1
|
Adeva-Andany MM, Adeva-Contreras L, Carneiro-Freire N, Ameneiros-Rodríguez E, Vila-Altesor M, Calvo-Castro I. The impact of high altitude (hypobaric hypoxia) on insulin resistance in humans. J Physiol Biochem 2025; 81:35-55. [PMID: 40019670 DOI: 10.1007/s13105-025-01069-8] [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: 06/22/2024] [Accepted: 01/30/2025] [Indexed: 03/01/2025]
Abstract
Exposure to hypobaric hypoxia (high altitude) diminishes systemic tissue oxygenation. Tissue hypoxia induces insulin resistance and a metabolic switch that reduces oxidative phosphorylation and glucose storage while enhancing glycolysis. Similarly to hypobaric hypoxia, insulin resistance develops in normal humans undergoing normobaric hypoxia and in patients with obstructive sleep apnea. Following acute exposure to high altitude, insulin resistance returns to baseline values upon returning to sea level or when compensatory mechanisms restore tissue oxygenation. However, insulin resistance persists in subjects unable to achieve sufficient oxygen delivery to tissues. Likewise, long-term residents at high altitude develop persistent insulin resistance when compensatory mechanisms do not attain adequate tissue oxygenation. Among these subjects, insulin resistance may cause clinical complications, such as hypertriglyceridemia, reduced HDL-c, visceral obesity, metabolic dysfunction-associated steatotic liver disease, essential hypertension, type 2 diabetes, subclinical vascular injury, cardiovascular disease, and kidney disease. Impaired tissue oxygenation allows the stabilization of hypoxia-inducible factor-1 (HIF-1), a transcription factor that modulates the transcriptional activity of a number of genes to coordinate the physiological responses to tissue hypoxia. Among them, HIF-1 downregulates PPARG, that codes peroxisome proliferator-activated receptor-gamma (PPAR-γ) and PPARGCA, that codes PPAR-γ coactivator-1α, in order to enable insulin resistance and the metabolic switch from oxidative phosphorylation toward glycolysis.
Collapse
Affiliation(s)
- María M Adeva-Andany
- Internal Medicine Department, Hospital General Juan Cardona, c/ Pardo Bazán s/n, Ferrol, 15406, Spain.
| | - Lucia Adeva-Contreras
- School of Medicine, Santiago de Compostela University, Santiago de Compostela, Galicia, Spain
| | - Natalia Carneiro-Freire
- Internal Medicine Department, Hospital General Juan Cardona, c/ Pardo Bazán s/n, Ferrol, 15406, Spain
| | - Eva Ameneiros-Rodríguez
- Internal Medicine Department, Hospital General Juan Cardona, c/ Pardo Bazán s/n, Ferrol, 15406, Spain
| | - Matilde Vila-Altesor
- Internal Medicine Department, Hospital General Juan Cardona, c/ Pardo Bazán s/n, Ferrol, 15406, Spain
| | - Isabel Calvo-Castro
- Internal Medicine Department, Hospital General Juan Cardona, c/ Pardo Bazán s/n, Ferrol, 15406, Spain
| |
Collapse
|
2
|
Robles-Oteíza C, Hastings K, Choi J, Sirois I, Ravi A, Expósito F, de Miguel F, Knight JR, López-Giráldez F, Choi H, Socci ND, Merghoub T, Awad M, Getz G, Gainor J, Hellmann MD, Caron É, Kaech SM, Politi K. Hypoxia is linked to acquired resistance to immune checkpoint inhibitors in lung cancer. J Exp Med 2025; 222:e20231106. [PMID: 39585348 PMCID: PMC11602551 DOI: 10.1084/jem.20231106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 04/29/2024] [Accepted: 09/27/2024] [Indexed: 11/26/2024] Open
Abstract
Despite the established use of immune checkpoint inhibitors (ICIs) to treat non-small cell lung cancer (NSCLC), only a subset of patients benefit from treatment and ∼50% of patients whose tumors respond eventually develop acquired resistance (AR). To identify novel drivers of AR, we generated murine Msh2 knock-out (KO) lung tumors that initially responded but eventually developed AR to anti-PD-1, alone or in combination with anti-CTLA-4. Resistant tumors harbored decreased infiltrating T cells and reduced cancer cell-intrinsic MHC-I and MHC-II levels, yet remained responsive to IFNγ. Resistant tumors contained extensive regions of hypoxia, and a hypoxia signature derived from single-cell transcriptional profiling of resistant cancer cells was associated with decreased progression-free survival in a cohort of NSCLC patients treated with anti-PD-1/PD-L1 therapy. Targeting hypoxic tumor regions using a hypoxia-activated pro-drug delayed AR to ICIs in murine Msh2 KO tumors. Thus, this work provides a rationale for targeting tumor metabolic features, such as hypoxia, in combination with immune checkpoint inhibition.
Collapse
Affiliation(s)
| | | | - Jungmin Choi
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Korea
| | | | - Arvind Ravi
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | | | - James R. Knight
- Yale Center for Genome Analysis, Yale University, New Haven, CT, USA
| | | | - Hyejin Choi
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nicholas D. Socci
- Marie-Josee and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Taha Merghoub
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program & Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Mark Awad
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Gad Getz
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Department of Pathology, Harvard Medical School, Boston, MA, USA
- Krantz Family Center for Cancer Research and Department of Pathology, Massachusetts Genral Hospital, Boston, MA, USA
| | - Justin Gainor
- Center for Thoracic Cancers, Massachusetts General Hospital, Boston, MA, USA
| | - Matthew D. Hellmann
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Étienne Caron
- CHU Sainte-Justine Research Center, Montreal, Canada
- Department of Pathology and Cellular Biology, Faculty of Medicine, Université de Montréal, Montreal, Canada
| | - Susan M. Kaech
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute, La Jolla, CA, USA
| | - Katerina Politi
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
- Departments of Pathology and Internal Medicine (Section of Medical Oncology), Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
3
|
Youssef E, Zhao S, Purcell C, Olson GL, El-Deiry WS. Targeting the SMURF2-HIF1α axis: a new frontier in cancer therapy. Front Oncol 2024; 14:1484515. [PMID: 39697237 PMCID: PMC11652374 DOI: 10.3389/fonc.2024.1484515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 11/14/2024] [Indexed: 12/20/2024] Open
Abstract
The SMAD-specific E3 ubiquitin protein ligase 2 (SMURF2) has emerged as a critical regulator in cancer biology, modulating the stability of Hypoxia-Inducible Factor 1-alpha (HIF1α) and influencing a network of hypoxia-driven pathways within the tumor microenvironment (TME). SMURF2 targets HIF1α for ubiquitination and subsequent proteasomal degradation, disrupting hypoxic responses that promote cancer cell survival, metabolic reprogramming, angiogenesis, and resistance to therapy. Beyond its role in HIF1α regulation, SMURF2 exerts extensive control over cellular processes central to tumor progression, including chromatin remodeling, DNA damage repair, ferroptosis, and cellular stress responses. Notably, SMURF2's ability to promote ferroptotic cell death through GSTP1 degradation offers an alternative pathway to overcome apoptosis resistance, expanding therapeutic options for refractory cancers. This review delves into the multifaceted interactions between SMURF2 and HIF1α, emphasizing how their interplay impacts metabolic adaptations like the Warburg effect, immune evasion, and therapeutic resistance. We discuss SMURF2's dual functionality as both a tumor suppressor and, in certain contexts, an oncogenic factor, underscoring its potential as a highly versatile therapeutic target. Furthermore, modulating the SMURF2-HIF1α axis presents an innovative approach to destabilize hypoxia-dependent pathways, sensitizing tumors to chemotherapy, radiotherapy, and immune-based treatments. However, the complexity of SMURF2's interactions necessitate a thorough assessment of potential off-target effects and challenges in specificity, which must be addressed to optimize its clinical application. This review concludes by proposing future directions for research into the SMURF2-HIF1α pathway, aiming to refine targeted strategies that exploit this axis and address the adaptive mechanisms of aggressive tumors, ultimately advancing the landscape of precision oncology.
Collapse
Affiliation(s)
- Emile Youssef
- Research & Development, SMURF-Therapeutics, Inc., Providence, RI, United States
- Medical & Pharmacovigilance, Kapadi, Inc., Raleigh, NC, United States
| | - Shuai Zhao
- Department of Pathology & Laboratory Medicine, Legorreta Cancer Center at Brown University, Providence, RI, United States
| | - Connor Purcell
- Department of Pathology & Laboratory Medicine, Legorreta Cancer Center at Brown University, Providence, RI, United States
| | - Gary L. Olson
- Medicinal Chemistry & Drug Discovery, Provid Pharmaceuticals, Inc., Monmouth Junction, NJ, United States
| | - Wafik S. El-Deiry
- Research & Development, SMURF-Therapeutics, Inc., Providence, RI, United States
- Department of Pathology & Laboratory Medicine, Legorreta Cancer Center at Brown University, Providence, RI, United States
| |
Collapse
|
4
|
Trusen S, Zimmermann JSA, Fries FN, Li Z, Chai N, Seitz B, Suiwal S, Amini M, Szentmáry N, Stachon T. Increased susceptibility of human limbal aniridia fibroblasts to oxidative stress. Exp Eye Res 2024; 248:110105. [PMID: 39303843 DOI: 10.1016/j.exer.2024.110105] [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: 05/19/2024] [Revised: 08/19/2024] [Accepted: 09/18/2024] [Indexed: 09/22/2024]
Abstract
Aniridia-associated keratopathy originates from a haploinsufficiency of the transcription factor PAX6 (PAX6+/-). In the corneal epithelium of PAX6+/- mice, a significant increase in oxidized proteins was observed, accompanied by impaired compensation for elevated oxidative stress (OS). The extent to which limbal fibroblast cells (LFCs) are affected by an increased susceptibility to OS in cases of congenital aniridia (AN) has not been determined, yet. Our aim was to examine the impact of OS on antioxidant enzyme expression in normal and AN-LFCs. Following isolation and culture of primary LFCs (n = 8) and AN-LFCs (n = 8), cells were treated with cobalt chloride for 48 h to chemically induce hypoxic conditions and OS. Subsequently, HIF-1α/-2α, PHD1/2, Nrf2, CAT, SOD1, PRDX6, and GPX1 gene expression was examined by qPCR. SOD1, PRDX6, and GPX1 protein levels were assessed from the cell lysate by Western blot. The induction of hypoxia led to reduced HIF-1α gene expression in both fibroblast groups (p≤0.008), while the decrease in PHD1 was limited to AN-LFCs (p = 0.0007). On the other hand, under hypoxic conditions, PHD2 showed higher mRNA expression in AN-LFCs compared to normal LFCs (p = 0.013). As a result of OS, the mRNA levels of Nrf2 (p<0.0001) and the antioxidant enzymes CAT (p = 0.005), SOD1 (p = 0.005), GPX1 (p = 0.002) decreased in AN-LFCs. This was accompanied by an increased protein expression of SOD1 (p = 0.019) and PRDX6 (p=0.0009). In the normal LFC group, the induced extent of OS had no impact on the gene (p≥0.151) and protein expression (p ≥ 0.629) of antioxidant enzymes, except for the GPX1 mRNA level (p = 0.027). AN-LFCs exhibit higher susceptibility to OS than normal LFCs. Therefore, in AN-LFCs, there are sustained alterations in gene and protein expression of antioxidative enzymes even after 48 h of CoCl2 treatment.
Collapse
Affiliation(s)
- Simon Trusen
- Dr. Rolf M. Schwiete Center for Limbal Stem Cell and Aniridia Research, Saarland University, Kirrberger Str. 100, 66424, Homburg/Saar, Germany.
| | - Julia Sarah Alexandra Zimmermann
- Dr. Rolf M. Schwiete Center for Limbal Stem Cell and Aniridia Research, Saarland University, Kirrberger Str. 100, 66424, Homburg/Saar, Germany
| | - Fabian Norbert Fries
- Dr. Rolf M. Schwiete Center for Limbal Stem Cell and Aniridia Research, Saarland University, Kirrberger Str. 100, 66424, Homburg/Saar, Germany; Department of Ophthalmology, Saarland University Medical Center, Kirrberger Str. 100, 66424, Homburg/Saar, Germany
| | - Zhen Li
- Dr. Rolf M. Schwiete Center for Limbal Stem Cell and Aniridia Research, Saarland University, Kirrberger Str. 100, 66424, Homburg/Saar, Germany
| | - Ning Chai
- Dr. Rolf M. Schwiete Center for Limbal Stem Cell and Aniridia Research, Saarland University, Kirrberger Str. 100, 66424, Homburg/Saar, Germany
| | - Berthold Seitz
- Department of Ophthalmology, Saarland University Medical Center, Kirrberger Str. 100, 66424, Homburg/Saar, Germany
| | - Shweta Suiwal
- Dr. Rolf M. Schwiete Center for Limbal Stem Cell and Aniridia Research, Saarland University, Kirrberger Str. 100, 66424, Homburg/Saar, Germany
| | - Maryam Amini
- Dr. Rolf M. Schwiete Center for Limbal Stem Cell and Aniridia Research, Saarland University, Kirrberger Str. 100, 66424, Homburg/Saar, Germany
| | - Nóra Szentmáry
- Dr. Rolf M. Schwiete Center for Limbal Stem Cell and Aniridia Research, Saarland University, Kirrberger Str. 100, 66424, Homburg/Saar, Germany
| | - Tanja Stachon
- Dr. Rolf M. Schwiete Center for Limbal Stem Cell and Aniridia Research, Saarland University, Kirrberger Str. 100, 66424, Homburg/Saar, Germany
| |
Collapse
|
5
|
László L, Kurilla A, Tilajka Á, Pancsa R, Takács T, Novák J, Buday L, Vas V. Unveiling epithelial plasticity regulation in lung cancer: Exploring the cross-talk among Tks4 scaffold protein partners. Mol Biol Cell 2024; 35:ar111. [PMID: 38985526 PMCID: PMC11321040 DOI: 10.1091/mbc.e24-03-0103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/24/2024] [Accepted: 07/01/2024] [Indexed: 07/12/2024] Open
Abstract
The epithelial-to-mesenchymal transition (EMT) represents a hallmark event in the evolution of lung cancer. This work aims to study a recently described EMT-regulating protein, Tks4, and to explore its potential as a prognostic biomarker in non-small cell lung cancer. In this study, we used CRISPR/Cas9 method to knockout (KO) Tks4 to study its functional roles in invadopodia formation, migration, and regulation of EMT marker expressions and we identified Tks4-interacting proteins. Tks4-KO A549 cells exhibited an EMT-like phenotype characterized by elongated morphology and increased expression of EMT markers. Furthermore, analyses of a large-scale lung cancer database and a patient-derived tissue array data revealed that the Tks4 mRNA level was decreased in more aggressive lung cancer stages. To understand the regulatory role of Tks4 in lung cancer, we performed a Tks4-interactome analysis via Tks4 immunoprecipitation-mass spectrometry on five different cell lines and identified CAPZA1 as a novel Tks4 partner protein. Thus, we propose that the absence of Tks4 leads to disruption of a connectome of multiple proteins and that the resulting undocking and likely mislocalization of signaling molecules impairs actin cytoskeleton rearrangement and activates EMT-like cell fate switches, both of which likely influence disease severity.
Collapse
Affiliation(s)
- Loretta László
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, 1117 Budapest, Hungary
- Doctoral School of Biology, Institute of Biology, ELTE Eötvös Loránd University, 1117 Budapest, Hungary
| | - Anita Kurilla
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, 1117 Budapest, Hungary
| | - Álmos Tilajka
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, 1117 Budapest, Hungary
- Doctoral School of Biology, Institute of Biology, ELTE Eötvös Loránd University, 1117 Budapest, Hungary
| | - Rita Pancsa
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, 1117 Budapest, Hungary
| | - Tamás Takács
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, 1117 Budapest, Hungary
- Doctoral School of Biology, Institute of Biology, ELTE Eötvös Loránd University, 1117 Budapest, Hungary
| | - Julianna Novák
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, 1117 Budapest, Hungary
| | - László Buday
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, 1117 Budapest, Hungary
- Department of Molecular Biology, Semmelweis University, 1094 Budapest, Hungary
| | - Virag Vas
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, 1117 Budapest, Hungary
| |
Collapse
|
6
|
Adeva-Andany MM, Domínguez-Montero A, Castro-Quintela E, Funcasta-Calderón R, Fernández-Fernández C. Hypoxia-Induced Insulin Resistance Mediates the Elevated Cardiovascular Risk in Patients with Obstructive Sleep Apnea: A Comprehensive Review. Rev Cardiovasc Med 2024; 25:231. [PMID: 39076340 PMCID: PMC11270082 DOI: 10.31083/j.rcm2506231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 04/07/2024] [Accepted: 04/12/2024] [Indexed: 07/31/2024] Open
Abstract
Patients with obstructive sleep apnea (OSA) experience insulin resistance and its clinical consequences, including hypertriglyceridemia, reduced high density lipoprotein-associated cholesterol (HDL-c), visceral adiposity, hepatic steatosis, increased epicardial fat thickness, essential hypertension, glucose intolerance, increased risk for type 2 diabetes, chronic kidney disease, subclinical vascular damage, and increased risk for cardiovascular events. Obesity is a major contributor to OSA. The prevalence of OSA is almost universal among patients with severe obesity undergoing bariatric surgery. However, insulin resistance and its clinical complications occur in OSA patients irrespective of general obesity (body mass index). In OSA patients, apnea episodes during sleep induce oxyhemoglobin desaturation and tissue hypoxia. Insulin resistance is an adaptive response to tissue hypoxia and develops in conditions with limited tissue oxygen supply, including healthy subjects exposed to hypobaric hypoxia (high altitude) and OSA patients. Indicators of oxyhemoglobin desaturation have been robustly and independently linked to insulin resistance and its clinical manifestations in patients with OSA. Insulin resistance mediates the elevated rate of type 2 diabetes, chronic kidney disease, and cardiovascular disease unexplained with traditional cardiovascular risk factors present in OSA patients. Pathophysiological processes underlying hypoxia-induced insulin resistance involve hypoxia inducible factor-1 upregulation and peroxisome proliferator-activated receptor-gamma (PPAR- γ ) downregulation. In human adipose tissue, PPAR- γ activity promotes glucose transport into adipocytes, lipid droplet biogenesis, and whole-body insulin sensitivity. Silencing of PPAR- γ in the adipose tissue reduces glucose uptake and fat accumulation into adipocytes and promotes insulin resistance. In conclusion, tissue hypoxia drives insulin resistance and its clinical consequences in patients with OSA, regardless of body mass index.
Collapse
|
7
|
Hall CHT, Lanis JM, Dowdell AS, Murphy EM, Bhagavatula G, Neuhart RM, Vijaya Sai KY, Colgan SP. Fundamental role for the creatine kinase pathway in protection from murine colitis. Mucosal Immunol 2023; 16:817-825. [PMID: 37716510 PMCID: PMC12070598 DOI: 10.1016/j.mucimm.2023.09.002] [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: 03/06/2023] [Revised: 09/08/2023] [Accepted: 09/10/2023] [Indexed: 09/18/2023]
Abstract
Inflammatory diseases of the digestive tract, including inflammatory bowel disease, cause metabolic stress within mucosal tissue. Creatine is a key energetic regulator. We previously reported a loss of creatine kinases (CKs) and the creatine transporter expression in inflammatory bowel disease patient intestinal biopsy samples and that creatine supplementation was protective in a dextran sulfate sodium (DSS) colitis mouse model. In the present studies, we evaluated the role of CK loss in active inflammation using the DSS colitis model. Mice lacking expression of CK brain type/CK mitochondrial form (CKdKO) showed increased susceptibility to DSS colitis (weight loss, disease activity, permeability, colon length, and histology). In a broad cytokine profiling, CKdKO mice expressed near absent interferon gamma (IFN-γ) levels. We identified losses in IFN-γ production from CD4+ and CD8+ T cells isolated from CKdKO mice. Addback of IFN-γ during DSS treatment resulted in partial protection for CKdKO mice. Extensions of these studies identified basal stabilization of the transcription factor hypoxia-inducible factor in CKdKO splenocytes and pharmacological stabilization of hypoxia-inducible factor resulted in reduced IFN-γ production by control splenocytes. Thus, the loss of IFN-γ production by CD4+ and CD8+ T cells in CKdKO mice resulted in increased colitis susceptibility and indicates that CK is protective in active mucosal inflammation.
Collapse
Affiliation(s)
- Caroline H T Hall
- Division of Gastroenterology, Hepatology and Nutrition, Children's Hospital Colorado and University of Colorado, Aurora, Colorado, USA; Division of Gastroenterology and Hepatology, Department of Medicine, Mucosal Inflammation Program and University of Colorado, Aurora, Colorado, USA
| | - Jordi M Lanis
- Division of Gastroenterology and Hepatology, Department of Medicine, Mucosal Inflammation Program and University of Colorado, Aurora, Colorado, USA; Department of Immunology and Microbiology, University of Colorado, Aurora, Colorado, USA
| | - Alexander S Dowdell
- Division of Gastroenterology and Hepatology, Department of Medicine, Mucosal Inflammation Program and University of Colorado, Aurora, Colorado, USA; Rocky Mountain Veterans Hospital, Aurora, Colorado, USA
| | - Emily M Murphy
- Division of Gastroenterology, Hepatology and Nutrition, Children's Hospital Colorado and University of Colorado, Aurora, Colorado, USA; Division of Gastroenterology and Hepatology, Department of Medicine, Mucosal Inflammation Program and University of Colorado, Aurora, Colorado, USA
| | - Geetha Bhagavatula
- Division of Gastroenterology, Hepatology and Nutrition, Children's Hospital Colorado and University of Colorado, Aurora, Colorado, USA; Division of Gastroenterology and Hepatology, Department of Medicine, Mucosal Inflammation Program and University of Colorado, Aurora, Colorado, USA
| | - Rane M Neuhart
- Division of Gastroenterology, Hepatology and Nutrition, Children's Hospital Colorado and University of Colorado, Aurora, Colorado, USA; Division of Gastroenterology and Hepatology, Department of Medicine, Mucosal Inflammation Program and University of Colorado, Aurora, Colorado, USA
| | - Kiranmayee Yenugudhati Vijaya Sai
- Division of Gastroenterology and Hepatology, Department of Medicine, Mucosal Inflammation Program and University of Colorado, Aurora, Colorado, USA; Rocky Mountain Veterans Hospital, Aurora, Colorado, USA
| | - Sean P Colgan
- Division of Gastroenterology and Hepatology, Department of Medicine, Mucosal Inflammation Program and University of Colorado, Aurora, Colorado, USA; Rocky Mountain Veterans Hospital, Aurora, Colorado, USA.
| |
Collapse
|
8
|
Nsiah NY, Morgan AB, Donkor N, Inman DM. Long-term HIF-1α stabilization reduces respiration, promotes mitophagy, and results in retinal cell death. Sci Rep 2023; 13:20541. [PMID: 37996657 PMCID: PMC10667534 DOI: 10.1038/s41598-023-47942-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 11/20/2023] [Indexed: 11/25/2023] Open
Abstract
Ocular hypertension during glaucoma can lead to hypoxia, activation of the HIF transcription factors, and a metabolic shift toward glycolysis. This study aims to test whether chronic HIF activation and the attendant metabolic reprogramming can initiate glaucoma-associated pathology independently of ocular hypertension. HIF-1α stabilization was induced in mice for 2 and 4 weeks by inhibiting prolyl hydroxylases using the small molecule Roxadustat. HIF-1α stabilization and the expression of its downstream bioenergetic targets were investigated in the retina by immunofluorescence, capillary electrophoresis, and biochemical enzyme activity assays. Roxadustat dosing resulted in significant stabilization of HIF-1α in the retina by 4 weeks, and upregulation in glycolysis-associated proteins (GLUT3, PDK-1) and enzyme activity in both neurons and glia. Accordingly, succinate dehydrogenase, mitochondrial marker MTCO1, and citrate synthase activity were significantly decreased at 4 weeks, while mitophagy was significantly increased. TUNEL assay showed significant apoptosis of cells in the retina, and PERG amplitude was significantly decreased with 4 weeks of HIF-1α stabilization. A significant increase in AMPK activation and glial hypertrophy, concomitant with decreases in retinal ganglion cell function and inner retina cell death suggests that chronic HIF-1α stabilization alone is detrimental to retina metabolic homeostasis and cellular survival.
Collapse
Affiliation(s)
- Nana Yaa Nsiah
- Department of Pharmaceutical Sciences, North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX, USA
- Genentech, South San Francisco, CA, USA
| | - Autumn B Morgan
- Department of Pharmaceutical Sciences, North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Nina Donkor
- Department of Pharmaceutical Sciences, North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Denise M Inman
- Department of Pharmaceutical Sciences, North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX, USA.
| |
Collapse
|
9
|
Huang T, Fakurazi S, Cheah PS, Ling KH. REST Targets JAK-STAT and HIF-1 Signaling Pathways in Human Down Syndrome Brain and Neural Cells. Int J Mol Sci 2023; 24:9980. [PMID: 37373133 DOI: 10.3390/ijms24129980] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/02/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Down syndrome (DS) is the most frequently diagnosed chromosomal disorder of chromosome 21 (HSA21) aneuploidy, characterized by intellectual disability and reduced lifespan. The transcription repressor, Repressor Element-1 Silencing Transcription factor (REST), which acts as an epigenetic regulator, is a crucial regulator of neuronal and glial gene expression. In this study, we identified and investigated the role of REST-target genes in human brain tissues, cerebral organoids, and neural cells in Down syndrome. Gene expression datasets generated from healthy controls and DS samples of human brain tissues, cerebral organoids, NPC, neurons, and astrocytes were retrieved from the Gene Ontology (GEO) and Sequence Read Archive (SRA) databases. Differential expression analysis was performed on all datasets to produce differential expression genes (DEGs) between DS and control groups. REST-targeted DEGs were subjected to functional ontologies, pathways, and network analyses. We found that REST-targeted DEGs in DS were enriched for the JAK-STAT and HIF-1 signaling pathways across multiple distinct brain regions, ages, and neural cell types. We also identified REST-targeted DEGs involved in nervous system development, cell differentiation, fatty acid metabolism and inflammation in the DS brain. Based on the findings, we propose REST as the critical regulator and a promising therapeutic target to modulate homeostatic gene expression in the DS brain.
Collapse
Affiliation(s)
- Tan Huang
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - Sharida Fakurazi
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - Pike-See Cheah
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - King-Hwa Ling
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
- Malaysian Research Institute on Ageing (MyAgeingTM), Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| |
Collapse
|
10
|
Mikami Y, Grubb BR, Rogers TD, Dang H, Asakura T, Kota P, Gilmore RC, Okuda K, Morton LC, Sun L, Chen G, Wykoff JA, Ehre C, Vilar J, van Heusden C, Livraghi-Butrico A, Gentzsch M, Button B, Stutts MJ, Randell SH, O’Neal WK, Boucher RC. Chronic airway epithelial hypoxia exacerbates injury in muco-obstructive lung disease through mucus hyperconcentration. Sci Transl Med 2023; 15:eabo7728. [PMID: 37285404 PMCID: PMC10664029 DOI: 10.1126/scitranslmed.abo7728] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 05/17/2023] [Indexed: 06/09/2023]
Abstract
Unlike solid organs, human airway epithelia derive their oxygen from inspired air rather than the vasculature. Many pulmonary diseases are associated with intraluminal airway obstruction caused by aspirated foreign bodies, virus infection, tumors, or mucus plugs intrinsic to airway disease, including cystic fibrosis (CF). Consistent with requirements for luminal O2, airway epithelia surrounding mucus plugs in chronic obstructive pulmonary disease (COPD) lungs are hypoxic. Despite these observations, the effects of chronic hypoxia (CH) on airway epithelial host defense functions relevant to pulmonary disease have not been investigated. Molecular characterization of resected human lungs from individuals with a spectrum of muco-obstructive lung diseases (MOLDs) or COVID-19 identified molecular features of chronic hypoxia, including increased EGLN3 expression, in epithelia lining mucus-obstructed airways. In vitro experiments using cultured chronically hypoxic airway epithelia revealed conversion to a glycolytic metabolic state with maintenance of cellular architecture. Chronically hypoxic airway epithelia unexpectedly exhibited increased MUC5B mucin production and increased transepithelial Na+ and fluid absorption mediated by HIF1α/HIF2α-dependent up-regulation of β and γENaC (epithelial Na+ channel) subunit expression. The combination of increased Na+ absorption and MUC5B production generated hyperconcentrated mucus predicted to perpetuate obstruction. Single-cell and bulk RNA sequencing analyses of chronically hypoxic cultured airway epithelia revealed transcriptional changes involved in airway wall remodeling, destruction, and angiogenesis. These results were confirmed by RNA-in situ hybridization studies of lungs from individuals with MOLD. Our data suggest that chronic airway epithelial hypoxia may be central to the pathogenesis of persistent mucus accumulation in MOLDs and associated airway wall damage.
Collapse
Affiliation(s)
- Yu Mikami
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Barbara R. Grubb
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Troy D. Rogers
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Hong Dang
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Takanori Asakura
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Pradeep Kota
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Rodney C. Gilmore
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Kenichi Okuda
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Lisa C. Morton
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Ling Sun
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Gang Chen
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Jason A. Wykoff
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Camille Ehre
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Juan Vilar
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Catharina van Heusden
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | | | - Martina Gentzsch
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Brian Button
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - M. Jackson Stutts
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Scott H. Randell
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Wanda K. O’Neal
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Richard C. Boucher
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| |
Collapse
|
11
|
Mayro B, Hoj JP, Cerda-Smith CG, Hutchinson HM, Caminear MW, Thrash HL, Winter PS, Wardell SE, McDonnell DP, Wu C, Wood KC, Pendergast AM. ABL kinases regulate the stabilization of HIF-1α and MYC through CPSF1. Proc Natl Acad Sci U S A 2023; 120:e2210418120. [PMID: 37040401 PMCID: PMC10120083 DOI: 10.1073/pnas.2210418120] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 02/07/2023] [Indexed: 04/12/2023] Open
Abstract
The hypoxia-inducible factor 1-α (HIF-1α) enables cells to adapt and respond to hypoxia (Hx), and the activity of this transcription factor is regulated by several oncogenic signals and cellular stressors. While the pathways controlling normoxic degradation of HIF-1α are well understood, the mechanisms supporting the sustained stabilization and activity of HIF-1α under Hx are less clear. We report that ABL kinase activity protects HIF-1α from proteasomal degradation during Hx. Using a fluorescence-activated cell sorting (FACS)-based CRISPR/Cas9 screen, we identified HIF-1α as a substrate of the cleavage and polyadenylation specificity factor-1 (CPSF1), an E3-ligase which targets HIF-1α for degradation in the presence of an ABL kinase inhibitor in Hx. We show that ABL kinases phosphorylate and interact with CUL4A, a cullin ring ligase adaptor, and compete with CPSF1 for CUL4A binding, leading to increased HIF-1α protein levels. Further, we identified the MYC proto-oncogene protein as a second CPSF1 substrate and show that active ABL kinase protects MYC from CPSF1-mediated degradation. These studies uncover a role for CPSF1 in cancer pathobiology as an E3-ligase antagonizing the expression of the oncogenic transcription factors, HIF-1α and MYC.
Collapse
Affiliation(s)
- Benjamin Mayro
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC27710
| | - Jacob P. Hoj
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC27710
| | - Christian G. Cerda-Smith
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC27710
| | - Haley M. Hutchinson
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC27710
| | - Michael W. Caminear
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC27710
| | - Hannah L. Thrash
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC27710
| | - Peter S. Winter
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC27710
| | - Suzanne E. Wardell
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC27710
| | - Donald P. McDonnell
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC27710
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC27710
| | - Colleen Wu
- Department of Orthopedic Surgery, Duke University School of Medicine, Durham, NC27710
| | - Kris C. Wood
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC27710
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC27710
| | - Ann Marie Pendergast
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC27710
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC27710
| |
Collapse
|
12
|
The NRSF/REST transcription factor in hallmarks of cancer: From molecular mechanisms to clinical relevance. Biochimie 2023; 206:116-134. [PMID: 36283507 DOI: 10.1016/j.biochi.2022.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 10/07/2022] [Accepted: 10/17/2022] [Indexed: 11/23/2022]
Abstract
The RE-1 silencing transcription factor (REST), or neuron restrictive silencing factor (NRSF), was first identified as a repressor of neuronal genes in non-neuronal tissue. Interestingly, this transcription factor may act as a tumor suppressor or an oncogenic role in developing neuroendocrine and other tumors in patients. The hallmarks of cancer include six biological processes, including proliferative signaling, evasion of growth suppressors, resistance to cell death, replicative immortality, inducing angiogenesis, and activating invasion and metastasis. In addition to two emerging hallmarks, the reprogramming of energy metabolism and evasion of the immune response are all implicated in the development of human tumors. It is essential to know the role of these processes as they will affect the outcome of alternatives for cancer treatment. Various studies in this review demonstrate that NRSF/REST affects the different hallmarks of cancer that could position NRSF/REST as an essential target in the therapy and diagnosis of certain types of cancer.
Collapse
|
13
|
Liebe H, Schlegel C, Cai X, Golubkova A, Loerke C, Leiva T, Hunter CJ. Apical-Out Enteroids as an Innovative Model for Necrotizing Enterocolitis. J Surg Res 2023; 283:1106-1116. [PMID: 36915002 PMCID: PMC10014931 DOI: 10.1016/j.jss.2022.11.048] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 11/15/2022] [Accepted: 11/20/2022] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Necrotizing enterocolitis (NEC) is a gastrointestinal disease of premature neonates. We previously validated a NEC enteroid model derived from human infant intestinal tissue. Typical enteroid configuration is basolateral-out (BO) without direct access to the luminal (apical) surface. Apical access is necessary to allow physiologic comparison of pathogen interaction with the intestinal epithelial barrier. We hypothesize that apical-out (AO) enteroids will provide a relevant NEC model to study this relationship. METHODS Following the institutional review board approval (#11610-11611), neonatal intestinal tissue was collected from surgical specimens. Stem cells were collected; enteroids were generated and grown to maturity in BO conformation then everted to AO. Enteroids were untreated or treated for 24 h with 100 μg/mL lipopolysaccharide and hypoxia. Protein and gene expression were analyzed for inflammatory markers, tight junction (TJ) proteins and permeability characteristic of NEC. RESULTS Apical TJ protein zonula occludens-1 and basolateral protein β-catenin immunofluorescence confirmed AO configuration. Treated AO enteroids had significantly increased messenger RNA (P = 0.001) and protein levels (P < 0.0001) of tumor necrosis factor-α compared to controls. Corrected total cell fluorescence of toll-like receptor 4 was significantly increased in treated AO enteroids compared to control (P = 0.002). Occludin was found to have significantly decreased messenger RNA in treated AO enteroids (P = 0.003). Expression of other TJ proteins claudins-1, -4 and zonula occludens-1 was significantly decreased in treated AO enteroids (P < 0.05). CONCLUSIONS AO enteroids present an innovative model for NEC with increased inflammation and gut barrier restructuring. This model allows for a biologically relevant investigation of the interaction between the pathogen and the intestinal epithelial barrier in NEC.
Collapse
Affiliation(s)
- Heather Liebe
- Division of Pediatric Surgery, Oklahoma Children's Hospital, Oklahoma City, Oklahoma.
| | - Camille Schlegel
- The University of Oklahoma Health Sciences Center, Department of Surgery, Oklahoma City, Oklahoma
| | - Xue Cai
- The University of Oklahoma Health Sciences Center, Department of Surgery, Oklahoma City, Oklahoma
| | - Alena Golubkova
- Division of Pediatric Surgery, Oklahoma Children's Hospital, Oklahoma City, Oklahoma
| | | | - Tyler Leiva
- Division of Pediatric Surgery, Oklahoma Children's Hospital, Oklahoma City, Oklahoma
| | - Catherine J Hunter
- Division of Pediatric Surgery, Oklahoma Children's Hospital, Oklahoma City, Oklahoma
| |
Collapse
|
14
|
Sun Z, Han J, Wang J. Circular RNA PIP5K1A promotes glycolysis and malignancy of non-small cell lung cancer via miR-656-3p/GBE1 axis under hypoxia. Mol Cell Toxicol 2023. [DOI: 10.1007/s13273-023-00343-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2023]
|
15
|
Hypoxia-Driven Changes in a Human Intestinal Organoid Model and the Protective Effects of Hydrolyzed Whey. Nutrients 2023; 15:nu15020393. [PMID: 36678267 PMCID: PMC9863820 DOI: 10.3390/nu15020393] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/08/2023] [Accepted: 01/09/2023] [Indexed: 01/15/2023] Open
Abstract
Many whey proteins, peptides and protein-derived amino acids have been suggested to improve gut health through their anti-oxidant, anti-microbial, barrier-protective and immune-modulating effects. Interestingly, although the degree of hydrolysis influences peptide composition and, thereby, biological function, this important aspect is often overlooked. In the current study, we aimed to investigate the effects of whey protein fractions with different degrees of enzymatic hydrolysis on the intestinal epithelium in health and disease with a novel 2D human intestinal organoid (HIO) monolayer model. In addition, we aimed to assess the anti-microbial activity and immune effects of the whey protein fractions. Human intestinal organoids were cultured from adult small intestines, and a model enabling apical administration of nutritional components during hypoxia-induced intestinal inflammation and normoxia (control) in crypt-like and villus-like HIO was established. Subsequently, the potential beneficial effects of whey protein isolate (WPI) and two whey protein hydrolysates with a 27.7% degree of hydrolysis (DH28) and a 50.9% degree of hydrolysis (DH51) were assessed. In addition, possible immune modulatory effects on human peripheral immune cells and anti-microbial activity on four microbial strains of the whey protein fractions were investigated. Exposure to DH28 prevented paracellular barrier loss of crypt-like HIO following hypoxia-induced intestinal inflammation with a concomitant decrease in hypoxia inducible factor 1 alpha (HIF1α) mRNA expression. WPI increased Treg numbers and Treg expression of cluster of differentiation 25 (CD25) and CD69 and reduced CD4+ T cell proliferation, whereas no anti-microbial effects were observed. The observed biological effects were differentially mediated by diverse whey protein fractions, indicating that (degree of) hydrolysis influences their biological effects. Moreover, these new insights may provide opportunities to improve immune tolerance and promote intestinal health.
Collapse
|
16
|
Oxidative Stress Response's Kinetics after 60 Minutes at Different (30% or 100%) Normobaric Hyperoxia Exposures. Int J Mol Sci 2022; 24:ijms24010664. [PMID: 36614106 PMCID: PMC9821105 DOI: 10.3390/ijms24010664] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/22/2022] [Accepted: 12/26/2022] [Indexed: 01/03/2023] Open
Abstract
Oxygen is a powerful trigger for cellular reactions and is used in many pathologies, including oxidative stress. However, the effects of oxygen over time and at different partial pressures remain poorly understood. In this study, the metabolic responses of normobaric oxygen intake for 1 h to mild (30%) and high (100%) inspired fractions were investigated. Fourteen healthy non-smoking subjects (7 males and 7 females; age: 29.9 ± 11.1 years, height: 168.2 ± 9.37 cm; weight: 64.4 ± 12.3 kg; BMI: 22.7 ± 4.1) were randomly assigned in the two groups. Blood samples were taken before the intake at 30 min, 2 h, 8 h, 24 h, and 48 h after the single oxygen exposure. The level of oxidation was evaluated by the rate of reactive oxygen species (ROS) and the levels of isoprostane. Antioxidant reactions were observed by total antioxidant capacity (TAC), superoxide dismutase (SOD), and catalase (CAT). The inflammatory response was measured using interleukin-6 (IL-6), neopterin, creatinine, and urates. Oxidation markers increased from 30 min on to reach a peak at 8 h. From 8 h post intake, the markers of inflammation took over, and more significantly with 100% than with 30%. This study suggests a biphasic response over time characterized by an initial "permissive oxidation" followed by increased inflammation. The antioxidant protection system seems not to be the leading actor in the first place. The kinetics of enzymatic reactions need to be better studied to establish therapeutic, training, or rehabilitation protocols aiming at a more targeted use of oxygen.
Collapse
|
17
|
Intermittent hypoxia enhances the expression of hypoxia inducible factor HIF1A through histone demethylation. J Biol Chem 2022; 298:102536. [PMID: 36174675 PMCID: PMC9597902 DOI: 10.1016/j.jbc.2022.102536] [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: 02/24/2022] [Revised: 09/14/2022] [Accepted: 09/15/2022] [Indexed: 11/29/2022] Open
Abstract
The cellular response to hypoxia is regulated through enzymatic oxygen sensors, including the prolyl hydroxylases, which control degradation of the well-known hypoxia inducible factors (HIFs). Other enzymatic oxygen sensors have been recently identified, including members of the KDM histone demethylase family. Little is known about how different oxygen-sensing pathways interact and if this varies depending on the form of hypoxia, such as chronic or intermittent. In this study, we investigated how two proposed cellular oxygen-sensing systems, HIF-1 and KDM4A, KDM4B, and KDM4C, respond in cells exposed to rapid forms of intermittent hypoxia (minutes) and compared to chronic hypoxia (hours). We found that intermittent hypoxia increases HIF-1α protein through a pathway distinct from chronic hypoxia, involving the KDM4A, KDM4B, and KDM4C histone lysine demethylases. Intermittent hypoxia increases the quantity and activity of KDM4A, KDM4B, and KDM4C, resulting in a decrease in histone 3 lysine 9 (H3K9) trimethylation near the HIF1A locus. We demonstrate that this contrasts with chronic hypoxia, which decreases KDM4A, KDM4B, and KDM4C activity, leading to hypertrimethylation of H3K9 globally and at the HIF1A locus. Altogether, we found that demethylation of histones bound to the HIF1A gene in intermittent hypoxia increases HIF1A mRNA expression, which has the downstream effect of increasing overall HIF-1 activity and expression of HIF target genes. This study highlights how multiple oxygen-sensing pathways can interact to regulate and fine tune the cellular hypoxic response depending on the period and length of hypoxia.
Collapse
|
18
|
Manella G, Ezagouri S, Champigneulle B, Gaucher J, Mendelson M, Lemarie E, Stauffer E, Pichon A, Howe CA, Doutreleau S, Golik M, Verges S, Asher G. The human blood transcriptome exhibits time-of-day-dependent response to hypoxia: Lessons from the highest city in the world. Cell Rep 2022; 40:111213. [PMID: 35977481 PMCID: PMC9396531 DOI: 10.1016/j.celrep.2022.111213] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 05/27/2022] [Accepted: 07/22/2022] [Indexed: 11/16/2022] Open
Abstract
High altitude exposes humans to hypobaric hypoxia, which induces various physiological and molecular changes. Recent studies point toward interaction between circadian rhythms and the hypoxic response, yet their human relevance is lacking. Here, we examine the effect of different high altitudes in conjunction with time of day on human whole-blood transcriptome upon an expedition to the highest city in the world, La Rinconada, Peru, which is 5,100 m above sea level. We find that high altitude vastly affects the blood transcriptome and, unexpectedly, does not necessarily follow a monotonic response to altitude elevation. Importantly, we observe daily variance in gene expression, especially immune-related genes, which is largely altitude dependent. Moreover, using a digital cytometry approach, we estimate relative changes in abundance of different cell types and find that the response of several immune cell types is time- and altitude dependent. Taken together, our data provide evidence for interaction between the transcriptional response to hypoxia and the time of day in humans. Low oxygen availability upon high altitude vastly affects human blood transcriptome The transcriptomic changes upon altitude elevation are not necessarily monotonic The daily variance in gene expression is dependent on altitude The response of several immune cell types is time- and altitude dependent
Collapse
Affiliation(s)
- Gal Manella
- Department of Biomolecular Sciences, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Saar Ezagouri
- Department of Biomolecular Sciences, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Benoit Champigneulle
- HP2 Laboratory, INSERM U1300, Grenoble Alpes University, CHU Grenoble Alpes, Grenoble, France
| | - Jonathan Gaucher
- HP2 Laboratory, INSERM U1300, Grenoble Alpes University, CHU Grenoble Alpes, Grenoble, France
| | - Monique Mendelson
- HP2 Laboratory, INSERM U1300, Grenoble Alpes University, CHU Grenoble Alpes, Grenoble, France
| | - Emeline Lemarie
- HP2 Laboratory, INSERM U1300, Grenoble Alpes University, CHU Grenoble Alpes, Grenoble, France
| | - Emeric Stauffer
- Laboratoire Interuniversitaire de Biologie de la Motricité (LIBM) EA7424, Team "Biologie vasculaire et du globule rouge", Université Claude Bernard Lyon 1, Université de Lyon, France; Laboratoire d'Excellence du Globule Rouge (Labex GR-Ex), PRES Sorbonne, Paris, France
| | - Aurélien Pichon
- Laboratoire MOVE, STAPS, Université de Poitiers, Poitiers, France
| | - Connor A Howe
- Centre for Heart, Lung and Vascular Health, School of Health and Exercise Sciences, University of British Columbia - Okanagan, Kelowna, BC, Canada
| | - Stéphane Doutreleau
- HP2 Laboratory, INSERM U1300, Grenoble Alpes University, CHU Grenoble Alpes, Grenoble, France
| | - Marina Golik
- Department of Biomolecular Sciences, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Samuel Verges
- HP2 Laboratory, INSERM U1300, Grenoble Alpes University, CHU Grenoble Alpes, Grenoble, France.
| | - Gad Asher
- Department of Biomolecular Sciences, Weizmann Institute of Science, 7610001 Rehovot, Israel.
| |
Collapse
|
19
|
Kiessling E, Peters F, Ebner LJ, Merolla L, Samardzija M, Baumgartner MR, Grimm C, Froese DS. HIF1 and DROSHA are involved in MMACHC repression in hypoxia. Biochim Biophys Acta Gen Subj 2022; 1866:130175. [DOI: 10.1016/j.bbagen.2022.130175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 05/03/2022] [Accepted: 05/23/2022] [Indexed: 11/25/2022]
|
20
|
Davis L, Recktenwald M, Hutt E, Fuller S, Briggs M, Goel A, Daringer N. Targeting HIF-2α in the Tumor Microenvironment: Redefining the Role of HIF-2α for Solid Cancer Therapy. Cancers (Basel) 2022; 14:1259. [PMID: 35267567 PMCID: PMC8909461 DOI: 10.3390/cancers14051259] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/26/2022] [Accepted: 02/26/2022] [Indexed: 02/06/2023] Open
Abstract
Inadequate oxygen supply, or hypoxia, is characteristic of the tumor microenvironment and correlates with poor prognosis and therapeutic resistance. Hypoxia leads to the activation of the hypoxia-inducible factor (HIF) signaling pathway and stabilization of the HIF-α subunit, driving tumor progression. The homologous alpha subunits, HIF-1α and HIF-2α, are responsible for mediating the transcription of a multitude of critical proteins that control proliferation, angiogenic signaling, metastasis, and other oncogenic factors, both differentially and sequentially regulating the hypoxic response. Post-translational modifications of HIF play a central role in its behavior as a mediator of transcription, as well as the temporal transition from HIF-1α to HIF-2α that occurs in response to chronic hypoxia. While it is evident that HIF-α is highly dynamic, HIF-2α remains vastly under-considered. HIF-2α can intensify the behaviors of the most aggressive tumors by adapting the cell to oxidative stress, thereby promoting metastasis, tissue remodeling, angiogenesis, and upregulating cancer stem cell factors. The structure, function, hypoxic response, spatiotemporal dynamics, and roles in the progression and persistence of cancer of this HIF-2α molecule and its EPAS1 gene are highlighted in this review, alongside a discussion of current therapeutics and future directions.
Collapse
Affiliation(s)
- Leah Davis
- Department of Biomedical Engineering, Rowan University, 201 Mullica Hill Rd, Glassboro, NJ 08028, USA
| | - Matthias Recktenwald
- Department of Biomedical Engineering, Rowan University, 201 Mullica Hill Rd, Glassboro, NJ 08028, USA
| | - Evan Hutt
- Department of Biomedical Engineering, Rowan University, 201 Mullica Hill Rd, Glassboro, NJ 08028, USA
| | - Schuyler Fuller
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Madison Briggs
- Department of Biomedical Engineering, Rowan University, 201 Mullica Hill Rd, Glassboro, NJ 08028, USA
| | - Arnav Goel
- Department of Biomedical Engineering, Rowan University, 201 Mullica Hill Rd, Glassboro, NJ 08028, USA
| | - Nichole Daringer
- Department of Biomedical Engineering, Rowan University, 201 Mullica Hill Rd, Glassboro, NJ 08028, USA
| |
Collapse
|
21
|
Liu Q, Palmgren VA, Danen EHJ, Le Dévédec SE. Acute vs. chronic vs. intermittent hypoxia in breast Cancer: a review on its application in in vitro research. Mol Biol Rep 2022; 49:10961-10973. [PMID: 36057753 PMCID: PMC9618509 DOI: 10.1007/s11033-022-07802-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 07/15/2022] [Indexed: 11/25/2022]
Abstract
Hypoxia has been linked to elevated instances of therapeutic resistance in breast cancer. The exposure of proliferating cancer cells to hypoxia has been shown to induce an aggressive phenotype conducive to invasion and metastasis. Regions of the primary tumors in the breast may be exposed to different types of hypoxia including acute, chronic or intermittent. Intermittent hypoxia (IH), also called cyclic hypoxia, is caused by exposure to cycles of hypoxia and reoxygenation (H-R cycles). Importantly, there is currently no consensus amongst the scientific community on the total duration of hypoxia, the oxygen level, and the possible presence of H-R cycles. In this review, we discuss current methods of hypoxia research, to explore how exposure regimes used in experiments are connected to signaling by different hypoxia inducible factors (HIFs) and to distinct cellular responses in the context of the hallmarks of cancer. We highlight discrepancies in the existing literature on hypoxia research within the field of breast cancer in particular and propose a clear definition of acute, chronic, and intermittent hypoxia based on HIF activation and cellular responses: (i) acute hypoxia is when the cells are exposed for no more than 24 h to an environment with 1% O2 or less; (ii) chronic hypoxia is when the cells are exposed for more than 48 h to an environment with 1% O2 or less and (iii) intermittent hypoxia is when the cells are exposed to at least two rounds of hypoxia (1% O2 or less) separated by at least one period of reoxygenation by exposure to normoxia (8.5% O2 or higher). Our review provides for the first time a guideline for definition of hypoxia related terms and a clear foundation for hypoxia related in vitro (breast) cancer research.
Collapse
Affiliation(s)
- Qiuyu Liu
- Division of Drug Discovery and Safety, Leiden Academic Centre of Drug Research, Leiden University, Leiden, The Netherlands
| | - Victoria A.C. Palmgren
- Division of Drug Discovery and Safety, Leiden Academic Centre of Drug Research, Leiden University, Leiden, The Netherlands
| | - Erik HJ Danen
- Division of Drug Discovery and Safety, Leiden Academic Centre of Drug Research, Leiden University, Leiden, The Netherlands
| | - Sylvia E. Le Dévédec
- Division of Drug Discovery and Safety, Leiden Academic Centre of Drug Research, Leiden University, Leiden, The Netherlands
| |
Collapse
|
22
|
A PDMS-Based Interdigitated Platform for Trophoblast Invasion Study Under Oxygen Stress Conditions. BIOCHIP JOURNAL 2021. [DOI: 10.1007/s13206-021-00035-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
23
|
Shen T, Xia W, Min S, Yang Z, Cheng L, Wang W, Zhan Q, Shao F, Zhang X, Wang Z, Zhang Y, Shen G, Zhang H, Wu LL, Yu GY, Kong QP, Wang X. A pair of long intergenic non-coding RNA LINC00887 variants act antagonistically to control Carbonic Anhydrase IX transcription upon hypoxia in tongue squamous carcinoma progression. BMC Biol 2021; 19:192. [PMID: 34493285 PMCID: PMC8422755 DOI: 10.1186/s12915-021-01112-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 07/30/2021] [Indexed: 12/11/2022] Open
Abstract
Background Long noncoding RNAs (lncRNAs) are important regulators in tumor progression. However, their biological functions and underlying mechanisms in hypoxia adaptation remain largely unclear. Results Here, we established a correlation between a Chr3q29-derived lncRNA gene and tongue squamous carcinoma (TSCC) by genome-wide analyses. Using RACE, we determined that two novel variants of this lncRNA gene are generated in TSCC, namely LINC00887_TSCC_short (887S) and LINC00887_TSCC_long (887L). RNA-sequencing in 887S or 887L loss-of-function cells identified their common downstream target as Carbonic Anhydrase IX (CA9), a gene known to be upregulated by hypoxia during tumor progression. Mechanistically, our results showed that the hypoxia-augmented 887S and constitutively expressed 887L functioned in opposite directions on tumor progression through the common target CA9. Upon normoxia, 887S and 887L interacted. Upon hypoxia, the two variants were separated. Each RNA recognized and bound to their responsive DNA cis-acting elements on CA9 promoter: 887L activated CA9’s transcription through recruiting HIF1α, while 887S suppressed CA9 through DNMT1-mediated DNA methylation. Conclusions We provided hypoxia-permitted functions of two antagonistic lncRNA variants to fine control the hypoxia adaptation through CA9. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-021-01112-2.
Collapse
Affiliation(s)
- Tao Shen
- Department of Geriatrics, Gerontology Institute of Anhui Province, The First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Anhui Provincial Key Laboratory of Tumor Immunotherapy and Nutrition Therapy, Hefei, China.,Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, China
| | - Wangxiao Xia
- State Key Laboratory of Genetic Resources and Evolution/Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, The Chinese Academy of Sciences, Kunming, 650223, China
| | - Sainan Min
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Zixuan Yang
- Department of Geriatrics, Gerontology Institute of Anhui Province, The First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Anhui Provincial Key Laboratory of Tumor Immunotherapy and Nutrition Therapy, Hefei, China.,Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, China
| | - Lehua Cheng
- State Key Laboratory of Genetic Resources and Evolution/Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, The Chinese Academy of Sciences, Kunming, 650223, China
| | - Wei Wang
- Department of Geriatrics, Gerontology Institute of Anhui Province, The First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Anhui Provincial Key Laboratory of Tumor Immunotherapy and Nutrition Therapy, Hefei, China.,Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, China
| | - Qianxi Zhan
- Department of Geriatrics, Gerontology Institute of Anhui Province, The First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Anhui Provincial Key Laboratory of Tumor Immunotherapy and Nutrition Therapy, Hefei, China.,Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, China
| | - Fanghong Shao
- Department of Geriatrics, Gerontology Institute of Anhui Province, The First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Anhui Provincial Key Laboratory of Tumor Immunotherapy and Nutrition Therapy, Hefei, China.,Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, China
| | - Xuehan Zhang
- Department of Geriatrics, Gerontology Institute of Anhui Province, The First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Anhui Provincial Key Laboratory of Tumor Immunotherapy and Nutrition Therapy, Hefei, China.,Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, China
| | - Zhiyu Wang
- Department of Medical Oncology, Affiliated Hospital of Hebei University, Baoding, 071000, China
| | - Yan Zhang
- School of Health Services Management, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Guodong Shen
- Department of Geriatrics, Gerontology Institute of Anhui Province, The First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Anhui Provincial Key Laboratory of Tumor Immunotherapy and Nutrition Therapy, Hefei, China
| | - Huafeng Zhang
- Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, China
| | - Li-Ling Wu
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, 100191, China
| | - Guang-Yan Yu
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Qing-Peng Kong
- State Key Laboratory of Genetic Resources and Evolution/Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, The Chinese Academy of Sciences, Kunming, 650223, China. .,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China. .,KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming, 650223, China.
| | - Xiangting Wang
- Department of Geriatrics, Gerontology Institute of Anhui Province, The First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China. .,Anhui Provincial Key Laboratory of Tumor Immunotherapy and Nutrition Therapy, Hefei, China. .,Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
24
|
David BT, Curtin JJ, Brown JL, Coutts DJC, Boles NC, Hill CE. Treatment with hypoxia-mimetics protects cultured rat Schwann cells against oxidative stress-induced cell death. Glia 2021; 69:2215-2234. [PMID: 34019306 PMCID: PMC11848739 DOI: 10.1002/glia.24019] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 05/05/2021] [Accepted: 05/06/2021] [Indexed: 12/14/2022]
Abstract
Schwann cell (SC) grafts promote axon regeneration in the injured spinal cord, but transplant efficacy is diminished by a high death rate in the first 2-3 days postimplantation. Both hypoxic preconditioning and pharmacological induction of the cellular hypoxic response can drive cellular adaptations and improve transplant survival in a number of disease/injury models. Hypoxia-inducible factor 1 alpha (HIF-1α), a regulator of the cellular response to hypoxia, is implicated in preconditioning-associated protection. HIF-1α cellular levels are regulated by the HIF-prolyl hydroxylases (HIF-PHDs). Pharmacological inhibition of the HIF-PHDs mimics hypoxic preconditioning and provides a method to induce adaptive hypoxic responses without direct exposure to hypoxia. In this study, we show that hypoxia-mimetics, deferoxamine (DFO) and adaptaquin (AQ), enhance HIF-1α stability and HIF-1α target gene expression. Expression profiling of hypoxia-related genes demonstrates that HIF-dependent and HIF-independent expression changes occur. Analyses of transcription factor binding sites identify several candidate transcriptional co-regulators that vary in SCs along with HIF-1α. Using an in vitro model system, we show that hypoxia-mimetics are potent blockers of oxidative stress-induced death in SCs. In contrast, traditional hypoxic preconditioning was not protective. The robust protection induced by pharmacological preconditioning, particularly with DFO, indicates that pharmacological induction of hypoxic adaptations could be useful for promoting transplanted SC survival. These agents may also be more broadly useful for protecting SCs, as oxidative stress is a major pathway that drives cellular damage in the context of neurological injury and disease, including demyelinating diseases and peripheral neuropathies.
Collapse
Affiliation(s)
- Brian T. David
- Burke Neurological Institute, White Plains, New York
- Weill Cornell Medicine, Feil Family Brain and Mind Research Institute, New York, New York
| | - Jessica J. Curtin
- Burke Neurological Institute, White Plains, New York
- Weill Cornell Medicine, Feil Family Brain and Mind Research Institute, New York, New York
| | - Jennifer L. Brown
- Burke Neurological Institute, White Plains, New York
- Weill Cornell Medicine, Feil Family Brain and Mind Research Institute, New York, New York
| | - David J. C. Coutts
- Burke Neurological Institute, White Plains, New York
- Weill Cornell Medicine, Feil Family Brain and Mind Research Institute, New York, New York
| | | | - Caitlin E. Hill
- Burke Neurological Institute, White Plains, New York
- Weill Cornell Medicine, Feil Family Brain and Mind Research Institute, New York, New York
- Neural Stem Cell Institute, Rensselaer, New York
| |
Collapse
|
25
|
Ma X, Wang C, Chen J, Wei D, Yu F, Sun J. circAGFG1 sponges miR-28-5p to promote non-small-cell lung cancer progression through modulating HIF-1α level. Open Med (Wars) 2021; 16:703-717. [PMID: 34013042 PMCID: PMC8111482 DOI: 10.1515/med-2021-0269] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 03/18/2021] [Accepted: 03/22/2021] [Indexed: 12/14/2022] Open
Abstract
Circular RNAs (circRNAs) have gained much attention for their crucial regulatory roles in human diseases and cancers. However, the role and the mechanism of circRNA ArfGAP with FG repeats 1 (circAGFG1) in non-small-cell lung cancer (NSCLC) are still largely unknown. circAGFG1 was highly expressed in NSCLC, and high expression of circAGFG1 was closely related to the low survival rate of NSCLC patients. circAGFG1 knockdown inhibited the proliferation, migration, and invasion and promoted the apoptosis of NSCLC cells. circAGFG1 bound to miR-28-5p in NSCLC cells, and circAGFG1 promoted NSCLC progression partly through sponging miR-28-5p in vitro. HIF-1α was a target of miR-28-5p, and miR-28-5p overexpression-mediated influences in NSCLC cells were partly overturned by the addition of HIF-1α overexpression plasmid. circAGFG1/miR-28-5p/HIF-1α axis regulated cellular glycolytic metabolism in NSCLC cells. circAGFG1 silencing restrained the xenograft tumor growth in vivo. circAGFG1 promoted the proliferation, migration, and invasion and suppressed the apoptosis of NSCLC cells through accelerating the glycolysis via miR-28-5p/HIF-1α axis.
Collapse
Affiliation(s)
- Xiaoan Ma
- Department of Respiratory Medicine, No. 215 Hospital of Shaanxi Nuclear Industry, Xianyang 712000, Shannxi, China
| | - Cuijie Wang
- Department of Respiratory Medicine, No. 215 Hospital of Shaanxi Nuclear Industry, Xianyang 712000, Shannxi, China
| | - Juan Chen
- Department of Respiratory Medicine, No. 215 Hospital of Shaanxi Nuclear Industry, Xianyang 712000, Shannxi, China
| | - Dan Wei
- Department of Respiratory Medicine, No. 215 Hospital of Shaanxi Nuclear Industry, Xianyang 712000, Shannxi, China
| | - Fei Yu
- Department of Respiratory Medicine, No. 215 Hospital of Shaanxi Nuclear Industry, Xianyang 712000, Shannxi, China
| | - Juan Sun
- Department of Respiratory Medicine, No. 215 Hospital of Shaanxi Nuclear Industry, Xianyang 712000, Shannxi, China
| |
Collapse
|
26
|
Cerezo-Magaña M, Christianson HC, van Kuppevelt TH, Forsberg-Nilsson K, Belting M. Hypoxic Induction of Exosome Uptake through Proteoglycan-Dependent Endocytosis Fuels the Lipid Droplet Phenotype in Glioma. Mol Cancer Res 2020; 19:528-540. [PMID: 33288734 DOI: 10.1158/1541-7786.mcr-20-0560] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 10/13/2020] [Accepted: 12/02/2020] [Indexed: 11/16/2022]
Abstract
As an adaptive response to hypoxic stress, aggressive tumors rewire their metabolic phenotype into increased malignant behavior through extracellular lipid scavenging and storage in lipid droplets (LD). However, the underlying mechanisms and potential lipid source retrieved in the hypoxic tumor microenvironment remain poorly understood. Here, we show that exosome-like extracellular vesicles (EV), known as influential messengers in the tumor microenvironment, may also serve anabolic functions by transforming hypoxic, patient-derived human glioblastoma cell lines into the LD+ phenotype. EVs were internalized via a hypoxia-sensitive, endocytic mechanism that fueled LD formation through direct lipid transfer, and independently of fatty acid synthase activity. EVs can enter cells through multiple and yet ill-defined pathways. On a mechanistic level, we found that hypoxia-mediated EV uptake depends on increased heparan sulfate proteoglycan (HSPG) endocytosis that preferentially followed the lipid raft pathway. The functional relevance of HSPG was evidenced by the reversal of EV-mediated LD loading by targeting of HSPG receptor function. IMPLICATIONS: Together, our data extend the multifaceted role of EVs in cancer biology by showing their LD-inducing capacity in hypoxic glioma cells. Moreover, these findings highlight a potential function for HSPG-mediated endocytosis as a salvage pathway for EV retrieval during tumor stress conditions.
Collapse
Affiliation(s)
- Myriam Cerezo-Magaña
- Department of Clinical Sciences, Section of Oncology, Lund University, Lund, Sweden
| | | | - Toin H van Kuppevelt
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands
| | - Karin Forsberg-Nilsson
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Mattias Belting
- Department of Clinical Sciences, Section of Oncology, Lund University, Lund, Sweden. .,Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden.,Skåne University Hospital, Lund, Sweden
| |
Collapse
|
27
|
Kang GJ, Xie A, Liu H, Dudley SC. MIR448 antagomir reduces arrhythmic risk after myocardial infarction by upregulating the cardiac sodium channel. JCI Insight 2020; 5:140759. [PMID: 33108349 PMCID: PMC7714400 DOI: 10.1172/jci.insight.140759] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 10/21/2020] [Indexed: 12/17/2022] Open
Abstract
Cardiac ischemia is associated with arrhythmias; however, effective therapies are currently limited. The cardiac voltage-gated sodium channel α subunit (SCN5A), encoding the Nav1.5 current, plays a key role in the cardiac electrical conduction and arrhythmic risk. Here, we show that hypoxia reduces Nav1.5 through effects on a miR, miR-448. miR-448 expression is increased in ischemic cardiomyopathy. miR-448 has a conserved binding site in 3′-UTR of SCN5A. miR-448 binding to this site suppressed SCN5A expression and sodium currents. Hypoxia-induced HIF-1α and NF-κB were major transcriptional regulators for MIR448. Moreover, hypoxia relieved MIR448 transcriptional suppression by RE1 silencing transcription factor. Therefore, miR-448 inhibition reduced arrhythmic risk after myocardial infarction. Here, we show that ischemia drove miR-448 expression, reduced Nav1.5 current, and increased arrhythmic risk. Arrhythmic risk was improved by preventing Nav1.5 downregulation, suggesting a new approach to antiarrhythmic therapy. Ischemic induction of miR-448 negatively regulates the cardiac sodium channel Nav1.5, and inhibiting miR-448 raises Nav1.5 and reduces arrhythmic risk after myocardial infarction in mice.
Collapse
|
28
|
Nimje MA, Patir H, Tirpude RK, Reddy PK, Kumar B. Physiological and oxidative stress responses to intermittent hypoxia training in Sprague Dawley rats. Exp Lung Res 2020; 46:376-392. [PMID: 32930002 DOI: 10.1080/01902148.2020.1821263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
AIM Rapid ascent to high altitude and inability to acclimatize lead to high-altitude illnesses. Intermittent hypoxia (IH) conditioning has been hypothesized as a non-pharmacological strategy aiming to improve adaptive responses during high altitude ascent. In the recent years, IH training (IHT) has become increasingly popular among recreational and professional athletes owing to its ability to mitigate high altitude related problems. This study aimed at exploring the role of IHT in altitude acclimatization. METHODS Male Sprague Dawley rats were subjected to IHT for 4 h consecutively for 5 days at 12% FiO2 under normobaric conditions. To assess the effect of IHT in hypoxic acclimatization, animals were further exposed to extreme hypoxia (EH) at 8% FiO2. Oxygen saturation (SpO2), respiratory rate and heart rate were recorded during the exposure. Oxidative stress (ROS, MDA, and 4-HNE) and histopathological examinations were studied in the lung tissue sections. Hypoxia biomarkers, HIF-1α, EPO, VEGF, and BPGM were evaluated through western blotting in the lung tissue. RESULTS Assessment of the IHT showed that SpO2 levels were found to be higher in the IH trained rats with a statistical difference of p < 0.01 in the first hour of hypoxia exposure as compared to the untrained rats. There was a significantly higher (p < 0.001) generation of ROS and MDA in the untrained rats as compared to the trained rats. Lipid peroxidation markers and systemic inflammatory marker were found to be expressed at much higher level in the untrained rats. There was a higher expression of HIF-1α (1.24-fold ↑), VEGF (1.14-fold ↑) and decrease in EPO (1.43-fold ↓) in the untrained rats as compared to trained rats. CONCLUSIONS Preconditioning with IHT resulted in the reduction in hypoxia induced oxidative stress during extreme hypoxia exposure and thus, maintaining redox balance as well as adjustment in the physiological changes in rats.
Collapse
Affiliation(s)
- Megha A Nimje
- Defence Institute of Physiology and Allied Sciences (DIPAS), (DRDO), Timarpur, Delhi, India
| | - Himadri Patir
- Defence Institute of Physiology and Allied Sciences (DIPAS), (DRDO), Timarpur, Delhi, India
| | - Rajesh Kumar Tirpude
- Defence Institute of Physiology and Allied Sciences (DIPAS), (DRDO), Timarpur, Delhi, India
| | - Prasanna K Reddy
- Defence Institute of Physiology and Allied Sciences (DIPAS), (DRDO), Timarpur, Delhi, India
| | - Bhuvnesh Kumar
- Defence Institute of Physiology and Allied Sciences (DIPAS), (DRDO), Timarpur, Delhi, India
| |
Collapse
|
29
|
Bustelo M, Barkhuizen M, van den Hove DLA, Steinbusch HWM, Bruno MA, Loidl CF, Gavilanes AWD. Clinical Implications of Epigenetic Dysregulation in Perinatal Hypoxic-Ischemic Brain Damage. Front Neurol 2020; 11:483. [PMID: 32582011 PMCID: PMC7296108 DOI: 10.3389/fneur.2020.00483] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 05/04/2020] [Indexed: 12/18/2022] Open
Abstract
Placental and fetal hypoxia caused by perinatal hypoxic-ischemic events are major causes of stillbirth, neonatal morbidity, and long-term neurological sequelae among surviving neonates. Brain hypoxia and associated pathological processes such as excitotoxicity, apoptosis, necrosis, and inflammation, are associated with lasting disruptions in epigenetic control of gene expression contributing to neurological dysfunction. Recent studies have pointed to DNA (de)methylation, histone modifications, and non-coding RNAs as crucial components of hypoxic-ischemic encephalopathy (HIE). The understanding of epigenetic dysregulation in HIE is essential in the development of new clinical interventions for perinatal HIE. Here, we summarize our current understanding of epigenetic mechanisms underlying the molecular pathology of HI brain damage and its clinical implications in terms of new diagnostic, prognostic, and therapeutic tools.
Collapse
Affiliation(s)
- Martín Bustelo
- Department of Pediatrics, Maastricht University Medical Center (MUMC), Maastricht, Netherlands.,Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, Netherlands.,Instituto de Ciencias Biomédicas, Facultad de Ciencias Médicas, Universidad Católica de Cuyo, San Juan, Argentina.,Laboratorio de Neuropatología Experimental, Facultad de Medicina, Instituto de Biología Celular y Neurociencias "Prof. E. De Robertis" (IBCN), Universidad de Buenos Aires, CONICET, Buenos Aires, Argentina
| | - Melinda Barkhuizen
- Department of Pediatrics, Maastricht University Medical Center (MUMC), Maastricht, Netherlands
| | - Daniel L A van den Hove
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, Netherlands.,Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Würzburg, Germany
| | - Harry Wilhelm M Steinbusch
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, Netherlands
| | - Martín A Bruno
- Instituto de Ciencias Biomédicas, Facultad de Ciencias Médicas, Universidad Católica de Cuyo, San Juan, Argentina
| | - C Fabián Loidl
- Instituto de Ciencias Biomédicas, Facultad de Ciencias Médicas, Universidad Católica de Cuyo, San Juan, Argentina.,Laboratorio de Neuropatología Experimental, Facultad de Medicina, Instituto de Biología Celular y Neurociencias "Prof. E. De Robertis" (IBCN), Universidad de Buenos Aires, CONICET, Buenos Aires, Argentina
| | - Antonio W Danilo Gavilanes
- Department of Pediatrics, Maastricht University Medical Center (MUMC), Maastricht, Netherlands.,Facultad de Ciencias Médicas, Instituto de Investigación e Innovación de Salud Integral, Universidad Católica de Santiago de Guayaquil, Guayaquil, Ecuador
| |
Collapse
|
30
|
Sadeghi F, Kardar GA, Bolouri MR, Nasri F, Sadri M, Falak R. Overexpression of bHLH domain of HIF-1 failed to inhibit the HIF-1 transcriptional activity in hypoxia. Biol Res 2020; 53:25. [PMID: 32503642 PMCID: PMC7275393 DOI: 10.1186/s40659-020-00293-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 05/18/2020] [Indexed: 01/22/2023] Open
Abstract
Background Hypoxia inducible factor-1 (HIF-1) is considered as the most activated transcriptional factor in response to low oxygen level or hypoxia. HIF-1 binds the hypoxia response element (HRE) sequence in the promoter of different genes, mainly through the bHLH domain and activates the transcription of genes, especially those involved in angiogenesis and EMT. Considering the critical role of bHLH in binding HIF-1 to the HRE sequence, we hypothesized that bHLH could be a promising candidate to be targeted in hypoxia condition. Methods We inserted an inhibitory bHLH (ibHLH) domain in a pIRES2-EGFP vector and transfected HEK293T cells with either the control vector or the designed construct. The ibHLH domain consisted of bHLH domains of both HIF-1a and Arnt, capable of competing with HIF-1 in binding to HRE sequences. The transfected cells were then treated with 200 µM of cobalt chloride (CoCl2) for 48 h to induce hypoxia. Real-time PCR and western blot were performed to evaluate the effect of ibHLH on the genes and proteins involved in angiogenesis and EMT. Results Hypoxia was successfully induced in the HEK293T cell line as the gene expression of VEGF, vimentin, and β-catenin were significantly increased after treatment of untransfected HEK293T cells with 200 µM CoCl2. The gene expression of VEGF, vimentin, and β-catenin and protein level of β-catenin were significantly decreased in the cells transfected with either control or ibHLH vectors in hypoxia. However, ibHLH failed to be effective on these genes and the protein level of β-catenin, when compared to the control vector. We also observed that overexpression of ibHLH had more inhibitory effect on gene and protein expression of N-cadherin compared to the control vector. However, it was not statistically significant. Conclusion bHLH has been reported to be an important domain involved in the DNA binding activity of HIF. However, we found that targeting this domain is not sufficient to inhibit the endogenous HIF-1 transcriptional activity. Further studies about the function of critical domains of HIF-1 are necessary for developing a specific HIF-1 inhibitor.
Collapse
Affiliation(s)
- Fatemeh Sadeghi
- Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Gholam Ali Kardar
- Immunology Asthma & Allergy Research Institute, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mohammad Reza Bolouri
- Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Farzad Nasri
- Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Sadri
- Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Falak
- Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran. .,Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
31
|
Abstract
Human survival is dependent upon the continuous delivery of O2 to each cell in the body in sufficient amounts to meet metabolic requirements, primarily for ATP generation by oxidative phosphorylation. Hypoxia-inducible factors (HIFs) regulate the transcription of thousands of genes to balance O2 supply and demand. The HIFs are negatively regulated by O2-dependent hydrox-ylation and ubiquitination by prolyl hydroxylase domain (PHD) proteins and the von Hippel-Lindau (VHL) protein. Germline mutations in the genes encoding VHL, HIF-2α, and PHD2 cause hereditary erythrocytosis, which is characterized by polycythemia and pulmonary hypertension and is caused by increased HIF activity. Evolutionary adaptation to life at high altitude is associated with unique genetic variants in the genes encoding HIF-2α and PHD2 that blunt the erythropoietic and pulmonary vascular responses to hypoxia.
Collapse
Affiliation(s)
- Gregg L Semenza
- Departments of Genetic Medicine, Oncology, Pediatrics, Radiation Oncology, Medicine, and Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA;
| |
Collapse
|
32
|
Jia Y, Guo Y, Jin Q, Qu H, Qi D, Song P, Zhang X, Wang X, Xu W, Dong Y, Liang Y, Quan C. A SUMOylation-dependent HIF-1α/CLDN6 negative feedback mitigates hypoxia-induced breast cancer metastasis. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:42. [PMID: 32093760 PMCID: PMC7038627 DOI: 10.1186/s13046-020-01547-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 02/14/2020] [Indexed: 12/21/2022]
Abstract
Background We have previously described CLDN6 as a tumor suppressor gene in breast cancer. Here, a new finding is that CLDN6 was upregulated under hypoxia, a commonly recognized factor that promotes tumor metastasis. In this study, we aim to explain this confusing finding and delineate the role of CLDN6 in the breast cancer metastasis induced by hypoxia. Methods RNAi and ChIP assays were used to confirm that CLDN6 is transcriptional regulated by HIF-1α. mRNA seq and KEGG analysis were performed to define the downstream pathways of CLDN6. The roles of the CLDN6/SENP1/HIF-1α signaling on tumor metastasis were evaluated by function experiments and clinical samples. Finally, the possible transcription factor of SENP1 was suspected and then validated by ChIP assay. Results We demonstrated a previously unrecognized negative feedback loop exists between CLDN6 and HIF-1α. CLDN6 was transcriptionally up-regulated by HIF-1α under hypoxia. On the other hand, in cytoplasm CLDN6 combines and retains β-catenin, a transcription factor of SENP1, causing β-catenin degradation and preventing its nuclear translocation. This process reduced SENP1 expression and prevented the deSUMOylation of HIF-1α, ultimately leading to HIF-1α degradation and breast cancer metastasis suppression. Conclusions Our data provide a molecular mechanistic insight indicating that CLDN6 loss may lead to elevated HIF-1α-driven breast cancer metastasis in a SUMOylation-dependent manner.
Collapse
Affiliation(s)
- Yiyang Jia
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, Jilin, 130021, People's Republic of China
| | - Yantong Guo
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, Jilin, 130021, People's Republic of China
| | - Qiu Jin
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, Jilin, 130021, People's Republic of China
| | - Huinan Qu
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, Jilin, 130021, People's Republic of China
| | - Da Qi
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, Jilin, 130021, People's Republic of China
| | - Peiye Song
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, Jilin, 130021, People's Republic of China
| | - Xiaoli Zhang
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, Jilin, 130021, People's Republic of China
| | - Xinqi Wang
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, Jilin, 130021, People's Republic of China
| | - Wenhong Xu
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, Jilin, 130021, People's Republic of China
| | - Yuan Dong
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, Jilin, 130021, People's Republic of China
| | - Yingying Liang
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, Jilin, 130021, People's Republic of China
| | - Chengshi Quan
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, Jilin, 130021, People's Republic of China.
| |
Collapse
|
33
|
Bclaf1 is a direct target of HIF-1 and critically regulates the stability of HIF-1α under hypoxia. Oncogene 2020; 39:2807-2818. [PMID: 32029898 DOI: 10.1038/s41388-020-1185-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 12/10/2019] [Accepted: 01/23/2020] [Indexed: 01/04/2023]
Abstract
Hypoxic stress is intimately connected with tumor progression, with hypoxia-inducible factor-1α (HIF-1α) being a critical regulator in this process. HIF-1α is stabilized in response to hypoxia, which is required for the induction of gene transcriptions important for hypoxic adaptation. Bclaf1 is a multifunctional protein involved in tumorigenesis, however, its role in this process is not well characterized. Here we report Bclaf1 is a direct transcriptional target of HIF-1 and upregulated in multiple cell lines during hypoxia. Importantly, we found Bclaf1 is involved in the stabilization of HIF-1α during long-term hypoxic treatments. Compared with the control cells, the protein level and stability of HIF-1α in Bclaf1 knockdown or knockout cells is greatly compromised after long-term hypoxic treatments, concomitant with the impaired inductions of HIF-1 target gene transcription. Bclaf1 knockout HeLa cells exhibit a reduced tumor growth in mice xenografts, in which the expressions of HIF-1α and its target genes are also decreased. Bclaf1 binds to HIF-1α in the nucleus, and this interaction is required for Bclaf1 to stabilize HIF-1α in hypoxic condition. These results uncover a positive feedback loop, HIF-1-Bclaf1, that sustains HIF-1 activity during long-term hypoxic conditions by binding to and protecting HIF-1α from degradation, and suggest that Bclaf1 may promote tumor progression by enhancing HIF-1α stability.
Collapse
|
34
|
Mitochondrial MUL1 E3 ubiquitin ligase regulates Hypoxia Inducible Factor (HIF-1α) and metabolic reprogramming by modulating the UBXN7 cofactor protein. Sci Rep 2020; 10:1609. [PMID: 32005965 PMCID: PMC6994496 DOI: 10.1038/s41598-020-58484-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 12/18/2019] [Indexed: 11/15/2022] Open
Abstract
MUL1 is a multifunctional E3 ubiquitin ligase anchored in the outer mitochondrial membrane with its RING finger domain facing the cytoplasm. MUL1 participates in various biological pathways involved in apoptosis, mitochondrial dynamics, and innate immune response. The unique topology of MUL1 enables it to “sense” mitochondrial stress in the intermembrane mitochondrial space and convey these signals through the ubiquitination of specific cytoplasmic substrates. We have identified UBXN7, the cofactor protein of the CRL2VHL ligase complex, as a specific substrate of MUL1 ligase. CRL2VHL ligase complex regulates HIF-1α protein levels under aerobic (normoxia) or anaerobic (hypoxia) conditions. Inactivation of MUL1 ligase leads to accumulation of UBXN7, with concomitant increase in HIF-1α protein levels, reduction in oxidative phosphorylation, and increased glycolysis. We describe a novel pathway that originates in the mitochondria and operates upstream of the CRL2VHL ligase complex. Furthermore, we delineate the mechanism by which the mitochondria, through MUL1 ligase, can inhibit the CRL2VHL complex leading to high HIF-1α protein levels and a metabolic shift to glycolysis under normoxic conditions.
Collapse
|
35
|
McQuaig R, Dixit P, Yamauchi A, Van Hout I, Papannarao JB, Bunton R, Parry D, Davis P, Katare R. Combination of Cardiac Progenitor Cells From the Right Atrium and Left Ventricle Exhibits Synergistic Paracrine Effects In Vitro. Cell Transplant 2020; 29:963689720972328. [PMID: 33153286 PMCID: PMC7784587 DOI: 10.1177/0963689720972328] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 10/14/2020] [Accepted: 10/19/2020] [Indexed: 12/26/2022] Open
Abstract
Cardiovascular diseases, such as ischemic heart disease, remain the most common cause of death worldwide. Regenerative medicine with stem cell therapy is a promising tool for cardiac repair. Combination of different cell types has been shown to improve the therapeutic potential, which is thought to be due to synergistic or complimentary reparative effects. We investigated if the combination of cardiac progenitor cells (CPCs) of right atrial appendage (RAA) and left ventricle (LV) that are isolated from the same patient exert synergistic or complimentary paracrine effects for apoptotic cell death and angiogenesis in an in vitro model. Flow cytometry analysis showed that both RAA and LV CPCs expressed the mesenchymal cell markers CD90 and CD105, and were predominantly negative for the hematopoietic cell marker, CD34. Analysis of conditioned media (CM) collected from the CPCs cultured either alone or in combination in serum-deprived hypoxic conditions to simulate ischemia showed marked increase in the level of pro-survival hepatocyte growth factor and pro-angiogenic vascular endothelial growth factor-A in the combined RAA and LV CPC group. Next, to determine the therapeutic potential of CM, AC16 human ventricular cardiomyocytes and human umbilical vein endothelial cells (HUVECs) were treated with CM. Results showed a significant reduction in hypoxia-induced apoptosis of human cardiomyocytes treated with CM collected from combined RAA and LV CPC group. Similarly, matrigel assay showed a significantly increased tube length formed by HUVECs when treated with CM from combined RAA and LV CPC group. Our study provided evidence that the combination of RAA CPCs and LV CPCs may have superior therapeutic effects due to synergistic paracrine effects for cardiac repair. Therefore, in vivo studies are warranted to determine if a combination of different stem cell types have greater therapeutic potential than single-cell therapies.
Collapse
Affiliation(s)
- Ryan McQuaig
- Department of Physiology-HeartOtago, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Parul Dixit
- Department of Physiology-HeartOtago, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Atsushi Yamauchi
- Department of Physiology-HeartOtago, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Isabelle Van Hout
- Department of Physiology-HeartOtago, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Jayanthi Bellae Papannarao
- Department of Physiology-HeartOtago, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Richard Bunton
- Department of Cardiothoracic Surgery and Medicine, Dunedin School of Medicine, University of Otago, New Zealand
| | - Dominic Parry
- Department of Cardiothoracic Surgery and Medicine, Dunedin School of Medicine, University of Otago, New Zealand
| | - Philip Davis
- Department of Cardiothoracic Surgery and Medicine, Dunedin School of Medicine, University of Otago, New Zealand
| | - Rajesh Katare
- Department of Physiology-HeartOtago, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
36
|
Bhuria V, Xing J, Scholta T, Bui KC, Nguyen MLT, Malek NP, Bozko P, Plentz RR. Hypoxia induced Sonic Hedgehog signaling regulates cancer stemness, epithelial-to-mesenchymal transition and invasion in cholangiocarcinoma. Exp Cell Res 2019; 385:111671. [PMID: 31634481 DOI: 10.1016/j.yexcr.2019.111671] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 10/04/2019] [Accepted: 10/05/2019] [Indexed: 12/17/2022]
Abstract
Aberrant activation of Sonic Hedgehog (SHH) pathway has been implicated in a variety of cancers including cholangiocarcinoma (CC); however, the influencing factors are still unknown. Additionally, intratumoral hypoxia is known to contribute towards therapeutic resistance through modulatory effects on various pathways. In this study, we investigated the relationship between hypoxia and SHH pathway activation and the effect of this interplay on cancer stemness and epithelial-to- mesenchymal transition (EMT) during cholangiocarcinogenesis. Hypoxia promoted SHH pathway activation, evidenced by upregulated SHH and SMO levels, and enhanced glioma-associated oncogene homolog 1 (GLI1) nuclear translocation; whereas silencing of HIF-1α impaired SHH upregulation. Hypoxia also enhanced the expression of cancer stem cell (CSC) transcription factors (NANOG, Oct4, SOX2), CD133 and EMT markers (N-cadherin, Vimentin), thereby supporting invasion. Cyclopamine treatment suppressed hypoxia induced SHH pathway activation, consequently reducing invasiveness by downregulating the expression of CSC transcription factors, CD133 and EMT. Cyclopamine induced apoptosis in CC cells under hypoxia, suggesting that hypoxia induced activation of SHH pathway has modulatory effects on CC progression. Therefore, SHH signaling is proposed as a target for CC treatment, which is refractory to standard chemotherapy.
Collapse
Affiliation(s)
- Vikas Bhuria
- Department of Internal Medicine I, Medical University Hospital, Tübingen, Germany
| | - Jun Xing
- Department of Internal Medicine I, Medical University Hospital, Tübingen, Germany
| | - Tim Scholta
- Department of Internal Medicine I, Medical University Hospital, Tübingen, Germany
| | - Khac Cuong Bui
- Department of Internal Medicine I, Medical University Hospital, Tübingen, Germany
| | - Mai Ly Thi Nguyen
- Department of Internal Medicine I, Medical University Hospital, Tübingen, Germany
| | - Nisar P Malek
- Department of Internal Medicine I, Medical University Hospital, Tübingen, Germany
| | - Przemyslaw Bozko
- Department of Internal Medicine I, Medical University Hospital, Tübingen, Germany.
| | - Ruben R Plentz
- Department of Internal Medicine I, Medical University Hospital, Tübingen, Germany; Department of Internal Medicine II, Bremen-Nord Hospital, Bremen, Germany.
| |
Collapse
|
37
|
Saxena K, Jolly MK. Acute vs. Chronic vs. Cyclic Hypoxia: Their Differential Dynamics, Molecular Mechanisms, and Effects on Tumor Progression. Biomolecules 2019; 9:E339. [PMID: 31382593 PMCID: PMC6722594 DOI: 10.3390/biom9080339] [Citation(s) in RCA: 129] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 07/31/2019] [Accepted: 08/01/2019] [Indexed: 02/07/2023] Open
Abstract
Hypoxia has been shown to increase the aggressiveness and severity of tumor progression. Along with chronic and acute hypoxic regions, solid tumors contain regions of cycling hypoxia (also called intermittent hypoxia or IH). Cyclic hypoxia is mimicked in vitro and in vivo by periodic exposure to cycles of hypoxia and reoxygenation (H-R cycles). Compared to chronic hypoxia, cyclic hypoxia has been shown to augment various hallmarks of cancer to a greater extent: angiogenesis, immune evasion, metastasis, survival etc. Cycling hypoxia has also been shown to be the major contributing factor in increasing the risk of cancer in obstructive sleep apnea (OSA) patients. Here, we first compare and contrast the effects of acute, chronic and intermittent hypoxia in terms of molecular pathways activated and the cellular processes affected. We highlight the underlying complexity of these differential effects and emphasize the need to investigate various combinations of factors impacting cellular adaptation to hypoxia: total duration of hypoxia, concentration of oxygen (O2), and the presence of and frequency of H-R cycles. Finally, we summarize the effects of cycling hypoxia on various hallmarks of cancer highlighting their dependence on the abovementioned factors. We conclude with a call for an integrative and rigorous analysis of the effects of varying extents and durations of hypoxia on cells, including tools such as mechanism-based mathematical modelling and microfluidic setups.
Collapse
Affiliation(s)
- Kritika Saxena
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, 560012, India
| | - Mohit Kumar Jolly
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, 560012, India.
| |
Collapse
|
38
|
PIN1 transcript variant 2 acts as a long non-coding RNA that controls the HIF-1-driven hypoxic response. Sci Rep 2019; 9:10599. [PMID: 31332228 PMCID: PMC6646326 DOI: 10.1038/s41598-019-47071-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 07/10/2019] [Indexed: 02/08/2023] Open
Abstract
The transcription factor HIF-1 induces the expression of genes that are essential for cell survival and oxygen homeostasis in hypoxic conditions. The prolyl isomerase Pin1 plays a role in the regulation of HIF-1α. However, the mechanism by which Pin1 controls HIF-1α remains controversial. Surprisingly, we here show that a PIN1 transcript downregulates HIF-1α as a long non-coding RNA. Pin1-silencing siRNAs augmented the hypoxia-induced expression of HIF-1α, thereby upregulating the expression of HIF-1 target genes. However, the overexpression of Pin1 protein did not inhibit the hypoxic expression of HIF-1α. Pin1 restoration in Pin1-depleted cells also failed to reverse the induction of HIF-1α by Pin1 knockdown. Unexpectedly, HIF-1α was found to be induced by both siRNAs for PIN1 transcript variants 1/2 and that for PIN1 transcript variants 2/3, indicating that the PIN1 transcript variant 2 (PIN1-v2) is responsible for HIF-1α induction. Mechanistically, PIN1-v2, which is classified as a long non-coding RNA due to early termination of translation, was evaluated to inhibit the transcription of HIF1A gene. In conclusion, PIN1-v2 may function in balancing the HIF-1-driven gene expression under hypoxia.
Collapse
|
39
|
Morris-Blanco KC, Kim T, Bertogliat MJ, Mehta SL, Chokkalla AK, Vemuganti R. Inhibition of the Epigenetic Regulator REST Ameliorates Ischemic Brain Injury. Mol Neurobiol 2019; 56:2542-2550. [PMID: 30039336 PMCID: PMC6344325 DOI: 10.1007/s12035-018-1254-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 07/17/2018] [Indexed: 01/03/2023]
Abstract
Cerebral ischemia is known to activate the repressor element-1 (RE1)-silencing transcription factor (REST) which silences neural genes via epigenetic remodeling and promotes neurodegeneration. We presently determined if REST inhibition derepresses target genes involved in synaptic plasticity and promotes functional outcome after experimental stroke. Following transient focal ischemia induced by middle cerebral artery occlusion (MCAO) in adult rats, REST expression was upregulated significantly from 12 h to 1 day of reperfusion compared to sham control. At 1 day of reperfusion, REST protein levels were increased and observed in the nuclei of neurons in the peri-infarct cortex. REST knockdown by intracerebral REST siRNA injection significantly reduced the post-ischemic expression of REST and increased the expression of several REST target genes, compared to control siRNA group. REST inhibition also decreased post-ischemic markers of apoptosis, reduced cortical infarct volume, and improved post-ischemic functional recovery on days 5 and 7 of reperfusion compared to the control siRNA group. REST knockdown resulted in a global increase in synaptic plasticity gene expression at 1 day of reperfusion compared to the control siRNA group and significantly increased several synaptic plasticity genes containing RE-1 sequences in their regulatory regions. These results demonstrate that direct inhibition of the epigenetic remodeler REST prevents secondary brain damage in the cortex and improves functional outcome potentially via de-repression of plasticity-related genes after stroke.
Collapse
Affiliation(s)
- Kahlilia C Morris-Blanco
- Department of Neurological Surgery, University of Wisconsin-Madison, Mail Code CSC-8660, 600 Highland Ave., Madison, WI, 53792, USA
- William S. Middleton Veterans Administration Hospital, Madison, WI, USA
| | - TaeHee Kim
- Department of Neurological Surgery, University of Wisconsin-Madison, Mail Code CSC-8660, 600 Highland Ave., Madison, WI, 53792, USA
- William S. Middleton Veterans Administration Hospital, Madison, WI, USA
| | - Mario J Bertogliat
- Department of Neurological Surgery, University of Wisconsin-Madison, Mail Code CSC-8660, 600 Highland Ave., Madison, WI, 53792, USA
| | - Suresh L Mehta
- Department of Neurological Surgery, University of Wisconsin-Madison, Mail Code CSC-8660, 600 Highland Ave., Madison, WI, 53792, USA
- William S. Middleton Veterans Administration Hospital, Madison, WI, USA
| | - Anil K Chokkalla
- Department of Neurological Surgery, University of Wisconsin-Madison, Mail Code CSC-8660, 600 Highland Ave., Madison, WI, 53792, USA
- Cellular and Molecular Pathology Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Raghu Vemuganti
- Department of Neurological Surgery, University of Wisconsin-Madison, Mail Code CSC-8660, 600 Highland Ave., Madison, WI, 53792, USA.
- William S. Middleton Veterans Administration Hospital, Madison, WI, USA.
- Cellular and Molecular Pathology Program, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
40
|
Huang P, Chen CS, Yang YH, Chou MC, Chang YH, Lai CL, Chen HY, Liu CK. REST rs3796529 Genotype and Rate of Functional Deterioration in Alzheimer's Disease. Aging Dis 2019; 10:94-101. [PMID: 30705771 PMCID: PMC6345341 DOI: 10.14336/ad.2018.0116] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 01/16/2018] [Indexed: 01/22/2023] Open
Abstract
Recently, REST (RE1-silencing transcription factor) gene has been shown to be lost in Alzheimer’s disease (AD), and a missense minor REST allele rs3796529-T has been shown to reduce the rate of hippocampal volume loss. However, whether the REST rs3796529 genotype is associated with the rate of functional deterioration in AD is unknown. A total of 584 blood samples from Taiwanese patients with AD were collected from January 2002 to December 2013. The diagnosis of AD was based on the National Institute of Neurological and Communicative Disorders and Stroke and the Alzheimer’s Disease and Related Disorders Association criteria. The allele frequency of rs3796529-T was compared between the AD cohort and 993 individuals from the general population in Taiwan. Kaplan-Meier analysis, the log rank test and a multivariate Cox model were then used to evaluate the association between rs3796529-T and functional deterioration in the AD cohort. The allele frequency of rs3796529-T was significantly lower in the AD cohort compared to the general population cohort (36.82% vs. 40.73%, p=0.029). Kaplan-Meier analysis and the log rank test showed that the AD patients carrying the rs3796529 T/T genotype had a longer progression-free survival than those with the C/C genotype (p=0.012). In multivariate analysis, the rs3796529 T/T genotype (adjusted HR=0.593, 95% CI: 0.401-0.877, p=0.009) was an independent protective factor for functional deterioration. The rs3796529 T/T genotype was associated with slower functional deterioration in patients with AD. This finding may lead to a to better understanding of the molecular pathways involved, and prompt further development of novel biomarkers to monitor AD.
Collapse
Affiliation(s)
- Poyin Huang
- 1Department of Neurology, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,2Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,3Ph.D. Program in Translational Medicine, Kaohsiung Medical University and Academia Sinica, Taiwan.,4Department of Neurology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Cheng-Sheng Chen
- 5Department of Psychiatry, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yuan-Han Yang
- 6Department of Neurology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung, Taiwan
| | - Mei-Chuan Chou
- 6Department of Neurology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung, Taiwan
| | - Ya-Hsuan Chang
- 7Institute of Statistical Science, Academia Sinica, Taipei, Taiwan
| | - Chiou-Lian Lai
- 1Department of Neurology, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hsuan-Yu Chen
- 3Ph.D. Program in Translational Medicine, Kaohsiung Medical University and Academia Sinica, Taiwan.,7Institute of Statistical Science, Academia Sinica, Taipei, Taiwan.,8Graduate institute of medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ching-Kuan Liu
- 2Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
41
|
Martinez CA, Kerr B, Jin C, Cistulli PA, Cook KM. Obstructive Sleep Apnea Activates HIF-1 in a Hypoxia Dose-Dependent Manner in HCT116 Colorectal Carcinoma Cells. Int J Mol Sci 2019; 20:ijms20020445. [PMID: 30669593 PMCID: PMC6359625 DOI: 10.3390/ijms20020445] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 01/17/2019] [Accepted: 01/18/2019] [Indexed: 12/18/2022] Open
Abstract
Obstructive sleep apnea (OSA) affects a significant proportion of the population and is linked to increased rates of cancer development and a worse cancer outcome. OSA is characterized by nocturnal intermittent hypoxia and animal models of OSA-like intermittent hypoxia show increased tumor growth and metastasis. Advanced tumors typically have regions of chronic hypoxia, activating the transcription factor, HIF-1, which controls the expression of genes involved in cancer progression. Rapid intermittent hypoxia from OSA has been proposed to increase HIF-1 activity and this may occur in tumors. The effect of exposing a developing tumor to OSA-like intermittent hypoxia is largely unknown. We have built a cell-based model of physiological OSA tissue oxygenation in order to study the effects of intermittent hypoxia in HCT116 colorectal cancer cells. We found that HIF-1α increases following intermittent hypoxia and that the expression of HIF-target genes increases, including those involved in glycolysis, the hypoxic pathway and extracellular matrix remodeling. Expression of these genes acts as a 'hypoxic' signature which is associated with a worse prognosis. The total dose of hypoxia determined the magnitude of change in the hypoxic signature rather than the frequency or duration of hypoxia-reoxygenation cycles per se. Finally, transcription of HIF1A mRNA differs in response to chronic and intermittent hypoxia suggesting that HIF-1α may be regulated at the transcriptional level in intermittent hypoxia and not just by the post-translational oxygen-dependent degradation pathway seen in chronic hypoxia.
Collapse
Affiliation(s)
- Chloe-Anne Martinez
- Charles Perkins Centre, Faculty of Medicine and Health, Northern Clinical School, The University of Sydney, Sydney NSW 2006, Australia.
| | - Bernadette Kerr
- Charles Perkins Centre, Faculty of Medicine and Health, Northern Clinical School, The University of Sydney, Sydney NSW 2006, Australia.
| | - Charley Jin
- Charles Perkins Centre, Faculty of Medicine and Health, Northern Clinical School, The University of Sydney, Sydney NSW 2006, Australia.
| | - Peter A Cistulli
- Charles Perkins Centre, Faculty of Medicine and Health, Northern Clinical School, The University of Sydney, Sydney NSW 2006, Australia.
- Department of Respiratory and Sleep Medicine, Royal North Shore Hospital, Sydney 2065, Australia.
| | - Kristina M Cook
- Charles Perkins Centre, Faculty of Medicine and Health, Northern Clinical School, The University of Sydney, Sydney NSW 2006, Australia.
| |
Collapse
|
42
|
Callegari K, Maegawa S, Bravo-Alegria J, Gopalakrishnan V. Pharmacological inhibition of LSD1 activity blocks REST-dependent medulloblastoma cell migration. Cell Commun Signal 2018; 16:60. [PMID: 30227871 PMCID: PMC6145331 DOI: 10.1186/s12964-018-0275-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 09/13/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Medulloblastoma (MB) is the most common malignant brain tumor in children. Current problems in the clinic include metastasis, recurrence, and treatment-related sequelae that highlight the need for targeted therapies. Epigenetic perturbations are an established hallmark of human MB and expression of Lysine Specific Demethylase 1 (LSD1) is elevated in MBs compared to normal tissue, suggesting that LSD1 inhibitors may have efficacy against human MB tumors. METHODS Expression of LSD1 was examined across a publicly-available database and correlated with patient outcomes. Sonic Hedgehog (SHH) MB samples were clustered based on expression of LSD1 and LSD1-associated RE-1 silencing transcription factor (REST) target genes as well as genes involved in metastasis. Resulting clusters were examined for patient outcomes associated with LSD1 and REST expression. Human SHH MB cell lines were transduced with a REST-transgene to create isogenic cell pairs. In vitro viability and cell migration assays were used to examine the effect of LSD1 knockdown or inhibition on these parameters. RESULTS We demonstrate that subsets of SHH MB tumors have elevated LSD1 expression coincident with increased expression of its deubiquitylase, USP7, and REST. Patients with co-elevation of USP7, REST, and LSD1 have poorer outcomes compared to those with lower expression of these genes. In SHH MB cell lines, REST elevation increased cell growth and LSD1 protein levels. Surprisingly, while genetic loss of LSD1 reduced cell viability, pharmacological targeting of its activity using LSD1 inhibitors did not affect cell viability. However, a reduction in REST-dependent cell migration was seen in wound healing, suggesting that REST-LSD1 interaction regulates cell migration. Ingenuity pathway analyses validated these findings and identified Hypoxia Inducible Factor 1 alpha (HIF1A) as a potential target. In line with this, ectopic expression of HIF1A rescued the loss of migration seen following LSD1 inhibition. CONCLUSIONS A subset of SHH patients display increased levels of LSD1 and REST, which is associated with poor outcomes. REST elevation in MB in conjunction with elevated LSD1 promotes MB cell migration. LSD1 inhibition blocks REST-dependent cell migration of MB cells in a HIF1A-dependent manner.
Collapse
Affiliation(s)
- Keri Callegari
- Department of Pediatrics, University of Texas M.D. Anderson Cancer Center, Unit 853, 1515 Holcombe Blvd, Houston, TX, 77030, USA.,The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Austin, USA
| | - Shinji Maegawa
- Department of Pediatrics, University of Texas M.D. Anderson Cancer Center, Unit 853, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Javiera Bravo-Alegria
- Department of Pediatrics, University of Texas M.D. Anderson Cancer Center, Unit 853, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Vidya Gopalakrishnan
- Department of Pediatrics, University of Texas M.D. Anderson Cancer Center, Unit 853, 1515 Holcombe Blvd, Houston, TX, 77030, USA. .,Department of Molecular and Cellular Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, USA. .,Department of Center for Cancer Epigenetics, University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, USA. .,Department of Brain Tumor Center, University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, USA. .,The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Austin, USA.
| |
Collapse
|
43
|
Impact of the hypoxic phenotype on the uptake and efflux of nanoparticles by human breast cancer cells. Sci Rep 2018; 8:12318. [PMID: 30120388 PMCID: PMC6098061 DOI: 10.1038/s41598-018-30517-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 07/30/2018] [Indexed: 12/30/2022] Open
Abstract
Breast cancer cells adapt to the hypoxic tumoral environment by undergoing changes in metabolism, cell signalling, endo-lysosomal receptor uptake and recycling. The resulting hypoxic cell phenotype has the potential to undermine the therapeutic efficacy of nanomedicines designed for endocytic uptake and specific intracellular trafficking. The aim of this study was to examine the impact of hypoxia and simulated reperfusion on the in vitro uptake and release of nanomedicines by human breast cancer cells. Cells were exposed to a hypoxic preconditioning treatment in 1% oxygen for 6 and 24 hours to induce temporal changes in the hypoxic circuit (e.g. HIF-1α expression). The preconditioned cells were then dosed with nanoparticles for 45 or 180 minutes emulating nanomedicine access following tumor reperfusion. Hypoxic preconditioning significantly increased nanoparticle retention by up to 10% when compared to normoxic cultures, with the greatest relative difference between normoxic and hypoxic cultures occurring with a 45 minute dosing interval. Exocytosis studies indicated that the preconditioned cells had a significantly increased nanoparticle efflux (up to 9%) when compared to normoxic cells. Overall, we were able to show that hypoxic preconditioning regulates both the endocytosis and exocytosis of nanomedicines in human breast cancer cells.
Collapse
|
44
|
Okamura Y, Mekata T, Elshopakey GE, Itami T. Molecular characterization and gene expression analysis of hypoxia-inducible factor and its inhibitory factors in kuruma shrimp Marsupenaeus japonicus. FISH & SHELLFISH IMMUNOLOGY 2018; 79:168-174. [PMID: 29753689 DOI: 10.1016/j.fsi.2018.05.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Revised: 05/02/2018] [Accepted: 05/08/2018] [Indexed: 06/08/2023]
Abstract
In shrimp aquaculture, overcrowded farming causes fluctuations in dissolved oxygen concentrations. Low-oxygen conditions (hypoxia) affect shrimp growth. Hypoxia-inducible factor (HIF) is a transcriptional factor in the basic helix-loop-helix/PAS family and is activated in response to hypoxic stress. However, little is known about HIF and other inhibitors of the HIF pathway in crustaceans. In this study, we cloned MjHIF-1α, an inhibitory factor, MjFIH-1 (factor inhibiting HIF-1α), and MjVHL (Von Hippel-Lindau tumor suppressor) from kuruma shrimp (Marsupenaeus japonicus). MjVHL is the first crustacean VHL ortholog to be cloned. MjHIF-1α, MjFIH-1, and MjVHL exhibit significant sequence similarity and share key functional domains with previously described vertebrate and invertebrate genes. As a result of gene expression analysis in various tissues, MjHIF-1α and MjVHL were more highly expressed in the intestine than in any other organ tissues. In hypoxia experiments, HIF-induced expression levels of MjHIF-1α in the hypoxic group increased significantly for 24 h after initiating hypoxia stimulation and expression of MjVHL decreased significantly for 6 h after hypoxia stimulation (P < 0.05).
Collapse
Affiliation(s)
- Yo Okamura
- Department of Marine Biology and Environmental Sciences, Faculty of Agriculture, University of Miyazaki, Miyazaki, 889-2192, Japan.
| | - Tohru Mekata
- National Research Institute of Aquaculture, Japan Fisheries Research and Education Agency, Mie, 516-0193, Japan.
| | - Gehad Elsaid Elshopakey
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35516, Egypt.
| | - Toshiaki Itami
- Department of Marine Biology and Environmental Sciences, Faculty of Agriculture, University of Miyazaki, Miyazaki, 889-2192, Japan.
| |
Collapse
|
45
|
Lee GY, Shin SH, Shin HW, Chun YS, Park JW. NDRG3 lowers the metastatic potential in prostate cancer as a feedback controller of hypoxia-inducible factors. Exp Mol Med 2018; 50:1-13. [PMID: 29760417 PMCID: PMC5951909 DOI: 10.1038/s12276-018-0089-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 02/23/2018] [Accepted: 02/27/2018] [Indexed: 12/20/2022] Open
Abstract
Expression of hypoxia-inducible factors (HIFs) and N-myc downstream-regulated gene 3 (NDRG3) are oxygen-dependently regulated by prolyl hydroxylase domain (PHD) enzymes. Little is known about the role of NDRG3 in the cellular adaptation to hypoxia, whereas the roles of HIFs are well understood. In this study, we investigated how NDRG3 affects the hypoxic response in prostate cancer cells. Compared with HIF-1α, hypoxic induction of NDRG3 was observed at a later phase. NDRG3 reduced hypoxic expression of HIF-1α by inhibiting AKT-driven translation of HIF1A mRNA. In addition, NDRG3 functionally inhibited HIF-1 by dissociating the coactivator p300 from HIF-1α. Accordingly, NDRG3 may fine-tune the HIF-1 signaling pathway to cope with long-term hypoxia. Of the diverse effects of HIF-1α on cancer progression, hypoxia-induced cell migration was investigated. In transwell chambers, NDRG3 negatively regulated the migration and invasion of prostate cancer cells under hypoxia. An informatics analysis using Gene Expression Omnibus (GEO) revealed that NDRG3 downregulation is associated with prostate cancer metastasis and high expression of HIF-1 downstream genes. In cancer tissue arrays, NDRG3 expression was lower in prostate cancer tissues with a Gleason score of 8 or greater and was inversely correlated with HIF-1α expression. Therefore, NDRG3 may have an anti-metastatic function in prostate cancer under a hypoxic microenvironment. A protein activated under the low oxygen conditions typical of tumor surroundings has an anti-metastatic function in prostate cancer cells. Jong-Wan Park and colleagues from Seoul National University College of Medicine, South Korea, showed in prostate cancer cells that NDRG3, a protein activated under low oxygen conditions, works to fine-tune the signaling of another oxygen-sensitive protein to deal with the long-term stress imposed by cellular conditions. As a consequence, migration and invasion of prostate cancer cells are repressed. The researchers then looked at NDRG3 expression levels in tissue samples from men with prostate cancer and found that the more aggressive the disease the lower the NDRG3 activity. The results point to NDRG3 as a tumor suppressor in prostate cancer and suggest drug strategies designed to boost its function could aid in the treatment of this disease.
Collapse
Affiliation(s)
- Ga Young Lee
- Department of Biomedical Science, Seoul National University College of Medicine, Seoul, Korea.,Department of Pharmacology, Seoul National University College of Medicine, Seoul, Korea.,Cancer Research Institute and Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Seung-Hyun Shin
- Department of Biomedical Science, Seoul National University College of Medicine, Seoul, Korea.,Department of Pharmacology, Seoul National University College of Medicine, Seoul, Korea.,Cancer Research Institute and Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Hyun-Woo Shin
- Department of Biomedical Science, Seoul National University College of Medicine, Seoul, Korea.,Department of Pharmacology, Seoul National University College of Medicine, Seoul, Korea.,Cancer Research Institute and Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Yang-Sook Chun
- Department of Biomedical Science, Seoul National University College of Medicine, Seoul, Korea.,Cancer Research Institute and Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Jong-Wan Park
- Department of Biomedical Science, Seoul National University College of Medicine, Seoul, Korea. .,Department of Pharmacology, Seoul National University College of Medicine, Seoul, Korea. .,Cancer Research Institute and Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
46
|
Batie M, Del Peso L, Rocha S. Hypoxia and Chromatin: A Focus on Transcriptional Repression Mechanisms. Biomedicines 2018; 6:biomedicines6020047. [PMID: 29690561 PMCID: PMC6027312 DOI: 10.3390/biomedicines6020047] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 03/26/2018] [Accepted: 04/19/2018] [Indexed: 12/20/2022] Open
Abstract
Hypoxia or reduced oxygen availability has been studied extensively for its ability to activate specific genes. Hypoxia-induced gene expression is mediated by the HIF transcription factors, but not exclusively so. Despite the extensive knowledge about how hypoxia activates genes, much less is known about how hypoxia promotes gene repression. In this review, we discuss the potential mechanisms underlying hypoxia-induced transcriptional repression responses. We highlight HIF-dependent and independent mechanisms as well as the potential roles of dioxygenases with functions at the nucleosome and DNA level. Lastly, we discuss recent evidence regarding the involvement of transcriptional repressor complexes in hypoxia.
Collapse
Affiliation(s)
- Michael Batie
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Crown Street, Liverpool L697ZB, UK.
| | - Luis Del Peso
- Department of Biochemistry, Institute of Biomedical Research, Autonomous Madrid University, Arturo Duperier, 4. 28029 Madrid, Spain.
| | - Sonia Rocha
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Crown Street, Liverpool L697ZB, UK.
| |
Collapse
|
47
|
HOXA9 inhibits HIF-1α-mediated glycolysis through interacting with CRIP2 to repress cutaneous squamous cell carcinoma development. Nat Commun 2018; 9:1480. [PMID: 29662084 PMCID: PMC5902613 DOI: 10.1038/s41467-018-03914-5] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 03/22/2018] [Indexed: 01/18/2023] Open
Abstract
Glycolytic reprogramming is a typical feature of many cancers; however, key regulators of glucose metabolism reengineering are poorly understood, especially in cutaneous squamous cell carcinoma (cSCC). Here, Homeobox A9 (HOXA9), a direct target of onco-miR-365, is identified to be significantly downregulated in cSCC tumors and cell lines. HOXA9 acts as a tumor suppressor and inhibits glycolysis in cSCC in vitro and in vivo by negatively regulating HIF-1α and its downstream glycolytic regulators, HK2, GLUT1 and PDK1. Mechanistic studies show that HOXA9-CRIP2 interaction at glycolytic gene promoters impeds HIF-1α binding, repressing gene expression in trans. Our results reveal a miR-365-HOXA9-HIF-1α regulatory axis that contributes to the enhanced glycolysis in cSCC development and may represent an intervention target for cSCC therapy. Hypoxia-inducible transcription factor HIF-1α promotes glycolysis allowing cell survival under stress. Here the authors show, using both cell lines and animal models, that in cutaneous squamous cell carcinoma HOXA9 acts as a tumor suppressor and inhibits glycolysis by associating with CRIP2 to repress HIF-1α binding to target genes.
Collapse
|
48
|
Pickel C, Taylor CT, Scholz CC. Genetic Knockdown and Pharmacologic Inhibition of Hypoxia-Inducible Factor (HIF) Hydroxylases. Methods Mol Biol 2018; 1742:1-14. [PMID: 29330785 DOI: 10.1007/978-1-4939-7665-2_1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Reduced oxygen supply that does not satisfy tissue and cellular demand (hypoxia) regularly occurs both in health and disease. Hence, the capacity for cellular oxygen sensing is of vital importance for each cell to be able to alter its energy metabolism and promote adaptation to hypoxia. The hypoxia-inducible factor (HIF) prolyl hydroxylases 1-3 (PHD1-3) and the asparagine hydroxylase factor-inhibiting HIF (FIH) are the primary cellular oxygen sensors, which confer cellular oxygen-dependent sensitivity upon HIF as well as other hypoxia-sensitive pathways, such as nuclear factor κB (NF-κB). Studying these enzymes allows us to understand the oxygen-dependent regulation of cellular processes and has led to the development of several putative novel therapeutics, which are currently in clinical trials for the treatment of anemia associated with kidney disease. Pharmacologic inhibition and genetic knockdown are commonly established techniques in protein biochemistry and are used to investigate the activity and function of proteins. Here, we describe specific protocols for the knockdown and inhibition of the HIF prolyl hydroxylases 1-3 (PHD1-3) and the asparagine hydroxylase factor-inhibiting HIF (FIH) using RNA interference (RNAi) and hydroxylase inhibitors, respectively. These techniques are essential tools for the analysis of the function of the HIF hydroxylases, allowing the investigation and discovery of novel functions and substrates of these enzymes.
Collapse
Affiliation(s)
- Christina Pickel
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Cormac T Taylor
- Conway Institute of Biomolecular and Biomedical Research, School of Medicine, Charles Institute and Systems Biology Ireland, University College Dublin, Belfield, Dublin, Ireland
| | - Carsten C Scholz
- Institute of Physiology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
49
|
Cavadas MAS, Taylor CT, Cheong A. Acquisition of Temporal HIF Transcriptional Activity Using a Secreted Luciferase Assay. Methods Mol Biol 2018; 1742:37-44. [PMID: 29330788 DOI: 10.1007/978-1-4939-7665-2_4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Here we describe a simple method based on secreted luciferase driven by a hypoxia-inducible factor (HIF) response element (HRE) that allows the acquisition of dynamic and high-throughput data on HIF transcriptional activity during hypoxia and pharmacological activation of HIF. The sensitivity of the assay allows for the secreted luciferase to be consecutively sampled (as little as 1% of the total supernatant) over an extended time period, thus allowing the acquisition of time-resolved HIF transcriptional activity.
Collapse
Affiliation(s)
- Miguel A S Cavadas
- Systems Biology Ireland, University College Dublin, Dublin, Ireland
- Conway Institute of Biomolecular and Biomedical Research, School of Medicine, University College Dublin, Dublin, Ireland
- Instituto Gulbenkian de Ciencia, Oeiras, Portugal
| | - Cormac T Taylor
- Systems Biology Ireland, University College Dublin, Dublin, Ireland
- Conway Institute of Biomolecular and Biomedical Research, School of Medicine, University College Dublin, Dublin, Ireland
| | - Alex Cheong
- Systems Biology Ireland, University College Dublin, Dublin, Ireland.
- Conway Institute of Biomolecular and Biomedical Research, School of Medicine, University College Dublin, Dublin, Ireland.
- Life and Health Sciences, Aston University, Birmingham, UK.
| |
Collapse
|
50
|
Ziółkowska-Suchanek I, Mosor M, Podralska M, Iżykowska K, Gabryel P, Dyszkiewicz W, Słomski R, Nowak J. FAM13A as a Novel Hypoxia-Induced Gene in Non-Small Cell Lung Cancer. J Cancer 2017; 8:3933-3938. [PMID: 29187867 PMCID: PMC5705994 DOI: 10.7150/jca.20342] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 08/04/2017] [Indexed: 11/24/2022] Open
Abstract
Several genome-wide association studies (GWASs), have identified that FAM13A and IREB2 loci are associated with lung cancer, but the mechanisms by which these genes contribute to lung diseases susceptibility, especially in hypoxia context, are unknown. Hypoxia has been identified as a major negative factor for tumor progression in clinical observation. It has been suggested, that lower oxygen tension, may modulate the IREB2 and FAM13A activity. However, the role of these genes in hypoxia response has not been explained. To precise the role of these genes in hypoxia response, we analyzed the FAM13A and IREB2 expression, in lung cancer cells in vitro and lung cancer tissue fragments cultured ex vivo. Three cell lines: non-small cell lung cancer (A549, CORL-105), human lung fibroblasts (HL) and 37 lung cancer tissue fragments were analyzed. The expression of IREB2, FAM13A and HIF1α after sustained 72 hours of hypoxia versus normal oxygen concentration were analyzed by TaqMan® Gene Expression Assays and Western Blot. The expression of FAM13A was significantly up-regulated by hypoxia in two lung cancer cell lines (A549, CORL-105, P<0.001), both at the level of protein and mRNA, and in lung cancer tissue fragments (P=0.0004). The IREB2 was down-regulated after hypoxia in A549 cancer cells (P<0.001). Conclusions: We found that FAM13A overexpression in human lung cancer cell lines overlapped with hypoxia effect on lung cancer tissues. FAM13A is strongly induced by hypoxia and may be identified as a novel hypoxia-induced gene in non-small cell lung cancer.
Collapse
Affiliation(s)
| | - Maria Mosor
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszyńska 32, 60-479 Poznań, Poland
| | - Marta Podralska
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszyńska 32, 60-479 Poznań, Poland
| | - Katarzyna Iżykowska
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszyńska 32, 60-479 Poznań, Poland
| | - Piotr Gabryel
- Department of Thoracic Surgery, University of Medical Sciences, Szamarzewskiego 62, 60-569 Poznań
| | - Wojciech Dyszkiewicz
- Department of Thoracic Surgery, University of Medical Sciences, Szamarzewskiego 62, 60-569 Poznań
| | - Ryszard Słomski
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszyńska 32, 60-479 Poznań, Poland.,Department of Biochemistry and Biotechnology, Poznan University of Life Sciences , Dojazd 11, 60-632 Poznan, Poland
| | - Jerzy Nowak
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszyńska 32, 60-479 Poznań, Poland
| |
Collapse
|