1
|
Gurol KC, Jursa T, Cho EJ, Fast W, Dalby KN, Smith DR, Mukhopadhyay S. PHD2 enzyme is an intracellular manganese sensor that initiates the homeostatic response against elevated manganese. Proc Natl Acad Sci U S A 2024; 121:e2402538121. [PMID: 38905240 PMCID: PMC11214094 DOI: 10.1073/pnas.2402538121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 05/24/2024] [Indexed: 06/23/2024] Open
Abstract
Intracellular sensors detect changes in levels of essential metals to initiate homeostatic responses. But, a mammalian manganese (Mn) sensor is unknown, representing a major gap in understanding of Mn homeostasis. Using human-relevant models, we recently reported that: 1) the primary homeostatic response to elevated Mn is upregulation of hypoxia-inducible factors (HIFs), which increases expression of the Mn efflux transporter SLC30A10; and 2) elevated Mn blocks the prolyl hydroxylation of HIFs by prolyl hydroxylase domain (PHD) enzymes, which otherwise targets HIFs for degradation. Thus, the mammalian mechanism for sensing elevated Mn likely relates to PHD inhibition. Moreover, 1) Mn substitutes for a catalytic iron (Fe) in PHD structures; and 2) exchangeable cellular levels of Fe and Mn are comparable. Therefore, we hypothesized that elevated Mn directly inhibits PHD by replacing its catalytic Fe. In vitro assays using catalytically active PHD2, the primary PHD isoform, revealed that Mn inhibited, and Fe supplementation rescued, PHD2 activity. However, a mutation in PHD2 (D315E) that selectively reduced Mn binding without substantially impacting Fe binding or enzymatic activity resulted in complete insensitivity of PHD2 to Mn in vitro. Additionally, hepatic cells expressing full-length PHD2D315E were less sensitive to Mn-induced HIF activation and SLC30A10 upregulation than PHD2wild-type. These results: 1) define a fundamental Mn sensing mechanism for controlling Mn homeostasis-elevated Mn inhibits PHD2, which functions as a Mn sensor, by outcompeting its catalytic Fe, and PHD2 inhibition activates HIF signaling to up-regulate SLC30A10; and 2) identify a unique mode of metal sensing that may have wide applicability.
Collapse
Affiliation(s)
- Kerem C. Gurol
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX78712
| | - Thomas Jursa
- Department of Microbiology and Environmental Toxicology, University of California at Santa Cruz, Santa Cruz, CA95064
| | - Eun Jeong Cho
- College of Pharmacy, Targeted Therapeutic Drug Discovery and Development Program, The University of Texas at Austin, Austin, TX78712
| | - Walter Fast
- Division of Chemical Biology and Drug Discovery, College of Pharmacy, The University of Texas at Austin, Austin, TX78712
| | - Kevin N. Dalby
- College of Pharmacy, Targeted Therapeutic Drug Discovery and Development Program, The University of Texas at Austin, Austin, TX78712
- Division of Chemical Biology and Drug Discovery, College of Pharmacy, The University of Texas at Austin, Austin, TX78712
| | - Donald R. Smith
- Department of Microbiology and Environmental Toxicology, University of California at Santa Cruz, Santa Cruz, CA95064
| | - Somshuvra Mukhopadhyay
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX78712
| |
Collapse
|
2
|
Kong G, Hua H, Lu Y, Yan K, Zheng Y, Jia Z, Guo H, Li M, Jin Y, Liu Z. Roxadustat ameliorates experimental colitis in mice by regulating macrophage polarization through increasing HIF level. Biochim Biophys Acta Gen Subj 2024; 1868:130548. [PMID: 38158022 DOI: 10.1016/j.bbagen.2023.130548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 11/10/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024]
Abstract
BACKGROUND Gastro-intestinal (GI) tract inflammation is as a result of inflammatory hypoxia which is also induced by long-standing group of disorders like inflammatory-bowel disease (IBD). Regulation of GI immune homeostasis by macrophage involves hypoxia-inducible factor (HIF). As inhibitor of HIF prolyl hydroxylase, roxadustat (ROX) increases the levels of HIF. METHODS We induced experimental colitis (EC) model in mice via dextran-sulfate sodium (DSS) to evaluate ROX role in above-mentioned disease. RESULTS ROX ameliorated EC in mice by blocking colonic length shorten and loss of body weight, thereby reducing scores of disease-activity index (DAI) and histopathology. ROX significantly reduced inflammatory cytokines levels, suppressed M1 and increased M2 macrophage polarization in colonic tissues. Besides, ROX blocked declining hematocrit (HCT) level in blood and increased HIF-1-α and HIF-2-α level in colonic tissues. The inhibitor of HIF-1- α, KC7F2 decreased body weight and colonic length in ROX-treated DSS mice. Meanwhile, DAI scores and histopathology in KC7F2 treated DSS mice were markedly higher than that of treatment with ROX alone. KC7F2 treatments also significantly increased inflammatory cytokines levels, respectively promoted and reduced polarization of M1 and M2 macrophages in colonic tissue from ROX treated mice. Further, KC7F2 treatments inhibited ROX induced HCT level increasing in blood and decreased HIF-1-α and HIF-2-α level in colonic tissue. CONCLUSION Collectively, we discovered that ROX ameliorated EC in mice by regulating macrophage polarization through promotion of HIF expression. GENERAL SIGNIFICANCE Taken together, we developed a new application of ROX, which provides new ideas and a scientific basis for IBD treatment.
Collapse
Affiliation(s)
- Guiping Kong
- Department of Gastroenterology, Children's Hospital of Nanjing Medical University, 210008 Nanjing, Jiangsu, China
| | - Hu Hua
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, 210008 Nanjing, Jiangsu, China; Department of Nephrology, Children's Hospital of Nanjing Medical University, 210008 Nanjing, Jiangsu, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, 211166 Nanjing, Jiangsu, China
| | - Yan Lu
- Department of Gastroenterology, Children's Hospital of Nanjing Medical University, 210008 Nanjing, Jiangsu, China
| | - Kunlong Yan
- Department of Gastroenterology, Children's Hospital of Nanjing Medical University, 210008 Nanjing, Jiangsu, China
| | - Yucan Zheng
- Department of Gastroenterology, Children's Hospital of Nanjing Medical University, 210008 Nanjing, Jiangsu, China
| | - Zhanjun Jia
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, 210008 Nanjing, Jiangsu, China; Department of Nephrology, Children's Hospital of Nanjing Medical University, 210008 Nanjing, Jiangsu, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, 211166 Nanjing, Jiangsu, China
| | - Hongmei Guo
- Department of Gastroenterology, Children's Hospital of Nanjing Medical University, 210008 Nanjing, Jiangsu, China
| | - Mei Li
- Department of Gastroenterology, Children's Hospital of Nanjing Medical University, 210008 Nanjing, Jiangsu, China
| | - Yu Jin
- Department of Gastroenterology, Children's Hospital of Nanjing Medical University, 210008 Nanjing, Jiangsu, China
| | - Zhifeng Liu
- Department of Gastroenterology, Children's Hospital of Nanjing Medical University, 210008 Nanjing, Jiangsu, China.
| |
Collapse
|
3
|
Caballano-Infantes E, Cahuana GM, Bedoya FJ, Salguero-Aranda C, Tejedo JR. The Role of Nitric Oxide in Stem Cell Biology. Antioxidants (Basel) 2022; 11:497. [PMID: 35326146 PMCID: PMC8944807 DOI: 10.3390/antiox11030497] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 02/24/2022] [Accepted: 02/25/2022] [Indexed: 02/07/2023] Open
Abstract
Nitric oxide (NO) is a gaseous biomolecule endogenously synthesized with an essential role in embryonic development and several physiological functions, such as regulating mitochondrial respiration and modulation of the immune response. The dual role of NO in embryonic stem cells (ESCs) has been previously reported, preserving pluripotency and cell survival or inducing differentiation with a dose-dependent pattern. In this line, high doses of NO have been used in vitro cultures to induce focused differentiation toward different cell lineages being a key molecule in the regenerative medicine field. Moreover, optimal conditions to promote pluripotency in vitro are essential for their use in advanced therapies. In this sense, the molecular mechanisms underlying stemness regulation by NO have been studied intensively over the current years. Recently, we have reported the role of low NO as a hypoxia-like inducer in pluripotent stem cells (PSCs), which supports using this molecule to maintain pluripotency under normoxic conditions. In this review, we stress the role of NO levels on stem cells (SCs) fate as a new approach for potential cell therapy strategies. Furthermore, we highlight the recent uses of NO in regenerative medicine due to their properties regulating SCs biology.
Collapse
Affiliation(s)
- Estefanía Caballano-Infantes
- Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, 41013 Seville, Spain; (G.M.C.); (F.J.B.)
- Department of Regeneration and Cell Therapy, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Seville-CSIC, 41092 Seville, Spain
| | - Gladys Margot Cahuana
- Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, 41013 Seville, Spain; (G.M.C.); (F.J.B.)
- Biomedical Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Francisco Javier Bedoya
- Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, 41013 Seville, Spain; (G.M.C.); (F.J.B.)
- Biomedical Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Carmen Salguero-Aranda
- Department of Pathology, Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital, CSIC-University of Seville, 41013 Seville, Spain;
- Spanish Biomedical Research Network Centre in Oncology-CIBERONC, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Normal and Pathological Cytology and Histology, School of Medicine, University of Seville, 41004 Seville, Spain
| | - Juan R. Tejedo
- Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, 41013 Seville, Spain; (G.M.C.); (F.J.B.)
- Biomedical Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM, Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
4
|
Caballano-Infantes E, Díaz I, Hitos AB, Cahuana GM, Martínez-Ruiz A, Soria-Juan B, Rodríguez-Griñolo R, Hmadcha A, Martín F, Soria B, Tejedo JR, Bedoya FJ. Stemness of Human Pluripotent Cells: Hypoxia-Like Response Induced by Low Nitric Oxide. Antioxidants (Basel) 2021; 10:antiox10091408. [PMID: 34573040 PMCID: PMC8472328 DOI: 10.3390/antiox10091408] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 08/20/2021] [Accepted: 08/27/2021] [Indexed: 12/20/2022] Open
Abstract
The optimization of conditions to promote the stemness of pluripotent cells in vitro is instrumental for their use in advanced therapies. We show here that exposure of human iPSCs and human ESCs to low concentrations of the chemical NO donor DETA/NO leads to stabilization of hypoxia-inducible factors (HIF-1α and HIF-2α) under normoxia, with this effect being dependent on diminished Pro 402 hydroxylation and decreased degradation by the proteasome. Moreover, the master genes of pluripotency, NANOG and OCT-4, were upregulated. NO also induces a shift in the metabolic profile of PSCs, with an increased expression of hypoxia response genes in glycolysis. Furthermore, a reduction in the mitochondrial membrane potential with lower oxygen consumption and increased expression of mitochondrial fusion regulators, such as DRP1, was observed. The results reported here indicate that NO mimics hypoxia response in human PSCs and enhances their stemness properties when cultured under normoxic conditions.
Collapse
Affiliation(s)
- Estefanía Caballano-Infantes
- Department of Regeneration and Cell Therapy, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Seville-CSIC, 41013 Seville, Spain; (I.D.); (A.B.H.); (A.H.); (F.M.); (J.R.T.)
- Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, 41013 Seville, Spain;
- Correspondence: (E.C.-I.); (F.J.B.)
| | - Irene Díaz
- Department of Regeneration and Cell Therapy, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Seville-CSIC, 41013 Seville, Spain; (I.D.); (A.B.H.); (A.H.); (F.M.); (J.R.T.)
- Biomedical Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM, Instituto de Salud Carlos III, 08036 Madrid, Spain;
| | - Ana Belén Hitos
- Department of Regeneration and Cell Therapy, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Seville-CSIC, 41013 Seville, Spain; (I.D.); (A.B.H.); (A.H.); (F.M.); (J.R.T.)
- Biomedical Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM, Instituto de Salud Carlos III, 08036 Madrid, Spain;
| | - Gladys Margot Cahuana
- Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, 41013 Seville, Spain;
- Biomedical Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM, Instituto de Salud Carlos III, 08036 Madrid, Spain;
| | - Antonio Martínez-Ruiz
- Unidad de Investigación, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria Princesa (IIS-IP), E-28009 Madrid, Spain;
| | | | - Rosario Rodríguez-Griñolo
- Departamento de Economía, Métodos Cuantitativo e Historia Económica, Universidad Pablo de Olavide, 41013 Seville, Spain;
| | - Abdelkrim Hmadcha
- Department of Regeneration and Cell Therapy, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Seville-CSIC, 41013 Seville, Spain; (I.D.); (A.B.H.); (A.H.); (F.M.); (J.R.T.)
- Biomedical Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM, Instituto de Salud Carlos III, 08036 Madrid, Spain;
| | - Franz Martín
- Department of Regeneration and Cell Therapy, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Seville-CSIC, 41013 Seville, Spain; (I.D.); (A.B.H.); (A.H.); (F.M.); (J.R.T.)
- Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, 41013 Seville, Spain;
- Biomedical Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM, Instituto de Salud Carlos III, 08036 Madrid, Spain;
| | - Bernat Soria
- Biomedical Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM, Instituto de Salud Carlos III, 08036 Madrid, Spain;
- ISABIAL and Institute of Bioengineering, University Miguel Hernández de Elche, 03010 Alicante, Spain
| | - Juan R. Tejedo
- Department of Regeneration and Cell Therapy, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Seville-CSIC, 41013 Seville, Spain; (I.D.); (A.B.H.); (A.H.); (F.M.); (J.R.T.)
- Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, 41013 Seville, Spain;
- Biomedical Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM, Instituto de Salud Carlos III, 08036 Madrid, Spain;
| | - Francisco Javier Bedoya
- Department of Regeneration and Cell Therapy, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Seville-CSIC, 41013 Seville, Spain; (I.D.); (A.B.H.); (A.H.); (F.M.); (J.R.T.)
- Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, 41013 Seville, Spain;
- Biomedical Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM, Instituto de Salud Carlos III, 08036 Madrid, Spain;
- Correspondence: (E.C.-I.); (F.J.B.)
| |
Collapse
|
5
|
Khan H, Anshu A, Prasad A, Roy S, Jeffery J, Kittipongdaja W, Yang DT, Schieke SM. Metabolic Rewiring in Response to Biguanides Is Mediated by mROS/HIF-1a in Malignant Lymphocytes. Cell Rep 2020; 29:3009-3018.e4. [PMID: 31801069 DOI: 10.1016/j.celrep.2019.11.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 10/11/2019] [Accepted: 11/04/2019] [Indexed: 12/20/2022] Open
Abstract
Metabolic flexibility allows cells to adapt to various environments and limits the efficacy of metabolic drugs. Therapeutic targeting of cancer metabolism relies on defining limiting requirements and vulnerabilities in the highly dynamic metabolic network. Here, we characterize the metabolic reprogramming and identify cancer-specific metabolic vulnerabilities in response to the pharmacological inhibition of mitochondrial complex I. Our work reveals the adaptation mechanism in malignant lymphocytes providing resistance against the biguanides phenformin and metformin by transcriptionally reprogramming glucose metabolism. Metabolic adaptation to complex I inhibition is mediated by mitochondrial reactive oxygen species (mROS) serving as a mitochondrial stress signal activating hypoxia-inducible factor-1a (HIF-1a). Inhibition of the mROS/HIF-1a axis through antioxidants or direct suppression of HIF-1a selectively disrupts metabolic adaptation and survival during complex I dysfunction in malignant lymphocytes. Our results identify HIF-1a signaling as a critical factor in resistance against biguanide-induced mitochondrial dysfunction, allowing selective targeting of metabolic pathways in leukemia and lymphoma.
Collapse
Affiliation(s)
- Hamidullah Khan
- Department of Dermatology, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Ashish Anshu
- Department of Dermatology, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Aman Prasad
- Department of Dermatology, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Sushmita Roy
- Department of Dermatology, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Justin Jeffery
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | | | - David T Yang
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Stefan M Schieke
- Department of Dermatology, University of Wisconsin-Madison, Madison, WI 53706, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA.
| |
Collapse
|
6
|
Han F, Wu G, Han S, Li Z, Jia Y, Bai L, Li X, Wang K, Yang F, Zhang J, Wang X, Guan H, Linlin S, Han J, Hu D. Hypoxia-inducible factor prolyl-hydroxylase inhibitor roxadustat (FG-4592) alleviates sepsis-induced acute lung injury. Respir Physiol Neurobiol 2020; 281:103506. [PMID: 32726645 DOI: 10.1016/j.resp.2020.103506] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 07/07/2020] [Accepted: 07/19/2020] [Indexed: 12/17/2022]
Abstract
Acute lung injury (ALI) is one of the most severe outcomes of sepsis which still waiting for effective treatment method. Roxadustat (FG-4592) which is often used for treatment of anemia in patients with chronic kidney disease (CKD), its affection on LPS-induced ALI haven't been evaluated. MH-S and MLE-12 cell injury and ALI mouse model was induced LPS. Several assays were used to explore the role of FG-4592 in reducing the damage caused by LPS. FG-4592 treatment significantly upregulated HIF-1α and HO-1 and strikingly attenuated inflammation in vivo and in vitro. Furthermore, septic mice overexpressing HIF-1α had high level of survival rate and lower expression of inflammatory factors while down-regulation can enhance the damage of LPS. HIF-1α has a protective effect on acute lung injury in LPS induced septic mice. FG-4592 treatment remarkably ameliorated the LPS-induced lung injury through the stabilization of HIF-1α. Besides the role in treating CKD anemia, the clinical use of FG-4592 also might be extended to ALI.
Collapse
Affiliation(s)
- Fu Han
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, China
| | - Gaofeng Wu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, China
| | - Shichao Han
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, China
| | - Zhenzhen Li
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, China
| | - Yanhui Jia
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, China
| | - Lu Bai
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, China
| | - Xiaoqiang Li
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, China
| | - Kejia Wang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, China
| | - Fangfang Yang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, China
| | - Jian Zhang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, China
| | - Xujie Wang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, China
| | - Hao Guan
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, China
| | - Su Linlin
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, China
| | - Juntao Han
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, China.
| | - Dahai Hu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, China.
| |
Collapse
|
7
|
Sadaghianloo N, Contenti J, Dardik A, Mazure NM. Role of Hypoxia and Metabolism in the Development of Neointimal Hyperplasia in Arteriovenous Fistulas. Int J Mol Sci 2019; 20:ijms20215387. [PMID: 31671790 PMCID: PMC6862436 DOI: 10.3390/ijms20215387] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 10/24/2019] [Accepted: 10/25/2019] [Indexed: 12/11/2022] Open
Abstract
For patients with end-stage renal disease requiring hemodialysis, their vascular access is both their lifeline and their Achilles heel. Despite being recommended as primary vascular access, the arteriovenous fistula (AVF) shows sub-optimal results, with about 50% of patients needing a revision during the year following creation. After the AVF is created, the venous wall must adapt to new environment. While hemodynamic changes are responsible for the adaptation of the extracellular matrix and activation of the endothelium, surgical dissection and mobilization of the vein disrupt the vasa vasorum, causing wall ischemia and oxidative stress. As a consequence, migration and proliferation of vascular cells participate in venous wall thickening by a mechanism of neointimal hyperplasia (NH). When aggressive, NH causes stenosis and AVF dysfunction. In this review we show how hypoxia, metabolism, and flow parameters are intricate mechanisms responsible for the development of NH and stenosis during AVF maturation.
Collapse
Affiliation(s)
- Nirvana Sadaghianloo
- Centre de Méditerranéen de Médecine Moléculaire (C3M), Université Côte d'Azur, INSERM U1065, 151 Route de St Antoine de Ginestière, BP2 3194, 06204 Nice CEDEX 03, France.
- Department of Vascular Surgery, Centre Hospitalier Universitaire de Nice, 06000 Nice, France.
| | - Julie Contenti
- Centre de Méditerranéen de Médecine Moléculaire (C3M), Université Côte d'Azur, INSERM U1065, 151 Route de St Antoine de Ginestière, BP2 3194, 06204 Nice CEDEX 03, France.
- Department of Emergency Medicine, Centre Hospitalier Universitaire de Nice, 06000 Nice, France.
| | - Alan Dardik
- Department of Surgery and the Vascular Biology and Therapeutics Program, Yale University, New Haven, CT 06520, USA.
- Department of Surgery, VA Connecticut Healthcare Systems, West Haven, CT 06516, USA.
| | - Nathalie M Mazure
- Centre de Méditerranéen de Médecine Moléculaire (C3M), Université Côte d'Azur, INSERM U1065, 151 Route de St Antoine de Ginestière, BP2 3194, 06204 Nice CEDEX 03, France.
- Department of Vascular Surgery, Centre Hospitalier Universitaire de Nice, 06000 Nice, France.
| |
Collapse
|
8
|
Abstract
Immune cell populations determine the balance between ongoing damage and repair following tissue injury. Cells responding to a tissue-damaged environment have significant bioenergetic and biosynthetic needs. In addition to supporting these needs, metabolic pathways govern the function of pro-repair immune cells, including regulatory T cells and tissue macrophages. In this Review, we explore how specific features of the tissue-damaged environment such as hypoxia, oxidative stress, and nutrient depletion serve as metabolic cues to promote or impair the reparative functions of immune cell populations. Hypoxia, mitochondrial DNA stress, and altered redox balance each contribute to mechanisms regulating the response to tissue damage. For example, hypoxia induces changes in regulatory T cell and macrophage metabolic profiles, including generation of 2-hydroxyglutarate, which inhibits demethylase reactions to modulate cell fate and function. Reactive oxygen species abundant in oxidative environments cause damage to mitochondrial DNA, initiating signaling pathways that likewise control pro-repair cell function. Nutrient depletion following tissue damage also affects pro-repair cell function through metabolic signaling pathways, specifically those sensitive to the redox state of the cell. The study of immunometabolism as an immediate sensor and regulator of the tissue-damaged environment provides opportunities to consider mechanisms that facilitate healthy repair of tissue injury.
Collapse
|
9
|
Sadiku P, Walmsley SR. Hypoxia and the regulation of myeloid cell metabolic imprinting: consequences for the inflammatory response. EMBO Rep 2019; 20:embr.201847388. [PMID: 30872317 PMCID: PMC6500960 DOI: 10.15252/embr.201847388] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 12/21/2018] [Accepted: 02/20/2019] [Indexed: 12/19/2022] Open
Abstract
Inflamed and infected tissue sites are characterised by oxygen and nutrient deprivation. The cellular adaptations to insufficient oxygenation, hypoxia, are mainly regulated by a family of transcription factors known as hypoxia-inducible factors (HIFs). The protein members of the HIF signalling pathway are critical regulators of both the innate and adaptive immune responses, and there is an increasing body of evidence to suggest that the elicited changes occur through cellular metabolic reprogramming. Here, we review the literature on innate immunometabolism to date and discuss the role of hypoxia in innate cell metabolic reprogramming, and how this determines immune responses.
Collapse
Affiliation(s)
- Pranvera Sadiku
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Sarah R Walmsley
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
10
|
Keeley TP, Mann GE. Defining Physiological Normoxia for Improved Translation of Cell Physiology to Animal Models and Humans. Physiol Rev 2019; 99:161-234. [PMID: 30354965 DOI: 10.1152/physrev.00041.2017] [Citation(s) in RCA: 216] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The extensive oxygen gradient between the air we breathe (Po2 ~21 kPa) and its ultimate distribution within mitochondria (as low as ~0.5-1 kPa) is testament to the efforts expended in limiting its inherent toxicity. It has long been recognized that cell culture undertaken under room air conditions falls short of replicating this protection in vitro. Despite this, difficulty in accurately determining the appropriate O2 levels in which to culture cells, coupled with a lack of the technology to replicate and maintain a physiological O2 environment in vitro, has hindered addressing this issue thus far. In this review, we aim to address the current understanding of tissue Po2 distribution in vivo and summarize the attempts made to replicate these conditions in vitro. The state-of-the-art techniques employed to accurately determine O2 levels, as well as the issues associated with reproducing physiological O2 levels in vitro, are also critically reviewed. We aim to provide the framework for researchers to undertake cell culture under O2 levels relevant to specific tissues and organs. We envisage that this review will facilitate a paradigm shift, enabling translation of findings under physiological conditions in vitro to disease pathology and the design of novel therapeutics.
Collapse
Affiliation(s)
- Thomas P Keeley
- King's British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, Faculty of Life Sciences and Medicine, King's College London , London , United Kingdom
| | - Giovanni E Mann
- King's British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, Faculty of Life Sciences and Medicine, King's College London , London , United Kingdom
| |
Collapse
|
11
|
Chauhan P, Saha B. Metabolic regulation of infection and inflammation. Cytokine 2018; 112:1-11. [PMID: 30472107 DOI: 10.1016/j.cyto.2018.11.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 10/30/2018] [Accepted: 11/13/2018] [Indexed: 12/11/2022]
Abstract
Immunometabolic framework provides a way to understand the immune regulation via cell intrinsic metabolic fluxes and metabolites during infections, tumors, and inflammatory disorders. During these diseases, the immune cells are activated requiring more energy and moderating their metabolic functions. The two categories of metabolic alterations are therefore causally associated with energy derivation and cellular functions. Pathogens, tumors and inflammation target energy metabolism, primarily glucose uptake, glucose catabolism, gluconeogenesis for continuing lipid metabolism through mainstream pathways such as glycolysis, tricarboxylic acid cycle, mitochondrial respiration and pentose phosphate pathway. Many biosynthetic pathways such as those of cholesterol, ceramide, sphingolipids, and fatty acids are altered explaining the metabolic interface in molecular pathogenesis in various infectious and non-infectious inflammatory diseases. The emerging immune-metabolic framework also identifies the key regulatory elements such as metabolites, signalling intermediates and transcription factors. These regulatory elements play key roles in deciding the fate of an infection, tumor or autoimmune diseases. The original research articles and the review articles in this Special issue of Cytokine on "Infection, Inflammation and Immunometabolomes" highlight these aspects of metabolic reprogramming and the role of some 'metabolomic regulators' in controlling the outcome of infectious and non-infectious diseases. In this Editorial, we introduce the readers to these articles discussing the elements in immune-metabolic framework.
Collapse
Affiliation(s)
- Prashant Chauhan
- National Centre for Cell Science, Ganeshkhind, Pune 411007, India
| | - Bhaskar Saha
- Trident Academy of Creative Technology, Bhubaneswar 750019, India
| |
Collapse
|
12
|
Vitamin C and immune cell function in inflammation and cancer. Biochem Soc Trans 2018; 46:1147-1159. [PMID: 30301842 PMCID: PMC6195639 DOI: 10.1042/bst20180169] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 08/16/2018] [Accepted: 08/21/2018] [Indexed: 12/14/2022]
Abstract
Vitamin C (ascorbate) is maintained at high levels in most immune cells and can affect many aspects of the immune response. Intracellular levels generally respond to variations in plasma ascorbate availability, and a combination of inadequate intake and increased turnover during severe stress can result in low plasma ascorbate status. Intracellular ascorbate supports essential functions and, in particular, acts as an enzyme cofactor for Fe- or Cu-containing oxygenases. Newly discovered enzymes in this family regulate cell metabolism and epigenetics, and dysregulation of their activity can affect cell phenotype, growth and survival pathways, and stem cell phenotype. This brief overview details some of the recent advances in our understanding of how ascorbate availability can affect the hydroxylases controlling the hypoxic response and the DNA and histone demethylases. These processes play important roles in the regulation of the immune system, altering cell survival pathways, metabolism and functions.
Collapse
|
13
|
Chen X, Zhou J, Li X, Wang X, Lin Y, Wang X. Exosomes derived from hypoxic epithelial ovarian cancer cells deliver microRNAs to macrophages and elicit a tumor-promoted phenotype. Cancer Lett 2018; 435:80-91. [PMID: 30098399 DOI: 10.1016/j.canlet.2018.08.001] [Citation(s) in RCA: 210] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 07/27/2018] [Accepted: 08/03/2018] [Indexed: 02/07/2023]
Abstract
Recently, cancer has been considered to be a complex system that includes the tumor microenvironment (TME). Tumor-associated macrophages (TAMs) are the most common immune-related stromal cells in the TME, and communication between cancer cells and TAMs is crucial for the progression of epithelial ovarian cancer (EOC). In this study, we revealed that exosomes derived from EOC cells remodel macrophages to a tumor-promoted phenotype, namely TAMs. In addition, hypoxic microenvironments have been postulated to facilitate this process in the TME, and hypoxia-inducible factors (HIFs) play an important role in this process. We found that TAMs educated by hypoxic exosomes derived from EOC cells promote tumor proliferation and migration in a feedback loop. Based on microarray analysis of normoxic and hypoxic exosomes, we discovered that a panel of miRNAs was enriched in hypoxic exosomes. And these three highly expressed miRNAs were induced by hypoxia via HIFs. In this study, we revealed that under hypoxic conditions, EOC cell-derived exosomes deliver miRNAs to induce M2 macrophage polarization, which promotes EOC cell proliferation and migration. This study suggests that these exosomes and associated miRNAs might serve as targets for novel treatments or diagnostic biomarkers for EOC.
Collapse
Affiliation(s)
- Xin Chen
- Department of Gynecology and Obstetrics, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 1665 Kong Jiang Road, 200092, Shanghai, China
| | - Jieru Zhou
- Department of Gynecology and Obstetrics, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 1665 Kong Jiang Road, 200092, Shanghai, China
| | - Xiaoduan Li
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, 201204, Shanghai, China
| | - Xinjing Wang
- Department of Gynecology and Obstetrics, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 1665 Kong Jiang Road, 200092, Shanghai, China
| | - Yingying Lin
- Department of Neurosurgery, RenJi Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 200127, China.
| | - Xipeng Wang
- Department of Gynecology and Obstetrics, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 1665 Kong Jiang Road, 200092, Shanghai, China.
| |
Collapse
|
14
|
Makhdoumi P, Abnous K, Mehri S, Etemad L, Imenshahidi M, Karimi G. Oral deferiprone administration ameliorates cisplatin-induced nephrotoxicity in rats. ACTA ACUST UNITED AC 2018; 70:1357-1368. [PMID: 30051477 DOI: 10.1111/jphp.12990] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Accepted: 07/07/2018] [Indexed: 01/13/2023]
Abstract
OBJECTIVES Cisplatin is one of the widely used antitumour agents with major clinical side effect, nephrotoxicity. We showed the role of iron in cisplatin-induced nephrotoxicity that entrance to the cell via transferrin receptor (TfR) as a gatekeeper for iron uptake. We also examined the effect of iron chelator deferiprone against this toxicity. METHODS Thirty male Wistar rats were randomly divided into six groups. Group I (saline orally for 10 days); group II (saline orally for 10 days plus single injection of cisplatin 7 mg/kg, intraperitoneally on 5th day); groups III, IV and V (deferiprone 50, 100 and 200 mg/kg orally for 10 days, respectively, plus cisplatin on 5th day). Group VI (deferiprone, orally). RESULTS Deferiprone provided functional and significant histological-proven protection in group IV. Deferiprone attenuated the increased creatinine, BUN, malondialdehyde and iron concentrations in cisplatin-injected animals. The increased amounts of TfR and decreased levels of HIF-1α and related anti-apoptotic genes expression in cisplatin-treated animals were improved by deferiprone. CONCLUSIONS The results supported a role for iron in cisplatin-induced nephrotoxicity and TfR may serve as an important source of iron. Based on these findings, deferiprone pretreatment may play a role in preventing cisplatin-induced nephropathy in cancer patient.
Collapse
Affiliation(s)
- Pouran Makhdoumi
- Student Research Committee, Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad, Iran.,Pharmaceutical Research Center, Mashhad, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Soghra Mehri
- Department of Pharmacodynamics and Toxicology, Pharmacy School, Mashhad University of Medical Sciences, Mashhad, Iran.,Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Leila Etemad
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohsen Imenshahidi
- Department of Pharmacodynamics and Toxicology, Pharmacy School, Mashhad University of Medical Sciences, Mashhad, Iran.,Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gholamreza Karimi
- Department of Pharmacodynamics and Toxicology, Pharmacy School, Mashhad University of Medical Sciences, Mashhad, Iran.,Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
15
|
Deepa A, Naveena K, Anindya R. DNA repair activity of Fe(II)/2OG-dependent dioxygenases affected by low iron level in Saccharomyces cerevisiae. FEMS Yeast Res 2018; 18:4847889. [DOI: 10.1093/femsyr/foy014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Accepted: 02/07/2018] [Indexed: 12/15/2022] Open
Affiliation(s)
- Akula Deepa
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Sangareddy-502285, India
| | - Kodipelli Naveena
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Sangareddy-502285, India
| | - Roy Anindya
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Sangareddy-502285, India
| |
Collapse
|
16
|
Abstract
PURPOSE OF REVIEW Fibroblast growth factor 23 (FGF23) is a hormone secreted by osteocytes and osteoblasts that regulates phosphorus and vitamin D homeostasis. FGF23 levels increase progressively in chronic kidney disease (CKD), and FGF23 excess might be a causal factor of left ventricular hypertrophy, CKD progression and death. Therefore, understanding the molecular mechanisms that control FGF23 production is critical to design therapies to lower FGF23 levels. The present review focuses on the role of inflammatory stimuli on FGF23 regulation and summarizes recent studies that support a novel framework linking inflammation to FGF23 regulation. RECENT FINDINGS Inflammation and iron deficiency, which are common occurrences in CKD, have emerged as novel FGF23 regulators. Recent findings show that inflammation increases FGF23 production in bone through direct and iron-related indirect mechanisms. In these settings, hypoxia-inducible factor (HIF)-1α orchestrates FGF23 transcription in response to inflammation and is primarily responsible for coordinating FGF23 production and cleavage. SUMMARY We demonstrate that inflammation increases FGF23 production and may contribute to elevated FGF23 levels in CKD. Osseous HIF-1α may represent a therapeutic target to lower FGF23 levels in CKD patients and minimize the negative consequences associated with FGF23 excess.
Collapse
|
17
|
Wohlrab C, Phillips E, Dachs GU. Vitamin C Transporters in Cancer: Current Understanding and Gaps in Knowledge. Front Oncol 2017; 7:74. [PMID: 28484682 PMCID: PMC5402541 DOI: 10.3389/fonc.2017.00074] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 04/06/2017] [Indexed: 11/14/2022] Open
Abstract
Sufficient uptake and whole body distribution of vitamin C (ascorbate) is essential for many biochemical processes, including some that are vital for tumor growth and spread. Uptake of ascorbate into cancer cells is modulated by availability, tumor blood flow, tissue diffusion parameters, and ascorbate transport proteins. Uptake into cells is mediated by two families of transport proteins, namely, the solute carrier gene family 23, consisting of sodium-dependent vitamin C transporters (SVCTs) 1 and 2, and the SLC2 family of glucose transporters (GLUTs). GLUTs transport the oxidized form of the vitamin, dehydroascorbate (DHA), which is present at negligible to low physiological levels. SVCT1 and 2 are capable of accumulating ascorbate against a concentration gradient from micromolar concentrations outside to millimolar levels inside of cells. Investigating the expression and regulation of SVCTs in cancer has only recently started to be included in studies focused on the role of ascorbate in tumor formation, progression, and response to therapy. This review gives an overview of the current, limited knowledge of ascorbate transport across membranes, as well as tissue distribution, gene expression, and the relevance of SVCTs in cancer. As tumor ascorbate accumulation may play a role in the anticancer activity of high dose ascorbate treatment, further research into ascorbate transport in cancer tissue is vital.
Collapse
Affiliation(s)
- Christina Wohlrab
- Mackenzie Cancer Research Group, Department of Pathology, University of Otago, Christchurch, New Zealand
| | - Elisabeth Phillips
- Mackenzie Cancer Research Group, Department of Pathology, University of Otago, Christchurch, New Zealand
| | - Gabi U Dachs
- Mackenzie Cancer Research Group, Department of Pathology, University of Otago, Christchurch, New Zealand
| |
Collapse
|
18
|
Zheng QQ, Zhao YS, Guo J, Zhao SD, Song LX, Fei CM, Zhang Z, Li X, Chang CK. Iron overload promotes erythroid apoptosis through regulating HIF-1a/ROS signaling pathway in patients with myelodysplastic syndrome. Leuk Res 2017; 58:55-62. [PMID: 28460338 DOI: 10.1016/j.leukres.2017.04.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 04/16/2017] [Accepted: 04/19/2017] [Indexed: 12/15/2022]
Abstract
Erythroid apoptosis increases significantly in myelodysplastic syndrome (MDS) patients with iron overload, but the underlying mechanism is not fully clear. In this study, we aim to explore the effect of HIF-1a/ROS on erythroid apoptosis in MDS patients with iron overload. We found that iron overload injured cellular functions through up-regulating ROS levels in MDS/AML cells, including inhibited cell viability, increased cell apoptosis and blocked cell cycle at G0/G1 phase. Interestingly, overexpression of hypoxia inducible factor-1a (HIF-1a), which was under-expressed in iron overload models, reduced ROS levels and attenuated cell damage caused by iron overload in MDS/AML cells. And gene knockdown of HIF-1a got the similar results as iron overload in MDS/AML cells. Furthermore, iron overload caused high erythroid apoptosis was closely related with ROS in MDS patients. Importantly, the HIF-1a protein levels of erythrocytes elevated obviously after incubation with desferrioxamine (DFO) from MDS patients with iron overload, accompanied by ROS levels inhibited and erythroid apoptosis reduced. Taken together, our findings determine that the HIF-1a/ROS signaling pathway plays a key role in promoting erythroid apoptosis in MDS patients with iron overload.
Collapse
Affiliation(s)
- Qing-Qing Zheng
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - You-Shan Zhao
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Juan Guo
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Si-da Zhao
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Lu-Xi Song
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Cheng-Ming Fei
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Zheng Zhang
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Xiao Li
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Chun-Kang Chang
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China.
| |
Collapse
|
19
|
Socco S, Bovee RC, Palczewski MB, Hickok JR, Thomas DD. Epigenetics: The third pillar of nitric oxide signaling. Pharmacol Res 2017; 121:52-58. [PMID: 28428114 DOI: 10.1016/j.phrs.2017.04.011] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 04/10/2017] [Indexed: 12/14/2022]
Abstract
Nitric oxide (NO), the endogenously produced free radical signaling molecule, is generally thought to function via its interactions with heme-containing proteins, such as soluble guanylyl cyclase (sGC), or by the formation of protein adducts containing nitrogen oxide functional groups (such as S-nitrosothiols, 3-nitrotyrosine, and dinitrosyliron complexes). These two types of interactions result in a multitude of down-stream effects that regulate numerous functions in physiology and disease. Of the numerous purported NO signaling mechanisms, epigenetic regulation has gained considerable interest in recent years. There is now abundant experimental evidence to establish NO as an endogenous epigenetic regulator of gene expression and cell phenotype. Nitric oxide has been shown to influence key aspects of epigenetic regulation that include histone posttranslational modifications, DNA methylation, and microRNA levels. Studies across disease states have observed NO-mediated regulation of epigenetic protein expression and enzymatic activity resulting in remodeling of the epigenetic landscape to ultimately influence gene expression. In addition to the well-established pathways of NO signaling, epigenetic mechanisms may provide much-needed explanations for poorly understood context-specific effects of NO. These findings provide more insight into the molecular mechanisms of NO signaling and increase our ability to dissect its functional role(s) in specific micro-environments in health and disease. This review will summarize the current state of NO signaling via epigenetic mechanisms (the "third pillar" of NO signaling).
Collapse
Affiliation(s)
- Samantha Socco
- Department of Medicinal Chemistry & Pharmacognosy, University of Illinois at Chicago, 60612, USA
| | - Rhea C Bovee
- Department of Medicinal Chemistry & Pharmacognosy, University of Illinois at Chicago, 60612, USA
| | - Marianne B Palczewski
- Department of Medicinal Chemistry & Pharmacognosy, University of Illinois at Chicago, 60612, USA
| | - Jason R Hickok
- Department of Medicinal Chemistry & Pharmacognosy, University of Illinois at Chicago, 60612, USA
| | - Douglas D Thomas
- Department of Medicinal Chemistry & Pharmacognosy, University of Illinois at Chicago, 60612, USA.
| |
Collapse
|
20
|
Wu HT, Kuo YC, Hung JJ, Huang CH, Chen WY, Chou TY, Chen Y, Chen YJ, Chen YJ, Cheng WC, Teng SC, Wu KJ. K63-polyubiquitinated HAUSP deubiquitinates HIF-1α and dictates H3K56 acetylation promoting hypoxia-induced tumour progression. Nat Commun 2016; 7:13644. [PMID: 27934968 PMCID: PMC5155157 DOI: 10.1038/ncomms13644] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 10/20/2016] [Indexed: 02/06/2023] Open
Abstract
Intratumoural hypoxia induces HIF-1α and promotes tumour progression, metastasis and treatment resistance. HIF-1α stability is regulated by VHL-E3 ligase-mediated ubiquitin-dependent degradation; however, the hypoxia-regulated deubiquitinase that stabilizes HIF-1α has not been identified. Here we report that HAUSP (USP7) deubiquitinase deubiquitinates HIF-1α to increase its stability, induce epithelial-mesenchymal transition and promote metastasis. Hypoxia induces K63-linked polyubiquitinated HAUSP at lysine 443 to enhance its functions. Knockdown of HAUSP decreases acetylation of histone 3 lysine 56 (H3K56Ac). K63-polyubiquitinated HAUSP interacts with a ubiquitin receptor CBP to specifically mediate H3K56 acetylation. ChIP-seq analysis of HAUSP and HIF-1α binding reveals two motifs responsive to hypoxia. HectH9 is the E3 ligase for HAUSP and a prognostic marker together with HIF-1α. This report demonstrates that hypoxia-induced K63-polyubiquitinated HAUSP deubiquitinates HIF-1α and causes CBP-mediated H3K56 acetylation on HIF-1α target gene promoters to promote EMT/metastasis, further defining HAUSP as a therapeutic target in hypoxia-induced tumour progression.
Collapse
Affiliation(s)
- Han-Tsang Wu
- Research Center for Tumor Medical Science, Graduate Institutes of Biomedical Sciences and New Drug Development, China Medical University, Taichung 404, Taiwan
| | - Yi-Chih Kuo
- Research Center for Tumor Medical Science, Graduate Institutes of Biomedical Sciences and New Drug Development, China Medical University, Taichung 404, Taiwan
| | - Jung-Jyh Hung
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 112, Taiwan.,Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei 112, Taiwan
| | - Chi-Hung Huang
- Taiwan Advance Biopharm (TABP), Inc., Xizhi city, New Taipei City 221, Taiwan
| | - Wei-Yi Chen
- Institute of Biochemistry &Molecular Biology, National Yang-Ming University, Taipei 112, Taiwan
| | - Teh-Ying Chou
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 112, Taiwan.,Department of Pathology, Taipei Veterans General Hospital, Taipei 112, Taiwan
| | - Yeh Chen
- Department of Biotechnology, Hungkuang University, Taichung 433, Taiwan
| | - Yi-Ju Chen
- Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan
| | - Yu-Ju Chen
- Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan
| | - Wei-Chung Cheng
- Research Center for Tumor Medical Science, Graduate Institutes of Biomedical Sciences and New Drug Development, China Medical University, Taichung 404, Taiwan
| | - Shu-Chun Teng
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Kou-Juey Wu
- Research Center for Tumor Medical Science, Graduate Institutes of Biomedical Sciences and New Drug Development, China Medical University, Taichung 404, Taiwan.,Department of Medical Research, China Medical University Hospital, Taichung 404, Taiwan
| |
Collapse
|
21
|
Abstract
Oxygen availability, along with the abundance of nutrients (such as glucose, glutamine, lipids and albumin), fluctuates significantly during tumour evolution and the recruitment of blood vessels, leukocytes and reactive fibroblasts to complex tumour microenvironments. As such, hypoxia and concomitant nutrient scarcity affect large gene expression programmes, signalling pathways, diverse metabolic reactions and various stress responses. This Review summarizes our current understanding of how these adaptations are integrated in hypoxic tumour cells and their role in disease progression.
Collapse
Affiliation(s)
- Michael S. Nakazawa
- Abramson Family Cancer Research Institute, Philadelphia, PA 19104, USA
- Department of Cancer Biology, Philadelphia, PA, USA
| | - Brian Keith
- Abramson Family Cancer Research Institute, Philadelphia, PA 19104, USA
- Department of Cancer Biology, Philadelphia, PA, USA
| | - M. Celeste Simon
- Abramson Family Cancer Research Institute, Philadelphia, PA 19104, USA
- Department of Cell and Developmental Biology, Philadelphia, PA 19104, USA
| |
Collapse
|
22
|
Vasudevan D, Bovee RC, Thomas DD. Nitric oxide, the new architect of epigenetic landscapes. Nitric Oxide 2016; 59:54-62. [PMID: 27553128 DOI: 10.1016/j.niox.2016.08.002] [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: 07/10/2016] [Accepted: 08/18/2016] [Indexed: 12/13/2022]
Abstract
Nitric oxide (NO) is an endogenously produced signaling molecule with multiple regulatory functions in physiology and disease. The most studied molecular mechanisms underlying the biological functions of NO include its reaction with heme proteins and regulation of protein activity via modification of thiol residues. A significant number of transcriptional responses and phenotypes observed in NO microenvironments, however, still lack mechanistic understanding. Recent studies shed new light on NO signaling by revealing its influence on epigenetic changes within the cell. Epigenetic alterations are important determinants of transcriptional responses and cell phenotypes, which can relay heritable information during cell division. As transcription across the genome is highly sensitive to these upstream epigenetic changes, this mode of NO signaling provides an alternate explanation for NO-mediated gene expression changes and phenotypes. This review will provide an overview of the interplay between NO and epigenetics as well as emphasize the unprecedented importance of these pathways to explain phenotypic effects associated with biological NO synthesis.
Collapse
Affiliation(s)
- Divya Vasudevan
- Department of Urology, Weill Cornell Medical College, New York, NY 10021, USA
| | - Rhea C Bovee
- Department of Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Douglas D Thomas
- Department of Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago, Chicago, IL 60612, USA.
| |
Collapse
|
23
|
Frise MC, Cheng HY, Nickol AH, Curtis MK, Pollard KA, Roberts DJ, Ratcliffe PJ, Dorrington KL, Robbins PA. Clinical iron deficiency disturbs normal human responses to hypoxia. J Clin Invest 2016; 126:2139-50. [PMID: 27140401 PMCID: PMC4887172 DOI: 10.1172/jci85715] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 03/10/2016] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Iron bioavailability has been identified as a factor that influences cellular hypoxia sensing, putatively via an action on the hypoxia-inducible factor (HIF) pathway. We therefore hypothesized that clinical iron deficiency would disturb integrated human responses to hypoxia. METHODS We performed a prospective, controlled, observational study of the effects of iron status on hypoxic pulmonary hypertension. Individuals with absolute iron deficiency (ID) and an iron-replete (IR) control group were exposed to two 6-hour periods of isocapnic hypoxia. The second hypoxic exposure was preceded by i.v. infusion of iron. Pulmonary artery systolic pressure (PASP) was serially assessed with Doppler echocardiography. RESULTS Thirteen ID individuals completed the study and were age- and sex-matched with controls. PASP did not differ by group or study day before each hypoxic exposure. During the first 6-hour hypoxic exposure, the rise in PASP was 6.2 mmHg greater in the ID group (absolute rises 16.1 and 10.7 mmHg, respectively; 95% CI for difference, 2.7-9.7 mmHg, P = 0.001). Intravenous iron attenuated the PASP rise in both groups; however, the effect was greater in ID participants than in controls (absolute reductions 11.1 and 6.8 mmHg, respectively; 95% CI for difference in change, -8.3 to -0.3 mmHg, P = 0.035). Serum erythropoietin responses to hypoxia also differed between groups. CONCLUSION Clinical iron deficiency disturbs normal responses to hypoxia, as evidenced by exaggerated hypoxic pulmonary hypertension that is reversed by subsequent iron administration. Disturbed hypoxia sensing and signaling provides a mechanism through which iron deficiency may be detrimental to human health. TRIAL REGISTRATION ClinicalTrials.gov (NCT01847352). FUNDING M.C. Frise is the recipient of a British Heart Foundation Clinical Research Training Fellowship (FS/14/48/30828). K.L. Dorrington is supported by the Dunhill Medical Trust (R178/1110). D.J. Roberts was supported by R&D funding from National Health Service (NHS) Blood and Transplant and a National Institute for Health Research (NIHR) Programme grant (RP-PG-0310-1004). This research was funded by the NIHR Oxford Biomedical Research Centre Programme.
Collapse
Affiliation(s)
- Matthew C. Frise
- University of Oxford, Department of Physiology, Anatomy and Genetics, Oxford, United Kingdom
| | - Hung-Yuan Cheng
- University of Oxford, Department of Physiology, Anatomy and Genetics, Oxford, United Kingdom
| | - Annabel H. Nickol
- Oxford University Hospitals NHS Foundation Trust, Oxford Centre for Respiratory Medicine, Churchill Hospital, Oxford, United Kingdom
| | - M. Kate Curtis
- University of Oxford, Department of Physiology, Anatomy and Genetics, Oxford, United Kingdom
| | - Karen A. Pollard
- University of Oxford, Department of Physiology, Anatomy and Genetics, Oxford, United Kingdom
| | - David J. Roberts
- University of Oxford, Nuffield Department of Clinical Laboratory Sciences, and National Health Service Blood and Transplant Oxford Centre, John Radcliffe Hospital, Oxford, United Kingdom
| | - Peter J. Ratcliffe
- University of Oxford, Nuffield Department of Medicine, Henry Wellcome Building for Molecular Physiology, Old Road Campus, Headington, Oxford, United Kingdom
| | - Keith L. Dorrington
- University of Oxford, Department of Physiology, Anatomy and Genetics, Oxford, United Kingdom
| | - Peter A. Robbins
- University of Oxford, Department of Physiology, Anatomy and Genetics, Oxford, United Kingdom
| |
Collapse
|
24
|
The LINK-A lncRNA activates normoxic HIF1α signalling in triple-negative breast cancer. Nat Cell Biol 2016; 18:213-24. [PMID: 26751287 PMCID: PMC4791069 DOI: 10.1038/ncb3295] [Citation(s) in RCA: 425] [Impact Index Per Article: 47.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 11/30/2015] [Indexed: 12/12/2022]
Abstract
Although long noncoding RNAs (lncRNAs) predominately reside in nuclear and exert their functions in many biological processes, their potential involvement in cytoplasmic signal transduction remains unexplored. Here, we identified a cytoplasmic lncRNA, Long-Intergenic Noncoding RNA for Kinase Activation (LINK-A), which mediates HB-EGF triggered, EGFR:GPNMB heterodimer-dependent HIF1α phosphorylation at Tyr565 and Ser797 by BRK and LRRK2 respectively. These events cause HIF1α stabilization, HIF1α-p300 interaction, and activation of HIF1α transcriptional programs under normoxic conditions. Mechanistically, LINK-A facilitates the recruitment of BRK to EGFR:GPNMB complex and BRK kinase activation. The BRK-dependent HIF1α Tyr565 phosphorylation interferes with Pro564 hydroxylation, leading to normoxic HIF1α stabilization. Both LINK-A and LINK-A-dependent signaling pathway activation correlate with TNBC, promoting breast cancer glycolysis reprogramming and tumorigenesis. Our findings illustrate the magnitude and diversity of cytoplasmic lncRNAs in signal transduction and highlight the important roles of lncRNAs in cancer.
Collapse
|
25
|
Siegert I, Schödel J, Nairz M, Schatz V, Dettmer K, Dick C, Kalucka J, Franke K, Ehrenschwender M, Schley G, Beneke A, Sutter J, Moll M, Hellerbrand C, Wielockx B, Katschinski DM, Lang R, Galy B, Hentze MW, Koivunen P, Oefner PJ, Bogdan C, Weiss G, Willam C, Jantsch J. Ferritin-Mediated Iron Sequestration Stabilizes Hypoxia-Inducible Factor-1α upon LPS Activation in the Presence of Ample Oxygen. Cell Rep 2015; 13:2048-55. [PMID: 26628374 DOI: 10.1016/j.celrep.2015.11.005] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 09/29/2015] [Accepted: 11/01/2015] [Indexed: 12/11/2022] Open
Abstract
Both hypoxic and inflammatory conditions activate transcription factors such as hypoxia-inducible factor (HIF)-1α and nuclear factor (NF)-κB, which play a crucial role in adaptive responses to these challenges. In dendritic cells (DC), lipopolysaccharide (LPS)-induced HIF1α accumulation requires NF-κB signaling and promotes inflammatory DC function. The mechanisms that drive LPS-induced HIF1α accumulation under normoxia are unclear. Here, we demonstrate that LPS inhibits prolyl hydroxylase domain enzyme (PHD) activity and thereby blocks HIF1α degradation. Of note, LPS-induced PHD inhibition was neither due to cosubstrate depletion (oxygen or α-ketoglutarate) nor due to increased levels of reactive oxygen species, fumarate, and succinate. Instead, LPS inhibited PHD activity through NF-κB-mediated induction of the iron storage protein ferritin and subsequent decrease of intracellular available iron, a critical cofactor of PHD. Thus, hypoxia and LPS both induce HIF1α accumulation via PHD inhibition but deploy distinct molecular mechanisms (lack of cosubstrate oxygen versus deprivation of co-factor iron).
Collapse
Affiliation(s)
- Isabel Siegert
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Johannes Schödel
- Department of Nephrology and Hypertension, Universitätsklinikum Erlangen and Friedrich-Alexander Universität (FAU), 91054 Erlangen, Germany
| | - Manfred Nairz
- Department of Internal Medicine VI, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Valentin Schatz
- Institute of Clinical Microbiology and Hygiene, University Hospital of Regensburg and University of Regensburg, 93053 Regensburg, Germany
| | - Katja Dettmer
- Institute of Functional Genomics, University of Regensburg, 93053 Regensburg, Germany
| | - Christopher Dick
- Institute of Clinical Microbiology and Hygiene, University Hospital of Regensburg and University of Regensburg, 93053 Regensburg, Germany
| | - Joanna Kalucka
- Department of Nephrology and Hypertension, Universitätsklinikum Erlangen and Friedrich-Alexander Universität (FAU), 91054 Erlangen, Germany
| | - Kristin Franke
- Heisenberg Research Group, Department of Clinical Pathobiochemistry, Institute of Clinical Chemistry and Laboratory Medicine, University of Technology, 01307 Dresden, Germany
| | - Martin Ehrenschwender
- Institute of Clinical Microbiology and Hygiene, University Hospital of Regensburg and University of Regensburg, 93053 Regensburg, Germany
| | - Gunnar Schley
- Department of Nephrology and Hypertension, Universitätsklinikum Erlangen and Friedrich-Alexander Universität (FAU), 91054 Erlangen, Germany
| | - Angelika Beneke
- Institute of Cardiovascular Physiology, University Medical Center, Georg-August-University Göttingen, 37073 Göttingen, Germany
| | - Jörg Sutter
- Department of Chemistry and Pharmacy, Inorganic Chemistry, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Matthias Moll
- Department of Chemistry and Pharmacy, Inorganic Chemistry, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Claus Hellerbrand
- Department of Internal Medicine I, University of Regensburg, 93053 Regensburg, Germany
| | - Ben Wielockx
- Heisenberg Research Group, Department of Clinical Pathobiochemistry, Institute of Clinical Chemistry and Laboratory Medicine, University of Technology, 01307 Dresden, Germany
| | - Dörthe M Katschinski
- Institute of Cardiovascular Physiology, University Medical Center, Georg-August-University Göttingen, 37073 Göttingen, Germany
| | - Roland Lang
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Bruno Galy
- Division of Virus-Associated Carcinogenesis, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | | | - Peppi Koivunen
- Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine, Oulu Center for Cell-Matrix Research, University of Oulu, 90014 Oulu, Finland
| | - Peter J Oefner
- Institute of Functional Genomics, University of Regensburg, 93053 Regensburg, Germany
| | - Christian Bogdan
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Günter Weiss
- Department of Internal Medicine VI, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Carsten Willam
- Department of Nephrology and Hypertension, Universitätsklinikum Erlangen and Friedrich-Alexander Universität (FAU), 91054 Erlangen, Germany
| | - Jonathan Jantsch
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany; Institute of Clinical Microbiology and Hygiene, University Hospital of Regensburg and University of Regensburg, 93053 Regensburg, Germany.
| |
Collapse
|
26
|
Owusu BY, Vaid M, Kaler P, Klampfer L. Prognostic and Predictive Significance of Stromal Fibroblasts and Macrophages in Colon Cancer. BIOMARKERS IN CANCER 2015; 7:29-37. [PMID: 26568685 PMCID: PMC4631158 DOI: 10.4137/bic.s25247] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 09/28/2015] [Accepted: 09/29/2015] [Indexed: 12/19/2022]
Abstract
Colon cancer development and malignant progression are driven by genetic and epigenetic alterations in tumor cells and by factors from the tumor microenvironment. Cancer cells become reliant on the activity of specific oncogenes and on prosurvival and proliferative signals they receive from the abnormal environment they create and reside in. Accordingly, the response to anticancer therapy is determined by genetic and epigenetic changes that are intrinsic to tumor cells and by the factors present in the tumor microenvironment. Recent advances in the understanding of the involvement of the tumor microenvironment in tumor progression and therapeutic response are optimizing the application of prognostic and predictive factors in colon cancer. Moreover, new targets in the tumor microenvironment that are amenable to therapeutic intervention have been identified. Because stromal cells are with rare exceptions genetically stable, the tumor microenvironment has emerged as a preferred target for therapeutic drugs. In this review, we discuss the role of stromal fibroblasts and macrophages in colon cancer progression and in the response of colon cancer patients to therapy.
Collapse
Affiliation(s)
- Benjamin Y Owusu
- Department of Oncology, Drug Discovery Division, Southern Research Institute, Birmingham, AL, USA
| | - Mudit Vaid
- Department of Oncology, Drug Discovery Division, Southern Research Institute, Birmingham, AL, USA
| | - Pawan Kaler
- Department of Oncology, Drug Discovery Division, Southern Research Institute, Birmingham, AL, USA
| | - Lidija Klampfer
- Department of Oncology, Drug Discovery Division, Southern Research Institute, Birmingham, AL, USA
| |
Collapse
|
27
|
Nagaraju GP, Bramhachari PV, Raghu G, El-Rayes BF. Hypoxia inducible factor-1α: Its role in colorectal carcinogenesis and metastasis. Cancer Lett 2015; 366:11-8. [PMID: 26116902 DOI: 10.1016/j.canlet.2015.06.005] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 05/29/2015] [Accepted: 06/09/2015] [Indexed: 01/05/2023]
Abstract
Tumor growth creates a hypoxic microenvironment, which promotes angiogenesis and aggressive tumor growth and invasion. HIF1α is a central molecule involved in mediating these effects of hypoxia. In colorectal cancer (CRC), hypoxia stabilizes the transcription factor HIF1α, leading to the expression of genes that are involved in tumor vascularization, metastasis/migration, cell survival and chemo-resistance. Therefore, HIF1α is a rational target for the development of new therapeutics for CRC. This article reviews the central role of HIF1α in CRC angiogenesis, metastasis, and progression as well as the strategies to target HIF1α stabilization.
Collapse
Affiliation(s)
- Ganji Purnachandra Nagaraju
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
| | | | - Godi Raghu
- Department of Biotechnology, Krishna University, Machilipatnam, AP-521001, India
| | - Bassel F El-Rayes
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
28
|
Frise MC, Robbins PA. Iron, oxygen, and the pulmonary circulation. J Appl Physiol (1985) 2015; 119:1421-31. [PMID: 26066825 PMCID: PMC4683351 DOI: 10.1152/japplphysiol.00179.2015] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 06/05/2015] [Indexed: 12/24/2022] Open
Abstract
The human pulmonary vasculature vasoconstricts in response to a reduction in alveolar oxygen tension, a phenomenon termed hypoxic pulmonary vasoconstriction (HPV). This review describes the time course of this behavior, which occurs in distinct phases, and then explores the importance for HPV of the hypoxia-inducible factor (HIF) pathway. Next, the HIF-hydroxylase enzymes that act as molecular oxygen sensors within the HIF pathway are discussed. These enzymes are particularly sensitive to intracellular iron availability, which confers iron-sensing properties on the HIF pathway. Human studies of iron chelation and supplementation are then reviewed. These demonstrate that the iron sensitivity of the HIF pathway evident from in vitro experiments is relevant to human pulmonary vascular physiology. Next, the importance of iron status in high-altitude illness and chronic cardiopulmonary disease is explored, and the therapeutic potential of intravenous iron discussed. The review concludes by highlighting some further complexities that arise from interactions between the HIF pathway and other intracellular iron-sensing mechanisms.
Collapse
Affiliation(s)
- Matthew C Frise
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Peter A Robbins
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
29
|
Thomas DD. Breathing new life into nitric oxide signaling: A brief overview of the interplay between oxygen and nitric oxide. Redox Biol 2015; 5:225-233. [PMID: 26056766 PMCID: PMC4473092 DOI: 10.1016/j.redox.2015.05.002] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 05/18/2015] [Indexed: 02/04/2023] Open
Abstract
Nitric oxide (•NO, nitrogen monoxide) is one of the most unique biological signaling molecules associated with a multitude of physiologic and pathological conditions. In order to fully appreciate its numerous roles, it is essential to understand its basic biochemical properties. Most signaling effector molecules such as steroids or proteins have a significant life-span and function through classical receptor–ligand interactions. •NO, however, is a short-lived free-radical gas that only reacts with two types of molecules under biological conditions; metals and other free radicals. These simple interactions can lead to a myriad of complex intermediates which in turn have their own phenotypic effects. For these reasons, responses to •NO often appear to be random or contradictory when outcomes are compared across various experimental settings. This article will serve as a brief overview of the chemical, biological, and microenvironmental factors that dictate •NO signaling with an emphasis on •NO metabolism. The prominent role that oxygen (dioxygen, O2) plays in •NO metabolism and how it influences the biological effects of •NO will be highlighted. This information and these concepts are intended to help students and investigators think about the interpretation of data from experiments where biological effects of •NO are being elucidated. Oxygen is a major determinant of the rates of nitric oxide synthesis and metabolism. Under biological conditions nitric oxide only reacts with metals and other free radicals. Oxygen determines the half-life, concentration, and diffusional distance of nitric oxide. Proteins respond to nitric oxide in a concentration and time-dependent manner. Oxygen and the redox environment will greatly influence signaling responses to nitric oxide.
Collapse
Affiliation(s)
- Douglas D Thomas
- Department of Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago, 833 S. Wood Street, MC 781, Chicago, IL 60612, USA.
| |
Collapse
|
30
|
Zengeya TT, Kulkarni RA, Meier JL. Modular synthesis of cell-permeating 2-ketoglutarate esters. Org Lett 2015; 17:2326-9. [PMID: 25915096 DOI: 10.1021/acs.orglett.5b00737] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Cell-permeating esters of 2-ketoglutarate (2-KG) have been synthesized through a convergent sequence from two modules in two and three steps, respectively. This route provides access to a full series of mono- and disubstituted 2-KG esters, enabling us to define the effect of regioisomeric masking on metabolite release and antihypoxic activity in cell-based assays. In addition to providing insight into the biological activity of cell permeable 2-KG esters, the straightforward and modular nature of this synthetic route may prove useful for the development of next-generation 2-KG analogues for diagnostic and therapeutic applications.
Collapse
Affiliation(s)
- Thomas T Zengeya
- Chemical Biology Laboratory, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Rhushikesh A Kulkarni
- Chemical Biology Laboratory, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Jordan L Meier
- Chemical Biology Laboratory, National Cancer Institute, Frederick, Maryland 21702, United States
| |
Collapse
|
31
|
Vasudevan D, Thomas DD. Insights into the diverse effects of nitric oxide on tumor biology. VITAMINS AND HORMONES 2015; 96:265-98. [PMID: 25189391 DOI: 10.1016/b978-0-12-800254-4.00011-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Among its many roles in cellular biology, nitric oxide (·NO) has long been associated with cancers both as a protumorigenic and as an antitumorigenic agent. The dual nature of this signaling molecule in varied settings is attributable to its temporal and concentration-dependent effects that produce different phenotypes. The steady-state ·NO concentration within the cell is a balance between its rate of enzymatic synthesis from the three nitric oxide synthase (NOS) isoforms and consumption via numerous metabolic pathways and demonstrates strong dependence on the tissue oxygen concentration. NOS expression and ·NO production are often deregulated and associated with numerous types of cancers with dissimilar prognostic outcomes. ·NO influences several facets of tumor initiation and progression including DNA damage, chronic inflammation, angiogenesis, epithelial-mesenchymal transition, and metastasis, to name a few. The role of ·NO as an epigenetic modulator has also recently emerged and has potentially important mechanistic implications in regulating transcription of oncogenes and tumor-suppressor genes. ·NO-derived cellular adducts such as dinitrosyliron complexes and the formation of higher nitrogen oxides further alter its cellular behavior. Among anticancer strategies, the use of NOS as a prognostic biomarker and modulation of ·NO production for therapeutic benefit have gained importance over the past decade. Numerous ·NO-releasing drugs and NOS inhibitors have been evaluated in preclinical and clinical settings to arrest tumor growth. Taken together, ·NO affects various arms of cancer signaling networks. An overview of this complex interplay is provided in this chapter.
Collapse
Affiliation(s)
- Divya Vasudevan
- Department of Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Douglas D Thomas
- Department of Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago, Chicago, Illinois, USA.
| |
Collapse
|
32
|
Talbot NP, Croft QP, Curtis MK, Turner BE, Dorrington KL, Robbins PA, Smith TG. Contrasting effects of ascorbate and iron on the pulmonary vascular response to hypoxia in humans. Physiol Rep 2014; 2:e12220. [PMID: 25501423 PMCID: PMC4332205 DOI: 10.14814/phy2.12220] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 10/29/2014] [Accepted: 10/30/2014] [Indexed: 11/24/2022] Open
Abstract
Hypoxia causes an increase in pulmonary artery pressure. Gene expression controlled by the hypoxia-inducible factor (HIF) family of transcription factors plays an important role in the underlying pulmonary vascular responses. The hydroxylase enzymes that regulate HIF are highly sensitive to varying iron availability, and iron status modifies the pulmonary vascular response to hypoxia, possibly through its effects on HIF. Ascorbate (vitamin C) affects HIF hydroxylation in a similar manner to iron and may therefore have similar pulmonary effects. This study investigated the possible contribution of ascorbate availability to hypoxic pulmonary vasoconstriction in humans. Seven healthy volunteers undertook a randomized, controlled, double-blind, crossover protocol which studied the effects of high-dose intravenous ascorbic acid (total 6 g) on the pulmonary vascular response to 5 h of sustained hypoxia. Systolic pulmonary artery pressure (SPAP) was assessed during hypoxia by Doppler echocardiography. Results were compared with corresponding data from a similar study investigating the effect of intravenous iron, in which SPAP was measured in seven healthy volunteers during 8 h of sustained hypoxia. Consistent with other studies, iron supplementation profoundly inhibited hypoxic pulmonary vasoconstriction (P < 0.001). In contrast, supraphysiological supplementation of ascorbate did not affect the increase in pulmonary artery pressure induced by several hours of hypoxia (P = 0.61). We conclude that ascorbate does not interact with hypoxia and the pulmonary circulation in the same manner as iron. Whether the effects of iron are HIF-mediated remains unknown, and the extent to which ascorbate contributes to HIF hydroxylation in vivo is also unclear.
Collapse
Affiliation(s)
- Nick P. Talbot
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, U.K
| | - Quentin P. Croft
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, U.K
| | - M. Kate Curtis
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, U.K
| | - Brandon E. Turner
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, U.K
| | - Keith L. Dorrington
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, U.K
| | - Peter A. Robbins
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, U.K
| | - Thomas G. Smith
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, U.K
- Nuffield Division of Anaesthetics, John Radcliffe Hospital, University of Oxford, Oxford, U.K
| |
Collapse
|
33
|
Genetic and functional evidence for a role for SLC11A1 in susceptibility to otitis media in early childhood in a Western Australian population. INFECTION GENETICS AND EVOLUTION 2013; 16:411-8. [DOI: 10.1016/j.meegid.2013.03.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 03/14/2013] [Accepted: 03/16/2013] [Indexed: 11/19/2022]
|
34
|
Hickok JR, Vasudevan D, Antholine WE, Thomas DD. Nitric oxide modifies global histone methylation by inhibiting Jumonji C domain-containing demethylases. J Biol Chem 2013; 288:16004-15. [PMID: 23546878 DOI: 10.1074/jbc.m112.432294] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Methylation of lysine residues on histone tails is an important epigenetic modification that is dynamically regulated through the combined effects of methyltransferases and demethylases. The Jumonji C domain Fe(II) α-ketoglutarate family of proteins performs the majority of histone demethylation. We demonstrate that nitric oxide ((•)NO) directly inhibits the activity of the demethylase KDM3A by forming a nitrosyliron complex in the catalytic pocket. Exposing cells to either chemical or cellular sources of (•)NO resulted in a significant increase in dimethyl Lys-9 on histone 3 (H3K9me2), the preferred substrate for KDM3A. G9a, the primary methyltransferase acting on H3K9me2, was down-regulated in response to (•)NO, and changes in methylation state could not be accounted for by methylation in general. Furthermore, cellular iron sequestration via dinitrosyliron complex formation correlated with increased methylation. The mRNA of several histone demethylases and methyltransferases was also differentially regulated in response to (•)NO. Taken together, these data reveal three novel and distinct mechanisms whereby (•)NO can affect histone methylation as follows: direct inhibition of Jumonji C demethylase activity, reduction in iron cofactor availability, and regulation of expression of methyl-modifying enzymes. This model of (•)NO as an epigenetic modulator provides a novel explanation for nonclassical gene regulation by (•)NO.
Collapse
Affiliation(s)
- Jason R Hickok
- Department of Medicinal Chemistry, University of Illinois at Chicago, Chicago, Illinois 60612, USA
| | | | | | | |
Collapse
|
35
|
Ratcliffe PJ. Oxygen sensing and hypoxia signalling pathways in animals: the implications of physiology for cancer. J Physiol 2013; 591:2027-42. [PMID: 23401619 DOI: 10.1113/jphysiol.2013.251470] [Citation(s) in RCA: 217] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Studies of regulation of the haematopoietic growth factor erythropoietin led to the unexpected discovery of a widespread system of direct oxygen sensing that regulates gene expression in animals. The oxygen-sensitive signal is generated by a series of non-haem Fe(II)- and 2-oxoglutarate-dependent dioxygenases that catalyse the post-translational hydroxylation of specific residues in the transcription factor hypoxia-inducible factor (HIF). These hydroxylations promote both oxygen-dependent degradation and oxygen-dependent inactivation of HIF, but are suppressed in hypoxia, leading to the accumulation of HIF and assembly of an active transcriptional complex in hypoxic cells. Hypoxia-inducible factor activates an extensive transcriptional cascade that interfaces with other cell signalling pathways, microRNA networks and RNA-protein translational control systems. The relationship of these cellular signalling pathways to the integrated physiology of oxygen homeostasis and the implication of dysregulating these massive physiological pathways in diseases such as cancer are discussed.
Collapse
Affiliation(s)
- Peter J Ratcliffe
- Henry Wellcome Building for Molecular Physiology, Old Road Campus, University of Oxford, Oxford OX3 7BN, UK.
| |
Collapse
|
36
|
The inflammatory microenvironment in hepatocellular carcinoma: a pivotal role for tumor-associated macrophages. BIOMED RESEARCH INTERNATIONAL 2012; 2013:187204. [PMID: 23533994 PMCID: PMC3591180 DOI: 10.1155/2013/187204] [Citation(s) in RCA: 301] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2012] [Accepted: 08/13/2012] [Indexed: 02/06/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the most common and aggressive human cancers worldwide. HCC is an example of inflammation-related cancer and represents a paradigm of the relation occurring between tumor microenvironment and tumor development. Tumor-associated macrophages (TAMs) are a major component of leukocyte infiltrate of tumors and play a pivotal role in tumor progression of inflammation-related cancer, including HCC. Several studies indicate that, in the tumor microenvironment, TAMs acquire an M2-polarized phenotype and promote angiogenesis, metastasis, and suppression of adaptive immunity through the expression of cytokines, chemokines, growth factors, and matrix metalloproteases. Indeed, an established M2 macrophage population has been associated with poor prognosis in HCC. The molecular links that connect cancer cells and TAMs are not completely known, but recent studies have demonstrated that NF-κB, STAT-3, and HIF-1 signaling pathways play key roles in this crosstalk. In this paper, we discuss the current knowledge about the role of TAMs in HCC development, highlighting the role of TAM-derived cytokines, chemokines, and growth factors in the initiation and progression of liver cancer and outlining the signaling pathways involved in the interplay between cancer cells and TAMs.
Collapse
|
37
|
Niecknig H, Tug S, Reyes BD, Kirsch M, Fandrey J, Berchner-Pfannschmidt U. Role of reactive oxygen species in the regulation of HIF-1 by prolyl hydroxylase 2 under mild hypoxia. Free Radic Res 2012; 46:705-17. [PMID: 22360728 DOI: 10.3109/10715762.2012.669041] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The function and survival of eukaryotic cells depends on a constant and sufficient oxygen supply. Cells recognize and respond to hypoxia by accumulation of the transcription factor hypoxia-inducible factor 1 (HIF-1), composed of an oxygen-sensitive HIF-1α and a constitutive HIF-1β subunit. Besides physiology, HIF-1 induction is involved in major pathological processes such as cardiovascular disease, inflammation and cancer, which are associated with the formation of reactive oxygen species (ROS). ROS have been reported to affect HIF-1 activity but the role for ROS in regulating HIF-1 has not been definitely settled. In order to shed light on the redox-regulation of HIF-1 by ROS, we studied the impact of exogenous ROS treatment (H(2)O(2)) on HIF-1α and HIF-1 regulatory protein prolyl hydroxylase 2 (PHD2) in the human osteosarcoma cell line U2OS. At early reaction periods, H(2)O(2) induced HIF-1α but at prolonged observation phases the opposite occurred. Herein, modulation of PHD activity appeared to be the key element, because knockdown and inhibition of the PHD2 prevented reduction of HIF-1α. However, H(2)O(2) treatment constantly suppressed HIF-1 transactivation at all time-points. Our data indicate a dual redox regulation of HIF-1α protein amount with a constant suppression of HIF-1 target gene expression by ROS.
Collapse
Affiliation(s)
- Helene Niecknig
- Institut für Physiologie, Universität Duisburg-Essen, Germany
| | | | | | | | | | | |
Collapse
|
38
|
Spagnuolo RD, Recalcati S, Tacchini L, Cairo G. Role of hypoxia-inducible factors in the dexrazoxane-mediated protection of cardiomyocytes from doxorubicin-induced toxicity. Br J Pharmacol 2011; 163:299-312. [PMID: 21232037 DOI: 10.1111/j.1476-5381.2011.01208.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND AND PURPOSE Iron aggravates the cardiotoxicity of doxorubicin, a widely used anticancer anthracycline, and the iron chelator dexrazoxane is the only agent protecting against doxorubicin cardiotoxicity; however, the mechanisms underlying the role of iron in doxorubicin-mediated cardiotoxicity and the protective role of dexrazoxane remain to be established. As iron is required for the degradation of hypoxia-inducible factors (HIF), which control the expression of antiapoptotic and protective genes, we tested the hypothesis that dexrazoxane-dependent HIF activation may mediate the cardioprotective effect of dexrazoxane. EXPERIMENTAL APPROACH Cell death, protein levels (by immunoblotting) and HIF-mediated transcription (using reporter constructs) were evaluated in the rat H9c2 cardiomyocyte cell line exposed to low doses of doxorubicin with or without dexrazoxane pretreatment. HIF levels were genetically manipulated by transfecting dominant-negative mutants or short hairpin RNA. KEY RESULTS Treatment with dexrazoxane induced HIF-1α and HIF-2α protein levels and transactivation capacity in H9c2 cells. It also prevented the induction of cell death and apoptosis by exposure of H9c2 cells to clinically relevant concentrations of doxorubicin. Suppression of HIF activity strongly reduced the protective effect of dexrazoxane. Conversely, HIF-1α overexpression protected against doxorubicin-mediated cell death and apoptosis also in cells not exposed to the chelator. Exposure to dexrazoxane increased the expression of the HIF-regulated, antiapoptotic proteins survivin, Mcl1 and haem oxygenase. CONCLUSIONS AND IMPLICATIONS Our results showing HIF-dependent prevention of doxorubicin toxicity in dexrazoxane-treated H9c2 cardiomyocytes suggest that HIF activation may be a mechanism contributing to the protective effect of dexrazoxane against anthracycline cardiotoxicity.
Collapse
Affiliation(s)
- R D Spagnuolo
- Department of Human Morphology and Biomedical Sciences 'Città Studi', University of Milan, Milano, Italy
| | | | | | | |
Collapse
|
39
|
Giusti S, Fiszer de Plazas S. Neuroprotection by hypoxic preconditioning involves upregulation of hypoxia-inducible factor-1 in a prenatal model of acute hypoxia. J Neurosci Res 2011; 90:468-78. [PMID: 21953610 DOI: 10.1002/jnr.22766] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2011] [Revised: 06/14/2011] [Accepted: 07/19/2011] [Indexed: 12/21/2022]
Abstract
The molecular pathways underlying the neuroprotective effects of preconditioning are promising, potentially drugable targets to promote cell survival. However, these pathways are complex and are not yet fully understood. In this study we have established a paradigm of hypoxic preconditioning based on a chick embryo model of normobaric acute hypoxia previously developed by our group. With this model, we analyzed the role of hypoxia-inducible factor-1α (HIF-1α) stabilization during preconditioning in HIF-1 signaling after the hypoxic injury and in the development of a neuroprotective effect against the insult. To this end, we used a pharmacological approach, based on the in vivo administration of positive (Fe(2+), ascorbate) and negative (CoCl(2)) modulators of the activity of HIF-prolyl hydroxylases (PHDs), the main regulators of HIF-1. We have found that preconditioning has a reinforcing effect on HIF-1 accumulation during the subsequent hypoxic injury. In addition, we have also demonstrated that HIF-1 induction during hypoxic preconditioning is necessary to obtain an enhancement in HIF-1 accumulation and to develop a tolerance against a subsequent hypoxic injury. We provide in vivo evidence that administration of Fe(2+) and ascorbate modulates HIF accumulation, suggesting that PHDs might be targets for neuroprotection in the CNS.
Collapse
Affiliation(s)
- Sebastián Giusti
- Institute of Cell Biology and Neuroscience Prof. E De Robertis, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | | |
Collapse
|
40
|
Bourseau-Guilmain E, Griveau A, Benoit JP, Garcion E. The importance of the stem cell marker prominin-1/CD133 in the uptake of transferrin and in iron metabolism in human colon cancer Caco-2 cells. PLoS One 2011; 6:e25515. [PMID: 21966538 PMCID: PMC3180456 DOI: 10.1371/journal.pone.0025515] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2011] [Accepted: 09/07/2011] [Indexed: 12/22/2022] Open
Abstract
As the pentaspan stem cell marker CD133 was shown to bind cholesterol and to localize in plasma membrane protrusions, we investigated a possible function for CD133 in endocytosis. Using the CD133 siRNA knockdown strategy and non-differentiated human colon cancer Caco-2 cells that constitutively over-expressed CD133, we provide for the first time direct evidence for a role of CD133 in the intracellular accumulation of fluorescently labeled extracellular compounds. Assessed using AC133 monoclonal antibody, CD133 knockdown was shown to improve Alexa488-transferrin (Tf) uptake in Caco-2 cells but had no impact on FITC-dextran or FITC-cholera-toxin. Absence of effect of the CD133 knockdown on Tf recycling established a role for CD133 in inhibiting Tf endocytosis rather than in stimulating Tf exocytosis. Use of previously identified inhibitors of known endocytic pathways and the positive impact of CD133 knockdown on cellular uptake of clathrin-endocytosed synthetic lipid nanocapsules supported that CD133 impact on endocytosis was primarily ascribed to the clathrin pathway. Also, cholesterol extraction with methyl-β-cyclodextrine up regulated Tf uptake at greater intensity in the CD133high situation than in the CD133low situation, thus suggesting a role for cholesterol in the inhibitory effect of CD133 on endocytosis. Interestingly, cell treatment with the AC133 antibody down regulated Tf uptake, thus demonstrating that direct extracellular binding to CD133 could affect endocytosis. Moreover, flow cytometry and confocal microscopy established that down regulation of CD133 improved the accessibility to the TfR from the extracellular space, providing a mechanism by which CD133 inhibited Tf uptake. As Tf is involved in supplying iron to the cell, effects of iron supplementation and deprivation on CD133/AC133 expression were investigated. Both demonstrated a dose-dependent down regulation here discussed to the light of transcriptional and post-transciptional effects. Taken together, these data extend our knowledge of the function of CD133 and underline the interest of further exploring the CD133-Tf-iron network.
Collapse
Affiliation(s)
- Erika Bourseau-Guilmain
- Laboratoire d'Ingénierie de la Vectorisation Particulaire, Inserm, UMR-S 646, Université d'Angers, Angers, France
| | - Audrey Griveau
- Laboratoire d'Ingénierie de la Vectorisation Particulaire, Inserm, UMR-S 646, Université d'Angers, Angers, France
| | - Jean-Pierre Benoit
- Laboratoire d'Ingénierie de la Vectorisation Particulaire, Inserm, UMR-S 646, Université d'Angers, Angers, France
| | - Emmanuel Garcion
- Laboratoire d'Ingénierie de la Vectorisation Particulaire, Inserm, UMR-S 646, Université d'Angers, Angers, France
- * E-mail:
| |
Collapse
|
41
|
Palmitoyl ascorbate liposomes and free ascorbic acid: comparison of anticancer therapeutic effects upon parenteral administration. Pharm Res 2011; 29:375-83. [PMID: 21845505 DOI: 10.1007/s11095-011-0557-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2011] [Accepted: 08/02/2011] [Indexed: 10/17/2022]
Abstract
PURPOSE To evaluate and compare anticancer therapeutic effect of palmitoyl ascorbate liposomes (PAL) and free ascorbic acid (AA). METHODS Liposomes incorporating palmitoyl ascorbate (PA) were prepared and evaluated for PA content by HPLC. To elucidate mechanism of action of cell death in vitro, effect of various H(2)O(2) scavengers and metal chelators on PA-mediated cytotoxicity was studied. Effect of various combinations of PAL and free AA on in vitro cytotoxicity was evaluated on 4T1 cells. In vivo, PAL formulation was modified with polyethylene glycol; effect of PEGylation on in vitro cytotoxicity was evaluated. Biodistribution of PEG-PAL formulation was investigated in female Balb/c mice bearing murine mammary carcinoma (4T1 cells). In vivo anticancer activity of PEG-PAL (PEG-PAL equivalent to 20 mg/kg of PA injected intravenously on alternate days) was compared with free AA therapy in same model. RESULTS PEG-PAL treatment was significantly more effective than free AA treatment in slowing tumor growth. CONCLUSIONS Nanoparticle formulations incorporating PA can kill cancer cells in vitro. The mechanism of PA cytotoxicity is based on production of extracellular reactive oxygen species and involves intracellular transition metals.
Collapse
|
42
|
Affiliation(s)
- Holger K Eltzschig
- Department of Anesthesiology, University of Colorado Denver, Aurora, CO 80045, USA.
| | | |
Collapse
|
43
|
Kim SR, Lee KS, Park HS, Park SJ, Min KH, Moon H, Puri KD, Lee YC. HIF-1α inhibition ameliorates an allergic airway disease via VEGF suppression in bronchial epithelium. Eur J Immunol 2010; 40:2858-69. [PMID: 20827786 DOI: 10.1002/eji.200939948] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Hypoxia-inducible factor-1α (HIF-1α) plays a critical role in immune and inflammatory responses. One of the HIF-1α target genes is vascular endothelial growth factor (VEGF), which is a potent stimulator of inflammation, airway remodeling, and physiologic dysregulation in allergic airway diseases. Using OVA-treated mice and murine tracheal epithelial cells, the signaling networks involved in HIF-1α activation and the role of HIF-1α in the pathogenesis of allergic airway disease were investigated. Transfection of airway epithelial cells with HIF-1α siRNA suppressed VEGF expression. In addition, the increased levels of HIF-1α and VEGF in lung tissues after OVA inhalation were substantially decreased by an HIF-1α inhibitor, 2-methoxyestradiol. Our data also show that the increased numbers of inflammatory cells, increased airway hyperresponsiveness, levels of IL-4, IL-5, IL-13, and vascular permeability in the lungs after OVA inhalation were significantly reduced by 2-methoxyestradiol or a VEGF inhibitor, CBO-P11. Moreover, we found that inhibition of the PI3K p110δ isoform (PI3K-δ) or HIF-1α reduced OVA-induced HIF-1α activation in airway epithelial cells. These findings indicate that HIF-1α inhibition may attenuate antigen-induced airway inflammation and hyperresponsiveness through the modulation of vascular leakage mediated by VEGF, and that PI3K-δ signaling may be involved in the allergen-induced HIF-1α activation.
Collapse
Affiliation(s)
- So Ri Kim
- Department of Internal Medicine and Research Center for Pulmonary Disorders, Chonbuk National University Medical School, Jeonju, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Xu M, Kashanchi F, Foster A, Rotimi J, Turner W, Gordeuk VR, Nekhai S. Hepcidin induces HIV-1 transcription inhibited by ferroportin. Retrovirology 2010; 7:104. [PMID: 21126372 PMCID: PMC3022686 DOI: 10.1186/1742-4690-7-104] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2010] [Accepted: 12/02/2010] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Physiological regulation of cellular iron involves iron export by the membrane protein, ferroportin, the expression of which is induced by iron and negatively modulated by hepcidin. We previously showed that iron chelation is associated with decreased HIV-1 transcription. We hypothesized that increased iron export by ferroportin might be associated with decreased HIV-1 transcription, and degradation of ferroportin by hepcidin might in turn induce HIV-1 transcription and replication. Here, we analyzed the effect of ferroportin and hepcidin on HIV-1 transcription. RESULTS Expression of ferroportin was associated with reduced HIV-1 transcription in 293T cells and addition of hepcidin to ferroportin-expressing cells counteracted this effect. Furthermore, exposure of promonocytic THP-1 cells to hepcidin was associated with decreased ferroportin expression, increased intracellular iron and induction of reporter luciferase gene expression. Finally, exposure of human primary macrophages and CD4+ T cells to hepcidin and iron was also associated with induction of viral production. CONCLUSION Our results suggest that the interplay between ferroportin-mediated iron export and hepcidin-mediated degradation of ferroportin might play a role in the regulation of HIV-1 transcription and may be important for understanding of HIV-1 pathogenesis.
Collapse
Affiliation(s)
- Min Xu
- Center for Sickle Cell Disease, Department of Medicine, Howard University, Washington, DC 20060, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Illingworth CJR, Loenarz C, Schofield CJ, Domene C. Chemical Basis for the Selectivity of the von Hippel Lindau Tumor Suppressor pVHL for Prolyl-Hydroxylated HIF-1α. Biochemistry 2010; 49:6936-44. [DOI: 10.1021/bi100358t] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Christopher J. R. Illingworth
- Department of Chemistry, Physical and Theoretical Chemistry Laboratory, University of Oxford, South Parks Road, Oxford OX1 3QZ, U.K
| | - Christoph Loenarz
- Chemistry Research Laboratory and Oxford Centre for Integrative Systems Biology, Department of Chemistry, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, U.K
| | - Christopher J. Schofield
- Chemistry Research Laboratory and Oxford Centre for Integrative Systems Biology, Department of Chemistry, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, U.K
| | - Carmen Domene
- Department of Chemistry, Physical and Theoretical Chemistry Laboratory, University of Oxford, South Parks Road, Oxford OX1 3QZ, U.K
| |
Collapse
|
46
|
Goven D, Boutten A, Leçon-Malas V, Marchal-Sommé J, Soler P, Boczkowski J, Bonay M. Induction of heme oxygenase-1, biliverdin reductase and H-ferritin in lung macrophage in smokers with primary spontaneous pneumothorax: role of HIF-1alpha. PLoS One 2010; 5:e10886. [PMID: 20526373 PMCID: PMC2878337 DOI: 10.1371/journal.pone.0010886] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2010] [Accepted: 05/05/2010] [Indexed: 12/27/2022] Open
Abstract
Background Few data concern the pathophysiology of primary spontaneous pneumothorax (PSP), which is associated with alveolar hypoxia/reoxygenation. This study tested the hypothesis that PSP is associated with oxidative stress in lung macrophages. We analysed expression of the oxidative stress marker 4-HNE; the antioxidant and anti-inflammatory proteins heme oxygenase-1 (HO-1), biliverdin reductase (BVR) and heavy chain of ferritin (H-ferritin); and the transcription factors controlling their expression Nrf2 and HIF-1α, in lung samples from smoker and nonsmoker patients with PSP (PSP-S and PSP-NS), cigarette smoke being a risk factor of recurrence of the disease. Methodology/Principal Findings mRNA was assessed by RT-PCR and proteins by western blot, immunohistochemistry and confocal laser analysis. 4-HNE, HO-1, BVR and H-ferritin were increased in macrophages from PSP-S as compared to PSP-NS and controls (C). HO-1 increase was associated with increased expression of HIF-1α mRNA and protein in alveolar macrophages in PSP-S patients, whereas Nrf2 was not modified. To understand the regulation of HO-1, BVR and H-ferritin, THP-1 macrophages were exposed to conditions mimicking conditions in C, PSP-S and PSP-NS patients: cigarette smoke condensate (CS) or air exposure followed or not by hypoxia/reoxygenation. Silencing RNA experiments confirmed that HIF-1α nuclear translocation was responsible for HO-1, BVR and H-ferritin induction mediated by CS and hypoxia/reoxygenation. Conclusions/Significance PSP in smokers is associated with lung macrophage oxidative stress. The response to this condition involves HIF-1α-mediated induction of HO-1, BVR and H-ferritin.
Collapse
Affiliation(s)
- Delphine Goven
- Inserm, U700, Faculté de Médecine-Site Bichat, Université Denis Diderot-Paris 7, Paris, France
| | - Anne Boutten
- Inserm, U700, Faculté de Médecine-Site Bichat, Université Denis Diderot-Paris 7, Paris, France
- Services de Biochimie A, Hôpital Bichat, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Véronique Leçon-Malas
- Services de Biochimie A, Hôpital Bichat, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Joëlle Marchal-Sommé
- Inserm, U700, Faculté de Médecine-Site Bichat, Université Denis Diderot-Paris 7, Paris, France
| | - Paul Soler
- Inserm, U700, Faculté de Médecine-Site Bichat, Université Denis Diderot-Paris 7, Paris, France
| | - Jorge Boczkowski
- Centre d'Investigation Clinique 007, Paris, France
- Inserm, U955, Faculté de Médecine, Groupe Hospitalier Universitaire Albert Chenevier - Henri Mondor, Créteil, France
- Faculté de Médecine, Université Paris 12, Créteil, France
| | - Marcel Bonay
- Inserm, U700, Faculté de Médecine-Site Bichat, Université Denis Diderot-Paris 7, Paris, France
- Service de Physiologie-Explorations Fonctionnelles, Hôpital Bichat, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
- * E-mail:
| |
Collapse
|
47
|
Billing AM, Fack F, Turner JD, Muller CP. Cortisol is a potent modulator of lipopolysaccharide-induced interferon signaling in macrophages. Innate Immun 2010; 17:302-20. [PMID: 20501517 DOI: 10.1177/1753425910369269] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The effects of cortisol (CORT) on resting and lipopolysaccharide (LPS)-activated monocyte-derived THP-1 macrophages were investigated by proteomics. Forty-seven proteins were found to be modulated, 20 by CORT, 11 by LPS, and 16 by CORT and LPS. Cortisol-sensitive chaperones and cytoskeletal proteins were mostly repressed. HCLS1, MGN, and MX1 were new proteins identified to be under the transcriptional control of this steroid and new CORT-sensitive variants of MX1, SYWC and IFIT3 were found. FKBP51, a known CORT target gene, showed the strongest response to CORT and synergism with LPS. In resting THP-1 macrophages, 18 proteins were modulated by CORT, with 15 being down-regulated. Activation of macrophages by LPS was associated with enhanced expression of immune response and metabolic proteins. In activated macrophages, CORT had a more equilibrated effect and almost all metabolism-related proteins were up-regulated, whereas immune response proteins were mostly down-regulated. The majority of the LPS up-regulated immune response-related proteins are known interferon (IFN) target genes (IFIT3, MX1, SYWC, PSME2) suggesting activation of the IRF3 signaling pathway. They were all suppressed by CORT. This is the first proteomics study to investigate the effects of CORT on activated immune cells.
Collapse
Affiliation(s)
- Anja M Billing
- Institute of Immunology, CRP-Santé/National Public Health Laboratory, 20A rue Auguste Lumiére, Luxembourg, Grand Duchy of Luxembourg
| | | | | | | |
Collapse
|
48
|
Nagel S, Talbot NP, Mecinović J, Smith TG, Buchan AM, Schofield CJ. Therapeutic manipulation of the HIF hydroxylases. Antioxid Redox Signal 2010; 12:481-501. [PMID: 19754349 DOI: 10.1089/ars.2009.2711] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The hypoxia-inducible factor (HIF) family of transcription factors is responsible for coordinating the cellular response to low oxygen levels in animals. By regulating the expression of a large array of target genes during hypoxia, these proteins also direct adaptive changes in the hematopoietic, cardiovascular, and respiratory systems. They also play roles in pathological processes, including tumorogenesis. In recent years, several oxygenases have been identified as key molecular oxygen sensors within the HIF system. The HIF hydroxylases regulate the stability and transcriptional activity of the HIF-alpha subunit by catalyzing hydroxylation of specific proline and asparaginyl residues, respectively. They require oxygen and 2-oxoglutarate (2OG) as co-substrates, and depend upon non-heme ferrous iron (Fe(II)) as a cofactor. This article summarizes current understanding of the biochemistry of the HIF hydroxylases, identifies targets for their pharmacological manipulation, and discusses their potential in the therapeutic manipulation of the HIF system.
Collapse
Affiliation(s)
- Simon Nagel
- Acute Stroke Programme, Nuffield Department of Clinical Medicine, John Radcliffe Hospital, Headington, Oxford, United Kingdom
| | | | | | | | | | | |
Collapse
|
49
|
Abstract
The ability of iron to accept or donate electrons, coupled with the ability of oxygen to act as an electron acceptor, renders both elements essential to normal cellular biology. However, these same chemical properties allow free iron in solution to generate toxic free radicals, particularly in combination with oxygen. Thus, closely interwoven homeostatic mechanisms have evolved to regulate both iron and oxygen concentrations at the systemic and the cellular levels. Systemically, iron levels are regulated through hepcidin-mediated uptake of iron in the duodenum, whereas intracellular free-iron levels are controlled through iron-regulatory proteins (IRPs). Cardiorespiratory changes increase systemic oxygen delivery, whereas at a cellular level, many responses to altered oxygen levels are coordinated by hypoxia-inducible factor (HIF). However, the mechanisms of iron homeostasis also are regulated by oxygen availability, with alterations in both hepcidin and IRP activity. In addition, many genes involved in iron homeostasis are direct targets of HIF. Furthermore, HIF activation is modulated by intracellular iron, through regulation of hydroxylase activity, which requires iron as a cofactor. In addition, HIF-2alpha translation is controlled by IRP activity, providing another level of interdependence between iron and oxygen homeostasis.
Collapse
Affiliation(s)
- David R Mole
- Henry Wellcome Building of Molecular Physiology, University of Oxford, Oxford, England.
| |
Collapse
|
50
|
Ebbesen P, Pettersen EO, Gorr TA, Jobst G, Williams K, Kieninger J, Wenger RH, Pastorekova S, Dubois L, Lambin P, Wouters BG, Van Den Beucken T, Supuran CT, Poellinger L, Ratcliffe P, Kanopka A, Görlach A, Gasmann M, Harris AL, Maxwell P, Scozzafava A. Taking advantage of tumor cell adaptations to hypoxia for developing new tumor markers and treatment strategies. J Enzyme Inhib Med Chem 2009; 24 Suppl 1:1-39. [PMID: 19005871 DOI: 10.1080/14756360902784425] [Citation(s) in RCA: 150] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Cancer cells in hypoxic areas of solid tumors are to a large extent protected against the action of radiation as well as many chemotherapeutic drugs. There are, however, two different aspects of the problem caused by tumor hypoxia when cancer therapy is concerned: One is due to the chemical reactions that molecular oxygen enters into therapeutically targeted cells. This results in a direct chemical protection against therapy by the hypoxic microenvironment, which has little to do with cellular biological regulatory processes. This part of the protective effect of hypoxia has been known for more than half a century and has been studied extensively. However, in recent years there has been more focus on the other aspect of hypoxia, namely the effect of this microenvironmental condition on selecting cells with certain genetic prerequisites that are negative with respect to patient prognosis. There are adaptive mechanisms, where hypoxia induces regulatory cascades in cells resulting in a changed metabolism or changes in extracellular signaling. These processes may lead to changes in cellular intrinsic sensitivity to treatment irrespective of oxygenation and, furthermore, may also have consequences for tissue organization. Thus, the adaptive mechanisms induced by hypoxia itself may have a selective effect on cells, with a fine-tuned protection against damage and stress of many kinds. It therefore could be that the adaptive mechanisms may take advantage of for new tumor labeling/imaging and treatment strategies. One of the Achilles' heels of hypoxia research has always been the exact measurements of tissue oxygenation as well as the control of oxygenation in biological tumor models. Thus, development of technology that can ease this control is vital in order to study mechanisms and perform drug development under relevant conditions. An integrated EU Framework project 2004-2009, termed EUROXY, demonstrates several pathways involved in transcription and translation control of the hypoxic cell phenotype and evidence of cross-talk with responses to pH and redox changes. The carbonic anhydrase isoenzyme CA IX was selected for further studies due to its expression on the surface of many types of hypoxic tumors. The effort has led to marketable culture flasks with sensors and incubation equipment, and the synthesis of new drug candidates against new molecular targets. New labeling/imaging methods for cancer diagnosing and imaging of hypoxic cancer tissue are now being tested in xenograft models and are also in early clinical testing, while new potential anti-cancer drugs are undergoing tests using xenografted tumor cancers. The present article describes the above results in individual consortium partner presentations.
Collapse
Affiliation(s)
- Peter Ebbesen
- Laboratory for Stem Cell Research, Aalborg University, Aarhus, Denmark.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|