1
|
Magavern EF, Kapil V, Saxena M, Gupta A, Caulfield MJ. Use of Genomics to Develop Novel Therapeutics and Personalize Hypertension Therapy. Arterioscler Thromb Vasc Biol 2024; 44:784-793. [PMID: 38385287 DOI: 10.1161/atvbaha.123.319220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
Hypertension is a prevalent public health problem, contributing to >10 million deaths annually. Though multiple therapeutics exist, many patients suffer from treatment-resistant hypertension or try several medications before achieving blood pressure control. Genomic advances offer mechanistic understanding of blood pressure variability, therapeutic targets, therapeutic response, and promise a stratified approach to treatment of primary hypertension. Cyclic guanosine monophosphate augmentation, aldosterone synthase inhibitors, and angiotensinogen blockade with silencing RNA and antisense therapies are among the promising novel approaches. Pharmacogenomic studies have also been done to explore the genetic bases underpinning interindividual variability in response to existing therapeutics. A polygenic approach using risk scores is likely to be the next frontier in stratifying responses to existing therapeutics.
Collapse
Affiliation(s)
- Emma F Magavern
- Centre of Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, United Kingdom
| | - Vikas Kapil
- Centre of Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, United Kingdom
| | - Manish Saxena
- Centre of Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, United Kingdom
| | - Ajay Gupta
- Centre of Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, United Kingdom
| | - Mark J Caulfield
- Centre of Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, United Kingdom
| |
Collapse
|
2
|
Smeir M, Chumala P, Katselis GS, Liu L. Lymphocyte-Specific Protein 1 Regulates Expression and Stability of Endothelial Nitric Oxide Synthase. Biomolecules 2024; 14:111. [PMID: 38254711 PMCID: PMC10813790 DOI: 10.3390/biom14010111] [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: 11/04/2023] [Revised: 12/14/2023] [Accepted: 01/06/2024] [Indexed: 01/24/2024] Open
Abstract
Nitric oxide (NO), synthesized by endothelial nitric oxide synthase (eNOS), plays a critical role in blood pressure regulation. Genome-wide association studies have identified genetic susceptibility loci for hypertension in human lymphocyte-specific protein 1 (LSP1) gene. LSP1 is recognized as modulator of leukocyte extravasation, and endothelial permeability, however, the role of LSP1 in regulation of NO signaling within endothelial cells (ECs) remains unknown. The present study investigated the role of LSP1 in the regulation of eNOS expression and activity utilizing human macrovascular ECs in vitro and LSP1 knockout (KO) mice. In ECs, specific CRISPR-Cas9 genomic editing deleted LSP1 and caused downregulation of eNOS expression. LSP1 gain-of-function through adenovirus-mediated gene transfer was associated with enhanced expression of eNOS. Co-immunoprecipitation and confocal fluorescence microscopy revealed that eNOS and LSP1 formed a protein complex under basal conditions in ECs. Furthermore, LSP1 deficiency in mice promoted significant upregulation and instability of eNOS. Utilizing a mass-spectrometry-based bottom-up proteomics approach, we identified novel truncated forms of eNOS in immunoprecipitates from LSP1 KO aortae. Our experimental data suggest an important role of endothelial LSP1 in regulation of eNOS expression and activity within human ECs and murine vascular tissues.
Collapse
Affiliation(s)
- Musstafa Smeir
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK S7N 5E5, Canada;
| | - Paulos Chumala
- Department of Medicine, Canadian Center for Rural and Agricultural Health, University of Saskatchewan, Saskatoon, SK S7N 2Z4, Canada; (P.C.); (G.S.K.)
| | - George S. Katselis
- Department of Medicine, Canadian Center for Rural and Agricultural Health, University of Saskatchewan, Saskatoon, SK S7N 2Z4, Canada; (P.C.); (G.S.K.)
| | - Lixin Liu
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK S7N 5E5, Canada;
| |
Collapse
|
3
|
Suvorava T, Metry S, Pick S, Kojda G. Alterations in endothelial nitric oxide synthase activity and their relevance to blood pressure. Biochem Pharmacol 2022; 205:115256. [DOI: 10.1016/j.bcp.2022.115256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 09/12/2022] [Accepted: 09/13/2022] [Indexed: 12/15/2022]
|
4
|
Xiao X, Huang S, Chen S, Wang Y, Sun Q, Xu X, Li Y. Mechanisms of cytokine release syndrome and neurotoxicity of CAR T-cell therapy and associated prevention and management strategies. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:367. [PMID: 34794490 PMCID: PMC8600921 DOI: 10.1186/s13046-021-02148-6] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 10/20/2021] [Indexed: 02/08/2023]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has yielded impressive outcomes and transformed treatment algorithms for hematological malignancies. To date, five CAR T-cell products have been approved by the US Food and Drug Administration (FDA). Nevertheless, some significant toxicities pose great challenges to the development of CAR T-cell therapy, most notably cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS). Understanding the mechanisms underlying these toxicities and establishing prevention and treatment strategies are important. In this review, we summarize the mechanisms underlying CRS and ICANS and provide potential treatment and prevention strategies.
Collapse
Affiliation(s)
- Xinyi Xiao
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, People's Republic of China
| | - Shengkang Huang
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, People's Republic of China
| | - Sifei Chen
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, People's Republic of China
| | - Yazhuo Wang
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, People's Republic of China.,Medical College of Rehabilitation, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Qihang Sun
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510623, People's Republic of China
| | - Xinjie Xu
- 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, People's Republic of China.
| | - Yuhua Li
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, People's Republic of China. .,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, Guangdong, 510005, People's Republic of China.
| |
Collapse
|
5
|
Mansouri V, Yazdanpanah N, Rezaei N. The immunologic aspects of cytokine release syndrome and graft versus host disease following CAR T cell therapy. Int Rev Immunol 2021; 41:649-668. [PMID: 34607523 DOI: 10.1080/08830185.2021.1984449] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Chimeric antigen receptor (CAR) T cells are the pioneers of cancer immunotherapy, which to this date have several FDA-approved products. They have been substantially improved since their first introduction in 1993 and have shown promising results regardless of their inevitable side effects. Cytokine release syndrome (CRS), the most common toxicity after CAR T cell treatment, is affiliated to a systemic inflammation through surge of cytokines, mainly IL-6, IL-1, and INF-γ. Furthermore, difference between histocompatibility antigens activates the graft versus host disease (GvHD) effect of the allogenic CAR T cells against the host cells. Immunological reactions induced by CAR T cells in the form of CRS or GvHD is necessary for fostering good responses, while excess reactions can potentially threaten patient life. In this review, we first describe the history, applications, and structure of CAR T cells, followed by a comprehensive review of CRS regarding its definition, management, and immunological aspects. Finally, we discuss about the clinical aspects of CRS and GvHD after CAR T cell therapy and how to harness anti-tumoral effects, while mitigating the adverse effects.
Collapse
Affiliation(s)
- Vahid Mansouri
- Gene Therapy Research Center, Digestive Diseases Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Niloufar Yazdanpanah
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Shi J, Liu S, Guo Y, Liu S, Xu J, Pan L, Hu Y, Liu Y, Cheng Y. Association between eNOS rs1799983 polymorphism and hypertension: a meta-analysis involving 14,185 cases and 13,407 controls. BMC Cardiovasc Disord 2021; 21:385. [PMID: 34372765 PMCID: PMC8351409 DOI: 10.1186/s12872-021-02192-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 07/30/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Essential hypertension is a complex disease determined by the interaction of genetic and environmental factors, eNOS is considered to be one of the susceptible genes for hypertension. Our study aimed to evaluate the association between eNOS rs1799983 polymorphism and hypertension, and to provide evidence for the etiology of hypertension. METHODS Case-control studies of eNOS rs1799983 polymorphism and hypertension were included by searching PubMed, Embase, Web of Science, Medline, Scopus, WanFang datebase, Vip datebase, and CNKI database according to PRISMA guideline. Eligible data were extracted and pooled, and were analyzed using R software based on five different genetic models. RESULTS A total of 60 eligible articles involving 14,185 cases and 13,407 controls were finally selected. We found significant association between eNOS rs1799983 polymorphism and hypertension under any genetic model (T vs G: OR = 1.44, 95% CI 1.26-1.63; GT vs GG: OR 1.34, 95% CI 1.18-1.52; TT vs GG: OR 1.80, 95% CI 1.41-2.31; GT + TT vs GG: OR 1.42, 95% CI 1.25-1.63; TT vs GG + GT: OR 1.68, 95% CI 1.35-2.08; GT vs GG + TT: OR 1.24, 95% CI 1.11-1.40). CONCLUSIONS We found that eNOS rs1799983 polymorphism is associated with the increased risk of hypertension under any genetic model. Moreover, investigations of gene-gene and gene-environment interactions are needed to give more insight into the association between eNOS rs1799983 polymorphism and hypertension.
Collapse
Affiliation(s)
- Jikang Shi
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, 130021, China
| | - Siyu Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, 130021, China
| | - Yanbo Guo
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, 130021, China
| | - Sainan Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, 130021, China
| | - Jiayi Xu
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, 130021, China
| | - Lingfeng Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, 130021, China
| | - Yueyang Hu
- Department of Children and Adolescence Health, School of Public Health, Jilin University, Changchun, 130021, Jilin, China
| | - Yawen Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, 130021, China.
| | - Yi Cheng
- The Cardiovascular Center, The First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
7
|
Cai Z, Yuan S, Zhong Y, Deng L, Li J, Tan X, Feng J. Amelioration of Endothelial Dysfunction in Diabetes: Role of Takeda G Protein-Coupled Receptor 5. Front Pharmacol 2021; 12:637051. [PMID: 33995040 PMCID: PMC8113688 DOI: 10.3389/fphar.2021.637051] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 02/22/2021] [Indexed: 12/25/2022] Open
Abstract
Diabetes mellitus (DM) eventually leads to chronic vascular complications, resulting in cardiovascular diseases. DM-associated endothelial dysfunction (ED) plays an important role in the development of chronic vascular complications. Low endothelial nitric oxide synthase (eNOS) activity, inflammation, and oxidative stress all contribute to ED. The G protein-coupled receptor Takeda G protein-coupled receptor 5 (TGR5) is a membrane receptor for bile acids that plays an important role in the regulation of glucose metabolism. Recent studies have shown that TGR5 is involved in the regulation of various mediators of ED, which suggests that TGR5 may represent a target for the treatment of DM-associated ED. In this review, we summarize the principal mechanisms of DM-associated ED, then propose TGR5 as a novel therapeutic target on the basis of its mechanistic involvement, and suggest potential directions for future research.
Collapse
Affiliation(s)
- Zhengyao Cai
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Suxin Yuan
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Yi Zhong
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Li Deng
- Department of Rheumatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jiafu Li
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Xiaoqiu Tan
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Jian Feng
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| |
Collapse
|
8
|
Wu Y, Ding Y, Ramprasath T, Zou MH. Oxidative Stress, GTPCH1, and Endothelial Nitric Oxide Synthase Uncoupling in Hypertension. Antioxid Redox Signal 2021; 34:750-764. [PMID: 32363908 PMCID: PMC7910417 DOI: 10.1089/ars.2020.8112] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 04/24/2020] [Accepted: 04/24/2020] [Indexed: 02/07/2023]
Abstract
Significance: Hypertension has major health consequences, which is associated with endothelial dysfunction. Endothelial nitric oxide synthase (eNOS)-produced nitric oxide (NO) signaling in the vasculature plays an important role in maintaining vascular homeostasis. Considering the importance of NO system, this review aims to provide a brief overview of the biochemistry of members of NO signaling, including GTPCH1 [guanosine 5'-triphosphate (GTP) cyclohydrolase 1], tetrahydrobiopterin (BH4), and eNOS. Recent Advances: Being NO signaling activators and regulators of eNOS signaling, BH4 treatment is getting widespread attention either as potential therapeutic agents or as preventive agents. Recent clinical trials also support that BH4 treatment could be considered a promising therapeutic in hypertension. Critical Issues: Under conditions of BH4 depletion, eNOS-generated superoxides trigger pathological events. Abnormalities in NO availability and BH4 deficiency lead to disturbed redox regulation causing pathological events. This disturbed signaling influences the development of systemic hypertension as well as pulmonary hypertension. Future Directions: Considering the importance of BH4 and NO to improve the translational significance, it is essential to continue research on this field to manipulate BH4 to increase the efficacy for treating hypertension. Thus, this review also examines the current state of knowledge on the effects of eNOS activators on preclinical models and humans to utilize this information for potential therapy.
Collapse
Affiliation(s)
- Yin Wu
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia, USA
| | - Ye Ding
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia, USA
| | - Tharmarajan Ramprasath
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia, USA
| | - Ming-Hui Zou
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia, USA
| |
Collapse
|
9
|
Cotta Filho CK, Oliveira-Paula GH, Rondon Pereira VC, Lacchini R. Clinically relevant endothelial nitric oxide synthase polymorphisms and their impact on drug response. Expert Opin Drug Metab Toxicol 2020; 16:927-951. [DOI: 10.1080/17425255.2020.1804857] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
| | | | | | - Riccardo Lacchini
- Department of Psychiatric Nursing and Human Sciences, University of Sao Paulo, Ribeirao Preto, Brazil
| |
Collapse
|
10
|
Hao Z, Li R, Meng L, Han Z, Hong Z. Macrophage, the potential key mediator in CAR-T related CRS. Exp Hematol Oncol 2020; 9:15. [PMID: 32665874 PMCID: PMC7349474 DOI: 10.1186/s40164-020-00171-5] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 06/20/2020] [Indexed: 01/05/2023] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy is a new frontier in cancer therapy. The toxicity of cytokine release syndrome (CRS) has become one of the major challenges that limits the wider use of CAR T cells to fight cancer. Exploration of CRS pathogenesis and treatment is becoming the main focus of ongoing studies. Myeloid-derived macrophages were found to play a critical role in CRS pathogenesis, and these cells mediate the major production of core cytokines, including IL-6, IL-1 and interferon (IFN)-γ. Colocalization of macrophages and CAR T cells was also identified as necessary for inducing CRS, and CD40L-CD40 signaling might be the key cell–cell interaction in the tumor microenvironment. Macrophages might also take part in endocrine and self-amplified catecholamine loops that can directly activate cytokine production and release by macrophages during CRS. In addition to tocilizumab and corticosteroids, several novel CRS therapies targeting macrophage-centered pathways have shown much potential, including GM-CSF blockade and administration of atrial natriuretic peptide (ANP) and α-methyltyrosine (metyrosine, MTR). In the present review, we summarized the role of macrophages in CRS and new developments in therapeutic strategies for CRS-associated toxicities.
Collapse
Affiliation(s)
- Zhaonian Hao
- The Second Clinical School Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 Hubei China
| | - Ruyuan Li
- The Second Clinical School Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 Hubei China
| | - Li Meng
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jie Fang Avenue, Hankou, Wuhan, 430030 Hubei China
| | - Zhiqiang Han
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jie Fang Avenue, Hankou, Wuhan, 430030 Hubei China
| | - Zhenya Hong
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jie Fang Avenue, Hankou, Wuhan, 430030 Hubei China
| |
Collapse
|
11
|
Wu H, Lu L, Chen J, Zhang C, Liu W, Zhuang S. Inhibited Nitric Oxide Production of Human Endothelial Nitric Oxide Synthase by Nitrated and Oxygenated Polycyclic Aromatic Hydrocarbons. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2020; 54:2922-2930. [PMID: 32022550 DOI: 10.1021/acs.est.9b07163] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Nitrated and oxygenated polycyclic aromatic hydrocarbons (NPAHs and OPAHs) from the direct atmospheric emission or the degradation of parent PAHs are increasingly recognized because of their potential health risks. Herein, we investigated the effects of four NPAHs/OPAHs (1-NNAP, 9-NANT, 9,10-AQ, and 9-FLU) and their parent PAHs (NAP, ANT, and FLU) on endothelium function with regard to endothelial nitric oxide synthase (eNOS) and endothelium-derived nitric oxide (NO) production in human umbilical vein endothelial cells. The eNOS enzymatic activity and NO production were promoted by NAP, ANT, and FLU; however, eNOS activity was dropped by 52.8, 52.1, 52.5, and 44.5%, and NO production was decreased by 31.1, 50.3, 65.0, and 35.0% after 24 h exposure to 0.01 μM 1-NNAP, 9-NANT, 9,10-AQ, and 9-FLU, respectively. The mRNA expression of eNOS and protein expression of phosphorylated eNOS (Ser1177) were increased by three PAHs but decreased by four NPAHs/OPAHs. The 100 ns molecular dynamics simulations reveal the conformational alteration in the key propionate of heme upon the binding of NPAHs/OPAHs. Our findings provide the first in silico and in vitro evidence for the potential endothelial dysfunction of nitrated and oxygenated PAHs. The health risk implications of NPAHs/OPAHs and corresponding parent PAHs warrant further research.
Collapse
Affiliation(s)
- Hao Wu
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
| | - Liping Lu
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jiayan Chen
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
| | - Chunlong Zhang
- Department of Environmental Sciences, University of Houston-Clear Lake, 2700 Bay Area Blvd., Houston 77058, Texas, United States
| | - Weiping Liu
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
- Research Center for Air Pollution and Health, Zhejiang University, Hangzhou 310058, China
| | - Shulin Zhuang
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
12
|
Dick BP, McMahan R, Knowles T, Becker L, Gharib SA, Vaisar T, Wietecha T, O'Brien KD, Bornfeldt KE, Chait A, Kim F. Hematopoietic Cell-Expressed Endothelial Nitric Oxide Protects the Liver From Insulin Resistance. Arterioscler Thromb Vasc Biol 2020; 40:670-681. [PMID: 31996027 DOI: 10.1161/atvbaha.119.313648] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
OBJECTIVE Mice genetically deficient in endothelial nitric oxide synthase (Nos3-/-) have fasting hyperinsulinemia and hepatic insulin resistance, indicating the importance of Nos3 (nitric oxide synthase) in maintaining metabolic homeostasis. Although the current paradigm holds that these metabolic effects are derived specifically from the expression of Nos3 in the endothelium, it has been established that bone marrow-derived cells also express Nos3. The aim of this study was to investigate whether bone marrow-derived cell Nos3 is important in maintaining metabolic homeostasis. Approach and Results: To test the hypothesis that bone marrow-derived cell Nos3 contributes to metabolic homeostasis, we generated chimeric male mice deficient or competent for Nos3 expression in circulating blood cells. These mice were placed on a low-fat diet for 5 weeks, a time period which is known to induce hepatic insulin resistance in global Nos3-deficient mice but not in wild-type C57Bl/6 mice. Surprisingly, we found that the absence of Nos3 in the bone marrow-derived component is associated with hepatic insulin resistance and that restoration of Nos3 in the bone marrow-derived component in global Nos3-deficient mice is sufficient to restore hepatic insulin sensitivity. Furthermore, we found that overexpression of Nos3 in bone marrow-derived component in wild-type mice attenuates the development of hepatic insulin resistance during high-fat feeding. Finally, compared with wild-type macrophages, the loss of macrophage Nos3 is associated with increased inflammatory responses to lipopolysaccharides and reduced anti-inflammatory responses to IL-4, a macrophage phenotype associated with the development of hepatic and systemic insulin resistance. CONCLUSIONS These results would suggest that the metabolic and hepatic consequences of high-fat feeding are mediated by loss of Nos3/nitric oxide actions in bone marrow-derived cells, not in endothelial cells.
Collapse
Affiliation(s)
- Brian P Dick
- From the Department of Medicine, University of Washington, Seattle (B.P.D., R.M., T.K., S.A.G., T.V., T.W., K.D.O., K.E.B., A.C., F.K.)
| | - Ryan McMahan
- From the Department of Medicine, University of Washington, Seattle (B.P.D., R.M., T.K., S.A.G., T.V., T.W., K.D.O., K.E.B., A.C., F.K.)
| | - Taft Knowles
- From the Department of Medicine, University of Washington, Seattle (B.P.D., R.M., T.K., S.A.G., T.V., T.W., K.D.O., K.E.B., A.C., F.K.)
| | - Lev Becker
- Ben May Department for Cancer Research, University of Chicago, IL (L.B.)
| | - Sina A Gharib
- From the Department of Medicine, University of Washington, Seattle (B.P.D., R.M., T.K., S.A.G., T.V., T.W., K.D.O., K.E.B., A.C., F.K.)
| | - Tomas Vaisar
- From the Department of Medicine, University of Washington, Seattle (B.P.D., R.M., T.K., S.A.G., T.V., T.W., K.D.O., K.E.B., A.C., F.K.)
| | - Tomasz Wietecha
- From the Department of Medicine, University of Washington, Seattle (B.P.D., R.M., T.K., S.A.G., T.V., T.W., K.D.O., K.E.B., A.C., F.K.)
| | - Kevin D O'Brien
- From the Department of Medicine, University of Washington, Seattle (B.P.D., R.M., T.K., S.A.G., T.V., T.W., K.D.O., K.E.B., A.C., F.K.)
| | - Karin E Bornfeldt
- From the Department of Medicine, University of Washington, Seattle (B.P.D., R.M., T.K., S.A.G., T.V., T.W., K.D.O., K.E.B., A.C., F.K.)
| | - Alan Chait
- From the Department of Medicine, University of Washington, Seattle (B.P.D., R.M., T.K., S.A.G., T.V., T.W., K.D.O., K.E.B., A.C., F.K.)
| | - Francis Kim
- From the Department of Medicine, University of Washington, Seattle (B.P.D., R.M., T.K., S.A.G., T.V., T.W., K.D.O., K.E.B., A.C., F.K.)
| |
Collapse
|
13
|
Wood KC, Durgin BG, Schmidt HM, Hahn SA, Baust JJ, Bachman T, Vitturi DA, Ghosh S, Ofori-Acquah SF, Mora AL, Gladwin MT, Straub AC. Smooth muscle cytochrome b5 reductase 3 deficiency accelerates pulmonary hypertension development in sickle cell mice. Blood Adv 2019; 3:4104-4116. [PMID: 31821458 PMCID: PMC6963246 DOI: 10.1182/bloodadvances.2019000621] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 10/29/2019] [Indexed: 01/26/2023] Open
Abstract
Pulmonary and systemic vasculopathies are significant risk factors for early morbidity and death in patients with sickle cell disease (SCD). An underlying mechanism of SCD vasculopathy is vascular smooth muscle (VSM) nitric oxide (NO) resistance, which is mediated by NO scavenging reactions with plasma hemoglobin (Hb) and reactive oxygen species that can oxidize soluble guanylyl cyclase (sGC), the NO receptor. Prior studies show that cytochrome b5 reductase 3 (CYB5R3), known as methemoglobin reductase in erythrocytes, functions in VSM as an sGC heme iron reductase critical for reducing and sensitizing sGC to NO and generating cyclic guanosine monophosphate for vasodilation. Therefore, we hypothesized that VSM CYB5R3 deficiency accelerates development of pulmonary hypertension (PH) in SCD. Bone marrow transplant was used to create SCD chimeric mice with background smooth muscle cell (SMC)-specific tamoxifen-inducible Cyb5r3 knockout (SMC R3 KO) and wild-type (WT) control. Three weeks after completing tamoxifen treatment, we observed 60% knockdown of pulmonary arterial SMC CYB5R3, 5 to 6 mm Hg elevated right-ventricular (RV) maximum systolic pressure (RVmaxSP) and biventricular hypertrophy in SS chimeras with SMC R3 KO (SS/R3KD) relative to WT (SS/R3WT). RV contractility, heart rate, hematological parameters, and cell-free Hb were similar between groups. When identically generated SS/R3 chimeras were studied 12 weeks after completing tamoxifen treatment, RVmaxSP in SS/R3KD had not increased further, but RV hypertrophy relative to SS/R3WT persisted. These are the first studies to establish involvement of SMC CYB5R3 in SCD-associated development of PH, which can exist in mice by 5 weeks of SMC CYB5R3 protein deficiency.
Collapse
Affiliation(s)
- Katherine C Wood
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, Department of Medicine
| | - Brittany G Durgin
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, Department of Medicine
| | - Heidi M Schmidt
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, Department of Medicine
- Department of Pharmacology and Chemical Biology
| | - Scott A Hahn
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, Department of Medicine
| | - Jeffrey J Baust
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, Department of Medicine
| | - Tim Bachman
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, Department of Medicine
| | - Dario A Vitturi
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, Department of Medicine
- Department of Pharmacology and Chemical Biology
| | - Samit Ghosh
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, Department of Medicine
| | - Solomon F Ofori-Acquah
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, Department of Medicine
- Division of Hematology and Oncology, Department of Medicine, and
| | - Ana L Mora
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, Department of Medicine
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Mark T Gladwin
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, Department of Medicine
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Adam C Straub
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, Department of Medicine
- Department of Pharmacology and Chemical Biology
| |
Collapse
|
14
|
Choi B, Shin MK, Kim EY, Park JE, Lee H, Kim SW, Song JK, Chang EJ. Elevated Neuropeptide Y in Endothelial Dysfunction Promotes Macrophage Infiltration and Smooth Muscle Foam Cell Formation. Front Immunol 2019; 10:1701. [PMID: 31379881 PMCID: PMC6657015 DOI: 10.3389/fimmu.2019.01701] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 07/08/2019] [Indexed: 12/19/2022] Open
Abstract
Endothelial dysfunction has been linked to vascular inflammation and foam cell formation but the underlying mechanisms still remain unclear. We sought to define the factors inducing inflammation and smooth muscle foam cell formation under endothelial dysfunction using endothelial nitric oxide synthase (eNOS)-deficient mice. Vascular smooth muscle cells (VSMCs) from eNOS-deficient mice displayed increased expression of macrophage-related genes and elevated lipid uptake. Neuropeptide Y (NPY) was upregulated in the aorta from the eNOS-deficient mice and promoted macrophage chemotaxis toward VSMCs while enhancing the activity of matrix metalloproteinase-3. Notably, NPY induced lipid uptake in VSMCs, facilitating smooth muscle foam cell formation, in association with enhanced expression of genes related to modified low-density lipoprotein uptake and macrophages. NPY was augmented by inflammatory pentraxin 3 (PTX3) in VSMCs. PTX3 enhanced macrophage migratory capacity through the NPY/neuropeptide Y receptor axis and this effect was attenuated by pharmacological inhibition with a receptor-specific antagonist. These observations suggest that endothelial dysfunction leads to the elevation of NPY that amplifies vascular inflammation by increasing inflammatory cell chemotaxis and triggers smooth muscle foam cell formation.
Collapse
Affiliation(s)
- Bongkun Choi
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea.,Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Min-Kyung Shin
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea.,Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Eun-Young Kim
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea.,Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Ji-Eun Park
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea.,Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Halim Lee
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Seong Who Kim
- Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea.,Department of Biochemistry and Molecular Biology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Jae-Kwan Song
- Division of Cardiology, Asan Medical Center, Research Institute for Valvular Heart Disease University of Ulsan College of Medicine, Seoul, South Korea
| | - Eun-Ju Chang
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea.,Stem Cell Immunomodulation Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea.,Department of Biochemistry and Molecular Biology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| |
Collapse
|
15
|
Luo Y, Wang Y, Luo W. C allele of -786 T>C polymorphism in the promoter region of endothelial nitric oxide synthase is responsible for endothelial dysfunction in the patients with rheumatoid arthritis. J Cell Biochem 2019; 121:363-370. [PMID: 31209933 DOI: 10.1002/jcb.29184] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 02/26/2019] [Accepted: 02/28/2019] [Indexed: 11/10/2022]
Abstract
BACKGROUND This study aimed to explore the roles of endothelial nitric oxide synthase (eNOS) in the control of metastasis of infection with endothelial dysfunction, as well as the roles of -786T>C polymorphism in eNOS promoter in the control of metastasis of endothelial function. METHOD In-silicon analysis and luciferase assay were used to identify the location of -786>C on the promoter of eNOS. Subsequently, real-time PCR and Western-blot were used to determine the expression level of eNOS. Ultrasound examination was used to detect baseline brachial artery diameter and flow-mediated dilation of patients in different treat groups. RESULTS -786T>C was located on the promoter of eNOS, and the luciferase activity of cells transfected with -786-C allele was much higher than empty vector, while even higher subsequent to transfection of -786-T allele. In addition, the result of ultrasound examination showed that the baseline brachial artery diameter was comparable between patients genotyped as TT, TC and CC, while the flow-mediated dilation of patients genotyped as TC was much higher compared with CC group, and the flow-mediated dilation of patients genotyped as TT even higher than TC group. We found eNOS messenger RNA and protein with TT genotype was significantly higher compared with other genotypes. And the production of NO was remarkably higher in TT groups compared with TC and CC, while the production of NO in TC and CC groups were similar. CONCLUSION These findings indicated that down-expression of -786T>C located on the promoter of eNOS is associated with an increased risk of endothelial dysfunction.
Collapse
Affiliation(s)
- Yanli Luo
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yang Wang
- Department of Internal Medicine, Yixing People's Hospital, Jiangsu, China
| | - Wanjun Luo
- Department of Cardiovascualr Surgery, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
16
|
Yokoyama M, Shimizu I, Nagasawa A, Yoshida Y, Katsuumi G, Wakasugi T, Hayashi Y, Ikegami R, Suda M, Ota Y, Okada S, Fruttiger M, Kobayashi Y, Tsuchida M, Kubota Y, Minamino T. p53 plays a crucial role in endothelial dysfunction associated with hyperglycemia and ischemia. J Mol Cell Cardiol 2019; 129:105-117. [PMID: 30790589 DOI: 10.1016/j.yjmcc.2019.02.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 02/12/2019] [Accepted: 02/16/2019] [Indexed: 12/23/2022]
Abstract
p53 is a guardian of the genome that protects against carcinogenesis. There is accumulating evidence that p53 is activated with aging. Such activation has been reported to contribute to various age-associated pathologies, but its role in vascular dysfunction is largely unknown. The aim of this study was to investigate whether activation of endothelial p53 has a pathological effect in relation to endothelial function. We established endothelial p53 loss-of-function and gain-of-function models by breeding endothelial-cell specific Cre mice with floxed Trp53 or floxed Mdm2/Mdm4 mice, respectively. Then we induced diabetes by injection of streptozotocin. In the diabetic state, endothelial p53 expression was markedly up-regulated and endothelium-dependent vasodilatation was significantly impaired. Impairment of vasodilatation was significantly ameliorated in endothelial p53 knockout (EC-p53 KO) mice, and deletion of endothelial p53 also significantly enhanced the induction of angiogenesis by ischemia. Conversely, activation of endothelial p53 by deleting Mdm2/Mdm4 reduced both endothelium-dependent vasodilatation and ischemia-induced angiogenesis. Introduction of p53 into human endothelial cells up-regulated the expression of phosphatase and tensin homolog (PTEN), thereby reducing phospho-eNOS levels. Consistent with these results, the beneficial impact of endothelial p53 deletion on endothelial function was attenuated in EC-p53 KO mice with an eNOS-deficient background. These results show that endothelial p53 negatively regulates endothelium-dependent vasodilatation and ischemia-induced angiogenesis, suggesting that inhibition of endothelial p53 could be a novel therapeutic target in patients with metabolic disorders.
Collapse
Affiliation(s)
- Masataka Yokoyama
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Ippei Shimizu
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan; Division of Molecular Aging and Cell Biology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Ayako Nagasawa
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan; Department of Thoracic and Cardiovascular Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Yohko Yoshida
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan; Division of Molecular Aging and Cell Biology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Goro Katsuumi
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Takayuki Wakasugi
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Yuka Hayashi
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Ryutaro Ikegami
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Masayoshi Suda
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Yusuke Ota
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Sho Okada
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Marcus Fruttiger
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| | - Yoshio Kobayashi
- Department of Cardiovascular Medicine, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Masanori Tsuchida
- Department of Thoracic and Cardiovascular Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Yoshiaki Kubota
- Department of Anatomy, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan.
| |
Collapse
|
17
|
Durgin BG, Straub AC. Redox control of vascular smooth muscle cell function and plasticity. J Transl Med 2018; 98:1254-1262. [PMID: 29463879 PMCID: PMC6102093 DOI: 10.1038/s41374-018-0032-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 12/15/2017] [Accepted: 12/18/2017] [Indexed: 02/07/2023] Open
Abstract
Vascular smooth muscle cells (SMC) play a major role in vascular diseases, such as atherosclerosis and hypertension. It has long been established in vitro that contractile SMC can phenotypically switch to function as proliferative and/or migratory cells in response to stimulation by oxidative stress, growth factors, and inflammatory cytokines. Reactive oxygen species (ROS) are oxidative stressors implicated in driving vascular diseases, shifting cell bioenergetics, and increasing SMC proliferation, migration, and apoptosis. In this review, we summarize our current knowledge of how disruptions to redox balance can functionally change SMC and how this may influence vascular disease pathogenesis. Specifically, we focus on our current understanding of the role of vascular nicotinamide adenine dinucleotide phosphate (NADPH) oxidases (NOX) 1, 4, and 5 in SMC function. We also review the evidence implicating mitochondrial fission in SMC phenotypic transitions and mitochondrial fusion in maintenance of SMC homeostasis. Finally, we discuss the importance of the redox regulation of the soluble guanylate cyclase (sGC)-cyclic guanosine monophosphate (cGMP)-protein kinase G (PKG) pathway as a potential oxidative and therapeutic target for regulating SMC function.
Collapse
Affiliation(s)
- Brittany G Durgin
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Adam C Straub
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
18
|
CAR T cell-induced cytokine release syndrome is mediated by macrophages and abated by IL-1 blockade. Nat Med 2018; 24:731-738. [PMID: 29808005 DOI: 10.1038/s41591-018-0041-7] [Citation(s) in RCA: 945] [Impact Index Per Article: 135.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 03/14/2018] [Indexed: 01/06/2023]
Abstract
Chimeric antigen receptor (CAR) therapy targeting CD19 is an effective treatment for refractory B cell malignancies, especially acute lymphoblastic leukemia (ALL) 1 . Although a majority of patients will achieve a complete response following a single infusion of CD19-targeted CAR-modified T cells (CD19 CAR T cells)2-4, the broad applicability of this treatment is hampered by severe cytokine release syndrome (CRS), which is characterized by fever, hypotension and respiratory insufficiency associated with elevated serum cytokines, including interleukin-6 (IL-6)2,5. CRS usually occurs within days of T cell infusion at the peak of CAR T cell expansion. In ALL, it is most frequent and more severe in patients with high tumor burden2-4. CRS may respond to IL-6 receptor blockade but can require further treatment with high dose corticosteroids to curb potentially lethal severity2-9. Improved therapeutic and preventive treatments require a better understanding of CRS physiopathology, which has so far remained elusive. Here we report a murine model of CRS that develops within 2-3 d of CAR T cell infusion and that is potentially lethal and responsive to IL-6 receptor blockade. We show that its severity is mediated not by CAR T cell-derived cytokines, but by IL-6, IL-1 and nitric oxide (NO) produced by recipient macrophages, which enables new therapeutic interventions.
Collapse
|
19
|
Interaction between nitric oxide and renal α1-adrenoreceptors mediated vasoconstriction in rats with left ventricular hypertrophyin Wistar Kyoto rats. PLoS One 2018; 13:e0189386. [PMID: 29447158 PMCID: PMC5844246 DOI: 10.1371/journal.pone.0189386] [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: 07/03/2017] [Accepted: 11/26/2017] [Indexed: 01/19/2023] Open
Abstract
Left ventricular hypertrophy (LVH) is associated with decreased responsiveness of
renal α1-adrenoreceptors subtypes to adrenergic agonists. Nitric
oxide donors are known to have antihypertrophic effects however their impact on
responsiveness of renal α1-adrenoreceptors subtypes is unknown. This
study investigated the impact of nitric oxide (NO) and its potential interaction
with the responsiveness of renal α1-adrenoreceptors subtypes to
adrenergic stimulation in rats with left ventricular hypertrophy (LVH). This
study also explored the impact of NO donor on CSE expression in normal and LVH
kidney. LVH was induced using isoprenaline and caffeine in drinking water for 2
weeks while NO donor (L-arginine, 1.25g/Lin drinking water) was given for 5
weeks. Intrarenal noradrenaline, phenylephrine and methoxamine responses were
determined in the absence and presence of selective α1-adrenoceptor
antagonists, 5- methylurapidil (5-MeU), chloroethylclonidine (CeC) and BMY 7378.
Renal cortical endothelial nitric oxide synthase mRNA was upregulated 7 fold
while that of cystathione γ lyase was unaltered in the NO treated LVH rats
(LVH-NO) group compared to LVH group. The responsiveness of renal
α1A, α1B and α1D-adrenoceptors in the low dose
and high dose phases of 5-MeU, CEC and BMY7378 to adrenergic agonists was
increased along with cGMP in the kidney of LVH-NO group. These findings suggest
that exogenous NO precursor up-regulated the renal eNOS/NO/cGMP pathway in LVH
rats and resulted in augmented α1A, α1B and α1D
adrenoreceptors responsiveness to the adrenergic agonists. There is a positive
interaction between H2S and NO production in normal animals but this
interaction appears absent in LVH animals.
Collapse
|
20
|
Azevedo AMM, Brites-Anselmi G, Pinheiro LC, de Almeida Belo V, Coeli-Lacchini FB, Molina CAF, de Andrade MF, Tucci S, Hirsch E, Tanus-Santos JE, Lacchini R. Relationship between asymmetric dimethylarginine, nitrite and genetic polymorphisms: Impact on erectile dysfunction therapy. Nitric Oxide 2017; 71:44-51. [DOI: 10.1016/j.niox.2017.10.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 10/11/2017] [Accepted: 10/22/2017] [Indexed: 01/24/2023]
|
21
|
Sofronova SI, Gaynullina DK, Shvetsova AA, Borzykh AA, Selivanova EK, Kostyunina DS, Sharova AP, Martyanov AA, Tarasova OS. Antenatal/early postnatal hypothyroidism alters arterial tone regulation in 2-week-old rats. J Endocrinol 2017; 235:137-151. [PMID: 28794003 DOI: 10.1530/joe-17-0225] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 08/09/2017] [Indexed: 01/05/2023]
Abstract
The mechanisms of vascular alterations resulting from early thyroid hormones deficiency are poorly understood. We tested the hypothesis that antenatal/early postnatal hypothyroidism would alter the activity of endothelial NO pathway and Rho-kinase pathway, which are specific for developing vasculature. Dams were treated with propylthiouracil (PTU, 7 ppm) in drinking water during gestation and 2 weeks after delivery, and their progeny had normal body weight but markedly reduced blood levels of thyroid hormones (ELISA). Small arteries from 2-week-old male pups were studied using wire myography, qPCR and Western blotting. Mesenteric arteries of PTU pups, compared to controls, demonstrated smaller maximum response to α1-adrenergic agonist methoxamine and reduced mRNA contents of smooth muscle differentiation markers α-actin and SERCA2A. Inhibition of basal NO synthesis by l-NNA led to tonic contraction of mesenteric arteries and augmented their contractile responses to methoxamine; both l-NNA effects were impaired in PTU pups. PTU pups demonstrated lower blood level of NO metabolites compared to control group (Griess reaction). Rho-kinase inhibitor Y27632 strongly reduced mesenteric arteries responses to methoxamine in PTU pups, that was accompanied by elevated Rho-kinase content in their arteries in comparison to control ones. Unlike mesenteric, saphenous arteries of PTU pups, compared to controls, had no changes in α-actin and SERCA2A contents and in responses to l-NNA and Y27632. In conclusion, thyroid hormones deficiency suppresses the anticontractile effect of NO and potentiates the procontractile Rho-kinase effects in mesenteric arteries of 2-week-old pups. Such alterations disturb perinatal cardiovascular homeostasis and might lead to cardiovascular pathologies in adulthood.
Collapse
Affiliation(s)
- Svetlana I Sofronova
- Institute for Biomedical ProblemsRussian Academy of Sciences, Moscow, Russia
- Faculty of BiologyM.V. Lomonosov Moscow State University, Moscow, Russia
| | - Dina K Gaynullina
- Institute for Biomedical ProblemsRussian Academy of Sciences, Moscow, Russia
- Faculty of BiologyM.V. Lomonosov Moscow State University, Moscow, Russia
- Department of PhysiologyRussian National Research Medical University, Moscow, Russia
| | - Anastasia A Shvetsova
- Institute for Biomedical ProblemsRussian Academy of Sciences, Moscow, Russia
- Faculty of BiologyM.V. Lomonosov Moscow State University, Moscow, Russia
| | - Anna A Borzykh
- Institute for Biomedical ProblemsRussian Academy of Sciences, Moscow, Russia
| | - Ekaterina K Selivanova
- Institute for Biomedical ProblemsRussian Academy of Sciences, Moscow, Russia
- Faculty of BiologyM.V. Lomonosov Moscow State University, Moscow, Russia
| | - Daria S Kostyunina
- Institute for Biomedical ProblemsRussian Academy of Sciences, Moscow, Russia
- Faculty of BiologyM.V. Lomonosov Moscow State University, Moscow, Russia
| | - Anna P Sharova
- Institute for Biomedical ProblemsRussian Academy of Sciences, Moscow, Russia
| | - Andrey A Martyanov
- Institute for Biomedical ProblemsRussian Academy of Sciences, Moscow, Russia
- Faculty of BiologyM.V. Lomonosov Moscow State University, Moscow, Russia
| | - Olga S Tarasova
- Institute for Biomedical ProblemsRussian Academy of Sciences, Moscow, Russia
- Faculty of BiologyM.V. Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
22
|
Selective impairment of blood pressure reduction by endothelial nitric oxide synthase dimer destabilization in mice. J Hypertens 2017; 35:76-88. [PMID: 27861245 DOI: 10.1097/hjh.0000000000001127] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
OBJECTIVES Endothelial dysfunction and oxidative stress are associated with hypertension but whether endothelial superoxide may play a role in the early development of essential hypertension remains uncertain. We investigated whether endothelial nitric oxide synthase (eNOS)-derived endothelial oxidative stress is involved in the regulation of SBP. METHODS Wild-type eNOS [mice with endothelium-specific overexpression of bovine endothelial NO-synthase (eNOS-Tg)] or a novel dimer-destabilized eNOS-mutant harboring a partially disrupted zinc-finger [mice with endothelium-specific overexpression of destabilized bovine eNOS destabilized by replacement of Cys 101 to Ala (C101A-eNOS-Tg)] was introduced in C57BL/6 in an endothelial-specific manner. Mice were monitored for aortic endothelium-dependent relaxation, SBP, levels of superoxide and several posttranslational modifications indicating activity and/or increased vascular oxidative stress. Some groups of mice underwent voluntary exercise training for 4 weeks or treatment with the superoxide dismutase mimetic Tempol. RESULTS C101A-eNOS-Tg showed significantly increased superoxide generation, protein-tyrosine-nitration and eNOS-tyrosine-nitration, eNOS-S-glutathionylation, eNOS phosphorylation and AMP kinase-α phosphorylation at Thr172 in aorta, skeletal muscle, left ventricular myocardium and lung as compared with eNOS-Tg and wild-type controls. Exercise training increased phosphorylation of eNOS at Ser and AMP kinase-α in wild-type. These physiologic adaptations were absent in C101A-eNOS-Tg. Maximal aortic endothelium-dependent relaxation was similar in all strains. C101A-eNOS-Tg displayed normal SBP despite higher levels of eNOS, whereas eNOS-Tg showed significant hypotension. Tempol completely reversed the occurring protein modifications and significantly reduced SBP in C101A-eNOS-Tg but not in wild-type. CONCLUSION Oxidative stress generated by endothelial-specific expression of genetically destabilized C101A-eNOS selectively prevents SBP-reducing activity of vascular eNOS, while having no effect on aortic endothelium-dependent relaxation. These data suggest that oxidative stress in microvascular endothelium may play a role for the development of essential hypertension.
Collapse
|
23
|
Hirata KI. Mitsuhiro Yokoyama, MD, PhD 1943-2016. Circ J 2017; 81:901-902. [PMID: 28603179 DOI: 10.1253/circj.cj-17-0504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Ken-Ichi Hirata
- The Cardiovascular Medicine Department, Kobe University Graduate School of Medicine
| |
Collapse
|
24
|
Xie X, Shi X, Xun X, Rao L. Endothelial nitric oxide synthase gene single nucleotide polymorphisms and the risk of hypertension: A meta-analysis involving 63,258 subjects. Clin Exp Hypertens 2017; 39:175-182. [PMID: 28287883 DOI: 10.1080/10641963.2016.1235177] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Xiaochuan Xie
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiaohan Shi
- Division of Reproductive Medical Center, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiaoshuang Xun
- West China School of Public Health, Sichuan University, Chengdu, Sichuan, China
| | - Li Rao
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
25
|
Kalinowski L, Janaszak-Jasiecka A, Siekierzycka A, Bartoszewska S, Woźniak M, Lejnowski D, Collawn JF, Bartoszewski R. Posttranscriptional and transcriptional regulation of endothelial nitric-oxide synthase during hypoxia: the role of microRNAs. Cell Mol Biol Lett 2016; 21:16. [PMID: 28536619 PMCID: PMC5415778 DOI: 10.1186/s11658-016-0017-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 08/18/2016] [Indexed: 02/07/2023] Open
Abstract
Understanding the cellular pathways that regulate endothelial nitric oxide (eNOS, NOS3) expression and consequently nitric oxide (NO) bioavailability during hypoxia is a necessary aspect in the development of novel treatments for cardiovascular disorders. eNOS expression and eNOS-dependent NO cellular signaling during hypoxia promote an equilibrium of transcriptional and posttranscriptional molecular mechanisms that belong to both proapoptotic and survival pathways. Furthermore, NO bioavailability results not only from eNOS levels, but also relies on the presence of eNOS substrate and cofactors, the phosphorylation status of eNOS, and the presence of reactive oxygen species (ROS) that can inactivate eNOS. Since both NOS3 levels and these signaling pathways can also be a subject of posttranscriptional modulation by microRNAs (miRNAs), this class of short noncoding RNAs contribute another level of regulation for NO bioavailability. As miRNA antagomirs or specific target protectors could be used in therapeutic approaches to regulate NO levels, either by changing NOS3 mRNA stability or through factors governing eNOS activity, it is critical to understand their role in governing eNOS activity during hypoxa. In contrast to a large number of miRNAs reported to the change eNOS expression during hypoxia, only a few miRNAs modulate eNOS activity. Furthermore, impaired miRNA biogenesis leads to NOS3 mRNA stabilization under hypoxia. Here we discuss the recent studies that define miRNAs’ role in maintaining endothelial NO bioavailability emphasizing those miRNAs that directly modulate NOS3 expression or eNOS activity.
Collapse
Affiliation(s)
- Leszek Kalinowski
- Department of Medical Laboratory Diagnostics and Central Bank of Frozen Tissues & Genetic Specimens, Medical University of Gdansk, Debinki 7, 80-211 Gdansk, Poland
| | - Anna Janaszak-Jasiecka
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Hallera 107, 80-416 Gdansk, Poland
| | - Anna Siekierzycka
- Department of Medical Laboratory Diagnostics and Central Bank of Frozen Tissues & Genetic Specimens, Medical University of Gdansk, Debinki 7, 80-211 Gdansk, Poland
| | - Sylwia Bartoszewska
- Department of Inorganic Chemistry, Medical University of Gdansk, Gdansk, Poland
| | - Marcin Woźniak
- Department of Medical Laboratory Diagnostics and Central Bank of Frozen Tissues & Genetic Specimens, Medical University of Gdansk, Debinki 7, 80-211 Gdansk, Poland
| | - Dawid Lejnowski
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Hallera 107, 80-416 Gdansk, Poland
| | - James F Collawn
- Department of Cell Biology, Developmental, and Integrative, University of Alabama at Birmingham, Birmingham, USA
| | - Rafal Bartoszewski
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Hallera 107, 80-416 Gdansk, Poland
| |
Collapse
|
26
|
Chandra SB, Mohan S, Ford BM, Huang L, Janardhanan P, Deo KS, Cong L, Muir ER, Duong TQ. Targeted overexpression of endothelial nitric oxide synthase in endothelial cells improves cerebrovascular reactivity in Ins2Akita-type-1 diabetic mice. J Cereb Blood Flow Metab 2016; 36:1135-42. [PMID: 26661212 PMCID: PMC4908624 DOI: 10.1177/0271678x15612098] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 09/23/2015] [Indexed: 11/16/2022]
Abstract
Reduced bioavailability of nitric oxide due to impaired endothelial nitric oxide synthase (eNOS) activity is a leading cause of endothelial dysfunction in diabetes. Enhancing eNOS activity in diabetes is a potential therapeutic target. This study investigated basal cerebral blood flow and cerebrovascular reactivity in wild-type mice, diabetic mice (Ins2(Akita+/-)), nondiabetic eNOS-overexpressing mice (TgeNOS), and the cross of two transgenic mice (TgeNOS-Ins2(Akita+/-)) at six months of age. The cross was aimed at improving eNOS expression in diabetic mice. The major findings were: (i) Body weights of Ins2(Akita+/-) and TgeNOS-Ins2(Akita+/-) were significantly different from wild-type and TgeNOS mice. Blood pressure of TgeNOS mice was lower than wild-type. (ii) Basal cerebral blood flow of the TgeNOS group was significantly higher than cerebral blood flow of the other three groups. (iii) The cerebrovascular reactivity in the Ins2(Akita+/-) mice was significantly lower compared with wild-type, whereas that in the TgeNOS-Ins2(Akita+/-) was significantly higher compared with the Ins2(Akita+/-) and TgeNOS groups. Overexpression of eNOS rescued cerebrovascular dysfunction in diabetic animals, resulting in improved cerebrovascular reactivity. These results underscore the possible role of eNOS in vascular dysfunction in the brain of diabetic mice and support the notion that enhancing eNOS activity in diabetes is a potential therapeutic target.
Collapse
Affiliation(s)
- Saurav B Chandra
- Research Imaging Institute, University of Texas Health Science Center, San Antonio, TX, USA
| | - Sumathy Mohan
- Department of Pathology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Bridget M Ford
- Research Imaging Institute, University of Texas Health Science Center, San Antonio, TX, USA
| | - Lei Huang
- Research Imaging Institute, University of Texas Health Science Center, San Antonio, TX, USA
| | - Preethi Janardhanan
- Department of Pathology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Kaiwalya S Deo
- Research Imaging Institute, University of Texas Health Science Center, San Antonio, TX, USA
| | - Linlin Cong
- Research Imaging Institute, University of Texas Health Science Center, San Antonio, TX, USA
| | - Eric R Muir
- Research Imaging Institute, University of Texas Health Science Center, San Antonio, TX, USA Department of Ophthalmology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Timothy Q Duong
- Research Imaging Institute, University of Texas Health Science Center, San Antonio, TX, USA Department of Ophthalmology, University of Texas Health Science Center, San Antonio, TX, USA
| |
Collapse
|
27
|
Park K, Mima A, Li Q, Rask-Madsen C, He P, Mizutani K, Katagiri S, Maeda Y, Wu IH, Khamaisi M, Preil SR, Maddaloni E, Sørensen D, Rasmussen LM, Huang PL, King GL. Insulin decreases atherosclerosis by inducing endothelin receptor B expression. JCI Insight 2016; 1:e86574. [PMID: 27200419 PMCID: PMC4869734 DOI: 10.1172/jci.insight.86574] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Endothelial cell (EC) insulin resistance and dysfunction, caused by diabetes, accelerates atherosclerosis. It is unknown whether specifically enhancing EC-targeted insulin action can decrease atherosclerosis in diabetes. Accordingly, overexpressing insulin receptor substrate-1 (IRS1) in the endothelia of Apoe-/- mice (Irs1/Apoe-/-) increased insulin signaling and function in the aorta. Atherosclerosis was significantly reduced in Irs1/ApoE-/- mice on diet-induced hyperinsulinemia and hyperglycemia. The mechanism of insulin's enhanced antiatherogenic actions in EC was related to remarkable induction of NO action, which increases endothelin receptor B (EDNRB) expression and intracellular [Ca2+]. Using the mice with knockin mutation of eNOS, which had Ser1176 mutated to alanine (AKI), deleting the only known mechanism for insulin to activate eNOS/NO pathway, we observed that IRS1 overexpression in the endothelia of Aki/ApoE-/- mice significantly decreased atherosclerosis. Interestingly, endothelial EDNRB expression was selectively reduced in intima of arteries from diabetic patients and rodents. However, endothelial EDNRB expression was upregulated by insulin via P13K/Akt pathway. Finally EDNRB deletion in EC of Ldlr-/- and Irs1/Ldlr-/- mice decreased NO production and accelerated atherosclerosis, compared with Ldlr-/- mice. Accelerated atherosclerosis in diabetes may be reduced by improving insulin signaling selectively via IRS1/Akt in the EC by inducing EDNRB expression and NO production.
Collapse
Affiliation(s)
- Kyoungmin Park
- Dianne Nunnally Hoppes Laboratory Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Akira Mima
- Dianne Nunnally Hoppes Laboratory Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Qian Li
- Dianne Nunnally Hoppes Laboratory Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Christian Rask-Madsen
- Dianne Nunnally Hoppes Laboratory Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Pingnian He
- Department of Cellular and Molecular Physiology, Penn State Hershey College of Medicine, Hershey, Pennsylvania, USA
| | - Koji Mizutani
- Dianne Nunnally Hoppes Laboratory Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Sayaka Katagiri
- Dianne Nunnally Hoppes Laboratory Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Yasutaka Maeda
- Dianne Nunnally Hoppes Laboratory Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - I-Hsien Wu
- Dianne Nunnally Hoppes Laboratory Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Mogher Khamaisi
- Dianne Nunnally Hoppes Laboratory Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Simone Rordam Preil
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, University of Southern Denmark, Odense, Denmark
| | - Ernesto Maddaloni
- Dianne Nunnally Hoppes Laboratory Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Ditte Sørensen
- Dianne Nunnally Hoppes Laboratory Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, University of Southern Denmark, Odense, Denmark
- Danish Diabetes Academy, Odense, Denmark
| | - Lars Melholt Rasmussen
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, University of Southern Denmark, Odense, Denmark
| | - Paul L. Huang
- Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - George L. King
- Dianne Nunnally Hoppes Laboratory Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
28
|
Shelite TR, Liang Y, Wang H, Mendell NL, Trent BJ, Sun J, Gong B, Xu G, Hu H, Bouyer DH, Soong L. IL-33-Dependent Endothelial Activation Contributes to Apoptosis and Renal Injury in Orientia tsutsugamushi-Infected Mice. PLoS Negl Trop Dis 2016; 10:e0004467. [PMID: 26943125 PMCID: PMC4778942 DOI: 10.1371/journal.pntd.0004467] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Accepted: 01/27/2016] [Indexed: 01/23/2023] Open
Abstract
Endothelial cells (EC) are the main target for Orientia tsutsugamushi infection and EC dysfunction is a hallmark of severe scrub typhus in patients. However, the molecular basis of EC dysfunction and its impact on infection outcome are poorly understood. We found that C57BL/6 mice that received a lethal dose of O. tsutsugamushi Karp strain had a significant increase in the expression of IL-33 and its receptor ST2L in the kidneys and liver, but a rapid reduction of IL-33 in the lungs. We also found exacerbated EC stress and activation in the kidneys of infected mice, as evidenced by elevated angiopoietin (Ang) 2/Ang1 ratio, increased endothelin 1 (ET-1) and endothelial nitric oxide synthase (eNOS) expression. Such responses were significantly attenuated in the IL-33-/- mice. Importantly, IL-33-/- mice also had markedly attenuated disease due to reduced EC stress and cellular apoptosis. To confirm the biological role of IL-33, we challenged wild-type (WT) mice with a sub-lethal dose of O. tsutsugamushi and gave mice recombinant IL-33 (rIL-33) every 2 days for 10 days. Exogenous IL-33 significantly increased disease severity and lethality, which correlated with increased EC stress and activation, increased CXCL1 and CXCL2 chemokines, but decreased anti-apoptotic gene BCL-2 in the kidneys. To further examine the role of EC stress, we infected human umbilical vein endothelial cells (HUVEC) in vitro. We found an infection dose-dependent increase in the expression of IL-33, ST2L soluble ST2 (sST2), and the Ang2/Ang1 ratio at 24 and 48 hours post-infection. This study indicates a pathogenic role of alarmin IL-33 in a murine model of scrub typhus and highlights infection-triggered EC damage and IL-33-mediated pathological changes during the course of Orientia infection.
Collapse
Affiliation(s)
- Thomas R Shelite
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Yuejin Liang
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Hui Wang
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Nicole L Mendell
- Department of Pathology, Center for Biodefense and Emerging Infectious Diseases, Center for Tropical Diseases, Sealy Center for Vaccine Development, Institute of Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Brandon J Trent
- Department of Pathology, Center for Biodefense and Emerging Infectious Diseases, Center for Tropical Diseases, Sealy Center for Vaccine Development, Institute of Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Jiaren Sun
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Bin Gong
- Department of Pathology, Center for Biodefense and Emerging Infectious Diseases, Center for Tropical Diseases, Sealy Center for Vaccine Development, Institute of Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Guang Xu
- Department of Pathology, Center for Biodefense and Emerging Infectious Diseases, Center for Tropical Diseases, Sealy Center for Vaccine Development, Institute of Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Haitao Hu
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Donald H Bouyer
- Department of Pathology, Center for Biodefense and Emerging Infectious Diseases, Center for Tropical Diseases, Sealy Center for Vaccine Development, Institute of Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Lynn Soong
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America.,Department of Pathology, Center for Biodefense and Emerging Infectious Diseases, Center for Tropical Diseases, Sealy Center for Vaccine Development, Institute of Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| |
Collapse
|
29
|
Sofronova SI, Borzykh AA, Gaynullina DK, Kuzmin IV, Shvetsova AA, Lukoshkova EV, Tarasova OS. Endothelial nitric oxide weakens arterial contractile responses and reduces blood pressure during early postnatal development in rats. Nitric Oxide 2016; 55-56:1-9. [PMID: 26923819 DOI: 10.1016/j.niox.2016.02.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 01/22/2016] [Accepted: 02/19/2016] [Indexed: 01/01/2023]
Abstract
OBJECTIVE During maturation the vascular system undergoes structural and functional remodeling. At the systemic level it results in a gradual increase of arterial blood pressure during postnatal ontogenesis. The mechanisms of maintaining the blood pressure at a comparatively low level during the early postnatal development are not completely understood. Recently we showed that the hindlimb arteries of young (1-2 wk-old) rats exhibited an enhanced endothelial NO-pathway activity, which weakened their contractile responsiveness compared to the arteries of adult rats. Here we tested the hypothesis that an increased tonic endothelial NO production can take place in the whole vascular system leading to a decreased level of systemic blood pressure in young rats. DESIGN AND METHODS Segments of small mesenteric, saphenous, sural and intrarenal arteries were isolated from the young (2 wk-old), juvenile (4 wk-old) and adult (10-12 wk-old) male rats and tested in a wire isometric myograph. Anticontractile effect of NO was evaluated by the effects of NOS inhibitor L-NNA on both arterial spontaneous tone and constrictor responses to methoxamine (α1-adrenoceptor agonist). In addition, eNOS and arginase-2 mRNA expression in arterial preparations by qPCR and serum nitrite/nitrate levels by Griess reaction were estimated. Blood pressure with an intra-carotid artery catheter was measured in conscious rats. RESULTS In all arteries of 2 wk rats except the renal ones, L-NNA exposure resulted in a considerable tonic contraction and a remarkable enhancement of contractile responses to methoxamine. The effect of L-NNA gradually decreased with age and by 10-12 weeks became very small in the mesenteric arteries and disappeared in the sural and saphenous arteries. Although no difference in eNOS mRNA expression was found, the content of arginase-2 mRNA was significantly lower in young rats compared to adults. Serum levels of NO metabolites were two-fold higher in 2 wk-old rats than in adult rats. Along with that, arterial blood pressure was by half lower but rose more prominently after administration of l-NAME in young rats than in adults. CONCLUSIONS In young rats, tonic release of NO by the endothelium considerably weakens contractile responses of arteries supplying intestine, skin and skeletal muscles, which receive a high proportion of the cardiac output. Such anticontractile effect of NO can be an important mechanism responsible for the blood pressure reduction in immature circulatory system.
Collapse
Affiliation(s)
- Svetlana I Sofronova
- Faculty of Biology, M.V. Lomonosov Moscow State University, Leninskie Gory 1-12, 119234, Moscow, Russia; Institute for Biomedical Problems, Russian Academy of Sciences, Khoroshevskoe shosse 76A, 123007, Moscow, Russia.
| | - Anna A Borzykh
- Institute for Biomedical Problems, Russian Academy of Sciences, Khoroshevskoe shosse 76A, 123007, Moscow, Russia
| | - Dina K Gaynullina
- Faculty of Biology, M.V. Lomonosov Moscow State University, Leninskie Gory 1-12, 119234, Moscow, Russia; Institute for Biomedical Problems, Russian Academy of Sciences, Khoroshevskoe shosse 76A, 123007, Moscow, Russia; Department of Physiology, Russian National Research Medical University, Ostrovitianova str. 1, 117997, Moscow, Russia
| | - Ilya V Kuzmin
- Faculty of Biology, M.V. Lomonosov Moscow State University, Leninskie Gory 1-12, 119234, Moscow, Russia; Institute for Biomedical Problems, Russian Academy of Sciences, Khoroshevskoe shosse 76A, 123007, Moscow, Russia
| | - Anastasia A Shvetsova
- Faculty of Biology, M.V. Lomonosov Moscow State University, Leninskie Gory 1-12, 119234, Moscow, Russia; Institute for Biomedical Problems, Russian Academy of Sciences, Khoroshevskoe shosse 76A, 123007, Moscow, Russia
| | - Elena V Lukoshkova
- Institute for Biomedical Problems, Russian Academy of Sciences, Khoroshevskoe shosse 76A, 123007, Moscow, Russia; Institute of Experimental Cardiology, Russian Cardiology Research Center, 3rd Cherepkovskaya Street 15a, 121552, Moscow, Russia
| | - Olga S Tarasova
- Faculty of Biology, M.V. Lomonosov Moscow State University, Leninskie Gory 1-12, 119234, Moscow, Russia; Institute for Biomedical Problems, Russian Academy of Sciences, Khoroshevskoe shosse 76A, 123007, Moscow, Russia
| |
Collapse
|
30
|
Disruption of Physiological Balance Between Nitric Oxide and Endothelium-Dependent Hyperpolarization Impairs Cardiovascular Homeostasis in Mice. Arterioscler Thromb Vasc Biol 2016; 36:97-107. [DOI: 10.1161/atvbaha.115.306499] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 10/26/2015] [Indexed: 01/09/2023]
|
31
|
Krishnan M, Janardhanan P, Roman L, Reddick RL, Natarajan M, van Haperen R, Habib SL, de Crom R, Mohan S. Enhancing eNOS activity with simultaneous inhibition of IKKβ restores vascular function in Ins2(Akita+/-) type-1 diabetic mice. J Transl Med 2015; 95:1092-104. [PMID: 26214584 DOI: 10.1038/labinvest.2015.96] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Revised: 04/24/2015] [Accepted: 05/28/2015] [Indexed: 12/13/2022] Open
Abstract
The balance of nitric oxide (NO) versus superoxide generation has a major role in the initiation and progression of endothelial dysfunction. Under conditions of high glucose, endothelial nitric oxide synthase (eNOS) functions as a chief source of superoxide rather than NO. In order to improve NO bioavailability within the vessel wall in type-1 diabetes, we investigated treatment strategies that improve eNOS phosphorylation and NO-dependent vasorelaxation. We evaluated methods to increase the eNOS activity by (1) feeding Ins2(Akita) spontaneously diabetic (type-1) mice with l-arginine in the presence of sepiapterin, a precursor of tetrahydrobiopterin; (2) preventing eNOS/NO deregulation by the inclusion of inhibitor kappa B kinase beta (IKKβ) inhibitor, salsalate, in the diet regimen in combination with l-arginine and sepiapterin; and (3) independently increasing eNOS expression to improve eNOS activity and associated NO production through generating Ins2(Akita) diabetic mice that overexpress human eNOS predominantly in vascular endothelial cells. Our results clearly demonstrated that diet supplementation with l-arginine, sepiapterin along with salsalate improved phosphorylation of eNOS and enhanced vasorelaxation of thoracic/abdominal aorta in type-1 diabetic mice. More interestingly, despite the overexpression of eNOS, the in-house generated transgenic eNOS-GFP (TgeNOS-GFP)-Ins2(Akita) cross mice showed an unanticipated effect of reduced eNOS phosphorylation and enhanced superoxide production. Our results demonstrate that enhancement of endogenous eNOS activity by nutritional modulation is more beneficial than increasing the endogenous expression of eNOS by gene therapy modalities.
Collapse
Affiliation(s)
- Manickam Krishnan
- Department of Pathology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Preethi Janardhanan
- Department of Pathology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Linda Roman
- Department of Biochemistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Robert L Reddick
- Department of Pathology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Mohan Natarajan
- Department of Pathology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Rien van Haperen
- Department of Cell Biology and Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Samy L Habib
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Rini de Crom
- Department of Cell Biology and Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Sumathy Mohan
- Department of Pathology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| |
Collapse
|
32
|
Suvorava T, Nagy N, Pick S, Lieven O, Rüther U, Dao VTV, Fischer JW, Weber M, Kojda G. Impact of eNOS-Dependent Oxidative Stress on Endothelial Function and Neointima Formation. Antioxid Redox Signal 2015; 23:711-23. [PMID: 25764009 PMCID: PMC4580305 DOI: 10.1089/ars.2014.6059] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 03/03/2015] [Accepted: 03/11/2015] [Indexed: 12/14/2022]
Abstract
AIMS Vascular oxidative stress generated by endothelial NO synthase (eNOS) was observed in experimental and clinical cardiovascular disease, but its relative importance for vascular pathologies is unclear. We investigated the impact of eNOS-dependent vascular oxidative stress on endothelial function and on neointimal hyperplasia. RESULTS A dimer-destabilized mutant of bovine eNOS where cysteine 101 was replaced by alanine was cloned and introduced into an eNOS-deficient mouse strain (eNOS-KO) in an endothelial-specific manner. Destabilization of mutant eNOS in cells and eNOS-KO was confirmed by the reduced dimer/monomer ratio. Purified mutant eNOS and transfected cells generated less citrulline and NO, respectively, while superoxide generation was enhanced. In eNOS-KO, introduction of mutant eNOS caused a 2.3-3.7-fold increase in superoxide and peroxynitrite formation in the aorta and myocardium. This was completely blunted by an NOS inhibitor. Nevertheless, expression of mutant eNOS in eNOS-KO completely restored maximal aortic endothelium-dependent relaxation to acetylcholine. Neointimal hyperplasia induced by carotid binding was much larger in eNOS-KO than in mutant eNOS-KO and C57BL/6, while the latter strains showed comparable hyperplasia. Likewise, vascular remodeling was blunted in eNOS-KO only. INNOVATION Our results provide the first in vivo evidence that eNOS-dependent oxidative stress is unlikely to be an initial cause of impaired endothelium-dependent vasodilation and/or a pathologic factor promoting intimal hyperplasia. These findings highlight the importance of other sources of vascular oxidative stress in cardiovascular disease. CONCLUSION eNOS-dependent oxidative stress is unlikely to induce functional vascular damage as long as concomitant generation of NO is preserved. This underlines the importance of current and new therapeutic strategies in improving endothelial NO generation.
Collapse
Affiliation(s)
- Tatsiana Suvorava
- Institute for Pharmacology and Clinical Pharmacology, Heinrich-Heine-University, Düsseldorf, Germany
| | - Nadine Nagy
- Institute for Pharmacology and Clinical Pharmacology, Heinrich-Heine-University, Düsseldorf, Germany
| | - Stephanie Pick
- Institute for Pharmacology and Clinical Pharmacology, Heinrich-Heine-University, Düsseldorf, Germany
| | - Oliver Lieven
- Institute for Animal Developmental and Molecular Biology, Heinrich-Heine-University, Düsseldorf, Germany
| | - Ulrich Rüther
- Institute for Animal Developmental and Molecular Biology, Heinrich-Heine-University, Düsseldorf, Germany
| | - Vu Thao-Vi Dao
- Institute for Pharmacology and Clinical Pharmacology, Heinrich-Heine-University, Düsseldorf, Germany
| | - Jens W. Fischer
- Institute for Pharmacology and Clinical Pharmacology, Heinrich-Heine-University, Düsseldorf, Germany
| | - Martina Weber
- Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia
| | - Georg Kojda
- Institute for Pharmacology and Clinical Pharmacology, Heinrich-Heine-University, Düsseldorf, Germany
| |
Collapse
|
33
|
Abstract
AbstractThe bovine mammary gland is a dynamic and complex organ composed of various cell types that work together for the purpose of milk synthesis and secretion. A layer of endothelial cells establishes the blood–milk barrier, which exists to facilitate the exchange of solutes and macromolecules necessary for optimal milk production. During bacterial challenge, however, endothelial cells divert some of their lactation function to protect the underlying tissue from damage by initiating inflammation. At the onset of inflammation, endothelial cells tightly regulate the movement of plasma components and leukocytes into affected tissue. Unfortunately, endothelial dysfunction as a result of exacerbated or sustained inflammation can negatively affect both barrier integrity and the health of surrounding extravascular tissue. The objective of this review is to highlight the role of endothelial cells in supporting milk production and regulating optimal inflammatory responses. The consequences of endothelial dysfunction and sustained inflammation on milk synthesis and secretion are discussed. Given the important role of endothelial cells in orchestrating the inflammatory response, a better understanding of endothelial function during mastitis may support development of targeted therapies to protect bovine mammary tissue and mammary endothelium.
Collapse
|
34
|
Lee WJ, Tateya S, Cheng AM, Rizzo-DeLeon N, Wang NF, Handa P, Wilson CL, Clowes AW, Sweet IR, Bomsztyk K, Schwartz MW, Kim F. M2 Macrophage Polarization Mediates Anti-inflammatory Effects of Endothelial Nitric Oxide Signaling. Diabetes 2015; 64:2836-46. [PMID: 25845662 PMCID: PMC4512216 DOI: 10.2337/db14-1668] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 03/21/2015] [Indexed: 12/18/2022]
Abstract
Endothelial nitric oxide (NO) signaling plays a physiological role in limiting obesity-associated insulin resistance and inflammation. This study was undertaken to investigate whether this NO effect involves polarization of macrophages toward an anti-inflammatory M2 phenotype. Mice with transgenic endothelial NO synthase overexpression were protected against high-fat diet (HFD)-induced hepatic inflammation and insulin resistance, and this effect was associated with reduced proinflammatory M1 and increased anti-inflammatory M2 activation of Kupffer cells. In cell culture studies, exposure of macrophages to endothelial NO similarly reduced inflammatory (M1) and increased anti-inflammatory (M2) gene expression. Similar effects were induced by macrophage overexpression of vasodilator-stimulated phosphoprotein (VASP), a key downstream mediator of intracellular NO signaling. Conversely, VASP deficiency induced proinflammatory M1 macrophage activation, and the transplantation of bone marrow from VASP-deficient donor mice into normal recipients caused hepatic inflammation and insulin resistance resembling that induced in normal mice by consumption of an HFD. These data suggest that proinflammatory macrophage M1 activation and macrophage-mediated inflammation are tonically inhibited by NO → VASP signal transduction, and that reduced NO → VASP signaling is involved in the effect of HFD feeding to induce M1 activation of Kupffer cells and associated hepatic inflammation. Our data implicate endothelial NO → VASP signaling as a physiological determinant of macrophage polarization and show that signaling via this pathway is required to prevent hepatic inflammation and insulin resistance.
Collapse
Affiliation(s)
- Woo Je Lee
- Department of Medicine, University of Washington, Seattle, WA Diabetes and Obesity Center of Excellence, University of Washington, Seattle, WA
| | - Sanshiro Tateya
- Department of Medicine, University of Washington, Seattle, WA Diabetes and Obesity Center of Excellence, University of Washington, Seattle, WA
| | - Andrew M Cheng
- Department of Medicine, University of Washington, Seattle, WA Diabetes and Obesity Center of Excellence, University of Washington, Seattle, WA
| | - Norma Rizzo-DeLeon
- Department of Medicine, University of Washington, Seattle, WA Diabetes and Obesity Center of Excellence, University of Washington, Seattle, WA
| | - Nicholas F Wang
- Department of Medicine, University of Washington, Seattle, WA Diabetes and Obesity Center of Excellence, University of Washington, Seattle, WA
| | - Priya Handa
- Department of Medicine, University of Washington, Seattle, WA Diabetes and Obesity Center of Excellence, University of Washington, Seattle, WA
| | - Carole L Wilson
- Department of Pathology, University of Washington, Seattle, WA
| | | | - Ian R Sweet
- Department of Medicine, University of Washington, Seattle, WA Diabetes and Obesity Center of Excellence, University of Washington, Seattle, WA
| | - Karol Bomsztyk
- Department of Medicine, University of Washington, Seattle, WA
| | - Michael W Schwartz
- Department of Medicine, University of Washington, Seattle, WA Diabetes and Obesity Center of Excellence, University of Washington, Seattle, WA
| | - Francis Kim
- Department of Medicine, University of Washington, Seattle, WA Diabetes and Obesity Center of Excellence, University of Washington, Seattle, WA
| |
Collapse
|
35
|
Suvorava T, Stegbauer J, Thieme M, Pick S, Friedrich S, Rump LC, Hohlfeld T, Kojda G. Sustained hypertension despite endothelial-specific eNOS rescue in eNOS-deficient mice. Biochem Biophys Res Commun 2015; 458:576-583. [PMID: 25680465 DOI: 10.1016/j.bbrc.2015.01.152] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 01/28/2015] [Indexed: 02/07/2023]
Abstract
The aim of the study was to evaluate the possible contribution of non-endothelial eNOS to the regulation of blood pressure (BP). To accomplish this, a double transgenic strain expressing eNOS exclusively in the vascular endothelium (eNOS-Tg/KO) has been generated by endothelial-specific targeting of bovine eNOS in eNOS-deficient mice (eNOS-KO). Expression of eNOS was evaluated in aorta, myocardium, kidney, brain stem and skeletal muscle. Organ bath studies revealed a complete normalization of aortic reactivity to acetylcholine, phenylephrine and the NO-donors in eNOS-Tg/KO. Function of eNOS in resistance arteries was demonstrated by acute i.v. infusion of acetylcholine and the NOS-inhibitor L-NAME. Acetylcholine decreased mean arterial pressure in all strains but eNOS-KO responded significantly less sensitive as compared eNOS-Tg/KO and C57BL/6. Likewise, acute i.v. L-NAME application elevated mean arterial pressure in C57BL/6 and eNOS-Tg/KO, but not in eNOS-KO. In striking contrast to these findings, mean, systolic and diastolic BP in eNOS-Tg/KO remained significantly elevated and was similar to values of eNOS-KO. Chronic oral treatment with L-NAME increased BP to the level of eNOS-KO only in C57BL/6, but had no effect on hypertension in eNOS-KO and eNOS-Tg/KO. Taken together, functional reconstitution of eNOS in the vasculature of eNOS-KO not even partially lowered BP. These data suggest that the activity of eNOS expressed in non-vascular tissue might play a role in physiologic BP regulation.
Collapse
Affiliation(s)
- Tatsiana Suvorava
- Institute of Pharmacology and Clinical Pharmacology, Heinrich-Heine-University, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Johannes Stegbauer
- Department of Nephrology, University Hospital, Heinrich-Heine-University, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Manuel Thieme
- Department of Nephrology, University Hospital, Heinrich-Heine-University, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Stephanie Pick
- Institute of Pharmacology and Clinical Pharmacology, Heinrich-Heine-University, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Sebastian Friedrich
- Department of Nephrology, University Hospital, Heinrich-Heine-University, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Lars C Rump
- Department of Nephrology, University Hospital, Heinrich-Heine-University, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Thomas Hohlfeld
- Institute of Pharmacology and Clinical Pharmacology, Heinrich-Heine-University, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Georg Kojda
- Institute of Pharmacology and Clinical Pharmacology, Heinrich-Heine-University, Universitätsstr. 1, 40225 Düsseldorf, Germany.
| |
Collapse
|
36
|
Lacchini R, Muniz JJ, Nobre YTDA, Cologna AJ, Martins ACP, Tanus-Santos JE. nNOS polymorphisms are associated with responsiveness to sildenafil in clinical and postoperative erectile dysfunction. Pharmacogenomics 2015; 15:775-84. [PMID: 24897285 DOI: 10.2217/pgs.14.30] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
AIM Sildenafil potentiates the nitric oxide (NO) signaling pathway. Since neuronal NOS is very important in the penis, we assessed whether NOS1 polymorphisms are associated with altered responsiveness to sildenafil in erectile dysfunction (ED). MATERIALS & METHODS Patients (n = 137) were divided as clinical ED or postoperative ED. They were subdivided as good responders or poor responders to sildenafil, and genotypes for rs41279104 and rs2682826 NOS1 polymorphisms were determined. RESULTS We found that the rs41279104 CT genotype was associated with good responders in postoperative ED patients, while rs2682826 CT genotype was associated with good responders in postoperative ED, and the TT genotype associated with good responders in both groups. Finally, the CT haplotype was associated with good responders in postoperative ED. CONCLUSION NOS1 polymorphisms are associated with responsiveness to sildenafil in ED. Original submitted 20 November 2013; Revision submitted 31 January 2014.
Collapse
Affiliation(s)
- Riccardo Lacchini
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Av. Bandeirantes, 3900, 14049-900 Ribeirao Preto, SP, Brazil.
| | | | | | | | | | | |
Collapse
|
37
|
Sansbury BE, Bhatnagar A, Hill BG. Impact of nutrient excess and endothelial nitric oxide synthase on the plasma metabolite profile in mice. Front Physiol 2014; 5:453. [PMID: 25505420 PMCID: PMC4243488 DOI: 10.3389/fphys.2014.00453] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 11/03/2014] [Indexed: 01/08/2023] Open
Abstract
An increase in calorie consumption is associated with the recent rise in obesity prevalence. However, our current understanding of the effects of nutrient excess on major metabolic pathways appears insufficient to develop safe and effective metabolic interventions to prevent obesity. Hence, we sought to identify systemic metabolic changes caused by nutrient excess and to determine how endothelial nitric oxide synthase (eNOS)—which has anti-obesogenic properties—affects systemic metabolism by measuring plasma metabolites. Wild-type (WT) and eNOS transgenic (eNOS-TG) mice were placed on low fat or high fat diets for 6 weeks, and plasma metabolites were measured using an unbiased metabolomic approach. High fat feeding in WT mice led to significant increases in fat mass, which was associated with significantly lower plasma levels of 1,5-anhydroglucitol, lysophospholipids, 3-dehydrocarnitine, and bile acids, as well as branched chain amino acids (BCAAs) and their metabolites. Plasma levels of several lipids including sphingomyelins, stearoylcarnitine, dihomo-linoleate and metabolites associated with oxidative stress were increased by high fat diet. In comparison with low fat-fed WT mice, eNOS-TG mice showed lower levels of several free fatty acids, but in contrast, the levels of bile acids, amino acids, and BCAA catabolites were increased. When placed on a high fat diet, eNOS overexpressing mice showed remarkably higher levels of plasma bile acids and elevated levels of plasma BCAAs and their catabolites compared with WT mice. Treatment with GW4064, an inhibitor of bile acid synthesis, decreased plasma bile acid levels but was not sufficient to reverse the anti-obesogenic effects of eNOS overexpression. These findings reveal unique metabolic changes in response to high fat diet and eNOS overexpression and suggest that the anti-obesity effects of eNOS are likely independent of changes in the bile acid pool.
Collapse
Affiliation(s)
- Brian E Sansbury
- Division of Cardiology, Department of Medicine, Institute of Molecular Cardiology, University of Louisville Louisville, KY, USA ; Department of Medicine, Diabetes and Obesity Center, University of Louisville Louisville, KY, USA ; Department of Physiology and Biophysics, University of Louisville Louisville, KY, USA
| | - Aruni Bhatnagar
- Division of Cardiology, Department of Medicine, Institute of Molecular Cardiology, University of Louisville Louisville, KY, USA ; Department of Medicine, Diabetes and Obesity Center, University of Louisville Louisville, KY, USA ; Department of Physiology and Biophysics, University of Louisville Louisville, KY, USA ; Department of Biochemistry and Molecular Biology, University of Louisville Louisville, KY, USA
| | - Bradford G Hill
- Division of Cardiology, Department of Medicine, Institute of Molecular Cardiology, University of Louisville Louisville, KY, USA ; Department of Medicine, Diabetes and Obesity Center, University of Louisville Louisville, KY, USA ; Department of Physiology and Biophysics, University of Louisville Louisville, KY, USA ; Department of Biochemistry and Molecular Biology, University of Louisville Louisville, KY, USA
| |
Collapse
|
38
|
Okaniwa N, Sasaki M, Mizushima T, Ogasawara N, Funaki Y, Joh T, Kasugai K. eNOS plays an important role in the regulation of colonic inflammation: a novel therapeutic target and a predictive marker for the prognosis of ulcerative colitis. Free Radic Res 2014; 49:35-44. [PMID: 25329334 DOI: 10.3109/10715762.2014.977788] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND We reported that deficiency of the eNOS protein exacerbates colitis induced by dextran sodium sulfate (DSS-induced colitis). However, the role of eNOS in colitis remains controversial. Therefore, we studied how over-expression of eNOS affected this inflammatory condition, using vascular endothelial cells and mice as in vitro and in vivo models, respectively. Furthermore, we investigated the influence of a polymorphism in the eNOS gene on the clinical features of ulcerative colitis (UC). METHODS We examined the effect of eNOS overexpression on the expression of adhesion molecules in the endothelium and assessed the degree of DSS-induced colitis in eNOS transgenic (eNOS-Tg) mice. We also investigated the relationship between a polymorphism in the eNOS gene and clinical features of patients with UC. RESULTS The expression of adhesion molecules, under inflammatory conditions, was attenuated in eNOS gene-transfected vascular endothelial cells, as measured by western blot analysis. Symptoms of DSS-induced colitis were likewise attenuated in eNOS-Tg mice, which exhibited lower weight loss, mortality, histological damage (by inflammation score and crypt damage score), and colonic myeloperoxidase activity, tumor necrosis factor-α expression, and MAdCAM-1 expression than in wild-type mice. Furthermore, there was a significant relationship between intractable cases of UC and a polymorphism in the eNOS gene promoter (c.-786 T > C) that decreases eNOS expression. CONCLUSION The eNOS gene plays an important role in the regulation of colonic inflammation. The level of eNOS expression may be a predictive marker for prognosis of UC, and eNOS expression may be a novel therapeutic target.
Collapse
Affiliation(s)
- N Okaniwa
- Department of Gastroenterology, Aichi Medical University School of Medicine , Nagakute , Japan
| | | | | | | | | | | | | |
Collapse
|
39
|
Straub AC, Butcher JT, Billaud M, Mutchler SM, Artamonov MV, Nguyen AT, Johnson T, Best AK, Miller MP, Palmer LA, Columbus L, Somlyo AV, Le TH, Isakson BE. Hemoglobin α/eNOS coupling at myoendothelial junctions is required for nitric oxide scavenging during vasoconstriction. Arterioscler Thromb Vasc Biol 2014; 34:2594-600. [PMID: 25278292 DOI: 10.1161/atvbaha.114.303974] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Hemoglobin α (Hb α) and endothelial nitric oxide synthase (eNOS) form a macromolecular complex at myoendothelial junctions; the functional role of this interaction remains undefined. To test if coupling of eNOS and Hb α regulates nitric oxide signaling, vascular reactivity, and blood pressure using a mimetic peptide of Hb α to disrupt this interaction. APPROACH AND RESULTS In silico modeling of Hb α and eNOS identified a conserved sequence of interaction. By mutating portions of Hb α, we identified a specific sequence that binds eNOS. A mimetic peptide of the Hb α sequence (Hb α X) was generated to disrupt this complex. Using in vitro binding assays with purified Hb α and eNOS and ex vivo proximity ligation assays on resistance arteries, we have demonstrated that Hb α X significantly decreased interaction between eNOS and Hb α. Fluorescein isothiocyanate labeling of Hb α X revealed localization to holes in the internal elastic lamina (ie, myoendothelial junctions). To test the functional effects of Hb α X, we measured cyclic guanosine monophosphate and vascular reactivity. Our results reveal augmented cyclic guanosine monophosphate production and altered vasoconstriction with Hb α X. To test the in vivo effects of these peptides on blood pressure, normotensive and hypertensive mice were injected with Hb α X, which caused a significant decrease in blood pressure; injection of Hb α X into eNOS(-/-) mice had no effect. CONCLUSIONS These results identify a novel sequence on Hb α that is important for Hb α/eNOS complex formation and is critical for nitric oxide signaling at myoendothelial junctions.
Collapse
Affiliation(s)
- Adam C Straub
- From the Department of Pharmacology and Chemical Biology (A.C.S.) and Heart, Lung, Blood and Vascular Medicine Institute (A.C.S., A.T.N., M.P.M.), University of Pittsburgh, PA; Robert M. Berne Cardiovascular Research Center (J.T.B., M.B., S.M.M., T.J., A.K.B., B.E.I.), Department of Molecular Physiology and Biophysics (M.B., M.V.A., A.V.S., B.E.I.), Deparment of Pediatrics (L.A.P.), Department of Chemistry (L.C.), and Department of Medicine (T.H.L.), University of Virginia, Charlottesville
| | - Joshua T Butcher
- From the Department of Pharmacology and Chemical Biology (A.C.S.) and Heart, Lung, Blood and Vascular Medicine Institute (A.C.S., A.T.N., M.P.M.), University of Pittsburgh, PA; Robert M. Berne Cardiovascular Research Center (J.T.B., M.B., S.M.M., T.J., A.K.B., B.E.I.), Department of Molecular Physiology and Biophysics (M.B., M.V.A., A.V.S., B.E.I.), Deparment of Pediatrics (L.A.P.), Department of Chemistry (L.C.), and Department of Medicine (T.H.L.), University of Virginia, Charlottesville
| | - Marie Billaud
- From the Department of Pharmacology and Chemical Biology (A.C.S.) and Heart, Lung, Blood and Vascular Medicine Institute (A.C.S., A.T.N., M.P.M.), University of Pittsburgh, PA; Robert M. Berne Cardiovascular Research Center (J.T.B., M.B., S.M.M., T.J., A.K.B., B.E.I.), Department of Molecular Physiology and Biophysics (M.B., M.V.A., A.V.S., B.E.I.), Deparment of Pediatrics (L.A.P.), Department of Chemistry (L.C.), and Department of Medicine (T.H.L.), University of Virginia, Charlottesville
| | - Stephanie M Mutchler
- From the Department of Pharmacology and Chemical Biology (A.C.S.) and Heart, Lung, Blood and Vascular Medicine Institute (A.C.S., A.T.N., M.P.M.), University of Pittsburgh, PA; Robert M. Berne Cardiovascular Research Center (J.T.B., M.B., S.M.M., T.J., A.K.B., B.E.I.), Department of Molecular Physiology and Biophysics (M.B., M.V.A., A.V.S., B.E.I.), Deparment of Pediatrics (L.A.P.), Department of Chemistry (L.C.), and Department of Medicine (T.H.L.), University of Virginia, Charlottesville
| | - Mykhaylo V Artamonov
- From the Department of Pharmacology and Chemical Biology (A.C.S.) and Heart, Lung, Blood and Vascular Medicine Institute (A.C.S., A.T.N., M.P.M.), University of Pittsburgh, PA; Robert M. Berne Cardiovascular Research Center (J.T.B., M.B., S.M.M., T.J., A.K.B., B.E.I.), Department of Molecular Physiology and Biophysics (M.B., M.V.A., A.V.S., B.E.I.), Deparment of Pediatrics (L.A.P.), Department of Chemistry (L.C.), and Department of Medicine (T.H.L.), University of Virginia, Charlottesville
| | - Anh T Nguyen
- From the Department of Pharmacology and Chemical Biology (A.C.S.) and Heart, Lung, Blood and Vascular Medicine Institute (A.C.S., A.T.N., M.P.M.), University of Pittsburgh, PA; Robert M. Berne Cardiovascular Research Center (J.T.B., M.B., S.M.M., T.J., A.K.B., B.E.I.), Department of Molecular Physiology and Biophysics (M.B., M.V.A., A.V.S., B.E.I.), Deparment of Pediatrics (L.A.P.), Department of Chemistry (L.C.), and Department of Medicine (T.H.L.), University of Virginia, Charlottesville
| | - Tyler Johnson
- From the Department of Pharmacology and Chemical Biology (A.C.S.) and Heart, Lung, Blood and Vascular Medicine Institute (A.C.S., A.T.N., M.P.M.), University of Pittsburgh, PA; Robert M. Berne Cardiovascular Research Center (J.T.B., M.B., S.M.M., T.J., A.K.B., B.E.I.), Department of Molecular Physiology and Biophysics (M.B., M.V.A., A.V.S., B.E.I.), Deparment of Pediatrics (L.A.P.), Department of Chemistry (L.C.), and Department of Medicine (T.H.L.), University of Virginia, Charlottesville
| | - Angela K Best
- From the Department of Pharmacology and Chemical Biology (A.C.S.) and Heart, Lung, Blood and Vascular Medicine Institute (A.C.S., A.T.N., M.P.M.), University of Pittsburgh, PA; Robert M. Berne Cardiovascular Research Center (J.T.B., M.B., S.M.M., T.J., A.K.B., B.E.I.), Department of Molecular Physiology and Biophysics (M.B., M.V.A., A.V.S., B.E.I.), Deparment of Pediatrics (L.A.P.), Department of Chemistry (L.C.), and Department of Medicine (T.H.L.), University of Virginia, Charlottesville
| | - Megan P Miller
- From the Department of Pharmacology and Chemical Biology (A.C.S.) and Heart, Lung, Blood and Vascular Medicine Institute (A.C.S., A.T.N., M.P.M.), University of Pittsburgh, PA; Robert M. Berne Cardiovascular Research Center (J.T.B., M.B., S.M.M., T.J., A.K.B., B.E.I.), Department of Molecular Physiology and Biophysics (M.B., M.V.A., A.V.S., B.E.I.), Deparment of Pediatrics (L.A.P.), Department of Chemistry (L.C.), and Department of Medicine (T.H.L.), University of Virginia, Charlottesville
| | - Lisa A Palmer
- From the Department of Pharmacology and Chemical Biology (A.C.S.) and Heart, Lung, Blood and Vascular Medicine Institute (A.C.S., A.T.N., M.P.M.), University of Pittsburgh, PA; Robert M. Berne Cardiovascular Research Center (J.T.B., M.B., S.M.M., T.J., A.K.B., B.E.I.), Department of Molecular Physiology and Biophysics (M.B., M.V.A., A.V.S., B.E.I.), Deparment of Pediatrics (L.A.P.), Department of Chemistry (L.C.), and Department of Medicine (T.H.L.), University of Virginia, Charlottesville
| | - Linda Columbus
- From the Department of Pharmacology and Chemical Biology (A.C.S.) and Heart, Lung, Blood and Vascular Medicine Institute (A.C.S., A.T.N., M.P.M.), University of Pittsburgh, PA; Robert M. Berne Cardiovascular Research Center (J.T.B., M.B., S.M.M., T.J., A.K.B., B.E.I.), Department of Molecular Physiology and Biophysics (M.B., M.V.A., A.V.S., B.E.I.), Deparment of Pediatrics (L.A.P.), Department of Chemistry (L.C.), and Department of Medicine (T.H.L.), University of Virginia, Charlottesville
| | - Avril V Somlyo
- From the Department of Pharmacology and Chemical Biology (A.C.S.) and Heart, Lung, Blood and Vascular Medicine Institute (A.C.S., A.T.N., M.P.M.), University of Pittsburgh, PA; Robert M. Berne Cardiovascular Research Center (J.T.B., M.B., S.M.M., T.J., A.K.B., B.E.I.), Department of Molecular Physiology and Biophysics (M.B., M.V.A., A.V.S., B.E.I.), Deparment of Pediatrics (L.A.P.), Department of Chemistry (L.C.), and Department of Medicine (T.H.L.), University of Virginia, Charlottesville
| | - Thu H Le
- From the Department of Pharmacology and Chemical Biology (A.C.S.) and Heart, Lung, Blood and Vascular Medicine Institute (A.C.S., A.T.N., M.P.M.), University of Pittsburgh, PA; Robert M. Berne Cardiovascular Research Center (J.T.B., M.B., S.M.M., T.J., A.K.B., B.E.I.), Department of Molecular Physiology and Biophysics (M.B., M.V.A., A.V.S., B.E.I.), Deparment of Pediatrics (L.A.P.), Department of Chemistry (L.C.), and Department of Medicine (T.H.L.), University of Virginia, Charlottesville
| | - Brant E Isakson
- From the Department of Pharmacology and Chemical Biology (A.C.S.) and Heart, Lung, Blood and Vascular Medicine Institute (A.C.S., A.T.N., M.P.M.), University of Pittsburgh, PA; Robert M. Berne Cardiovascular Research Center (J.T.B., M.B., S.M.M., T.J., A.K.B., B.E.I.), Department of Molecular Physiology and Biophysics (M.B., M.V.A., A.V.S., B.E.I.), Deparment of Pediatrics (L.A.P.), Department of Chemistry (L.C.), and Department of Medicine (T.H.L.), University of Virginia, Charlottesville.
| |
Collapse
|
40
|
Renshall LJ, Dilworth MR, Greenwood SL, Sibley CP, Wareing M. In vitro assessment of mouse fetal abdominal aortic vascular function. Am J Physiol Regul Integr Comp Physiol 2014; 307:R746-54. [PMID: 25056105 PMCID: PMC4166756 DOI: 10.1152/ajpregu.00058.2014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Fetal growth restriction (FGR) affects 3–8% of human pregnancies. Mouse models have provided important etiological data on FGR; they permit the assessment of treatment strategies on the physiological function of both mother and her developing offspring. Our study aimed to 1) develop a method to assess vascular function in fetal mice and 2) as a proof of principle ascertain whether a high dose of sildenafil citrate (SC; Viagra) administered to the pregnant dam affected fetal vascular reactivity. We developed a wire myography methodology for evaluation of fetal vascular function in vitro using the placenta-specific insulin-like growth factor II (Igf2) knockout mouse (P0; a model of FGR). Vascular function was determined in abdominal aortas isolated from P0 and wild-type (WT) fetuses at embryonic day (E) 18.5 of gestation. A subset of dams received SC 0.8 mg/ml via drinking water from E12.5; data were compared with water-only controls. Using wire myography, we found that fetal aortic rings exhibited significant agonist-induced contraction, and endothelium-dependent and endothelium-independent relaxation. Sex-specific alterations in reactivity were noted in both strains. Maternal treatment with SC significantly attenuated endothelium-dependent and endothelium-independent relaxation of fetal aortic rings. Mouse fetal abdominal aortas reproducibly respond to vasoactive agents. Study of these vessels in mouse genetic models of pregnancy complications may 1) help to delineate early signs of abnormal vascular reactivity and 2) inform whether treatments given to the mother during pregnancy may impact upon fetal vascular function.
Collapse
Affiliation(s)
- Lewis J Renshall
- Maternal and Fetal Health Research Centre, Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, Manchester, United Kingdom; and St. Mary's Hospital, Central Manchester University Hospitals National Health Service Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Mark R Dilworth
- Maternal and Fetal Health Research Centre, Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, Manchester, United Kingdom; and St. Mary's Hospital, Central Manchester University Hospitals National Health Service Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Susan L Greenwood
- Maternal and Fetal Health Research Centre, Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, Manchester, United Kingdom; and St. Mary's Hospital, Central Manchester University Hospitals National Health Service Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Colin P Sibley
- Maternal and Fetal Health Research Centre, Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, Manchester, United Kingdom; and St. Mary's Hospital, Central Manchester University Hospitals National Health Service Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Mark Wareing
- Maternal and Fetal Health Research Centre, Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, Manchester, United Kingdom; and St. Mary's Hospital, Central Manchester University Hospitals National Health Service Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| |
Collapse
|
41
|
Geoffrion M, Du X, Irshad Z, Vanderhyden BC, Courville K, Sui G, D'Agati VD, Ott-Braschi S, Rabbani N, Thornalley PJ, Brownlee M, Milne RW. Differential effects of glyoxalase 1 overexpression on diabetic atherosclerosis and renal dysfunction in streptozotocin-treated, apolipoprotein E-deficient mice. Physiol Rep 2014; 2:2/6/e12043. [PMID: 24920125 PMCID: PMC4208644 DOI: 10.14814/phy2.12043] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The reactive dicarbonyls, glyoxal and methylglyoxal (MG), increase in diabetes and may participate in the development of diabetic complications. Glyoxal and MG are detoxified by the sequential activities of glyoxalase 1 (GLO1) and glyoxalase 2. To determine the contribution of these dicarbonyls to the etiology of complications, we have genetically manipulated GLO1 levels in apolipoprotein E‐null (Apoe−/−) mice. Male Apoe−/− mice, hemizygous for a human GLO1 transgene (GLO1TGApoe−/− mice) or male nontransgenic Apoe−/− litter mates were injected with streptozotocin or vehicle and 6 or 20 weeks later, aortic atherosclerosis was quantified. The GLO1 transgene lessened streptozotocin (STZ)‐induced increases in immunoreactive hydroimidazolone (MG‐H1). Compared to nondiabetic mice, STZ‐treated GLO1TGApoe−/− and Apoe−/− mice had increased serum cholesterol and triglycerides and increased atherosclerosis at both times after diabetes induction. While the increased GLO1 activity in the GLO1TGApoe−/− mice failed to protect against diabetic atherosclerosis, it lessened glomerular mesangial expansion, prevented albuminuria and lowered renal levels of dicarbonyls and protein glycation adducts. Aortic atherosclerosis was also quantified in 22‐week‐old, male normoglycemic Glo1 knockdown mice on an Apoe−/− background (Glo1KDApoe−/− mice), an age at which Glo1KD mice exhibit albuminuria and renal pathology similar to that of diabetic mice. In spite of ~75% decrease in GLO1 activity and increased aortic MG‐H1, the Glo1KDApoe−/− mice did not show increased atherosclerosis compared to age‐matched Apoe−/− mice. Thus, manipulation of GLO1 activity does not affect the development of early aortic atherosclerosis in Apoe−/− mice but can dictate the onset of kidney disease independently of blood glucose levels. Increased levels of methylglyoxal and methylglyoxal‐derived advanced glycation end products may contribute to the development of diabetic complications. We show that overexpression of an enzyme that participates in the pathway of methylglyoxal detoxification, glyoxalase 1, protects streptozotocin‐treated, apolipoprotein E‐deficient mice from diabetic kidney disease but not from diabetes‐induced accelerated aortic atherosclerosis.
Collapse
Affiliation(s)
- Michèle Geoffrion
- Atherosclerosis, Genetics and Cell Biology Group, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Xueliang Du
- Diabetes Research Center, Albert Einstein College of Medicine, Bronx, New York
| | - Zehra Irshad
- Clinical Sciences Research Laboratories, Warwick Medical School, University of Warwick, Coventry, U.K
| | - Barbara C Vanderhyden
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Kerri Courville
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Guangzhi Sui
- Diabetes Research Center, Albert Einstein College of Medicine, Bronx, New York
| | | | - Sylvie Ott-Braschi
- Atherosclerosis, Genetics and Cell Biology Group, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Naila Rabbani
- Clinical Sciences Research Laboratories, Warwick Medical School, University of Warwick, Coventry, U.K
| | - Paul J Thornalley
- Clinical Sciences Research Laboratories, Warwick Medical School, University of Warwick, Coventry, U.K
| | - Michael Brownlee
- Diabetes Research Center, Albert Einstein College of Medicine, Bronx, New York
| | - Ross W Milne
- Atherosclerosis, Genetics and Cell Biology Group, University of Ottawa Heart Institute, Ottawa, Ontario, Canada Department of Pathology and Laboratory Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
42
|
Kamezaki F, Tsutsui M, Takahashi M, Sonoda S, Kubo T, Fujino Y, Adachi T, Abe H, Takeuchi M, Mayumi T, Otsuji Y. Plasma levels of nitric oxide metabolites are markedly reduced in normotensive men with electrocardiographically determined left ventricular hypertrophy. Hypertension 2014; 64:516-22. [PMID: 24914203 DOI: 10.1161/hypertensionaha.114.03287] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Recent studies have revealed that electrocardiographically determined left ventricular hypertrophy (ECG-LVH) is a risk factor for cardiovascular death not only in hypertensive patients but also in normotensive subjects. However, the underlying mechanisms remain to be elucidated. In this study, we tested our hypothesis that normotensive subjects with ECG-LVH have reduced nitric oxide production. A total of 840 Japanese male workers were enrolled, and 579 eligible subjects were studied. ECG-LVH was assessed according to the Sokolow-Lyon voltage criteria and the Cornell voltage-duration product. The median level of plasma NOx (nitrite plus nitrate), a marker of systemic nitric oxide production, was markedly lower in the normotensive subjects with ECG-LVH (n=73) than in those without (n=506), and the clinical characteristics were significantly different between the 2 groups (each P<0.05). Importantly, a one-to-one propensity score matching analysis showed similar markedly lower median plasma NOx level in the normotensive subjects with ECG-LVH compared with that observed in the matched normotensive subjects without ECG-LVH (P<0.05). Furthermore, the tertiles of the plasma NOx levels were inversely correlated with the prevalence and severity of ECG-LVH (both P<0.05). The lower plasma NOx levels were associated with significantly higher plasma 8-isoprostane levels, a marker of systemic lipid peroxidation (P<0.05). These results provide the first evidence that normotensive subjects with ECG-LVH exhibit defective nitric oxide production, along with increased oxidative stress. Our findings may thus explain, at least in part, a potential mechanism underlying the increased risk of cardiovascular death in normotensive individuals with ECG-LVH.
Collapse
Affiliation(s)
- Fumihiko Kamezaki
- From the Department of Emergency Medicine (F.K., T.M.), Second Department of Internal Medicine (M. Takahashi, S.S., H.A., M. Takeuchi, Y.O.), and Department of Public Health (T.K., Y.F.), School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan; Department of Pharmacology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan (M. Tsutsui); and Laboratory of Clinical Pharmaceutics, Gifu Pharmaceutical University, Gifu, Japan (T.A.).
| | - Masato Tsutsui
- From the Department of Emergency Medicine (F.K., T.M.), Second Department of Internal Medicine (M. Takahashi, S.S., H.A., M. Takeuchi, Y.O.), and Department of Public Health (T.K., Y.F.), School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan; Department of Pharmacology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan (M. Tsutsui); and Laboratory of Clinical Pharmaceutics, Gifu Pharmaceutical University, Gifu, Japan (T.A.)
| | - Masao Takahashi
- From the Department of Emergency Medicine (F.K., T.M.), Second Department of Internal Medicine (M. Takahashi, S.S., H.A., M. Takeuchi, Y.O.), and Department of Public Health (T.K., Y.F.), School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan; Department of Pharmacology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan (M. Tsutsui); and Laboratory of Clinical Pharmaceutics, Gifu Pharmaceutical University, Gifu, Japan (T.A.)
| | - Shinjo Sonoda
- From the Department of Emergency Medicine (F.K., T.M.), Second Department of Internal Medicine (M. Takahashi, S.S., H.A., M. Takeuchi, Y.O.), and Department of Public Health (T.K., Y.F.), School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan; Department of Pharmacology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan (M. Tsutsui); and Laboratory of Clinical Pharmaceutics, Gifu Pharmaceutical University, Gifu, Japan (T.A.)
| | - Tatsuhiko Kubo
- From the Department of Emergency Medicine (F.K., T.M.), Second Department of Internal Medicine (M. Takahashi, S.S., H.A., M. Takeuchi, Y.O.), and Department of Public Health (T.K., Y.F.), School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan; Department of Pharmacology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan (M. Tsutsui); and Laboratory of Clinical Pharmaceutics, Gifu Pharmaceutical University, Gifu, Japan (T.A.)
| | - Yoshihisa Fujino
- From the Department of Emergency Medicine (F.K., T.M.), Second Department of Internal Medicine (M. Takahashi, S.S., H.A., M. Takeuchi, Y.O.), and Department of Public Health (T.K., Y.F.), School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan; Department of Pharmacology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan (M. Tsutsui); and Laboratory of Clinical Pharmaceutics, Gifu Pharmaceutical University, Gifu, Japan (T.A.)
| | - Tetsuo Adachi
- From the Department of Emergency Medicine (F.K., T.M.), Second Department of Internal Medicine (M. Takahashi, S.S., H.A., M. Takeuchi, Y.O.), and Department of Public Health (T.K., Y.F.), School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan; Department of Pharmacology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan (M. Tsutsui); and Laboratory of Clinical Pharmaceutics, Gifu Pharmaceutical University, Gifu, Japan (T.A.)
| | - Haruhiko Abe
- From the Department of Emergency Medicine (F.K., T.M.), Second Department of Internal Medicine (M. Takahashi, S.S., H.A., M. Takeuchi, Y.O.), and Department of Public Health (T.K., Y.F.), School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan; Department of Pharmacology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan (M. Tsutsui); and Laboratory of Clinical Pharmaceutics, Gifu Pharmaceutical University, Gifu, Japan (T.A.)
| | - Masaaki Takeuchi
- From the Department of Emergency Medicine (F.K., T.M.), Second Department of Internal Medicine (M. Takahashi, S.S., H.A., M. Takeuchi, Y.O.), and Department of Public Health (T.K., Y.F.), School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan; Department of Pharmacology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan (M. Tsutsui); and Laboratory of Clinical Pharmaceutics, Gifu Pharmaceutical University, Gifu, Japan (T.A.)
| | - Toshihiko Mayumi
- From the Department of Emergency Medicine (F.K., T.M.), Second Department of Internal Medicine (M. Takahashi, S.S., H.A., M. Takeuchi, Y.O.), and Department of Public Health (T.K., Y.F.), School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan; Department of Pharmacology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan (M. Tsutsui); and Laboratory of Clinical Pharmaceutics, Gifu Pharmaceutical University, Gifu, Japan (T.A.)
| | - Yutaka Otsuji
- From the Department of Emergency Medicine (F.K., T.M.), Second Department of Internal Medicine (M. Takahashi, S.S., H.A., M. Takeuchi, Y.O.), and Department of Public Health (T.K., Y.F.), School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan; Department of Pharmacology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan (M. Tsutsui); and Laboratory of Clinical Pharmaceutics, Gifu Pharmaceutical University, Gifu, Japan (T.A.)
| |
Collapse
|
43
|
Hermida N, Balligand JL. Low-density lipoprotein-cholesterol-induced endothelial dysfunction and oxidative stress: the role of statins. Antioxid Redox Signal 2014; 20:1216-37. [PMID: 23924077 DOI: 10.1089/ars.2013.5537] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
SIGNIFICANCE Cardiovascular diseases (CVD) represent a major public health burden. High low-density lipoprotein (LDL)-cholesterol is a recognized pathogenic factor for atherosclerosis, and its complications and statins represent the most potent and widely used therapeutic approach to prevent and control these disorders. RECENT ADVANCES A number of clinical and experimental studies concur to identify endothelial dysfunction as a primary step in the development of atherosclerosis, as well as a risk factor for subsequent clinical events. Oxidant stress resulting from chronic elevation of plasma LDL-cholesterol (LDL-chol) is a major contributor to both endothelial dysfunction and its complications, for example, through alterations of endothelial nitric oxide signaling. CRITICAL ISSUES Statin treatment reduces morbidity and mortality of CVD, but increasing evidence questions that this is exclusively through reduction of plasma LDL-chol. The identification of ancillary effects on (cardio)vascular biology, for example, through their modulation of oxidative stress, will not only increase our understanding of their mechanisms of action, with a potential broadening of their indication(s), but also lead to the identification of new molecular targets for future therapeutic developments in CVD. FUTURE DIRECTIONS Further characterization of molecular pathways targeted by statins, for example, not directly mediated by changes in plasma lipid concentrations, should enable a more comprehensive approach to the pathogenesis of (cardio)vascular disease, including, for example, epigenetic regulation and fine tuning of cell metabolism.
Collapse
Affiliation(s)
- Nerea Hermida
- 1 Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Experimentale et Clinique (IREC), Université catholique de Louvain , Brussels, Belgium
| | | |
Collapse
|
44
|
Gazi E, Temiz A, Altun B, Barutcu A, Silan F, Colkesen Y, Ozdemir O. Endothelial function and germ-line ACE I/D, eNOS and PAI-1 gene profiles in patients with coronary slow flow in the Canakkale population: multiple thrombophilic gene profiles in coronary slow flow. Cardiovasc J Afr 2014; 25:9-14. [PMID: 24435163 PMCID: PMC3959185 DOI: 10.5830/cvja-2013-083] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2011] [Accepted: 08/14/2013] [Indexed: 11/25/2022] Open
Abstract
Background We examined the effects of ACE, PAI-1 and eNOS gene polymorphisms on endothelial function. The genes are related to atherosclerosis and endothelial dysfunction in coronary slow flow (CSF). Methods Thirty-three patients with angiographically proven CSF and 48 subjects with normal coronary flow were enrolled in this study. Coronary flow patterns were determined by the thrombolysis in myocardial infarction (TIMI) frame count method. Endothelial function was assessed in the brachial artery by endothelium-dependent flow-mediated dilatation (FMD). PAI-1 4G/5G, eNOS T-786C and ACE I/D polymorphisms were determined by polymerase chain reaction (PCR) amplification. Results No difference was found between the groups regarding age, heart rate and blood pressure. Males were more prevalent among patients with CSF than control subjects (58.8 vs 29.8%, p = 0.009). Mean TIMI frame counts were significantly higher in CSF patients (24.2 ± 4.0 vs 13.1 ± 2.5 fpm, p = 0.001). FMD was significantly lower in CSF patients than in the controls (4.9 ± 6.6 vs 7.9 ± 5.6%, p = 0.029). TIMI frame count and FMD were found to be negatively correlated in a correlation analysis (r = –0.269, p = 0.015). PAI-1 4G/5G, eNOS T-786C and ACE I/D polymorphisms were similar in the two groups. Conclusions This study showed that endothelial function was impaired in patients with CSF. PAI-1, ACE and eNOS polymorphisms were not related to CSF in our study population.
Collapse
Affiliation(s)
- Emine Gazi
- Department of Cardiology, Faculty of Medicine, Canakkale Onsekiz mart University, Canakkale, Turkey
| | - Ahmet Temiz
- Department of Cardiology, Faculty of Medicine, Canakkale Onsekiz mart University, Canakkale, Turkey
| | - Burak Altun
- Department of Cardiology, Faculty of Medicine, Canakkale Onsekiz mart University, Canakkale, Turkey
| | - Ahmet Barutcu
- Department of Cardiology, Faculty of Medicine, Canakkale Onsekiz mart University, Canakkale, Turkey
| | - Fatma Silan
- Department of Medical Genetics, Faculty of Medicine, Canakkale Onsekiz Mart University, Canakkale, Turkey
| | - Yucel Colkesen
- Department of Cardiology, Faculty of Medicine, Canakkale Onsekiz mart University, Canakkale, Turkey
| | - Ozturk Ozdemir
- Department of Medical Genetics, Faculty of Medicine, Canakkale Onsekiz Mart University, Canakkale, Turkey
| |
Collapse
|
45
|
Galluccio E, Cassina L, Russo I, Gelmini F, Setola E, Rampoldi L, Citterio L, Rossodivita A, Kamami M, Colombo A, Alfieri O, Carini M, Bosi E, Trovati M, Piatti P, Monti LD, Casari G. A novel truncated form of eNOS associates with altered vascular function. Cardiovasc Res 2013; 101:492-502. [DOI: 10.1093/cvr/cvt267] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
46
|
Abstract
Pulmonary hypertension in human patients can result from increased pulmonary vascular tone, pressure transferred from the systemic circulation, dropout of small pulmonary vessels, occlusion of vessels with thrombi or intimal lesions, or some combination of all of these. Different animal models have been designed to reflect these different mechanistic origins of disease. Pulmonary hypertension models may be roughly grouped into tone-related models, inflammation-related models, and genetic models with unusual or mixed mechanism. Models of tone generally use hypoxia as a base, and then modify this with either genetic modifications (SOD, NOS, and caveolin) or with drugs (Sugen), although some genetic modifications of tone-related pathways can result in spontaneous pulmonary hypertension (Hph-1). Inflammation-related models can use either toxic chemicals (monocrotaline, bleomycin), live pathogens (stachybotrys, schistosomiasis), or genetic modifications (IL-6, VIP). Additional genetic models rely on alterations in metabolism (adiponectin), cell migration (S100A4), the serotonin pathway, or the BMP pathway. While each of these shares molecular and pathologic symptoms with different classes of human pulmonary hypertension, in most cases the molecular etiology of human pulmonary hypertension is unknown, and so the relationship between any model and human disease is unclear. There is thus no best animal model of pulmonary hypertension; instead, investigators must select the model most related to the specific pathology they are studying.
Collapse
Affiliation(s)
- James West
- Vanderbilt University Medical Center, Nashville, Tennessee, USA.
| | | |
Collapse
|
47
|
Shahabi P, Siest G, Visvikis-siest S. Influence of inflammation on cardiovascular protective effects of cytochrome P450 epoxygenase-derived epoxyeicosatrienoic acids. Drug Metab Rev 2013; 46:33-56. [DOI: 10.3109/03602532.2013.837916] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
48
|
Madamanchi NR, Runge MS. Redox signaling in cardiovascular health and disease. Free Radic Biol Med 2013; 61:473-501. [PMID: 23583330 PMCID: PMC3883979 DOI: 10.1016/j.freeradbiomed.2013.04.001] [Citation(s) in RCA: 149] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Revised: 03/05/2013] [Accepted: 04/02/2013] [Indexed: 02/07/2023]
Abstract
Spatiotemporal regulation of the activity of a vast array of intracellular proteins and signaling pathways by reactive oxygen species (ROS) governs normal cardiovascular function. However, data from experimental and animal studies strongly support that dysregulated redox signaling, resulting from hyperactivation of various cellular oxidases or mitochondrial dysfunction, is integral to the pathogenesis and progression of cardiovascular disease (CVD). In this review, we address how redox signaling modulates the protein function, the various sources of increased oxidative stress in CVD, and the labyrinth of redox-sensitive molecular mechanisms involved in the development of atherosclerosis, hypertension, cardiac hypertrophy and heart failure, and ischemia-reperfusion injury. Advances in redox biology and pharmacology for inhibiting ROS production in specific cell types and subcellular organelles combined with the development of nanotechnology-based new in vivo imaging systems and targeted drug delivery mechanisms may enable fine-tuning of redox signaling for the treatment and prevention of CVD.
Collapse
Affiliation(s)
- Nageswara R Madamanchi
- McAllister Heart Institute, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Marschall S Runge
- McAllister Heart Institute, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
49
|
Genetic deletion of aquaporin-1 results in microcardia and low blood pressure in mouse with intact nitric oxide-dependent relaxation, but enhanced prostanoids-dependent relaxation. Pflugers Arch 2013; 466:237-51. [PMID: 23873354 DOI: 10.1007/s00424-013-1325-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2013] [Revised: 06/27/2013] [Accepted: 06/28/2013] [Indexed: 12/21/2022]
Abstract
The water channels, aquaporins (AQPs) are key mediators of transcellular fluid transport. However, their expression and role in cardiac tissue is poorly characterized. Particularly, AQP1 was suggested to transport other molecules (nitric oxide (NO), hydrogen peroxide (H2O2)) with potential major bearing on cardiovascular physiology. We therefore examined the expression of all AQPs and the phenotype of AQP1 knockout mice (vs. wild-type littermates) under implanted telemetry in vivo, as well as endothelium-dependent relaxation in isolated aortas and resistance vessels ex vivo. Four aquaporins were expressed in wild-type heart tissue (AQP1, AQP7, AQP4, AQP8) and two aquaporins in aortic and mesenteric vessels (AQP1-AQP7). AQP1 was expressed in endothelial as well as cardiac and vascular muscle cells and co-segregated with caveolin-1. AQP1 knockout (KO) mice exhibited a prominent microcardia and decreased myocyte transverse dimensions despite no change in capillary density. Both male and female AQP1 KO mice had lower mean BP, which was not attributable to altered water balance or autonomic dysfunction (from baroreflex and frequency analysis of BP and HR variability). NO-dependent BP variability was unperturbed. Accordingly, endothelium-derived hyperpolarizing factor (EDH(F)) or NO-dependent relaxation were unchanged in aorta or resistance vessels ex vivo. However, AQP1 KO mesenteric vessels exhibited an increase in endothelial prostanoids-dependent relaxation, together with increased expression of COX-2. This enhanced relaxation was abrogated by COX inhibition. We conclude that AQP1 does not regulate the endothelial EDH or NO-dependent relaxation ex vivo or in vivo, but its deletion decreases baseline BP together with increased prostanoids-dependent relaxation in resistance vessels. Strikingly, this was associated with microcardia, unrelated to perturbed angiogenesis. This may raise interest for new inhibitors of AQP1 and their use to treat hypertrophic cardiac remodeling.
Collapse
|
50
|
Kelsey L, Flenniken AM, Qu D, Funnell APW, Pearson R, Zhou YQ, Voronina I, Berberovic Z, Wood G, Newbigging S, Weiss ES, Wong M, Quach I, Yeh SYS, Deshwar AR, Scott IC, McKerlie C, Henkelman M, Backx P, Simpson J, Osborne L, Rossant J, Crossley M, Bruneau B, Adamson SL. ENU-induced mutation in the DNA-binding domain of KLF3 reveals important roles for KLF3 in cardiovascular development and function in mice. PLoS Genet 2013; 9:e1003612. [PMID: 23874215 PMCID: PMC3708807 DOI: 10.1371/journal.pgen.1003612] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Accepted: 05/22/2013] [Indexed: 12/23/2022] Open
Abstract
KLF3 is a Krüppel family zinc finger transcription factor with widespread tissue expression and no previously known role in heart development. In a screen for dominant mutations affecting cardiovascular function in N-ethyl-N-nitrosourea (ENU) mutagenized mice, we identified a missense mutation in the Klf3 gene that caused aortic valvular stenosis and partially penetrant perinatal lethality in heterozygotes. All homozygotes died as embryos. In the first of three zinc fingers, a point mutation changed a highly conserved histidine at amino acid 275 to arginine (Klf3H275R). This change impaired binding of the mutant protein to KLF3's canonical DNA binding sequence. Heterozygous Klf3H275R mutants that died as neonates had marked biventricular cardiac hypertrophy with diminished cardiac chambers. Adult survivors exhibited hypotension, cardiac hypertrophy with enlarged cardiac chambers, and aortic valvular stenosis. A dominant negative effect on protein function was inferred by the similarity in phenotype between heterozygous Klf3H275R mutants and homozygous Klf3 null mice. However, the existence of divergent traits suggested the involvement of additional interactions. We conclude that KLF3 plays diverse and important roles in cardiovascular development and function in mice, and that amino acid 275 is critical for normal KLF3 protein function. Future exploration of the KLF3 pathway provides a new avenue for investigating causative factors contributing to cardiovascular disorders in humans. Cardiac defects are among the most common malformations in humans. Most causative genetic mutations remain unknown. To discover new causative genes important in cardiovascular development and function, we examined 1770 mice with randomly mutated genes and found a mutant with aortic valvular stenosis, and increased risk of fetal and neonatal death. Using linkage analysis and sequencing, we identified a protein-altering point mutation in the gene regulatory protein KLF3. Mice that survived into adulthood with one mutant copy of the Klf3 gene had low arterial blood pressure, enlarged hearts, and increased mortality due to heart failure. When both copies of the Klf3 gene was mutant, then embryos had heart defects, and all died before birth. KLF3 had no previously known role in heart development so to confirm these findings, we (1) knocked down klf3 expression in zebrafish embryos and (2) examined mice with a mutation that effectively eliminated the KLF3 protein. In both cases, cardiovascular dysfunction was observed. In conclusion, we have discovered that KLF3 plays diverse and important roles in cardiovascular development and function in mice. Future exploration of the KLF3 pathway provides a new avenue for investigating causative factors contributing to cardiovascular disorders in humans.
Collapse
Affiliation(s)
- Lois Kelsey
- Samuel Lunenfeld Research Institute of Mount Sinai Hospital, Toronto, Ontario, Canada
- Centre for Modeling Human Disease, Toronto Centre for Phenogenomics, Toronto, Ontario, Canada
| | - Ann M. Flenniken
- Samuel Lunenfeld Research Institute of Mount Sinai Hospital, Toronto, Ontario, Canada
- Centre for Modeling Human Disease, Toronto Centre for Phenogenomics, Toronto, Ontario, Canada
| | - Dawei Qu
- Samuel Lunenfeld Research Institute of Mount Sinai Hospital, Toronto, Ontario, Canada
- Centre for Modeling Human Disease, Toronto Centre for Phenogenomics, Toronto, Ontario, Canada
| | - Alister P. W. Funnell
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Richard Pearson
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Yu-Qing Zhou
- Mouse Imaging Centre, Toronto Centre for Phenogenomics, Toronto, Ontario, Canada
- The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Irina Voronina
- Samuel Lunenfeld Research Institute of Mount Sinai Hospital, Toronto, Ontario, Canada
- Centre for Modeling Human Disease, Toronto Centre for Phenogenomics, Toronto, Ontario, Canada
| | - Zorana Berberovic
- Samuel Lunenfeld Research Institute of Mount Sinai Hospital, Toronto, Ontario, Canada
- Centre for Modeling Human Disease, Toronto Centre for Phenogenomics, Toronto, Ontario, Canada
| | - Geoffrey Wood
- Centre for Modeling Human Disease, Toronto Centre for Phenogenomics, Toronto, Ontario, Canada
- The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Susan Newbigging
- Centre for Modeling Human Disease, Toronto Centre for Phenogenomics, Toronto, Ontario, Canada
- The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Edward S. Weiss
- Centre for Modeling Human Disease, Toronto Centre for Phenogenomics, Toronto, Ontario, Canada
| | - Michael Wong
- Mouse Imaging Centre, Toronto Centre for Phenogenomics, Toronto, Ontario, Canada
- The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Ivan Quach
- Samuel Lunenfeld Research Institute of Mount Sinai Hospital, Toronto, Ontario, Canada
| | - S. Y. Sandy Yeh
- Samuel Lunenfeld Research Institute of Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Ashish R. Deshwar
- The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Ian C. Scott
- The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Heart and Stroke Richard Lewar Centre of Excellence, Toronto, Ontario, Canada
| | - Colin McKerlie
- Centre for Modeling Human Disease, Toronto Centre for Phenogenomics, Toronto, Ontario, Canada
- The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Mark Henkelman
- Mouse Imaging Centre, Toronto Centre for Phenogenomics, Toronto, Ontario, Canada
- The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Peter Backx
- Heart and Stroke Richard Lewar Centre of Excellence, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Jeremy Simpson
- Heart and Stroke Richard Lewar Centre of Excellence, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Lucy Osborne
- Centre for Modeling Human Disease, Toronto Centre for Phenogenomics, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Janet Rossant
- Centre for Modeling Human Disease, Toronto Centre for Phenogenomics, Toronto, Ontario, Canada
- The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Merlin Crossley
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Benoit Bruneau
- Gladstone Institute of Cardiovascular Disease, Department of Pediatrics, and Cardiovascular Research Institute, University of California, San Francisco, California, United States of America
| | - S. Lee Adamson
- Samuel Lunenfeld Research Institute of Mount Sinai Hospital, Toronto, Ontario, Canada
- Centre for Modeling Human Disease, Toronto Centre for Phenogenomics, Toronto, Ontario, Canada
- Heart and Stroke Richard Lewar Centre of Excellence, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Department of Obstetrics and Gynaecology, University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|