1
|
Shaydakov ME, Diaz JA, Eklöf B, Lurie F. Venous valve hypoxia as a possible mechanism of deep vein thrombosis: a scoping review. INT ANGIOL 2024; 43:309-322. [PMID: 38864688 DOI: 10.23736/s0392-9590.24.05170-8] [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: 06/13/2024]
Abstract
INTRODUCTION The pathogenesis of deep vein thrombosis (DVT) has been explained by an interplay between a changed blood composition, vein wall alteration, and blood flow abnormalities. A comprehensive investigation of these components of DVT pathogenesis has substantially promoted our understanding of thrombogenesis in the venous system. Meanwhile, the process of DVT initiation remains obscure. This systematic review aims to collect, analyze, and synthesize the published evidence to propose hypoxia as a possible trigger of DVT. EVIDENCE ACQUISITION An exhaustive literature search was conducted across multiple electronic databased including PubMed, EMBASE, Scopus, and Web of Science to identify studies pertinent to the research hypothesis. The search was aimed at exploring the connection between hypoxia, reoxygenation, and the initiation of deep vein thrombosis (DVT). The following key words were used: "deep vein thrombosis," "venous thrombosis," "venous thromboembolism," "hypoxia," "reoxygenation," "venous valve," and "venous endothelium." Reviews, case reports, editorials, and letters were excluded. EVIDENCE SYNTHESIS Based on the systematic search outcome, 156 original papers relevant to the issue were selected for detailed review. These studies encompassed a range of experimental and observational clinical research, focusing on various aspects of DVT, including the anatomical, physiological, and cellular bases of the disease. A number of studies suggested limitations in the traditional understanding of Virchow's triad as an acceptable explanation for DVT initiation. Emerging evidence points to more complex interactions and additional factors that may be critical in the early stages of thrombogenesis. The role of venous valves has been recognized but remains underappreciated, with several studies indicating that these sites may act as primary loci for thrombus formation. A collection of studies describes the effects of hypoxia on venous endothelial cells at the cellular and molecular levels. Hypoxia influences several pathways that regulate endothelial cell permeability, inflammatory response, and procoagulation activity, underpinning the endothelial dysfunction noted in DVT. CONCLUSIONS Hypoxia of the venous valve may serve as an independent hypothesis to outline the DVT triggering process. Further research projects in this field may discover new molecular pathways responsible for the disease and suggest new therapeutic targets.
Collapse
Affiliation(s)
- Maxim E Shaydakov
- Division of Vascular Surgery, University of Pittsburgh Medical Center, Pittsburg, PA, USA -
| | - Jose A Diaz
- Division of Surgical Research, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Fedor Lurie
- Jobst Vascular Institute, ProMedica Health System, Toledo, OH, USA
| |
Collapse
|
2
|
Xiao W, Shrimali N, Oldham WM, Clish CB, He H, Wong SJ, Wertheim BM, Arons E, Haigis MC, Leopold JA, Loscalzo J. Branched chain α-ketoacids aerobically activate HIF1α signaling in vascular cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.29.595538. [PMID: 38853866 PMCID: PMC11160772 DOI: 10.1101/2024.05.29.595538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Hypoxia-inducible factor 1α (HIF1α) is a master regulator of numerous biological processes under low oxygen tensions. Yet, the mechanisms and biological consequences of aerobic HIF1α activation by intrinsic factors, particularly in primary cells remain elusive. Here, we show that HIF1α signaling is activated in several human primary vascular cells under ambient oxygen tensions, and in vascular smooth muscle cells (VSMCs) of normal human lung tissue, which contributed to a relative resistance to further enhancement of glycolytic activity in hypoxia. Mechanistically, aerobic HIFα activation is mediated by paracrine secretion of three branched chain α-ketoacids (BCKAs), which suppress prolyl hydroxylase domain-containing protein 2 (PHD2) activity via direct inhibition and via lactate dehydrogenase A (LDHA)-mediated generation of L-2-hydroxyglutarate (L2HG). Metabolic dysfunction induced by BCKAs was observed in the lungs of rats with pulmonary arterial hypertension (PAH) and in pulmonary artery smooth muscle cells (PASMCs) from idiopathic PAH patients. BCKA supplementation stimulated glycolytic activity and promoted a phenotypic switch to the synthetic phenotype in PASMCs of normal and PAH subjects. In summary, we identify BCKAs as novel signaling metabolites that activate HIF1α signaling in normoxia and that the BCKA-HIF1α pathway modulates VSMC function and may be relevant to pulmonary vascular pathobiology.
Collapse
Affiliation(s)
- Wusheng Xiao
- Divisions of Cardiovascular Medicine and Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, School of Public Health, Peking University, Beijing 100191, China
- Key Laboratory of State Administration of Traditional Chinese Medicine for Compatibility Toxicology, School of Public Health, Peking University, Beijing 100191, China
| | - Nishith Shrimali
- Divisions of Cardiovascular Medicine and Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - William M. Oldham
- Divisions of Cardiovascular Medicine and Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Clary B. Clish
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA
| | - Huamei He
- Divisions of Cardiovascular Medicine and Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Samantha J. Wong
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Bradley M. Wertheim
- Divisions of Cardiovascular Medicine and Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Elena Arons
- Divisions of Cardiovascular Medicine and Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Marcia C. Haigis
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Jane A. Leopold
- Divisions of Cardiovascular Medicine and Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Joseph Loscalzo
- Divisions of Cardiovascular Medicine and Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
3
|
Al-Saidi A, Alzaim IF, Hammoud SH, Al Arab G, Abdalla S, Mougharbil N, Eid AH, El-Yazbi AF. Interruption of perivascular and perirenal adipose tissue thromboinflammation rescues prediabetic cardioautonomic and renovascular deterioration. Clin Sci (Lond) 2024; 138:289-308. [PMID: 38381744 DOI: 10.1042/cs20231186] [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: 09/28/2023] [Revised: 02/20/2024] [Accepted: 02/21/2024] [Indexed: 02/23/2024]
Abstract
The cardiovascular and renovascular complications of metabolic deterioration are associated with localized adipose tissue dysfunction. We have previously demonstrated that metabolic impairment delineated the heightened vulnerability of both the perivascular (PVAT) and perirenal adipose tissue (PRAT) depots to hypoxia and inflammation, predisposing to cardioautonomic, vascular and renal deterioration. Interventions either addressing underlying metabolic disturbances or halting adipose tissue dysfunction rescued the observed pathological and functional manifestations. Several lines of evidence implicate adipose tissue thromboinflammation, which entails the activation of the proinflammatory properties of the blood clotting cascade, in the pathogenesis of metabolic and cardiovascular diseases. Despite offering valuable tools to interrupt the thromboinflammatory cycle, there exists a significant knowledge gap regarding the potential pleiotropic effects of anticoagulant drugs on adipose inflammation and cardiovascular function. As such, a systemic investigation of the consequences of PVAT and PRAT thromboinflammation and its interruption in the context of metabolic disease has not been attempted. Here, using an established prediabetic rat model, we demonstrate that metabolic disturbances are associated with PVAT and PRAT thromboinflammation in addition to cardioautonomic, vascular and renal functional decline. Administration of rivaroxaban, a FXa inhibitor, reduced PVAT and PRAT thromboinflammation and ameliorated the cardioautonomic, vascular and renal deterioration associated with prediabetes. Our present work outlines the involvement of PVAT and PRAT thromboinflammation during early metabolic derangement and offers novel perspectives into targeting adipose tissue thrombo-inflammatory pathways for the management its complications in future translational efforts.
Collapse
Affiliation(s)
- Aya Al-Saidi
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Ibrahim F Alzaim
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Safaa H Hammoud
- Department of Pharmacology and Therapeutics, Beirut Arab University, Beirut, Lebanon
| | - Ghida Al Arab
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Samaya Abdalla
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Nahed Mougharbil
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Ahmed F El-Yazbi
- Faculty of Pharmacy, Department of Pharmacology and Toxicology, Alexandria University, Alexandria, Egypt
- Faculty of Pharmacy and Research and Innovation Hub, Alamein International University, Alamein, Matrouh, Egypt
| |
Collapse
|
4
|
Nguyen TVH, Bergmann U, Kietzmann T, Mennerich D. Protein kinase B/AKT phosphorylates hypoxia-inducible factor-3α1 in response to insulin, promoting cell growth and migration. Front Cell Dev Biol 2023; 11:1250000. [PMID: 38020884 PMCID: PMC10665492 DOI: 10.3389/fcell.2023.1250000] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Hypoxia-inducible factors (HIFs) are best known for their roles in the adaptation to low oxygen environments. Besides hypoxia, HIF-1/2 α-subunits are also regulated by various non-hypoxic stimuli including insulin which can act via the PI3K/protein kinase B (PKB) signaling pathway. However, with respect to insulin little is known about HIF-3α. We aimed to investigate this relationship and found that insulin stimulates HIF-3α expression under both normal and low oxygen conditions. Blocking PKB activity reversed the effects of insulin, indicating that HIF-3α is a direct target of PKB. We identified serine 524, located in the oxygen-dependent degradation domain of HIF-3α, as a phosphorylation site of PKB. Mutating serine 524 impaired binding of PKB to HIF-3α and its ubiquitination, suggesting that PKB regulates HIF-3α stability through phosphorylation, thereby affecting important cellular processes such as cell viability and cell adhesion. Importantly, we discovered that this phosphorylation site also influenced insulin-dependent cell migration. These findings shed light on a novel mechanism by which insulin affects PKB-dependent HIF-3α expression and activity, with potential implications in metabolic diseases and cancer.
Collapse
Affiliation(s)
| | | | - Thomas Kietzmann
- Faculty of Biochemistry and Molecular Medicine, and Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Daniela Mennerich
- Faculty of Biochemistry and Molecular Medicine, and Biocenter Oulu, University of Oulu, Oulu, Finland
| |
Collapse
|
5
|
Kietzmann T. Vitamin C: From nutrition to oxygen sensing and epigenetics. Redox Biol 2023; 63:102753. [PMID: 37263060 PMCID: PMC10245123 DOI: 10.1016/j.redox.2023.102753] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/09/2023] [Accepted: 05/16/2023] [Indexed: 06/03/2023] Open
Abstract
Vitamin C is unbeatable - at least when it comes to sales. Of all the vitamin preparations, those containing vitamin C sell best. This is surprising because vitamin C deficiency is extremely rare. Nevertheless, there is still controversy about whether the additional intake of vitamin C supplements is essential for our health. In this context, the possible additional benefit is in most cases merely reduced to the known effect as an antioxidant. However, new findings in recent years on the mechanisms of oxygen-sensing and epigenetic control underpin the multifaceted role of vitamin C in a biological context and have therefore renewed interest in it. In the present article, therefore, known facts are linked to these new key data. In addition, available clinical data on vitamin C use of cancer therapy are summarized.
Collapse
Affiliation(s)
- Thomas Kietzmann
- University of Oulu, Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, P.O. Box 3000, 90014, Oulu, Finland.
| |
Collapse
|
6
|
Sutanto H, Soegiarto G. Risk of Thrombosis during and after a SARS-CoV-2 Infection: Pathogenesis, Diagnostic Approach, and Management. Hematol Rep 2023; 15:225-243. [PMID: 37092518 PMCID: PMC10123679 DOI: 10.3390/hematolrep15020024] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 03/07/2023] [Accepted: 03/24/2023] [Indexed: 04/05/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) increases the risk of thromboembolic events, especially in patients with severe infections requiring intensive care and cardiorespiratory support. COVID-19 patients with thromboembolic complications have a higher risk of death, and if they survive, these complications are expected to negatively affect these patients’ quality of life. Moreover, recent data reported that the risk of thromboembolism remains high months after a COVID-19 infection. Therefore, understanding the pathogenesis of thrombosis in the setting of COVID-19 may facilitate the early prevention and treatment of COVID-19-associated thromboembolism to reduce concomitant morbidity, mortality, and disability. This review will first discuss the clinical characteristics of COVID-19 infections, particularly with regard to the underlying pathophysiology. Then, the pathogenesis of COVID-19-associated thrombosis at the molecular and cellular levels will be comprehensively reviewed. Next, the clinical manifestations of venous and arterial thromboembolism in COVID-19 as well as the potential benefits of several laboratory markers of thrombosis will be further discussed. Lastly, the preventive and therapeutic management of thromboembolism during and after COVID-19 will also be explained.
Collapse
Affiliation(s)
- Henry Sutanto
- Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia
- Department of Internal Medicine, Dr. Soetomo Teaching Hospital, Surabaya 60286, Indonesia
| | - Gatot Soegiarto
- Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia
- Department of Internal Medicine, Dr. Soetomo Teaching Hospital, Surabaya 60286, Indonesia
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia
| |
Collapse
|
7
|
Jing W, Liu C, Su C, Liu L, Chen P, Li X, Zhang X, Yuan B, Wang H, Du X. Role of reactive oxygen species and mitochondrial damage in rheumatoid arthritis and targeted drugs. Front Immunol 2023; 14:1107670. [PMID: 36845127 PMCID: PMC9948260 DOI: 10.3389/fimmu.2023.1107670] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 01/30/2023] [Indexed: 02/11/2023] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterized by synovial inflammation, pannus formation, and bone and cartilage damage. It has a high disability rate. The hypoxic microenvironment of RA joints can cause reactive oxygen species (ROS) accumulation and mitochondrial damage, which not only affect the metabolic processes of immune cells and pathological changes in fibroblastic synovial cells but also upregulate the expression of several inflammatory pathways, ultimately promoting inflammation. Additionally, ROS and mitochondrial damage are involved in angiogenesis and bone destruction, thereby accelerating RA progression. In this review, we highlighted the effects of ROS accumulation and mitochondrial damage on inflammatory response, angiogenesis, bone and cartilage damage in RA. Additionally, we summarized therapies that target ROS or mitochondria to relieve RA symptoms and discuss the gaps in research and existing controversies, hoping to provide new ideas for research in this area and insights for targeted drug development in RA.
Collapse
Affiliation(s)
- Weiyao Jing
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, China
| | - Cui Liu
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, China
| | - Chenghong Su
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, China
| | - Limei Liu
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, China
| | - Ping Chen
- Department of Rheumatic and Bone Disease, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, China
| | - Xiangjun Li
- Department of Rheumatic and Bone Disease, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, China
| | - Xinghua Zhang
- Department of Acupuncture, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, China
| | - Bo Yuan
- Department of Acupuncture and Pain, Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Haidong Wang
- Department of Rheumatic and Bone Disease, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, China
| | - Xiaozheng Du
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, China
| |
Collapse
|
8
|
Kurniansyah N, Wallace DA, Zhang Y, Yu B, Cade B, Wang H, Ochs-Balcom HM, Reiner AP, Ramos AR, Smith JD, Cai J, Daviglus M, Zee PC, Kaplan R, Kooperberg C, Rich SS, Rotter JI, Gharib SA, Redline S, Sofer T. An integrated multi-omics analysis of sleep-disordered breathing traits implicates P2XR4 purinergic signaling. Commun Biol 2023; 6:125. [PMID: 36721044 PMCID: PMC9889381 DOI: 10.1038/s42003-023-04520-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 01/23/2023] [Indexed: 02/01/2023] Open
Abstract
Sleep Disordered Breathing (SDB) is a common disease associated with increased risk for cardiometabolic, cardiovascular, and cognitive diseases. How SDB affects the molecular environment is still poorly understood. We study the association of three SDB measures with gene expression measured using RNA-seq in multiple blood tissues from the Multi-Ethnic Study of Atherosclerosis. We develop genetic instrumental variables for the associated transcripts as polygenic risk scores (tPRS), then generalize and validate the tPRS in the Women's Health Initiative. We measure the associations of the validated tPRS with SDB and serum metabolites in Hispanic Community Health Study/Study of Latinos. Here we find differential gene expression by blood cell type in relation to SDB traits and link P2XR4 expression to average oxyhemoglobin saturation during sleep and butyrylcarnitine (C4) levels. These findings can be used to develop interventions to alleviate the effect of SDB on the human molecular environment.
Collapse
Affiliation(s)
- Nuzulul Kurniansyah
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
| | - Danielle A Wallace
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
| | - Ying Zhang
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
| | - Bing Yu
- Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Brian Cade
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
| | - Heming Wang
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
| | - Heather M Ochs-Balcom
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - Alexander P Reiner
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, The State University of New York, Buffalo, NY, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Alberto R Ramos
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Joshua D Smith
- Northwest Genomic Center, University of Washington, Seattle, WA, USA
| | - Jianwen Cai
- Department of Biostatistics, University of North Carolina, at Chapel Hill, NC, USA
| | - Martha Daviglus
- Institute for Minority Health Research, University of Illinois at Chicago, Chicago, IL, USA
| | - Phyllis C Zee
- Division of Sleep Medicine, Department of Neurology, Northwestern University, Chicago, IL, USA
| | - Robert Kaplan
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Epidemiology & Population Health, Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Sina A Gharib
- Computational Medicine Core, Center for Lung Biology, UW Medicine Sleep Center, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Susan Redline
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
| | - Tamar Sofer
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA.
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA.
- Departments of Medicine and of Biostatistics, Harvard University, Boston, MA, USA.
| |
Collapse
|
9
|
Zheng H, Huang N, Lin JQ, Yan LY, Jiang QG, Yang WZ. Effect and mechanism of pirfenidone combined with 2-methoxy-estradiol perfusion through portal vein on hepatic artery hypoxia-induced hepatic fibrosis. Adv Med Sci 2023; 68:46-53. [PMID: 36610261 DOI: 10.1016/j.advms.2022.12.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 09/20/2022] [Accepted: 12/07/2022] [Indexed: 01/07/2023]
Abstract
PURPOSE The aim of this study was to explore the effect and mechanism of pirfenidone (PFD) combined with 2-methoxyestradiol (2-ME2) perfusion through portal vein on hepatic artery hypoxia-induced hepatic fibrosis. MATERIALS AND METHODS Sprague-Dawley rats were divided into 5 groups (n = 3/group): control group, hepatic artery ligation (HAL) group, HAL + PFD (portal vein perfusion of PFD) group, HAL+2-ME2 (portal vein perfusion of 2-ME2) group and HAL + PFD+2-ME2 group depending on whether they received HAL and/or portal vein perfusion (PFD and/or 2-ME2). Livers were harvested for pathology, western blotting (WB), and quantitative real-time PCR (qRT-PCR). RESULTS Sirius red staining showed that portal vein perfusion of drugs resulted in degradation of liver fibrosis. Immunohistochemistry showed decreased hypoxia-inducible factor-1 α (HIF-1α) and α-smooth muscle actin (α-SMA) after portal intravenous drugs infusion compared with HAL group (P < 0.05). WB analysis showed increased Smad7 in HAL + PFD group compared with HAL group (P < 0.05). qRT-PCR analysis showed decreased matrix metallo-proteinase 2 (MMP2), transforming growth factor β1 (TGF-β1), monocyte chemoattractant protein-1 (MCP-1), and Collagen I mRNA in HAL + PFD group except for tissue inhibitor of metalloproteinase-1 (TIMP-1) compared with HAL group (P < 0.05). Compared with HAL + PFD group, the addition of 2-ME2 did not lead to better results in qRT-PCR analysis. CONCLUSIONS The portal vein perfusion of PFD significantly reduced the hepatic artery hypoxia-induced fibrosis degree in treated rats by down-regulating the expression of HIF-1α, α-SMA, MMP2, TGF-β1, MCP-1, and Collagen I, as well as up-regulating the TIMP-1 expression and Smad7 protein level. Combined 2-ME2 infusion was not better than PFD alone.
Collapse
Affiliation(s)
- Hui Zheng
- Department of Interventional Radiology, Fujian Medical University Union Hospital, Fuzhou, Fujian, People's Republic of China
| | - Ning Huang
- Department of Interventional Radiology, Fujian Medical University Union Hospital, Fuzhou, Fujian, People's Republic of China
| | - Jun-Qing Lin
- Department of Interventional Radiology, Fujian Medical University Union Hospital, Fuzhou, Fujian, People's Republic of China
| | - Le-Ye Yan
- Department of Interventional Radiology, Fujian Medical University Union Hospital, Fuzhou, Fujian, People's Republic of China
| | - Qing-Gui Jiang
- Department of Interventional Radiology, Fujian Medical University Union Hospital, Fuzhou, Fujian, People's Republic of China; Department of Interventional Therapy, Xiamen Humanity Hospital, Xiamen, Fujian, People's Republic of China
| | - Wei-Zhu Yang
- Department of Interventional Radiology, Fujian Medical University Union Hospital, Fuzhou, Fujian, People's Republic of China.
| |
Collapse
|
10
|
Iacobini C, Vitale M, Haxhi J, Pesce C, Pugliese G, Menini S. Mutual Regulation between Redox and Hypoxia-Inducible Factors in Cardiovascular and Renal Complications of Diabetes. Antioxidants (Basel) 2022; 11:2183. [PMID: 36358555 PMCID: PMC9686572 DOI: 10.3390/antiox11112183] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/31/2022] [Accepted: 11/02/2022] [Indexed: 08/30/2023] Open
Abstract
Oxidative stress and hypoxia-inducible factors (HIFs) have been implicated in the pathogenesis of diabetic cardiovascular and renal diseases. Reactive oxygen species (ROS) mediate physiological and pathophysiological processes, being involved in the modulation of cell signaling, differentiation, and survival, but also in cyto- and genotoxic damage. As master regulators of glycolytic metabolism and oxygen homeostasis, HIFs have been largely studied for their role in cell survival in hypoxic conditions. However, in addition to hypoxia, other stimuli can regulate HIFs stability and transcriptional activity, even in normoxic conditions. Among these, a regulatory role of ROS and their byproducts on HIFs, particularly the HIF-1α isoform, has received growing attention in recent years. On the other hand, HIF-1α and HIF-2α exert mutually antagonistic effects on oxidative damage. In diabetes, redox-mediated HIF-1α deregulation contributes to the onset and progression of cardiovascular and renal complications, and recent findings suggest that deranged HIF signaling induced by hyperglycemia and other cellular stressors associated with metabolic disorders may cause mitochondrial dysfunction, oxidative stress, and inflammation. Understanding the mechanisms of mutual regulation between HIFs and redox factors and the specific contribution of the two main isoforms of HIF-α is fundamental to identify new therapeutic targets for vascular complications of diabetes.
Collapse
Affiliation(s)
- Carla Iacobini
- Department of Clinical and Molecular Medicine, “La Sapienza” University, 00189 Rome, Italy
| | - Martina Vitale
- Department of Clinical and Molecular Medicine, “La Sapienza” University, 00189 Rome, Italy
| | - Jonida Haxhi
- Department of Clinical and Molecular Medicine, “La Sapienza” University, 00189 Rome, Italy
| | - Carlo Pesce
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetic and Maternal Infantile Sciences (DINOGMI), Department of Excellence of MIUR, University of Genoa Medical School, 16132 Genoa, Italy
| | - Giuseppe Pugliese
- Department of Clinical and Molecular Medicine, “La Sapienza” University, 00189 Rome, Italy
| | - Stefano Menini
- Department of Clinical and Molecular Medicine, “La Sapienza” University, 00189 Rome, Italy
| |
Collapse
|
11
|
Ferrer JLM, Garcia RL. Antioxidant Systems, lncRNAs, and Tunneling Nanotubes in Cell Death Rescue from Cigarette Smoke Exposure. Cells 2022; 11:2277. [PMID: 35892574 PMCID: PMC9330437 DOI: 10.3390/cells11152277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/17/2022] [Accepted: 07/19/2022] [Indexed: 12/10/2022] Open
Abstract
Cigarette smoke is a rich source of carcinogens and reactive oxygen species (ROS) that can damage macromolecules including DNA. Repair systems can restore DNA integrity. Depending on the duration or intensity of stress signals, cells may utilize various survival and adaptive mechanisms. ROS levels are kept in check through redundant detoxification processes controlled largely by antioxidant systems. This review covers and expands on the mechanisms available to cigarette smoke-exposed cancer cells for restoring the redox balance. These include multiple layers of transcriptional control, each of which is posited to be activated upon reaching a particular stress threshold, among them the NRF2 pathway, the AP-1 and NF-kB pathways, and, finally, TP53, which triggers apoptosis if extreme toxicity is reached. The review also discusses long noncoding RNAs, which have been implicated recently in regulating oxidative stress-with roles in ROS detoxification, the inflammatory response, oxidative stress-induced apoptosis, and mitochondrial oxidative phosphorylation. Lastly, the emerging roles of tunneling nanotubes in providing additional mechanisms for metabolic rescue and the regulation of redox imbalance are considered, further highlighting the expanded redox reset arsenal available to cells.
Collapse
Affiliation(s)
| | - Reynaldo L. Garcia
- Disease Molecular Biology and Epigenetics Laboratory, National Institute of Molecular Biology and Biotechnology, University of the Philippines Diliman, Quezon City 1101, Philippines;
| |
Collapse
|
12
|
Zolotoff C, Puech C, Roche F, Perek N. Effects of intermittent hypoxia with thrombin in an in vitro model of human brain endothelial cells and their impact on PAR-1/PAR-3 cleavage. Sci Rep 2022; 12:12305. [PMID: 35853902 PMCID: PMC9296553 DOI: 10.1038/s41598-022-15592-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 06/27/2022] [Indexed: 11/09/2022] Open
Abstract
Patients with obstructive sleep apnea/hypopnea (OSA) are at high risk of cerebrovascular diseases leading to cognitive impairment. The oxidative stress generated by intermittent hypoxia (IH) could lead to an increase in blood-brain barrier (BBB) permeability, an essential interface for the protection of the brain. Moreover, in patients with OSA, blood coagulation could be increased leading to cardiovascular complications. Thrombin is a factor found increased in these populations that exerts various cellular effects through activation of protease activated receptors (PARs). Thus, we have evaluated in an in vitro BBB model the association of IH with thrombin at two concentrations. We measured the apparent BBB permeability, expression of tight junctions, ROS production, HIF-1α expression, and cleavage of PAR-1/PAR-3. Pre-treatment with dabigatran was performed. IH and higher thrombin concentrations altered BBB permeability: high levels of HIF-1α expression, ROS and PAR-1 activation compared to PAR-3 in such conditions. Conversely, lower concentration of thrombin associated with IH appear to have a protective effect on BBB with a significant cleavage of PAR-3. Dabigatran reversed the deleterious effect of thrombin at high concentrations but also suppressed the beneficial effect of low dose thrombin. Therefore, thrombin and PARs represent novel attractive targets to prevent BBB opening in OSA.
Collapse
Affiliation(s)
- Cindy Zolotoff
- INSERM, U1059, Sainbiose, Dysfonction Vasculaire et Hémostase, Université Jean Monnet Saint-Etienne, Saint-Priest-en-Jarez, France. .,Faculté de Médecine - Campus Santé Innovations, 10 Rue de la Marandière, 42270, Saint-Priest-en-Jarez, France.
| | - Clémentine Puech
- INSERM, U1059, Sainbiose, Dysfonction Vasculaire et Hémostase, Université Jean Monnet Saint-Etienne, Saint-Priest-en-Jarez, France
| | - Frédéric Roche
- INSERM, U1059, Sainbiose, Dysfonction Vasculaire et Hémostase, Université Jean Monnet Saint-Etienne, Saint-Priest-en-Jarez, France.,Service de Physiologie Clinique Et de L'Exercice, Centre VISAS, CHU Saint Etienne, Saint-Priest-en-Jarez, France
| | - Nathalie Perek
- INSERM, U1059, Sainbiose, Dysfonction Vasculaire et Hémostase, Université Jean Monnet Saint-Etienne, Saint-Priest-en-Jarez, France
| |
Collapse
|
13
|
Badran M, Gozal D. PAI-1: A Major Player in the Vascular Dysfunction in Obstructive Sleep Apnea? Int J Mol Sci 2022; 23:5516. [PMID: 35628326 PMCID: PMC9141273 DOI: 10.3390/ijms23105516] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/05/2022] [Accepted: 05/12/2022] [Indexed: 02/04/2023] Open
Abstract
Obstructive sleep apnea is a chronic and prevalent condition that is associated with endothelial dysfunction, atherosclerosis, and imposes excess overall cardiovascular risk and mortality. Despite its high prevalence and the susceptibility of CVD patients to OSA-mediated stressors, OSA is still under-recognized and untreated in cardiovascular practice. Moreover, conventional OSA treatments have yielded either controversial or disappointing results in terms of protection against CVD, prompting the need for the identification of additional mechanisms and associated adjuvant therapies. Plasminogen activator inhibitor-1 (PAI-1), the primary inhibitor of tissue-type plasminogen activator (tPA) and urinary-type plasminogen activator (uPA), is a key regulator of fibrinolysis and cell migration. Indeed, elevated PAI-1 expression is associated with major cardiovascular adverse events that have been attributed to its antifibrinolytic activity. However, extensive evidence indicates that PAI-1 can induce endothelial dysfunction and atherosclerosis through complex interactions within the vasculature in an antifibrinolytic-independent matter. Elevated PAI-1 levels have been reported in OSA patients. However, the impact of PAI-1 on OSA-induced CVD has not been addressed to date. Here, we provide a comprehensive review on the mechanisms by which OSA and its most detrimental perturbation, intermittent hypoxia (IH), can enhance the transcription of PAI-1. We also propose causal pathways by which PAI-1 can promote atherosclerosis in OSA, thereby identifying PAI-1 as a potential therapeutic target in OSA-induced CVD.
Collapse
Affiliation(s)
- Mohammad Badran
- Department of Child Health and Child Health Research Institute, School of Medicine, University of Missouri, 400 N Keene St, Suite 010, Columbia, MO 65201, USA;
| | - David Gozal
- Department of Child Health and Child Health Research Institute, School of Medicine, University of Missouri, 400 N Keene St, Suite 010, Columbia, MO 65201, USA;
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO 65201, USA
| |
Collapse
|
14
|
Gkotinakou IM, Mylonis I, Tsakalof A. Vitamin D and Hypoxia: Points of Interplay in Cancer. Cancers (Basel) 2022; 14:cancers14071791. [PMID: 35406562 PMCID: PMC8997790 DOI: 10.3390/cancers14071791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/29/2022] [Accepted: 03/30/2022] [Indexed: 11/16/2022] Open
Abstract
Vitamin D is a hormone that, through its action, elicits a broad spectrum of physiological responses ranging from classic to nonclassical actions such as bone morphogenesis and immune function. In parallel, many studies describe the antiproliferative, proapoptotic, antiangiogenic effects of calcitriol (the active hormonal form) that contribute to its anticancer activity. Additionally, epidemiological data signify the inverse correlation between vitamin D levels and cancer risk. On the contrary, tumors possess several adaptive mechanisms that enable them to evade the anticancer effects of calcitriol. Such maladaptive processes are often a characteristic of the cancer microenvironment, which in solid tumors is frequently hypoxic and elicits the overexpression of Hypoxia-Inducible Factors (HIFs). HIF-mediated signaling not only contributes to cancer cell survival and proliferation but also confers resistance to anticancer agents. Taking into consideration that calcitriol intertwines with signaling events elicited by the hypoxic status cells, this review examines their interplay in cellular signaling to give the opportunity to better understand their relationship in cancer development and their prospect for the treatment of cancer.
Collapse
Affiliation(s)
| | - Ilias Mylonis
- Correspondence: (I.M.); (A.T.); Tel.: +30-2410-685578 (I.M. & A.T)
| | - Andreas Tsakalof
- Correspondence: (I.M.); (A.T.); Tel.: +30-2410-685578 (I.M. & A.T)
| |
Collapse
|
15
|
Bahraini M, Dorgalaleh A. The Impact of SARS-CoV-2 Infection on Blood Coagulation and Fibrinolytic Pathways: A Review of Prothrombotic Changes Caused by COVID-19. Semin Thromb Hemost 2021; 48:19-30. [PMID: 34695858 DOI: 10.1055/s-0041-1736166] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The cardinal pathology of coronavirus disease 2019 (COVID-19) is a primary infection of pulmonary tract cells by severe acute respiratory syndrome coronavirus 2, provoking a local inflammatory response, often accompanied by cytokine storm and acute respiratory distress syndrome, especially in patients with severe disease. Systemic propagation of the disease may associate with thrombotic events, including deep vein thrombosis, pulmonary embolism, and thrombotic microangiopathy, which are important causes of morbidity and mortality in patients with COVID-19. This narrative review describes current knowledge of the pathophysiological mechanisms of COVID-19-associated coagulopathy, with focus on prothrombotic changes in hemostatic mediators, including plasma levels of clotting factors, natural anticoagulants, components of fibrinolytic system, and platelets. It will also highlight the central role of endothelial cells in COVID-19-associated coagulopathy. This narrative review discusses also potential therapeutic strategies for managing thrombotic complications. Awareness by medical experts of contributors to the pathogenesis of thrombotic events in COVID-19 is imperative to develop therapeutics not limited to regular anticoagulants. Instituting cooperation among medical personnel and researchers may lessen this novel virus' impact now, and in the event of recurrence.
Collapse
Affiliation(s)
- Mehran Bahraini
- Department of Hematology and Blood Transfusion, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Akbar Dorgalaleh
- Department of Hematology and Blood Transfusion, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
16
|
Celik D, Kantarci A. Vascular Changes and Hypoxia in Periodontal Disease as a Link to Systemic Complications. Pathogens 2021; 10:1280. [PMID: 34684229 PMCID: PMC8541389 DOI: 10.3390/pathogens10101280] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/27/2021] [Accepted: 10/02/2021] [Indexed: 12/13/2022] Open
Abstract
The hypoxic microenvironment caused by oral pathogens is the most important cause of the disruption of dynamic hemostasis between the oral microbiome and the immune system. Periodontal infection exacerbates the inflammatory response with increased hypoxia and causes vascular changes. The chronicity of inflammation becomes systemic as a link between oral and systemic diseases. The vascular network plays a central role in controlling infection and regulating the immune response. In this review, we focus on the local and systemic vascular network change mechanisms of periodontal inflammation and the pathological processes of inflammatory diseases. Understanding how the vascular network influences the pathology of periodontal diseases and the systemic complication associated with this pathology is essential for the discovery of both local and systemic proactive control mechanisms.
Collapse
Affiliation(s)
- Dilek Celik
- Immunology Division, Health Sciences Institute, Trakya University, Edirne 22100, Turkey;
| | - Alpdogan Kantarci
- Forsyth Institute, Cambridge, MA 02142, USA
- School of Dental Medicine, Harvard University, Boston, MA 02142, USA
| |
Collapse
|
17
|
Takaishi K, Kinoshita H, Kawashima S, Kawahito S. Human Vascular Smooth Muscle Function and Oxidative Stress Induced by NADPH Oxidase with the Clinical Implications. Cells 2021; 10:cells10081947. [PMID: 34440716 PMCID: PMC8393371 DOI: 10.3390/cells10081947] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 01/05/2023] Open
Abstract
Among reactive oxygen species, superoxide mediates the critical vascular redox signaling, resulting in the regulation of the human cardiovascular system. The reduced form of nicotinamide adenine dinucleotide phosphate oxidase (NADPH oxidase, NOX) is the source of superoxide and relates to the crucial intracellular pathology and physiology of vascular smooth muscle cells, including contraction, proliferation, apoptosis, and inflammatory response. Human vascular smooth muscle cells express NOX1, 2, 4, and 5 in physiological and pathological conditions, and those enzymes play roles in most cardiovascular disorders caused by hypertension, diabetes, inflammation, and arteriosclerosis. Various physiologically active substances, including angiotensin II, stimulate NOX via the cytosolic subunits’ translocation toward the vascular smooth muscle cell membrane. As we have shown, some pathological stimuli such as high glucose augment the enzymatic activity mediated by the phosphatidylinositol 3-kinase-Akt pathway, resulting in the membrane translocation of cytosolic subunits of NOXs. This review highlights and details the roles of human vascular smooth muscle NOXs in the pathophysiology and clinical aspects. The regulation of the enzyme expressed in the vascular smooth muscle cells may lead to the prevention and treatment of human cardiovascular diseases.
Collapse
Affiliation(s)
- Kazumi Takaishi
- Department of Dental Anesthesiology, Graduate School of Biomedical Sciences, Tokushima University, 3-18-15, Kuramoto, Tokushima 770-8504, Japan; (K.T.); (S.K.)
| | - Hiroyuki Kinoshita
- Department of Anesthesiology, Graduate School of Biomedical Sciences, Tokushima University, 3-18-15, Kuramoto, Tokushima 770-8504, Japan
- Department of Anesthesiology and Intensive Care, School of Medicine, Hamamatsu University, 1-20-1, Handayama, Hamamatsu City 431-3192, Japan;
- Correspondence: ; Tel.: +81-53-436-1251
| | - Shingo Kawashima
- Department of Anesthesiology and Intensive Care, School of Medicine, Hamamatsu University, 1-20-1, Handayama, Hamamatsu City 431-3192, Japan;
| | - Shinji Kawahito
- Department of Dental Anesthesiology, Graduate School of Biomedical Sciences, Tokushima University, 3-18-15, Kuramoto, Tokushima 770-8504, Japan; (K.T.); (S.K.)
| |
Collapse
|
18
|
Machcinska S, Kopcewicz M, Bukowska J, Walendzik K, Gawronska-Kozak B. Impairment of the Hif-1α regulatory pathway in Foxn1-deficient (Foxn1 -/- ) mice affects the skin wound healing process. FASEB J 2021; 35:e21289. [PMID: 33475195 DOI: 10.1096/fj.202001907r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 11/13/2020] [Accepted: 12/03/2020] [Indexed: 02/06/2023]
Abstract
Hypoxia and hypoxia-regulated factors (eg, hypoxia-inducible factor-1α [Hif-1α], factor inhibiting Hif-1α [Fih-1], thioredoxin-1 [Trx-1], aryl hydrocarbon receptor nuclear translocator 2 [Arnt-2]) have essential roles in skin wound healing. Using Foxn1-/- mice that can heal skin injuries in a unique scarless manner, we investigated the interaction between Foxn1 and hypoxia-regulated factors. The Foxn1-/- mice displayed impairments in the regulation of Hif-1α, Trx-1, and Fih-1 but not Arnt-2 during the healing process. An analysis of wounded skin showed that the skin of the Foxn1-/- mice healed in a scarless manner, displaying rapid re-epithelialization and an increase in transforming growth factor β (Tgfβ-3) and collagen III expression. An in vitro analysis revealed that Foxn1 overexpression in keratinocytes isolated from the skin of the Foxn1-/- mice led to reduced Hif-1α expression in normoxic but not hypoxic cultures and inhibited Fih-1 expression exclusively under hypoxic conditions. These data indicate that in the skin, Foxn1 affects hypoxia-regulated factors that control the wound healing process and suggest that under normoxic conditions, Foxn1 is a limiting factor for Hif-1α.
Collapse
Affiliation(s)
- Sylwia Machcinska
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Marta Kopcewicz
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Joanna Bukowska
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Katarzyna Walendzik
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Barbara Gawronska-Kozak
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| |
Collapse
|
19
|
Li LM, Chen C, Ran RX, Huang JT, Sun HL, Zeng C, Zhang Z, Zhang W, Liu SM. Loss of TARBP2 Drives the Progression of Hepatocellular Carcinoma via miR-145-SERPINE1 Axis. Front Oncol 2021; 11:620912. [PMID: 34249676 PMCID: PMC8265608 DOI: 10.3389/fonc.2021.620912] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 03/09/2021] [Indexed: 12/13/2022] Open
Abstract
The clinical outcomes of hepatocellular carcinoma (HCC) remain dismal. Elucidating the molecular mechanisms for the progression of aggressive HCC holds the promise for developing novel intervention strategies. The transactivation response element RNA-binding protein (TRBP/TARBP2), a key component of microRNA (miRNA) processing and maturation machinery has been shown to play conflicting roles in tumor development and progression. We sought to investigate the expression of TARBP2 in HCC using well-characterized HCC cell lines, patient-derived tissues and blood samples. Additionally, the potential prognostic and diagnostic value of TARBP2 in HCC were analyzed using Kaplan-Meier plots and ROC curve. Cell counting kit-8 (CCK-8), wound healing and transwell assays examined the ability of TARBP2 to induce cell proliferation, migration, and invasion in HCC cell lines. RNA sequencing was applied to identify the downstream elements of TARBP2. The interaction of potential targets of TARBP2, miR-145 and serpin family E member 1 (SERPINE1), was assessed using luciferase reporter assay. TARBP2 expression was down-regulated in HCC cell lines relative to normal hepatocyte cells, with a similar pattern further confirmed in tissue and blood samples. Notably, the loss of TARBP2 was demonstrated to promote proliferation, migration, and invasion in HCC cell lines. Interestingly, the reduction of TARBP2 was shown to result in the upregulation of SERPINE1, also known as plasminogen activator inhibitor (PAI-1), which is a vital gene of the HIF-1 signaling pathway. Knockdown of SERPINE1 rescued the TARBP2-lost phenotype. Moreover, TARBP2 depletion induced the upregulation of SERPINE1 through reducing the processing of miR-145, which directly targets SERPINE1. Finally, overexpression of miR-145 repressed SERPINE1 and rescued the functions in sh-TARBP2 HCC cells. Our findings underscore a linear TARBP2-miR-145-SERPINE1 pathway that drives HCC progression, with the potential as a novel intervention target for aggressive HCC.
Collapse
Affiliation(s)
- Li-Man Li
- Department of Clinical Laboratory, Center for Gene Diagnosis, and Program of Clinical Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Chang Chen
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Ruo-Xi Ran
- Department of Clinical Laboratory, Center for Gene Diagnosis, and Program of Clinical Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jing-Tao Huang
- Department of Clinical Laboratory, Center for Gene Diagnosis, and Program of Clinical Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China.,Department of Clinical Laboratory, Renmin Hospital, Wuhan University, Wuhan, China
| | - Hui-Lung Sun
- Department of Chemistry and Institute for Biophysical Dynamics, Howard Hughes Medical Institute, The University of Chicago, Chicago, IL, United States
| | - Chang Zeng
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Zhou Zhang
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Wei Zhang
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States.,Institute of Precision Medicine, Jining Medical University, Jining, China
| | - Song-Mei Liu
- Department of Clinical Laboratory, Center for Gene Diagnosis, and Program of Clinical Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Province Key Laboratory of Allergy and Immunology, Wuhan, China
| |
Collapse
|
20
|
Slivka D, Dumke C, Hailes W, Ruby B. Impact of Hypoxic Exercise Recovery on Skeletal Muscle Glycogen and Gene Expression. High Alt Med Biol 2021; 22:300-307. [PMID: 34142871 DOI: 10.1089/ham.2021.0028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Slivka, Dustin, Charles Dumke, Walter Hailes, and Brent Ruby. Impact of hypoxic exercise recovery on skeletal muscle glycogen and gene expression. High Alt Med Biol. 22:300-307, 2021. Background: The impact of altitude during recovery from exercise is largely unknown. The purpose of this study was to determine the acute gene response and muscle glycogen re-synthesis after exercise when exposed to simulated high altitude during recovery. Materials and Methods: Twelve male participants (age, 25 ± 2 years; height, 178 ± 7 cm; weight, 78.8 ± 7.8 kg; VO2peak, 4.25 ± 0.59 l/min; Wpeak 307 ± 44 W; and body fat, 13.1% ± 1.2%) completed two trials (random order), which consisted of cycling for 90 minutes in laboratory conditions and then recovering for 6 hours in laboratory conditions (975 m; normoxia) or at a high simulated altitude (5,000 m; hypoxia). Results: Skeletal muscle biopsies from the vastus lateralis were obtained before exercise, after exercise, and 6 hours after exercise for the measurement of metabolic gene expression and muscle glycogen. Muscle glycogen decreased with exercise (61% ± 13%, p < 0.05) and increased with recovery (78% ± 35%, p < 0.05) with no difference between trials (p > 0.05). Hypoxia-inducible factor (HIF)-1α, HIF-2α, optic atrophy gene 1 (OPA-1), mitofusin 2 (MFN-2), and peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC-1α) gene expression were suppressed after altitude exposure (p < 0.05), while mitochondrial fission 1 protein (FIS-1), phosphofructokinase (PFK), Cytochrome c oxidase (COX), and hexokinase (HK) were unaffected by altitude exposure (p > 0.05). Conclusions: High-altitude exposure during recovery from exercise inhibits gene expression associated with mitochondrial development without affecting muscle glycogen re-synthesis.
Collapse
Affiliation(s)
- Dustin Slivka
- School of Health and Kinesiology, University of Nebraska at Omaha, Omaha, Nebraska, USA
| | - Charles Dumke
- School of Integrative Physiology and Athletic Training, University of Montana, Missoula, Montana, USA
| | - Walter Hailes
- School of Integrative Physiology and Athletic Training, University of Montana, Missoula, Montana, USA
| | - Brent Ruby
- School of Integrative Physiology and Athletic Training, University of Montana, Missoula, Montana, USA
| |
Collapse
|
21
|
The pleiotropic effects of antithrombotic drugs in the metabolic-cardiovascular-neurodegenerative disease continuum: impact beyond reduced clotting. Clin Sci (Lond) 2021; 135:1015-1051. [PMID: 33881143 DOI: 10.1042/cs20201445] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 04/07/2021] [Accepted: 04/13/2021] [Indexed: 12/25/2022]
Abstract
Antithrombotic drugs are widely used for primary and secondary prevention, as well as treatment of many cardiovascular disorders. Over the past few decades, major advances in the pharmacology of these agents have been made with the introduction of new drug classes as novel therapeutic options. Accumulating evidence indicates that the beneficial outcomes of some of these antithrombotic agents are not solely related to their ability to reduce thrombosis. Here, we review the evidence supporting established and potential pleiotropic effects of four novel classes of antithrombotic drugs, adenosine diphosphate (ADP) P2Y12-receptor antagonists, Glycoprotein IIb/IIIa receptor Inhibitors, and Direct Oral Anticoagulants (DOACs), which include Direct Factor Xa (FXa) and Direct Thrombin Inhibitors. Specifically, we discuss the molecular evidence supporting such pleiotropic effects in the context of cardiovascular disease (CVD) including endothelial dysfunction (ED), atherosclerosis, cardiac injury, stroke, and arrhythmia. Importantly, we highlight the role of DOACs in mitigating metabolic dysfunction-associated cardiovascular derangements. We also postulate that DOACs modulate perivascular adipose tissue inflammation and thus, may reverse cardiovascular dysfunction early in the course of the metabolic syndrome. In this regard, we argue that some antithrombotic agents can reverse the neurovascular damage in Alzheimer's and Parkinson's brain and following traumatic brain injury (TBI). Overall, we attempt to provide an up-to-date comprehensive review of the less-recognized, beneficial molecular aspects of antithrombotic therapy beyond reduced thrombus formation. We also make a solid argument for the need of further mechanistic analysis of the pleiotropic effects of antithrombotic drugs in the future.
Collapse
|
22
|
Mylonis I, Chachami G, Simos G. Specific Inhibition of HIF Activity: Can Peptides Lead the Way? Cancers (Basel) 2021; 13:cancers13030410. [PMID: 33499237 PMCID: PMC7865418 DOI: 10.3390/cancers13030410] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/15/2021] [Accepted: 01/16/2021] [Indexed: 12/19/2022] Open
Abstract
Simple Summary Cancer cells in solid tumors often experience lack of oxygen (hypoxia), which they overcome with the help of hypoxia inducible transcription factors (HIFs). When HIFs are activated, they stimulate the expression of many genes and cause the production of proteins that help cancer cells grow and migrate even in the presence of very little oxygen. Many experiments have shown that agents that block the activity of HIFs (HIF inhibitors) can prevent growth of cancer cells under hypoxia and, subsequently, hinder formation of malignant tumors or metastases. Most small chemical HIF inhibitors lack the selectivity required for development of safe anticancer drugs. On the other hand, peptides derived from HIFs themselves can be very selective HIF inhibitors by disrupting specific associations of HIFs with cellular components that are essential for HIF activation. This review discusses the nature of available peptide HIF inhibitors and their prospects as effective pharmaceuticals against cancer. Abstract Reduced oxygen availability (hypoxia) is a characteristic of many disorders including cancer. Central components of the systemic and cellular response to hypoxia are the Hypoxia Inducible Factors (HIFs), a small family of heterodimeric transcription factors that directly or indirectly regulate the expression of hundreds of genes, the products of which mediate adaptive changes in processes that include metabolism, erythropoiesis, and angiogenesis. The overexpression of HIFs has been linked to the pathogenesis and progression of cancer. Moreover, evidence from cellular and animal models have convincingly shown that targeting HIFs represents a valid approach to treat hypoxia-related disorders. However, targeting transcription factors with small molecules is a very demanding task and development of HIF inhibitors with specificity and therapeutic potential has largely remained an unattainable challenge. Another promising approach to inhibit HIFs is to use peptides modelled after HIF subunit domains known to be involved in protein–protein interactions that are critical for HIF function. Introduction of these peptides into cells can inhibit, through competition, the activity of endogenous HIFs in a sequence and, therefore also isoform, specific manner. This review summarizes the involvement of HIFs in cancer and the approaches for targeting them, with a special focus on the development of peptide HIF inhibitors and their prospects as highly-specific pharmacological agents.
Collapse
Affiliation(s)
- Ilias Mylonis
- Laboratory of Biochemistry, Faculty of Medicine, University of Thessaly, 41500 Larissa, Greece;
- Correspondence: (I.M.); (G.S.)
| | - Georgia Chachami
- Laboratory of Biochemistry, Faculty of Medicine, University of Thessaly, 41500 Larissa, Greece;
| | - George Simos
- Laboratory of Biochemistry, Faculty of Medicine, University of Thessaly, 41500 Larissa, Greece;
- Gerald Bronfman Department of Oncology, Faculty of Medicine, McGill University, Montreal, QC H4A 3T2, Canada
- Correspondence: (I.M.); (G.S.)
| |
Collapse
|
23
|
Şimşek F, Işık Ü, Aktepe E, Kılıç F, Şirin FB, Bozkurt M. Comparison of Serum VEGF, IGF-1, and HIF-1α Levels in Children with Autism Spectrum Disorder and Healthy Controls. J Autism Dev Disord 2021; 51:3564-3574. [PMID: 33389301 DOI: 10.1007/s10803-020-04820-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/26/2020] [Indexed: 01/13/2023]
Abstract
The aim of this study was to determine whether serum VEGF, IGF-1, and HIF-1α levels differed between Autism Spectrum Disorder (ASD) patients and healthy controls. A total of 40 children with ASD and 40 healthy controls aged 4-12 years were included. Serum levels of VEGF, IGF-1, and HIF-1α were measured using commercial enzyme-linked immunosorbent assay kits. Serum IGF-1 levels were found to be statistically significantly higher in the ASD group than in the control group. Serum HIF-1α levels were borderline significantly lower in the ASD group. There was no statistically significant difference in serum VEGF levels between the two groups. IGF-1 and HIF-1α may play a potential role in the etiopathogenesis of ASD.
Collapse
Affiliation(s)
- Fulya Şimşek
- Department of Child and Adolescent Psychiatry, Suleyman Demirel University Medicine Faculty, Çünür, East Campus, Isparta, 32260, Turkey
| | - Ümit Işık
- Department of Child and Adolescent Psychiatry, Suleyman Demirel University Medicine Faculty, Çünür, East Campus, Isparta, 32260, Turkey.
| | - Evrim Aktepe
- Department of Child and Adolescent Psychiatry, Suleyman Demirel University Medicine Faculty, Çünür, East Campus, Isparta, 32260, Turkey
| | - Faruk Kılıç
- Department of Psychiatry, Suleyman Demirel University Medicine Faculty, Isparta, Turkey
| | - Fevziye Burcu Şirin
- Department of Biochemistry, Suleyman Demirel University Medicine Faculty, Isparta, Turkey
| | - Mustafa Bozkurt
- Department of Biochemistry, Suleyman Demirel University Medicine Faculty, Isparta, Turkey
| |
Collapse
|
24
|
Abstract
Neuroanatomic and functional studies show the paraventricular (PVN) of the hypothalamus to have a central role in the autonomic control that supports cardiovascular regulation. Direct and indirect projections from the PVN preautonomic neurons to the sympathetic preganglionic neurons in the spinal cord modulate sympathetic activity. The preautonomic neurons of the PVN adjust their level of activation in response to afferent signals arising from peripheral viscerosensory receptors relayed through the nucleus tractus solitarius. The prevailing sympathetic tone is a balance between excitatory and inhibitory influences that arises from the preautonomic PVN neurons. Under physiologic conditions, tonic sympathetic inhibition driven by a nitric oxide-γ-aminobutyric acid-mediated mechanism is dominant, but in pathologic situation such as heart failure there is a switch from inhibition to sympathoexcitation driven by glutamate and angiotensin II. Angiotensin II, reactive oxygen species, and hypoxia as a result of myocardial infarction/ischemia alter the tightly regulated posttranslational protein-protein interaction of CAPON (carboxy-terminal postsynaptic density protein ligand of neuronal nitric oxide synthase (NOS1)) and PIN (protein inhibitor of NOS1) signaling mechanism. Within the preautonomic neurons of the PVN, the disruption of CAPON and PIN signaling leads to a downregulation of NOS1 expression and reduced NO bioavailability. These data support the notion that CAPON-PIN dysregulation of NO bioavailability is a major contributor to the pathogenesis of sympathoexcitation in heart failure.
Collapse
Affiliation(s)
- Susan Pyner
- Department of Biosciences, Durham University, Durham, United Kingdom.
| |
Collapse
|
25
|
Ye F, Garton HJL, Hua Y, Keep RF, Xi G. The Role of Thrombin in Brain Injury After Hemorrhagic and Ischemic Stroke. Transl Stroke Res 2020; 12:496-511. [PMID: 32989665 DOI: 10.1007/s12975-020-00855-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/22/2020] [Accepted: 09/23/2020] [Indexed: 02/06/2023]
Abstract
Thrombin is increased in the brain after hemorrhagic and ischemic stroke primarily due to the prothrombin entry from blood either with a hemorrhage or following blood-brain barrier disruption. Increasing evidence indicates that thrombin and its receptors (protease-activated receptors (PARs)) play a major role in brain pathology following ischemic and hemorrhagic stroke (including intracerebral, intraventricular, and subarachnoid hemorrhage). Thrombin and PARs affect brain injury via multiple mechanisms that can be detrimental or protective. The cleavage of prothrombin into thrombin is the key step of hemostasis and thrombosis which takes place in every stroke and subsequent brain injury. The extravascular effects and direct cellular interactions of thrombin are mediated by PARs (PAR-1, PAR-3, and PAR-4) and their downstream signaling in multiple brain cell types. Such effects include inducing blood-brain-barrier disruption, brain edema, neuroinflammation, and neuronal death, although low thrombin concentrations can promote cell survival. Also, thrombin directly links the coagulation system to the immune system by activating interleukin-1α. Such effects of thrombin can result in both short-term brain injury and long-term functional deficits, making extravascular thrombin an understudied therapeutic target for stroke. This review examines the role of thrombin and PARs in brain injury following hemorrhagic and ischemic stroke and the potential treatment strategies which are complicated by their role in both hemostasis and brain.
Collapse
Affiliation(s)
- Fenghui Ye
- Department of Neurosurgery, University of Michigan, R5018 Biomedical Science Research Building, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Hugh J L Garton
- Department of Neurosurgery, University of Michigan, R5018 Biomedical Science Research Building, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Ya Hua
- Department of Neurosurgery, University of Michigan, R5018 Biomedical Science Research Building, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, R5018 Biomedical Science Research Building, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Guohua Xi
- Department of Neurosurgery, University of Michigan, R5018 Biomedical Science Research Building, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA.
| |
Collapse
|
26
|
Kusadasi N, Sikma M, Huisman A, Westerink J, Maas C, Schutgens R. A Pathophysiological Perspective on the SARS-CoV-2 Coagulopathy. Hemasphere 2020; 4:e457. [PMID: 32885147 PMCID: PMC7430228 DOI: 10.1097/hs9.0000000000000457] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 06/22/2020] [Indexed: 01/08/2023] Open
Abstract
Recent evidence is focusing on the presence of a hypercoagulable state with development of both venous and arterial thromboembolic complications in patients infected with SARS-CoV-2. The ongoing activation of coagulation related to the severity of the illness is further characterized by thrombotic microangiopathy and endotheliitis. These microangiopathic changes cannot be classified as classical disseminated intravascular coagulation (DIC). In this short review we describe the interaction between coagulation and inflammation with focus on the possible mechanisms that might be involved in SARS-CoV-2 infection associated coagulopathy in the critically ill.
Collapse
Affiliation(s)
- Nuray Kusadasi
- Department of Intensive Care Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Maaike Sikma
- Department of Intensive Care Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
- Dutch Poisons Information Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Albert Huisman
- Central Diagnostic Laboratory, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jan Westerink
- Department of Vascular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Coen Maas
- Central Diagnostic Laboratory, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Roger Schutgens
- Van Creveldkliniek, Benign Hematology Center, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
27
|
Targeting perivascular and epicardial adipose tissue inflammation: therapeutic opportunities for cardiovascular disease. Clin Sci (Lond) 2020; 134:827-851. [PMID: 32271386 DOI: 10.1042/cs20190227] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 03/20/2020] [Accepted: 03/30/2020] [Indexed: 02/07/2023]
Abstract
Major shifts in human lifestyle and dietary habits toward sedentary behavior and refined food intake triggered steep increase in the incidence of metabolic disorders including obesity and Type 2 diabetes. Patients with metabolic disease are at a high risk of cardiovascular complications ranging from microvascular dysfunction to cardiometabolic syndromes including heart failure. Despite significant advances in the standards of care for obese and diabetic patients, current therapeutic approaches are not always successful in averting the accompanying cardiovascular deterioration. There is a strong relationship between adipose inflammation seen in metabolic disorders and detrimental changes in cardiovascular structure and function. The particular importance of epicardial and perivascular adipose pools emerged as main modulators of the physiology or pathology of heart and blood vessels. Here, we review the peculiarities of these two fat depots in terms of their origin, function, and pathological changes during metabolic deterioration. We highlight the rationale for pharmacological targeting of the perivascular and epicardial adipose tissue or associated signaling pathways as potential disease modifying approaches in cardiometabolic syndromes.
Collapse
|
28
|
Yang T, Zelikin AN, Chandrawati R. Enzyme Mimics for the Catalytic Generation of Nitric Oxide from Endogenous Prodrugs. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e1907635. [PMID: 32372556 DOI: 10.1002/smll.201907635] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 02/19/2020] [Indexed: 06/11/2023]
Abstract
The highly diverse biological roles of nitric oxide (NO) in both physiological and pathophysiological processes have prompted great interest in the use of NO as a therapeutic agent in various biomedical applications. NO can exert either protective or deleterious effects depending on its concentration and the location where it is delivered or generated. This double-edged attribute, together with the short half-life of NO in biological systems, poses a major challenge to the realization of the full therapeutic potential of this molecule. Controlled release strategies show an admirable degree of precision with regard to the spatiotemporal dosing of NO but are disadvantaged by the finite NO deliverable payload. In turn, enzyme-prodrug therapy techniques afford enhanced deliverable payload but are troubled by the inherent low stability of natural enzymes, as well as the requirement to control pharmacokinetics for the exogenous prodrugs. The past decade has seen the advent of a new paradigm in controlled delivery of NO, namely localized bioconversion of the endogenous prodrugs of NO, specifically by enzyme mimics. These early developments are presented, successes of this strategy are highlighted, and possible future work on this avenue of research is critically discussed.
Collapse
Affiliation(s)
- Tao Yang
- School of Chemical Engineering and Australian Centre for Nanomedicine (ACN), The University of New South Wales (UNSW Sydney), Sydney, NSW, 2052, Australia
| | - Alexander N Zelikin
- Department of Chemistry and iNANO Interdisciplinary Nanoscience Center, Aarhus University, Aarhus, C 8000, Denmark
| | - Rona Chandrawati
- School of Chemical Engineering and Australian Centre for Nanomedicine (ACN), The University of New South Wales (UNSW Sydney), Sydney, NSW, 2052, Australia
| |
Collapse
|
29
|
Dey A, Prabhudesai S, Zhang Y, Rao G, Thirugnanam K, Hossen MN, Dwivedi SKD, Ramchandran R, Mukherjee P, Bhattacharya R. Cystathione β-synthase regulates HIF-1α stability through persulfidation of PHD2. SCIENCE ADVANCES 2020; 6:eaaz8534. [PMID: 32937467 PMCID: PMC7458453 DOI: 10.1126/sciadv.aaz8534] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 05/20/2020] [Indexed: 05/10/2023]
Abstract
The stringent expression of the hypoxia inducible factor-1α (HIF-1α) is critical to a variety of pathophysiological conditions. We reveal that, in normoxia, enzymatic action of cystathionine β-synthase (CBS) produces H2S, which persulfidates prolyl hydroxylase 2 (PHD2) at residues Cys21 and Cys33 (zinc finger motif), augmenting prolyl hydroxylase activity. Depleting endogenous H2S either by hypoxia or by inhibiting CBS via chemical or genetic means reduces persulfidation of PHD2 and inhibits activity, preventing hydroxylation of HIF-1α, resulting in stabilization. Our in vitro findings are further supported by the depletion of CBS in the zebrafish model that exhibits axis defects and abnormal intersegmental vessels. Exogenous H2S supplementation rescues both in vitro and in vivo phenotypes. We have identified the persulfidated residues and defined their functional significance in regulating the activity of PHD2 via point mutations. Thus, the CBS/H2S/PHD2 axis may provide therapeutic opportunities for pathologies associated with HIF-1α dysregulation in chronic diseases.
Collapse
Affiliation(s)
- Anindya Dey
- Department of Obstetrics and Gynecology, Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Shubhangi Prabhudesai
- Department of Pediatrics, Department of Obstetrics and Gynecology, Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Yushan Zhang
- Department of Pathology, Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Geeta Rao
- Department of Pathology, Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Karthikeyan Thirugnanam
- Department of Pediatrics, Department of Obstetrics and Gynecology, Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Md Nazir Hossen
- Department of Pathology, Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Shailendra Kumar Dhar Dwivedi
- Department of Obstetrics and Gynecology, Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Ramani Ramchandran
- Department of Pediatrics, Department of Obstetrics and Gynecology, Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Priyabrata Mukherjee
- Department of Pathology, Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA.
| | - Resham Bhattacharya
- Department of Obstetrics and Gynecology, Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA.
| |
Collapse
|
30
|
Low-Molecular-Weight Heparin Reduces Ventilation-Induced Lung Injury through Hypoxia Inducible Factor-1α in a Murine Endotoxemia Model. Int J Mol Sci 2020; 21:ijms21093097. [PMID: 32353952 PMCID: PMC7247708 DOI: 10.3390/ijms21093097] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 04/02/2020] [Accepted: 04/27/2020] [Indexed: 12/18/2022] Open
Abstract
Patients with sepsis frequently require mechanical ventilation (MV) to survive. However, MV has been shown to induce the production of proinflammatory cytokines, causing ventilator-induced lung injury (VILI). It has been demonstrated that hypoxia-inducible factor (HIF)-1α plays a crucial role in inducing both apoptotic and inflammatory processes. Low-molecular-weight heparin (LMWH) has been shown to have anti-inflammatory activities. However, the effects of HIF-1α and LMWH on sepsis-related acute lung injury (ALI) have not been fully delineated. We hypothesized that LMWH would reduce lung injury, production of free radicals and epithelial apoptosis through the HIF-1α pathway. Male C57BL/6 mice were exposed to 6-mL/kg or 30-mL/kg MV for 5 h. Enoxaparin, 4 mg/kg, was administered subcutaneously 30 min before MV. We observed that MV with endotoxemia induced microvascular permeability; interleukin-6, tumor necrosis factor-α, macrophage inflammatory protein-2 and vascular endothelial growth factor protein production; neutrophil infiltration; oxidative loads; HIF-1α mRNA activation; HIF-1α expression; bronchial epithelial apoptosis; and decreased respiratory function in mice (p < 0.05). Endotoxin-induced augmentation of VILI and epithelial apoptosis were reduced in the HIF-1α-deficient mice and in the wild-type mice following enoxaparin administration (p < 0.05). Our data suggest that enoxaparin reduces endotoxin-augmented MV-induced ALI, partially by inhibiting the HIF-1α pathway.
Collapse
|
31
|
Kračun D, Klop M, Knirsch A, Petry A, Kanchev I, Chalupsky K, Wolf CM, Görlach A. NADPH oxidases and HIF1 promote cardiac dysfunction and pulmonary hypertension in response to glucocorticoid excess. Redox Biol 2020; 34:101536. [PMID: 32413743 PMCID: PMC7226895 DOI: 10.1016/j.redox.2020.101536] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 03/31/2020] [Accepted: 04/04/2020] [Indexed: 12/22/2022] Open
Abstract
Cardiovascular side effects are frequent problems accompanying systemic glucocorticoid therapy, although the underlying mechanisms are not fully resolved. Reactive oxygen species (ROS) have been shown to promote various cardiovascular diseases although the link between glucocorticoid and ROS signaling has been controversial. As the family of NADPH oxidases has been identified as important source of ROS in the cardiovascular system we investigated the role of NADPH oxidases in response to the synthetic glucocorticoid dexamethasone in the cardiovascular system in vitro and in vivo in mice lacking functional NADPH oxidases due to a mutation in the gene coding for the essential NADPH oxidase subunit p22phox. We show that dexamethasone induced NADPH oxidase-dependent ROS generation, leading to vascular proliferation and angiogenesis due to activation of the transcription factor hypoxia-inducible factor-1 (HIF1). Chronic treatment of mice with low doses of dexamethasone resulted in the development of systemic hypertension, cardiac hypertrophy and left ventricular dysfunction, as well as in pulmonary hypertension and pulmonary vascular remodeling. In contrast, mice deficient in p22phox-dependent NADPH oxidases were protected against these cardiovascular side effects. Mechanistically, dexamethasone failed to upregulate HIF1α levels in these mice, while vascular HIF1α deficiency prevented pulmonary vascular remodeling. Thus, p22phox-dependent NADPH oxidases and activation of the HIF pathway are critical elements in dexamethasone-induced cardiovascular pathologies and might provide interesting targets to limit cardiovascular side effects in patients on chronic glucocorticoid therapy.
Collapse
Affiliation(s)
- Damir Kračun
- Experimental and Molecular Pediatric Cardiology, Department of Pediatric Cardiology and Congenital Heart Diseases, German Heart Center Munich at the Technical University Munich, Munich, 80636, Germany
| | - Mathieu Klop
- Experimental and Molecular Pediatric Cardiology, Department of Pediatric Cardiology and Congenital Heart Diseases, German Heart Center Munich at the Technical University Munich, Munich, 80636, Germany
| | - Anna Knirsch
- Experimental and Molecular Pediatric Cardiology, Department of Pediatric Cardiology and Congenital Heart Diseases, German Heart Center Munich at the Technical University Munich, Munich, 80636, Germany
| | - Andreas Petry
- Experimental and Molecular Pediatric Cardiology, Department of Pediatric Cardiology and Congenital Heart Diseases, German Heart Center Munich at the Technical University Munich, Munich, 80636, Germany
| | - Ivan Kanchev
- Experimental and Molecular Pediatric Cardiology, Department of Pediatric Cardiology and Congenital Heart Diseases, German Heart Center Munich at the Technical University Munich, Munich, 80636, Germany
| | - Karel Chalupsky
- Experimental and Molecular Pediatric Cardiology, Department of Pediatric Cardiology and Congenital Heart Diseases, German Heart Center Munich at the Technical University Munich, Munich, 80636, Germany; Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the ASCR, v. v. i., Prague, Czech Republic
| | - Cordula M Wolf
- Department of Pediatric Cardiology and Congenital Heart Diseases, German Heart Center Munich at the Technical University Munich, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Agnes Görlach
- Experimental and Molecular Pediatric Cardiology, Department of Pediatric Cardiology and Congenital Heart Diseases, German Heart Center Munich at the Technical University Munich, Munich, 80636, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.
| |
Collapse
|
32
|
Gutmann C, Siow R, Gwozdz AM, Saha P, Smith A. Reactive Oxygen Species in Venous Thrombosis. Int J Mol Sci 2020; 21:E1918. [PMID: 32168908 PMCID: PMC7139897 DOI: 10.3390/ijms21061918] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/02/2020] [Accepted: 03/04/2020] [Indexed: 01/03/2023] Open
Abstract
Reactive oxygen species (ROS) have physiological roles as second messengers, but can also exert detrimental modifications on DNA, proteins and lipids if resulting from enhanced generation or reduced antioxidant defense (oxidative stress). Venous thrombus (DVT) formation and resolution are influenced by ROS through modulation of the coagulation, fibrinolysis, proteolysis and the complement system, as well as the regulation of effector cells such as platelets, endothelial cells, erythrocytes, neutrophils, mast cells, monocytes and fibroblasts. Many conditions that carry an elevated risk of venous thrombosis, such as the Antiphospholipid Syndrome, have alterations in their redox homeostasis. Dietary and pharmacological antioxidants can modulate several important processes involved in DVT formation, but their overall effect is unknown and there are no recommendations regarding their use. The development of novel antioxidant treatments that aim to abrogate the formation of DVT or promote its resolution will depend on the identification of targets that enable ROS modulation confined to their site of interest in order to prevent off-target effects on physiological redox mechanisms. Subgroups of patients with increased systemic oxidative stress might benefit from unspecific antioxidant treatment, but more clinical studies are needed to bring clarity to this issue.
Collapse
Affiliation(s)
- Clemens Gutmann
- King’s British Heart Foundation Centre, King’s College London, 125 Coldharbour Lane, London SE5 9NU, UK;
| | - Richard Siow
- Vascular Biology & Inflammation Section, School of Cardiovascular Medicine & Sciences, British Heart Foundation of Research Excellence, King’s College London, SE1 9NH, UK;
| | - Adam M. Gwozdz
- Academic Department of Surgery, School of Cardiovascular Medicine & Sciences, British Heart Foundation of Research Excellence, King’s College London, London SE1 7EH, UK; (A.M.G.); (P.S.)
| | - Prakash Saha
- Academic Department of Surgery, School of Cardiovascular Medicine & Sciences, British Heart Foundation of Research Excellence, King’s College London, London SE1 7EH, UK; (A.M.G.); (P.S.)
| | - Alberto Smith
- Academic Department of Surgery, School of Cardiovascular Medicine & Sciences, British Heart Foundation of Research Excellence, King’s College London, London SE1 7EH, UK; (A.M.G.); (P.S.)
| |
Collapse
|
33
|
Knock GA. NADPH oxidase in the vasculature: Expression, regulation and signalling pathways; role in normal cardiovascular physiology and its dysregulation in hypertension. Free Radic Biol Med 2019; 145:385-427. [PMID: 31585207 DOI: 10.1016/j.freeradbiomed.2019.09.029] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/29/2019] [Accepted: 09/23/2019] [Indexed: 02/06/2023]
Abstract
The last 20-25 years have seen an explosion of interest in the role of NADPH oxidase (NOX) in cardiovascular function and disease. In vascular smooth muscle and endothelium, NOX generates reactive oxygen species (ROS) that act as second messengers, contributing to the control of normal vascular function. NOX activity is altered in response to a variety of stimuli, including G-protein coupled receptor agonists, growth-factors, perfusion pressure, flow and hypoxia. NOX-derived ROS are involved in smooth muscle constriction, endothelium-dependent relaxation and smooth muscle growth, proliferation and migration, thus contributing to the fine-tuning of blood flow, arterial wall thickness and vascular resistance. Through reversible oxidative modification of target proteins, ROS regulate the activity of protein tyrosine phosphatases, kinases, G proteins, ion channels, cytoskeletal proteins and transcription factors. There is now considerable, but somewhat contradictory evidence that NOX contributes to the pathogenesis of hypertension through oxidative stress. Specific NOX isoforms have been implicated in endothelial dysfunction, hyper-contractility and vascular remodelling in various animal models of hypertension, pulmonary hypertension and pulmonary arterial hypertension, but also have potential protective effects, particularly NOX4. This review explores the multiplicity of NOX function in the healthy vasculature and the evidence for and against targeting NOX for antihypertensive therapy.
Collapse
Affiliation(s)
- Greg A Knock
- Dpt. of Inflammation Biology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King's College London, UK.
| |
Collapse
|
34
|
Chiu H, Lee H, Lee K, Zhao Y, Hsu CY, Shyu W. Mechanisms of ischaemic neural progenitor proliferation: a regulatory role of the HIF‐1α‐CBX7 pathway. Neuropathol Appl Neurobiol 2019; 46:391-405. [PMID: 31630421 DOI: 10.1111/nan.12585] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 10/08/2019] [Indexed: 01/14/2023]
Affiliation(s)
- H.‐Y. Chiu
- Children’s Hospital China Medical University and Hospital Taichung Taiwan
| | - H.‐T. Lee
- Department of Neurosurgery Taichung Veterans General Hospital Taichung Taiwan
- Graduate Institute of Medical Sciences National Defense Medical Center Taipei Taiwan
| | - K.‐H. Lee
- Eshelman School of Pharmacy University of North Carolina Chapel Hill NC USA
| | - Y. Zhao
- Eshelman School of Pharmacy University of North Carolina Chapel Hill NC USA
| | - C. Y. Hsu
- Graduate Institute of Biomedical Science and Drug Development Center China Medical University Taichung Taiwan
| | - W.C. Shyu
- Graduate Institute of Biomedical Science and Drug Development Center China Medical University Taichung Taiwan
- Translational Medicine Research Center China Medical University & Hospital Taichung Taiwan
- Department of Neurology China Medical University & Hospital Taichung Taiwan
- Department of Occupational Therapy Asia University Taichung Taiwan
| |
Collapse
|
35
|
Rafikova O, Al Ghouleh I, Rafikov R. Focus on Early Events: Pathogenesis of Pulmonary Arterial Hypertension Development. Antioxid Redox Signal 2019; 31:933-953. [PMID: 31169021 PMCID: PMC6765063 DOI: 10.1089/ars.2018.7673] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 06/03/2019] [Accepted: 06/03/2019] [Indexed: 12/17/2022]
Abstract
Significance: Pulmonary arterial hypertension (PAH) is a progressive disease of the lung vasculature characterized by the proliferation of all vascular wall cell types, including endothelial, smooth muscle, and fibroblasts. The disease rapidly advances into a form with extensive pulmonary vascular remodeling, leading to a rapid increase in pulmonary vascular resistance, which results in right heart failure. Recent Advances: Most current research in the PAH field has been focused on the late stage of the disease, largely due to an urgent need for patient treatment options in clinics. Further, the pathobiology of PAH is multifaceted in the advanced disease, and there has been promising recent progress in identifying various pathological pathways related to the late clinical picture. Critical Issues: Early stage PAH still requires additional attention from the scientific community, and although the survival of patients with early diagnosis is comparatively higher, the disease develops in patients asymptomatically, making it difficult to identify and treat early. Future Directions: There are several reasons to focus on the early stage of PAH. First, the complexity of late stage disease, owing to multiple pathways being activated in a complex system with intra- and intercellular signaling, leads to an unclear picture of the key contributors to the pathobiology. Second, an understanding of early pathophysiological events can increase the ability to identify PAH patients earlier than what is currently possible. Third, the prompt diagnosis of PAH would allow for the therapy to start earlier, which has proved to be a more successful strategy, and it ensures better survival in PAH patients.
Collapse
Affiliation(s)
- Olga Rafikova
- Division of Endocrinology, Department of Medicine, University of Arizona, Tucson, Arizona
| | - Imad Al Ghouleh
- Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ruslan Rafikov
- Division of Endocrinology, Department of Medicine, University of Arizona, Tucson, Arizona
| |
Collapse
|
36
|
Sadaghianloo N, Contenti J, Dardik A, Mazure NM. Role of Hypoxia and Metabolism in the Development of Neointimal Hyperplasia in Arteriovenous Fistulas. Int J Mol Sci 2019; 20:ijms20215387. [PMID: 31671790 PMCID: PMC6862436 DOI: 10.3390/ijms20215387] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 10/24/2019] [Accepted: 10/25/2019] [Indexed: 12/11/2022] Open
Abstract
For patients with end-stage renal disease requiring hemodialysis, their vascular access is both their lifeline and their Achilles heel. Despite being recommended as primary vascular access, the arteriovenous fistula (AVF) shows sub-optimal results, with about 50% of patients needing a revision during the year following creation. After the AVF is created, the venous wall must adapt to new environment. While hemodynamic changes are responsible for the adaptation of the extracellular matrix and activation of the endothelium, surgical dissection and mobilization of the vein disrupt the vasa vasorum, causing wall ischemia and oxidative stress. As a consequence, migration and proliferation of vascular cells participate in venous wall thickening by a mechanism of neointimal hyperplasia (NH). When aggressive, NH causes stenosis and AVF dysfunction. In this review we show how hypoxia, metabolism, and flow parameters are intricate mechanisms responsible for the development of NH and stenosis during AVF maturation.
Collapse
Affiliation(s)
- Nirvana Sadaghianloo
- Centre de Méditerranéen de Médecine Moléculaire (C3M), Université Côte d'Azur, INSERM U1065, 151 Route de St Antoine de Ginestière, BP2 3194, 06204 Nice CEDEX 03, France.
- Department of Vascular Surgery, Centre Hospitalier Universitaire de Nice, 06000 Nice, France.
| | - Julie Contenti
- Centre de Méditerranéen de Médecine Moléculaire (C3M), Université Côte d'Azur, INSERM U1065, 151 Route de St Antoine de Ginestière, BP2 3194, 06204 Nice CEDEX 03, France.
- Department of Emergency Medicine, Centre Hospitalier Universitaire de Nice, 06000 Nice, France.
| | - Alan Dardik
- Department of Surgery and the Vascular Biology and Therapeutics Program, Yale University, New Haven, CT 06520, USA.
- Department of Surgery, VA Connecticut Healthcare Systems, West Haven, CT 06516, USA.
| | - Nathalie M Mazure
- Centre de Méditerranéen de Médecine Moléculaire (C3M), Université Côte d'Azur, INSERM U1065, 151 Route de St Antoine de Ginestière, BP2 3194, 06204 Nice CEDEX 03, France.
- Department of Vascular Surgery, Centre Hospitalier Universitaire de Nice, 06000 Nice, France.
| |
Collapse
|
37
|
Mohamed R, Janke R, Guo W, Cao Y, Zhou Y, Zheng W, Babaahmadi-Rezaei H, Xu S, Kamato D, Little PJ. GPCR transactivation signalling in vascular smooth muscle cells: role of NADPH oxidases and reactive oxygen species. VASCULAR BIOLOGY (BRISTOL, ENGLAND) 2019; 1:R1-R11. [PMID: 32923966 PMCID: PMC7439842 DOI: 10.1530/vb-18-0004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Accepted: 07/23/2019] [Indexed: 02/02/2023]
Abstract
The discovery and extension of G-protein-coupled receptor (GPCR) transactivation-dependent signalling has enormously broadened the GPCR signalling paradigm. GPCRs can transactivate protein tyrosine kinase receptors (PTKRs) and serine/threonine kinase receptors (S/TKRs), notably the epidermal growth factor receptor (EGFR) and transforming growth factor-β type 1 receptor (TGFBR1), respectively. Initial comprehensive mechanistic studies suggest that these two transactivation pathways are distinct. Currently, there is a focus on GPCR inhibitors as drug targets, and they have proven to be efficacious in vascular diseases. With the broadening of GPCR transactivation signalling, it is therefore important from a therapeutic perspective to find a common transactivation pathway of EGFR and TGFBR1 that can be targeted to inhibit complex pathologies activated by the combined action of these receptors. Reactive oxygen species (ROS) are highly reactive molecules and they act as second messengers, thus modulating cellular signal transduction pathways. ROS are involved in different mechanisms of GPCR transactivation of EGFR. However, the role of ROS in GPCR transactivation of TGFBR1 has not yet been studied. In this review, we will discuss the involvement of ROS in GPCR transactivation-dependent signalling.
Collapse
Affiliation(s)
- Raafat Mohamed
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Queensland, Australia
- Department of Basic Sciences, College of Dentistry, University of Mosul, Mosul, Iraq
| | - Reearna Janke
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Wanru Guo
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Yingnan Cao
- Department of Pharmacy, Xinhua College of Sun Yat-sen University, Guangzhou, China
| | - Ying Zhou
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Wenhua Zheng
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Hossein Babaahmadi-Rezaei
- Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Atherosclerosis Research Center, Ahvaz, Iran
| | - Suowen Xu
- Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Danielle Kamato
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Queensland, Australia
- Department of Pharmacy, Xinhua College of Sun Yat-sen University, Guangzhou, China
| | - Peter J Little
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Queensland, Australia
- Department of Pharmacy, Xinhua College of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
38
|
Schoos A, Gabriel C, Knab VM, Fux DA. Activation of HIF-1 α by δ-Opioid Receptors Induces COX-2 Expression in Breast Cancer Cells and Leads to Paracrine Activation of Vascular Endothelial Cells. J Pharmacol Exp Ther 2019; 370:480-489. [PMID: 31300611 DOI: 10.1124/jpet.119.257501] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 06/24/2019] [Indexed: 01/05/2023] Open
Abstract
Opioids promote tumor angiogenesis in mammary malignancies, but the underlying signaling mechanism is largely unknown. The current study investigated the hypothesis that stimulation of δ-opioid receptors (DOR) in breast cancer (BCa) cells activates the hypoxia-inducible factor 1α (HIF-1α), which triggers synthesis and release of diverse angiogenic factors. Immunoblotting revealed that incubation of human MCF-7 and T47D breast cancer cells with the DOR agonist d-Ala2,d-Leu5-enkephalin (DADLE) resulted in a transient accumulation and thus activation of HIF-1α DADLE-induced HIF-1α activation preceded PI3K/Akt stimulation and was blocked by the DOR antagonist naltrindole and naloxone, pertussis toxin, different phosphoinositide 3-kinase (PI3K) inhibitors, and the Akt inhibitor Akti-1/2. Whereas DADLE exposure had no effect on the expression and secretion of vascular endothelial growth factor (VEGF) in BCa cells, an increased abundance of cyclooxygenase-2 (COX-2) and release of prostaglandin E2 (PGE2) was detected. DADLE-induced COX-2 expression was also observed in three-dimensional cultured MCF-7 cells and impaired by PI3K/Akt inhibitors and the HIF-1α inhibitor echinomycin. Supernatant from DADLE-treated MCF-7 cells triggered sprouting of endothelial (END) cells, which was blocked when MCF-7 cells were pretreated with echinomycin or the COX-2 inhibitor celecoxib. Also no sprouting was observed when END cells were exposed to the PGE2 receptor antagonist PF-04418948. The findings together indicate that DOR stimulation in BCa cells leads to PI3K/Akt-dependent HIF-1α activation and COX-2 expression, which trigger END cell sprouting by paracrine activation of PGE2 receptors. These findings provide a potential mechanism of opioid-driven tumor angiogenesis and thus therapeutic targets to combat the tumor-angiogenic opioid effect. SIGNIFICANCE STATEMENT: Opioids are indispensable analgesics for treating cancer-related pain. However, opioids were found to promote tumor growth and metastasis, which questions the use of these potent pain-relieving drugs in cancer patients. Enhanced tumor vascularization after opioid treatment implies that tumor progression results from angiogenic opioid effects. Thus, understanding the signaling mechanism of opioid-driven tumor angiogenesis helps to identify therapeutic targets to combat these undesired tumor effects. The present study reveals that stimulation of δ-opioid receptors in breast cancer cells leads to an activation of HIF-1α and expression of COX-2 via PI3K/Akt stimulation, which results in a paracrine activation of vascular endothelial cells by prostaglandin E2 receptors.
Collapse
Affiliation(s)
- Alexandra Schoos
- Division Clinical Pharmacology, Institute of Pharmacology and Toxicology (A.S., V.M.K., D.A.F.) and Institute of Pathology and Forensic Veterinary Medicine (C.G.), University of Veterinary Medicine Vienna, Vienna, Austria
| | - Cordula Gabriel
- Division Clinical Pharmacology, Institute of Pharmacology and Toxicology (A.S., V.M.K., D.A.F.) and Institute of Pathology and Forensic Veterinary Medicine (C.G.), University of Veterinary Medicine Vienna, Vienna, Austria
| | - Vanessa M Knab
- Division Clinical Pharmacology, Institute of Pharmacology and Toxicology (A.S., V.M.K., D.A.F.) and Institute of Pathology and Forensic Veterinary Medicine (C.G.), University of Veterinary Medicine Vienna, Vienna, Austria
| | - Daniela A Fux
- Division Clinical Pharmacology, Institute of Pharmacology and Toxicology (A.S., V.M.K., D.A.F.) and Institute of Pathology and Forensic Veterinary Medicine (C.G.), University of Veterinary Medicine Vienna, Vienna, Austria
| |
Collapse
|
39
|
Chaurasia SN, Kushwaha G, Kulkarni PP, Mallick RL, Latheef NA, Mishra JK, Dash D. Platelet HIF-2α promotes thrombogenicity through PAI-1 synthesis and extracellular vesicle release. Haematologica 2019; 104:2482-2492. [PMID: 31004026 PMCID: PMC6959171 DOI: 10.3324/haematol.2019.217463] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 04/17/2019] [Indexed: 01/03/2023] Open
Abstract
Oxygen-compromised environments, such as high altitude, are associated with platelet hyperactivity. Platelets confined within the relatively impervious core of an aggregate/thrombus have restricted access to oxygen, yet they continue to perform energy-intensive procoagulant activities that sustain the thrombus. Studying platelet signaling under hypoxia is, therefore, critical to our understanding of the mechanistic basis of thrombus stability. We report here that hypoxia-inducible factor (HIF)-2α is translated from pre-existing mRNA and stabilized against proteolytic degradation in enucleate platelets exposed to hypoxia. Hypoxic stress, too, stimulates platelets to synthesize plasminogen-activator inhibitor-1 (PAI-1) and shed extracellular vesicles, both of which potentially contribute to the prothrombotic phenotype associated with hypoxia. Stabilization of HIF-α by administering hypoxia-mimetics to mice accelerates thrombus formation in mesenteric arterioles. In agreement, platelets from patients with chronic obstructive pulmonary disease and high altitude residents exhibiting thrombogenic attributes have abundant expression of HIF-2α and PAI- 1. Thus, targeting platelet hypoxia signaling could be an effective anti-thrombotic strategy.
Collapse
Affiliation(s)
- Susheel N Chaurasia
- Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Geeta Kushwaha
- Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Paresh P Kulkarni
- Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Ram L Mallick
- Department of Biochemistry, Birat Medical College & Teaching Hospital, Biratnagar, Nepal
| | - Nazmy A Latheef
- Department of Tuberculosis & Respiratory Diseases, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Jai K Mishra
- Department of Tuberculosis & Respiratory Diseases, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Debabrata Dash
- Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| |
Collapse
|
40
|
Yamaguchi R, Sakamoto A, Yamaguchi R, Haraguchi M, Narahara S, Sugiuchi H, Katoh T, Yamaguchi Y. Di-(2-Ethylhexyl) Phthalate Promotes Release of Tissue Factor-Bearing Microparticles From Macrophages via the TGFβ1/Smad/PAI-1 Signaling Pathway. Am J Med Sci 2019; 357:492-506. [PMID: 30910165 DOI: 10.1016/j.amjms.2019.02.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 01/26/2019] [Accepted: 02/07/2019] [Indexed: 01/03/2023]
Abstract
BACKGROUND Plasminogen activator inhibitor type 1 promotes formation of endothelial microparticles with procoagulant activity. However, it remains unclear whether di-(2-ethylhexyl) phthalate, a peroxisome proliferator-activated receptor α agonist, influences microparticle formation. MATERIALS AND METHODS The effect of di-(2-ethylhexyl) phthalate on release of tissue factor-bearing microparticles was investigated using human M1 macrophages. RESULTS Exposure of M1 macrophages to di-(2-ethylhexyl) phthalate significantly upregulated expression of plasminogen activator inhibitor type 1, whereas incubation of macrophages with small interfering RNA for peroxisome proliferator-activated receptor α attenuated it. Di-(2-ethylhexyl) phthalate significantly increased the tissue factor protein level in culture supernatants of M1 macrophages, but not M2 macrophages. After purification of proteins by centrifugal filtration, western blotting detected 2 high molecular weight bands of tissue factor-bearing microparticles in culture supernatants of M1 macrophages. The upper band showed binding to factor VIIa and tissue factor pathway inhibitor, unlike the lower band. This suggested heterogeneity of the procoagulant activity of tissue factor-bearing microparticles, presumably dependent upon encryption/decryption of tissue factor. Phosphatidylserine contributes to tissue factor decryption, and western blotting revealed that the density of phosphatidylserine was reduced in the upper tissue factor band compared with the lower band. Di-(2-ethylhexyl) phthalate also upregulated transforming growth factor-β1 protein production by M1 macrophages. Moreover, silencing of Smad2, Smad3 or Smad4 attenuated plasminogen activator inhibitor type 1 expression and tissue factor-release from macrophages after di-(2-ethylhexyl) phthalate stimulation. CONCLUSIONS Di-(2-ethylhexyl) phthalate promotes formation of tissue factor-bearing microparticles in human M1 macrophages via the transforming growth factor-β1/Smad/ plasminogen activator inhibitor type 1 signaling pathway.
Collapse
Affiliation(s)
- Rui Yamaguchi
- Department of Public Health, Faculty of Life Sciences, Kumamoto University School of Medicine, Kumamoto, Japan; Graduate School of Medical Science, Kumamoto Health Science University, Kumamoto, Japan
| | - Arisa Sakamoto
- Graduate School of Medical Science, Kumamoto Health Science University, Kumamoto, Japan
| | - Reona Yamaguchi
- Department of Neuroscience, Graduate School of Medicine and Faculty of Medicine, Kyoto University, Kyoto, Japan
| | - Misa Haraguchi
- Graduate School of Medical Science, Kumamoto Health Science University, Kumamoto, Japan
| | - Shinji Narahara
- Graduate School of Medical Science, Kumamoto Health Science University, Kumamoto, Japan
| | - Hiroyuki Sugiuchi
- Graduate School of Medical Science, Kumamoto Health Science University, Kumamoto, Japan
| | - Takahiko Katoh
- Department of Public Health, Faculty of Life Sciences, Kumamoto University School of Medicine, Kumamoto, Japan
| | - Yasuo Yamaguchi
- Graduate School of Medical Science, Kumamoto Health Science University, Kumamoto, Japan.
| |
Collapse
|
41
|
Xia J, Ozaki I, Matsuhashi S, Kuwashiro T, Takahashi H, Anzai K, Mizuta T. Mechanisms of PKC-Mediated Enhancement of HIF-1α Activity and its Inhibition by Vitamin K2 in Hepatocellular Carcinoma Cells. Int J Mol Sci 2019; 20:ijms20051022. [PMID: 30813635 PMCID: PMC6429062 DOI: 10.3390/ijms20051022] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 02/20/2019] [Accepted: 02/21/2019] [Indexed: 01/27/2023] Open
Abstract
Hypoxia-inducible factor 1 (HIF-1) plays important roles in cancer cell biology. HIF-1α is reportedly activated by several factors, including protein kinase C (PKC), in addition to hypoxia. We investigated the role of PKC isoforms and the effects of vitamin K2 (VK2) in the activation process of HIF-1α. Human hepatocellular carcinoma (HCC)-derived Huh7 cells were cultured under normoxic and hypoxic (1% O₂) conditions with or without the PKC stimulator TPA. The expression, transcriptional activity and nuclear translocation of HIF-1α were examined under treatment with PKC inhibitors, siRNAs against each PKC isoform and VK2. Hypoxia increased the expression and activity of HIF-1α. TPA increased the HIF-1α activity several times under both normoxic and hypoxic conditions. PKC-δ siRNA-mediated knockdown, PKC-δ inhibitor (rottlerin) and pan-PKC inhibitor (Ro-31-8425) suppressed the expression and transcriptional activity of HIF-1α. VK2 significantly inhibited the TPA-induced HIF-1α transcriptional activity and suppressed the expression and nuclear translocation of HIF-1α induced by TPA without altering the HIF-1α mRNA levels. These data indicate that PKC-δ enhances the HIF-1α transcriptional activity by increasing the nuclear translocation, and that VK2 might suppress the HIF-1α activation through the inhibition of PKC in HCC cells.
Collapse
Affiliation(s)
- Jinghe Xia
- Department of Internal Medicine, Saga Medical School, Saga University, 5-1-1 Nabeshima, Saga 849-8501, Japan.
| | - Iwata Ozaki
- Department of Internal Medicine, Saga Medical School, Saga University, 5-1-1 Nabeshima, Saga 849-8501, Japan.
- Health Administration Center, Saga Medical School, Saga University, 5-1-1 Nabeshima, Saga 849-8501, Japan.
| | - Sachiko Matsuhashi
- Department of Internal Medicine, Saga Medical School, Saga University, 5-1-1 Nabeshima, Saga 849-8501, Japan.
| | - Takuya Kuwashiro
- Department of Internal Medicine, Saga Medical School, Saga University, 5-1-1 Nabeshima, Saga 849-8501, Japan.
| | - Hirokazu Takahashi
- Department of Internal Medicine, Saga Medical School, Saga University, 5-1-1 Nabeshima, Saga 849-8501, Japan.
| | - Keizo Anzai
- Department of Internal Medicine, Saga Medical School, Saga University, 5-1-1 Nabeshima, Saga 849-8501, Japan.
| | - Toshihiko Mizuta
- Department of Internal Medicine, Fujikawa Hospital, 1-2-6 Matsubara, Saga 840-0831, Japan.
| |
Collapse
|
42
|
Bai Y, Gong X, Dou C, Cao Z, Dong S. Redox control of chondrocyte differentiation and chondrogenesis. Free Radic Biol Med 2019; 132:83-89. [PMID: 30394290 DOI: 10.1016/j.freeradbiomed.2018.10.443] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 10/14/2018] [Accepted: 10/26/2018] [Indexed: 11/24/2022]
Abstract
Chondrogenesis involves the recruitment and migration of mesenchymal cells, mesenchymal condensation, and chondrocyte differentiation and hypertrophy. Multiple factors precisely regulate chondrogenesis. Recent studies have demonstrated that the redox status of chondrocytes plays an essential role in the regulation of chondrocyte differentiation and chondrogenesis. Reactive oxygen species (ROS) and reactive nitrogen species (RNS) are important factors that change the intracellular redox status. Physiological levels of ROS/RNS act as intracellular signals in chondrocytes, and oxidative stress impairs the metabolism of chondrocytes. Under physiological conditions, the balance between ROS/RNS production and elimination ensures that redox-sensitive signalling proteins function correctly. The redox homeostasis of chondrocytes ensures that they respond appropriately to endogenous and exogenous stimuli. This review focuses on the redox regulation of key signalling pathways and transcription factors that control chondrogenesis and chondrocyte differentiation. Additionally, the mechanism by which ROS/RNS regulate signalling proteins and transcription factors in chondrocytes is also reviewed.
Collapse
Affiliation(s)
- Yun Bai
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Gaotanyan Street No.30, Chongqing 400038, China
| | - Xiaoshan Gong
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Gaotanyan Street No.30, Chongqing 400038, China
| | - Ce Dou
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Gaotanyan Street No.30, Chongqing 400038, China
| | - Zhen Cao
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Gaotanyan Street No.30, Chongqing 400038, China
| | - Shiwu Dong
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Gaotanyan Street No.30, Chongqing 400038, China; State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University, Chongqing 400038, China.
| |
Collapse
|
43
|
Abstract
SIGNIFICANCE G protein-coupled receptors (GPCR) are the largest group of cell surface receptors, which link cells to their environment. Reactive oxygen species (ROS) can act as important cellular signaling molecules. The family of NADPH oxidases generates ROS in response to activated cell surface receptors. Recent Advances: Various signaling pathways linking GPCRs and activation of NADPH oxidases have been characterized. CRITICAL ISSUES Still, a more detailed analysis of G proteins involved in the GPCR-mediated activation of NADPH oxidases is needed. In addition, a more precise discrimination of NADPH oxidase activation due to either upregulation of subunit expression or post-translational subunit modifications is needed. Also, the role of noncanonical modulators of NADPH oxidase activation in the response to GPCRs awaits further analyses. FUTURE DIRECTIONS As GPCRs are one of the most popular classes of investigational drug targets, further detailing of G protein-coupled mechanisms in the activation mechanism of NADPH oxidases as well as better understanding of the link between newly identified NADPH oxidase interaction partners and GPCR signaling will provide new opportunities for improved efficiency and decreased off target effects of therapies targeting GPCRs.
Collapse
Affiliation(s)
- Andreas Petry
- 1 Experimental and Molecular Pediatric Cardiology, German Heart Center Munich , TU Munich, Munich, Germany
| | - Agnes Görlach
- 1 Experimental and Molecular Pediatric Cardiology, German Heart Center Munich , TU Munich, Munich, Germany .,2 DZHK (German Centre for Cardiovascular Research) , Partner Site Munich, Munich Heart Alliance, Munich, Germany
| |
Collapse
|
44
|
Zhang Z, Trautz B, Kračun D, Vogel F, Weitnauer M, Hochkogler K, Petry A, Görlach A. Stabilization of p22phox by Hypoxia Promotes Pulmonary Hypertension. Antioxid Redox Signal 2019; 30:56-73. [PMID: 30044141 DOI: 10.1089/ars.2017.7482] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
AIMS Hypoxia and reactive oxygen species (ROS) have been shown to play a role in the pathogenesis of pulmonary hypertension (PH), a potentially fatal disorder characterized by pulmonary vascular remodeling, elevated pulmonary arterial pressure, and right ventricular hypertrophy. However, how they are linked in the context of PH is not completely understood. We, therefore, investigated the role of the NADPH oxidase subunit p22phox in the response to hypoxia both in vitro and in vivo. RESULTS We found that hypoxia decreased ubiquitinylation and proteasomal degradation of p22phox dependent on prolyl hydroxylases (PHDs) and the E3 ubiquitin ligase protein von Hippel Lindau (pVHL), which resulted in p22phox stabilization and accumulation. p22phox promoted vascular proliferation, migration, and angiogenesis under normoxia and hypoxia. Increased levels of p22phox were also detected in lungs and hearts from mice with hypoxia-induced PH. Mice harboring a point mutation (Y121H) in the p22phox gene, which resulted in decreased p22phox stability and subsequent loss of this protein, were protected against hypoxia-induced PH. Mechanistically, p22phox contributed to ROS generation under normoxia, hypoxia, and hypoxia/reoxygenation. p22phox increased the levels and activity of HIF1α, the major cellular regulator of hypoxia adaptation, under normoxia and hypoxia, possibly by decreasing the levels of the PHD cofactors ascorbate and iron(II), and it contributed to the downregulation of the tumor suppressor miR-140 by hypoxia. INNOVATION These data identify p22phox as an important regulator of the hypoxia response both in vitro and in vivo. CONCLUSION p22phox-dependent NADPH oxidases contribute to the pathophysiology of PH induced by hypoxia.
Collapse
Affiliation(s)
- Zuwen Zhang
- 1 Experimental and Molecular Pediatric Cardiology, German Heart Center Munich at the Technical University Munich, Munich, Germany
| | - Benjamin Trautz
- 1 Experimental and Molecular Pediatric Cardiology, German Heart Center Munich at the Technical University Munich, Munich, Germany
| | - Damir Kračun
- 1 Experimental and Molecular Pediatric Cardiology, German Heart Center Munich at the Technical University Munich, Munich, Germany
| | - Frederick Vogel
- 1 Experimental and Molecular Pediatric Cardiology, German Heart Center Munich at the Technical University Munich, Munich, Germany
| | - Michael Weitnauer
- 1 Experimental and Molecular Pediatric Cardiology, German Heart Center Munich at the Technical University Munich, Munich, Germany
| | - Katharina Hochkogler
- 1 Experimental and Molecular Pediatric Cardiology, German Heart Center Munich at the Technical University Munich, Munich, Germany .,2 DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance , Munich, Germany
| | - Andreas Petry
- 1 Experimental and Molecular Pediatric Cardiology, German Heart Center Munich at the Technical University Munich, Munich, Germany
| | - Agnes Görlach
- 1 Experimental and Molecular Pediatric Cardiology, German Heart Center Munich at the Technical University Munich, Munich, Germany .,2 DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance , Munich, Germany
| |
Collapse
|
45
|
Carvalho C, Moreira PI. Oxidative Stress: A Major Player in Cerebrovascular Alterations Associated to Neurodegenerative Events. Front Physiol 2018; 9:806. [PMID: 30018565 PMCID: PMC6037979 DOI: 10.3389/fphys.2018.00806] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 06/08/2018] [Indexed: 12/19/2022] Open
Abstract
The brain is one of the most exquisite organs in the body with high metabolic demands, and requires a tight regulation of the surrounding environment. This tight control is exerted by the neurovascular unit (NVU) comprising different cell types, where endothelial cells play the commander-in-chief role. Thus, it is assumable that even slight perturbations in NVU might affect, in some cases irreversibly, brain homeostasis and health. In this line, recent findings support the two-hit vascular hypothesis for neurodegenerative conditions, where vascular dysfunction underlies the development of neurodegenerative diseases, such as Alzheimer’s disease (AD). Knowing that endothelial cells are rich in mitochondria and nicotinamide adenine dinucleotide phosphate (NADPH) oxidases, two major reactive oxygen species (ROS) sources, this review aims to gather information on how oxidative stress is in the front line of vascular alterations observed in brain aging and neurodegenerative conditions, particularly AD. Also, a brief discussion about the therapeutic strategies aimed to protect against cerebrovascular diseases is included.
Collapse
Affiliation(s)
- Cristina Carvalho
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Paula I Moreira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Laboratory of Physiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
46
|
Pulmonary Arterial Hypertension and Endothelial Dysfunction Is Linked to NADPH Oxidase-Derived Superoxide Formation in Venous Thrombosis and Pulmonary Embolism in Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:1860513. [PMID: 29983855 PMCID: PMC6015670 DOI: 10.1155/2018/1860513] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 02/11/2018] [Accepted: 02/28/2018] [Indexed: 12/30/2022]
Abstract
Pulmonary embolism (PE) results from deep vein thrombosis (DVT) and can lead to chronic thromboembolic pulmonary hypertension (CTEPH) involving vascular dysfunction. Mechanisms are incompletely understood, in part due to lack of mouse models. We induced PE in C57BL/6 mice by intravenous injection of thrombin (166 U/kg BW), confirmed by a sudden bradycardia, bradypnea, and an increase in pulmonary artery (PA) pressure observed by high-frequency ultrasound. While symptoms resolved rapidly after single thrombin application, repeated PEs resulted in sustained PA-pressure increase, increased PA superoxide formation assessed by oxidative fluorescent microtopography, increased PA gp91phox expression, and endothelial dysfunction assessed by isometric tension studies of isolated PA segments after 24 hours. DVT was modeled in C57BL/6 mice by ligation of the inferior vena cava (IVC). Importantly, small pulmonary emboli could be detected along with a mild phenotype of PA endothelial dysfunction and oxidative stress in the absence of PA-pressure elevation. mRNA expression of plasminogen activator inhibitor-1 was increased in PAs of mice with recurrent PE after repetitive thrombin injections and to a lesser extent in DVT mice. In summary, our data suggest that PA endothelial dysfunction, induced by gp91phox-derived ROS, is an early event upon repetitive PE. This phenomenon might help to elucidate the mechanisms of PA dysfunction in the pathogenesis of CTEPH.
Collapse
|
47
|
AlAbdi L, He M, Yang Q, Norvil AB, Gowher H. The transcription factor Vezf1 represses the expression of the antiangiogenic factor Cited2 in endothelial cells. J Biol Chem 2018; 293:11109-11118. [PMID: 29794136 PMCID: PMC6052231 DOI: 10.1074/jbc.ra118.002911] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 05/19/2018] [Indexed: 01/05/2023] Open
Abstract
Formation of the vasculature by angiogenesis is critical for proper development, but angiogenesis also contributes to the pathogenesis of various disorders, including cancer and cardiovascular diseases. Vascular endothelial zinc finger 1 (Vezf1), is a Krüppel-like zinc finger protein that plays a vital role in vascular development. However, the mechanism by which Vezf1 regulates this process is not fully understood. Here, we show that Vezf1−/− mouse embryonic stem cells (ESC) have significantly increased expression of a stem cell factor, Cbp/p300-interacting transactivator 2 (Cited2). Compared with WT ESCs, Vezf1−/− ESCs inefficiently differentiated into endothelial cells (ECs), which exhibited defects in the tube-formation assay. These defects were due to reduced activation of EC-specific genes concomitant with lower enrichment of histone 3 acetylation at Lys27 (H3K27) at their promoters. We hypothesized that overexpression of Cited2 in Vezf1−/− cells sequesters P300/CBP away from the promoters of proangiogenic genes and thereby contributes to defective angiogenesis in these cells. This idea was supported by the observation that shRNA-mediated depletion of Cited2 significantly reduces the angiogenic defects in the Vezf1−/− ECs. In contrast to previous studies that have focused on the role of Vezf1 as a transcriptional activator of proangiogenic genes, our findings have revealed a role for Vezf1 in modulating the expression of the antiangiogenic factor Cited2. Vezf1 previously has been characterized as an insulator protein, and our results now provide insights into the mechanism, indicating that Vezf1 can block inappropriate, nonspecific interactions of promoters with cis-located enhancers, preventing aberrant promoter activation.
Collapse
Affiliation(s)
| | - Ming He
- From the Department of Biochemistry and
| | | | | | - Humaira Gowher
- From the Department of Biochemistry and .,Purdue University Center for Cancer Research, Purdue University, West Lafayette, Indiana 47907
| |
Collapse
|
48
|
Heikal L, Ghezzi P, Mengozzi M, Ferns G. Assessment of HIF-1α expression and release following endothelial injury in-vitro and in-vivo. Mol Med 2018; 24:22. [PMID: 30134815 PMCID: PMC6016879 DOI: 10.1186/s10020-018-0026-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 05/07/2018] [Indexed: 11/10/2022] Open
Abstract
Background Endothelial injury is an early and enduring feature of cardiovascular disease. Inflammation and hypoxia may be responsible for this, and are often associated with the up-regulation of several transcriptional factors that include Hypoxia Inducible Factor-1 (HIF-1). Although it has been reported that HIF-1α is detectable in plasma, it is known to be unstable. Our aim was to optimize an assay for HIF-1α to be applied to in vitro and in vivo applications, and to use this assay to assess the release kinetics of HIF-1α following endothelial injury. Methods An ELISA for the measurement of HIF-1α in cell-culture medium and plasma was optimized, and the assay was used to determine the best conditions for sample collection and storage. The results of the ELISA were validated using Western blotting and immunohistochemistry (IHC). In vitro, a standardized injury was produced in a monolayer of rat aortic endothelial cells (RAECs) and intracellular HIF-1α was measured at intervals over 24 h. In vivo, a rat angioplasty model was used. The right carotid artery was injured using a 2F Fogarty balloon catheter. HIF-1α was measured in the plasma and in the arterial tissue (0, 1, 2, 3 and 5 days post injury). Results The HIF-1α ELISA had a limit of detection of 2.7 pg/mL and was linear up to 1000 pg/ mL. Between and within-assay, the coefficient of variation values were less than 15%. HIF-1α was unstable in cell lysates and plasma, and it was necessary to add a protease inhibitor immediately after collection, and to store samples at -80 °C prior to analysis. The dynamics of HIF-1α release were different for the in vitro and in vivo models. In vitro, HIF-1α reached maximum concentrations approximately 2 h post injury, whereas peak values in plasma and tissues occurred approximately 2 days post injury, in the balloon injury model. Conclusion HIF-1α can be measured in plasma, but this requires careful sample collection and storage. The carotid artery balloon injury model is associated with the transient release of HIF-1α into the circulation that probably reflects the hypoxia induced in the artery wall. Electronic supplementary material The online version of this article (10.1186/s10020-018-0026-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lamia Heikal
- Brighton and Sussex Medical School Department of Clinical and experimental investigation, University of Sussex, Falmer East Sussex, Brighton, BN1 9PS, UK
| | - Pietro Ghezzi
- Brighton and Sussex Medical School Department of Clinical and experimental investigation, University of Sussex, Falmer East Sussex, Brighton, BN1 9PS, UK
| | - Manuela Mengozzi
- Brighton and Sussex Medical School Department of Clinical and experimental investigation, University of Sussex, Falmer East Sussex, Brighton, BN1 9PS, UK
| | - Gordon Ferns
- Brighton and Sussex Medical School Department of Clinical and experimental investigation, University of Sussex, Falmer East Sussex, Brighton, BN1 9PS, UK. .,Brighton and Sussex Medical School Department of Medical Education, Mayfield House, Falmer East Sussex, Brighton, BN1 9PH, UK.
| |
Collapse
|
49
|
Kossmann S, Lagrange J, Jäckel S, Jurk K, Ehlken M, Schönfelder T, Weihert Y, Knorr M, Brandt M, Xia N, Li H, Daiber A, Oelze M, Reinhardt C, Lackner K, Gruber A, Monia B, Karbach SH, Walter U, Ruggeri ZM, Renné T, Ruf W, Münzel T, Wenzel P. Platelet-localized FXI promotes a vascular coagulation-inflammatory circuit in arterial hypertension. Sci Transl Med 2018; 9:9/375/eaah4923. [PMID: 28148841 DOI: 10.1126/scitranslmed.aah4923] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 07/05/2016] [Accepted: 12/12/2016] [Indexed: 01/05/2023]
Abstract
Multicellular interactions of platelets, leukocytes, and the blood vessel wall support coagulation and precipitate arterial and venous thrombosis. High levels of angiotensin II cause arterial hypertension by a complex vascular inflammatory pathway that requires leukocyte recruitment and reactive oxygen species production and is followed by vascular dysfunction. We delineate a previously undescribed, proinflammatory coagulation-vascular circuit that is a major regulator of vascular tone, blood pressure, and endothelial function. In mice with angiotensin II-induced hypertension, tissue factor was up-regulated, as was thrombin-dependent endothelial cell vascular cellular adhesion molecule 1 expression and integrin αMβ2- and platelet-dependent leukocyte adhesion to arterial vessels. The resulting vascular inflammation and dysfunction was mediated by activation of thrombin-driven factor XI (FXI) feedback, independent of factor XII. The FXI receptor glycoprotein Ibα on platelets was required for this thrombin feedback activation in angiotensin II-infused mice. Inhibition of FXI synthesis with an antisense oligonucleotide was sufficient to prevent thrombin propagation on platelets, vascular leukocyte infiltration, angiotensin II-induced endothelial dysfunction, and arterial hypertension in mice and rats. Antisense oligonucleotide against FXI also reduced the increased blood pressure and attenuated vascular and kidney dysfunction in rats with established arterial hypertension. Further, platelet-localized thrombin generation was amplified in an FXI-dependent manner in patients with uncontrolled arterial hypertension, suggesting that platelet-localized thrombin generation may serve as an inflammatory marker of high blood pressure. Our results outline a coagulation-inflammation circuit that promotes vascular dysfunction, and highlight the possible utility of FXI-targeted anticoagulants in treating hypertension, beyond their application as antithrombotic agents in cardiovascular disease.
Collapse
Affiliation(s)
- Sabine Kossmann
- Center for Thrombosis and Hemostasis Mainz, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany.,Center for Cardiology, Cardiology I, University Medical Center Mainz, 55131 Mainz, Germany
| | - Jeremy Lagrange
- Center for Thrombosis and Hemostasis Mainz, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Sven Jäckel
- Center for Thrombosis and Hemostasis Mainz, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Kerstin Jurk
- Center for Thrombosis and Hemostasis Mainz, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Moritz Ehlken
- Center for Thrombosis and Hemostasis Mainz, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany.,Center for Cardiology, Cardiology I, University Medical Center Mainz, 55131 Mainz, Germany
| | - Tanja Schönfelder
- Center for Thrombosis and Hemostasis Mainz, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Yvonne Weihert
- Center for Cardiology, Cardiology I, University Medical Center Mainz, 55131 Mainz, Germany
| | - Maike Knorr
- Center for Cardiology, Cardiology I, University Medical Center Mainz, 55131 Mainz, Germany
| | - Moritz Brandt
- Center for Cardiology, Cardiology I, University Medical Center Mainz, 55131 Mainz, Germany
| | - Ning Xia
- Department of Pharmacology, University Medical Center Mainz, 55131 Mainz, Germany
| | - Huige Li
- Department of Pharmacology, University Medical Center Mainz, 55131 Mainz, Germany
| | - Andreas Daiber
- Center for Cardiology, Cardiology I, University Medical Center Mainz, 55131 Mainz, Germany
| | - Matthias Oelze
- Center for Cardiology, Cardiology I, University Medical Center Mainz, 55131 Mainz, Germany
| | - Christoph Reinhardt
- Center for Thrombosis and Hemostasis Mainz, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Karl Lackner
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Mainz, 55131 Mainz, Germany
| | - Andras Gruber
- Department of Biomedical Engineering, Oregon Health and Science University, 3303 Southwest Bond Avenue, CH13B, Portland, OR 97239, USA.,Aronora Inc., 4640 Southwest Macadam Avenue, Suite 200A, Portland, OR 97239, USA
| | - Brett Monia
- Ionis Pharmaceuticals Inc., 2855 Gazelle Court, Carlsbad, CA 92010, USA
| | - Susanne H Karbach
- Center for Cardiology, Cardiology I, University Medical Center Mainz, 55131 Mainz, Germany
| | - Ulrich Walter
- Center for Thrombosis and Hemostasis Mainz, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Zaverio M Ruggeri
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Thomas Renné
- Department of Molecular Medicine and Surgery, L1:00, Karolinska Institutet, SE-171 71 Stockholm, Sweden.,Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Wolfram Ruf
- Center for Thrombosis and Hemostasis Mainz, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany.,Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037, USA.,DZHK (German Center for Cardiovascular Research), Partner Site Rhine Main, University Medical Center Mainz, 55131 Mainz, Germany
| | - Thomas Münzel
- Center for Cardiology, Cardiology I, University Medical Center Mainz, 55131 Mainz, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Rhine Main, University Medical Center Mainz, 55131 Mainz, Germany
| | - Philip Wenzel
- Center for Thrombosis and Hemostasis Mainz, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany. .,Center for Cardiology, Cardiology I, University Medical Center Mainz, 55131 Mainz, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Rhine Main, University Medical Center Mainz, 55131 Mainz, Germany
| |
Collapse
|
50
|
Sharma NM, Cunningham CJ, Zheng H, Liu X, Patel KP. Hypoxia-Inducible Factor-1α Mediates Increased Sympathoexcitation via Glutamatergic N-Methyl-d-Aspartate Receptors in the Paraventricular Nucleus of Rats With Chronic Heart Failure. Circ Heart Fail 2017; 9:CIRCHEARTFAILURE.116.003423. [PMID: 27810863 DOI: 10.1161/circheartfailure.116.003423] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 10/13/2016] [Indexed: 12/30/2022]
Abstract
BACKGROUND Increased sympathetic outflow is a major contributor to the progression of chronic heart failure (CHF). Potentiation of glutamatergic tone has been causally related to the sympathoexcitation in CHF. Specifically, an increase in the N-methyl-d-aspartate-type 1 receptor (NMDA-NR1) expression within the paraventricular nucleus (PVN) is critically linked to the increased sympathoexcitation during CHF. However, the molecular mechanism(s) for the upregulation of NMDA-NR1 remains unexplored. We hypothesized that hypoxia via hypoxia-inducible factor 1α (HIF-1α) might contribute to the augmentation of the NMDA-NR1-mediated sympathoexcitatory responses from the PVN in CHF. METHODS AND RESULTS Immunohistochemistry staining, mRNA, and protein for hypoxia-inducible factor 1α were upregulated within the PVN of left coronary artery-ligated CHF rats. In neuronal cell line (NG108-15) in vitro, hypoxia caused a significant increase in mRNA and protein for HIF-1α (2-fold) with the concomitant increase in NMDA-NR1 mRNA, protein levels, and glutamate-induced Ca+ influx. Chromatin immunoprecipitation assay identified HIF-1α binding to NMDA-NR1 promoter during hypoxia. Silencing of HIF-1α in NG108 cells leads to a significant decrease in expression of NMDA-NR1, suggesting that expression of HIF-1α is necessary for the upregulation of NMDA-NR1. Consistent with these observations, HIF-1α silencing within the PVN abrogated the increased basal sympathetic tone and sympathoexcitatory responses to microinjection of NMDA in the PVN of rats with CHF. CONCLUSIONS These results uncover a critical role for HIF-1 in the upregulation of NMDA-NR1 to mediate sympathoexcitation in CHF. We conclude that subtle hypoxia within the PVN may act as a metabolic cue to modulate sympathoexcitation during CHF.
Collapse
Affiliation(s)
- Neeru M Sharma
- From the Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha
| | - Craig J Cunningham
- From the Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha
| | - Hong Zheng
- From the Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha
| | - Xuefei Liu
- From the Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha
| | - Kaushik P Patel
- From the Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha.
| |
Collapse
|