1
|
Zuo Y, Li B, Gao M, Xiong R, He R, Li N, Geng Q. Novel insights and new therapeutic potentials for macrophages in pulmonary hypertension. Respir Res 2024; 25:147. [PMID: 38555425 PMCID: PMC10981837 DOI: 10.1186/s12931-024-02772-8] [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] [Received: 07/23/2023] [Accepted: 03/13/2024] [Indexed: 04/02/2024] Open
Abstract
Inflammation and immune processes underlie pulmonary hypertension progression. Two main different activated phenotypes of macrophages, classically activated M1 macrophages and alternatively activated M2 macrophages, are both involved in inflammatory processes related to pulmonary hypertension. Recent advances suggest that macrophages coordinate interactions among different proinflammatory and anti-inflammatory mediators, and other cellular components such as smooth muscle cells and fibroblasts. In this review, we summarize the current literature on the role of macrophages in the pathogenesis of pulmonary hypertension, including the origin of pulmonary macrophages and their response to triggers of pulmonary hypertension. We then discuss the interactions among macrophages, cytokines, and vascular adventitial fibroblasts in pulmonary hypertension, as well as the potential therapeutic benefits of macrophages in this disease. Identifying the critical role of macrophages in pulmonary hypertension will contribute to a comprehensive understanding of this pathophysiological abnormality, and may provide new perspectives for pulmonary hypertension management.
Collapse
Affiliation(s)
- Yifan Zuo
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Boyang Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Minglang Gao
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Rui Xiong
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Ruyuan He
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Ning Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| |
Collapse
|
2
|
Figueroa M, Hall S, Mattia V, Mendoza A, Brown A, Xiong Y, Mukherjee R, Jones JA, Richardson W, Ruddy JM. Vascular smooth muscle cell mechanotransduction through serum and glucocorticoid inducible kinase-1 promotes interleukin-6 production and macrophage accumulation in murine hypertension. JVS Vasc Sci 2023; 4:100124. [PMID: 37920479 PMCID: PMC10618507 DOI: 10.1016/j.jvssci.2023.100124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 08/01/2023] [Indexed: 11/04/2023] Open
Abstract
Objective The objective of this investigation was to demonstrate that in vivo induction of hypertension (HTN) and in vitro cyclic stretch of aortic vascular smooth muscle cells (VSMCs) can cause serum and glucocorticoid-inducible kinase (SGK-1)-dependent production of cytokines to promote macrophage accumulation that may promote vascular pathology. Methods HTN was induced in C57Bl/6 mice with angiotensin II infusion (1.46 mg/kg/day × 21 days) with or without systemic infusion of EMD638683 (2.5 mg/kg/day × 21 days), a selective SGK-1 inhibitor. Systolic blood pressure was recorded. Abdominal aortas were harvested to quantify SGK-1 activity (pSGK-1/SGK-1) by immunoblot. Flow cytometry quantified the abundance of CD11b+/F480+ cells (macrophages). Plasma interleukin (IL)-6 and monocyte chemoattractant protein-1 (MCP-1) was assessed by enzyme-linked immunosorbent assay. Aortic VSMCs from wild-type mice were subjected to 12% biaxial cyclic stretch (Stretch) for 3 or 12 hours with or without EMD638683 (10 μM) and with or without SGK-1 small interfering RNA with subsequent quantitative polymerase chain reaction for IL-6 and MCP-1 expression. IL-6 and MCP-1 in culture media were analyzed by enzyme-linked immunosorbent assay. Aortic VSMCs from SGK-1flox+/+ mice were transfected with Cre-Adenovirus to knockdown SGK-1 (SGK-1KD VSMCs) and underwent parallel tension experimentation. Computational modeling was used to simulate VSMC signaling. Statistical analysis included analysis of variance with significance at a P value of <.05. Results SGK-1 activity, abundance of CD11b+/F4-80+ cells, and plasma IL-6 were increased in the abdominal aorta of mice with HTN and significantly reduced by treatment with EMD638683. This outcome mirrored the increased abundance of IL-6 in media from Stretch C57Bl/6 VSMCs and attenuation of the effect with EMD638683 or SGK-1 small interfering RNA. C57Bl/6 VSMCs also responded to Stretch with increased MCP-1 expression and secretion into the culture media. Further supporting the integral role of mechanical signaling through SGK-1, target gene expression and cytokine secretion was unchanged in SGK-1KD VSMCs with Stretch, and computer modeling confirmed SGK-1 as an intersecting node of signaling owing to mechanical strain and angiotensin II. Conclusions Mechanical activation of SGK-1 in aortic VSMCs can promote inflammatory signaling and increased macrophage abundance, therefore this kinase warrants further exploration as a pharmacotherapeutic target to abrogate hypertensive vascular pathology.
Collapse
Affiliation(s)
- Mario Figueroa
- Division of Vascular Surgery, Medical University of South Carolina, Charleston, SC
| | - SarahRose Hall
- Division of Vascular Surgery, Medical University of South Carolina, Charleston, SC
| | - Victoria Mattia
- Division of Vascular Surgery, Medical University of South Carolina, Charleston, SC
| | - Alex Mendoza
- Division of Vascular Surgery, Medical University of South Carolina, Charleston, SC
| | - Adam Brown
- Division of Vascular Surgery, Medical University of South Carolina, Charleston, SC
| | - Ying Xiong
- Division of Cardiothoracic Surgery, Medical University of South Carolina, Charleston, SC
| | - Rupak Mukherjee
- Division of Cardiothoracic Surgery, Medical University of South Carolina, Charleston, SC
| | - Jeffrey A. Jones
- Division of Cardiothoracic Surgery, Medical University of South Carolina, Charleston, SC
- Ralph H. Johnson VA Medical Center, Charleston, SC
| | - William Richardson
- Department of Chemical Engineering, University of Arkansas, Fayetteville, AK
| | - Jean Marie Ruddy
- Division of Vascular Surgery, Medical University of South Carolina, Charleston, SC
- Ralph H. Johnson VA Medical Center, Charleston, SC
| |
Collapse
|
3
|
Inhibition of Serum- and Glucocorticoid-Regulated Protein Kinase-1 Aggravates Imiquimod-Induced Psoriatic Dermatitis and Enhances Proinflammatory Cytokine Expression through the NF-kB Pathway. J Invest Dermatol 2023; 143:954-964. [PMID: 36623704 DOI: 10.1016/j.jid.2022.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 11/16/2022] [Accepted: 12/20/2022] [Indexed: 01/09/2023]
Abstract
Although the anti-inflammatory effect of serum- and glucocorticoid-regulated protein kinase 1 (SGK1) has been established in other diseases, the possible regulatory role of SGK1 in psoriasis and the underlying molecular mechanisms remain largely unknown. In this study, we found that SGK1 expression was decreased in macrophages from patients with psoriasis. Moreover, a specific pharmacological SGK1 inhibitor, EMD638683, significantly enhanced imiquimod-mediated toll-like receptor 7/8 activity and proinflammatory cytokine production in RAW264.7 cells, and this result was confirmed by Sgk1 small interfering RNA. Further mechanistic data showed that SGK1 inhibition increased the phosphorylation of Bruton's agammaglobulinemia tyrosine kinase; moreover, Bruton's agammaglobulinemia tyrosine kinase inhibition abrogated the proinflammatory effects of the SGK1 inhibitor on toll-like receptor 7/8 activation, thereby validating that SGK1 inhibition enhances the toll-like receptor 7/8 pathway by increasing Bruton's agammaglobulinemia tyrosine kinase phosphorylation. In addition, our in vivo results showed that SGK1 inhibition significantly increased the secretion of proinflammatory cytokines, including IL-1β, IL-6, and TNF-α, and the infiltration of T helper 17 cells in an imiquimod-induced psoriasis mouse model. Altogether, these results show that SGK1 plays a critical role in the pathogenesis of psoriasis by modulating inflammatory responses in skin lesions, indicating that SGK1‒Bruton's agammaglobulinemia tyrosine kinase signaling could be a novel therapeutic target for the control of psoriasis.
Collapse
|
4
|
Lu RQ, Zhang YY, Zhao HQ, Guo RQ, Jiang ZX, Guo R. SGK1, a Critical Regulator of Immune Modulation and Fibrosis and a Potential Therapeutic Target in Chronic Graft-Versus-Host Disease. Front Immunol 2022; 13:822303. [PMID: 35222400 PMCID: PMC8866649 DOI: 10.3389/fimmu.2022.822303] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 01/25/2022] [Indexed: 11/24/2022] Open
Abstract
Patients with severe chronic graft-versus-host disease (cGVHD) always experience debilitating tissue injury and have poorer quality of life and shorter survival time. The early stage of cGVHD is characterized by inflammation, which eventually leads to extensive tissue fibrosis in various organs, such as skin and lung, eventually inducing scleroderma-like changes and bronchiolitis obliterans syndrome. Here we review the functions of serum/glucocorticoid regulated kinase 1 (SGK1), a hub molecule in multiple signal transduction pathways and cell phosphorylation cascades, which has important roles in cell proliferation and ion channel regulation, and its relevance in cGVHD. SGK1 phosphorylates the ubiquitin ligase, NEDD4, and induces Th cells to differentiate into Th17 and Th2 phenotypes, hinders Treg development, and promotes inflammatory fibrosis. Phosphorylation of NEDD4 by SGK1 also leads to up-regulation of the transcription factor SMAD2/3, thereby amplifying the fibrosis-promoting effect of TGF-β. SGK1 also up-regulates the inflammatory transcription factor, nuclear factor-κB (NF-κB), which in turn stimulates the expression of multiple inflammatory mediators, including connective tissue growth factor. Overexpression of SGK1 has been observed in various fibrotic diseases, including pulmonary fibrosis, diabetic renal fibrosis, liver cirrhosis, hypertensive cardiac fibrosis, peritoneal fibrosis, and Crohn’s disease. In addition, SGK1 inhibitors can attenuate, or even reverse, the effect of fibrosis, and may be used to treat inflammatory conditions and/or fibrotic diseases, such as cGVHD, in the future.
Collapse
Affiliation(s)
- Run-Qing Lu
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yin-Yin Zhang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hai-Qiu Zhao
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Rong-Qun Guo
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhong-Xing Jiang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Rong Guo
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
5
|
The role of immune cells in pulmonary hypertension: Focusing on macrophages. Hum Immunol 2021; 83:153-163. [PMID: 34844784 DOI: 10.1016/j.humimm.2021.11.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 11/14/2021] [Accepted: 11/15/2021] [Indexed: 01/06/2023]
Abstract
Pulmonary hypertension (PH) is a life-threatening pathological state with elevated pulmonary arterial pressure, resulting in right ventricular failure and heart functional failure. Analyses of human samples and rodent models of pH support the infiltration of various immune cells, including neutrophils, mast cells, dendritic cells, B-cells, T-cells, and natural killer cells, to the lungs and pulmonary perivascular regions and their involvement in the PH development. There is evidence that macrophages are presented in the pulmonary lesions of pH patients as first-line myeloid leucocytes. Macrophage accumulation and presence, both M1 and M2 phenotypes, is a distinctive hallmark of pH which plays a pivotal role in pulmonary artery remodeling through various cellular and molecular interactions and mechanisms, including CCL2 and CX3CL1 chemokines, adventitial fibroblasts, glucocorticoid-regulated kinase 1 (SGK1), crosstalk with other immune cells, leukotriene B4 (LTB4), bone morphogenetic protein receptor 2 (BMPR2), macrophage migration inhibitory factor (MIF), and thrombospondin-1 (TSP-1). In this paper, we reviewed the molecular mechanisms and the role of immune cells and responses are involved in PH development. We also summarized the polarization of macrophages in response to different stimuli and their pathological role and their infiltration in the lung of pH patients and animal models.
Collapse
|
6
|
Chen S, Yan D, Qiu A. The role of macrophages in pulmonary hypertension: Pathogenesis and targeting. Int Immunopharmacol 2020; 88:106934. [PMID: 32889242 DOI: 10.1016/j.intimp.2020.106934] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 08/22/2020] [Accepted: 08/22/2020] [Indexed: 12/13/2022]
Abstract
Pulmonary hypertension (PH) is a pathophysiological disorder that can complicate most cardiovascular and respiratory diseases and may involve multiple clinical conditions, but its pathogenesis is poorly understood. Despite recent developments in the management of PH, there is an urgent need for new ways to effectively treat PH and reduce the risk of further complications. Recent studies have shown that dysregulated immunity underlies the development of PH. Myeloid cells, including monocytes and macrophages, participate in immune homeostasis and the adaptive immune response, but the function and production of these cells in PH is not well understood. A prominent pathological feature of pH is the accumulation of macrophages near the arterioles of the lung, indicating that pulmonary inflammation mediated by lung perivascular macrophages is a key driver of pulmonary remodelling, which leads to increased right ventricular systolic pressure. An improved understanding of the roles macrophages play in immune responses associated with PH may lead to new therapeutic targets. In this review, we highlight the relationship between macrophages and PH, the molecular mechanisms involved, and the recent advances in targeting these processes to treat PH.
Collapse
Affiliation(s)
- Shanshan Chen
- Department of Respiratory and Critical Care Medicine, Yancheng Third People's Hospital, The Affiliated Yancheng Hospital of Southeast University Medical College, Jiangsu, China
| | - Dongmei Yan
- Department of Clinical Laboratory, Yancheng Third People's Hospital, The Affiliated Yancheng Hospital of Southeast University Medical College, Jiangsu, China
| | - Aimin Qiu
- Department of Respiratory and Critical Care Medicine, Yancheng Third People's Hospital, The Affiliated Yancheng Hospital of Southeast University Medical College, Jiangsu, China.
| |
Collapse
|
7
|
An increase in alveolar fluid clearance induced by hyperinsulinemia in obese rats with LPS-induced acute lung injury. Respir Physiol Neurobiol 2020; 279:103470. [PMID: 32474115 DOI: 10.1016/j.resp.2020.103470] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/07/2020] [Accepted: 05/25/2020] [Indexed: 01/11/2023]
Abstract
A lower mortality rate is observed in obese patients with acute lung injury (ALI), which is referred to as the obesity paradox, in several studies and recent meta-analyses. Hyperinsulinemia is characterized as the primary effect of obesity, and exogenous insulin attenuates LPS-induced pulmonary edema. The detailed mechanism responsible for the effect of hyperinsulinemia on pulmonary edema and alveolar filling needs to be elucidated. SD rats were fed with a high-fat diet (HFD) for a total of 14 weeks. SD rats were anesthetized and intraperitoneally injected with 10 mg/kg lipopolysaccharide (LPS), while control rats received only saline vehicle. Insulin receptor antagonist S961 (20 nmol/kg) was given by the tail vein and serum, and glucocorticoid-induced protein kinase-1 (SGK-1) inhibitor EMD638683 (20 mg/kg) was administrated intragastrically prior to LPS exposure. The lungs were isolated for the measurement of alveolar fluid clearance. The protein expression of epithelial sodium channel (ENaC) was detected by Western blot. Insulin level in serum was significantly higher in HFD rats compared with normal diet rats in the presence or absence of LPS pretreatment. Hyperinsulinemia induced by high fat feeding increased alveolar fluid clearance and the abundance of α-ENaC, β-ENaC, and γ-ENaC in both normal rats and ALI rats. Moreover, these effects were reversed in response to S961. EMD638683 prevented the simulation of alveolar fluid clearance and protein expression of ENaC in HFD rats with ALI. These findings suggest that hyperinsulinemia induced by obesity results in the stimulation of alveolar fluid clearance via the upregulation of the abundance of ENaC in clinical acute lung injury, whereas theses effects are prevented by an SGK-1 inhibitor.
Collapse
|
8
|
Maestro I, Boya P, Martinez A. Serum- and glucocorticoid-induced kinase 1, a new therapeutic target for autophagy modulation in chronic diseases. Expert Opin Ther Targets 2020; 24:231-243. [PMID: 32067528 DOI: 10.1080/14728222.2020.1730328] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Introduction: Autophagy, a basic cellular degradation pathway essential for survival, is altered both in aging and in many chronic human diseases, including infections, cancer, heart disease, and neurodegeneration. Identifying new therapeutic targets for the control and modulation of autophagy events is therefore of utmost importance in drug discovery. Serum and glucocorticoid activated kinase 1 (SGK1), known for decades for its role in ion channel modulation, is now known to act as a switch for autophagy homeostasis, and has emerged as a novel and important therapeutic target likely to attract considerable research attention in the coming years.Areas covered: In this general review of SGK1 we describe the kinase's structure and its roles in physiological and pathological contexts. We also discuss small-molecule modulators of SGK1 activity. These modulators are of particular interest to medicinal chemists and pharmacists seeking to develop more potent and selective drug candidates for SGK1, which, despite its key role in autophagy, remains relatively understudied.Expert opinion: The main future challenges in this area are (i) deciphering the role of SGK1 in selective autophagy processes (e.g. mitophagy, lipophagy, and aggrephagy); (ii) identifying selective allosteric modulators of SGK1 with specific biological functions; and (iii) conducting first-in-man clinical studies.
Collapse
Affiliation(s)
- Inés Maestro
- Centro de Investigaciones Biológicas Margarita Salas-CSIC, Madrid, Spain
| | - Patricia Boya
- Centro de Investigaciones Biológicas Margarita Salas-CSIC, Madrid, Spain
| | - Ana Martinez
- Centro de Investigaciones Biológicas Margarita Salas-CSIC, Madrid, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
9
|
Sun CC, Zhang LC, Gao CL, Zhang HR, Yu RL, Kang CM. Design and screening of SGK1, Src dual inhibitors using pharmacophore models, molecular docking, and molecular dynamics simulation. NEW J CHEM 2020. [DOI: 10.1039/d0nj02249g] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Serum and glucocorticoid-regulated protein kinase 1 that can promote the growth of tumor cells is highly expressed in many tumors. Sarcoma gene plays an important role in the pathogenesis of cancer and is an important kinase in tumor cell expression pathways.
Collapse
Affiliation(s)
- Chuan-ce Sun
- College of Chemical Engineering
- Qingdao University of Science and Technology
- Qingdao
- China
| | - Li-chuan Zhang
- College of Chemical Engineering
- Qingdao University of Science and Technology
- Qingdao
- China
| | - Cheng-long Gao
- College of Chemical Engineering
- Qingdao University of Science and Technology
- Qingdao
- China
| | - Hao-ran Zhang
- College of Chemical Engineering
- Qingdao University of Science and Technology
- Qingdao
- China
| | - Ri-lei Yu
- Key Laboratory of Marine Drugs
- Chinese Ministry of Education
- School of Medicine and Pharmacy
- Ocean University of China
- Qingdao
| | - Cong-min Kang
- College of Chemical Engineering
- Qingdao University of Science and Technology
- Qingdao
- China
| |
Collapse
|
10
|
Zhao M, Chen N, Li X, Lin L, Chen X. MiR-19a modulates hypoxia-mediated cell proliferation and migration via repressing PTEN in human pulmonary arterial smooth muscle. Life Sci 2019; 239:116928. [PMID: 31682848 DOI: 10.1016/j.lfs.2019.116928] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 09/23/2019] [Accepted: 10/01/2019] [Indexed: 12/16/2022]
Abstract
AIM The dysfunction of human pulmonary arterial smooth muscle cells (HPASMCs) has been suggested to participate in the pathophysiology of pulmonary arterial hypertension (PAH). This study determined miR-19a expression in hypoxia-induced HPASMCs and explored the mechanistic actions of miR-19a in hypoxia-induced HPASMC proliferation and migration. METHODS QRT-PCR and western blot assays respectively determined the mRNA and protein expression of miR-19a, phosphatase and tensin homolog (PTEN) and hypoxia-inducible factor-1 alpha (HIF-1α). In vitro functional assays determined HPASMC proliferation and migration, respectively. Luciferase reporter assay determined interaction between miR-19a and PTEN. The knockdown effects of miR-19a on PAH were confirmed in in vivo mice model. RESULTS Hypoxia treatment time-dependently up-regulated miR-19a expression and enhanced cell proliferation in HPASMCs. MiR-19a overexpression increased cell proliferation and migration of HPASMCs, while repression of miR-19a reduced cell proliferative and migratory potentials of hypoxia-treated HPASMCs. Bioinformatics analysis and luciferase reporter assay showed that PTEN 3' untranslated region was targeted by miR-19a, and miR-19a repressed the mRNA and protein expression of PTEN in HPASMCs. Further rescue studies revealed that miR-19a regulated proliferative and migratory potentials of hypoxia-treated HPASMCs via suppressing PTEN expression. In addition, HIF-1α was identified as one of the mediators for the hypoxia-induced aberrant expression levels of miR-19a and PTEN. MiR-19a overexpression enhanced PI3K/AKT signaling, which was attenuated by enforced expression of PTEN in HPASMCs. More importantly, knockdown of miR-19 attenuated the chronic hypoxia-induced PAH in in vivo mice model. CONCLUSION This study presented a novel mechanistic action of miR-19a-mediated cell proliferation and migration of HPASMCs.
Collapse
Affiliation(s)
- Mei Zhao
- Department of Pharmacy, Sanya Central Hospital (The Third People's Hospital of Hainan Province), Sanya City, Hainan Province, China.
| | - Ni Chen
- Department of Pharmacy, The Second Affiliated Hospital of Hainan Medical University, Haikou City, Hainan Province, China.
| | - Xuelian Li
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin City, Heilongjiang Province, China.
| | - Ling Lin
- Department of Cardiovascular Medicine, Sanya Central Hospital (The Third People's Hospital of Hainan Province), Sanya City, Hainan Province, China.
| | - Xin Chen
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin City, Heilongjiang Province, China.
| |
Collapse
|
11
|
Huang W, Cheng C, Shan W, Ding Z, Liu F, Lu W, He W, Xu J, Yin Z. Knockdown of SGK1 alleviates the IL‐1β‐induced chondrocyte anabolic and catabolic imbalance by activating FoxO1‐mediated autophagy in human chondrocytes. FEBS J 2019; 287:94-107. [PMID: 31330080 DOI: 10.1111/febs.15009] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 05/11/2019] [Accepted: 07/19/2019] [Indexed: 12/17/2022]
Affiliation(s)
- Wei Huang
- Department of Orthopaedics The First Affiliated Hospital of Anhui Medical University Hefei China
- Department of Orthopaedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine University of Science and Technology of China Hefei China
| | - Chao Cheng
- Department of Orthopaedics The First Affiliated Hospital of Anhui Medical University Hefei China
| | - Wen‐Shan Shan
- Department of Orthopaedics The First Affiliated Hospital of Anhui Medical University Hefei China
| | - Zhen‐Fei Ding
- Department of Orthopaedics The First Affiliated Hospital of Anhui Medical University Hefei China
| | - Fu‐En Liu
- Department of Orthopaedics The First Affiliated Hospital of Anhui Medical University Hefei China
| | - Wei Lu
- Department of Orthopaedics The First Affiliated Hospital of Anhui Medical University Hefei China
| | - Wei He
- School of Basic Medical Sciences Anhui Medical University Hefei China
| | - Jie‐Gou Xu
- School of Basic Medical Sciences Anhui Medical University Hefei China
| | - Zong‐Sheng Yin
- Department of Orthopaedics The First Affiliated Hospital of Anhui Medical University Hefei China
| |
Collapse
|
12
|
Du YN, Tang XF, Xu L, Chen WD, Gao PJ, Han WQ. SGK1-FoxO1 Signaling Pathway Mediates Th17/Treg Imbalance and Target Organ Inflammation in Angiotensin II-Induced Hypertension. Front Physiol 2018; 9:1581. [PMID: 30524295 PMCID: PMC6262360 DOI: 10.3389/fphys.2018.01581] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Accepted: 10/23/2018] [Indexed: 01/17/2023] Open
Abstract
It has been demonstrated that serum/glucocorticoid regulated kinase 1 (SGK1) and the downstream transcription factor forkhead box O1 (FoxO1) plays a critical role in the differentiation of T helper 17 cells/regulatory T cells (Th17/Treg). In the present study, we hypothesized that this SGK1-FoxO1 signaling pathway is involved in Th17/Treg imbalance and target organ damage in angiotensin II (AngII)-induced hypertensive mice. Results show that SGK1 inhibitor EMD638683 significantly reversed renal dysfunction and cardiac dysfunction in echocardiography as indicated by decreased blood urine nitrogen and serum creatinine in AngII-infused mice. Flow cytometric assay shows that there was significant Th17/Treg imbalance in spleen and in renal/cardiac infiltrating lymphocytes as indicated by the increased Th17 cells (CD4+-IL17A+ cells) and decreased Treg cells (CD4+-Foxp3+). Consistently, real-time PCR shows that Th17-related cytokines including IL-17A, IL-23, and tumor necrosis factor α (TNF-α) was increased and Treg-related cytokine IL-10 was decreased in renal and cardiac infiltrating lymphocytes in AngII-infused mice. Meanwhile, SGK1 protein level, as well as its phosphorylation and activity, was significantly increased in spleen in AngII-infused rats. Furthermore, it was found that splenic phosphorylated FoxO1 was significantly increased, whereas total FoxO1 in nuclear preparation was significantly decreased in AngII-infused mice, suggesting that increased FoxO1 phosphorylation initiate its translocation from cytoplasm to nucleus. Notably, all changes were significantly inhibited by the treatment of EMD638683. These results suggest that SGK1 was involved in Th17/Treg imbalance and target organ damage in AngII-induced hypertension.
Collapse
Affiliation(s)
- Ya-Nan Du
- Shanghai Key Laboratory of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Institute of Hypertension, Shanghai, China
| | - Xiao-Feng Tang
- Shanghai Key Laboratory of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Institute of Hypertension, Shanghai, China
| | - Lian Xu
- Shanghai Key Laboratory of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Laboratory of Vascular Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Wen-Dong Chen
- Shanghai Key Laboratory of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Institute of Hypertension, Shanghai, China
| | - Ping-Jin Gao
- Shanghai Key Laboratory of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Institute of Hypertension, Shanghai, China.,Laboratory of Vascular Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Wei-Qing Han
- Shanghai Key Laboratory of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Institute of Hypertension, Shanghai, China.,Laboratory of Vascular Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
13
|
Perakakis N, Ghaly W, Peradze N, Boutari C, Batirel S, Douglas VP, Mantzoros CS. Research advances in metabolism 2017. Metabolism 2018; 83:280-289. [PMID: 29378200 DOI: 10.1016/j.metabol.2018.01.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 01/19/2018] [Indexed: 11/19/2022]
Affiliation(s)
- Nikolaos Perakakis
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | - Wael Ghaly
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Department of Physiology, Fayoum University, Fayoum, Egypt
| | - Natia Peradze
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Chrysoula Boutari
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Saime Batirel
- Department of Medical Biochemistry, Faculty of Medicine, Marmara University, Istanbul 34854, Turkey; Genetic and Metabolic Diseases Research Center (GEMHAM), Marmara University, Istanbul 34854, Turkey
| | - Vivian Paraskevi Douglas
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Christos S Mantzoros
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
14
|
Kuebler WM, Bonnet S, Tabuchi A. Inflammation and autoimmunity in pulmonary hypertension: is there a role for endothelial adhesion molecules? (2017 Grover Conference Series). Pulm Circ 2018; 8:2045893218757596. [PMID: 29480134 PMCID: PMC5865459 DOI: 10.1177/2045893218757596] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
While pulmonary hypertension (PH) has traditionally not been considered as a disease that is directly linked to or, potentially, even caused by inflammation, a rapidly growing body of evidence has demonstrated the accumulation of a variety of inflammatory and immune cells in PH lungs, in and around the wall of remodeled pulmonary resistance vessels and in the vicinity of plexiform lesions, respectively. Concomitantly, abundant production and release of various inflammatory mediators has been documented in both PH patients and experimental models of PH. While these findings unequivocally demonstrate an inflammatory component in PH, they have fueled an intense and presently ongoing debate as to the nature of this inflammatory aspect: is it a mere bystander of or response to the actual disease process, or is it a pathomechanistic contributor or potentially even a trigger of endothelial injury, smooth muscle hypertrophy and hyperplasia, and the resulting lung vascular remodeling? In this review, we will discuss the present evidence for an inflammatory component in PH disease with a specific focus on the potential role of the endothelium in this scenario and highlight future avenues of experimental investigation which may lead to novel therapeutic interventions.
Collapse
Affiliation(s)
- Wolfgang M Kuebler
- 1 Charite Universitatsmedizin Berlin Institut fur Physiologie, Berlin, Germany
| | | | - Arata Tabuchi
- 1 Charite Universitatsmedizin Berlin Institut fur Physiologie, Berlin, Germany
| |
Collapse
|
15
|
Pullamsetti SS, Savai R. Macrophage Regulation during Vascular Remodeling: Implications for Pulmonary Hypertension Therapy. Am J Respir Cell Mol Biol 2018; 56:556-558. [PMID: 28459385 DOI: 10.1165/rcmb.2017-0033ed] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Affiliation(s)
- Soni Savai Pullamsetti
- 1 Max Planck Institute for Heart and Lung Research Department of Lung Development and Remodeling Member of the German Center for Lung Research (DZL) Bad Nauheim, Germany and.,2 Department of Internal Medicine Universities of Giessen and Marburg Lung Center Member of the DZL Justus Liebig University Giessen, Germany
| | - Rajkumar Savai
- 1 Max Planck Institute for Heart and Lung Research Department of Lung Development and Remodeling Member of the German Center for Lung Research (DZL) Bad Nauheim, Germany and.,2 Department of Internal Medicine Universities of Giessen and Marburg Lung Center Member of the DZL Justus Liebig University Giessen, Germany
| |
Collapse
|
16
|
Jia G, Habibi J, Aroor AR, Hill MA, DeMarco VG, Lee LE, Ma L, Barron BJ, Whaley-Connell A, Sowers JR. Enhanced endothelium epithelial sodium channel signaling prompts left ventricular diastolic dysfunction in obese female mice. Metabolism 2018; 78:69-79. [PMID: 28920862 DOI: 10.1016/j.metabol.2017.08.008] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 07/21/2017] [Accepted: 08/25/2017] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Enhanced activation of cell specific mineralocorticoid receptors (MRs) in obesity plays a key role in the development of cardiovascular disease including cardiac diastolic dysfunction as a critical prognosticator. Our previous investigations demonstrated that selective endothelium MR activation promotes a maladaptive inflammatory response and fibrosis in cardiovascular tissue in female mice fed a western diet (WD), and this was associated with expression and activation of the epithelial sodium channel on the surface of endothelial cells (EnNaC). However, the specific role of EnNaC signaling in the development of cardiac stiffness and diastolic dysfunction has not been examined. We hypothesized that targeted inhibition of EnNaC with low dose amiloride would prevent WD-induced diastolic dysfunction by suppressing abnormal endothelial permeability, inflammation and oxidative stress, and myocardial fibrosis. MATERIALS/METHODS Four week-old female C57BL6/J mice were fed a WD with or without a low dose of amiloride (1mg/kg/day) for 16weeks. Left ventricular cardiac function was evaluated by magnetic resonance imaging. In addition, we examined coronary vessel and cardiac remodeling, fibrosis, macrophage infiltration using immunohistochemistry, western blot and real time PCR. RESULTS Amiloride, an antagonist of EnNaC, attenuated WD-induced impairment of left ventricular initial filling rate and relaxation time. Cardiac diastolic dysfunction was associated with increases in coronary endothelium remodeling and permeability that paralleled WD-induced increases in F-actin and fibronectin, decreased expression of claudin-5 and occludin, and increased macrophage recruitment, M1 polarization, cardiac oxidative stress, fibrosis and maladaptive remodeling. CONCLUSION Our data support the concept that EnNaC activation mediates endothelium permeability which, in turn, promotes macrophage infiltration, M1 polarization, and oxidative stress, resulting in cardiac fibrosis and diastolic dysfunction in females with diet induced obesity.
Collapse
Affiliation(s)
- Guanghong Jia
- Diabetes and Cardiovascular Research Center, University of Missouri School of Medicine, Columbia, MO 65212, USA; Research Service, Harry S Truman Memorial Veterans Hospital, 800 Hospital Dr, Columbia, MO 65201, USA.
| | - Javad Habibi
- Diabetes and Cardiovascular Research Center, University of Missouri School of Medicine, Columbia, MO 65212, USA; Research Service, Harry S Truman Memorial Veterans Hospital, 800 Hospital Dr, Columbia, MO 65201, USA
| | - Annayya R Aroor
- Diabetes and Cardiovascular Research Center, University of Missouri School of Medicine, Columbia, MO 65212, USA; Research Service, Harry S Truman Memorial Veterans Hospital, 800 Hospital Dr, Columbia, MO 65201, USA
| | - Michael A Hill
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO 65212, USA; Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65212, USA
| | - Vincent G DeMarco
- Diabetes and Cardiovascular Research Center, University of Missouri School of Medicine, Columbia, MO 65212, USA; Research Service, Harry S Truman Memorial Veterans Hospital, 800 Hospital Dr, Columbia, MO 65201, USA; Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Li E Lee
- Research Service, Harry S Truman Memorial Veterans Hospital, 800 Hospital Dr, Columbia, MO 65201, USA; Department of Radiology, University of Missouri school of Medicine, Columbia, MO 65212, USA
| | - Lixin Ma
- Research Service, Harry S Truman Memorial Veterans Hospital, 800 Hospital Dr, Columbia, MO 65201, USA; Department of Radiology, University of Missouri school of Medicine, Columbia, MO 65212, USA
| | - Brady J Barron
- Diabetes and Cardiovascular Research Center, University of Missouri School of Medicine, Columbia, MO 65212, USA; Research Service, Harry S Truman Memorial Veterans Hospital, 800 Hospital Dr, Columbia, MO 65201, USA
| | - Adam Whaley-Connell
- Diabetes and Cardiovascular Research Center, University of Missouri School of Medicine, Columbia, MO 65212, USA; Research Service, Harry S Truman Memorial Veterans Hospital, 800 Hospital Dr, Columbia, MO 65201, USA
| | - James R Sowers
- Diabetes and Cardiovascular Research Center, University of Missouri School of Medicine, Columbia, MO 65212, USA; Research Service, Harry S Truman Memorial Veterans Hospital, 800 Hospital Dr, Columbia, MO 65201, USA; Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO 65212, USA; Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65212, USA.
| |
Collapse
|
17
|
Yin J, You S, Liu H, Chen L, Zhang C, Hu H, Xue M, Cheng W, Wang Y, Li X, Shi Y, Li N, Yan S, Li X. Role of P2X 7R in the development and progression of pulmonary hypertension. Respir Res 2017. [PMID: 28646872 PMCID: PMC5483271 DOI: 10.1186/s12931-017-0603-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is a devastating disease that lacks sufficient treatment. Studies have shown that the Nod-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome contributes to PAH pathogenesis, but the role of the upstream molecular P2X7 receptor (P2X7R) has remained unexplored. We investigated the role of P2X7R in the pathogenesis of PAH. METHODS AND RESULTS PH was induced by a single subcutaneous injection of monocrotaline (MCT) (60 mg/kg) on left pneumonectomised Sprague-Dawley rats, as validated by significant increases in pulmonary artery pressure and vessel wall thickness. Marked P2X7R was detected by predominant PA immunostaining in lungs from PH rats. Western blot revealed a significant increase in the protein levels of P2X7R as well as NLRP3 and caspase-1 in the diseased lung tissue compared with normal tissue. The rats received A-740003 (a selective P2X7 receptor antagonist, 30 mg/kg) daily starting from 1 week before or 2 weeks after MCT injection. Consequently, A-740003 reversed the NLRP3 inflammasome upregulation, significantly decreased the mean right ventricular (RV) pressure and RV hypertrophy, and reversed pulmonary arterial remodelling 4 weeks after MCT injection, as both a pretreatment and rescue intervention. Notably, A-740003 significantly reduced macrophage and pro-inflammatory cytokine levels, as measured via bronchoalveolar lavage. The recruitment of macrophages as well as collagen fibre deposition in the perivascular areas were also reduced, as confirmed by histological staining. CONCLUSIONS P2X7R contributes to the pathogenesis of PH, probably in association with activation of the NLRP3 inflammasome. Blockade of P2X7R might be applied as a novel therapeutic approach for the treatment of PAH.
Collapse
Affiliation(s)
- Jie Yin
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, No. 16766 Jingshi Road, Lixia District, Jinan, Shandong Province, China
| | - Shuling You
- Adicon Company, Department of Pathology, Wangkai Infectious Diseases Hospital of Zaozhuang City, Zaozhuang, Shandong Province, China
| | - Haopeng Liu
- Department of Neurosurgery, Zhangqiu People Hospital, Jinan, Shandong, China
| | - Li Chen
- Department of Gastroenterology, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chengdong Zhang
- Department of Orthopedics, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hesheng Hu
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, No. 16766 Jingshi Road, Lixia District, Jinan, Shandong Province, China
| | - Mei Xue
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, No. 16766 Jingshi Road, Lixia District, Jinan, Shandong Province, China
| | - Wenjuan Cheng
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, No. 16766 Jingshi Road, Lixia District, Jinan, Shandong Province, China
| | - Ye Wang
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, No. 16766 Jingshi Road, Lixia District, Jinan, Shandong Province, China
| | - Xinran Li
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, No. 16766 Jingshi Road, Lixia District, Jinan, Shandong Province, China
| | - Yugen Shi
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, No. 16766 Jingshi Road, Lixia District, Jinan, Shandong Province, China
| | - Nannan Li
- Department of Emergency, Shandong Provincial Qianfoshan Hospital, Shandong University of Traditional Chinese Medicine, No. 16766 Jingshi Road, Lixia District, Jinan, Shandong Province, China
| | - Suhua Yan
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, No. 16766 Jingshi Road, Lixia District, Jinan, Shandong Province, China.
| | - Xiaolu Li
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, No. 16766 Jingshi Road, Lixia District, Jinan, Shandong Province, China. .,Department of Emergency, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China.
| |
Collapse
|
18
|
Salt, aldosterone and extrarenal Na + - sensitive responses in pregnancy. Placenta 2017; 56:53-58. [PMID: 28094006 PMCID: PMC5526786 DOI: 10.1016/j.placenta.2017.01.100] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 12/30/2016] [Accepted: 01/09/2017] [Indexed: 12/23/2022]
Abstract
Outside of pregnancy excessive salt consumption is known to be harmful being linked to increased blood pressure and cardiovascular disease. However, pregnancy represents a major change to a woman's physiology resulting in an intimate adaptation to environmental conditions. It is now becoming apparent that salt is essential for a number of these changes during pregnancy including haematological, cardiac adaptations as well as directly influencing placental development and the uteroplacental immune environment. The present review discusses the important role that salt has during normal pregnancy and evidence will also be presented to show how the placenta may act as a salt sensing organ temporarily, yet substantially regulating maternal blood pressure. The placenta may function as an extrarenal regulator of maternal blood pressure. Na+handling in pregnancy is completely different to the non-pregnant situation. Na+may actually lower blood pressure in pregnancy affected with pre-eclampsia. Aldosterone is an important regulator of placental and fetal development. Na+ may compensate for aldosterone deficiency in pregnancy.
Collapse
|
19
|
Yin J, You S, Li N, Jiao S, Hu H, Xue M, Wang Y, Cheng W, Liu J, Xu M, Yan S, Li X. Lung-specific RNA interference of coupling factor 6, a novel peptide, attenuates pulmonary arterial hypertension in rats. Respir Res 2016; 17:99. [PMID: 27491388 PMCID: PMC4973057 DOI: 10.1186/s12931-016-0409-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 06/08/2016] [Indexed: 02/04/2023] Open
Abstract
Background Pulmonary arterial hypertension (PAH) is a progressive and life-threatening disease associated with high morbidity and mortality rates. However, the exact regulatory mechanism of PAH is unknown. Although coupling factor 6 (CF6) is known to function as a repressor, its role in PAH has not been explored. Here, we investigated the involvement of endogenous CF6 in the development of PAH. Methods PAH was induced with monocrotaline (MCT), as demonstrated by significant increases in pulmonary artery pressure and vessel wall thickness. The adeno-associated virus (AAV) carrying CF6 short hairpin RNA (shRNA) or control vector (2×1010 gp) was intratracheally transfected into the lungs of rats 2 weeks before or after MCT injection. Results A 2-6-fold increase in CF6 was observed in the lungs and circulation of the MCT-injected rats as confirmed by qRT-PCR and ELISA. Immunohistochemistry analysis revealed a small quantity of CF6 localized to endothelial cells (ECs) under physiological conditions spread to surrounding tissues in a paracrine manner in PAH lungs. Notably, CF6 shRNA effectively inhibited CF6 expression, abolished lung macrophage infiltration, reversed endothelial dysfunction and vascular remodeling, and ameliorated the severity of pulmonary hypertension and right ventricular dysfunction at 4 weeks both as a pretreatment and rescue intervention. In addition, the circulating and lung levels of 6-keto-PGF1a, a stable metabolite of prostacyclin, were reversed by CF6 inhibition, suggesting that the effect of CF6 inhibition may partly be mediated through prostacyclin. Conclusions CF6 contributes to the pathogenesis of PAH, probably in association with downregulation of prostacyclin. The blockage of CF6 might be applied as a novel therapeutic approach for PAH and PA remodeling. Electronic supplementary material The online version of this article (doi:10.1186/s12931-016-0409-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jie Yin
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, No. 16766 Jingshi Road, Lixia District, Jinan, Shandong Province, China
| | - Shuling You
- Department of Pathology, Adicon Company, Wangkai Infectious Diseases Hospital of Zaozhuang City, Zaozhuang, Shandong, China
| | - Nannan Li
- Department of Emergency, Shandong Provincial Qianfoshan Hospital, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Shouhai Jiao
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, No. 16766 Jingshi Road, Lixia District, Jinan, Shandong Province, China
| | - Hesheng Hu
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, No. 16766 Jingshi Road, Lixia District, Jinan, Shandong Province, China
| | - Mei Xue
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, No. 16766 Jingshi Road, Lixia District, Jinan, Shandong Province, China
| | - Ye Wang
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, No. 16766 Jingshi Road, Lixia District, Jinan, Shandong Province, China
| | - Wenjuan Cheng
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, No. 16766 Jingshi Road, Lixia District, Jinan, Shandong Province, China
| | - Ju Liu
- Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong Province, China
| | - Min Xu
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, No. 16766 Jingshi Road, Lixia District, Jinan, Shandong Province, China
| | - Suhua Yan
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, No. 16766 Jingshi Road, Lixia District, Jinan, Shandong Province, China.
| | - Xiaolu Li
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, No. 16766 Jingshi Road, Lixia District, Jinan, Shandong Province, China. .,Department of Emergency, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong Province, China.
| |
Collapse
|
20
|
Jia G, Habibi J, Aroor AR, Martinez-Lemus LA, DeMarco VG, Ramirez-Perez FI, Sun Z, Hayden MR, Meininger GA, Mueller KB, Jaffe IZ, Sowers JR. Endothelial Mineralocorticoid Receptor Mediates Diet-Induced Aortic Stiffness in Females. Circ Res 2016; 118:935-943. [PMID: 26879229 DOI: 10.1161/circresaha.115.308269] [Citation(s) in RCA: 135] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 02/12/2016] [Indexed: 12/13/2022]
Abstract
RATIONALE Enhanced activation of the mineralocorticoid receptors (MRs) in cardiovascular tissues increases oxidative stress, maladaptive immune responses, and inflammation with associated functional vascular abnormalities. We previously demonstrated that consumption of a Western diet (WD) for 16 weeks results in aortic stiffening, and that these abnormalities were prevented by systemic MR blockade in female mice. However, the cell-specific role of endothelial cell MR (ECMR) in these maladaptive vascular effects has not been explored. OBJECTIVE We hypothesized that specific deletion of the ECMR would prevent WD-induced increases in endothelial sodium channel activation, reductions in bioavailable nitric oxide, increased vascular remodeling, and associated increases in vascular stiffness in females. METHODS AND RESULTS Four-week-old female ECMR knockout and wild-type mice were fed either mouse chow or WD for 16 weeks. WD feeding resulted in aortic stiffness and endothelial dysfunction as determined in vivo by pulse wave velocity and ex vivo by atomic force microscopy, and wire and pressure myography. The WD-induced aortic stiffness was associated with enhanced endothelial sodium channel activation, attenuated endothelial nitric oxide synthase activation, increased oxidative stress, a proinflammatory immune response and fibrosis. Conversely, cell-specific ECMR deficiency prevented WD-induced aortic fibrosis and stiffness in conjunction with reductions in endothelial sodium channel activation, oxidative stress and macrophage proinflammatory polarization, restoration of endothelial nitric oxide synthase activation. CONCLUSIONS Increased ECMR signaling associated with consumption of a WD plays a key role in endothelial sodium channel activation, reduced nitric oxide production, oxidative stress, and inflammation that lead to aortic remodeling and stiffness in female mice.
Collapse
Affiliation(s)
- Guanghong Jia
- Diabetes and Cardiovascular Research Center, University of Missouri School of Medicine, Columbia, MO, 65212, USA.,Research Service, Harry S Truman Memorial Veterans Hospital, Research Service, 800 Hospital Dr, Columbia, MO, 65201, USA
| | - Javad Habibi
- Diabetes and Cardiovascular Research Center, University of Missouri School of Medicine, Columbia, MO, 65212, USA.,Research Service, Harry S Truman Memorial Veterans Hospital, Research Service, 800 Hospital Dr, Columbia, MO, 65201, USA
| | - Annayya R Aroor
- Diabetes and Cardiovascular Research Center, University of Missouri School of Medicine, Columbia, MO, 65212, USA.,Research Service, Harry S Truman Memorial Veterans Hospital, Research Service, 800 Hospital Dr, Columbia, MO, 65201, USA
| | - Luis A Martinez-Lemus
- Research Service, Harry S Truman Memorial Veterans Hospital, Research Service, 800 Hospital Dr, Columbia, MO, 65201, USA.,Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO, 65212, USA.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, 65212, USA
| | - Vincent G DeMarco
- Diabetes and Cardiovascular Research Center, University of Missouri School of Medicine, Columbia, MO, 65212, USA.,Research Service, Harry S Truman Memorial Veterans Hospital, Research Service, 800 Hospital Dr, Columbia, MO, 65201, USA.,Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO, 65212, USA
| | | | - Zhe Sun
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, 65212, USA
| | - Melvin R Hayden
- Diabetes and Cardiovascular Research Center, University of Missouri School of Medicine, Columbia, MO, 65212, USA.,Research Service, Harry S Truman Memorial Veterans Hospital, Research Service, 800 Hospital Dr, Columbia, MO, 65201, USA
| | - Gerald A Meininger
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO, 65212, USA.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, 65212, USA
| | | | - Iris Z Jaffe
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA
| | - James R Sowers
- Diabetes and Cardiovascular Research Center, University of Missouri School of Medicine, Columbia, MO, 65212, USA.,Research Service, Harry S Truman Memorial Veterans Hospital, Research Service, 800 Hospital Dr, Columbia, MO, 65201, USA.,Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO, 65212, USA.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, 65212, USA
| |
Collapse
|
21
|
Witte S, Bradley A, Enright AJ, Muljo SA. High-density P300 enhancers control cell state transitions. BMC Genomics 2015; 16:903. [PMID: 26546038 PMCID: PMC4636788 DOI: 10.1186/s12864-015-1905-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 09/08/2015] [Indexed: 12/11/2022] Open
Abstract
Background Transcriptional enhancers are frequently bound by a set of transcription factors that collaborate to activate lineage-specific gene expression. Recently, it was appreciated that a subset of enhancers comprise extended clusters dubbed stretch- or super-enhancers (SEs). These SEs are located near key cell identity genes, and enriched for non-coding genetic variations associated with disease. Previously, SEs have been defined as having the highest density of Med1, Brd4 or H3K27ac by ChIP-seq. The histone acetyltransferase P300 has been used as a marker of enhancers, but little is known about its binding to SEs. Results We establish that P300 marks a similar SE repertoire in embryonic stem cells as previously reported using Med1 and H3K27ac. We also exemplify a role for SEs in mouse T helper cell fate decision. Similarly, upon activation of macrophages by bacterial endotoxin, we found that many SE-associated genes encode inflammatory proteins that are strongly up-regulated. These SEs arise from small, low-density enhancers in unstimulated macrophages. We also identified expression quantitative trait loci (eQTL) in human monocytes that lie within such SEs. In macrophages and Th17 cells, inflammatory SEs can be perturbed either genetically or pharmacologically thus revealing new avenues to target inflammation. Conclusions Our findings support the notion that P300-marked SEs can help identify key nodes of transcriptional control during cell fate decisions. The SE landscape changes drastically during cell differentiation and cell activation. As these processes are crucial in immune responses, SEs may be useful in revealing novel targets for treating inflammatory diseases. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-1905-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Steven Witte
- Integrative Immunobiology Unit, Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA. .,Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK.
| | - Allan Bradley
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK.
| | - Anton J Enright
- EMBL - European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK.
| | - Stefan A Muljo
- Integrative Immunobiology Unit, Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
22
|
He J, Qi D, Wang DX, Deng W, Ye Y, Feng LH, Zhu T, Zhao Y, Zhang CR. Insulin upregulates the expression of epithelial sodium channel in vitro and in a mouse model of acute lung injury: Role of mTORC2/SGK1 pathway. Exp Cell Res 2015; 331:164-175. [DOI: 10.1016/j.yexcr.2014.09.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 09/03/2014] [Accepted: 09/18/2014] [Indexed: 01/04/2023]
|
23
|
Binger KJ, Linker RA, Muller DN, Kleinewietfeld M. Sodium chloride, SGK1, and Th17 activation. Pflugers Arch 2014; 467:543-50. [PMID: 25471348 DOI: 10.1007/s00424-014-1659-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 11/19/2014] [Accepted: 11/20/2014] [Indexed: 02/08/2023]
Abstract
The incidence of autoimmune diseases in Western civilizations is increasing rapidly, suggesting an influence of environmental factors, such as diet. The pathogenesis of several of these autoimmune diseases is characterized by aberrant activation of T helper 17 (Th17) cells. Recent reports have shown that the differentiation of Th17 cells is sensitive to changes in local microenvironments, in particular salt (NaCl) concentrations, in a molecular mechanism centered around the serum- and glucocorticoid-inducible kinase 1 (SGK1). In this review, we summarize the recently disclosed mechanisms by which salt has been shown to affect SGK1 and, subsequently, Th17 activation.
Collapse
Affiliation(s)
- Katrina J Binger
- Experimental and Clinical Research Center, an institutional cooperation between the Charité Medical Faculty and the Max-Delbrueck Center for Molecular Medicine, Berlin, 13125, Germany
| | | | | | | |
Collapse
|