institute of Cardiovascular Sciences, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, United Kingdom (T.M.A.M., D.O., E.J.C., L.N.); Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt (T.M.A.M.); Aston Research Centre for Healthy Ageing, School of Life and Health Sciences, Aston University, Birmingham, United Kingdom (J.E.B.); Department of Molecular Biology, Universidad Autónoma de Madrid, Madrid, Spain (P.G.-d.A.); and University of Luxembourg, Walferdange, Luxembourg (L.N.)
| | - Ludwig Neyses
- From the Molecular Pharmacology Group, School of Pharmacy (R.R.B., S.K., A.L.A.), Brain Tumor UK Neuro-oncology Research Centre (F.B.R.), and Oncology Group (W.W.), Research Institute in Healthcare Science, Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton, United Kingdom; Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (A.A., D.L.-M., A.E., J.O., B.C.O., P.G.-d.A., S.M.-M., J.M.R.); Human Genetics Department, Institute for Rare Diseases Research, Carlos III Health Institute, Madrid, Spain (A.A.); Institute of Cardiovascular Sciences, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, United Kingdom (T.M.A.M., D.O., E.J.C., L.N.); Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt (T.M.A.M.); Aston Research Centre for Healthy Ageing, School of Life and Health Sciences, Aston University, Birmingham, United Kingdom (J.E.B.); Department of Molecular Biology, Universidad Autónoma de Madrid, Madrid, Spain (P.G.-d.A.); and University of Luxembourg, Walferdange, Luxembourg (L.N.)
| | - Juan Miguel Redondo
- From the Molecular Pharmacology Group, School of Pharmacy (R.R.B., S.K., A.L.A.), Brain Tumor UK Neuro-oncology Research Centre (F.B.R.), and Oncology Group (W.W.), Research Institute in Healthcare Science, Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton, United Kingdom; Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (A.A., D.L.-M., A.E., J.O., B.C.O., P.G.-d.A., S.M.-M., J.M.R.); Human Genetics Department, Institute for Rare Diseases Research, Carlos III Health Institute, Madrid, Spain (A.A.); Institute of Cardiovascular Sciences, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, United Kingdom (T.M.A.M., D.O., E.J.C., L.N.); Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt (T.M.A.M.); Aston Research Centre for Healthy Ageing, School of Life and Health Sciences, Aston University, Birmingham, United Kingdom (J.E.B.); Department of Molecular Biology, Universidad Autónoma de Madrid, Madrid, Spain (P.G.-d.A.); and University of Luxembourg, Walferdange, Luxembourg (L.N.).
| | - Angel Luis Armesilla
- From the Molecular Pharmacology Group, School of Pharmacy (R.R.B., S.K., A.L.A.), Brain Tumor UK Neuro-oncology Research Centre (F.B.R.), and Oncology Group (W.W.), Research Institute in Healthcare Science, Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton, United Kingdom; Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (A.A., D.L.-M., A.E., J.O., B.C.O., P.G.-d.A., S.M.-M., J.M.R.); Human Genetics Department, Institute for Rare Diseases Research, Carlos III Health Institute, Madrid, Spain (A.A.); Institute of Cardiovascular Sciences, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, United Kingdom (T.M.A.M., D.O., E.J.C., L.N.); Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt (T.M.A.M.); Aston Research Centre for Healthy Ageing, School of Life and Health Sciences, Aston University, Birmingham, United Kingdom (J.E.B.); Department of Molecular Biology, Universidad Autónoma de Madrid, Madrid, Spain (P.G.-d.A.); and University of Luxembourg, Walferdange, Luxembourg (L.N.).
| |
Collapse
|
32
|
Sánchez SA, Méndez-Barbero N, Santos-Beneit AM, Esteban V, Jiménez-Borreguero LJ, Campanero MR, Redondo JM. Nonlinear optical 3-dimensional method for quantifying atherosclerosis burden. Circ Cardiovasc Imaging 2014; 7:566-9. [PMID: 24847011 DOI: 10.1161/circimaging.114.001753] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Susana A Sánchez
- From the Microscopy and Dynamic Imaging Unit (S.A.S., A.M.S.-B.), Department of Vascular Biology and Inflammation (N.M.-B., V.E., J.M.R.), and Department of Atherothrombosis and Imaging (L.J.J.-B.), Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; Cardiology Department, Hospital de la Princesa, Madrid, Spain (L.J.J.-B.); and Department of Cancer Biology, Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-UAM, Madrid, Spain (M.R.C.).
| | - Nerea Méndez-Barbero
- From the Microscopy and Dynamic Imaging Unit (S.A.S., A.M.S.-B.), Department of Vascular Biology and Inflammation (N.M.-B., V.E., J.M.R.), and Department of Atherothrombosis and Imaging (L.J.J.-B.), Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; Cardiology Department, Hospital de la Princesa, Madrid, Spain (L.J.J.-B.); and Department of Cancer Biology, Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-UAM, Madrid, Spain (M.R.C.)
| | - Antonio M Santos-Beneit
- From the Microscopy and Dynamic Imaging Unit (S.A.S., A.M.S.-B.), Department of Vascular Biology and Inflammation (N.M.-B., V.E., J.M.R.), and Department of Atherothrombosis and Imaging (L.J.J.-B.), Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; Cardiology Department, Hospital de la Princesa, Madrid, Spain (L.J.J.-B.); and Department of Cancer Biology, Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-UAM, Madrid, Spain (M.R.C.)
| | - Vanesa Esteban
- From the Microscopy and Dynamic Imaging Unit (S.A.S., A.M.S.-B.), Department of Vascular Biology and Inflammation (N.M.-B., V.E., J.M.R.), and Department of Atherothrombosis and Imaging (L.J.J.-B.), Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; Cardiology Department, Hospital de la Princesa, Madrid, Spain (L.J.J.-B.); and Department of Cancer Biology, Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-UAM, Madrid, Spain (M.R.C.)
| | - Luis J Jiménez-Borreguero
- From the Microscopy and Dynamic Imaging Unit (S.A.S., A.M.S.-B.), Department of Vascular Biology and Inflammation (N.M.-B., V.E., J.M.R.), and Department of Atherothrombosis and Imaging (L.J.J.-B.), Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; Cardiology Department, Hospital de la Princesa, Madrid, Spain (L.J.J.-B.); and Department of Cancer Biology, Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-UAM, Madrid, Spain (M.R.C.)
| | - Miguel R Campanero
- From the Microscopy and Dynamic Imaging Unit (S.A.S., A.M.S.-B.), Department of Vascular Biology and Inflammation (N.M.-B., V.E., J.M.R.), and Department of Atherothrombosis and Imaging (L.J.J.-B.), Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; Cardiology Department, Hospital de la Princesa, Madrid, Spain (L.J.J.-B.); and Department of Cancer Biology, Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-UAM, Madrid, Spain (M.R.C.).
| | - Juan Miguel Redondo
- From the Microscopy and Dynamic Imaging Unit (S.A.S., A.M.S.-B.), Department of Vascular Biology and Inflammation (N.M.-B., V.E., J.M.R.), and Department of Atherothrombosis and Imaging (L.J.J.-B.), Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; Cardiology Department, Hospital de la Princesa, Madrid, Spain (L.J.J.-B.); and Department of Cancer Biology, Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-UAM, Madrid, Spain (M.R.C.).
| |
Collapse
|
33
|
Minami T. Calcineurin-NFAT activation and DSCR-1 auto-inhibitory loop: how is homoeostasis regulated? J Biochem 2014; 155:217-26. [PMID: 24505143 DOI: 10.1093/jb/mvu006] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Calcineurin-nuclear factor of activated T cells (NFAT) signalling plays a critical role not only in the immune and nervous systems, but also in cardiovascular development and pathological endothelial cell activation during angiogenesis or inflammation. Studies in NFAT-null mice demonstrated that there is high redundancy between functions of the different NFAT family members. Deletion of only one NFAT causes mild phenotypes, but compound deletions of multiple NFAT family members leads to severe abnormalities in multiple organ systems. Genome-wide transcription analysis revealed that many NFAT target genes are related to cell growth and inflammation, whereas the gene most strongly induced by NFAT in endothelial cells is an auto-inhibitory molecule, Down syndrome critical region (DSCR)-1. The NFAT-DSCR-1 signalling axis may vary depending on the cell-type or signal dosage level under the microenvironment. In the endothelium, stable expression of the DSCR-1 short isoform attenuates septic inflammatory shock, tumour growth and tumour metastasis to lung. Moreover, dysfunction of DSCR-1 and the NFAT priming kinase, DYRK1A, prevents NFAT nuclear occupancy. This change in NFAT nuclear localization is responsible for many of the features of Down syndrome. Thus, fine-tuning of the NFAT-DSCR-1 negative feedback loop may enable therapeutic manipulation in vasculopathic diseases.
Collapse
Affiliation(s)
- Takashi Minami
- Div. of Vascular Biology, RCAST, The University of Tokyo, Tokyo 153-8904, Japan
| |
Collapse
|
34
|
Torac E, Gaman L, Atanasiu V. The regulator of calcineurin (RCAN1) an important factor involved in atherosclerosis and cardiovascular diseases development. J Med Life 2014; 7:481-7. [PMID: 25713607 PMCID: PMC4316123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2014] [Accepted: 09/20/2014] [Indexed: 11/23/2022] Open
Abstract
Atherosclerosis, one of the main causes of cardiovascular diseases, is a complex process that involves manifold factors. Besides the vascular lipids accumulation, inflammatory factors could be considered as a proatherogenic factor - RCAN1. RCAN1 is a regulator of calcineurin, both of them being calcium dependent proteins. Recent studies have shown that RCAN1 has an important role in heart valve development. In the same time researchers found that, the atherosclerotic plaques have an up-regulated RCAN1 gene expression. In the near future, it is desirable to elucidate the RCAN1 function and classify it as a possible biochemical marker to diagnose infancy atherosclerosis.
Collapse
Affiliation(s)
- E Torac
- Biochemistry Department, ”Carol Davila” University of Medicine and Pharmacy, Bucharest, Romania
| | - L Gaman
- Biochemistry Department, ”Carol Davila” University of Medicine and Pharmacy, Bucharest, Romania
| | - V Atanasiu
- Biochemistry Department, ”Carol Davila” University of Medicine and Pharmacy, Bucharest, Romania
| |
Collapse
|