1
|
Lin TK, Huang CR, Lin KJ, Hsieh YH, Chen SD, Lin YC, Chao AC, Yang DI. Potential Roles of Hypoxia-Inducible Factor-1 in Alzheimer's Disease: Beneficial or Detrimental? Antioxidants (Basel) 2024; 13:1378. [PMID: 39594520 PMCID: PMC11591038 DOI: 10.3390/antiox13111378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/04/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
The major pathological characteristics of Alzheimer's disease (AD) include senile plaques and neurofibrillary tangles (NFTs), which are mainly composed of aggregated amyloid-beta (Aβ) peptide and hyperphosphorylated tau protein, respectively. The excessive production of reactive oxygen species (ROS) and neuroinflammation are crucial contributing factors to the pathological mechanisms of AD. Hypoxia-inducible factor-1 (HIF-1) is a transcription factor critical for tissue adaption to low-oxygen tension. Growing evidence has suggested HIF-1 as a potential therapeutic target for AD; conversely, other experimental findings indicate that HIF-1 induction contributes to AD pathogenesis. These previous findings thus point to the complex, even contradictory, roles of HIF-1 in AD. In this review, we first introduce the general pathogenic mechanisms of AD as well as the potential pathophysiological roles of HIF-1 in cancer, immunity, and oxidative stress. Based on current experimental evidence in the literature, we then discuss the possible beneficial as well as detrimental mechanisms of HIF-1 in AD; these sections also include the summaries of multiple chemical reagents and proteins that have been shown to exert beneficial effects in AD via either the induction or inhibition of HIF-1.
Collapse
Affiliation(s)
- Tsu-Kung Lin
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833401, Taiwan; (T.-K.L.); (C.-R.H.); (S.-D.C.)
- College of Medicine, Chang Gung University, Taoyuan 333323, Taiwan
| | - Chi-Ren Huang
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833401, Taiwan; (T.-K.L.); (C.-R.H.); (S.-D.C.)
- College of Medicine, Chang Gung University, Taoyuan 333323, Taiwan
| | - Kai-Jung Lin
- Department of Family Medicine, National Taiwan University Hospital, Taipei 100225, Taiwan;
| | - Yi-Heng Hsieh
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan;
| | - Shang-Der Chen
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833401, Taiwan; (T.-K.L.); (C.-R.H.); (S.-D.C.)
| | - Yi-Chun Lin
- Department of Neurology, Taipei City Hospital Renai Branch, Taipei 106243, Taiwan;
| | - A-Ching Chao
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung 807377, Taiwan
- Department of Neurology, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
- Department of Sports Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
| | - Ding-I Yang
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan;
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| |
Collapse
|
2
|
He C, Li Q, Wu W, Liu K, Li X, Zheng H, Lai Y. Ferroptosis-associated genes and compounds in renal cell carcinoma. Front Immunol 2024; 15:1473203. [PMID: 39399506 PMCID: PMC11466770 DOI: 10.3389/fimmu.2024.1473203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 09/09/2024] [Indexed: 10/15/2024] Open
Abstract
As the main type of renal cell carcinoma (RCC), clear cell RCC (ccRCC) is often associated with the deletion or mutation of the von Hippel Lindau (VHL) gene, enhancement of glucose and lipid metabolism, and heterogeneity of the tumor microenvironment. VHL alterations in RCC cells lead to the activation of hypoxia-inducible factors and their downstream target vascular endothelial growth factor, and to the reprogramming of multiple cell death pathways and metabolic weakness, including ferroptosis, which are associated with targeted therapy or immunotherapy. The changes in biological metabolites (e.g., iron and lipids) support ferroptosis as a potential therapeutic strategy for RCC, while iron metabolism and ferroptosis regulation have been examined as anti-RCC agents in numerous studies, and various ferroptosis-related molecules have been shown to be related to the metastasis and prognosis of ccRCC. For example, glutathione peroxidase 4 and glutaminase inhibitors can inhibit pyrimidine synthesis and increase reactive oxygen species levels in VHL-deficient RCC cells. In addition, the release of damage-associated molecular patterns by tumor cells undergoing ferroptosis also mediates antitumor immunity, and immune therapy can synergize with targeted therapy or radiotherapy through ferroptosis. However, Inducing ferroptosis not only suppresses cancer, but also promotes cancer development due to its potential negative effects on anti-cancer immunity. Therefore, ferroptosis and various tumor microenviroment-related molecules may co-occur during the development and treatment of RCC, and further understanding of the interactions, core targets, and related drugs of ferroptosis may provide new combination drug strategies for RCC treatment. Here we summarize the key genes and compounds on ferroptosis and RCC in order to envision future treatment strategies and to provide sufficient information for overcoming RCC resistance through ferroptosis.
Collapse
Affiliation(s)
- Chengwu He
- Department of Urology, Shenzhen Shockwave Lithotripsy Research Institute, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Qingyi Li
- Department of Urology, Shenzhen Shockwave Lithotripsy Research Institute, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Weijia Wu
- Department of Urology, Shenzhen Shockwave Lithotripsy Research Institute, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Ke Liu
- Department of Urology, Shenzhen Shockwave Lithotripsy Research Institute, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Xingwen Li
- Tibet Future Biomedicine Company Limited, Golmud, Qinghai, China
| | - Hanxiong Zheng
- Department of Urology, Shenzhen Shockwave Lithotripsy Research Institute, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Yongchang Lai
- Department of Pharmaceutical Management, School of Medical Business, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| |
Collapse
|
3
|
Ahmed M, Semreen AM, Giddey AD, Ramadan WS, El-Awady R, Soares NC, El-Huneidi W, Bustanji Y, Alqudah MAY, Alzoubi KH, Semreen MH. Proteomic and metabolomic signatures of U87 glioblastoma cells treated with cisplatin and/or paclitaxel. Ann Med 2024; 55:2305308. [PMID: 38253025 PMCID: PMC10810643 DOI: 10.1080/07853890.2024.2305308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 12/19/2023] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND Glioblastoma (GBM) is a primary malignancy of the central nervous system and is classified as a grade IV astrocytoma by the World Health Organization (WHO). Although GBM rarely metastasizes, its prognosis remains poor. Moreover, the standard treatment for GBM, temozolomide (TMZ), is associated with chemoresistance, which is a major factor behind GBM-related deaths. Investigating drugs with repurposing potential in the context of GBM is worthwhile to bypass lengthy bench-to-bedside research. The field of omics has garnered significant interest in scientific research because of its potential to delineate the intricate regulatory network underlying tumor development. In particular, proteomic and metabolomic analyses are powerful approaches for the investigation of metabolic enzymes and intermediate metabolites since they represent the functional end of the cancer phenotype. METHODS We chose two of the most widely prescribed anticancer drugs, cisplatin and paclitaxel. To our knowledge, the current literature lacks studies examining their effects on metabolic and proteomic alterations in GBM. We employed the mass spectrometry technological platform 'UHPLC-Q-TOF-MS/MS' to examine the changes in the proteome and metabolome profiles of the U87 cell line with defined concentrations of cisplatin and/or paclitaxel via an untargeted approach. RESULTS A total of 1,419 distinct proteins and 90 metabolites were generated, and subsequent analysis was performed. We observed that upon treatment with cisplatin (9.5 μM), U87 cells exhibited apparent efforts to cope with this exogenous stressor, understanding the effect of paclitaxel (5.3 μM) on altering the transport machinery of the cell, and how the combination of cisplatin and/or paclitaxel suggests potential interactions with promising benefits in GBM therapeutics. CONCLUSION Our research provides a detailed map of alterations in response to cisplatin and paclitaxel treatment, provides crucial insights into the molecular basis of their action, and paves the way for further research to identify molecular targets for this elusive malignancy.
Collapse
Affiliation(s)
- Munazza Ahmed
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Ahlam M. Semreen
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Alexander D. Giddey
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Wafaa S. Ramadan
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Raafat El-Awady
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Nelson C. Soares
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
- Laboratory of Proteomics, Department of Human Genetics, National Institute of Health Doutor Ricardo Jorge (INSA), Lisbon, Portugal
| | - Waseem El-Huneidi
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Yasser Bustanji
- Department of Basic and Clinical Pharmacology, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- School of Pharmacy, The University of Jordan, Amman, Jordan
| | - Mohammad A. Y. Alqudah
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Karem H. Alzoubi
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Mohammad H. Semreen
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
4
|
Burtscher J, Mallet RT, Pialoux V, Millet GP, Burtscher M. Adaptive Responses to Hypoxia and/or Hyperoxia in Humans. Antioxid Redox Signal 2022; 37:887-912. [PMID: 35102747 DOI: 10.1089/ars.2021.0280] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Significance: Oxygen is indispensable for aerobic life, but its utilization exposes cells and tissues to oxidative stress; thus, tight regulation of cellular, tissue, and systemic oxygen concentrations is crucial. Here, we review the current understanding of how the human organism (mal-)adapts to low (hypoxia) and high (hyperoxia) oxygen levels and how these adaptations may be harnessed as therapeutic or performance enhancing strategies at the systemic level. Recent Advances: Hyperbaric oxygen therapy is already a cornerstone of modern medicine, and the application of mild hypoxia, that is, hypoxia conditioning (HC), to strengthen the resilience of organs or the whole body to severe hypoxic insults is an important preparation for high-altitude sojourns or to protect the cardiovascular system from hypoxic/ischemic damage. Many other applications of adaptations to hypo- and/or hyperoxia are only just emerging. HC-sometimes in combination with hyperoxic interventions-is gaining traction for the treatment of chronic diseases, including numerous neurological disorders, and for performance enhancement. Critical Issues: The dose- and intensity-dependent effects of varying oxygen concentrations render hypoxia- and/or hyperoxia-based interventions potentially highly beneficial, yet hazardous, although the risks versus benefits are as yet ill-defined. Future Directions: The field of low and high oxygen conditioning is expanding rapidly, and novel applications are increasingly recognized, for example, the modulation of aging processes, mood disorders, or metabolic diseases. To advance hypoxia/hyperoxia conditioning to clinical applications, more research on the effects of the intensity, duration, and frequency of altered oxygen concentrations, as well as on individual vulnerabilities to such interventions, is paramount. Antioxid. Redox Signal. 37, 887-912.
Collapse
Affiliation(s)
- Johannes Burtscher
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland.,Institute of Sport Sciences, University of Lausanne, Lausanne, Switzerland
| | - Robert T Mallet
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Vincent Pialoux
- Inter-University Laboratory of Human Movement Biology EA7424, University Claude Bernard Lyon 1, University of Lyon, Lyon, France
| | - Grégoire P Millet
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland.,Institute of Sport Sciences, University of Lausanne, Lausanne, Switzerland
| | - Martin Burtscher
- Department of Sport Science, University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
5
|
Ahmad IM, Dafferner AJ, O’Connell KA, Mehla K, Britigan BE, Hollingsworth MA, Abdalla MY. Heme Oxygenase-1 Inhibition Potentiates the Effects of Nab-Paclitaxel-Gemcitabine and Modulates the Tumor Microenvironment in Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2021; 13:2264. [PMID: 34066839 PMCID: PMC8125955 DOI: 10.3390/cancers13092264] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/22/2021] [Accepted: 05/03/2021] [Indexed: 12/27/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy with a poor prognosis. Tumor hypoxia plays an active role in promoting tumor progression, malignancy, and resistance to therapy in PDAC. We present evidence that nab-paclitaxel-gemcitabine (NPG) and/or a hypoxic tumor microenvironment (TME) up-regulate heme oxygenase-1 (HO-1), providing a survival advantage for tumors. Using PDAC cells in vitro and a PDAC mouse model, we found that NPG chemotherapy up-regulated expression of HO-1 in PDAC cells and increased its nuclear translocation. Inhibition of HO-1 with ZnPP and SnPP sensitized PDAC cells to NPG-induced cytotoxicity (p < 0.05) and increased apoptosis (p < 0.05). Additionally, HO-1 expression was increased in gemcitabine-resistant PDAC cells (p < 0.05), and HO-1 inhibition increased GEM-resistant PDAC sensitivity to NPG (p < 0.05). NPG combined with HO-1 inhibitor inhibited tumor size in an orthotopic model. In parallel, HO-1 inhibition abrogated the influx of macrophages and FoxP3+ cells, while increasing the proportion of CD8+ infiltration in the pancreatic tumors. These effects were mediated primarily by reducing expression of the immunosuppressive cytokine IL-10.
Collapse
Affiliation(s)
- Iman M. Ahmad
- College of Allied Health Professions, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Alicia J. Dafferner
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Kelly A. O’Connell
- Fred & Pamela Buffett Cancer Center, The Eppley Institute for Research in Cancer, University of Nebraska Medical Center, Omaha, NE 68198, USA; (K.A.O.); (K.M.); (M.A.H.)
| | - Kamiya Mehla
- Fred & Pamela Buffett Cancer Center, The Eppley Institute for Research in Cancer, University of Nebraska Medical Center, Omaha, NE 68198, USA; (K.A.O.); (K.M.); (M.A.H.)
| | - Bradley E. Britigan
- Veterans Affairs Medical Center-Nebraska Western Iowa, Department of Internal Medicine and Research Service, Omaha, NE 68105, USA;
| | - Michael A. Hollingsworth
- Fred & Pamela Buffett Cancer Center, The Eppley Institute for Research in Cancer, University of Nebraska Medical Center, Omaha, NE 68198, USA; (K.A.O.); (K.M.); (M.A.H.)
| | - Maher Y. Abdalla
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| |
Collapse
|
6
|
Costa DL, Amaral EP, Andrade BB, Sher A. Modulation of Inflammation and Immune Responses by Heme Oxygenase-1: Implications for Infection with Intracellular Pathogens. Antioxidants (Basel) 2020; 9:antiox9121205. [PMID: 33266044 PMCID: PMC7761188 DOI: 10.3390/antiox9121205] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/25/2020] [Accepted: 11/26/2020] [Indexed: 02/07/2023] Open
Abstract
Heme oxygenase-1 (HO-1) catalyzes the degradation of heme molecules releasing equimolar amounts of biliverdin, iron and carbon monoxide. Its expression is induced in response to stress signals such as reactive oxygen species and inflammatory mediators with antioxidant, anti-inflammatory and immunosuppressive consequences for the host. Interestingly, several intracellular pathogens responsible for major human diseases have been shown to be powerful inducers of HO-1 expression in both host cells and in vivo. Studies have shown that this HO-1 response can be either host detrimental by impairing pathogen control or host beneficial by limiting infection induced inflammation and tissue pathology. These properties make HO-1 an attractive target for host-directed therapy (HDT) of the diseases in question, many of which have been difficult to control using conventional antibiotic approaches. Here we review the mechanisms by which HO-1 expression is induced and how the enzyme regulates inflammatory and immune responses during infection with a number of different intracellular bacterial and protozoan pathogens highlighting mechanistic commonalities and differences with the goal of identifying targets for disease intervention.
Collapse
Affiliation(s)
- Diego L. Costa
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto 14049-900, São Paulo, Brazil
- Correspondence: ; Tel.: +55-16-3315-3061
| | - Eduardo P. Amaral
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; (E.P.A.); (A.S.)
| | - Bruno B. Andrade
- Wellcome Centre for Infectious Disease Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7925, South Africa;
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador 40296-710, Bahia, Brazil
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador 40210-320, Bahia, Brazil
- Curso de Medicina, Faculdade de Tecnologia e Ciências (UniFTC), Salvador 41741-590, Bahia, Brazil
- Curso de Medicina, Universidade Salvador (UNIFACS), Laureate International Universities, Salvador 41770-235, Bahia, Brazil
- Escola Bahiana de Medicina e Saúde Pública (EBMSP), Salvador 40290-000, Bahia, Brazil
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Alan Sher
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; (E.P.A.); (A.S.)
| |
Collapse
|
7
|
Dery KJ, Nakamura K, Kadono K, Hirao H, Kageyama S, Ito T, Kojima H, Kaldas FM, Busuttil RW, Kupiec‐Weglinski JW. Human Antigen R (HuR): A Regulator of Heme Oxygenase-1 Cytoprotection in Mouse and Human Liver Transplant Injury. Hepatology 2020; 72:1056-1072. [PMID: 31879990 PMCID: PMC8330638 DOI: 10.1002/hep.31093] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 12/10/2019] [Indexed: 01/16/2023]
Abstract
BACKGROUND AND AIMS Ischemia-reperfusion injury (IRI) represents a risk factor in liver transplantation (LT). We have shown that overexpression of heme oxygenase-1 (HO-1) mitigates hepatic IRI in LT recipients. Here, we hypothesized that human antigen R (HuR), the stabilizer of adenylate-uridylate (AU)-rich mRNAs, is required for hepatoprotection in LT. APPROACH AND RESULTS In an experimental arm, HuR/HO-1 protein expression was correlated with hepatic IRI phenotype. In an in vitro inflammation mimic model of hepatic warm IRI, induction of HuR/HO-1 and cytoplasmic localization following cytokine preconditioning were detected in primary hepatocyte cultures, whereas HuR silencing caused negative regulation of HO-1, followed by enhanced cytotoxicity. Using the HuR-inhibitor, we showed that HuR likely regulates HO-1 through its 3' untranslated region and causes neutrophil activation (CD69+/lymphocyte antigen 6 complex locus G [Ly6-G]). HuR silencing in bone marrow-derived macrophages decreased HO-1 expression, leading to the induction of proinflammatory cytokines/chemokines. RNA sequencing of HuR silenced transcripts under in vitro warm IRI revealed regulation of genes thymus cell antigen 1 (THY1), aconitate decarboxylase 1 (ACOD1), and Prostaglandin E Synthase (PTGES). HuR, but not hypoxia-inducible protein alpha, positively regulated HO-1 in warm, but not cold, hypoxia/reoxygenation conditions. HuR modulated HO-1 in primary hepatocytes, neutrophils, and macrophages under reperfusion. Adjunctive inhibition of HuR diminished microtubule-associated proteins 1A/1B light chain 3B (LC3B), a marker for autophagosome, under HO-1 regulation, suggesting a cytoprotective mechanism in hepatic IR. In a clinical arm, hepatic biopsies from 51 patients with LT were analyzed at 2 hours after reperfusion. Graft HuR expression was negatively correlated with macrophage (CD80/CD86) and neutrophil (Cathepsin G) markers. Hepatic IRI increased HuR/HO-1 expression and inflammatory genes. High HuR-expressing liver grafts showed lower serum alanine aminotransferase/serum aspartate aminotransferase levels and improved LT survival. CONCLUSIONS This translational study identifies HuR as a regulator of HO-1-mediated cytoprotection in sterile liver inflammation and a biomarker of ischemic stress resistance in LT.
Collapse
Affiliation(s)
- Kenneth J. Dery
- The Dumont‐UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas TransplantationDavid Geffen School of Medicine at UCLALos AngelesCA
| | - Kojiro Nakamura
- The Dumont‐UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas TransplantationDavid Geffen School of Medicine at UCLALos AngelesCA,Department of SurgeryKyoto UniversityKyotoJapan,Department of SurgeryNishi‐Kobe Medical CenterKobeJapan
| | - Kentaro Kadono
- The Dumont‐UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas TransplantationDavid Geffen School of Medicine at UCLALos AngelesCA
| | - Hirofumi Hirao
- The Dumont‐UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas TransplantationDavid Geffen School of Medicine at UCLALos AngelesCA
| | - Shoichi Kageyama
- The Dumont‐UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas TransplantationDavid Geffen School of Medicine at UCLALos AngelesCA
| | - Takahiro Ito
- The Dumont‐UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas TransplantationDavid Geffen School of Medicine at UCLALos AngelesCA
| | - Hidenobu Kojima
- The Dumont‐UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas TransplantationDavid Geffen School of Medicine at UCLALos AngelesCA
| | - Fady M. Kaldas
- The Dumont‐UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas TransplantationDavid Geffen School of Medicine at UCLALos AngelesCA
| | - Ronald W. Busuttil
- The Dumont‐UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas TransplantationDavid Geffen School of Medicine at UCLALos AngelesCA
| | - Jerzy W. Kupiec‐Weglinski
- The Dumont‐UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas TransplantationDavid Geffen School of Medicine at UCLALos AngelesCA
| |
Collapse
|
8
|
Halder A, Yadav K, Aggarwal A, Singhal N, Sandhir R. Activation of TNFR1 and TLR4 following oxygen glucose deprivation promotes mitochondrial fission in C6 astroglial cells. Cell Signal 2020; 75:109714. [PMID: 32693013 DOI: 10.1016/j.cellsig.2020.109714] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 06/30/2020] [Accepted: 07/12/2020] [Indexed: 12/14/2022]
Abstract
Astrocytes have emerged as active players in the innate immune response triggered by various types of insults. Recent literature suggests that mitochondria are key participants in innate immunity. The present study investigates the role of ischemia-induced innate immune response on p65/PGC-1α mediated mitochondrial dynamics in C6 astroglial cells. OGD conditions induced astroglial differentiation in C6 cells and increased the expression of hypoxia markers; HIF-1α, HO-1 and Cox4i2. OGD conditions resulted in induction of innate immune response in terms of expression of TNFR1 and TLR4 along with increase in IL-6 and TNF-α levels. OGD conditions resulted in decreased expression of I-κB with a concomitant increase in phos-p65 levels. The expression of PGC-1α, a key regulator of mitochondrial biogenesis, was also increased. Immunochemical staining suggested that phos-p65 and PGC-1α was co-localized. Studies on mitochondrial fusion (Mfn-1) and fission (DRP1) markers revealed shift toward fission. In addition, mitochondrial membrane potential decreased with increased DNA degradation and apoptosis confirming mitochondrial fission under OGD conditions. However, inhibition of phos-p65 by MG132 reduced the co-localization of phos-p65/ PGC-1α and significantly increased the Mfn-1 expression. The findings demonstrate the involvement of TNFR1 and TLR4 mediated immune response followed by interaction between phos-p65 and PGC-1α in promoting fission in C6 cells under hypoxic condition.
Collapse
Affiliation(s)
- Avishek Halder
- Department of Biochemistry, Basic Medical Science Block II, Panjab University, Chandigarh, India
| | - Kamalendra Yadav
- National Agri-Food Biotechnology Institute, Sector 81, Mohali, Punjab, India
| | - Aanchal Aggarwal
- National Agri-Food Biotechnology Institute, Sector 81, Mohali, Punjab, India
| | - Nitin Singhal
- National Agri-Food Biotechnology Institute, Sector 81, Mohali, Punjab, India
| | - Rajat Sandhir
- Department of Biochemistry, Basic Medical Science Block II, Panjab University, Chandigarh, India.
| |
Collapse
|
9
|
Rashid I, Baisvar VS, Singh M, Kumar R, Srivastava P, Kushwaha B, Pathak AK. Isolation and characterization of hypoxia inducible heme oxygenase 1 (HMOX1) gene in Labeo rohita. Genomics 2020; 112:2327-2333. [PMID: 31923615 DOI: 10.1016/j.ygeno.2020.01.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 11/05/2019] [Accepted: 01/06/2020] [Indexed: 11/29/2022]
Abstract
The HMOX1 gene plays role in several biological processes and is also responsive to hypoxia stress. Freshwater carp fish, Labeo rohita, is reported as hypoxia sensitive, but the information of annotated hypoxia genes in public domain is very scanty for this species. Here, an attempt was made to isolate and characterize HMOX1 gene in L. rohita using information from zebrafish. HMOX1 gene was obtained by mapping HMOX1 protein of zebrafish over assembled genome of L. rohita. Aligned region was used for designing primers for HMOX1 amplification. Eight overlapping sets of primers were designed for amplifying ~540 bp long successive overlapping fragments. Splicing of overlapping amplicons generated 3715 bp fragment that was confirmed as HMOX1 gene having full coding region with 6 exons between 184 and 2156 bp positions. HMOX1 characterization is an initiative for L. rohita genes annotation to support the characterization of new genes in the important species.
Collapse
Affiliation(s)
- Iliyas Rashid
- ICAR-National Bureau of Fish Genetic Resources, Canal Ring Road, P.O.- Dilkusha, Lucknow, Uttar Pradesh 226 002, India; AMITY Institute of Biotechnology, AMITY University, Uttar Pradesh, Lucknow Campus, Lucknow 226 028, Uttar Pradesh, India.
| | - Vishwamitra Singh Baisvar
- ICAR-National Bureau of Fish Genetic Resources, Canal Ring Road, P.O.- Dilkusha, Lucknow, Uttar Pradesh 226 002, India.
| | - Mahender Singh
- ICAR-National Bureau of Fish Genetic Resources, Canal Ring Road, P.O.- Dilkusha, Lucknow, Uttar Pradesh 226 002, India.
| | - Ravindra Kumar
- ICAR-National Bureau of Fish Genetic Resources, Canal Ring Road, P.O.- Dilkusha, Lucknow, Uttar Pradesh 226 002, India.
| | - Prachi Srivastava
- AMITY Institute of Biotechnology, AMITY University, Uttar Pradesh, Lucknow Campus, Lucknow 226 028, Uttar Pradesh, India.
| | - Basdeo Kushwaha
- ICAR-National Bureau of Fish Genetic Resources, Canal Ring Road, P.O.- Dilkusha, Lucknow, Uttar Pradesh 226 002, India.
| | - Ajey Kumar Pathak
- ICAR-National Bureau of Fish Genetic Resources, Canal Ring Road, P.O.- Dilkusha, Lucknow, Uttar Pradesh 226 002, India.
| |
Collapse
|
10
|
Faes C, Juban G, Aufradet E, Desgeorges T, Charrin E, Connes P, Mury P, Mattè A, De Franceschi L, Martin C, Pialoux V. Effects of hypoxia-reoxygenation stimuli on renal redox status and nuclear factor erythroid 2-related factor 2 pathway in sickle cell SAD mice. Exp Physiol 2019; 105:357-369. [PMID: 31805612 DOI: 10.1113/ep087969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 12/02/2019] [Indexed: 11/08/2022]
Abstract
NEW FINDINGS What is the central question of this study? What are the effects of repeated subclinical vaso-occlusions on nuclear factor erythroid 2 related factor 2 (Nrf2) and oxidative stress balance regulation in the kidney of transgenic SAD mice? What is the main finding and its importance? In response to hypoxia-reoxygenation, nuclear Nrf2 protein expression decreased in the kidney of SAD mice while haem oxygenase transcripts were increased. This suggest that in SAD mice, other transcription factors than Nrf2 could be involved in renal antioxidant gene regulation in response to hypoxia-reoxygenation. ABSTRACT Hypoxia-reoxygenation (H/R) stress is known to increase oxidative stress in transgenic sickle mice and can cause organ failure. Here we described the effects of H/R on nuclear factor erythroid 2-related factor 2 (Nrf2) as a putative regulator of redox status in the kidneys of SAD mice investigating Nrf2-regulated antioxidant enzymes. Transgenic SAD mice and healthy C57Bl/6J mice were exposed to 4 h of hypoxia followed by various times of reoxygenation at ambient air (2 or 6 h). Regardless of the conditions (i.e. normoxia or H/R), SAD mice expressed higher renal oxidative stress levels. Nuclear Nrf2 protein expression decreased after 2 h post-hypoxia only in the medulla region of the kidney and only in SAD mice. Simultaneously, haem oxygenase transcripts were affected by H/R stimulus with a significant enhancement after 2 h post-hypoxia. Similarly, hypoxia inducible factor-1α staining increased after 2 h post-hypoxia in SAD mice in both cortex and medulla areas. Our data confirm that the kidneys are organs that are particularly sensitive to H/R stimuli in sickle cell SAD mice. Also, these results suggest an effect of the duration of recovery period (short vs. long) and specific responses according to kidney areas, medulla vs. cortex, on Nrf2 expression in response to H/R stimuli in SAD mice.
Collapse
Affiliation(s)
- Camille Faes
- Interuniversity Laboratory of Human Movement Biology EA7424, Univ Lyon - University Claude Bernard Lyon 1, Villeurbanne, France.,Labex GR-Ex, Paris, France
| | - Gaëtan Juban
- Institut NeuroMyoGène CNRS UMR 5510, INSERM U1217, Univ Lyon - University Claude Bernard Lyon 1, Lyon, France
| | - Emeline Aufradet
- Interuniversity Laboratory of Human Movement Biology EA7424, Univ Lyon - University Claude Bernard Lyon 1, Villeurbanne, France
| | - Thibaut Desgeorges
- Institut NeuroMyoGène CNRS UMR 5510, INSERM U1217, Univ Lyon - University Claude Bernard Lyon 1, Lyon, France
| | - Emmanuelle Charrin
- Interuniversity Laboratory of Human Movement Biology EA7424, Univ Lyon - University Claude Bernard Lyon 1, Villeurbanne, France.,Labex GR-Ex, Paris, France
| | - Philippe Connes
- Interuniversity Laboratory of Human Movement Biology EA7424, Univ Lyon - University Claude Bernard Lyon 1, Villeurbanne, France.,Labex GR-Ex, Paris, France.,Institut Universitaire de France, Paris, France
| | - Pauline Mury
- Interuniversity Laboratory of Human Movement Biology EA7424, Univ Lyon - University Claude Bernard Lyon 1, Villeurbanne, France.,Labex GR-Ex, Paris, France
| | - Alessandro Mattè
- Department of Medicine, University of Verona and AOUI-Verona, Verona, Italy
| | | | - Cyril Martin
- Interuniversity Laboratory of Human Movement Biology EA7424, Univ Lyon - University Claude Bernard Lyon 1, Villeurbanne, France.,Labex GR-Ex, Paris, France
| | - Vincent Pialoux
- Interuniversity Laboratory of Human Movement Biology EA7424, Univ Lyon - University Claude Bernard Lyon 1, Villeurbanne, France.,Labex GR-Ex, Paris, France.,Institut Universitaire de France, Paris, France
| |
Collapse
|
11
|
Rafikova O, Williams ER, McBride ML, Zemskova M, Srivastava A, Nair V, Desai AA, Langlais PR, Zemskov E, Simon M, Mandarino LJ, Rafikov R. Hemolysis-induced Lung Vascular Leakage Contributes to the Development of Pulmonary Hypertension. Am J Respir Cell Mol Biol 2019; 59:334-345. [PMID: 29652520 DOI: 10.1165/rcmb.2017-0308oc] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Although hemolytic anemia-associated pulmonary hypertension (PH) and pulmonary arterial hypertension (PAH) are more common than the prevalence of idiopathic PAH alone, the role of hemolysis in the development of PAH is poorly characterized. We hypothesized that hemolysis independently contributes to PAH pathogenesis via endothelial barrier dysfunction with resulting perivascular edema and inflammation. Plasma samples from patients with and without PAH (both confirmed by right heart catheterization) were used to measure free hemoglobin (Hb) and its correlation with PAH severity. A sugen (50 mg/kg)/hypoxia (3 wk)/normoxia (2 wk) rat model was used to elucidate the role of free Hb/heme pathways in PAH. Human lung microvascular endothelial cells were used to study heme-mediated endothelial barrier effects. Our data indicate that patients with PAH have increased levels of free Hb in plasma that correlate with PAH severity. There is also a significant accumulation of free Hb and depletion of haptoglobin in the rat model. In rats, perivascular edema was observed at early time points concomitant with increased infiltration of inflammatory cells. Heme-induced endothelial permeability in human lung microvascular endothelial cells involved activation of the p38/HSP27 pathway. Indeed, the rat model also exhibited increased activation of p38/HSP27 during the initial phase of PH. Surprisingly, despite the increased levels of hemolysis and heme-mediated signaling, there was no heme oxygenase-1 activation. This can be explained by observed destabilization of HIF-1a during the first 2 weeks of PH regardless of hypoxic conditions. Our data suggest that hemolysis may play a significant role in PAH pathobiology.
Collapse
Affiliation(s)
- Olga Rafikova
- 1 Department of Medicine, Division of Endocrinology, and
| | | | | | | | | | - Vineet Nair
- 2 Division of Cardiology, Sarver Heart Center, Department of Medicine, University of Arizona, Tucson, Arizona; and
| | - Ankit A Desai
- 2 Division of Cardiology, Sarver Heart Center, Department of Medicine, University of Arizona, Tucson, Arizona; and
| | | | - Evgeny Zemskov
- 3 Department of Medicine, Division of Translational and Regenerative Medicine, University of Arizona College of Medicine, Tucson, Arizona
| | - Marc Simon
- 4 Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Ruslan Rafikov
- 1 Department of Medicine, Division of Endocrinology, and
| |
Collapse
|
12
|
Phelan JJ, Basdeo SA, Tazoll SC, McGivern S, Saborido JR, Keane J. Modulating Iron for Metabolic Support of TB Host Defense. Front Immunol 2018; 9:2296. [PMID: 30374347 PMCID: PMC6196273 DOI: 10.3389/fimmu.2018.02296] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 09/17/2018] [Indexed: 02/05/2023] Open
Abstract
Tuberculosis (TB) is the world's biggest infectious disease killer. The increasing prevalence of multidrug-resistant and extensively drug-resistant TB demonstrates that current treatments are inadequate and there is an urgent need for novel therapies. Research is now focused on the development of host-directed therapies (HDTs) which can be used in combination with existing antimicrobials, with a special focus on promoting host defense. Immunometabolic reprogramming is integral to TB host defense, therefore, understanding and supporting the immunometabolic pathways that are altered after infection will be important for the development of new HDTs. Moreover, TB pathophysiology is interconnected with iron metabolism. Iron is essential for the survival of Mycobacterium tuberculosis (Mtb), the bacteria that causes TB disease. Mtb struggles to replicate and persist in low iron environments. Iron chelation has therefore been suggested as a HDT. In addition to its direct effects on iron availability, iron chelators modulate immunometabolism through the stabilization of HIF1α. This review examines immunometabolism in the context of Mtb and its links to iron metabolism. We suggest that iron chelation, and subsequent stabilization of HIF1α, will have multifaceted effects on immunometabolic function and holds potential to be utilized as a HDT to boost the host immune response to Mtb infection.
Collapse
Affiliation(s)
- James J Phelan
- Department of Clinical Medicine, Trinity Centre for Health Sciences, Trinity Translational Medicine Institute, St. James's Hospital, Dublin, Ireland
| | - Sharee A Basdeo
- Department of Clinical Medicine, Trinity Centre for Health Sciences, Trinity Translational Medicine Institute, St. James's Hospital, Dublin, Ireland
| | - Simone C Tazoll
- Department of Clinical Medicine, Trinity Centre for Health Sciences, Trinity Translational Medicine Institute, St. James's Hospital, Dublin, Ireland
| | - Sadhbh McGivern
- Department of Clinical Medicine, Trinity Centre for Health Sciences, Trinity Translational Medicine Institute, St. James's Hospital, Dublin, Ireland
| | - Judit R Saborido
- Department of Clinical Medicine, Trinity Centre for Health Sciences, Trinity Translational Medicine Institute, St. James's Hospital, Dublin, Ireland
| | - Joseph Keane
- Department of Clinical Medicine, Trinity Centre for Health Sciences, Trinity Translational Medicine Institute, St. James's Hospital, Dublin, Ireland
| |
Collapse
|
13
|
Yuan X, Lee JW, Bowser JL, Neudecker V, Sridhar S, Eltzschig HK. Targeting Hypoxia Signaling for Perioperative Organ Injury. Anesth Analg 2018; 126:308-321. [PMID: 28759485 PMCID: PMC5735013 DOI: 10.1213/ane.0000000000002288] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Perioperative organ injury has a significant impact on surgical outcomes and presents a leading cause of death in the United States. Recent research has pointed out an important role of hypoxia signaling in the protection from organ injury, including for example myocardial infarction, acute respiratory distress syndrome, acute kidney, or gut injury. Hypoxia induces the stabilization of hypoxia-inducible factors (HIFs), thereby leading to the induction of HIF target genes, which facilitates adaptive responses to low oxygen. In this review, we focus on current therapeutic strategies targeting hypoxia signaling in various organ injury models and emphasize potential clinical approaches to integrate these findings into the care of surgical patients. Conceptually, there are 2 options to target the HIF pathway for organ protection. First, drugs became recently available that promote the stabilization of HIFs, most prominently via inhibition of prolyl hydroxylase. These compounds are currently trialed in patients, for example, for anemia treatment or prevention of ischemia and reperfusion injury. Second, HIF target genes (such as adenosine receptors) could be activated directly. We hope that some of these approaches may lead to novel pharmacologic strategies to prevent or treat organ injury in surgical patients.
Collapse
Affiliation(s)
- Xiaoyi Yuan
- Department of Anesthesiology, the University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Jae W. Lee
- Department of Anesthesiology, the University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Jessica L. Bowser
- Department of Anesthesiology, the University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Viola Neudecker
- Department of Anesthesiology, Clinic of the University of Munich, Munich, Germany
| | - Srikanth Sridhar
- Department of Anesthesiology, the University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Holger K. Eltzschig
- Department of Anesthesiology, the University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| |
Collapse
|
14
|
Ito H, Kurokawa H, Hirayama A, P Indo H, J Majima H, Matsui H. Cancer cell-specific mitochondrial reactive oxygen species promote non-heme iron uptake and enhance the proliferation of gastric epithelial cancer cell. J Clin Biochem Nutr 2017; 61:183-188. [PMID: 29203959 PMCID: PMC5703790 DOI: 10.3164/jcbn.17-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Accepted: 06/12/2017] [Indexed: 01/23/2023] Open
Abstract
Iron is an essential nutrient for life and is involved in many important processes such as oxygen transport and DNA synthesis. However, excess amounts of iron can cause carcinogenesis by producing reactive oxygen species. Thus, the cellular transport of iron must be tightly regulated. In the human body, iron may be present as heme or non-heme iron. The mechanisms governing the cellular transport of these forms have not been clearly elucidated. We previously reported that the expression of an important heme transporter, heme carrier protein 1 was regulated by cancer-specific reactive oxygen species derived from mitochondria. In this study, we have asked if mitochondrial reactive oxygen species may also be related with non-heme iron transport. In order to address this question, we have investigated the relationship between mitochondrial reactive oxygen species and accumulation of cellular non-heme iron in a rat gastric normal, cancer and manganese superoxide dismutase-overexpressing cancer cell line, in which reactive oxygen species from mitochondria are specifically scavenged. We have also analyzed the expression of divalent metal transporter 1 and ferroprotin, involved in the incorporation and excretion of non-heme iron, respectively, as well as a hypoxia-related transcription factor HIF-1α, to elucidate the molecular mechanism of non-heme iron accumulation.
Collapse
Affiliation(s)
- Hiromu Ito
- Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8575, Japan
| | - Hiromi Kurokawa
- Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8575, Japan
| | - Aki Hirayama
- Center for Integrative Medicine, Tsukuba University of Technology, 4-12-7 Kasuga, Tsukuba 305-8521, Japan
| | - Hiroko P Indo
- Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| | - Hideyuki J Majima
- Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| | - Hirofumi Matsui
- Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8575, Japan.,Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| |
Collapse
|
15
|
Lee YJ, Bernstock JD, Nagaraja N, Ko B, Hallenbeck JM. Global SUMOylation facilitates the multimodal neuroprotection afforded by quercetin against the deleterious effects of oxygen/glucose deprivation and the restoration of oxygen/glucose. J Neurochem 2016; 138:101-16. [PMID: 27087120 DOI: 10.1111/jnc.13643] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 04/05/2016] [Accepted: 04/06/2016] [Indexed: 12/13/2022]
Abstract
The putative neuroprotective properties of various flavonoids have long been reported. Among this class of chemicals, quercetin, a major flavone/flavonol naturally occurring in plants, deserves focused attention because of the myriad of beneficial effects observed in various in vitro and in vivo models of central nervous system damage/degeneration. However, the mechanisms governing the beneficial outcomes mediated by quercetin remain to be elucidated. In an effort to define the underlying molecular mechanisms, our study employed human/rat neuroblastoma cell lines (SHSY5Y and B35, respectively) and E18-derived rat primary cortical neurons upon which the effects of various flavonoids were examined. Of note, increases in the levels of global SUMOylation, a post-translational modification with the Small Ubiquitin-like MOdifier (SUMO) were pronounced. Quercetin treatment increased SUMOylation levels in both SHSY5Y cells and rat cortical neurons in a dose and time-dependent manner, possibly via the direct inactivation of certain SENPs (SUMO-specific isopeptidases). Of particular interest, cells treated with quercetin displayed increased tolerance to oxygen/glucose deprivation exposure, an in vitro model of ischemia. SHSY5Y cells treated with quercetin also increased the expression of Nrf2 (via a decrease in the levels of Keap1), heme oxygenase-1 (HO-1), and nitric oxide synthase 1 (NOS1), which provide further protection from oxidative stress. In addition, the increased SUMOylation of HIF-1α was noted and deemed to be significant. We hypothesize that SUMOylated HIF-1α plays a fundamental role in the protection afforded and may underlie some of quercetin's ability to protect cells from oxygen/glucose deprivation-induced cell death, via an up-regulation of HO-1 and NOS1, which ultimately leads to the induction of pro-life NOS1/protein kinase G signaling. Quercetin acts to increase survival in the face of ischemia via an increase of SENP3 expression, the possible inactivation of SENPs 1/2, and via a decrease in KEAP1 levels (thereby increasing Nrf2 stability). These changes may then lead to increase in HIF-1α SUMOylation and HO-1 activation, followed by an up-regulation of NOS1/PKG signaling. Pathways altered via quercetin treatment within our experimental system are represented by blue arrowheads. Solid black arrows represent relationships that have been explored while a dotted arrow represents a relationship that has yet to be confirmed.
Collapse
Affiliation(s)
- Yang-Ja Lee
- Stroke Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health (NINDS/NIH), Bethesda, Maryland, USA
| | - Joshua D Bernstock
- Stroke Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health (NINDS/NIH), Bethesda, Maryland, USA
| | - Nandakumar Nagaraja
- Stroke Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health (NINDS/NIH), Bethesda, Maryland, USA
| | - Brian Ko
- Stroke Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health (NINDS/NIH), Bethesda, Maryland, USA
| | - John M Hallenbeck
- Stroke Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health (NINDS/NIH), Bethesda, Maryland, USA
| |
Collapse
|
16
|
Fu Q, Colgan SP, Shelley CS. Hypoxia: The Force that Drives Chronic Kidney Disease. Clin Med Res 2016; 14:15-39. [PMID: 26847481 PMCID: PMC4851450 DOI: 10.3121/cmr.2015.1282] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 09/30/2015] [Indexed: 12/15/2022]
Abstract
In the United States the prevalence of end-stage renal disease (ESRD) reached epidemic proportions in 2012 with over 600,000 patients being treated. The rates of ESRD among the elderly are disproportionally high. Consequently, as life expectancy increases and the baby-boom generation reaches retirement age, the already heavy burden imposed by ESRD on the US health care system is set to increase dramatically. ESRD represents the terminal stage of chronic kidney disease (CKD). A large body of evidence indicating that CKD is driven by renal tissue hypoxia has led to the development of therapeutic strategies that increase kidney oxygenation and the contention that chronic hypoxia is the final common pathway to end-stage renal failure. Numerous studies have demonstrated that one of the most potent means by which hypoxic conditions within the kidney produce CKD is by inducing a sustained inflammatory attack by infiltrating leukocytes. Indispensable to this attack is the acquisition by leukocytes of an adhesive phenotype. It was thought that this process resulted exclusively from leukocytes responding to cytokines released from ischemic renal endothelium. However, recently it has been demonstrated that leukocytes also become activated independent of the hypoxic response of endothelial cells. It was found that this endothelium-independent mechanism involves leukocytes directly sensing hypoxia and responding by transcriptional induction of the genes that encode the β2-integrin family of adhesion molecules. This induction likely maintains the long-term inflammation by which hypoxia drives the pathogenesis of CKD. Consequently, targeting these transcriptional mechanisms would appear to represent a promising new therapeutic strategy.
Collapse
Affiliation(s)
- Qiangwei Fu
- Kabara Cancer Research Institute, La Crosse, WI
| | - Sean P Colgan
- Mucosal Inflammation Program and University of Colorado School of Medicine, Aurora, CO
| | - Carl Simon Shelley
- University of Wisconsin School of Medicine and Public Health, Madison, WI
| |
Collapse
|
17
|
Babu KR, Muckenthaler MU. miR-20a regulates expression of the iron exporter ferroportin in lung cancer. J Mol Med (Berl) 2015; 94:347-59. [PMID: 26560875 PMCID: PMC4803811 DOI: 10.1007/s00109-015-1362-3] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 10/20/2015] [Accepted: 10/26/2015] [Indexed: 12/25/2022]
Abstract
Abstract Ferroportin (FPN) exports iron from duodenal enterocytes, macrophages, and hepatocytes to maintain systemic iron homeostasis. In addition, FPN is expressed in various cancer cells. Here, we show that in lung cancer, FPN expression is regulated by miR-20a. Within the FPN-3′-untranslated region (3′UTR), we identify and experimentally validate three evolutionarily conserved target sites for the microRNA (miRNA) members of the miR-17 seed family, including miR-20a. Our analysis of RNA sequencing data from patients with lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC) revealed that FPN messenger RNA (mRNA) levels are significantly decreased in tumor compared to matched healthy tissue, while miR-20a levels are increased. A significant negative correlation of miR-20a and FPN expression was observed. Functional studies further demonstrate that FPN is post-transcriptionally regulated by miR-20a in non-small cell lung cancer (NSCLC) cells and that overexpression or knockdown of miR-20a or FPN affects NSCLC proliferation and colony formation. Taken together, our data suggest that increased expression of miR-20 in lung cancer may decrease iron export, leading to intracellular iron retention, which, in turn, favors cell proliferation. Key messages miR-20a controls expression of the iron exporter ferroportin (FPN) by binding to highly conserved target sites in its 3′UTR. Expression of miR-20a is inversely correlated to FPN in lung cancer. Low FPN expression stimulates proliferation and colony formation of non-small cell lung cancer (NSCLC) cells, possibly by increasing iron availability for cancer cell proliferation.
Electronic supplementary material The online version of this article (doi:10.1007/s00109-015-1362-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kamesh R Babu
- Department of Pediatric Hematology, Oncology, and Immunology, University of Heidelberg, Heidelberg, Germany.,Molecular Medicine Partnership Unit, University of Heidelberg, Heidelberg, Germany
| | - Martina U Muckenthaler
- Department of Pediatric Hematology, Oncology, and Immunology, University of Heidelberg, Heidelberg, Germany. .,Molecular Medicine Partnership Unit, University of Heidelberg, Heidelberg, Germany.
| |
Collapse
|
18
|
Boname JM, Bloor S, Wandel MP, Nathan JA, Antrobus R, Dingwell KS, Thurston TL, Smith DL, Smith JC, Randow F, Lehner PJ. Cleavage by signal peptide peptidase is required for the degradation of selected tail-anchored proteins. ACTA ACUST UNITED AC 2014; 205:847-62. [PMID: 24958774 PMCID: PMC4068138 DOI: 10.1083/jcb.201312009] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Intramembrane proteolytic cleavage by signal peptide peptidase is required for the turnover of some ER-resident, tail-anchored membrane proteins. The regulated turnover of endoplasmic reticulum (ER)–resident membrane proteins requires their extraction from the membrane lipid bilayer and subsequent proteasome-mediated degradation. Cleavage within the transmembrane domain provides an attractive mechanism to facilitate protein dislocation but has never been shown for endogenous substrates. To determine whether intramembrane proteolysis, specifically cleavage by the intramembrane-cleaving aspartyl protease signal peptide peptidase (SPP), is involved in this pathway, we generated an SPP-specific somatic cell knockout. In a stable isotope labeling by amino acids in cell culture–based proteomics screen, we identified HO-1 (heme oxygenase-1), the rate-limiting enzyme in the degradation of heme to biliverdin, as a novel SPP substrate. Intramembrane cleavage by catalytically active SPP provided the primary proteolytic step required for the extraction and subsequent proteasome-dependent degradation of HO-1, an ER-resident tail-anchored protein. SPP-mediated proteolysis was not limited to HO-1 but was required for the dislocation and degradation of additional tail-anchored ER-resident proteins. Our study identifies tail-anchored proteins as novel SPP substrates and a specific requirement for SPP-mediated intramembrane cleavage in protein turnover.
Collapse
Affiliation(s)
- Jessica M Boname
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY, England, UK Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, England, UK
| | - Stuart Bloor
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY, England, UK Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, England, UK
| | - Michal P Wandel
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY, England, UK Division of Protein and Nucleic Acid Chemistry, Medical Research Council Laboratory of Molecular Biology, Cambridge CB2 0QH, England, UK
| | - James A Nathan
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY, England, UK Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, England, UK
| | - Robin Antrobus
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY, England, UK
| | - Kevin S Dingwell
- Medical Research Council National Institute for Medical Research, London NW7 1AA, England, UK
| | - Teresa L Thurston
- Division of Protein and Nucleic Acid Chemistry, Medical Research Council Laboratory of Molecular Biology, Cambridge CB2 0QH, England, UK
| | - Duncan L Smith
- Cancer Research UK Manchester Institute, University of Manchester, Manchester M20 4B, England, UK
| | - James C Smith
- Medical Research Council National Institute for Medical Research, London NW7 1AA, England, UK
| | - Felix Randow
- Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, England, UK Division of Protein and Nucleic Acid Chemistry, Medical Research Council Laboratory of Molecular Biology, Cambridge CB2 0QH, England, UK
| | - Paul J Lehner
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY, England, UK Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, England, UK
| |
Collapse
|
19
|
Renal overexpression of atrial natriuretic peptide and hypoxia inducible factor-1α as adaptive response to a high salt diet. BIOMED RESEARCH INTERNATIONAL 2014; 2014:936978. [PMID: 24689065 PMCID: PMC3943195 DOI: 10.1155/2014/936978] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Revised: 12/31/2013] [Accepted: 01/06/2014] [Indexed: 01/11/2023]
Abstract
In the kidney, a high salt intake favors oxidative stress and hypoxia and causes the development of fibrosis. Both atrial natriuretic peptide (ANP) and hypoxia inducible factor (HIF-1α) exert cytoprotective effects. We tested the hypothesis that renal expression of ANP and HIF-1α is involved in a mechanism responding to the oxidative stress produced in the kidneys of rats chronically fed a high sodium diet. Sprague-Dawley rats were fed with a normal salt (0.4% NaCl) (NS) or a high salt (8% NaCl) (HS) diet for 3 weeks, with or without the administration of tempol (T), an inhibitor of oxidative stress, in the drinking water. We measured the mean arterial pressure (MAP), glomerular filtration rate (GFR), and urinary sodium excretion (UVNa). We evaluated the expression of ANP, HIF-1α, and transforming growth factor (TGF-β1) in renal tissues by western blot and immunohistochemistry. The animals fed a high salt diet showed increased MAP and UVNa levels and enhanced renal immunostaining of ANP, HIF-1α, and TGF-β1. The administration of tempol together with the sodium overload increased the natriuresis further and prevented the elevation of blood pressure and the increased expression of ANP, TGF-β1, and HIF-1α compared to their control. These findings suggest that HIF-1α and ANP, synthesized by the kidney, are involved in an adaptive mechanism in response to a sodium overload to prevent or attenuate the deleterious effects of the oxidative stress and the hypoxia on the development of fibrosis.
Collapse
|
20
|
Esteban MA, Maxwell PH. Manipulation of oxygen tensions forin vitrocell culture using a hypoxic workstation. Expert Rev Proteomics 2014; 2:307-14. [PMID: 16000078 DOI: 10.1586/14789450.2.3.307] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
It is increasingly clear that oxygen tension exerts potent effects on many biologic processes in a range well above that at which aerobic metabolism is compromised. Cell culture ex vivo is traditionally performed in unstirred liquid media at ambient oxygen concentrations in the laboratory, with no attention to the level of oxygen experienced by the cells. This is certainly not reflecting physiology, and oxygenation may be further altered during cell handling and extraction procedures. The hypoxia-inducible factor pathway illustrates the potential for oxygen tension to have dramatic effects in terms of post-translational modification of proteins, and to influence a broad range of cellular pathways including those involved in substrate transport, metabolic pathways, growth factor signaling and differentiation. While the standard laboratory approach may remain suitable for many biologic applications, there are other situations in which more attention to oxygenation will be appropriate. This review discusses a workstation that allows investigators to manipulate oxygenation.
Collapse
Affiliation(s)
- Miguel A Esteban
- Imperial College, Renal laboratory, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK.
| | | |
Collapse
|
21
|
Role of haem oxygenase in the renoprotective effects of soluble epoxide hydrolase inhibition in diabetic spontaneously hypertensive rats. Clin Sci (Lond) 2013; 125:349-59. [PMID: 23611540 DOI: 10.1042/cs20130003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
We have shown previously that inhibition of sEH (soluble epoxide hydrolase) increased EETs (epoxyeicosatrienoic acids) levels and reduced renal injury in diabetic mice and these changes were associated with induction of HO (haem oxygenase)-1. The present study determines whether the inhibition of HO negates the renoprotective effect of sEH inhibition in diabetic SHR (spontaneously hypertensive rats). After 6 weeks of induction of diabetes with streptozotocin, SHR were divided into the following groups: untreated, treated with the sEH inhibitor t-AUCB {trans-4-[4-(3-adamantan-1-yl-ureido)-cyclohexyloxy]-benzoic acid}, treated with the HO inhibitor SnMP (stannous mesoporphyrin), and treated with both inhibitors for 4 more weeks; non-diabetic SHR served as a control group. Induction of diabetes significantly increased renal sEH expression and decreased the renal EETs/DHETEs (dihydroxyeicosatrienoic acid) ratio without affecting HO-1 activity or expression in SHR. Inhibition of sEH with t-AUCB increased the renal EETs/DHETEs ratio and HO-1 activity in diabetic SHR; however, it did not significantly alter systolic blood pressure. Treatment of diabetic SHR with t-AUCB significantly reduced the elevation in urinary albumin and nephrin excretion, whereas co-administration of the HO inhibitor SnMP with t-AUCB prevented these changes. Immunohistochemical analysis revealed elevations in renal fibrosis as indicated by increased renal TGF-β (transforming growth factor β) levels and fibronectin expression in diabetic SHR and these changes were reduced with sEH inhibition. Co-administration of SnMP with t-AUCB prevented its ability to reduce renal fibrosis in diabetic SHR. In addition, SnMP treatment also prevented t-AUCB-induced decreases in renal macrophage infiltration, IL-17 expression and MCP-1 levels in diabetic SHR. These findings suggest that HO-1 induction is involved in the protective effect of sEH inhibition against diabetic renal injury.
Collapse
|
22
|
Upstream stimulatory factor 2 and hypoxia-inducible factor 2α (HIF2α) cooperatively activate HIF2 target genes during hypoxia. Mol Cell Biol 2012; 32:4595-610. [PMID: 22966206 DOI: 10.1128/mcb.00724-12] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
While the functions of hypoxia-inducible factor 1α (HIF1α)/aryl hydrocarbon receptor nuclear translocator (ARNT) and HIF2α/ARNT (HIF2) proteins in activating hypoxia-inducible genes are well established, the role of other transcription factors in the hypoxic transcriptional response is less clear. We report here for the first time that the basic helix-loop-helix-leucine-zip transcription factor upstream stimulatory factor 2 (USF2) is required for the hypoxic transcriptional response, specifically, for hypoxic activation of HIF2 target genes. We show that inhibiting USF2 activity greatly reduces hypoxic induction of HIF2 target genes in cell lines that have USF2 activity, while inducing USF2 activity in cells lacking USF2 activity restores hypoxic induction of HIF2 target genes. Mechanistically, USF2 activates HIF2 target genes by binding to HIF2 target gene promoters, interacting with HIF2α protein, and recruiting coactivators CBP and p300 to form enhanceosome complexes that contain HIF2α, USF2, CBP, p300, and RNA polymerase II on HIF2 target gene promoters. Functionally, the effect of USF2 knockdown on proliferation, motility, and clonogenic survival of HIF2-dependent tumor cells in vitro is phenocopied by HIF2α knockdown, indicating that USF2 works with HIF2 to activate HIF2 target genes and to drive HIF2-depedent tumorigenesis.
Collapse
|
23
|
Aharoni-Simon M, Anavi S, Beifuss U, Madar Z, Tirosh O. Nitric oxide, can it be only good? Increasing the antioxidant properties of nitric oxide in hepatocytes by YC-1 compound. Nitric Oxide 2012; 27:248-56. [PMID: 22955014 DOI: 10.1016/j.niox.2012.08.076] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2012] [Revised: 08/05/2012] [Accepted: 08/21/2012] [Indexed: 12/25/2022]
Abstract
The aim of the study was to evaluate the effect of Nitric oxide (NO) on redox changes and fat accumulation in hepatocytes. AML-12 hepatocytes were exposed to the NO donor Diethylenetriamine-NONOate (DETA-NO). DETA-NO led to a dose- and time-dependent increase in lipid accumulation in the cells, measured by Nile red fluorescence. Exposure of the cells to 1mM DETA-NO for 24h increased reactive oxygen species production, mainly peroxides. At the same time, NO induced elevation of reduced glutathione (GSH) and a mild activation of the antioxidant transcription factors Hypoxia-inducible factor 1α (HIF1α) and NF-E2 related factor 2 (Nrf-2). We used 100 μM YC-1 to inhibit HIF1α activity and induce activation of soluble Guanylate Cyclase (sGC). YC-1 alone did not affect fat accumulation, and only moderately increased the expression of Nrf-2-targeted genes Heme oxygenase 1 (Hmox1), NAD(P)H dehydrogenase (quinone 1) (Nqo1) and Glutathione S-transferase α1 (Gstα1). However, YC-1 abolished the negative effect of NO on fat accumulation when administered together. Strikingly, YC-1 potentiated the effect of NO on Nrf-2 activation, thus increasing dramatically the antioxidant properties of NO. Moreover, YC-1 intensified the effect of NO on the expression of peroxisome-proliferator-activated receptor-gamma co-activator 1α (PGC1α) and mitochondrial biogenesis markers. This study suggests that YC-1 may shift the deleterious effects of NO into the beneficial ones, and may improve the antioxidant properties of NO.
Collapse
Affiliation(s)
- Michal Aharoni-Simon
- The School of Nutritional Sciences, Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | | | | | | | | |
Collapse
|
24
|
Xu X, Kriegel AJ, Liu Y, Usa K, Mladinov D, Liu H, Fang Y, Ding X, Liang M. Delayed ischemic preconditioning contributes to renal protection by upregulation of miR-21. Kidney Int 2012; 82:1167-75. [PMID: 22785173 PMCID: PMC3777822 DOI: 10.1038/ki.2012.241] [Citation(s) in RCA: 138] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Delayed ischemic preconditioning effectively protects kidneys from ischemia-reperfusion injury but the mechanism underlying renal protection remains poorly understood. Here we examined the in vivo role of microRNA miR-21 in the renal protection conferred by delayed ischemic preconditioning in mice. A 15 minute renal ischemic preconditioning significantly increased the expression of miR-21 by 4 hours and substantially attenuated ischemia-reperfusion injury induced 4 days later. A locked nucleic acid-modified anti-miR-21 given at the time of ischemic preconditioning knocked down miR-21 and significantly exacerbated subsequent ischemia-reperfusion injury in the mouse kidney. Knockdown of miR-21 resulted in significant upregulation of programmed cell death protein 4, a pro-apoptotic target gene of miR-21, and substantially increased tubular cell apoptosis. Hypoxia inducible factor-1α in the kidney was activated after ischemic preconditioning and blockade of its activity with a decoy abolished the up-regulation of miR-21 in cultured human renal epithelial cells treated with the inducer cobalt chloride. In the absence of ischemic preconditioning, knockdown of miR-21 alone did not significantly affect ischemia-reperfusion injury in the mouse kidney. Thus, upregulation of miR-21 contributes to the protective effect of delayed ischemic preconditioning against subsequent renal ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Xialian Xu
- Division of Nephrology, Shanghai Medical College, Fudan University, Zhongshan Hospital, Shanghai, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Lin PH, Lan WM, Chau LY. TRC8 suppresses tumorigenesis through targeting heme oxygenase-1 for ubiquitination and degradation. Oncogene 2012; 32:2325-34. [PMID: 22689053 DOI: 10.1038/onc.2012.244] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The TRC8 gene, which was previously shown to be disrupted by a 3;8 chromosomal translocation in hereditary kidney cancer, encodes for an endoplasmic reticulum-resident E3 ligase. Studies have shown that TRC8 exhibits a tumor-suppressive effect through its E3-ligase activity. Therefore, the identification of its physiological substrates will provide important insights into the molecular mechanism underlying TRC8-mediated tumor suppression. Here we show that TRC8 targets heme oxygenase-1 (HO-1), an antioxidant enzyme highly expressed in various cancers, for ubiquitination and degradation. Ectopic TRC8 expression suppresses HO-1-induced cancer cell growth and migration/invasion. Conversely, HO-1 depletion reduced the tumorigenic and invasive capacities promoted by TRC8 knockdown. HO-1 downregulation in renal carcinoma cells induces a mitotic delay at G2/M phase by increasing the intracellular reactive oxygen species and the DNA-damage-induced checkpoint activation. These results highlight the tumorigenic role of HO-1 and the importance of TRC8-mediated HO-1 degradation in the control of cancer growth.
Collapse
Affiliation(s)
- P-H Lin
- Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
| | | | | |
Collapse
|
26
|
du Souich P, Fradette C. The effect and clinical consequences of hypoxia on cytochrome P450, membrane carrier proteins activity and expression. Expert Opin Drug Metab Toxicol 2011; 7:1083-100. [DOI: 10.1517/17425255.2011.586630] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
27
|
Yim HE, Kim JH, Yoo KH, Bae IS, Hong YS, Lee JW. Spironolactone and enalapril differentially up-regulate the expression of VEGF and heme oxygenase-1 in the neonatal rat kidney. Pediatr Res 2011; 69:378-83. [PMID: 21263376 DOI: 10.1203/pdr.0b013e3182114c38] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Both the renin-angiotensin-aldosterone system (RAAS) and hypoxia are vital physiological factors involved in the control of nephrogenesis and vascularization. We investigated the relationship between RAAS and hypoxia in the developing kidney. The expression of VEGF and heme oxygenase (HO)-1 related with the oxygen was analyzed in the enalapril- or spironolactone-treated neonatal rat kidneys. Enalapril (30 mg/kg/d) or spironolactone (200 mg/kg/d) was administered to newborn rat pups for 7 d. The newborn rats were injected i.p. with pimonidazole (200 mg/kg), a marker of severe tissue hypoxia, 1 h before killing. VEGF and HO-1 protein expression was significantly increased by immunoblots and immunohistochemistry in both the enalapril- and spironolactone-treated kidneys, compared with the controls (p < 0.05). HO-1 mRNA expression was increased in the spironolactone-treated group (p < 0.05). The immunoactivity of pimonidazole was not different from that of the controls in the enalapril-treated group, whereas it was increased in the spironolactone-treated group. The results of this study indicate that aldosterone blockade or angiotensin II inhibition in the developing rat kidney up-regulated renal VEGF and HO-1 expression regardless of the hypoxic conditions and may differentially modulate VEGF and HO-1 production.
Collapse
Affiliation(s)
- Hyung Eun Yim
- Department of Pediatrics, Korea University Medical Center, College of Medicine, Korea University, Seoul 152-703, Korea
| | | | | | | | | | | |
Collapse
|
28
|
Jiang YF, Chou CH, Lin EC, Chiu CH. Molecular characterization of hypoxia and hypoxia-inducible factor 1 alpha (HIF-1α) from Taiwan voles (Microtus kikuchii). Comp Biochem Physiol A Mol Integr Physiol 2010; 158:183-8. [PMID: 20937407 DOI: 10.1016/j.cbpa.2010.10.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2010] [Revised: 10/04/2010] [Accepted: 10/04/2010] [Indexed: 11/17/2022]
Abstract
Hypoxia-inducible factor 1 (HIF-1) is a transcription factor that senses and adapts cells to hypoxic environmental conditions. HIF-1 is composed of an oxygen-regulated α subunit (HIF-1α) and a constitutively expressed β subunit (HIF-1β). Taiwan voles (Microtus kikuchii) are an endemic species in Taiwan, found only in mountainous areas greater than 2000m above sea level. In this study, the full-length HIF-1α cDNA was cloned and sequenced from liver tissues of Taiwan voles. We found that HIF-1α of Taiwan voles had high sequence similarity to HIF-1α of other species. Sequence alignment of HIF-1α functional domains indicated basic helix-loop-helix (bHLH), PER-ARNT-SIM (PAS) and C-terminal transactivation (TAD-C) domains were conserved among species, but sequence variations were found between the oxygen-dependent degradation domains (ODDD). To measure Taiwan vole HIF-1α responses to hypoxia, animals were challenged with cobalt chloride, and HIF-1α mRNA and protein expression in brain, lung, heart, liver, kidney, and muscle was assessed by quantitative RT-PCR and Western blot analysis. Upon induction of hypoxic stress with cobalt chloride, an increase in HIF-1α mRNA levels was detected in lung, heart, kidney, and muscle tissue. In contrast, protein expression levels showed greater variation between individual animals. These results suggest that the regulation of HIF-1α may be important to the Taiwan vole under cobalt chloride treatments. But more details regarding the evolutionary effect of environmental pressure on HIF-1α primary sequence, HIF-1α function and regulation in Taiwan voles remain to be identified.
Collapse
Affiliation(s)
- Yi-Fan Jiang
- Department of Animal Science and Technology, College of Bio-Resources and Agriculture, National Taiwan University, Taipei 10617, Taiwan
| | | | | | | |
Collapse
|
29
|
Dai KY, Chan SH, Chang AY. Heme oxygenase-1 plays a pro-life role in experimental brain stem death via nitric oxide synthase I/protein kinase G signaling at rostral ventrolateral medulla. J Biomed Sci 2010; 17:72. [PMID: 20819234 PMCID: PMC2941487 DOI: 10.1186/1423-0127-17-72] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2010] [Accepted: 09/07/2010] [Indexed: 12/13/2022] Open
Abstract
Background Despite its clinical importance, a dearth of information exists on the cellular and molecular mechanisms that underpin brain stem death. A suitable neural substrate for mechanistic delineation on brain stem death resides in the rostral ventrolateral medulla (RVLM) because it is the origin of a life-and-death signal that sequentially increases (pro-life) and decreases (pro-death) to reflect the advancing central cardiovascular regulatory dysfunction during the progression towards brain stem death in critically ill patients. The present study evaluated the hypothesis that heme oxygnase-1 (HO-1) may play a pro-life role as an interposing signal between hypoxia-inducible factor-1 (HIF-1) and nitric oxide synthase I (NOS I)/protein kinase G (PKG) cascade in RVLM, which sustains central cardiovascular regulatory functions during brain stem death. Methods We performed cardiovascular, pharmacological, biochemical and confocal microscopy experiments in conjunction with an experimental model of brain stem death that employed microinjection of the organophosphate insecticide mevinphos (Mev; 10 nmol) bilaterally into RVLM of adult male Sprague-Dawley rats. Results Western blot analysis coupled with laser scanning confocal microscopy revealed that augmented HO-1 expression that was confined to the cytoplasm of RVLM neurons occurred preferentially during the pro-life phase of experimental brain stem death and was antagonized by immunoneutralization of HIF-1α or HIF-1β in RVLM. On the other hand, the cytoplasmic presence of HO-2 in RVLM neurons manifested insignificant changes during both phases. Furthermore, immunoneutralization of HO-1 or knockdown of ho-1 gene in RVLM blunted the augmented life-and-death signals exhibited during the pro-life phase. Those pretreatments also blocked the upregulated pro-life NOS I/PKG signaling without affecting the pro-death NOS II/peroxynitrite cascade in RVLM. Conclusions We conclude that transcriptional upregulation of HO-1 on activation by HIF-1 in RVLM plays a preferential pro-life role by sustaining central cardiovascular regulatory functions during brain stem death via upregulation of NOS I/PKG signaling pathway. Our results further showed that the pro-dead NOS II/peroxynitrite cascade in RVLM is not included in this repertoire of cellular events.
Collapse
Affiliation(s)
- Kuang-Yu Dai
- Center for Translational Research in Biomedical Sciences, Chang Gung Memorial, Hospital-Kaohsiung Medical Center, Kaohsiung County 83301, Taiwan
| | | | | |
Collapse
|
30
|
Abstract
Heme is an essential molecule in aerobic organisms. Heme consists of protoporphyrin IX and a ferrous (Fe(2+)) iron atom, which has high affinity for oxygen (O(2)). Hemoglobin, the major oxygen-carrying protein in blood, is the most abundant heme-protein in animals and humans. Hemoglobin consists of four globin subunits (alpha(2)beta(2)), with each subunit carrying a heme group. Ferrous (Fe(2+)) hemoglobin is easily oxidized in circulation to ferric (Fe(3+)) hemoglobin, which readily releases free hemin. Hemin is hydrophobic and intercalates into cell membranes. Hydrogen peroxide can split the heme ring and release "free" redox-active iron, which catalytically amplifies the production of reactive oxygen species. These oxidants can oxidize lipids, proteins, and DNA; activate cell-signaling pathways and oxidant-sensitive, proinflammatory transcription factors; alter protein expression; perturb membrane channels; and induce apoptosis and cell death. Heme-derived oxidants induce recruitment of leukocytes, platelets, and red blood cells to the vessel wall; oxidize low-density lipoproteins; and consume nitric oxide. Heme metabolism, extracellular and intracellular defenses against heme, and cellular cytoprotective adaptations are emphasized. Sickle cell disease, an archetypal example of hemolysis, heme-induced oxidative stress, and cytoprotective adaptation, is reviewed.
Collapse
Affiliation(s)
- John D Belcher
- Department of Medicine, Division of Hematology, Oncology and Transplantation, Minneapolis, Minnesota 55455, USA
| | | | | | | | | |
Collapse
|
31
|
He JZ, Ho JJD, Gingerich S, Courtman DW, Marsden PA, Ward ME. Enhanced translation of heme oxygenase-2 preserves human endothelial cell viability during hypoxia. J Biol Chem 2010; 285:9452-9461. [PMID: 20118244 DOI: 10.1074/jbc.m109.077230] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Heme oxygenases (HOs) -1 and -2 catalyze the breakdown of heme to release carbon monoxide, biliverdin, and ferrous iron, which may preserve cell function during oxidative stress. HO-1 levels decrease in endothelial cells exposed to hypoxia, whereas the effect of hypoxia on HO-2 expression is unknown. The current study was carried out to determine if hypoxia alters HO-2 protein levels in human endothelial cells and whether this enzyme plays a role in preserving their viability during hypoxic stress. Human umbilical vein endothelial cells (HUVECs), human aortic endothelial cells (HAECs), and human blood outgrowth endothelial cells were exposed to 21% or 1% O(2) for 48 or 16 h in the presence or absence of tumor necrosis factor-alpha (10 ng/ml) or H(2)O(2) (100 microm). In all three endothelial cell types HO-1 mRNA and protein levels were decreased following hypoxic incubation, whereas HO-2 protein levels were unaltered. In HUVECs HO-2 levels were maintained during hypoxia despite a 57% reduction in steady-state HO-2 mRNA level and a 43% reduction in total protein synthesis. Polysome profiling revealed increased HO-2 transcript association with polysomes during hypoxia consistent with enhanced translation of these transcripts. Importantly, inhibition of HO-2 expression by small interference RNA increased oxidative stress, exacerbated mitochondrial membrane depolarization, and enhanced caspase activation and apoptotic cell death in cells incubated under hypoxic but not normoxic conditions. These data indicate that HO-2 is important in maintaining endothelial viability and may preserve local regulation of vascular tone, thrombosis, and inflammatory responses during reductions in systemic oxygen delivery.
Collapse
Affiliation(s)
- Jeff Z He
- Terrence Donnelly Laboratories, Toronto, Ontario M5B 1W8; Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario M5B 1W8; Departments of Laboratory Medicine and Pathobiology, Toronto, Ontario M5S 1A8, Canada
| | - J J David Ho
- Medical Biophysics, Toronto, Ontario M5S 1A8, Canada
| | - Sheena Gingerich
- Nephrology, Department of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - David W Courtman
- Terrence Donnelly Laboratories, Toronto, Ontario M5B 1W8; Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario M5B 1W8
| | - Philip A Marsden
- Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario M5B 1W8; Nephrology, Department of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada.
| | - Michael E Ward
- Terrence Donnelly Laboratories, Toronto, Ontario M5B 1W8; Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario M5B 1W8; Departments of Laboratory Medicine and Pathobiology, Toronto, Ontario M5S 1A8, Canada; Divisions of Respirology, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
32
|
McCaig D, Lyall F. Heme oxygenase expression in human placental villous tissue in response to exposure to in vitro ischemia-reperfusion injury. Hypertens Pregnancy 2010; 28:256-72. [PMID: 19263288 DOI: 10.1080/10641950802132811] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Heme oxygenases (HO-1 and HO-2) are responsible for the production of carbon monoxide, a vasodilator. The products of heme oxygenase are also anti-oxidants. HO is expressed within the placenta and is important in controlling placental blood flow. HO can be sensitive to oxygen, with responses differing depending on the cell type. Recent studies have suggested that in preeclampsia, the placenta would be subjected to fluctuations in oxygen tension analogous to an ischemia-reperfusion injury. Thus the present study tested the hypothesis that HO-1 and or HO-2 expression in placental villous explants would be altered by an ischemic-reperfusion insult. Human term placental explants were exposed to hypoxia then re-oxygenation in 5% or 20% O(2) or repeated cycles of hypoxia-re-oxygenation. HO protein concentrations were assessed by Western blotting. No changes in HO-1 or HO-2 were found with any treatment protocol. Chemical induction of HO-1 was possible in explants showing that HO-1 induction in explants is possible. The results suggest that cells in term placental villous tissue do not respond to hypoxia-re-oxygenation by altering the amount of HO-1 or HO-2 protein.
Collapse
Affiliation(s)
- D McCaig
- Maternal and Fetal Medicine Section, Institute of Medical Genetics, Yorkhill, Glasgow, Scotland, United Kingdom
| | | |
Collapse
|
33
|
Young SC, Storm MV, Speed JS, Kelsen S, Tiller CV, Vera T, Drummond HA, Stec DE. Inhibition of biliverdin reductase increases ANG II-dependent superoxide levels in cultured renal tubular epithelial cells. Am J Physiol Regul Integr Comp Physiol 2009; 297:R1546-53. [PMID: 19759334 DOI: 10.1152/ajpregu.90933.2008] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Induction of heme oxygenase-1 (HO-1) in the renal medulla increases carbon monoxide and bilirubin production and decreases ANG II-mediated superoxide production. The goal of this study was to determine the importance of increases in bilirubin to the antioxidant effects of HO-1 induction in cultured mouse thick ascending loop of Henle (TALH) and inner medullary collecting duct (IMCD3) cells. Bilirubin levels were decreased by using small interfering RNAs (siRNAs) targeted to biliverdin reductase (BVR), which is the cellular enzyme responsible for the conversion of biliverdin to bilirubin. Treatment of cultured TALH or IMCD-3 cells with BVR siRNA (50 or 100 nM) resulted in an 80% decrease in the level of BVR protein and decreased cellular bilirubin levels from 46 +/- 5 to 23 +/- 4 nM (n = 4). We then determined the effects of inhibition of BVR on ANG II-mediated superoxide production. Superoxide production induced by ANG II (10(-9) M) significantly increased in both TALH and IMCD-3 cells. Treatment of TALH cells with BVR siRNA resulted in a significant increase in ouabain-sensitive rubidium uptake from 95 +/- 6 to 122 +/- 5% control (n = 4, P < 0.05). Lastly, inhibition of BVR with siRNA did not prevent the decrease in superoxide levels observed in cells pretreated with the HO-1 inducer, hemin. We conclude that decreased levels of cellular bilirubin increase ANG II-mediated superoxide production and sodium transport; however, increases in bilirubin are not necessary for HO-1 induction to attenuate ANG II-mediated superoxide production.
Collapse
Affiliation(s)
- Shelby C Young
- Department of Physiology and Biophysics, Center for Excellence in Cardiovascular-Renal Research, University of Mississippi Medical Center, Jackson, Mississippi 39216, USA
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
Heme oxygenase-1 (HO-1), the rate-limiting enzyme of heme degradation and antioxidant defense protein, is induced in the lungs of animals exposed to hyperoxia. However, high levels of HO-1 expression may be deleterious, thus necessitating tight regulation. Previous reports show maturational differences in rat HO-1 regulation in hyperoxia, as newborns do not up-regulate HO-1mRNA compared with adults. To better understand the differential response of lung HO-1 to hyperoxia, we exposed newborn and adult mice to >95% oxygen. The newborn lungs had reduced HO-1 mRNA induction compared with adults and newborn transgenic mice over-expressing luciferase driven by the 15 kb HO-1 promoter (HO-1/Luc Tg) had less increased light emission in hyperoxia compared with adults. Compared with adults, levels of the repressor of HO-1 transcription, Bach1, were higher in the neonatal lung as was nuclear protein-DNA binding to the antioxidant response element (ARE) from HO-1. Furthermore, at baseline and in hyperoxia, chromatin immunoprecipitation (ChIP) revealed increased Bach1 binding to the HO-1 distal enhancers (DEs) in the neonates compared with adults. These data suggest that elevated levels of Bach1 may help to limit HO-1 induction in the newborn at baseline and in response to oxidative stress.
Collapse
|
35
|
Navar LG, Arendshorst WJ, Pallone TL, Inscho EW, Imig JD, Bell PD. The Renal Microcirculation. Compr Physiol 2008. [DOI: 10.1002/cphy.cp020413] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
36
|
Lin PH, Chiang MT, Chau LY. Ubiquitin-proteasome system mediates heme oxygenase-1 degradation through endoplasmic reticulum-associated degradation pathway. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2008; 1783:1826-34. [PMID: 18544348 DOI: 10.1016/j.bbamcr.2008.05.008] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2007] [Revised: 05/13/2008] [Accepted: 05/13/2008] [Indexed: 11/15/2022]
Abstract
The present study investigated the cellular mechanism underlying the degradation of heme oxygenase-1 (HO-1), an endoplasmic reticulum (ER)-anchored protein. The turnover of HO-1 induced in vascular smooth muscle cells (VSMCs) was significantly attenuated by proteasome inhibitors, suggesting the involvement of a proteasome-mediated pathway. High molecular weight ubiquitin conjugates were co-immunoprecipitated with HO-1 from VSMCs after proteasome inhibition, and HO-1 ubiquitination was confirmed in HEK293 cells overexpressing His-tagged HO-1 and HA-tagged ubiquitin. Endogenous p97, an ATPase, and Ufd1, both implicated as essential components in the ER-associated degradation pathway (ERAD), were co-eluted with His-tagged HO-1 from metal affinity resin. Knockdown of either p97 or Ufd1 in HEK293 cells using specific siRNA significantly prolonged the half-life of endogenously induced HO-1 and slowed the degradation of ubiquitinated HO-1. HO-1 ubiquitination in HEK293 cells was enhanced by zinc chloride, but suppressed with a zinc chelator (N,N,N',N'-tetrakis(2-pyridylmethyl)ethyl-enediamine), suggesting the involvement of a RING-E3 ligase in this process. Collectively, these data indicate that HO-1 protein turnover is regulated by the ubiquitin-proteasome system through the ERAD pathway.
Collapse
Affiliation(s)
- Pu-Hua Lin
- Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | | | | |
Collapse
|
37
|
Wunderlich L, Paragh G, Wikonkál NM, Bánhegyi G, Kárpáti S, Mandl J. UVB induces a biphasic response of HIF-1α in cultured human keratinocytes. Exp Dermatol 2008; 17:335-42. [DOI: 10.1111/j.1600-0625.2007.00640.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
38
|
Li N, Chen L, Yi F, Xia M, Li PL. Salt-sensitive hypertension induced by decoy of transcription factor hypoxia-inducible factor-1alpha in the renal medulla. Circ Res 2008; 102:1101-8. [PMID: 18356541 DOI: 10.1161/circresaha.107.169201] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Hypoxia inducible factor (HIF)-1alpha, a transcription factor, is abundantly expressed in the renal medulla and regulates many oxygen-sensitive genes such as nitric oxide synthase, cyclooxygenase-2, and heme oxygenase-1. Given the important roles of these genes in the control of arterial pressure, the present study was to test the hypothesis that HIF-1alpha-mediated gene activation serves as an antihypertensive pathway by regulating renal medullary function and sodium excretion. HIF-1alpha decoy oligodeoxynucleotides (ODNs) or scrambled ODNs were transfected into the renal medulla in uninephrectomized Sprague-Dawley rats. Two weeks after ODN transfection, the HIF-1alpha binding activities were significantly inhibited by 45%, and high salt-induced increases of nitric oxide synthase-2 and heme oxygenase-1 transcriptions were also inhibited by 70% and 61% in the renal medulla from decoy rats. The natriuretic responses and increases of renal medullary blood flow responding to the elevations of renal perfusion pressure were significantly blunted by 50% and 37% in decoy rats. Intravenously acute sodium loading increased medullary blood flow and urinary sodium excretion, which was remarkably attenuated in decoy rats. In decoy rats, high salt intake caused a greater positive sodium balance. Consequently, arterial pressure was remarkably increased (from 118+/-1.9 to 154+/-6.3 mm Hg) in decoy rats but not in control rats when the rats were challenged with a high salt diet. There was no blood pressure change in decoy rats that were maintained in normal salt diet. In conclusion, HIF-1alpha-mediated gene activation importantly participates in the regulation of renal medullary function and long-term arterial blood pressure.
Collapse
Affiliation(s)
- Ningjun Li
- Department of Pharmacology & Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, PO Box 980613, Richmond, VA 23298, USA.
| | | | | | | | | |
Collapse
|
39
|
Courtney AE, Maxwell AP. Heme oxygenase 1: does it have a role in renal cytoprotection? Am J Kidney Dis 2008; 51:678-90. [PMID: 18371544 DOI: 10.1053/j.ajkd.2007.11.033] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2007] [Accepted: 11/28/2007] [Indexed: 12/13/2022]
Abstract
Heme oxygenase (HO) was first identified as the rate-limiting enzyme in the degradative pathway of heme, but is now recognized to be involved in diverse biological processes. Different isoforms of HO exist; HO-1 (HMOX1) is ubiquitously present in mammalian tissue with low constitutive expression under physiological conditions, but is upregulated in response to a variety of potentially noxious stimuli. HO-1, an integral component of an important cytoprotective mechanism, mediates its action through removal of heme, the generation of heme breakdown reaction products (biliverdin, free iron, and carbon monoxide), and modulation of key cellular molecules. Data from experimental models in which HO-1 was induced or inhibited, together with observations in genetically modified animals, showed a beneficial effect of HO-1 in several pathways leading to kidney injury. The discovery of a functional guanosine thymine tandem repeat polymorphism in the promoter region of the human HO-1 gene has stimulated clinical investigations in a variety of diseases. However, despite theoretical and experimental support for an important pathophysiological role for HO-1, the relevance of this polymorphism in native kidney or renal transplant function is equivocal. This article reviews the molecular genetics of HO-1, its myriad cytoprotective effects allied to how these are mediated, and relates these findings to experimental and clinical evidence of HO-1 involvement in renal disease.
Collapse
Affiliation(s)
- Aisling E Courtney
- Nephrology Research Group, Queen's University of Belfast, Belfast City Hospital, Belfast BT9 7AB, UK.
| | | |
Collapse
|
40
|
Navar LG, Arendshorst WJ, Pallone TL, Inscho EW, Imig JD, Bell PD. The Renal Microcirculation. Microcirculation 2008. [DOI: 10.1016/b978-0-12-374530-9.00015-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
|
41
|
Li N, Yi F, dos Santos EA, Donley DK, Li PL. Role of Renal Medullary Heme Oxygenase in the Regulation of Pressure Natriuresis and Arterial Blood Pressure. Hypertension 2007; 49:148-54. [PMID: 17075028 DOI: 10.1161/01.hyp.0000250086.06137.fb] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Recent studies have demonstrated that inhibition of renal medullary heme oxygenase (HO) activity and carbon monoxide (CO) significantly decreases renal medullary blood flow and sodium excretion. Given the crucial role of renal medullary blood flow in the control of pressure natriuresis, the present study was designed to determine whether renal medullary HO activity and resulting CO production participate in the regulation of pressure natriuresis and thereby the long-term control of arterial blood pressure. In anesthetized Sprague-Dawley rats, increases in renal perfusion pressure induced significant elevations of CO concentrations in the renal medulla. Renal medullary infusion of chromium mesoporphyrin (CrMP), an inhibitor of HO activity, remarkably inhibited HO activity and the renal perfusion pressure-dependent increases in CO levels in the renal medulla and significantly blunted pressure natriuresis. In conscious Sprague-Dawley rats, continuous infusion of CrMP into the renal medulla significantly increased mean arterial pressure (129±2.5 mm Hg in CrMP group versus 118±1.6 mm Hg in vehicle group) when animals were fed a normal salt diet (1% NaCl). After rats were switched to a high-salt diet (8% NaCl) for 10 days, CrMP-treated animals exhibited further increases in mean arterial pressure compared with CrMP-treated animals that were kept on normal salt diet (152±4.1 versus 130±4.2 mm Hg). These results suggest that renal medullary HO activity plays a crucial role in the control of pressure natriuresis and arterial blood pressure and that impairment of this HO/CO-mediated antihypertensive mechanism in the renal medulla may result in the development of hypertension.
Collapse
Affiliation(s)
- Ningjun Li
- Department of Pharmacology and Toxicology, Medical College of Virginia, Virginia Commonwealth University, Richmond 23298, USA.
| | | | | | | | | |
Collapse
|
42
|
|
43
|
Zhang G, Zhang F, Muh R, Yi F, Chalupsky K, Cai H, Li PL. Autocrine/paracrine pattern of superoxide production through NAD(P)H oxidase in coronary arterial myocytes. Am J Physiol Heart Circ Physiol 2006; 292:H483-95. [PMID: 16963617 DOI: 10.1152/ajpheart.00632.2006] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The present study tested the hypothesis that membrane-bound NAD(P)H oxidase in coronary arterial myocytes (CAMs) is capable of producing superoxide (O(2)(*-)) toward extracellular space to exert an autocrine- or paracrine-like action in these cells. Using a high-speed wavelength-switching fluorescent microscopic imaging technique, we simultaneously monitored the binding of dihydroethidium-oxidizing product to exogenous salmon testes DNA trapped outside CAMs and to nuclear DNA as indicators of extra- and intracellular O(2)(*-) production. It was found that a muscarinic agonist oxotremorine (OXO; 80 microM) increased O(2)(*-) levels more rapidly outside than inside CAMs. In the presence of superoxide dismutase (500 U/ml) plus catalase (400 U/ml) and NAD(P)H oxidase inhibitor diphenylene iodonium (50 microM) or apocynin (100 microM), these increases in extra- and intracellular O(2)(*-) levels were substantially abolished or attenuated. The O(2)(*-) increase outside CAMs was also confirmed by detecting oxidation of nitro blue tetrazolium and confocal microscopic localization of Matrigel-trapped OxyBURST H(2)HFF Green BSA staining around these cells. By electron spin resonance spectrometry, the extracellular accumulation of O(2)(*-) was demonstrated as a superoxide dismutase-sensitive component outside CAMs. Furthermore, RNA interference of NAD(P)H oxidase subunits Nox1 or p47 markedly blocked OXO-induced increases in both extra- and intracellular O(2)(*-) levels, whereas small inhibitory RNA of Nox4 only attenuated intracellular O(2)(*-) accumulation. These results suggest that Nox1 represents a major NAD(P)H oxidase isoform responsible for extracellular O(2)(*-) production. This rapid extracellular production of O(2)(*-) seems to be unique to OXO-induced M(1)-receptor activation, since ANG II-induced intra- and extracellular O(2)(*-) increases in parallel. It is concluded that the outward production of O(2)(*-) via NAD(P)H oxidase in CAMs may represent an important producing pattern for its autocrine or paracrine actions.
Collapse
Affiliation(s)
- Guo Zhang
- Dept. of Pharmacology and Toxicology, Medical College of Virginia, 410 North 12th St., Richmond, VA 23298, USA
| | | | | | | | | | | | | |
Collapse
|
44
|
Mense SM, Zhang L. Heme: a versatile signaling molecule controlling the activities of diverse regulators ranging from transcription factors to MAP kinases. Cell Res 2006; 16:681-92. [PMID: 16894358 DOI: 10.1038/sj.cr.7310086] [Citation(s) in RCA: 200] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Heme (iron protoporphyrin IX) is an essential molecule for numerous living organisms. Not only does it serve as a prosthetic group in enzymes, it also acts as a signaling molecule that controls diverse molecular and cellular processes ranging from signal transduction to protein complex assembly. Deficient heme synthesis or function impacts the hematopoietic, hepatic and nervous systems in humans. Recent studies have revealed a series of heme-regulated transcription factors and signal transducers including Hap1, a heme-activated transcription factor that mediates the effects of oxygen on gene transcription in the yeast Saccharomyces cerevisiae; Bach1, a transcriptional repressor that is negatively regulated by heme in mammalian cells; IRR, an iron regulatory protein that mediates the iron-dependant regulation of heme synthesis in the bacterium Bradyrhizobium japonicum; and heme-regulated inhibitor, an eucaryotic initiation factor 2alpha kinase that coordinates protein synthesis with heme availability in reticulocytes. In this review, we summarize the current knowledge about how heme controls the activity of these transcriptional regulators and signal transducers, and discuss diseases associated with defective heme synthesis, degradation and function.
Collapse
Affiliation(s)
- Sarah M Mense
- Department of Environmental Health Sciences, Columbia University, Mailman School of Public Health, New York, NY 10032, USA
| | | |
Collapse
|
45
|
Li N, Yi F, Sundy CM, Chen L, Hilliker ML, Donley DK, Muldoon DB, Li PL. Expression and actions of HIF prolyl-4-hydroxylase in the rat kidneys. Am J Physiol Renal Physiol 2006; 292:F207-16. [PMID: 16885149 DOI: 10.1152/ajprenal.00457.2005] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Hypoxia inducible factor (HIF) prolyl-4-hydroxylase domain-containing proteins (PHDs) promote the degradation of HIF-1alpha. Because HIF-1alpha is highly expressed in the renal medulla and HIF-1alpha-targeted genes such as nitric oxide synthase, cyclooxygenase, and heme oxygenase are important in the regulation of renal medullary function, we hypothesized that PHD regulates HIF-1alpha levels in the renal medulla and, thereby, participates in the control of renal Na(+) excretion. Using real-time RT-PCR, Western blot, and immunohistochemical analyses, we have demonstrated that all three isoforms of PHD, PHD1, PHD2, and PHD3, are expressed in the kidneys and that PHD2 is the most abundant isoform. Regionally, all PHDs exhibited much higher levels in renal medulla than cortex. A furosemide-induced increase in renal medullary tissue Po(2) significantly decreased PHD levels in renal medulla, whereas hypoxia significantly increased mRNA levels of PHDs in cultured renal medullary interstitial cells, indicating that O(2) regulates PHDs. Functionally, the PHD inhibitor l-mimosine (l-Mim, 50 mg x kg(-1) x day(-1) i.p. for 2 wk) substantially upregulated HIF-1alpha expression in the kidneys, especially in the renal medulla, and remarkably enhanced (by >80%) the natriuretic response to renal perfusion pressure in Sprague-Dawley rats. Inhibition of HIF transcriptional activity by renal medullary transfection of HIF-1alpha decoy oligodeoxynucleotides attenuated l-Mim-induced enhancement of pressure natriuresis, which confirmed that HIF-1alpha mediated the effect of l-Mim. These results indicate that highly expressed PHDs in the renal medulla make an important contribution to the control of renal Na(+) excretion through regulation of HIF-1alpha and its targeted genes.
Collapse
Affiliation(s)
- Ningjun Li
- Dept. of Pharmacology & Toxicology, Medical College of Virginia Campus, Virginia Commonwealth Univ., PO Box 980613, Richmond, VA 23298, USA.
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Neuhofer W, Beck FX. Survival in Hostile Environments: Strategies of Renal Medullary Cells. Physiology (Bethesda) 2006; 21:171-80. [PMID: 16714475 DOI: 10.1152/physiol.00003.2006] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Cells in the renal medulla exist in a hostile milieu characterized by wide variations in extracellular solute concentrations, low oxygen tensions, and abundant reactive oxygen species. This article reviews the strategies adopted by these cells to allow them to survive and fulfill their functions under these extreme conditions.
Collapse
|
47
|
Mense SM, Sengupta A, Zhou M, Lan C, Bentsman G, Volsky DJ, Zhang L. Gene expression profiling reveals the profound upregulation of hypoxia-responsive genes in primary human astrocytes. Physiol Genomics 2006; 25:435-49. [PMID: 16507782 DOI: 10.1152/physiolgenomics.00315.2005] [Citation(s) in RCA: 108] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Oxygen is vital for the development and survival of mammals. In response to hypoxia, the brain initiates numerous adaptive responses at the organ level as well as at the molecular and cellular levels, including the alteration of gene expression. Astrocytes play critical roles in the proper functioning of the brain; thus the manner in which astrocytes respond to hypoxia is likely important in determining the outcome of brain hypoxia. Here, we used microarray gene expression profiling and data-analysis algorithms to identify and analyze hypoxia-responsive genes in primary human astrocytes. We also compared gene expression patterns in astrocytes with those in human HeLa cells and pulmonary artery endothelial cells (ECs). Remarkably, in astrocytes, five times as many genes were induced as suppressed, whereas in HeLa and pulmonary ECs, as many as or more genes were suppressed than induced. More genes encoding hypoxia-inducible functions, such as glycolytic enzymes and angiogenic growth factors, were strongly induced in astrocytes compared with HeLa cells. Furthermore, gene ontology and computational algorithms revealed that many target genes of the EGF and insulin signaling pathways and the transcriptional regulators Myc, Jun, and p53 were selectively altered by hypoxia in astrocytes. Indeed, Western blot analysis confirmed that two major signal transducers mediating insulin and EGF action, Akt and MEK1/2, were activated by hypoxia in astrocytes. These results provide a global view of the signaling and regulatory network mediating oxygen regulation in human astrocytes.
Collapse
Affiliation(s)
- S M Mense
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York 10032, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Harrison EM, McNally SJ, Devey L, Garden OJ, Ross JA, Wigmore SJ. Insulin induces heme oxygenase-1 through the phosphatidylinositol 3-kinase/Akt pathway and the Nrf2 transcription factor in renal cells. FEBS J 2006; 273:2345-56. [PMID: 16704410 DOI: 10.1111/j.1742-4658.2006.05224.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Heme oxygenase-1 catalyzes the breakdown of heme and is protective in models of kidney transplantation. In this study we describe the induction of heme oxygenase-1 mRNA and protein by insulin. Following treatment with insulin, a five-fold increase in heme oxygenase-1 mRNA and a four-fold increase in protein expression were observed in renal adenocarcinoma cells; insulin-induced heme oxygenase-1 expression was also demonstrated in mouse primary tubular epithelial cells. The induction of heme oxygenase-1 in renal adenocarcinoma cells was blocked by actinomycin D and cycloheximide and was abolished by the phosphatidylinositol 3-kinase inhibitor, LY294002, but not by the inactive analog LY303511. Overexpressing a dominant-negative form of Akt abrogated the heme oxygenase-1-inducing effects of insulin, whereas cells transfected with a constitutively active Akt construct demonstrated an increase in heme oxygenase-1 promoter activity and protein expression. The transcription factor NF-E2-related factor-2 was found to translocate to the nucleus following insulin treatment in a phosphatidylinositol 3-kinase-dependent manner. Pretreatment with NF-E2-related factor-2 small-interfering RNA abolished insulin-induced heme oxygenase-1 induction. Insulin was also found to activate the mitogen-activated protein kinase cascades p38 and extracellular signal-related kinase; however, inhibition of these pathways with SB202190 and PD98059 did not alter insulin-induced heme oxygenase-1 expression. Thus, insulin induces heme oxygenase-1 mRNA and protein expression in renal cells in a phosphatidylinositol 3-kinase/Akt and NF-E2-related factor-2-dependent manner.
Collapse
Affiliation(s)
- Ewen M Harrison
- Tissue Injury and Repair Group, University of Edinburgh, UK.
| | | | | | | | | | | |
Collapse
|
49
|
Li N, Chen L, Muh RW, Li PL. Hyperhomocysteinemia associated with decreased renal transsulfuration activity in Dahl S rats. Hypertension 2006; 47:1094-100. [PMID: 16636197 DOI: 10.1161/01.hyp.0000219634.83928.6e] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Elevated plasma homocysteine (Hcys) has been reported to participate in the development of arterial and glomerular sclerosis in Dahl salt-sensitive hypertensive (SS) rats. The mechanism resulting in hyperhomocysteinemia in these animals remains unknown. Disposal of Hcys in the kidneys plays an important role in regulating the plasma Hcys level. We, therefore, examined the activities and expressions of the enzymes involved in the metabolism of Hcys in the kidneys of SS rats, compared with that of Brown Norway rats and SSBN13 rats, a consomic subcolony of SS rats that carries a substituted chromosome 13 from Brown Norway rats. High-performance liquid chromatography analysis demonstrated that plasma Hcys levels were significantly higher in SS rats. The conversion of S-adenosylhomocysteine into Hcys via S-adenosylhomocysteine hydrolase by renal tissue was not different among these 3 rat strains. However, the metabolic rate of Hcys into cysteine was markedly reduced in the SS rat kidneys. The mRNA and protein levels of cystathionine beta-synthase (CBS), one of the key enzymes in the transsulfuration pathway in the kidneys, were significantly lower in SS rats. In microdissected nephron segments, CBS mRNA was shown to be mainly present in renal proximal tubules (PTs). The mRNA levels of CBS in the PTs were also significantly decreased in SS rats, accompanied by a reduced CBS activity in PTs. We conclude that hyperhomocysteinemia is associated with a decreased activity and expression of CBS in renal PTs because of the defect of chromosome 13 in SS rats.
Collapse
Affiliation(s)
- Ningjun Li
- Department of Pharmacology and Toxicology, Medical College of Virginia, Virginia Commonwealth University, Richmond, VA 23298, USA.
| | | | | | | |
Collapse
|
50
|
Fleury C, Petit A, Mwale F, Antoniou J, Zukor DJ, Tabrizian M, Huk OL. Effect of cobalt and chromium ions on human MG-63 osteoblasts in vitro: morphology, cytotoxicity, and oxidative stress. Biomaterials 2006; 27:3351-60. [PMID: 16488005 DOI: 10.1016/j.biomaterials.2006.01.035] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2005] [Accepted: 01/24/2006] [Indexed: 10/25/2022]
Abstract
Recent studies demonstrated that Co(2+) and Cr(3+) ions induced cell mortality, TNF-alpha secretion, and oxidation of proteins in macrophages. However, little is known about the effects of corrosion products on the osteogenic cells, which have a crucial role in controlling bone remodeling. The aim of the present study was to investigate the effect of Co(2+) (0-10 ppm) and Cr(3+) (0-150 ppm) on human MG-63 osteoblast-like cells in term of cytotoxicity and oxidative stress. Microscopic analysis demonstrated changes in shape, size, and number of cells. Co(2+) had a greater effect on these parameters than Cr(3+). Cell counting showed a significant decrease in the number of MG-63 osteoblasts in a time- and dose-dependent manner, with Co(2+) more toxic than Cr(3+). The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) analysis also showed a decreased cellular activity in presence of Co(2+) and Cr(3+) ions. Oxidized and nitrated proteins, two markers of oxidative stress, were detected as single bands and revealed time- and dose-dependent protein modifications. We also studied the expression of three antioxidant enzymes. The expression of heme oxygenase-1 was increased by both ions after 24h, before decreasing gradually thereafter. Glutathione peroxidase expression was also increased in a concentration- and time-dependent manner by both Co(2+) and Cr(3+) ions. Co(2+) decreased catalase expression while Cr(3+) increased it in a dose- and time-dependent manner. In conclusion, this study demonstrated that Cr(3+) and Co(2+) have a cytotoxic effect on MG-63 osteoblasts and have the potential to modify their redox state.
Collapse
Affiliation(s)
- Cyrille Fleury
- Division of Orthopaedic Surgery, McGill University, Lady Davis Institute for Medical Research, The Sir Mortimer B. Davis-Jewish General Hospital, 3775 Chemin de la Côte Sainte Catherine, Montreal, Que., Canada H3 T 1E2
| | | | | | | | | | | | | |
Collapse
|