1
|
Liu Y, Ma X, Lei L, Wang L, Deng Q, Lu H, Li H, Tian S, Qin X, Zhang W, Sun Y. Smooth Muscle Cell-Specific LKB1 Protects Against Sugen 5416/Hypoxia-induced Pulmonary Hypertension through Inhibition of BMP4. Am J Respir Cell Mol Biol 2025; 72:169-180. [PMID: 39236291 DOI: 10.1165/rcmb.2023-0430oc] [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: 12/06/2023] [Accepted: 09/05/2024] [Indexed: 09/07/2024] Open
Abstract
Pulmonary hypertension (PH) is a life-threatening syndrome associated with hyperproliferation of pulmonary artery smooth muscle cells (PASMCs), which exhibit features similar to those of cancer cells. Currently, there is no curative treatment for PH. LKB1 is known as a tumor suppressor gene with an antiproliferative effect on cancer cells. However, its role and mechanism in the development of PH remain unclear. Gain- and loss-of-function strategies were used to elucidate the mechanisms of LKB1 in regulating the occurrence and progression of PH. Sugen 5416/hypoxia (SuHx) PH model was utilized for in vivo study. We observed a decreased expression of LKB1 not only in the lung vessels of the SuHx mouse model but also in human PASMCs (HPASMCs) exposed to hypoxia. Smooth muscle-specific LKB1 knockout significantly aggravated SuHx-induced PH in mice. RNA-sequencing analysis revealed a substantial increase in bone morphogenetic protein 4 (BMP4) in the aortas of LKB1SMKO mice compared with controls, identifying BMP4 as a novel target of LKB1. LKB1 knockdown in HPASMCs cultured under hypoxic conditions increased BMP4 protein level and HPASMC proliferation and migration. The coimmunoprecipitation analysis revealed that LKB1 directly modulates BMP4 protein degradation through phosphorylation. Therapeutically, suppressing BMP4 expression in smooth muscle cells alleviates PH in LKB1SMKO mice. Our findings demonstrate that LKB1 attenuates PH by enhancing the lysosomal degradation of BMP4, thus suppressing the proliferation and migration of HPASMCs. Modulating the LKB1-BMP4 axis in smooth muscle cells could be a promising therapeutic strategy of PH.
Collapse
MESH Headings
- Animals
- Protein Serine-Threonine Kinases/metabolism
- Protein Serine-Threonine Kinases/genetics
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/pathology
- Hypertension, Pulmonary/chemically induced
- Humans
- Bone Morphogenetic Protein 4/metabolism
- Bone Morphogenetic Protein 4/genetics
- Mice
- Mice, Knockout
- AMP-Activated Protein Kinases/metabolism
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Hypoxia/metabolism
- Hypoxia/complications
- Cell Proliferation
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Mice, Inbred C57BL
- AMP-Activated Protein Kinase Kinases/metabolism
- Male
- Disease Models, Animal
- Indoles
- Pyrroles
Collapse
Affiliation(s)
- Yan Liu
- State Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Xiaoping Ma
- State Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
- Department of Obstetrics and Gynecology, Liaocheng People's Hospital, Liaocheng, China
| | - Lingli Lei
- School of Clinical Medical Sciences, Clinical Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China; and
| | - Lin Wang
- Department of Cardiology, Jinan Central Hospital, Jinan, China
| | - Qiming Deng
- State Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Hanlin Lu
- State Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Hongxuan Li
- State Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Shuhui Tian
- School of Clinical Medical Sciences, Clinical Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China; and
| | - Xiaoteng Qin
- State Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Wencheng Zhang
- State Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Yuanyuan Sun
- State Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
2
|
Tan W, Wang Y, Li M, Zhao C, Hu Y, Gao R, Chen Z, Hu L, Li Q. A novel pyridine-2-one AMPK inhibitor: Discovery, mechanism, and in vivo evaluation in a hypoxic pulmonary arterial hypertension rat model. Eur J Med Chem 2025; 286:117266. [PMID: 39826489 DOI: 10.1016/j.ejmech.2025.117266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 01/03/2025] [Accepted: 01/08/2025] [Indexed: 01/22/2025]
Abstract
AMP-activated protein kinase (AMPK), a heterotrimeric serine-threonine kinase, has been identified as a promising target for regulating vascular remodeling in pulmonary arterial hypertension (PAH) due to its capacity to promote proliferation, autophagy, and anti-apoptosis in pulmonary artery smooth muscle cells (PASMCs). However, research into AMPK inhibitors is very limited. Herein, a virtual screening strategy was employed to identify CHEMBL3780091 as a lead compound for a series of novel AMPK inhibitors by exploring the structure-activity relationship around a specific pyridine-2-one scaffold. Subsequently, the most promising 13a was observed to exhibit excellent AMPK inhibitory activity and favorable anti-proliferative activity against PASMCs through the inhibition of the AMPK signaling pathway in vitro. Moreover, compound 13a significantly reduced right ventricular systolic pressure, attenuated vascular remodeling, and improved right heart function in hypoxia-induced PAH rats in vivo. In conclusion, this study provides a novel and potential lead compound for the study of AMPK inhibitors and a new direction for the development of PAH drugs that focus on improving vascular remodeling.
Collapse
Affiliation(s)
- Wenhua Tan
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China
| | - Yu Wang
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China
| | - Mengqi Li
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China
| | - Congke Zhao
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China
| | - Yuanbo Hu
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China
| | - Ruizhe Gao
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, 410013, Hunan, China
| | - Zhuo Chen
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China
| | - Liqing Hu
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, 410013, Hunan, China.
| | - Qianbin Li
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China.
| |
Collapse
|
3
|
Hou J, Nie Y, Wen Y, Hua S, Hou Y, He H, Sun S. The role and mechanism of AMPK in pulmonary hypertension. Ther Adv Respir Dis 2024; 18:17534666241271990. [PMID: 39136335 PMCID: PMC11322949 DOI: 10.1177/17534666241271990] [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: 10/16/2023] [Accepted: 06/28/2024] [Indexed: 08/16/2024] Open
Abstract
Pulmonary hypertension (PH) is a chronic progressive disease with high mortality. There has been more and more research focusing on the role of AMPK in PH. AMPK consists of three subunits-α, β, and γ. The crosstalk among these subunits ultimately leads to a delicate balance to affect PH, which results in conflicting conclusions about the role of AMPK in PH. It is still unclear how these subunits interfere with each other and achieve balance to improve or deteriorate PH. Several signaling pathways are related to AMPK in the treatment of PH, including AMPK/eNOS/NO pathway, Nox4/mTORC2/AMPK pathway, AMPK/BMP/Smad pathway, and SIRT3-AMPK pathway. Among these pathways, the role and mechanism of AMPK/eNOS/NO and Nox4/mTORC2/AMPK pathways are clearer than others, while the SIRT3-AMPK pathway remains still unclear in the treatment of PH. There are drugs targeting AMPK to improve PH, such as metformin (MET), MET combination, and rhodiola extract. In addition, several novel factors target AMPK for improving PH, such as ADAMTS8, TUFM, and Salt-inducible kinases. However, more researches are needed to explore the specific AMPK signaling pathways involved in these novel factors in the future. In conclusion, AMPK plays an important role in PH.
Collapse
Affiliation(s)
- Jing Hou
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, China
- Class Three & Class Eight, 2021Clinical Medicine, Kunming Medical University, Kunming, China
| | - Yu Nie
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, China
- Class Three & Class Eight, 2021Clinical Medicine, Kunming Medical University, Kunming, China
| | - Yiqiong Wen
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Shu Hua
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yunjiao Hou
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Huilin He
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Shibo Sun
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University, No. 295, Xichang Road, Wuhua District, Kunming 650032, China
| |
Collapse
|
4
|
Ning L, Zou Y, Li S, Cao Y, Xu B, Zhang S, Cai Y. Anti-PCSK9 Treatment Attenuates Liver Fibrosis via Inhibiting Hypoxia-Induced Autophagy in Hepatocytes. Inflammation 2023; 46:2102-2119. [PMID: 37466835 PMCID: PMC10673768 DOI: 10.1007/s10753-023-01865-8] [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: 04/05/2023] [Revised: 06/25/2023] [Accepted: 06/26/2023] [Indexed: 07/20/2023]
Abstract
Hypoxia and its induced autophagy are involved in the initiation and progression of liver fibrosis. Proprotein convertase subtilisin/kexin type 9 (PCSK9) has been recognized as a potential regulator of autophagy. Our previously reported study found that PCSK9 expression increased in liver fibrosis and that anti-PCSK9 treatment alleviated liver injury. This study aimed to investigate the mechanism of anti-PCSK9 treatment on liver fibrosis by inhibiting hypoxia-induced autophagy. Carbon tetrachloride-induced mouse liver fibrosis and mouse hepatocyte line AML12, cultured under the hypoxic condition, were established to undergo PCSK9 inhibition. The degree of liver fibrosis was shown with histological staining. The reactive oxygen species (ROS) generation was detected by flow cytometry. The expression of PCSK9, hypoxia-inducible factor-1α (HIF-1α), and autophagy-related proteins was examined using Western blot. The autophagic flux was assessed under immunofluorescence and transmission electron microscope. The mouse liver samples were investigated via RNA-sequencing to explore the underlying signaling pathway. The results showed that PCSK9 expression was upregulated with the development of liver fibrosis, which was accompanied by enhanced autophagy. In vitro data verified that PCSK9 increased via hypoxia and inflammation, accompanied by the hypoxia-induced autophagy increased. Then, the validation was acquired of the bidirectional interaction of hypoxia-ROS and PCSK9. The hypoxia reversal attenuated PCSK9 expression and autophagy. Additionally, anti-PCSK9 treatment alleviated liver inflammation and fibrosis, reducing hypoxia and autophagy in vivo. In mechanism, the AMPK/mTOR/ULK1 signaling pathway was identified as a target for anti-PCSK9 therapy. In conclusion, anti-PCSK9 treatment could alleviate liver inflammation and fibrosis by regulating AMPK/mTOR/ULK1 signaling pathway to reduce hypoxia-induced autophagy in hepatocytes.
Collapse
Affiliation(s)
- Liuxin Ning
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Liver Diseases, Shanghai, 200032, China
| | - Yanting Zou
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Liver Diseases, Shanghai, 200032, China
| | - Shuyu Li
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Liver Diseases, Shanghai, 200032, China
| | - Yue Cao
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Liver Diseases, Shanghai, 200032, China
| | - Beili Xu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Liver Diseases, Shanghai, 200032, China
| | - Shuncai Zhang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Liver Diseases, Shanghai, 200032, China
| | - Yu Cai
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Shanghai Institute of Liver Diseases, Shanghai, 200032, China.
| |
Collapse
|
5
|
Ren H, Dai R, Nik Nabil WN, Xi Z, Wang F, Xu H. Unveiling the dual role of autophagy in vascular remodelling and its related diseases. Biomed Pharmacother 2023; 168:115643. [PMID: 37839111 DOI: 10.1016/j.biopha.2023.115643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 09/18/2023] [Accepted: 10/03/2023] [Indexed: 10/17/2023] Open
Abstract
Vascular remodelling is an adaptive response to physiological and pathological stimuli that leads to structural and functional changes in the vascular intima, media, and adventitia. Pathological vascular remodelling is a hallmark feature of numerous vascular diseases, including atherosclerosis, hypertension, abdominal aortic aneurysm, pulmonary hypertension and preeclampsia. Autophagy is critical in maintaining cellular homeostasis, and its dysregulation has been implicated in the pathogenesis of various diseases, including vascular diseases. However, despite emerging evidence, the role of autophagy and its dual effects on vascular remodelling has garnered limited attention. Autophagy can exert protective and detrimental effects on the vascular intima, media and adventitia, thereby substantially influencing the course of vascular remodelling and its related vascular diseases. Currently, there has not been a review that thoroughly describes the regulation of autophagy in vascular remodelling and its impact on related diseases. Therefore, this review aimed to bridge this gap by focusing on the regulatory roles of autophagy in diseases related to vascular remodelling. This review also summarizes recent advancements in therapeutic agents targeting autophagy to regulate vascular remodelling. Additionally, this review offers an overview of recent breakthroughs in therapeutic agents targeting autophagy to regulate vascular remodelling. A deeper understanding of how autophagy orchestrates vascular remodelling can drive the development of targeted therapies for vascular diseases.
Collapse
Affiliation(s)
- Hangui Ren
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai 201203, China
| | - Rongchen Dai
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai 201203, China
| | - Wan Najbah Nik Nabil
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai 201203, China; Pharmaceutical Services Program, Ministry of Health, Selangor 46200, Malaysia
| | - Zhichao Xi
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai 201203, China
| | - Feng Wang
- Department of Neurology, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai 200137, China.
| | - Hongxi Xu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai 201203, China.
| |
Collapse
|
6
|
Liu Y, Zhu L, Ming Y, Wu Z, Zhang L, Chen Q, Qi Y. A role of TRIM59 in pulmonary hypertension: modulating the protein ubiquitylation modification. J Transl Med 2023; 21:821. [PMID: 37978515 PMCID: PMC10655329 DOI: 10.1186/s12967-023-04712-4] [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: 09/25/2023] [Accepted: 11/07/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND Pulmonary hypertension (PH), an infrequent disease, is characterized by excessive pulmonary vascular remodeling and proliferation of pulmonary artery smooth muscle cells (PASMCs). However, its underlying molecular mechanisms remain unclear. Uncovering its molecular mechanisms will be beneficial to the treatment of PH. METHODS Differently expressed genes (DEGs) in the lung tissues of PH patients were analyzed with a GEO dataset GSE113439. From these DEGs, we focused on TRIM59 which was highly expressed in PH patients. Subsequently, the expression of TRIM59 in the pulmonary arteries of PH patients, lung tissues of PH rat model and PASMCs cultured in a hypoxic condition was verified by quantitative real-time PCR (qPCR), western blot and immunohistochemistry. Furthermore, the role of TRIM59 in PAMSC proliferation and pathological changes in PH rats was assessed via gain-of-function and loss-of-function experiments. In addition, the transcriptional regulation of YAP1/TEAD4 on TRIM59 was confirmed by qPCR, western blot, luciferase reporter assay, ChIP and DNA pull-down. In order to uncover the underlying mechanisms of TRIM59, a protein ubiquitomics and a CoIP- HPLC-MS/MS were companied to identify the direct targets of TRIM59. RESULTS TRIM59 was highly expressed in the pulmonary arteries of PH patients and lung tissues of PH rats. Over-expression of TRIM59 accelerated the proliferation of PASMCs, while TRIM59 silencing resulted in the opposite results. Moreover, TRIM59 silencing mitigated the injuries in heart and lung and attenuated pulmonary vascular remodeling during PH. In addition, its transcription was positively regulated by YAP1/TEAD4. Then we further explored the underlying mechanisms of TRIM59 and found that TRIM59 overexpression resulted in an altered ubiquitylation of proteins. Accompanied with the results of CoIP- HPLC-MS/MS, 34 proteins were identified as the direct targets of TRIM59. CONCLUSION TRIM59 was highly expressed in PH patients and promoted the proliferation of PASMCs and pulmonary vascular remodeling, thus contributing to the pathogenesis of PH. It is indicated that TRIM59 may become a potential target for PH treatment.
Collapse
Affiliation(s)
- Yingli Liu
- Department of Pulmonary and Critical Care Medicine, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, People's Republic of China
| | - Li Zhu
- Department of Pulmonary and Critical Care Medicine, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, People's Republic of China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, People's Republic of China
| | - Yue Ming
- Department of Pulmonary and Critical Care Medicine, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, People's Republic of China
| | - Zhuhua Wu
- Department of Pulmonary and Critical Care Medicine, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, People's Republic of China
| | - Lili Zhang
- Department of Pulmonary and Critical Care Medicine, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, People's Republic of China
| | - Qi Chen
- Department of Pulmonary and Critical Care Medicine, Henan University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, People's Republic of China
| | - Yong Qi
- Department of Pulmonary and Critical Care Medicine, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Henan University People's Hospital, Zhengzhou, People's Republic of China.
| |
Collapse
|
7
|
Chong SJF, Zhu F, Dashevsky O, Mizuno R, Lai JX, Hackett L, Ryan CE, Collins MC, Iorgulescu JB, Guièze R, Penailillo J, Carrasco R, Hwang YC, Muñoz DP, Bouhaddou M, Lim YC, Wu CJ, Allan JN, Furman RR, Goh BC, Pervaiz S, Coppé JP, Mitsiades CS, Davids MS. Hyperphosphorylation of BCL-2 family proteins underlies functional resistance to venetoclax in lymphoid malignancies. J Clin Invest 2023; 133:e170169. [PMID: 37751299 PMCID: PMC10645378 DOI: 10.1172/jci170169] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 09/19/2023] [Indexed: 09/27/2023] Open
Abstract
The B cell leukemia/lymphoma 2 (BCL-2) inhibitor venetoclax is effective in chronic lymphocytic leukemia (CLL); however, resistance may develop over time. Other lymphoid malignancies such as diffuse large B cell lymphoma (DLBCL) are frequently intrinsically resistant to venetoclax. Although genomic resistance mechanisms such as BCL2 mutations have been described, this probably only explains a subset of resistant cases. Using 2 complementary functional precision medicine techniques - BH3 profiling and high-throughput kinase activity mapping - we found that hyperphosphorylation of BCL-2 family proteins, including antiapoptotic myeloid leukemia 1 (MCL-1) and BCL-2 and proapoptotic BCL-2 agonist of cell death (BAD) and BCL-2 associated X, apoptosis regulator (BAX), underlies functional mechanisms of both intrinsic and acquired resistance to venetoclax in CLL and DLBCL. Additionally, we provide evidence that antiapoptotic BCL-2 family protein phosphorylation altered the apoptotic protein interactome, thereby changing the profile of functional dependence on these prosurvival proteins. Targeting BCL-2 family protein phosphorylation with phosphatase-activating drugs rewired these dependencies, thus restoring sensitivity to venetoclax in a panel of venetoclax-resistant lymphoid cell lines, a resistant mouse model, and in paired patient samples before venetoclax treatment and at the time of progression.
Collapse
MESH Headings
- Mice
- Animals
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Drug Resistance, Neoplasm/genetics
- Proto-Oncogene Proteins c-bcl-2/genetics
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- bcl-X Protein/genetics
- Apoptosis Regulatory Proteins
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/pathology
- Cell Line, Tumor
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Apoptosis/genetics
- Myeloid Cell Leukemia Sequence 1 Protein/genetics
- Myeloid Cell Leukemia Sequence 1 Protein/metabolism
Collapse
Affiliation(s)
- Stephen Jun Fei Chong
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Fen Zhu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Olga Dashevsky
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Rin Mizuno
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Jolin X.H. Lai
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Liam Hackett
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Christine E. Ryan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Mary C. Collins
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - J. Bryan Iorgulescu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Romain Guièze
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Johany Penailillo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Ruben Carrasco
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Yeonjoo C. Hwang
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, California, USA
| | - Denise P. Muñoz
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, California, USA
| | - Mehdi Bouhaddou
- Department of Microbiology, Immunology and Molecular Genetics, UCLA, Los Angeles, California, USA
| | - Yaw Chyn Lim
- Cancer Science Institute, National University of Singapore, Singapore
| | - Catherine J. Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - John N. Allan
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, New York, USA
| | - Richard R. Furman
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, New York, USA
| | - Boon Cher Goh
- Cancer Science Institute, National University of Singapore, Singapore
| | - Shazib Pervaiz
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jean-Philippe Coppé
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, California, USA
| | - Constantine S. Mitsiades
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Matthew S. Davids
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
8
|
Bruns DR, McNair BD, Peelor FF, Borowik AK, Pranay A, Yusifov A, Miller BF. Skeletal and cardiac muscle have different protein turnover responses in a model of right heart failure. GeroScience 2023; 45:2545-2557. [PMID: 37118350 PMCID: PMC10651599 DOI: 10.1007/s11357-023-00777-7] [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: 02/08/2023] [Accepted: 03/20/2023] [Indexed: 04/30/2023] Open
Abstract
Right heart failure (RHF) is a common and deadly disease in aged populations. Extra-cardiac outcomes of RHF such as skeletal muscle atrophy contribute to morbidity and mortality. Despite the significance of maintaining right ventricular (RV) and muscle function, the mechanisms of RHF and muscle atrophy are unclear. Metformin (MET) improves cardiac and muscle function through the regulation of metabolism and the cellular stress response. However, whether MET is a viable therapeutic for RHF and muscle atrophy is not yet known. We used deuterium oxide labeling to measure individual protein turnover in the RV as well as subcellular skeletal muscle proteostasis in aged male mice subjected to 4 weeks of hypobaric hypoxia (HH)-induced RHF. Mice exposed to HH had elevated RV mass and impaired RV systolic function, neither of which was prevented by MET. HH resulted in a higher content of glycolytic, cardiac, and antioxidant proteins in the RV, most of which were inhibited by MET. The synthesis of these key RV proteins was generally unchanged by MET, suggesting MET accelerated protein breakdown. HH resulted in a loss of skeletal muscle mass due to inhibited protein synthesis alongside myofibrillar protein breakdown. MET did not impact HH-induced muscle protein turnover and did not prevent muscle wasting. Together, we show tissue-dependent responses to HH-induced RHF where the RV undergoes hypertrophic remodeling with higher expression of metabolic and stress response proteins. Skeletal muscle undergoes loss of protein mass and atrophy, primarily due to myofibrillar protein breakdown. MET did not prevent HH-induced RV dysfunction or muscle wasting, suggesting that the identification of other therapies to attenuate RHF and concomitant muscle atrophy is warranted.
Collapse
Affiliation(s)
- Danielle R Bruns
- Division of Kinesiology & Health, University of Wyoming, 1000 E. University Ave, Dept. 3196, Laramie, WY, 82071, USA.
- Wyoming WWAMI Medical Education, Laramie, WY, USA.
| | - Benjamin D McNair
- Division of Kinesiology & Health, University of Wyoming, 1000 E. University Ave, Dept. 3196, Laramie, WY, 82071, USA
| | - Frederick F Peelor
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Agnieszka K Borowik
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Atul Pranay
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Aykhan Yusifov
- Division of Kinesiology & Health, University of Wyoming, 1000 E. University Ave, Dept. 3196, Laramie, WY, 82071, USA
| | - Benjamin F Miller
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Oklahoma City VA Medical Center, Oklahoma City, OK, USA
| |
Collapse
|
9
|
McNair BD, Polson SM, Shorthill SK, Yusifov A, Walker LA, Weiser-Evans MCM, Kovacs EJ, Bruns DR. Metformin protects against pulmonary hypertension-induced right ventricular dysfunction in an age- and sex-specific manner independent of cardiac AMPK. Am J Physiol Heart Circ Physiol 2023; 325:H278-H292. [PMID: 37389952 PMCID: PMC10393374 DOI: 10.1152/ajpheart.00124.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/30/2023] [Accepted: 06/16/2023] [Indexed: 07/02/2023]
Abstract
Right ventricular (RV) function is the strongest predictor of survival in age-related heart failure as well as other clinical contexts in which aging populations suffer significant morbidity and mortality. However, despite the significance of maintaining RV function with age and disease, mechanisms of RV failure remain poorly understood and no RV-directed therapies exist. The antidiabetic drug and AMP-activated protein kinase (AMPK) activator metformin protects against left ventricular dysfunction, suggesting cardioprotective properties may translate to the RV. Here, we aimed to understand the impact of advanced age on pulmonary hypertension (PH)-induced right ventricular dysfunction. We further aimed to test whether metformin is cardioprotective in the RV and whether the protection afforded by metformin requires cardiac AMPK. We used a murine model of PH by exposing adult (4-6 mo) and aged (18 mo) male and female mice to hypobaric hypoxia (HH) for 4 wk. Cardiopulmonary remodeling was exacerbated in aged mice compared with adult mice as evidenced by elevated RV weight and impaired RV systolic function. Metformin attenuated HH-induced RV dysfunction but only in adult male mice. Metformin still protected the adult male RV even in the absence of cardiac AMPK. Together, we suggest that aging exacerbates PH-induced RV remodeling and that metformin may represent a therapeutic option for this disease in a sex- and age-dependent manner, but in an AMPK-independent manner. Ongoing efforts are aimed at elucidating the molecular basis for RV remodeling as well as delineating the mechanisms of cardioprotection provided by metformin in the absence of cardiac AMPK.NEW & NOTEWORTHY Right ventricular (RV) function predicts survival in age-related disease, yet mechanisms of RV failure are unclear. We show that aged mice undergo exacerbated RV remodeling compared with young. We tested the AMPK activator metformin to improve RV function and show that metformin attenuates RV remodeling only in adult male mice via a mechanism that does not require cardiac AMPK. Metformin is therapeutic for RV dysfunction in an age- and sex-specific manner independent of cardiac AMPK.
Collapse
Affiliation(s)
- Benjamin D McNair
- Division of Kinesiology and Health, University of Wyoming, Laramie, Wyoming, United States
| | - Sydney M Polson
- Division of Kinesiology and Health, University of Wyoming, Laramie, Wyoming, United States
| | - Samantha K Shorthill
- Division of Kinesiology and Health, University of Wyoming, Laramie, Wyoming, United States
| | - Aykhan Yusifov
- Division of Kinesiology and Health, University of Wyoming, Laramie, Wyoming, United States
| | - Lori A Walker
- Division of Cardiology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Mary C M Weiser-Evans
- Division of Renal Diseases and Hypertension, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Consortium for Fibrosis Research and Translation, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Elizabeth J Kovacs
- Division of GI, Trauma, and Endocrine Surgery, Department of Surgery, University of Colorado Denver, Aurora, Colorado, United States
- Burn Research Program, University of Colorado Denver, Aurora, Colorado, United States
- GI and Liver Innate Immune Program, University of Colorado Denver, Aurora, Colorado, United States
| | - Danielle R Bruns
- Division of Kinesiology and Health, University of Wyoming, Laramie, Wyoming, United States
- Wyoming WWAMI Medical Education Program, Laramie, Wyoming, United States
| |
Collapse
|
10
|
Flores K, Siques P, Brito J, Arribas SM. AMPK and the Challenge of Treating Hypoxic Pulmonary Hypertension. Int J Mol Sci 2022; 23:ijms23116205. [PMID: 35682884 PMCID: PMC9181235 DOI: 10.3390/ijms23116205] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/29/2022] [Accepted: 04/30/2022] [Indexed: 02/01/2023] Open
Abstract
Hypoxic pulmonary hypertension (HPH) is characterized by sustained elevation of pulmonary artery pressure produced by vasoconstriction and hyperproliferative remodeling of the pulmonary artery and subsequent right ventricular hypertrophy (RVH). The search for therapeutic targets for cardiovascular pathophysiology has extended in many directions. However, studies focused on mitigating high-altitude pulmonary hypertension (HAPH) have been rare. Because AMP-activated protein kinase (AMPK) is involved in cardiovascular and metabolic pathology, AMPK is often studied as a potential therapeutic target. AMPK is best characterized as a sensor of cellular energy that can also restore cellular metabolic homeostasis. However, AMPK has been implicated in other pathways with vasculoprotective effects. Notably, cellular metabolic stress increases the intracellular ADP/ATP or AMP/ATP ratio, and AMPK activation restores ATP levels by activating energy-producing catabolic pathways and inhibiting energy-consuming anabolic pathways, such as cell growth and proliferation pathways, promoting cardiovascular protection. Thus, AMPK activation plays an important role in antiproliferative, antihypertrophic and antioxidant pathways in the pulmonary artery in HPH. However, AMPK plays contradictory roles in promoting HPH development. This review describes the main findings related to AMPK participation in HPH and its potential as a therapeutic target. It also extrapolates known AMPK functions to discuss the less-studied HAPH context.
Collapse
Affiliation(s)
- Karen Flores
- Institute of Health Studies, University Arturo Prat, Av. Arturo Prat 2120, Iquique 1110939, Chile; (P.S.); (J.B.)
- Institute DECIPHER, German-Chilean Institute for Research on Pulmonary Hypoxia and Its Health Sequelae, 20251 Hamburg, Germany and Iquique 1100000, Chile
- Correspondence: ; Tel.: +56-572526392
| | - Patricia Siques
- Institute of Health Studies, University Arturo Prat, Av. Arturo Prat 2120, Iquique 1110939, Chile; (P.S.); (J.B.)
- Institute DECIPHER, German-Chilean Institute for Research on Pulmonary Hypoxia and Its Health Sequelae, 20251 Hamburg, Germany and Iquique 1100000, Chile
| | - Julio Brito
- Institute of Health Studies, University Arturo Prat, Av. Arturo Prat 2120, Iquique 1110939, Chile; (P.S.); (J.B.)
- Institute DECIPHER, German-Chilean Institute for Research on Pulmonary Hypoxia and Its Health Sequelae, 20251 Hamburg, Germany and Iquique 1100000, Chile
| | - Silvia M. Arribas
- Department of Physiology, University Autonoma of Madrid, 28049 Madrid, Spain;
| |
Collapse
|
11
|
Dillard J, Meng X, Nelin L, Liu Y, Chen B. Nitric oxide activates AMPK by modulating PDE3A in human pulmonary artery smooth muscle cells. Physiol Rep 2021; 8:e14559. [PMID: 32914566 PMCID: PMC7507575 DOI: 10.14814/phy2.14559] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 08/05/2020] [Indexed: 01/17/2023] Open
Abstract
Phosphodiesterase 3 (PDE3), of which there are two isoforms, PDE3A and PDE3B, hydrolyzes cAMP and cGMP—cyclic nucleotides important in the regulation of pulmonary vascular tone. PDE3 has been implicated in pulmonary hypertension unresponsive to nitric oxide (NO); however, contributions of the two isoforms are not known. Furthermore, adenosine monophosphate‐activated protein kinase (AMPK), a critical regulator of cellular energy homeostasis, has been shown to be modulated by PDE3 in varying cell types. While AMPK has recently been implicated in pulmonary hypertension pathogenesis, its role and regulation in the pulmonary vasculature remain to be elucidated. Therefore, we utilized human pulmonary artery smooth muscle cells (hPASMC) to test the hypothesis that NO increases PDE3 expression in an isoform‐specific manner, thereby activating AMPK and inhibiting hPASMC proliferation. We found that in hPASMC, NO treatment increased PDE3A protein expression and PDE3 activity with a concomitant decrease in cAMP concentrations and increase in AMPK phosphorylation. Knockdown of PDE3A using siRNA transfection blunted the NO‐induced AMPK activation, indicating that PDE3A plays an important role in AMPK regulation in hPASMC. Treatment with a soluble guanylate cyclase (sGC) stimulator increased PDE3A expression and AMPK activation similar to that seen with NO treatment, whereas treatment with a sGC inhibitor blunted the NO‐induced increase in PDE3A and AMPK activation. These results suggest that NO increases PDE3A expression, decreases cAMP, and activates AMPK via the sGC‐cGMP pathway. We speculate that NO‐induced increases in PDE3A and AMPK may have implications in the pathogenesis and the response to therapies in pulmonary hypertensive disorders.
Collapse
Affiliation(s)
- Julie Dillard
- Pulmonary Hypertension Group, Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Xiaomei Meng
- Pulmonary Hypertension Group, Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Leif Nelin
- Pulmonary Hypertension Group, Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.,Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Yusen Liu
- Pulmonary Hypertension Group, Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.,Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Bernadette Chen
- Pulmonary Hypertension Group, Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.,Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
| |
Collapse
|
12
|
Zhao Q, Song P, Zou MH. AMPK and Pulmonary Hypertension: Crossroads Between Vasoconstriction and Vascular Remodeling. Front Cell Dev Biol 2021; 9:691585. [PMID: 34169079 PMCID: PMC8217619 DOI: 10.3389/fcell.2021.691585] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 05/18/2021] [Indexed: 12/25/2022] Open
Abstract
Pulmonary hypertension (PH) is a debilitating and life-threatening disease characterized by increased blood pressure within the pulmonary arteries. Adenosine monophosphate-activated protein kinase (AMPK) is a heterotrimeric serine-threonine kinase that contributes to the regulation of metabolic and redox signaling pathways. It has key roles in the regulation of cell survival and proliferation. The role of AMPK in PH is controversial because both inhibition and activation of AMPK are preventive against PH development. Some clinical studies found that metformin, the first-line antidiabetic drug and the canonical AMPK activator, has therapeutic efficacy during treatment of early-stage PH. Other study findings suggest the use of metformin is preferentially beneficial for treatment of PH associated with heart failure with preserved ejection fraction (PH-HFpEF). In this review, we discuss the "AMPK paradox" and highlight the differential effects of AMPK on pulmonary vasoconstriction and pulmonary vascular remodeling. We also review the effects of AMPK activators and inhibitors on rescue of preexisting PH in animals and include a discussion of gender differences in the response to metformin in PH.
Collapse
Affiliation(s)
| | | | - Ming-Hui Zou
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA, United States
| |
Collapse
|
13
|
Wang L, Zhang X, Cao Y, Ma Q, Mao X, Xu J, Yang Q, Zhou Y, Lucas R, Fulton DJ, Su Y, Barman SA, Hong M, Liu Z, Huo Y. Mice with a specific deficiency of Pfkfb3 in myeloid cells are protected from hypoxia-induced pulmonary hypertension. Br J Pharmacol 2021; 178:1055-1072. [PMID: 33300142 DOI: 10.1111/bph.15339] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 11/09/2020] [Accepted: 11/23/2020] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND AND PURPOSE Macrophage infiltration into the lungs is a characteristic of pulmonary hypertension (PH). Glycolysis is the main metabolic pathway for macrophage activation. However, the effect of macrophage glycolysis on the development of PH remains unknown. We investigated the effect of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKBF3), a critical enzyme of macrophage glycolysis, on PH development. EXPERIMENTAL APPROACH Lung tissues from PH patients were examined by immunostaining with macrophage markers. PH was induced in Wistar rats with SU5416/hypoxia and in mice with hypoxia. Lungs and macrophages were isolated for analysis by RT-PCR, western blot, flow cytometry, and immunostaining. KEY RESULTS Expression of glycolytic molecules was increased in circulating peripheral blood mononuclear cells (PBMCs) and lung macrophages of PH patients. These results were also found in lung macrophages of SU5416/hypoxia (Su/Hx)-induced PH rats and hypoxia-induced PH mice. PH was ameliorated in myeloid-specific Pfkfb3-deficient mice (Pfkfb3ΔMϕ ) or mice treated with the PFKFB3 inhibitor 3PO, compared with their controls. Alveolar macrophages of PH Pfkfb3ΔMϕ mice produced lower levels of growth factors and pro-inflammatory cytokines than those of control mice. Circulating myeloid cells and lung myeloid cells were much fewer in PH Pfkfb3ΔMϕ mice than controls. Mechanistically, overexpression of Hif1a or Hif2a in bone marrow-derived macrophages (BMDMs) cultured with bone marrow of Pfkfb3ΔMϕ mice restored the decreased expression of pro-inflammatory cytokines and growth factors. CONCLUSIONS AND IMPLICATIONS Myeloid Pfkfb3 deficiency protects mice from PH, thereby suggesting that myeloid PFKFB3 is one of the important targets in the therapeutic effect of PFKFB3 inhibition in PH treatment.
Collapse
Affiliation(s)
- Lina Wang
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Xiaoyu Zhang
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Yapeng Cao
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Qian Ma
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Xiaoxiao Mao
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Jiean Xu
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Qiuhua Yang
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Yaqi Zhou
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Rudolf Lucas
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - David J Fulton
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Yunchao Su
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Scott A Barman
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Mei Hong
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Zhiping Liu
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Yuqing Huo
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| |
Collapse
|
14
|
Yoshida T, Matsuura K, Goya S, Ma D, Shimada K, Kitpipatkun P, Namiki R, Uemura A, Suzuki K, Tanaka R. Metformin prevents the development of monocrotaline-induced pulmonary hypertension by decreasing serum levels of big endothelin-1. Exp Ther Med 2020; 20:149. [PMID: 33093887 PMCID: PMC7571338 DOI: 10.3892/etm.2020.9278] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 01/16/2020] [Indexed: 12/20/2022] Open
Abstract
Pulmonary hypertension (PH) is a disease with poor prognosis, and it is characterized by the progressive elevation of pulmonary vascular resistance and pressure. Various factors are associated with the pathology of PH, including AMP-activated protein kinase (AMPK) deficiency. The present study aimed to evaluate the therapeutic effect of metformin, an AMPK activator, in a monocrotaline (MCT)-induced PH rat model. Rats were randomly divided into the following three groups: i) Saline-injected group (sham group); ii) monocrotaline (MCT)-injected group (PH group); iii) MCT-injected and metformin-treated group (MT group). Four weeks following MCT injection, cardiac ultrasonography, invasive hemodynamic measurements, measurement of serum levels of big endothelin-1 (big ET-1) and histological analysis were performed to evaluate the effect of metformin treatment in PH. Pulmonary arterial pressure and serum big ET-1 concentrations were reduced in the MT group compared with the PH group. Medial wall thickness and wall area of the pulmonary arterioles in the MT group were decreased compared with the PH group. Comparing the right heart functional parameters among groups revealed that the acceleration time/ejection time ratio improved in the MT group compared with the PH group. Thus, the present study demonstrated the efficacy of metformin in an MCT-induced PH rat model and suggested that metformin may be a valuable, potential novel therapeutic for the treatment of PH.
Collapse
Affiliation(s)
- Tomohiko Yoshida
- Department of Veterinary Surgery, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan
| | - Katsuhiro Matsuura
- Department of Veterinary Surgery, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan
| | - Seijirow Goya
- Department of Veterinary Surgery, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan
| | - Danfu Ma
- Department of Veterinary Surgery, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan
| | - Kazumi Shimada
- Department of Veterinary Surgery, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan
| | - Pitipat Kitpipatkun
- Department of Veterinary Surgery, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan
| | - Ryosuke Namiki
- Department of Veterinary Surgery, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan
| | - Akiko Uemura
- Department of Veterinary Surgery, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan
| | - Kazuhiko Suzuki
- Department of Veterinary Toxicology, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan
| | - Ryou Tanaka
- Department of Veterinary Surgery, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan
| |
Collapse
|
15
|
Hu L, Li L, Chang Q, Fu S, Qin J, Chen Z, Li X, Liu Q, Hu G, Li Q. Discovery of Novel Pyrazolo[3,4- b] Pyridine Derivatives with Dual Activities of Vascular Remodeling Inhibition and Vasodilation for the Treatment of Pulmonary Arterial Hypertension. J Med Chem 2020; 63:11215-11234. [PMID: 32914624 DOI: 10.1021/acs.jmedchem.0c01132] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Current pulmonary arterial hypertension (PAH) therapeutic strategies mainly focus on vascular relaxation with less emphasis on vascular remodeling, which results in poor prognosis. Hence, dual pathway regulators with vasodilation effect via soluble guanylate cyclase (sGC) stimulation and vascular remodeling regulation effect by AMP-activated protein kinase (AMPK) inhibition provide more advantages and potentialities. Herein, we designed and synthesized a series of novel pyrazolo[3,4-b] pyridine derivatives based on sGC stimulator and AMPK inhibitor scaffolds. In vitro, 2 exhibited moderate vasodilation activity and higher proliferation and migration suppressive effects compared to riociguat. In vivo, 2 significantly decreased right ventricular systolic pressure (RVSP), attenuated pulmonary artery medial thickness (PAMT), and right ventricular hypertrophy (RVH) in hypoxia-induced PAH rat models (i.g.). Given the unique advantages of significant vascular remodeling inhibition and moderate vascular relaxation based on the dual pathway regulation, we proposed 2 as a promising lead for anti-PAH drug discovery.
Collapse
Affiliation(s)
- Liqing Hu
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013 Hunan, China.,Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, 23298 Virginia, United States
| | - Lijun Li
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013 Hunan, China
| | - Qi Chang
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013 Hunan, China
| | - Songsen Fu
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013 Hunan, China
| | - Jia Qin
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013 Hunan, China
| | - Zhuo Chen
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013 Hunan, China
| | - Xiaohui Li
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013 Hunan, China
| | - Qinglian Liu
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, 23298 Virginia, United States
| | - Gaoyun Hu
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013 Hunan, China
| | - Qianbin Li
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013 Hunan, China
| |
Collapse
|
16
|
Hu L, Zhao R, Liu Q, Li Q. New Insights Into Heat Shock Protein 90 in the Pathogenesis of Pulmonary Arterial Hypertension. Front Physiol 2020; 11:1081. [PMID: 33041844 PMCID: PMC7522509 DOI: 10.3389/fphys.2020.01081] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 08/05/2020] [Indexed: 12/21/2022] Open
Affiliation(s)
- Liqing Hu
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Rui Zhao
- The First Clinical School, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qinglian Liu
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Qianbin Li
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
- *Correspondence: Qianbin Li,
| |
Collapse
|
17
|
Ornatowski W, Lu Q, Yegambaram M, Garcia AE, Zemskov EA, Maltepe E, Fineman JR, Wang T, Black SM. Complex interplay between autophagy and oxidative stress in the development of pulmonary disease. Redox Biol 2020; 36:101679. [PMID: 32818797 PMCID: PMC7451718 DOI: 10.1016/j.redox.2020.101679] [Citation(s) in RCA: 227] [Impact Index Per Article: 45.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/20/2020] [Accepted: 08/04/2020] [Indexed: 12/16/2022] Open
Abstract
The autophagic pathway involves the encapsulation of substrates in double-membraned vesicles, which are subsequently delivered to the lysosome for enzymatic degradation and recycling of metabolic precursors. Autophagy is a major cellular defense against oxidative stress, or related conditions that cause accumulation of damaged proteins or organelles. Selective forms of autophagy can maintain organelle populations or remove aggregated proteins. Dysregulation of redox homeostasis under pathological conditions results in excessive generation of reactive oxygen species (ROS), leading to oxidative stress and the associated oxidative damage of cellular components. Accumulating evidence indicates that autophagy is necessary to maintain redox homeostasis. ROS activates autophagy, which facilitates cellular adaptation and diminishes oxidative damage by degrading and recycling intracellular damaged macromolecules and dysfunctional organelles. The cellular responses triggered by oxidative stress include the altered regulation of signaling pathways that culminate in the regulation of autophagy. Current research suggests a central role for autophagy as a mammalian oxidative stress response and its interrelationship to other stress defense systems. Altered autophagy phenotypes have been observed in lung diseases such as chronic obstructive lung disease, acute lung injury, cystic fibrosis, idiopathic pulmonary fibrosis, and pulmonary arterial hypertension, and asthma. Understanding the mechanisms by which ROS regulate autophagy will provide novel therapeutic targets for lung diseases. This review highlights our current understanding on the interplay between ROS and autophagy in the development of pulmonary disease.
Collapse
Affiliation(s)
- Wojciech Ornatowski
- Department of Medicine, The University of Arizona Health Sciences, Tucson, AZ, USA
| | - Qing Lu
- Department of Medicine, The University of Arizona Health Sciences, Tucson, AZ, USA
| | | | - Alejandro E Garcia
- Department of Medicine, The University of Arizona Health Sciences, Tucson, AZ, USA
| | - Evgeny A Zemskov
- Department of Medicine, The University of Arizona Health Sciences, Tucson, AZ, USA
| | - Emin Maltepe
- Department of Pediatrics, The University of California, San Francisco, San Francisco, CA, USA
| | - Jeffrey R Fineman
- Department of Pediatrics, The University of California, San Francisco, San Francisco, CA, USA; Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Ting Wang
- Department of Internal Medicine, The University of Arizona Health Sciences, Phoenix, AZ, USA
| | - Stephen M Black
- Department of Medicine, The University of Arizona Health Sciences, Tucson, AZ, USA.
| |
Collapse
|
18
|
Wang HL, Tang FQ, Jiang YH, Zhu Y, Jian Z, Xiao YB. AMPKα2 deficiency exacerbates hypoxia-induced pulmonary hypertension by promoting pulmonary arterial smooth muscle cell proliferation. J Physiol Biochem 2020; 76:445-456. [PMID: 32592088 DOI: 10.1007/s13105-020-00742-4] [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] [Received: 03/14/2019] [Accepted: 04/23/2020] [Indexed: 12/13/2022]
Abstract
Increased evidence indicates that adenosine monophosphate-activated protein kinase (AMPK) plays a vital role in vascular homeostasis, especially under hypoxia, and protects against the progression of pulmonary hypertension (PH). However, the role of AMPK in the pathogenesis of PH remains to be clarified. In the present study, we confirmed that a loss of AMPKα2 exacerbated the development of PH by using hypoxia-induced PH model in AMPKα2 -/- mice. After a 4-week period of hypoxic exposure, AMPKα2 -/- mice exhibited more severe pulmonary vascular remodeling and pulmonary vascular smooth muscle cell (SMC) proliferation when compared with wild type (WT) mice. In vitro, AMPKα2 knockdown promoted the proliferation of pulmonary arterial smooth muscle cells (PASMCs) under hypoxia. This phenomenon was accompanied by upregulated Skp2 and downregulated p27kip1 expression and was abolished by rapamycin, an inhibitor of mTOR. These results indicate that AMPKα2 deficiency exacerbates hypoxia-induced PH by promoting PASMC proliferation via the mTOR/Skp2/p27kip1 signaling axis. Therefore, enhanced AMPKα2 activity might underlie a novel therapeutic strategy for the management of PH.
Collapse
Affiliation(s)
- Hai-Long Wang
- Department of Cardiovascular Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, People's Republic of China
| | - Fu-Qin Tang
- Department of Cardiovascular Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, People's Republic of China
| | - Yun-Han Jiang
- Department of Cardiovascular Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, People's Republic of China
| | - Yu Zhu
- Department of Cardiovascular Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, People's Republic of China
| | - Zhao Jian
- Department of Cardiovascular Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, People's Republic of China.
| | - Ying-Bin Xiao
- Department of Cardiovascular Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, People's Republic of China.
| |
Collapse
|
19
|
Bao YR, Chen JW, Jiang Y, Wang LH, Xue R, Qian JX, Zhang GX. Sodium Tanshinone II Sulfonate A Ameliorates Hypoxia-Induced Pulmonary Hypertension. Front Pharmacol 2020; 11:687. [PMID: 32508639 PMCID: PMC7253651 DOI: 10.3389/fphar.2020.00687] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 04/27/2020] [Indexed: 12/12/2022] Open
Abstract
Background Pulmonary hypertension (PH) remains a prevalent disease globally. Sodium tanshinone II sulfonate A (STS) has been used in clinical treatment of PH. Aims The aim of the present study was to investigate the effect of sodium STS treatment on hypoxia-induced PH and related mechanisms. Methods Male Sprague-Dawley rats were housed in a hypoxic chamber with an oxygen concentration of 10 ± 1% for 8 h a day over 21 days. Rats were treated with either STS (low-dose: 10 mg/kg or high-dose: 30 mg/kg) or LY294002 (which is an inhibitor of PI3K). Pulmonary arterial pressure (PAP) was measured, right ventricular hypertrophy parameters were monitored, lung edema parameters were measured, and pathological changes were observed by hematoxylin-eosin (HE) staining. Protein expressions of apoptosis, and PI3K/AKT/mTOR/autophagy pathways in rat lung tissue were examined by western blot. Levels of the pro-inflammatory factors IL-6, IL-8, TNF-α in lung tissues of rats were measured using an enzyme linked immunosorbent assay (ELISA). Results Results of our study demonstrate that persistent exposure to hypoxic conditions increased PAP, right ventricular hypertrophy, lung edema, parameters of lung vascular proliferation and decreased the ratio of Bax/Bcl-2. Furthermore, hypoxic conditions activated the PI3K/Akt/mTOR pathway, inhibited autophagy, and elevated abundance of inflammatory factors in rat lung tissue. Treatment with STS resulted in a dose-dependent decrease in PAP, right ventricular hypertrophy, lung edema, lung vascular proliferation and reversed hypoxia induced lung tissue protein expression and pro-inflammatory factors in rat lung tissue. In addition, hypoxia-induced increases in PAP, cardiac hypertrophy, and lung expression of the proteins PI3K/Akt/mTOR/autophagy pathway were partially reversed by treatment with LY294002. Conclusions STS alleviates hypoxia-induced PH by promoting apoptosis, inhibiting PI3K/AKT/mTOR pathway, up-regulating autophagy, and inhibiting inflammatory responses.
Collapse
Affiliation(s)
- Ya-Ru Bao
- Department of Physiology, Medical College of Soochow University, Suzhou, China
| | - Jing-Wei Chen
- Department of Internal Medicine, Suzhou TCM Hospital affiliated to Nanjing University of Chinese Medicine, Suzhou, China
| | - Yan Jiang
- Department of Physiology, Medical College of Soochow University, Suzhou, China
| | - Lin-Hui Wang
- Department of Physiology, Medical College of Soochow University, Suzhou, China
| | - Rong Xue
- Department of Physiology, Medical College of Soochow University, Suzhou, China
| | - Jin-Xian Qian
- Department of Respiratory and Critical Care Medicine, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| | - Guo-Xing Zhang
- Department of Physiology, Medical College of Soochow University, Suzhou, China
| |
Collapse
|
20
|
Wang Y, Zhang X, Chen W, Gao L, Li J, Song T, Chi J, Zhang X, Shi Z, Dong Y, Yin X, Liu Y. Cortistatin ameliorates Ang II-induced proliferation of vascular smooth muscle cells by inhibiting autophagy through SSTR3 and SSTR5. Life Sci 2020; 253:117726. [PMID: 32348837 DOI: 10.1016/j.lfs.2020.117726] [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] [Received: 10/08/2019] [Revised: 04/16/2020] [Accepted: 04/23/2020] [Indexed: 12/24/2022]
Abstract
AIMS Vascular smooth muscle cell (VSMC) proliferation plays a significant role in the development of various vascular disorders. However, the effect of cortistatin (CST) on VSMC proliferation remains unclear. Therefore, the purpose of our research aimed to study whether CST protected VSMCs from angiotensin II (Ang II)-induced proliferation and which mechanisms participated in the process. MAIN METHODS Cultured rat VSMCs were treated with Ang II with or without CST for 24 h. Cell proliferation rate was measured by cell counting kit-8 (CCK8) assay. The expressions of CST and its receptors were assessed by quantitative real-time PCR (qRT-PCR). The protein expression levels were analyzed by western blots. Immunofluorescence and transmission electron microscopy (TEM) were used to observe autophagy. KEY FINDINGS Our results showed that different concentrations of CST alleviated the Ang II-induced VSMC proliferation. The autophagy and reactive oxygen species (ROS) stimulated by Ang II were attenuated by CST. Furthermore, when the autophagy inhibitor 3-methyladenine (3-MA) was added, it exerted similar inhibition effects like CST, but didn't augment the protective role of CST on Ang II-induced VSMC autophagy and proliferation. Moreover, blocking somatostatin receptor 3 and 5 (SSTR3 and SSTR5) partially abrogated the suppressive effect of CST on Ang II-stimulated VSMC proliferation and autophagy. SIGNIFICANCE This study indicated that CST could ameliorate Ang II-stimulated VSMC proliferation by inhibiting autophagy partially through its receptors SSTR3 and SSTR5, providing a reasonable evidence for CST as a novel perspective therapeutic target of vascular diseases.
Collapse
Affiliation(s)
- Ying Wang
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xin Zhang
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wenjia Chen
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Lei Gao
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jihe Li
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Tao Song
- Department of Cadre, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jinyu Chi
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaohui Zhang
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhiyu Shi
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yanghong Dong
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xinhua Yin
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Yue Liu
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
21
|
De Munck DG, De Meyer GR, Martinet W. Autophagy as an emerging therapeutic target for age-related vascular pathologies. Expert Opin Ther Targets 2020; 24:131-145. [PMID: 31985292 DOI: 10.1080/14728222.2020.1723079] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Introduction: The incidence of age-related vascular diseases such as arterial stiffness, hypertension and atherosclerosis, is rising dramatically and is substantially impacting healthcare systems. Mounting evidence suggests that there is an important role for autophagy in maintaining (cardio)vascular health. Impaired vascular autophagy has been linked to arterial aging and the initiation of vascular disease.Areas covered: The function and implications of autophagy in vascular smooth muscle cells and endothelial cells are discussed in healthy blood vessels and arterial disease. Furthermore, we discuss current treatment options for vascular disease and their links with autophagy. A literature search was conducted in PubMed up to October 2019.Expert opinion: Although the therapeutic potential of inducing autophagy in age-related vascular pathologies is considerable, several issues should be addressed before autophagy induction can be clinically used to treat vascular disease. These issues include uncertainty regarding the most effective drug target as well as the lack of potency and selectivity of autophagy inducing drugs. Moreover, drug tolerance or autophagy mediated cell death have been reported as possible adverse effects. Special attention is required for determining the cause of autophagy deficiency to optimize the treatment strategy.
Collapse
Affiliation(s)
- Dorien G De Munck
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Guido Ry De Meyer
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Wim Martinet
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
22
|
Wu M, Wu Y, Huang J, Wu Y, Wu H, Jiang B, Zhuang J. Protein expression profile changes of lung tissue in patients with pulmonary hypertension. PeerJ 2020; 8:e8153. [PMID: 32030316 PMCID: PMC6996500 DOI: 10.7717/peerj.8153] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 11/04/2019] [Indexed: 12/17/2022] Open
Abstract
Background Pulmonary hypertension occurs in approximately 1% of the global population, and the prognosis for such patients may be poor. However, the mechanisms underlying the development of this disease remain unclear. Thus, understanding the development of pulmonary hypertension and finding new therapeutic targets and approaches are important for improved clinical outcomes. Methods Lung tissue specimens were collected from six patients with atrial septal defect and pulmonary hypertension (all women, with a mean age of 46.5 ± 4.7 years, and their condition could not be corrected with an internal medical occlusion device) and from nine control patients with lung cancer who underwent lobectomy (six men and three women, with a mean age of 56.7 ± 1.7 years). Isobaric tags for relative and absolute quantitation and liquid chromatography tandem mass spectrometry analyses were used to detect protein expression levels. Results We found 74 significantly upregulated and 88 significantly downregulated differentially expressed proteins between control and pulmonary hypertensive lung tissue specimens. Gene ontology analyses identified the top 20 terms in all three categories, that is, biological process, cellular component, and molecular function. Kyoto Encyclopedia of Genes and Genomes and protein–protein interaction analyses determined the top 10 signaling pathways and found that the six hub proteins associated with the differentially expressed upregulated proteins (PRKAA1, DHPR, ACTB, desmin, ACTG1, and ITGA1) were all involved in hypertrophic cardiomyopathy, arrhythmogenic right ventricular cardiomyopathy, and dilated cardiomyopathy. Conclusion Our results identified protein expression profile changes in lung tissue derived from patients with pulmonary hypertension, providing potential new biomarkers for clinical diagnosis and prognosis for patients with pulmonary hypertension and offering candidate protein targets for future therapeutic drug development.
Collapse
Affiliation(s)
- Min Wu
- Department of Cardiac Surgery, Guangdong Provincial People's Hospital, Guangzhou City, Guangdong Province, China
| | - Yijin Wu
- Department of Cardiac Surgery, Guangdong Provincial People's Hospital, Guangzhou City, Guangdong Province, China
| | - Jinsong Huang
- Department of Cardiac Surgery, Guangdong Provincial People's Hospital, Guangzhou City, Guangdong Province, China
| | - Yueheng Wu
- Department of Cardiac Surgery, Guangdong Provincial People's Hospital, Guangzhou City, Guangdong Province, China
| | - Hongmei Wu
- Department of Cardiac Surgery, Guangdong Provincial People's Hospital, Guangzhou City, Guangdong Province, China
| | - Benyuan Jiang
- Department of Cardiac Surgery, Guangdong Provincial People's Hospital, Guangzhou City, Guangdong Province, China
| | - Jian Zhuang
- Department of Cardiac Surgery, Guangdong Provincial People's Hospital, Guangzhou City, Guangdong Province, China
| |
Collapse
|
23
|
Zhang X, Liu Q, Zhang C, Sheng J, Li S, Li W, Yang X, Wang X, He S, Bai J, Zhu D. Puerarin prevents progression of experimental hypoxia-induced pulmonary hypertension via inhibition of autophagy. J Pharmacol Sci 2019; 141:97-105. [PMID: 31640920 DOI: 10.1016/j.jphs.2019.09.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 09/15/2019] [Accepted: 09/18/2019] [Indexed: 01/07/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is defined as elevation of mean pulmonary arterial pressure to ≥25 mmHg within the low pressure pulmonary circulatory system. PAH is characterized by obstructive vascular remodeling, partially due to excessive pulmonary arterial smooth muscle cell (PASMC) proliferation. Puerarin is a natural flavonoid isolated from the herb Radix puerariae, which has been widely used for the treatment of cardiovascular and cerebrovascular disorders and diabetes. However, how puerarin mediates autophagy in the progression of pulmonary vascular remodeling is unclear. In this study, we explored the effects of puerarin in a hypoxic pulmonary hypertension (PH) rat model using immunohistochemistry, and morphometric analyses of right ventricle. In addition, cell counting kit 8 assay, western blotting and flow cytometry were employed to test cell proliferation in PASMCs, and then autophagy was tested with mRFP-GFP-LC3 fluorescence microscopy and Western blot. We found that puerarin could alleviate hypoxia-induced PH in rats and improved pulmonary histopathology, and also reduced the expression of autophagy markers in vivo and in vitro. Moreover, puerarin also ameliorated hypoxia-induced PASMC proliferation in an autophagy-dependent manner. Overall, these findings demonstrated that puerarin could prevent hypoxia-induced PH in rats, possibly via reducing autophagy and suppressing cell proliferation.
Collapse
Affiliation(s)
- Xiaodan Zhang
- College of Pharmacy, Harbin University of Commerce, Harbin, 150076, PR China.
| | - Qi Liu
- College of Pharmacy, Harbin University of Commerce, Harbin, 150076, PR China.
| | - Chen Zhang
- College of Pharmacy, Harbin University of Commerce, Harbin, 150076, PR China.
| | - Jiejing Sheng
- College of Pharmacy, Harbin University of Commerce, Harbin, 150076, PR China.
| | - Songlin Li
- College of Pharmacy, Harbin University of Commerce, Harbin, 150076, PR China.
| | - Wendi Li
- College of Pharmacy, Harbin University of Commerce, Harbin, 150076, PR China.
| | - Xinying Yang
- College of Pharmacy, Harbin University of Commerce, Harbin, 150076, PR China.
| | - Xiaoying Wang
- Central Laboratory of Harbin Medical University (Daqing), Daqing, 163319, PR China.
| | - Siyu He
- Central Laboratory of Harbin Medical University (Daqing), Daqing, 163319, PR China.
| | - June Bai
- Central Laboratory of Harbin Medical University (Daqing), Daqing, 163319, PR China.
| | - Daling Zhu
- Central Laboratory of Harbin Medical University (Daqing), Daqing, 163319, PR China; College of Pharmacy, Harbin Medical University, Harbin, 150081, PR China; State Province Key Laboratories of Biomedicine-Pharmaceutics of China, Daqing, 163319, PR China.
| |
Collapse
|
24
|
Grootaert MOJ, Moulis M, Roth L, Martinet W, Vindis C, Bennett MR, De Meyer GRY. Vascular smooth muscle cell death, autophagy and senescence in atherosclerosis. Cardiovasc Res 2019; 114:622-634. [PMID: 29360955 DOI: 10.1093/cvr/cvy007] [Citation(s) in RCA: 383] [Impact Index Per Article: 63.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 01/17/2018] [Indexed: 12/14/2022] Open
Abstract
In the present review, we describe the causes and consequences of loss of vascular smooth muscle cells (VSMCs) or their function in advanced atherosclerotic plaques and discuss possible mechanisms such as cell death or senescence, and induction of autophagy to promote cell survival. We also highlight the potential use of pharmacological modulators of these processes to limit plaque progression and/or improve plaque stability. VSMCs play a pivotal role in atherogenesis. Loss of VSMCs via initiation of cell death leads to fibrous cap thinning and promotes necrotic core formation and calcification. VSMC apoptosis is induced by pro-inflammatory cytokines, oxidized low density lipoprotein, high levels of nitric oxide and mechanical injury. Apoptotic VSMCs are characterized by a thickened basal lamina surrounding the cytoplasmic remnants of the VSMC. Inefficient clearance of apoptotic VSMCs results in secondary necrosis and subsequent inflammation. A critical determinant in the VSMC stress response and phenotypic switching is autophagy, which is activated by various stimuli, including reactive oxygen and lipid species, cytokines, growth factors and metabolic stress. Successful autophagy stimulates VSMC survival, whereas reduced autophagy promotes age-related changes in the vasculature. Recently, an interesting link between autophagy and VSMC senescence has been uncovered. Defective VSMC autophagy accelerates not only the development of stress-induced premature senescence but also atherogenesis, albeit without worsening plaque stability. VSMC senescence in atherosclerosis is likely a result of replicative senescence and/or stress-induced premature senescence in response to DNA damaging and/or oxidative stress-inducing stimuli. The finding that VSMC senescence can promote atherosclerosis further illustrates that normal, adequate VSMC function is crucial in protecting the vessel wall against atherosclerosis.
Collapse
Affiliation(s)
- Mandy O J Grootaert
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Box 110, Addenbrooke's Centre for Clinical Investigation, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Manon Moulis
- INSERM, UMR-1048, Institute of Metabolic and Cardiovascular Diseases and University Paul Sabatier, F-31342 Toulouse, France
| | - Lynn Roth
- Laboratory of Physiopharmacology, University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium
| | - Wim Martinet
- Laboratory of Physiopharmacology, University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium
| | - Cécile Vindis
- INSERM, UMR-1048, Institute of Metabolic and Cardiovascular Diseases and University Paul Sabatier, F-31342 Toulouse, France
| | - Martin R Bennett
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Box 110, Addenbrooke's Centre for Clinical Investigation, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Guido R Y De Meyer
- Laboratory of Physiopharmacology, University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium
| |
Collapse
|
25
|
Zhang CF, Zhao FY, Xu SL, Liu J, Xing XQ, Yang J. Autophagy in pulmonary hypertension: Emerging roles and therapeutic implications. J Cell Physiol 2019; 234:16755-16767. [PMID: 30932199 DOI: 10.1002/jcp.28531] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 02/21/2019] [Accepted: 03/06/2019] [Indexed: 02/05/2023]
Abstract
Autophagy is an important mechanism for cellular self-digestion and basal homeostasis. This gene- and modulator-regulated pathway is conserved in cells. Recently, several studies have shown that autophagic dysfunction is associated with pulmonary hypertension (PH). However, the relationship between autophagy and PH remains controversial. In this review, we mainly introduce the effects of autophagy-related genes and some regulatory molecules on PH and the relationship between autophagy and PH under the conditions of hypoxia, monocrotaline injection, thromboembolic stress, oxidative stress, and other drugs and toxins. The effects of other autophagy-related drugs, such as chloroquine, 3-methyladenine, rapamycin, and other potential therapeutic drugs and targets, in PH are also described.
Collapse
Affiliation(s)
- Chun-Fang Zhang
- Department of Respiratory Medicine, The Fourth Affiliated Hospital of Kunming Medical University, The Second People's Hospital of Yunnan, Kunming, Yunnan, China
| | - Fang-Yun Zhao
- Department of Pharmacy, Yan'An Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China
| | - Shuang-Lan Xu
- Department of Respiratory Medicine, The Fourth Affiliated Hospital of Kunming Medical University, The Second People's Hospital of Yunnan, Kunming, Yunnan, China
| | - Jie Liu
- Department of Respiratory Medicine, The Fourth Affiliated Hospital of Kunming Medical University, The Second People's Hospital of Yunnan, Kunming, Yunnan, China
| | - Xi-Qian Xing
- Department of Respiratory Medicine, The Fourth Affiliated Hospital of Kunming Medical University, The Second People's Hospital of Yunnan, Kunming, Yunnan, China
| | - Jiao Yang
- First Department of Respiratory Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
26
|
Moral-Sanz J, Lewis SA, MacMillan S, Ross FA, Thomson A, Viollet B, Foretz M, Moran C, Hardie DG, Evans AM. The LKB1-AMPK-α1 signaling pathway triggers hypoxic pulmonary vasoconstriction downstream of mitochondria. Sci Signal 2018; 11:11/550/eaau0296. [PMID: 30279167 DOI: 10.1126/scisignal.aau0296] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Hypoxic pulmonary vasoconstriction (HPV), which aids ventilation-perfusion matching in the lungs, is triggered by mechanisms intrinsic to pulmonary arterial smooth muscles. The unique sensitivity of these muscles to hypoxia is conferred by mitochondrial cytochrome c oxidase subunit 4 isoform 2, the inhibition of which has been proposed to trigger HPV through increased generation of mitochondrial reactive oxygen species. Contrary to this model, we have shown that the LKB1-AMPK-α1 signaling pathway is critical to HPV. Spectral Doppler ultrasound revealed that deletion of the AMPK-α1 catalytic subunit blocked HPV in mice during mild (8% O2) and severe (5% O2) hypoxia, whereas AMPK-α2 deletion attenuated HPV only during severe hypoxia. By contrast, neither of these genetic manipulations affected serotonin-induced reductions in pulmonary vascular flow. HPV was also attenuated by reduced expression of LKB1, a kinase that activates AMPK during energy stress, but not after deletion of CaMKK2, a kinase that activates AMPK in response to increases in cytoplasmic Ca2+ Fluorescence imaging of acutely isolated pulmonary arterial myocytes revealed that AMPK-α1 or AMPK-α2 deletion did not affect mitochondrial membrane potential during normoxia or hypoxia. However, deletion of AMPK-α1, but not of AMPK-α2, blocked hypoxia from inhibiting KV1.5, the classical "oxygen-sensing" K+ channel in pulmonary arterial myocytes. We conclude that LKB1-AMPK-α1 signaling pathways downstream of mitochondria are critical for the induction of HPV, in a manner also supported by AMPK-α2 during severe hypoxia.
Collapse
Affiliation(s)
- Javier Moral-Sanz
- Centre for Discovery Brain Sciences and Cardiovascular Science, College of Medicine and Veterinary Medicine, Hugh Robson Building, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Sophronia A Lewis
- Centre for Discovery Brain Sciences and Cardiovascular Science, College of Medicine and Veterinary Medicine, Hugh Robson Building, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Sandy MacMillan
- Centre for Discovery Brain Sciences and Cardiovascular Science, College of Medicine and Veterinary Medicine, Hugh Robson Building, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Fiona A Ross
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK
| | - Adrian Thomson
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Benoit Viollet
- Institut Cochin, INSERM U1016, Sorbonne Paris cité, 75014 Paris, France.,CNRS UMR 8104, Sorbonne Paris cité, 75014 Paris, France.,Université Paris Descartes, Sorbonne Paris cité, 75014 Paris, France
| | - Marc Foretz
- Institut Cochin, INSERM U1016, Sorbonne Paris cité, 75014 Paris, France.,CNRS UMR 8104, Sorbonne Paris cité, 75014 Paris, France.,Université Paris Descartes, Sorbonne Paris cité, 75014 Paris, France
| | - Carmel Moran
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - D Grahame Hardie
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK
| | - A Mark Evans
- Centre for Discovery Brain Sciences and Cardiovascular Science, College of Medicine and Veterinary Medicine, Hugh Robson Building, University of Edinburgh, Edinburgh EH8 9XD, UK.
| |
Collapse
|
27
|
Dai J, Zhou Q, Chen J, Rexius-Hall ML, Rehman J, Zhou G. Alpha-enolase regulates the malignant phenotype of pulmonary artery smooth muscle cells via the AMPK-Akt pathway. Nat Commun 2018; 9:3850. [PMID: 30242159 PMCID: PMC6155017 DOI: 10.1038/s41467-018-06376-x] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 09/03/2018] [Indexed: 12/20/2022] Open
Abstract
The molecular mechanisms underlying the metabolic shift toward increased glycolysis observed in pulmonary artery smooth muscle cells (PASMC) during the pathogenesis of pulmonary arterial hypertension (PAH) are not fully understood. Here we show that the glycolytic enzyme α-enolase (ENO1) regulates the metabolic reprogramming and malignant phenotype of PASMC. We show that ENO1 levels are elevated in patients with associated PAH and in animal models of hypoxic pulmonary hypertension (HPH). The silencing or inhibition of ENO1 decreases PASMC proliferation and de-differentiation, and induces PASMC apoptosis, whereas the overexpression of ENO1 promotes a synthetic, de- differentiated, and apoptotic-resistant phenotype via the AMPK-Akt pathway. The suppression of ENO1 prevents the hypoxia-induced metabolic shift from mitochondrial respiration to glycolysis in PASMC. Finally, we find that pharmacological inhibition of ENO1 reverses HPH in mice and rats, suggesting ENO1 as a regulator of pathogenic metabolic reprogramming in HPH.
Collapse
Affiliation(s)
- Jingbo Dai
- Department of Pediatrics, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Qiyuan Zhou
- Department of Pediatrics, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Jiwang Chen
- Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Megan L Rexius-Hall
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Jalees Rehman
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, 60612, USA
- Division of Cardiology, Department of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Guofei Zhou
- Department of Pediatrics, University of Illinois at Chicago, Chicago, IL, 60612, USA.
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China.
| |
Collapse
|
28
|
|
29
|
Dai J, Zhou Q, Tang H, Chen T, Li J, Raychaudhuri P, Yuan JXJ, Zhou G. Smooth muscle cell-specific FoxM1 controls hypoxia-induced pulmonary hypertension. Cell Signal 2018; 51:119-129. [PMID: 30092353 DOI: 10.1016/j.cellsig.2018.08.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 08/02/2018] [Accepted: 08/02/2018] [Indexed: 12/17/2022]
Abstract
RATIONALE Forkhead box M1 (FoxM1) is a transcription factor that promotes cell proliferation by regulating a broad spectrum of genes that participate in cell cycle regulation, such as Cyclin B, CDC25B, and Aurora B Kinase. We have shown that hypoxia, a well-known stimulus for pulmonary hypertension (PH), induces FoxM1 in pulmonary artery smooth muscle cells (PASMC) in a HIF-dependent pathway, resulting in PASMC proliferation, while the suppression of FoxM1 prevents hypoxia-induced PASMC proliferation. However, the implications of FoxM1 in the development of PH remain less known. METHODS We determined FoxM1 levels in the lung samples of idiopathic PAH (pulmonary arterial hypertension) (IPAH) patients and hypoxia-induced PH mice. We generated constitutive and inducible smooth muscle cell (SMC)-specific FoxM1 knockdown or knockout mice as well as FoxM1 transgenic mice which overexpress FoxM1, and exposed them to hypoxia (10% O2, 90% N2) or normoxia (Room air, 21% oxygen) for four weeks, and measured PH indices. We also isolated mouse PASMC (mPASMC) and mouse embryonic fibroblasts (MEF) from these mice to examine the cell proliferation and expression levels of SMC contractile proteins. RESULTS We showed that in hypertensive human lungs or mouse lungs, FoxM1 levels were elevated. Constitutive knockout of FoxM1 in mouse SMC caused early lethality, whereas constitutive knockdown of FoxM1 in mouse SMC prevented hypoxia-induced PH and PASMC proliferation. Inducible knockout of FoxM1 in SMC reversed hypoxia-induced pulmonary artery wall remodeling in existing PH. Overexpression of FoxM1 enhanced hypoxia-induced pulmonary artery wall remodeling and right ventricular hypertrophy in mice. Alteration of FoxM1 status did not affect hypoxia-induced hypoxia-inducible factor (HIF) activity in mice. Knockout of FoxM1 decreased PASMC proliferation and induced expression of SMC contractile proteins and TGF-β/Smad3 signaling. CONCLUSIONS Our studies provide clear evidence that altered FoxM1 expression in PASMC contributes to PH and uncover a correlation between Smad3-dependent signaling in FoxM1-mediated proliferation and de-differentiation of PASMC.
Collapse
Affiliation(s)
- Jingbo Dai
- Department of Pediatrics, College of Medicine, University of Illinois at Chicago, Wood Street, Chicago, IL 60612, USA
| | - Qiyuan Zhou
- Department of Pediatrics, College of Medicine, University of Illinois at Chicago, Wood Street, Chicago, IL 60612, USA
| | - Haiyang Tang
- Department of Medicine, University of Arizona, Tucson, AZ, USA; State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Tianji Chen
- Department of Pediatrics, College of Medicine, University of Illinois at Chicago, Wood Street, Chicago, IL 60612, USA
| | - Jing Li
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, USA
| | - Pradip Raychaudhuri
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, USA
| | - Jason X-J Yuan
- Department of Medicine, University of Arizona, Tucson, AZ, USA
| | - Guofei Zhou
- Department of Pediatrics, College of Medicine, University of Illinois at Chicago, Wood Street, Chicago, IL 60612, USA; State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
30
|
Sui D, Zhou H, Wang F, Zhong M, Zhang W, Ti Y. Cell death-inducing DFF45-like effector C gene silencing alleviates pulmonary vascular remodeling in a type 2 diabetic rat model. J Diabetes Investig 2018; 9:741-752. [PMID: 29078040 PMCID: PMC6031506 DOI: 10.1111/jdi.12768] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 10/06/2017] [Accepted: 10/23/2017] [Indexed: 12/18/2022] Open
Abstract
AIMS/INTRODUCTION Cell death-inducing DFF45-like effector C (CIDEC) was proven to be closely associated with the development of insulin resistance and metabolic syndrome. We aimed to investigate whether CIDEC gene silencing could alleviate pulmonary vascular remodeling in a type 2 diabetes rat model. MATERIALS AND METHODS We built a type 2 diabetes rat model. An adenovirus harboring CIDEC small interfering ribonucleic acid was then injected into the jugular vein to silence the CIDEC gene. After hematoxylin-eosin and Sirius red staining, we detected indexes of the pulmonary arterioles remodeling. Immunohistochemical staining of proliferating cell nuclear antigen was used to evaluate the pulmonary arterial smooth muscle cell proliferation. Apoptosis was evaluated by terminal deoxynucleotidyl transferase dUTP nick end labeling reaction and western blotting. The levels of signaling pathway proteins expression were measured by western blotting analyses. RESULTS Histological analysis of the pulmonary artery showed that the thickness of the adventitia and medial layer increased notably in type 2 diabetes rats. Immunohistochemistry showed that more proliferating cell nuclear antigen-positive pulmonary arterial smooth muscle cells could be seen in type 2 diabetes rats; and after CIDEC gene silencing, proliferating cell nuclear antigen positive cells decreased accordingly. Cleaved caspase-3 and cleaved poly (adenosine diphosphate-ribose) polymerase measured by western blotting showed increased apoptosis with overexpressed CIDEC in diabetes. Terminal deoxynucleotidyl transferase dUTP nick end labeling reaction showed that the apoptosis mainly occurred in endothelial cells. Western blotting analysis showed CIDEC overexpression in rats with diabetes, and phosphorylated adenosine 5' monophosphate-activated protein kinase-α expression was significantly decreased. After CIDEC gene silencing, the expression of phosphorylated adenosine 5' monophosphate-activated protein kinase-α was upregulated. CONCLUSIONS The CIDEC/5' monophosphate-activated protein kinase signaling pathway could be a potential therapeutic candidate against pulmonary vascular diseases in type 2 diabetes patients.
Collapse
Affiliation(s)
- Dong‐xin Sui
- The Key Laboratory of Cardiovascular Remodeling and Function ResearchChinese Ministry of Education and Chinese Ministry of Health, and The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular MedicineDepartment of CardiologyQilu Hospital of Shandong UniversityJinanShandongChina
- Department of Respirationthe Second Hospital of Shandong UniversityJinanShandongChina
| | - Hui‐min Zhou
- The Key Laboratory of Cardiovascular Remodeling and Function ResearchChinese Ministry of Education and Chinese Ministry of Health, and The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular MedicineDepartment of CardiologyQilu Hospital of Shandong UniversityJinanShandongChina
| | - Feng Wang
- The Key Laboratory of Cardiovascular Remodeling and Function ResearchChinese Ministry of Education and Chinese Ministry of Health, and The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular MedicineDepartment of CardiologyQilu Hospital of Shandong UniversityJinanShandongChina
| | - Ming Zhong
- The Key Laboratory of Cardiovascular Remodeling and Function ResearchChinese Ministry of Education and Chinese Ministry of Health, and The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular MedicineDepartment of CardiologyQilu Hospital of Shandong UniversityJinanShandongChina
| | - Wei Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function ResearchChinese Ministry of Education and Chinese Ministry of Health, and The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular MedicineDepartment of CardiologyQilu Hospital of Shandong UniversityJinanShandongChina
| | - Yun Ti
- The Key Laboratory of Cardiovascular Remodeling and Function ResearchChinese Ministry of Education and Chinese Ministry of Health, and The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular MedicineDepartment of CardiologyQilu Hospital of Shandong UniversityJinanShandongChina
| |
Collapse
|
31
|
Southern BD, Scheraga RG, Olman MA. Impaired AMPK Activity Drives Age-Associated Acute Lung Injury after Hemorrhage. Am J Respir Cell Mol Biol 2018; 56:553-555. [PMID: 28459384 DOI: 10.1165/rcmb.2017-0023ed] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Affiliation(s)
- Brian D Southern
- 1 Cleveland Clinic Respiratory Institute Lerner Research Institute Department of Pathobiology Cleveland, Ohio
| | - Rachel G Scheraga
- 1 Cleveland Clinic Respiratory Institute Lerner Research Institute Department of Pathobiology Cleveland, Ohio
| | - Mitchell A Olman
- 1 Cleveland Clinic Respiratory Institute Lerner Research Institute Department of Pathobiology Cleveland, Ohio
| |
Collapse
|
32
|
Meng YY, Wu CW, Yu B, Li H, Chen M, Qi GX. PARP-1 Involvement in Autophagy and Their Roles in Apoptosis of Vascular Smooth Muscle Cells under Oxidative Stress. Folia Biol (Praha) 2018; 64:103-111. [PMID: 30394268 DOI: 10.14712/fb2018064030103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Autophagy and poly(ADP-ribose) polymerase 1 (PARP-1) are activated and involved in a series of cell processes under oxidative stress, which is associated with pathogenesis of atherosclerosis. Research on their relationship under oxidative stress has been limited. In this study, we aimed to investigate the activation, relationship, and role of autophagy and PARP-1 in vascular smooth muscle cell (VSMC) death under oxidative stress. This study explored the signal molecule PARP-1 and autophagy in VSMCs using gene silencing and the hydrogen peroxide (H2O2)-stimulated oxidative stress model. We observed that H2O2 could induce autophagy in VSMCs, and the inhibition of autophagy could protect VSMCs against oxidative stress-mediated cell death. Meanwhile, PARP-1 could also be activated by H2O2. Additionally, we analysed the regulatory role of PARP-1 in oxidative stress-mediated autophagy and found that PARP-1 was a novel factor involved in the H2O2-induced autophagy via the AMPK-mTOR pathway. Finally, PARP-1 inhibition protected VSMCs against caspase-dependent apoptosis. These data suggested that PARP-1 played a critical role in H2O2-mediated autophagy and both of them were involved in apoptosis of VSMCs.
Collapse
Affiliation(s)
- Y Y Meng
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - C W Wu
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - B Yu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China
| | - H Li
- Department of Cardiology, No.1 Central Hospital of Baoding, Baoding, Hebei Province, China
| | - M Chen
- Department of Geriatrics, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - G X Qi
- Department of Geriatrics, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| |
Collapse
|
33
|
Xiong PY, Potus F, Chan W, Archer SL. Models and Molecular Mechanisms of World Health Organization Group 2 to 4 Pulmonary Hypertension. Hypertension 2018; 71:34-55. [PMID: 29158355 PMCID: PMC5777609 DOI: 10.1161/hypertensionaha.117.08824] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Ping Yu Xiong
- From the Department of Medicine and Queen's Cardiopulmonary Unit (QCPU) (P.Y.X., F.P., W.C., S.L.A.) and Biomedical and Molecular Sciences (P.Y.X.), Queen's University, Kingston, Ontario, Canada
| | - Francois Potus
- From the Department of Medicine and Queen's Cardiopulmonary Unit (QCPU) (P.Y.X., F.P., W.C., S.L.A.) and Biomedical and Molecular Sciences (P.Y.X.), Queen's University, Kingston, Ontario, Canada
| | - Winnie Chan
- From the Department of Medicine and Queen's Cardiopulmonary Unit (QCPU) (P.Y.X., F.P., W.C., S.L.A.) and Biomedical and Molecular Sciences (P.Y.X.), Queen's University, Kingston, Ontario, Canada
| | - Stephen L Archer
- From the Department of Medicine and Queen's Cardiopulmonary Unit (QCPU) (P.Y.X., F.P., W.C., S.L.A.) and Biomedical and Molecular Sciences (P.Y.X.), Queen's University, Kingston, Ontario, Canada.
| |
Collapse
|
34
|
Autophagy in health and disease: focus on the cardiovascular system. Essays Biochem 2017; 61:721-732. [PMID: 29233881 DOI: 10.1042/ebc20170022] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 10/03/2017] [Accepted: 10/16/2017] [Indexed: 12/20/2022]
Abstract
Autophagy is a highly conserved mechanism of lysosome-mediated protein and organelle degradation that plays a crucial role in maintaining cellular homeostasis. In the last few years, specific functions for autophagy have been identified in many tissues and organs. In the cardiovascular system, autophagy appears to be essential to heart and vessel homeostasis and function; however defective or excessive autophagy activity seems to contribute to major cardiovascular disorders including heart failure (HF) or atherosclerosis. Here, we review the current knowledge on the role of cardiovascular autophagy in physiological and pathophysiological conditions.
Collapse
|
35
|
Hu H, Ding Y, Wang Y, Geng S, Liu J, He J, Lu Y, Li X, Yuan M, Zhu S, Zhao S. MitoK ATP channels promote the proliferation of hypoxic human pulmonary artery smooth muscle cells via the ROS/HIF/miR-210/ISCU signaling pathway. Exp Ther Med 2017; 14:6105-6112. [PMID: 29285165 DOI: 10.3892/etm.2017.5322] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 05/16/2017] [Indexed: 12/18/2022] Open
Abstract
Previous results have indicated that mitochondrial ATP-sensitive potassium (mitoKATP) channels are associated with the hypoxic proliferation of pulmonary artery smooth muscle cells (PASMCs). However, the mechanism underlying the promotive effects of mitoKATP channels on cell proliferation in response to hypoxia remains unknown. mitoKATP channel opening results in a collapse of mitochondrial membrane potential and generation of mitochondrial reactive oxygen species (ROS). As hypoxia-inducible factor-1α (HIF-1α) is a critical oxygen sensor and major transcriptional regulator of the hypoxic adaptive response, the current study assessed whether mitoKATP opening contributes to the chronic proliferation of human PASMCs (hPASMCs) in collaboration with HIF-1α and its downstream targets under hypoxic conditions. The present study demonstrated that there was crosstalk between mitoKATP channels and HIF-1α signaling in PASMCs under hypoxic conditions. The results suggest that mitoKATP channels are involved in the proliferation of PASMCs during hypoxia through upregulation of the ROS/HIF/microRNA-210/iron-sulfur cluster protein signaling pathway.
Collapse
Affiliation(s)
- Hongling Hu
- Department of Respiratory Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Yu Ding
- Key Laboratory for Molecular Diagnosis of Hubei, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China.,Central Laboratory, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Yang Wang
- Department of Respiratory Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Shuang Geng
- Department of Respiratory Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Jue Liu
- Department of Clinical Pharmacy, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Jinrong He
- Key Laboratory for Molecular Diagnosis of Hubei, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China.,Central Laboratory, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Yang Lu
- Department of Respiratory Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Xueying Li
- Department of Respiratory Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Mingli Yuan
- Department of Respiratory Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Shan Zhu
- Department of Respiratory Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Su Zhao
- Department of Respiratory Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| |
Collapse
|
36
|
Zhou Q, Dai J, Chen T, Dada LA, Zhang X, Zhang W, DeCamp MM, Winn RA, Sznajder JI, Zhou G. Downregulation of PKCζ/Pard3/Pard6b is responsible for lung adenocarcinoma cell EMT and invasion. Cell Signal 2017; 38:49-59. [PMID: 28652146 PMCID: PMC5555371 DOI: 10.1016/j.cellsig.2017.06.016] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 06/08/2017] [Accepted: 06/20/2017] [Indexed: 12/24/2022]
Abstract
Atypical protein kinase C ζ (PKCζ) forms an apico-basal polarity complex with Partitioning Defective (Pard) 3 and Pard6 to regulate normal epithelial cell apico-basolateral polarization. The dissociation of the PKCζ/Pard3/Pard6 complex is essential for the disassembly of the tight/adherens junction and epithelial-mesenchymal transition (EMT) that is critical for tumor spreading. Loss of cell polarity and epithelial organization is strongly correlated with malignancy and tumor progression in some other cancer types. However, it is unclear whether the PKCζ/Pard3/Pard6 complex plays a role in the progression of non-small-cell lung cancer (NSCLC). We found that hypoxia downregulated the PKCζ/Pard3/Pard6 complex, correlating with induction of lung cancer cell migration and invasion. Silencing of the PKCζ/Pard3/Pard6 polarity complex components induced lung cancer cell EMT, invasion, and colonization in vivo. Suppression of Pard3 was associated with altered expression of genes regulating wound healing, cell apoptosis/death and cell motility, and particularly upregulation of MAP3K1 and fibronectin which are known to contribute to lung cancer progression. Human lung adenocarcinoma tissues expressed less Pard6b and PKCζ than the adjacent normal tissues and in experimental mouse lung adenocarcinoma, the levels of Pard3 and PKCζ were also decreased. In addition, we showed that a methylation locus in the gene body of Pard3 is positively associated with the expression of Pard3 and that methylation of the Pard3 gene increased cellular sensitivity to carboplatin, a common chemotherapy drug. Suppression of Pard3 increased chemoresistance in lung cancer cells. Together, these results suggest that reduced expression of PKCζ/Pard3/Pard6 contributes to NSCLC EMT, invasion, and chemoresistance.
Collapse
Affiliation(s)
- Qiyuan Zhou
- Department of Pediatrics, University of Illinois at Chicago, Chicago, IL, USA
| | - Jingbo Dai
- Department of Pediatrics, University of Illinois at Chicago, Chicago, IL, USA
| | - Tianji Chen
- Department of Pediatrics, University of Illinois at Chicago, Chicago, IL, USA
| | - Laura A Dada
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Xu Zhang
- Division of Hematology and Oncology, University of Illinois at Chicago, Chicago, IL, USA
| | - Wei Zhang
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Malcolm M DeCamp
- The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Division of Thoracic Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Robert A Winn
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA; Cancer Center, University of Illinois at Chicago, Chicago, IL, USA
| | - Jacob I Sznajder
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Guofei Zhou
- Department of Pediatrics, University of Illinois at Chicago, Chicago, IL, USA; Cancer Center, University of Illinois at Chicago, Chicago, IL, USA; State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
37
|
Kyriakakis E, Frismantiene A, Dasen B, Pfaff D, Rivero O, Lesch KP, Erne P, Resink TJ, Philippova M. T-cadherin promotes autophagy and survival in vascular smooth muscle cells through MEK1/2/Erk1/2 axis activation. Cell Signal 2017; 35:163-175. [DOI: 10.1016/j.cellsig.2017.04.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 03/20/2017] [Accepted: 04/05/2017] [Indexed: 10/19/2022]
|
38
|
Abstract
The AMP-activated protein kinase (AMPK) is a key regulator of cellular and whole-body energy homeostasis, which acts to restore energy homoeostasis whenever cellular energy charge is depleted. Over the last 2 decades, it has become apparent that AMPK regulates several other cellular functions and has specific roles in cardiovascular tissues, acting to regulate cardiac metabolism and contractile function, as well as promoting anticontractile, anti-inflammatory, and antiatherogenic actions in blood vessels. In this review, we discuss the role of AMPK in the cardiovascular system, including the molecular basis of mutations in AMPK that alter cardiac physiology and the proposed mechanisms by which AMPK regulates vascular function under physiological and pathophysiological conditions.
Collapse
Affiliation(s)
- Ian P Salt
- From the Institute of Cardiovascular & Medical Sciences, College of Medical, Veterinary & Life Sciences, University of Glasgow, Scotland, United Kingdom (I.P.S.); and Division of Cell Signalling & Immunology, School of Life Sciences, University of Dundee, Scotland, United Kingdom (D.G.H.).
| | - D Grahame Hardie
- From the Institute of Cardiovascular & Medical Sciences, College of Medical, Veterinary & Life Sciences, University of Glasgow, Scotland, United Kingdom (I.P.S.); and Division of Cell Signalling & Immunology, School of Life Sciences, University of Dundee, Scotland, United Kingdom (D.G.H.)
| |
Collapse
|
39
|
Xue J, Nelin LD, Chen B. Hypoxia induces arginase II expression and increases viable human pulmonary artery smooth muscle cell numbers via AMPKα 1 signaling. Am J Physiol Lung Cell Mol Physiol 2017; 312:L568-L578. [PMID: 28213467 DOI: 10.1152/ajplung.00117.2016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 02/08/2017] [Accepted: 02/09/2017] [Indexed: 01/08/2023] Open
Abstract
Pulmonary artery smooth muscle cell (PASMC) proliferation is one of the hallmark features of hypoxia-induced pulmonary hypertension. With only supportive treatment options available for this life-threatening disease, treating and preventing the proliferation of PASMCs is a viable therapeutic option. A key promoter of hypoxia-induced increases in the number of viable human PASMCs is arginase II, with attenuation of viable cell numbers following pharmacologic inhibition or siRNA knockdown of the enzyme. Additionally, increased levels of arginase have been demonstrated in the pulmonary vasculature of patients with pulmonary hypertension. The signaling pathways responsible for the hypoxic induction of arginase II in PASMCs, however, remain unknown. Hypoxia is a recognized activator of AMPK, which is known to be expressed in human PASMCs (hPASMCs). Activation of AMPK by hypoxia has been shown to promote cell survival in PASMCs. In addition, pharmacologic agents targeting AMPK have been shown to attenuate chronic hypoxia-induced pulmonary hypertension in animal models. The present studies tested the hypothesis that hypoxia-induced arginase II expression in hPASMCs is mediated through AMPK signaling. We found that pharmacologic inhibitors of AMPK, as well as siRNA knockdown of AMPKα1, prevented hypoxia-induced arginase II. The hypoxia-induced increase in viable hPASMC numbers was also prevented following both pharmacologic inhibition and siRNA knockdown of AMPK. Furthermore, we demonstrate that overexpression of AMPK induced arginase II protein expression and viable cells numbers in hPASMCs.
Collapse
Affiliation(s)
- Jianjing Xue
- Pulmonary Hypertension Group, Center for Perinatal Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio; and
| | - Leif D Nelin
- Pulmonary Hypertension Group, Center for Perinatal Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio; and.,Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Bernadette Chen
- Pulmonary Hypertension Group, Center for Perinatal Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio; and .,Department of Pediatrics, The Ohio State University, Columbus, Ohio
| |
Collapse
|
40
|
Liu H, Li XZ, Peng M, Ji W, Zhao L, Li L, Zhang L, Si JQ, Ma KT. Role of gap junctions in the contractile response to agonists in the mesenteric resistance artery of rats with acute hypoxia. Mol Med Rep 2017; 15:1823-1831. [DOI: 10.3892/mmr.2017.6188] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 12/21/2016] [Indexed: 11/06/2022] Open
|
41
|
He Y, Cao X, Guo P, Li X, Shang H, Liu J, Xie M, Xu Y, Liu X. Quercetin induces autophagy via FOXO1-dependent pathways and autophagy suppression enhances quercetin-induced apoptosis in PASMCs in hypoxia. Free Radic Biol Med 2017; 103:165-176. [PMID: 27979659 DOI: 10.1016/j.freeradbiomed.2016.12.016] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Revised: 12/10/2016] [Accepted: 12/12/2016] [Indexed: 02/06/2023]
Abstract
Quercetin, an important dietary flavonoid has been demonstrated to potentially reverse or even prevent pulmonary arterial hypertension (PAH) progression. However, the effects of quercetin on apoptosis and autophagy in pulmonary arterial smooth muscle cells (PASMCs) have not yet been clearly elucidated. The current study found that quercetin significantly induce the apoptotic and autophagic capacities of PASMCs in vitro and in vivo in hypoxia. In addition, we found that quercetin increases FOXO1 (a major mediator in autophagy regulation) expression and transcriptional activity. Moreover, FOXO1 knockdown by siRNAs inhibited the phosphorylation of mTOR and 4E-BPI, which is downstream of P70-S6K, and markedly blocked quercetin-induced autophagy. We also observed that FOXO1-mediated autophagy was achieved via SESN3 not Rictor upregulation and after mTOR suppression. Furthermore, Treatment with autophagy-specific inhibitors could markedly enhance quercetin-induced apoptosis in PASMCs under hypoxia. Finally, quercetin in combination with autophagy inhibition treatment could enhance the therapeutic effects of quercetin in hypoxia-associated PAH in vivo. Taken together, quercetin could enhance hypoxia-induced autophagy through the FOXO1-SENS3-mTOR pathway in PASMCs. Combining quercetin and autophagy inhibitors may be a novel therapeutic strategy for treating hypoxia-associated PAH.
Collapse
Affiliation(s)
- Yuanzhou He
- Department of Respiratory Diseases, Tongji Hospital, Key Lab of Pulmonary Diseases of Health Ministry, Key Site of National Clinical Research Center for Respiratory Disease, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Xiaopei Cao
- Department of Respiratory Diseases, Tongji Hospital, Key Lab of Pulmonary Diseases of Health Ministry, Key Site of National Clinical Research Center for Respiratory Disease, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Pujian Guo
- Department of Respiratory Diseases, Tongji Hospital, Key Lab of Pulmonary Diseases of Health Ministry, Key Site of National Clinical Research Center for Respiratory Disease, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Xiaochen Li
- Department of Respiratory Diseases, Tongji Hospital, Key Lab of Pulmonary Diseases of Health Ministry, Key Site of National Clinical Research Center for Respiratory Disease, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Huihui Shang
- Department of Respiratory Diseases, Tongji Hospital, Key Lab of Pulmonary Diseases of Health Ministry, Key Site of National Clinical Research Center for Respiratory Disease, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Jin Liu
- Department of Respiratory Diseases, Tongji Hospital, Key Lab of Pulmonary Diseases of Health Ministry, Key Site of National Clinical Research Center for Respiratory Disease, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Min Xie
- Department of Respiratory Diseases, Tongji Hospital, Key Lab of Pulmonary Diseases of Health Ministry, Key Site of National Clinical Research Center for Respiratory Disease, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Yongjian Xu
- Department of Respiratory Diseases, Tongji Hospital, Key Lab of Pulmonary Diseases of Health Ministry, Key Site of National Clinical Research Center for Respiratory Disease, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Xiansheng Liu
- Department of Respiratory Diseases, Tongji Hospital, Key Lab of Pulmonary Diseases of Health Ministry, Key Site of National Clinical Research Center for Respiratory Disease, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China.
| |
Collapse
|
42
|
Hao MX, Wang X, Jiao KL. MicroRNA-17-5p mediates hypoxia-induced autophagy and inhibits apoptosis by targeting signal transducer and activator of transcription 3 in vascular smooth muscle cells. Exp Ther Med 2017; 13:935-941. [PMID: 28450922 PMCID: PMC5403340 DOI: 10.3892/etm.2017.4048] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Accepted: 10/18/2016] [Indexed: 12/03/2022] Open
Abstract
The aim of the present study was to investigate hypoxia-induced apoptosis and autophagy in vascular smooth muscle cells (VSMCs) and the underlying molecular mechanisms of microRNA (miR)-17-5p responses in an anaerobic environment. The results revealed that miR-17-5p expression was significantly upregulated in VSMCs subjected to hypoxic conditions (P<0.05) and lower miR-17-5p levels were observed in ethyl 3,4-dihydroxybenzoate-treated and hypoxia inducible factor-1 loss-of-function cells. Additionally, it was demonstrated that miR-17-5p is associated with hypoxia-induced autophagy, which was confirmed by upregulating the light chain 3-II/LC3-I ratio and downregulating nucleoporin p62. Cell apoptosis was inhibited in response to hypoxia, and levels of pro-apoptotic proteins B-cell lymphoma 2-associated X protein and p-caspase were markedly decreased when VSMCs were subjected to hypoxic conditions. Furthermore, expression of signal transducer and activator of transcription 3 (STAT3) decreased when cells were transfected with overexpressing miR-17-5p and subjected to hypoxic conditions, and the combination of miR-17-5p loss-of-function and hypoxia induced greater upregulation in the protein expression of STAT3 compared with a single treatment for hypoxia in VSMCs. In conclusion, miR-17-5p may be a novel hypoxia-responsive miR and hypoxia may induce protective autophagy and anti-apoptosis in VSMCs by targeting STAT3.
Collapse
Affiliation(s)
- Ming-Xiu Hao
- Department of Geriatrics, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Xing Wang
- Department of Endocrinology, Shanghai Pudong New Area Gongli Hospital, Second Military Medical University, Shanghai 200135, P.R. China
| | - Kun-Li Jiao
- Department of Cardiology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, P.R. China
| |
Collapse
|
43
|
Ramadan A, Al-Omran M, Verma S. The putative role of autophagy in the pathogenesis of abdominal aortic aneurysms. Atherosclerosis 2017; 257:288-296. [PMID: 28139205 DOI: 10.1016/j.atherosclerosis.2017.01.017] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 12/08/2016] [Accepted: 01/13/2017] [Indexed: 10/20/2022]
Abstract
Abdominal aortic aneurysms (AAA) are a significant cause of worldwide mortality and morbidity. While the histopathological characteristics of AAA are well documented, the cellular and molecular mechanisms involved in the pathogenesis of AAA are not entirely understood. Autophagy is a highly conserved basal cellular process in eukaryotic cells that involves the turnover of organelles and proteins. It is also activated as an adaptive response to stressful conditions to promote cell survival. While autophagy typically promotes pro-survival processes, it can sometimes lead to cellular demise. Preclinical studies have revealed autophagy to be a protective mechanism in certain vascular diseases with several autophagy-related genes reported to be markedly upregulated in human aneurysmal tissue. The role autophagy plays in the pathogenesis of AAA, however, remains poorly defined. In this review, we discuss the putative role of autophagy in AAA by reviewing several in vitro and in vivo studies that address the functional significance of autophagy in cells that are involved in the pathophysiology of AAA, amongst which are macrophages, smooth muscle and endothelial cells.
Collapse
Affiliation(s)
- Azza Ramadan
- Division of Cardiac Surgery, Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, ON, Canada
| | - Mohammed Al-Omran
- Division of Vascular Surgery, Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, ON, Canada; Department of Surgery, University of Toronto, ON, Canada; Department of Surgery, King Saud University, Riyadh, Saudi Arabia
| | - Subodh Verma
- Division of Cardiac Surgery, Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, ON, Canada; Department of Surgery, University of Toronto, ON, Canada.
| |
Collapse
|
44
|
Evans AM, Mahmoud AD, Moral-Sanz J, Hartmann S. The emerging role of AMPK in the regulation of breathing and oxygen supply. Biochem J 2016; 473:2561-72. [PMID: 27574022 PMCID: PMC5003690 DOI: 10.1042/bcj20160002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 04/20/2016] [Accepted: 05/03/2016] [Indexed: 01/25/2023]
Abstract
Regulation of breathing is critical to our capacity to accommodate deficits in oxygen availability and demand during, for example, sleep and ascent to altitude. It is generally accepted that a fall in arterial oxygen increases afferent discharge from the carotid bodies to the brainstem and thus delivers increased ventilatory drive, which restores oxygen supply and protects against hypoventilation and apnoea. However, the precise molecular mechanisms involved remain unclear. We recently identified as critical to this process the AMP-activated protein kinase (AMPK), which is key to the cell-autonomous regulation of metabolic homoeostasis. This observation is significant for many reasons, not least because recent studies suggest that the gene for the AMPK-α1 catalytic subunit has been subjected to natural selection in high-altitude populations. It would appear, therefore, that evolutionary pressures have led to AMPK being utilized to regulate oxygen delivery and thus energy supply to the body in the short, medium and longer term. Contrary to current consensus, however, our findings suggest that AMPK regulates ventilation at the level of the caudal brainstem, even when afferent input responses from the carotid body are normal. We therefore hypothesize that AMPK integrates local hypoxic stress at defined loci within the brainstem respiratory network with an index of peripheral hypoxic status, namely afferent chemosensory inputs. Allied to this, AMPK is critical to the control of hypoxic pulmonary vasoconstriction and thus ventilation-perfusion matching at the lungs and may also determine oxygen supply to the foetus by, for example, modulating utero-placental blood flow.
Collapse
Affiliation(s)
- A Mark Evans
- Centre for Integrative Physiology, College of Medicine and Veterinary Medicine, Hugh Robson Building, University of Edinburgh, Edinburgh EH8 9XD, U.K.
| | - Amira D Mahmoud
- Centre for Integrative Physiology, College of Medicine and Veterinary Medicine, Hugh Robson Building, University of Edinburgh, Edinburgh EH8 9XD, U.K
| | - Javier Moral-Sanz
- Centre for Integrative Physiology, College of Medicine and Veterinary Medicine, Hugh Robson Building, University of Edinburgh, Edinburgh EH8 9XD, U.K
| | - Sandy Hartmann
- Centre for Integrative Physiology, College of Medicine and Veterinary Medicine, Hugh Robson Building, University of Edinburgh, Edinburgh EH8 9XD, U.K
| |
Collapse
|
45
|
Moral-Sanz J, Mahmoud AD, Ross FA, Eldstrom J, Fedida D, Hardie DG, Evans AM. AMP-activated protein kinase inhibits Kv 1.5 channel currents of pulmonary arterial myocytes in response to hypoxia and inhibition of mitochondrial oxidative phosphorylation. J Physiol 2016; 594:4901-15. [PMID: 27062501 PMCID: PMC5009768 DOI: 10.1113/jp272032] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 03/26/2016] [Indexed: 12/29/2022] Open
Abstract
KEY POINTS Progression of hypoxic pulmonary hypertension is thought to be due, in part, to suppression of voltage-gated potassium channels (Kv ) in pulmonary arterial smooth muscle by hypoxia, although the precise molecular mechanisms have been unclear. AMP-activated protein kinase (AMPK) has been proposed to couple inhibition of mitochondrial metabolism by hypoxia to acute hypoxic pulmonary vasoconstriction and progression of pulmonary hypertension. Inhibition of complex I of the mitochondrial electron transport chain activated AMPK and inhibited Kv 1.5 channels in pulmonary arterial myocytes. AMPK activation by 5-aminoimidazole-4-carboxamide riboside, A769662 or C13 attenuated Kv 1.5 currents in pulmonary arterial myocytes, and this effect was non-additive with respect to Kv 1.5 inhibition by hypoxia and mitochondrial poisons. Recombinant AMPK phosphorylated recombinant human Kv 1.5 channels in cell-free assays, and inhibited K(+) currents when introduced into HEK 293 cells stably expressing Kv 1.5. These results suggest that AMPK is the primary mediator of reductions in Kv 1.5 channels following inhibition of mitochondrial oxidative phosphorylation during hypoxia and by mitochondrial poisons. ABSTRACT Progression of hypoxic pulmonary hypertension is thought to be due, in part, to suppression of voltage-gated potassium channels (Kv ) in pulmonary arterial smooth muscle cells that is mediated by the inhibition of mitochondrial oxidative phosphorylation. We sought to determine the role in this process of the AMP-activated protein kinase (AMPK), which is intimately coupled to mitochondrial function due to its activation by LKB1-dependent phosphorylation in response to increases in the cellular AMP:ATP and/or ADP:ATP ratios. Inhibition of complex I of the mitochondrial electron transport chain using phenformin activated AMPK and inhibited Kv currents in pulmonary arterial myocytes, consistent with previously reported effects of mitochondrial inhibitors. Myocyte Kv currents were also markedly inhibited upon AMPK activation by A769662, 5-aminoimidazole-4-carboxamide riboside and C13 and by intracellular dialysis from a patch-pipette of activated (thiophosphorylated) recombinant AMPK heterotrimers (α2β2γ1 or α1β1γ1). Hypoxia and inhibitors of mitochondrial oxidative phosphorylation reduced AMPK-sensitive K(+) currents, which were also blocked by the selective Kv 1.5 channel inhibitor diphenyl phosphine oxide-1 but unaffected by the presence of the BKCa channel blocker paxilline. Moreover, recombinant human Kv 1.5 channels were phosphorylated by AMPK in cell-free assays, and K(+) currents carried by Kv 1.5 stably expressed in HEK 293 cells were inhibited by intracellular dialysis of AMPK heterotrimers and by A769662, the effects of which were blocked by compound C. We conclude that AMPK mediates Kv channel inhibition by hypoxia in pulmonary arterial myocytes, at least in part, through phosphorylation of Kv 1.5 and/or an associated protein.
Collapse
Affiliation(s)
- Javier Moral-Sanz
- Centre for Integrative Physiology, College of Medicine and Veterinary Medicine, Hugh Robson Building, George Square, University of Edinburgh, Edinburgh, EH8 9XD, UK
| | - Amira D Mahmoud
- Centre for Integrative Physiology, College of Medicine and Veterinary Medicine, Hugh Robson Building, George Square, University of Edinburgh, Edinburgh, EH8 9XD, UK
| | - Fiona A Ross
- Division of Cell Signalling & Immunology, School of Life Sciences, University of Dundee, Dow Street, Dundee, DD1 5EH, UK
| | - Jodene Eldstrom
- Department of Anaesthesiology. Pharmacology and Therapeutics, University of British Columbia, 2350 Health Science Mall, Vancouver, Canada, V6T 1Z3
| | - David Fedida
- Department of Anaesthesiology. Pharmacology and Therapeutics, University of British Columbia, 2350 Health Science Mall, Vancouver, Canada, V6T 1Z3
| | - D Grahame Hardie
- Division of Cell Signalling & Immunology, School of Life Sciences, University of Dundee, Dow Street, Dundee, DD1 5EH, UK
| | - A Mark Evans
- Centre for Integrative Physiology, College of Medicine and Veterinary Medicine, Hugh Robson Building, George Square, University of Edinburgh, Edinburgh, EH8 9XD, UK
| |
Collapse
|
46
|
Dean A, Nilsen M, Loughlin L, Salt IP, MacLean MR. Metformin Reverses Development of Pulmonary Hypertension via Aromatase Inhibition. Hypertension 2016; 68:446-54. [DOI: 10.1161/hypertensionaha.116.07353] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 05/13/2016] [Indexed: 11/16/2022]
Abstract
Females are more susceptible to pulmonary arterial hypertension than males, although the reasons remain unclear. The hypoglycemic drug, metformin, is reported to have multiple actions, including the inhibition of aromatase and stimulation of AMP-activated protein kinase. Inhibition of aromatase using anastrazole is protective in experimental pulmonary hypertension but whether metformin attenuates pulmonary hypertension through this mechanism remains unknown. We investigated whether metformin affected aromatase activity and if it could reduce the development of pulmonary hypertension in the sugen 5416/hypoxic rat model. We also investigated its influence on proliferation in human pulmonary arterial smooth muscle cells. Metformin reversed right ventricular systolic pressure, right ventricular hypertrophy, and decreased pulmonary vascular remodeling in the rat. Furthermore, metformin increased rat lung AMP-activated protein kinase signaling, decreased lung and circulating estrogen levels, levels of aromatase, the estrogen metabolizing enzyme; cytochrome P450 1B1 and its transcription factor; the aryl hydrocarbon receptor. In human pulmonary arterial smooth muscle cells, metformin decreased proliferation and decreased estrogen synthesis by decreasing aromatase activity through the PII promoter site of
Cyp19a1
. Thus, we report for the first time that metformin can reverse pulmonary hypertension through inhibition of aromatase and estrogen synthesis in a manner likely to be mediated by AMP-activated protein kinase.
Collapse
Affiliation(s)
- Afshan Dean
- From the Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Margaret Nilsen
- From the Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Lynn Loughlin
- From the Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Ian P. Salt
- From the Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Margaret R. MacLean
- From the Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
47
|
Protective Roles of Endothelial AMP-Activated Protein Kinase Against Hypoxia-Induced Pulmonary Hypertension in Mice. Circ Res 2016; 119:197-209. [DOI: 10.1161/circresaha.115.308178] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 05/23/2016] [Indexed: 12/31/2022]
Abstract
Rationale:
Endothelial AMP-activated protein kinase (AMPK) plays an important role for vascular homeostasis, and its role is impaired by vascular inflammation. However, the role of endothelial AMPK in the pathogenesis of pulmonary arterial hypertension (PAH) remains to be elucidated.
Objective:
To determine the role of endothelial AMPK in the development of PAH.
Methods and Results:
Immunostaining showed that endothelial AMPK is downregulated in the pulmonary arteries of patients with PAH and hypoxia mouse model of pulmonary hypertension (PH). To elucidate the role of endothelial AMPK in PH, we used endothelial-specific AMPK-knockout mice (
eAMPK
–/–
), which were exposed to hypoxia. Under normoxic condition,
eAMPK
–/–
mice showed the normal morphology of pulmonary arteries compared with littermate controls (
eAMPK
flox/flox
). In contrast, development of hypoxia-induced PH was accelerated in
eAMPK
–/–
mice compared with controls. Furthermore, the exacerbation of PH in
eAMPK
–/–
mice was accompanied by reduced endothelial function, upregulation of growth factors, and increased proliferation of pulmonary artery smooth muscle cells. Importantly, conditioned medium from endothelial cells promoted pulmonary artery smooth muscle cell proliferation, which was further enhanced by the treatment with AMPK inhibitor. Serum levels of inflammatory cytokines, including tumor necrosis factor-α and interferon-γ were significantly increased in patients with PAH compared with healthy controls. Consistently, endothelial AMPK and cell proliferation were significantly reduced by the treatment with serum from patients with PAH compared with controls. Importantly, long-term treatment with metformin, an AMPK activator, significantly attenuated hypoxia-induced PH in mice.
Conclusions:
These results indicate that endothelial AMPK is a novel therapeutic target for the treatment of PAH.
Collapse
|
48
|
Singh N, Manhas A, Kaur G, Jagavelu K, Hanif K. Inhibition of fatty acid synthase is protective in pulmonary hypertension. Br J Pharmacol 2016; 173:2030-45. [PMID: 27061087 PMCID: PMC4882492 DOI: 10.1111/bph.13495] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Revised: 03/21/2016] [Accepted: 03/26/2016] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND AND PURPOSE In pulmonary hypertension (PH), similar to cancer, there is altered energy metabolism, apoptosis resistance and cellular proliferation leading to pulmonary vascular remodelling. Proliferating cells exhibit higher rate of de novo fatty acid synthesis to provide lipids for membrane formation and energy production. As inhibition of de novo fatty acid synthesis proved protective in cancer experimentally, therefore, it was hypothesized that modulation of de novo fatty acid synthesis by inhibition of fatty acid synthase (FAS) may prove beneficial for PH. EXPERIMENTAL APPROACH For in vitro studies, human pulmonary artery smooth muscle cells (HPASMCs) were exposed to hypoxia and to induce PH in vivo, rats were treated with monocrotaline (MCT). FAS was inhibited by siRNA (60 nM) and C75 (2 mg·kg(-1) , i.p. once a week for 5 weeks) in in vitro and in vivo studies respectively. RESULTS Increased expression and activity of FAS were observed in hypoxic HPASMCs and lungs of MCT-treated rats. Inhibition of FAS increased apoptosis and glucose oxidation, but decreased proliferation and markers of autophagy, glycolysis and insulin resistance in hypoxic HPASMCs. It also improved the mitochondrial functions as evident by increased level of ATP and restoration of normal level of ROS and membrane potential of mitochondria. In MCT-treated rats, FAS inhibition decreased right ventricular pressure, hypertrophy, pulmonary vascular remodelling (increased apoptosis and decreased proliferation of cells) and endothelial dysfunction in lungs. CONCLUSIONS Our results demonstrate that FAS activity is modulated in PH, and its inhibition may provide a new therapeutic approach to treat PH.
Collapse
Affiliation(s)
- Neetu Singh
- Division of PharmacologyCSIR‐Central Drug Research InstituteLucknowIndia
- Academy of Scientific and Innovative ResearchNew DelhiIndia
| | - Amit Manhas
- Division of PharmacologyCSIR‐Central Drug Research InstituteLucknowIndia
| | - Gurpreet Kaur
- Division of PharmacologyCSIR‐Central Drug Research InstituteLucknowIndia
| | - Kumaravelu Jagavelu
- Division of PharmacologyCSIR‐Central Drug Research InstituteLucknowIndia
- Academy of Scientific and Innovative ResearchNew DelhiIndia
| | - Kashif Hanif
- Division of PharmacologyCSIR‐Central Drug Research InstituteLucknowIndia
- Academy of Scientific and Innovative ResearchNew DelhiIndia
| |
Collapse
|
49
|
Wang X, Ibrahim YF, Das D, Zungu-Edmondson M, Shults NV, Suzuki YJ. Carfilzomib reverses pulmonary arterial hypertension. Cardiovasc Res 2016; 110:188-99. [PMID: 26952044 DOI: 10.1093/cvr/cvw047] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Accepted: 02/19/2016] [Indexed: 01/23/2023] Open
Abstract
AIMS Pulmonary arterial hypertension (PAH) remains a lethal disease with pronounced narrowing of pulmonary vessels due to abnormal cell growth. Agents that can reduce the pulmonary vascular thickness thus have therapeutic potential. The present study investigated the efficacy of carfilzomib (CFZ), a proteasome inhibitor and a cancer chemotherapeutic drug, on reversing PAH. METHODS AND RESULTS In two rat models of PAH, SU5416/hypoxia and SU5416/ovalbumin, CFZ effectively reversed pulmonary vascular remodelling with the promotion of apoptosis and autophagy. In human pulmonary artery smooth muscle cells, knocking down mediators of autophagy attenuated CFZ-induced cell death. The cell death role of autophagy was promoted by the participation of tumour protein p53-inducible nuclear protein 1. CFZ increased the protein ubiquitination, and siRNA knockdown of ubiquitin inhibited cell death, suggesting that CFZ-induced cell death is ubiquitin-dependent. Mass spectrometry demonstrated the ubiquitination of major vault protein and heat shock protein 90 in response to CFZ. The siRNA knockdown of these proteins enhanced CFZ-induced cell death, revealing that they are cell survival factors. CFZ reduced right-ventricular pressure and enhanced the efficacy of a vasodilator, sodium nitroprusside. While no indications of CFZ toxicity were observed in the right ventricle of PAH rats, apoptosis was promoted in the left ventricle. Apoptosis was prevented by dexrazoxane or by pifithrin-α without interfering with the efficacy of CFZ to reverse pulmonary vascular remodelling. CONCLUSION The addition of anti-tumour agents such as CFZ along with cardioprotectants to currently available vasodilators may be a promising way to improve PAH therapy.
Collapse
Affiliation(s)
- Xinhong Wang
- Department of Pharmacology and Physiology, Georgetown University Medical Center, 3900 Reservoir Road NW, Washington, DC 20057, USA Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Yasmine F Ibrahim
- Department of Pharmacology and Physiology, Georgetown University Medical Center, 3900 Reservoir Road NW, Washington, DC 20057, USA Department of Pharmacology, Minia University School of Medicine, Minia 61111, Egypt
| | - Dividutta Das
- Department of Pharmacology and Physiology, Georgetown University Medical Center, 3900 Reservoir Road NW, Washington, DC 20057, USA
| | - Makhosazane Zungu-Edmondson
- Department of Pharmacology and Physiology, Georgetown University Medical Center, 3900 Reservoir Road NW, Washington, DC 20057, USA
| | - Nataliia V Shults
- Department of Pharmacology and Physiology, Georgetown University Medical Center, 3900 Reservoir Road NW, Washington, DC 20057, USA
| | - Yuichiro J Suzuki
- Department of Pharmacology and Physiology, Georgetown University Medical Center, 3900 Reservoir Road NW, Washington, DC 20057, USA
| |
Collapse
|
50
|
Tai S, Hu XQ, Peng DQ, Zhou SH, Zheng XL. The roles of autophagy in vascular smooth muscle cells. Int J Cardiol 2016; 211:1-6. [PMID: 26954728 DOI: 10.1016/j.ijcard.2016.02.128] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 02/05/2016] [Accepted: 02/22/2016] [Indexed: 12/21/2022]
Abstract
Autophagy, which is an evolutionarily conserved mechanism and links to several cellular pathways, impacts vascular smooth muscle cells (VSMCs) survival and function. Activation of autophagy by intercellular and/or extracellular stimuli has protective effects on VSMCs against cell death, while on the contrary, overloading autophagy has been recognized as a deleterious process by excessive self-digestion. Alterations in autophagy has been documented in VSMC in response to various stimuli, resulting in modulation of VSMC functions, including proliferation, migration, matrix secretion, contraction/relaxation, and differentiation. Each of these changes in VSMC functions plays a critical role in the development of vascular diseases. Importantly, emerging evidence demonstrates that autophagy deficiency in VSMCs would contribute to atherosclerosis and restenosis, shedding novel light on therapeutic target of the vascular disorders. Herein, this review summarizes the recent progress associated with the roles of autophagy in VSMC and offers the perspectives to several challenges and future directions for further studies.
Collapse
Affiliation(s)
- Shi Tai
- Dept. of Biochemistry & Molecular Biology, Faculty of Medicine, Univ. of Calgary, Calgary, Alberta, Canada; Dept. of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xin-Qun Hu
- Dept. of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Dao-Quan Peng
- Dept. of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Sheng-Hua Zhou
- Dept. of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, China.
| | - Xi-Long Zheng
- Dept. of Biochemistry & Molecular Biology, Faculty of Medicine, Univ. of Calgary, Calgary, Alberta, Canada; Dept. of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, China.
| |
Collapse
|