1
|
Zhang S, Wang YS, Li Y, To KI, Zhang ET, Jin YH. Annexin A2 binds the 3'-UTR of H2AX mRNA and regulates histone-H2AX-derived hypoxia-inducible factor 1-alpha activation. Cell Signal 2025; 132:111781. [PMID: 40164417 DOI: 10.1016/j.cellsig.2025.111781] [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: 12/28/2024] [Revised: 03/21/2025] [Accepted: 03/28/2025] [Indexed: 04/02/2025]
Abstract
Annexin A2 (Anxa2), a multifunctional protein with RNA-binding capabilities, is frequently overexpressed in various tumors, and its expression is highly correlated with malignant progression. In this study, we demonstrate for the first time that Anxa2 was co-expressed with glycolytic genes, suggesting its potential role as a regulator of glycolysis. RNA-protein interaction assay revealed that Anxa2 interacted with 3'-UTR of H2AX mRNA and protected it from miRNA-mediated degradation. Up-regulated Histone-H2AX enhances the expression of glycolytic genes including GLUT1, HK2, PGK1, ENO1, PKM2, GAPDH and LDHA via stabilizing hypoxia-inducible factor 1-alpha (HIF1α), thereby accelerating lactic acid production and secretion. (20S) G-Rh2, a natural compound targeting Anxa2, significantly interfered the Anxa2-H2AX mRNA interaction, and inhibited subsequent glycolysis progression. We propose that Anxa2 acts as a novel regulator in glycolysis via enhancing H2AX expression, and (20S) G-Rh2 may exert its anti-cancer activity by targeting Anxa2-H2AX-HIF1α-glycolysis axis in human hepatoma HepG2 cells.
Collapse
Affiliation(s)
- Shiyin Zhang
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Yu-Shi Wang
- Department of Criminal Science and Technology, Jilin Police College, Changchun 130117, China
| | - Yang Li
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Kwang-Il To
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - En-Ting Zhang
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Ying-Hua Jin
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China.
| |
Collapse
|
2
|
Wang P, Rong Z, Li F, Ma S, Qiu H, Lian W, Li Z, Chen T, Zhong Q, Wang W, Sun G, Liu C, Ni L, Di X. Optimal injection sites for therapeutic angiogenesis: HGF-mediated regulation of HIF-1α via MAPK/PI3K pathways in hypoxic endothelial cells. Tissue Cell 2025; 95:102871. [PMID: 40132390 DOI: 10.1016/j.tice.2025.102871] [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: 11/11/2024] [Revised: 01/18/2025] [Accepted: 03/15/2025] [Indexed: 03/27/2025]
Abstract
Therapeutic angiogenesis offers a promising strategy for patients with critical limb-threatening ischemia (CLTI) who are unsuitable candidates for revascularization. However, the optimal administration sites for gene therapy agents, such as pCK-HGF-X7, remains undefined. Clinical trials commonly employ multiple intramuscular injections at sites of arterial occlusion; yet the necessity and efficacy of such extensive and repetitive protocols remains unclear. Targeted injections into ischemic tissues or their margins may improve therapeutic outcomes. Moreover, the molecular mechanisms by which hepatocyte growth factor (HGF)/c-Met signaling regulates hypoxia-inducible factor-1α (HIF-1α) expression under hypoxic conditions are not fully understood. This study aims to elucidate these molecular mechanisms in endothelial cells under hypoxic conditions and to identify the most effective injection sites for therapeutic angiogenesis agents. The effects of various HGF isoforms/complexes on human aortic endothelial cells (HAECs) were evaluated under normoxic and hypoxic conditions, focusing on proliferation, migration, and tube formation. Pathway inhibitors were used to explore the underlying mechanisms in hypoxic HAECs, and the findings were validated in a rat hindlimb ischemia model. Results demonstrated that HGF723 and HGF728 activated the mitogen-activated protein kinase (MAPK) and phosphatidylinositol 3-kinase (PI3K) pathways, regulating HIF expression and significantly enhanced endothelial cell proliferation, migration, and tube formation, particularly under hypoxia. Despite these cellular effects, HGF treatment did not significantly improve tissue perfusion or neovascularization in normal rat hindlimbs. However, in ischemic rat hindlimbs, it markedly promoted angiogenesis and improved tissue perfusion in the gastrocnemius muscle. These findings indicate that therapeutic angiogenesis agents should primarily target hypoxic tissues, extending to the interface between normoxic and hypoxic regions, to optimize treatment efficacy.
Collapse
Affiliation(s)
- Peng Wang
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, No. 1 Shuaifuyuan Hutong, Dongcheng District, Beijing 100730, China
| | - Zhihua Rong
- Department of Vascular Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Fengshi Li
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - ShanShan Ma
- Department of R&D Center, Beijing Northland Biotech co., LTD, No.5, Shangdi Kaituo Road,Haidian, Beijing 100085, China
| | - Hongjuan Qiu
- Department of R&D Center, Beijing Northland Biotech co., LTD, No.5, Shangdi Kaituo Road,Haidian, Beijing 100085, China
| | - Wenzhuo Lian
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, No. 1 Shuaifuyuan Hutong, Dongcheng District, Beijing 100730, China
| | - Zongshu Li
- Biomedical Engineering Facility of National Infrastructures for Translational Medicine, Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Tianqi Chen
- Biomedical Engineering Facility of National Infrastructures for Translational Medicine, Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Qing Zhong
- Biomedical Engineering Facility of National Infrastructures for Translational Medicine, Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Wenjing Wang
- Laboratory Animal Research Facility, National Infrastructures for Translational Medicine, Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Guoqiang Sun
- Department of Information Center, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, No. 1 Shuaifuyuan Hutong, Dongcheng District, Beijing 100730, China
| | - ChangWei Liu
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, No. 1 Shuaifuyuan Hutong, Dongcheng District, Beijing 100730, China
| | - Leng Ni
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, No. 1 Shuaifuyuan Hutong, Dongcheng District, Beijing 100730, China.
| | - Xiao Di
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, No. 1 Shuaifuyuan Hutong, Dongcheng District, Beijing 100730, China.
| |
Collapse
|
3
|
Liu L, Yu J, Liu Y, Xie L, Hu F, Liu H. Hypoxia-driven angiogenesis and metabolic reprogramming in vascular tumors. Front Cell Dev Biol 2025; 13:1572909. [PMID: 40443737 PMCID: PMC12119610 DOI: 10.3389/fcell.2025.1572909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 04/28/2025] [Indexed: 06/02/2025] Open
Abstract
Hypoxia is a hallmark of the tumor microenvironment (TME), and it plays a crucial role in the occurrence and progression in vascular tumors. Under hypoxic conditions, hypoxia-inducible factor 1-alpha (HIF-1α) is stabilized, inducing changes in the expression of various target genes involved in angiogenesis, metabolism, and cell survival. This includes the upregulation of pro-angiogenic factors like VEGF, which promotes the formation of dysfunctional blood vessels, contributing to the worsening of the hypoxic microenvironment. At the same time, hypoxia induces a metabolic shift toward glycolysis, even in the presence of oxygen, supporting tumor cell survival and proliferation by providing necessary energy and biosynthetic precursors. This review discusses the molecular mechanisms by which hypoxia regulates angiogenesis and metabolic reprogramming in vascular tumors, highlighting the intricate link between these processes, and explores potential therapeutic strategies to target these pathways in order to develop effective treatment strategies for patients.
Collapse
Affiliation(s)
- Lu Liu
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defect and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
- NHC Key Laboratory of Chronobiology (Sichuan University), Chengdu, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children’s Health, West China Second University Hospital, Sichuan University, Chengdu, China
- Sichuan Birth Defects Clinical Research Center, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Jiayun Yu
- Department of Radiotherapy, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, Sichuan University, Chengdu, China
| | - Yang Liu
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defect and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
- NHC Key Laboratory of Chronobiology (Sichuan University), Chengdu, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children’s Health, West China Second University Hospital, Sichuan University, Chengdu, China
- Sichuan Birth Defects Clinical Research Center, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Liang Xie
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defect and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
- NHC Key Laboratory of Chronobiology (Sichuan University), Chengdu, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children’s Health, West China Second University Hospital, Sichuan University, Chengdu, China
- Sichuan Birth Defects Clinical Research Center, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Fan Hu
- Key Laboratory of Birth Defect and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
- Department of Pediatric Cardiology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hanmin Liu
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defect and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
- NHC Key Laboratory of Chronobiology (Sichuan University), Chengdu, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children’s Health, West China Second University Hospital, Sichuan University, Chengdu, China
- Sichuan Birth Defects Clinical Research Center, West China Second University Hospital, Sichuan University, Chengdu, China
- Department of Pediatric Pulmonology and Immunology, WCSUH-Tianfu·Sichuan Provincial Children’s Hospital, Sichuan University, Meishan, China
| |
Collapse
|
4
|
Bryant P, McCann P, Namjou K, Sikavitsas V, Harrison R. Mid-IR laser measurement of acetaldehyde in the headspace gas of cell culture media samples from growth of breast cancer cells in hydrogel scaffolds. Anal Bioanal Chem 2025:10.1007/s00216-025-05899-9. [PMID: 40355762 DOI: 10.1007/s00216-025-05899-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 04/17/2025] [Accepted: 04/22/2025] [Indexed: 05/15/2025]
Abstract
A mid-IR laser absorption spectrometer configured with an interband cascade laser (ICL) was used to measure acetaldehyde concentrations in the headspace gas of cell culture media samples obtained from the culturing of breast cancer cells in a flow perfusion bioreactor. Measurements were performed by bubbling lab air through a media sample and passing the exhaust gas through a long optical path gas cell where rotational-vibrational modes of acetaldehyde were excited by the ICL. Acetaldehyde concentrations were determined from peak-to-peak voltages for the two strongest acetaldehyde absorption features within a spectral region spanning 1770.20 cm-1 to 1770.35 cm-1. Three different media samples obtained from cells cultured with the same nominal conditions had headspace acetaldehyde concentrations of 412 ppb, 513 ppb, and 390 ppb, while two media samples with imposed hypoxia or cobalt chloride stabilization of hypoxia inducible factor 1-alpha (HIF-1α) had higher acetaldehyde concentrations, 815 ppb and 536 ppb, respectively. These results establish experimental proof-of-concept for the ability of mid-IR laser absorption spectroscopy to measure acetaldehyde in cell culture media headspace, allowing observation of HIF-1α driven shifts in cancer cell metabolism.
Collapse
Affiliation(s)
- Parker Bryant
- School of Chemical, Biological and Materials Engineering, Gallogly College of Engineering, The University of Oklahoma, Norman, USA
| | - Patrick McCann
- School of Electrical and Computer Engineering, Gallogly College of Engineering, The University of Oklahoma, 110 West Boyd Street, Norman, OK, 73019-1102, USA.
| | - Khosrow Namjou
- School of Electrical and Computer Engineering, Gallogly College of Engineering, The University of Oklahoma, 110 West Boyd Street, Norman, OK, 73019-1102, USA
| | - Vassilios Sikavitsas
- School of Chemical, Biological and Materials Engineering, Gallogly College of Engineering, The University of Oklahoma, Norman, USA
| | - Roger Harrison
- School of Chemical, Biological and Materials Engineering, Gallogly College of Engineering, The University of Oklahoma, Norman, USA
| |
Collapse
|
5
|
Yang L, Chen Y, Wu Y. The hypoxia signaling pathway in the development of acute myeloid leukemia. Biomed Pharmacother 2025; 186:117999. [PMID: 40188762 DOI: 10.1016/j.biopha.2025.117999] [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: 01/13/2025] [Revised: 03/06/2025] [Accepted: 03/17/2025] [Indexed: 04/25/2025] Open
Abstract
Acute myeloid leukemia (AML) is the most common acute leukemia in adults. Although advances in targeted agents have greatly improved the prognosis of patients with AML in recent years, those who fail to achieve remission or relapse after remission are still in urgent need of novel therapeutic strategies. The hypoxia signaling pathway is involved in various biological processes, and hypoxia-inducible factor alpha (HIF-α) is considered a potential therapeutic target in AML. The bone marrow microenvironment is known to be in a state of chronic hypoxia, which is important for hematopoietic stem cells to maintain quiescence, and provides leukemic stem cells with a refuge from immune defenses and chemotherapeutic agents. Therefore, this review aims to explore the role of the HIF-α signaling pathway in the development of AML.
Collapse
Affiliation(s)
- Liqing Yang
- Fujian Provincial Key Laboratory on Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, Fujian 350001, China; Department of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China
| | - Yuanzhong Chen
- Fujian Provincial Key Laboratory on Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, Fujian 350001, China
| | - Yong Wu
- Fujian Provincial Key Laboratory on Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, Fujian 350001, China; Department of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China.
| |
Collapse
|
6
|
Lynch-Miller M, Lockow S, Dümmer K, Henneck T, Olmer R, Jaboreck MC, Mergani AO, Wandrey M, Branitzki-Heinemann K, Brogden G, Naim HY, Martin U, Schulz C, Talbot SR, Meurer M, Baumgärtner W, von Köckritz-Blickwede M. Characterization of 3D human pulmonary epithelial model morphology and oxygen status under normoxia and hypoxia. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119980. [PMID: 40315920 DOI: 10.1016/j.bbamcr.2025.119980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 04/05/2025] [Accepted: 04/28/2025] [Indexed: 05/04/2025]
Abstract
Infection generates localized hypoxia in affected tissue, inducing cellular survival responses and modulating inflammatory processes. Consideration of oxygen status as a parameter in in vitro infection research is therefore vital to the generation of physiologically relevant data within the 3R context. In this study, we characterize the culture morphology and oxygenation of liquid-liquid interface (LLI) permanent bronchial epithelial (Calu-3), classical air-liquid interface (cALI) Calu-3, and cALI human primary bronchial epithelial cell (hBEC) models under the normoxic conditions within standard incubators, commonly employed in in vitro work. We compare the normoxic state of these models to their hypoxic state to assess changes in the airway epithelial environment in response to oxygen deprivation, and the extent to which select hypoxia responses can be observed at the molecular level. Additional juxtapositions are drawn between Calu-3 LLI and cALI models and Calu-3 conventional monolayer (CM) and inverted air-liquid interface (iALI) models, due to their relevance for basic and specialized research, respectively. Epithelial complexity was observed to vary amongst the filter-based models, and all models were found to exhibit characteristic extracellular oxygen depletion patterns under normoxia. Importantly, the extracellular oxygen contents of Calu-3 LLI, cALI, and CM models significantly decreased during normoxic incubation. Specific hypoxia responses through stabilization of HIF-1α, HIF-2α, and/or HIF-3α and alteration of ACE2 protein levels differed in response to both culture format and cell type. Therefore, while all models examined provide valuable opportunities for in vitro exploration, variation in their morphological, physiological, and molecular characteristics necessitates careful consideration during experimental design.
Collapse
Affiliation(s)
- Maura Lynch-Miller
- Institute of Biochemistry, University of Veterinary Medicine, Foundation, Hannover, Germany; Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Foundation, Hannover, Germany
| | - Sandra Lockow
- Department of Pathology, University of Veterinary Medicine, Foundation, Hannover, Germany
| | - Katrin Dümmer
- Institute of Biochemistry, University of Veterinary Medicine, Foundation, Hannover, Germany; Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Foundation, Hannover, Germany
| | - Timo Henneck
- Institute of Biochemistry, University of Veterinary Medicine, Foundation, Hannover, Germany; Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Foundation, Hannover, Germany
| | - Ruth Olmer
- Leibniz Research Laboratories for Biotechnology and Artificial Organs, Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG), REBIRTH-Research Center for Translational Regenerative Medicine, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
| | - Mark-Christian Jaboreck
- Leibniz Research Laboratories for Biotechnology and Artificial Organs, Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG), REBIRTH-Research Center for Translational Regenerative Medicine, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
| | - AhmedElmontaser O Mergani
- Institute of Biochemistry, University of Veterinary Medicine, Foundation, Hannover, Germany; Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Foundation, Hannover, Germany
| | - Madita Wandrey
- Institute of Biochemistry, University of Veterinary Medicine, Foundation, Hannover, Germany; Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Foundation, Hannover, Germany
| | - Katja Branitzki-Heinemann
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Foundation, Hannover, Germany
| | - Graham Brogden
- Institute of Experimental Virology, TWINCORE, Center for Experimental and Clinical Infection Research Hannover, Hannover, Germany
| | - Hassan Y Naim
- Institute of Biochemistry, University of Veterinary Medicine, Foundation, Hannover, Germany
| | - Ulrich Martin
- Leibniz Research Laboratories for Biotechnology and Artificial Organs, Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG), REBIRTH-Research Center for Translational Regenerative Medicine, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
| | - Claudia Schulz
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Foundation, Hannover, Germany
| | - Steven R Talbot
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Marita Meurer
- Institute of Biochemistry, University of Veterinary Medicine, Foundation, Hannover, Germany; Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Foundation, Hannover, Germany
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine, Foundation, Hannover, Germany
| | - Maren von Köckritz-Blickwede
- Institute of Biochemistry, University of Veterinary Medicine, Foundation, Hannover, Germany; Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Foundation, Hannover, Germany.
| |
Collapse
|
7
|
Chiu DKC, Zhang X, Cheng BYL, Liu Q, Hayashi K, Yu B, Lee R, Zhang C, An X, Rajadas J, Reticker-Flynn NE, Rankin EB, Engleman EG. Tumor-derived erythropoietin acts as an immunosuppressive switch in cancer immunity. Science 2025; 388:eadr3026. [PMID: 40273234 PMCID: PMC12110762 DOI: 10.1126/science.adr3026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 12/20/2024] [Accepted: 03/05/2025] [Indexed: 04/26/2025]
Abstract
Successful cancer immunotherapy requires a patient to mount an effective immune response against tumors; however, many cancers evade the body's immune system. To investigate the basis for treatment failure, we examined spontaneous mouse models of hepatocellular carcinoma (HCC) with either an inflamed T cell-rich or a noninflamed T cell-deprived tumor microenvironment (TME). Our studies reveal that erythropoietin (EPO) secreted by tumor cells determines tumor immunotype. Tumor-derived EPO autonomously generates a noninflamed TME by interacting with its cognate receptor EPOR on tumor-associated macrophages (TAMs). EPO signaling prompts TAMs to become immunoregulatory through NRF2-mediated heme depletion. Removing either tumor-derived EPO or EPOR on TAMs leads to an inflamed TME and tumor regression independent of genotype, owing to augmented antitumor T cell immunity. Thus, the EPO/EPOR axis functions as an immunosuppressive switch for antitumor immunity.
Collapse
Affiliation(s)
| | - Xiangyue Zhang
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | | | - Qiang Liu
- Biomaterials and Advanced Drug Delivery Laboratory, Stanford University School of Medicine, Stanford, CA, USA
| | - Kazukuni Hayashi
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Bo Yu
- ImmunEdge Inc. Mountain View, California 94043, USA
| | - Ryan Lee
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Catherine Zhang
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Xiuli An
- Laboratory of Membrane Biology, New York Blood Center, New York, NY, 10065, USA
| | - Jayakumar Rajadas
- Biomaterials and Advanced Drug Delivery Laboratory, Stanford University School of Medicine, Stanford, CA, USA
| | - Nathan E Reticker-Flynn
- Department of Otolaryngology - Head and Neck Surgery, Stanford University, Stanford, CA 94305, USA
| | - Erinn B Rankin
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
- Stanford Cancer Institute, Stanford University, Palo Alto, CA 94305, USA
| | - Edgar G Engleman
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
- Stanford Cancer Institute, Stanford University, Palo Alto, CA 94305, USA
- Lead contact
| |
Collapse
|
8
|
Ogura Y, Sun X, Zhang Z, Kawata K, Wu J, Matsubara R, Ozeki AN, Taniue K, Onoguchi-Mizutani R, Adachi S, Nakayama K, Goda N, Akimitsu N. Fragile X messenger ribonucleoprotein 1 (FMRP) regulates glycolytic gene expression under chronic hypoxia in HCT116 cells. Sci Rep 2025; 15:13273. [PMID: 40246883 PMCID: PMC12006372 DOI: 10.1038/s41598-025-91828-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 02/24/2025] [Indexed: 04/19/2025] Open
Abstract
Oxygen shortage, known as hypoxia, occurs commonly in both physiological and pathological conditions. Transcriptional regulation by hypoxia-inducible factors is a dominant regulatory mechanism controlling hypoxia-responsive genes during acute hypoxia; however, recent studies suggest that post-transcriptional regulation, including RNA degradation, also involves hypoxia-induced gene expression during the chronic hypoxia. In this study, we developed a method to quantify the contributions of RNA synthesis and degradation to differential gene expression, and identified 102 genes mainly regulated via RNA degradation under chronic hypoxia in HCT116 cells. Bioinformatics analysis showed that the genes mainly regulated by RNA degradation were involved in glycolysis. We examined changes in the RNA-binding ability of RNA-binding proteins by RNA interactome capture and statistical analysis using public databases. We identified fragile X messenger ribonucleoprotein 1 (FMRP) as an RNA-binding protein involved in the chronic hypoxia-induced increase in mRNAs encoding rate-limiting enzymes. This study emphasizes the importance of post-transcriptional gene regulation under chronic hypoxia in HCT116 cells.
Collapse
Affiliation(s)
- Yoko Ogura
- Department of Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Xiaoning Sun
- Advanced Interdisciplinary Studies, Engineering Department, The University of Tokyo, Tokyo, Japan
| | - Zaijun Zhang
- Department of Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Kentaro Kawata
- Isotope Science Center, The University of Tokyo, Tokyo, 113-0032, Japan.
| | - Jinyu Wu
- Department of Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Ryuma Matsubara
- Isotope Science Center, The University of Tokyo, Tokyo, 113-0032, Japan
| | | | - Kenzui Taniue
- Isotope Science Center, The University of Tokyo, Tokyo, 113-0032, Japan
| | | | - Shungo Adachi
- Department of Proteomics, National Cancer Center Research Institute, Tokyo, 104-0045, Japan
| | - Koh Nakayama
- Department of Pharmacology, School of Medicine, Asahikawa Medical University, Hokkaido, 078-8510, Japan
| | - Nobuhito Goda
- Department of Life Science and Medical Bioscience, School of Advanced Science and Engineering, Waseda University, Tokyo, 162-8480, Japan
| | - Nobuyoshi Akimitsu
- Isotope Science Center, The University of Tokyo, Tokyo, 113-0032, Japan.
| |
Collapse
|
9
|
Du M, Pan X, Peng Y, Yang J, Pan S, Cheng R, Yang S, Yang Z, Pan J, Liu P, Zhang S, Zhang X. A Tandem Cytometry Platform for Single-Cell Analysis of Protein and Metabolites. Anal Chem 2025; 97:6962-6966. [PMID: 40130787 DOI: 10.1021/acs.analchem.5c00606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
Multiomics analysis at the single-cell level is essential for both fundamental research and clinical applications, with proteomics and metabolomics being particularly crucial for providing insights into cellular states and functions. The state of the art flow cytometry has shown great potential in identifying cellular proteins, while emerging metabolite mass spectrometry cytometry techniques address metabolite detection. Herein, we propose a tandem platform that integrates fluorescence flow cytometry with electrospray ionization mass spectrometry for one-step single-cell analysis of protein and metabolites. An algorithm was established to correlate multidimensional information in individual cells, with additional data processing modules designed to ensure accuracy and facilitate further analysis. The tandem cytometry platform demonstrated efficacy in profiling breast cancer cells, particularly under hypoxic conditions, revealing metabolic shifts with decreased glutathione and increased l-glutamine levels, indicative of hypoxia-inducible factor activity. This platform introduces a powerful analytical capability that promises to elevate the precision of cell-based diagnostics and therapeutic strategies.
Collapse
Affiliation(s)
- Murong Du
- Department of Chemistry, Tsinghua University, Beijing 100084, P. R. China
| | - Xingyu Pan
- Department of Chemistry, Tsinghua University, Beijing 100084, P. R. China
| | - Yamei Peng
- Institute of Microanalytical Systems, Department of Chemistry, Zhejiang University, Hangzhou 310058, P. R. China
| | - Jinlei Yang
- Department of Chemistry, Tsinghua University, Beijing 100084, P. R. China
| | - Siyuan Pan
- Department of Chemistry, Tsinghua University, Beijing 100084, P. R. China
| | - Runsong Cheng
- Department of Chemistry, Tsinghua University, Beijing 100084, P. R. China
| | - Shu Yang
- State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, P. R. China
| | - Zhenli Yang
- Cell Resource Center, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing 100730, P. R. China
| | - Jianzhang Pan
- Institute of Microanalytical Systems, Department of Chemistry, Zhejiang University, Hangzhou 310058, P. R. China
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou 310058, P. R. China
| | - Peng Liu
- State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, P. R. China
| | - Sichun Zhang
- Department of Chemistry, Tsinghua University, Beijing 100084, P. R. China
| | - Xinrong Zhang
- Department of Chemistry, Tsinghua University, Beijing 100084, P. R. China
| |
Collapse
|
10
|
Zhao S, Li H, Luo W, Hu Z, Wang Y, Liu T, Zhang Y, Dai R. WHOLE TRANSCRIPTION ANALYSIS IDENTIFIED THE REGULATION OF HYPOXIA-INDUCIBLE FACTORS IN MONOCYTES WITH IMMUNE SUPPRESSION: IMPLICATIONS FOR CLINICAL OUTCOMES. Shock 2025; 63:541-551. [PMID: 39405478 PMCID: PMC11939089 DOI: 10.1097/shk.0000000000002479] [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: 05/30/2024] [Revised: 07/08/2024] [Accepted: 09/03/2024] [Indexed: 03/21/2025]
Abstract
ABSTRACT Aims: Sepsis progression is marked by a complex immune response, where the involvement of hypoxia-inducible factors (HIFs) plays an uncertain role. The study aims to elucidate the involvement of HIF-1α in monocyte function during sepsis and its potential as a prognostic indicator. Methods and Results: Transcriptomic data from healthy individuals and septic patients in datasets GSE54514, GSE167363, and GSE46955 were analyzed. Additionally, human monocytes were employed to elucidate how HIF regulates immune responses in the context of sepsis. Septic nonsurvivors exhibited sustained upregulation of HIF-1α expression alongside compromised inflammatory response and antigen presentation, with downregulation of NF-κB and HLADRB1 genes associated with poor sepsis prognosis. Conversely, septic survivors displayed an increased proportion of classical monocytes and enhanced inflammation and expression of antigen presentation-related genes. During the recovery phase of sepsis, monocytes continued to demonstrate elevated HIF-1α expression. In cultured THP1 cells and septic CD14 + monocytes, HIF hindered inflammatory responses and antigen presentation, while also suppressing the proportion of classical monocytes after LPS stimulation. Mechanistically, HIF significantly attenuated LPS-induced immune responses in monocytes by inhibiting the phosphorylation of IKK. Conclusions: HIF in monocytes acts as a suppressor of immune-inflammatory responses and antigen presentation, and may serve as a negative molecular marker for sepsis development.
Collapse
Affiliation(s)
- Shuai Zhao
- Department of Anesthesiology, the Second Xiangya Hospital, Central South University, Changsha, China
- Anesthesiology Research Institute of Central South University, Changsha, China
| | - Hui Li
- Department of Anesthesiology, the Second Xiangya Hospital, Central South University, Changsha, China
- Anesthesiology Research Institute of Central South University, Changsha, China
| | - Wei Luo
- Department of Anesthesiology, the Second Xiangya Hospital, Central South University, Changsha, China
- Anesthesiology Research Institute of Central South University, Changsha, China
| | - Zhaolan Hu
- Department of Anesthesiology, the Second Xiangya Hospital, Central South University, Changsha, China
- Anesthesiology Research Institute of Central South University, Changsha, China
| | - Yulu Wang
- Department of Integrated Oncology, Center for Integrated Oncology, University Hospital of Bonn, Bonn, Germany
| | - Tao Liu
- Department of Anesthesiology, the Second Xiangya Hospital, Central South University, Changsha, China
- Anesthesiology Research Institute of Central South University, Changsha, China
| | - Yanling Zhang
- Department of Anesthesiology, the Second Xiangya Hospital, Central South University, Changsha, China
- Anesthesiology Research Institute of Central South University, Changsha, China
| | - RuPing Dai
- Department of Anesthesiology, the Second Xiangya Hospital, Central South University, Changsha, China
- Anesthesiology Research Institute of Central South University, Changsha, China
| |
Collapse
|
11
|
Gong L, Zhang H, Liu Y, Wang X, Xia R. Interactions Between Non-Coding RNAs and HIF-1alpha in the Context of Colorectal Cancer. Biomolecules 2025; 15:510. [PMID: 40305214 PMCID: PMC12024830 DOI: 10.3390/biom15040510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/17/2025] [Accepted: 03/30/2025] [Indexed: 05/02/2025] Open
Abstract
Hypoxia-inducible factor-1α (HIF-1α), a master regulator of cellular adaptation to hypoxia, drives colorectal cancer (CRC) progression by fueling angiogenesis, metastasis, and therapy resistance. Emerging evidence delineates intricate crosstalk between non-coding RNAs (ncRNAs)-including microRNAs, long non-coding RNAs, and circular RNAs-and HIF-1α, forming bidirectional regulatory networks that orchestrate CRC pathogenesis. By interacting with HIF-1α, these non-coding RNAs contribute to the orchestration of the aggressive hypoxic tumor microenvironment. Recent studies have evaluated the clinical potential of lncRNAs and miRNAs in the realms of non-invasive liquid biopsies and RNA-targeted therapies. This review offers a comprehensive synthesis of recent investigations into the mechanisms by which lncRNAs and miRNAs interact with HIF-1α to modulate CRC progression. Additionally, we further explore the clinical implications of ncRNA/HIF-1α crosstalk, emphasizing their potential as diagnostic biomarkers and therapeutic targets, while also spotlighting intriguing and promising areas of ncRNA research. Methods: In this study, our search strategy employed in databases such as PubMed, Web of Science, and EMBASE is as follows: we will specify search terms, including combinations of "non-coding RNA", "HIF-1α", and "colorectal cancer", along with a date range for the literature search (for example, from 2000 to 2025) to capture the most relevant and up-to-date research.
Collapse
Affiliation(s)
| | | | | | - Xianwang Wang
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China; (L.G.); (H.Z.); (Y.L.)
| | - Ruohan Xia
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China; (L.G.); (H.Z.); (Y.L.)
| |
Collapse
|
12
|
Cao Z, Li J, Hu W, Xu J, Zhao F, Wang Y, Qin S, Liu M, Wang P, Duan J, Zhou W, Ding Z, Tang S, Ma X, Wang L. Near-Infrared Imaging Agent ABSi-148 Alleviates CA IX-Mediated Hypoxic Fibrosis in Inflammation-Cancer Transition. Adv Healthc Mater 2025; 14:e2404935. [PMID: 40099420 DOI: 10.1002/adhm.202404935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 03/07/2025] [Indexed: 03/19/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains a formidable challenge due to its late diagnosis and intrinsic treatment resistance, exacerbates by its development from chronic inflammation to cancer transition (ICT). Here, this investigation aims to develop and evaluate ABSi-148, a novel near-infrared (NIR) agent targeting hypoxic carbonic anhydrase IX (CA IX), for its potential applications in ICT imaging and even PDAC treatment. ABSi-148 is synthesized from 4-(2-Aminoethyl) benzene sulfonamide (ABS), a sulfonamide derivative, conjugating with MHI-148 dye with merits of exceptional NIR-emitting traits, high biocompatibility, and deep tissue penetration imaging capability. It selectively accumulates in CoCl2-induced pancreatic stellate cells and pancreatic cancer cells via binding with transmembrane CA IX in vitro. Meanwhile, ABSi-148 effectively visualizes the early pancreatic lesion, and its long-term administration inhibits the progression of hypoxia-related fibrosis involved in pancreatic intraepithelial neoplasias (PanINs), and even PDAC progression in vivo. Besides, ABSi-148 monitors treatment efficacy and localizes hypoxic tumor regions, enhancing survival in tamoxifen combined with caerulein-induced KPC mice. Overall, ABSi-148 emerges as a theranostic NIR agent for precise diagnosis and targeted therapy in ICT of PDAC, promising to alleviate tumor progression and enhancing outcomes.
Collapse
Affiliation(s)
- Zhi Cao
- Department of Gastroenterology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, China, Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, 510631, China
- Department of Gastroenterology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Jingmin Li
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, China, Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, 510631, China
- Internal medicine department, Guangzhou women and children's Medical Center, Guangzhou, 510623, China
| | - Weibin Hu
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, China, Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, 510631, China
- Department of Nuclear Medicine, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 518037, China
| | - Jian Xu
- Stroke center, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510799, China
| | - Fengyun Zhao
- Cancer Research Institute of Zhongshan City, Zhongshan City People's Hospital, Zhongshan, 528403, China
| | - Yishu Wang
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, China, Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, 510631, China
| | - Shuanglin Qin
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Research Center for Precision Medication of Chinese Medicine, FuRong Laboratory, Hunan University of Chinese Medicine, Changsha, 410218, China
| | - Ming Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Ping Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510230, China
| | - Jingwei Duan
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, China, Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, 510631, China
| | - Wensheng Zhou
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Zhaowei Ding
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510230, China
| | - Shaohui Tang
- Department of Gastroenterology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Xiaodong Ma
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, China, Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, 510631, China
| | - Lisheng Wang
- Department of Gastroenterology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China
| |
Collapse
|
13
|
Wu M, Koester DC, Walkinshaw G, Ng D, Zhou X, Ho A, Tsao J, Barnes M, Brenner MC, Spong S, Nelson G, Gervasi DC, Vaisberg E, Sternlicht M, Sidhu P, Lin J, Ibrahim M, Thompson MD, Chou J, Pangilinan G, Makwana O, Wei Z, Signore PE, Del Balzo U, Hoch U, Ramurthy S. Discovery of Novel, Potent, Orally Bioavailable and Efficacious, Hypoxia-Inducible Factor Prolyl Hydroxylase Inhibitors for Hematopoietic Stem Cell Mobilization. J Med Chem 2025; 68:6386-6406. [PMID: 40047531 DOI: 10.1021/acs.jmedchem.4c02889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2025]
Abstract
Hematopoietic stem cell (HSC) mobilization is often difficult to achieve in patients suffering from multiple myeloma and non-Hodgkin's lymphoma. Granulocyte-colony stimulating factor (G-CSF) therapy alone has often not led to the desired outcomes. Herein, we describe the discovery of 7-cyclohexyl-4-hydroxy-8-oxo-N-(pyridazin-4-ylmethyl)-7,8-dihydro-2,7-naphthyridine-3-carboxamide 13, a hypoxia-inducible factor prolyl hydroxylase (HIF-PH) inhibitor, which was discovered by focusing on drug-like properties. Building on a previous discovery that HIF-PH inhibitors can enhance HSC mobilization in combination with G-CSF, we optimized 13 to exhibit high PHD2 potency, improved solubility, and an optimized PK profile. 13 was effective at enhancing G-CSF-induced HSC mobilization in mice at a dose of 2 mg/kg.
Collapse
Affiliation(s)
- Min Wu
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - Dennis C Koester
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - Gail Walkinshaw
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - Danny Ng
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - Xiaoti Zhou
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - Angel Ho
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - Jenny Tsao
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - Michael Barnes
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - Mitchell C Brenner
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - Suzanne Spong
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - Grace Nelson
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - David C Gervasi
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - Elena Vaisberg
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - Mark Sternlicht
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - Parmjeet Sidhu
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - Jack Lin
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - Mohamed Ibrahim
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - Michael D Thompson
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - James Chou
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - Gerardo Pangilinan
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - Om Makwana
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - Zhihua Wei
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - Pierre E Signore
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - Ughetta Del Balzo
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - Ute Hoch
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| | - Savithri Ramurthy
- FibroGen Inc., 409 Illinois Street, San Francisco, California 94154, United States
| |
Collapse
|
14
|
Wang C, Fan X, Shi Y, Tang F. Radiation-Induced Brain Injury with Special Reference to Astrocytes as a Therapeutic Target. J Integr Neurosci 2025; 24:25907. [PMID: 40152565 DOI: 10.31083/jin25907] [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: 07/30/2024] [Revised: 10/22/2024] [Accepted: 11/06/2024] [Indexed: 03/29/2025] Open
Abstract
Radiotherapy is one of the primary modalities for oncologic treatment and has been utilized at least once in over half of newly diagnosed cancer patients. Cranial radiotherapy has significantly enhanced the long-term survival rates of patients with brain tumors. However, radiation-induced brain injury, particularly hippocampal neuronal damage along with impairment of neurogenesis, inflammation, and gliosis, adversely affects the quality of life for these patients. Astrocytes, a type of glial cell that are abundant in the brain, play essential roles in maintaining brain homeostasis and function. Despite their importance, the pathophysiological changes in astrocytes induced by radiation have not been thoroughly investigated, and no systematic or comprehensive review addressing the effects of radiation on astrocytes and related diseases has been conducted. In this paper, we review current studies on the neurophysiological roles of astrocytes following radiation exposure. We describe the pathophysiological changes in astrocytes, including astrogliosis, astrosenescence, and the associated cellular and molecular mechanisms. Additionally, we summarize the roles of astrocytes in radiation-induced impairments of neurogenesis and the blood-brain barrier (BBB). Based on current research, we propose that brain astrocytes may serve as potential therapeutic targets for treating radiation-induced brain injury (RIBI) and subsequent neurological and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Caiping Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 226001 Nantong, Jiangsu, China
- Radiation Physiology Laboratory, Singapore Nuclear Research and Safety Initiative, National University of Singapore, 138602 Singapore, Singapore
| | - Xingjuan Fan
- Department of Neurology, Affiliated Hospital of Nantong University, 226001 Nantong, Jiangsu, China
| | - Yunwei Shi
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 226001 Nantong, Jiangsu, China
- Radiation Physiology Laboratory, Singapore Nuclear Research and Safety Initiative, National University of Singapore, 138602 Singapore, Singapore
| | - Fengru Tang
- Radiation Physiology Laboratory, Singapore Nuclear Research and Safety Initiative, National University of Singapore, 138602 Singapore, Singapore
| |
Collapse
|
15
|
Benak D, Alanova P, Holzerova K, Chalupova M, Opletalova B, Kolar F, Pavlinkova G, Hlavackova M. Epitranscriptomic regulation of HIF-1: bidirectional regulatory pathways. Mol Med 2025; 31:105. [PMID: 40102715 PMCID: PMC11917031 DOI: 10.1186/s10020-025-01149-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Accepted: 03/03/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND Epitranscriptomics, the study of RNA modifications such as N6-methyladenosine (m6A), provides a novel layer of gene expression regulation with implications for numerous biological processes, including cellular adaptation to hypoxia. Hypoxia-inducible factor-1 (HIF-1), a master regulator of the cellular response to low oxygen, plays a critical role in adaptive and pathological processes, including cancer, ischemic heart disease, and metabolic disorders. Recent discoveries accent the dynamic interplay between m6A modifications and HIF-1 signaling, revealing a complex bidirectional regulatory network. While the roles of other RNA modifications in HIF-1 regulation remain largely unexplored, emerging evidence suggests their potential significance. MAIN BODY This review examines the reciprocal regulation between HIF-1 and epitranscriptomic machinery, including m6A writers, readers, and erasers. HIF-1 modulates the expression of key m6A components, while its own mRNA is regulated by m6A modifications, positioning HIF-1 as both a regulator and a target in this system. This interaction enhances our understanding of cellular hypoxic responses and opens avenues for clinical applications in treating conditions like cancer and ischemic heart disease. Promising progress has been made in developing selective inhibitors targeting the m6A-HIF-1 regulatory axis. However, challenges such as off-target effects and the complexity of RNA modification dynamics remain significant barriers to clinical translation. CONCLUSION The intricate interplay between m6A and HIF-1 highlights the critical role of epitranscriptomics in hypoxia-driven processes. Further research into these regulatory networks could drive therapeutic innovation in cancer, ischemic heart disease, and other hypoxia-related conditions. Overcoming challenges in specificity and off-target effects will be essential for realizing the potential of these emerging therapies.
Collapse
Affiliation(s)
- Daniel Benak
- Laboratory of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Petra Alanova
- Laboratory of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Kristyna Holzerova
- Laboratory of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Miloslava Chalupova
- Laboratory of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Barbora Opletalova
- Laboratory of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Frantisek Kolar
- Laboratory of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Gabriela Pavlinkova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology, Czech Academy of Sciences, Vestec, Czech Republic
| | - Marketa Hlavackova
- Laboratory of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic.
| |
Collapse
|
16
|
Lykkesfeldt J, Carr AC, Tveden-Nyborg P. The pharmacology of vitamin C. Pharmacol Rev 2025; 77:100043. [PMID: 39986139 DOI: 10.1016/j.pharmr.2025.100043] [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: 02/07/2024] [Accepted: 01/14/2025] [Indexed: 02/24/2025] Open
Abstract
Ascorbic acid, the reduced form of vitamin C, is a ubiquitous small carbohydrate. Despite decades of focused research, new metabolic functions of this universal electron donor are still being discovered and add to the complexity of our view of vitamin C in human health. Although praised as an unsurpassed water-soluble antioxidant in plasma and cells, the most interesting functions of vitamin C seem to be its roles as specific electron donor in numerous biological reactions ranging from the well-known hydroxylation of proline to cofactor for the epigenetic master regulators ten-eleven translocation enzymes and Jumonji domain-containing histone-lysine demethylases. Some of these functions may have important implications for disease prevention and treatment and have spiked renewed interest in, eg, vitamin C's potential in cancer therapy. Moreover, some fundamental pharmacokinetic properties of vitamin C remain to be established including if other mechanisms than passive diffusion governs the efflux of ascorbate anions from the cell. Taken together, there still seems to be much to learn about the pharmacology of vitamin C and its role in health and disease. This review explores new avenues of vitamin C and integrates our present knowledge of its pharmacology. SIGNIFICANCE STATEMENT: Vitamin C is involved in multiple biological reactions of which most are essential to human health. Hundreds of millions of people are considered deficient in vitamin C according to accepted guidelines, but little is known about the long-term consequences. Although the complexity of vitamin C's physiology and pharmacology has been widely disregarded in clinical studies for decades, it seems clear that a deeper understanding of particularly its pharmacology holds the key to unravel and possibly exploit the potential of vitamin C in disease prevention and therapy.
Collapse
Affiliation(s)
- Jens Lykkesfeldt
- Section of Biomedicine, Department of Veterinary and Animal Science, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Anitra C Carr
- Nutrition in Medicine Research Group, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Pernille Tveden-Nyborg
- Section of Biomedicine, Department of Veterinary and Animal Science, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
17
|
McDermott A, Tavassoli A. Hypoxia-inducible transcription factors: architects of tumorigenesis and targets for anticancer drug discovery. Transcription 2025; 16:86-117. [PMID: 39470609 PMCID: PMC11970764 DOI: 10.1080/21541264.2024.2417475] [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: 05/21/2024] [Revised: 10/10/2024] [Accepted: 10/12/2024] [Indexed: 10/30/2024] Open
Abstract
Hypoxia-inducible factors (HIFs) play a pivotal role as master regulators of tumor survival and growth, controlling a wide array of cellular processes in response to hypoxic stress. Clinical data correlates upregulated HIF-1 and HIF-2 levels with an aggressive tumor phenotype and poor patient outcome. Despite extensive validation as a target in cancer, pharmaceutical targeting of HIFs, particularly the interaction between α and βsubunits that forms the active transcription factor, has proved challenging. Nonetheless, many indirect inhibitors of HIFs have been identified, targeting diverse parts of this pathway. Significant strides have also been made in the development of direct inhibitors of HIF-2, exemplified by the FDA approval of Belzutifan for the treatment of metastatic clear cell renal carcinoma. While efforts to target HIF-1 using various therapeutic modalities have shown promise, no clinical candidates have yet emerged. This review aims to provide insights into the intricate and extensive role played by HIFs in cancer, and the ongoing efforts to develop therapeutic agents against this target.
Collapse
Affiliation(s)
| | - Ali Tavassoli
- School of Chemistry, University of Southampton, Southampton, UK
| |
Collapse
|
18
|
Qiao Z, Nguyen LC, Yang D, Dann C, Thomas DM, Henn M, Valdespino A, Swenson CS, Oakes SA, Rosner MR, Moellering RE. Direct inhibition of tumor hypoxia response with synthetic transcriptional repressors. Nat Chem Biol 2025; 21:247-255. [PMID: 39215099 DOI: 10.1038/s41589-024-01716-z] [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: 11/03/2023] [Accepted: 08/01/2024] [Indexed: 09/04/2024]
Abstract
Many oncogenic transcription factors (TFs) are considered to be undruggable because of their reliance on large protein-protein and protein-DNA interfaces. TFs such as hypoxia-inducible factors (HIFs) and X-box-binding protein 1 (XBP1) are induced by hypoxia and other stressors in solid tumors and bind to unfolded protein response element (UPRE) and hypoxia-induced response element (HRE) motifs to control oncogenic gene programs. Here, we report a strategy to create synthetic transcriptional repressors (STRs) that mimic the basic leucine zipper domain of XBP1 and recognize UPRE and HRE motifs. A lead molecule, STR22, binds UPRE and HRE DNA sequences with high fidelity and competes with both TFs in cells. Under hypoxia, STR22 globally suppresses HIF1α binding to HRE-containing promoters and enhancers, inhibits hypoxia-induced gene expression and blocks protumorigenic phenotypes in triple-negative breast cancer (TNBC) cells. In vivo, intratumoral and systemic STR22 treatment inhibited hypoxia-dependent gene expression, primary tumor growth and metastasis of TNBC tumors. These data validate a novel strategy to target the tumor hypoxia response through coordinated inhibition of TF-DNA binding.
Collapse
Affiliation(s)
- Zeyu Qiao
- Department of Chemistry, The University of Chicago, Chicago, IL, USA
- Institute for Genomics and Systems Biology, The University of Chicago, Chicago, IL, USA
| | - Long C Nguyen
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL, USA
| | - Dongbo Yang
- Department of Chemistry, The University of Chicago, Chicago, IL, USA
- Institute for Genomics and Systems Biology, The University of Chicago, Chicago, IL, USA
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL, USA
| | - Christopher Dann
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL, USA
| | - Deborah M Thomas
- Department of Chemistry, The University of Chicago, Chicago, IL, USA
- Institute for Genomics and Systems Biology, The University of Chicago, Chicago, IL, USA
| | - Madeline Henn
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL, USA
| | - Andrea Valdespino
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL, USA
| | - Colin S Swenson
- Department of Chemistry, The University of Chicago, Chicago, IL, USA
- Institute for Genomics and Systems Biology, The University of Chicago, Chicago, IL, USA
| | - Scott A Oakes
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - Marsha Rich Rosner
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL, USA.
| | - Raymond E Moellering
- Department of Chemistry, The University of Chicago, Chicago, IL, USA.
- Institute for Genomics and Systems Biology, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
19
|
Wang L, Nakamura A. Where are we in targeting hypoxia-induced pathways in inflammatory arthritis? Current understanding, insights, and future directions. Int Immunopharmacol 2025; 146:113883. [PMID: 39718060 DOI: 10.1016/j.intimp.2024.113883] [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: 09/30/2024] [Revised: 11/30/2024] [Accepted: 12/15/2024] [Indexed: 12/25/2024]
Abstract
INTRODUCTION Joint tissues affected by inflammatory arthritis (IA) create hypoxic microenvironments that sustain the inflammatory response. Although targeting molecules in hypoxia-induced pathways has provided valuable insights into potential novel therapies for various types of IA, progress remains preclinical, and no clinical trials have been conducted for IA. METHODS A literature search was conducted to create a narrative review exploring the role of hypoxia and its signaling pathways in IA pathogenesis, as well as the potential and future directions for IA therapies that target hypoxia-induced molecules before moving forward to clinical applications. RESULTS Hypoxia is a prevalent feature of the IA synovial microenvironment and contributes to disease progression. Various studies and preclinical models demonstrate how hypoxia-inducible factors, vascular endothelial growth factors, and matrix metalloproteinases, among other molecules, influence rheumatoid arthritis, axial spondyloarthritis, psoriatic arthritis, and juvenile idiopathic arthritis. Despite these findings, drug development targeting these molecules in IA has been limited due to challenges in delineating the mechanistic pathways of hypoxia, the distinct roles of hypoxia-induced molecules depending on anatomical sites, and concerns regarding pharmacokinetics and patient safety. However, given that hypoxia-induced molecule-targeting therapies have been successfully approved for treating cancers and cardiovascular diseases, further research is needed to advance the application of similar medications in IA. CONCLUSIONS Given the pathogenic effects of hypoxic microenvironments in IA, it is imperative to continue gathering compelling evidence to advance hypoxia-induced therapies. Furthermore, elucidating the safety and efficacy of such drugs in various preclinical models, in collaboration with chemists and the pharmaceutical industry, is crucial for accelerating the development of novel, optimized treatment methods.
Collapse
Affiliation(s)
- Lisa Wang
- Department of Medicine, Division of Rheumatology, Queen's University, Kingston, Ontario, Canada; Faculty of Health Sciences, School of Medicine, Queen's University, Kingston, Ontario, Canada.
| | - Akihiro Nakamura
- Department of Medicine, Division of Rheumatology, Queen's University, Kingston, Ontario, Canada; Faculty of Health Sciences, School of Medicine, Queen's University, Kingston, Ontario, Canada; Translational Institute of Medicine, Department of Medicine, Queen's University, Ontario, Canada; Rheumatology Clinic, Kingston Health Science Centre, Kingston, Ontario, Canada.
| |
Collapse
|
20
|
Rodriguez R, Müller S, Colombeau L, Solier S, Sindikubwabo F, Cañeque T. Metal Ion Signaling in Biomedicine. Chem Rev 2025; 125:660-744. [PMID: 39746035 PMCID: PMC11758815 DOI: 10.1021/acs.chemrev.4c00577] [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: 08/02/2024] [Revised: 11/10/2024] [Accepted: 12/10/2024] [Indexed: 01/04/2025]
Abstract
Complex multicellular organisms are composed of distinct tissues involving specialized cells that can perform specific functions, making such life forms possible. Species are defined by their genomes, and differences between individuals within a given species directly result from variations in their genetic codes. While genetic alterations can give rise to disease-causing acquisitions of distinct cell identities, it is now well-established that biochemical imbalances within a cell can also lead to cellular dysfunction and diseases. Specifically, nongenetic chemical events orchestrate cell metabolism and transcriptional programs that govern functional cell identity. Thus, imbalances in cell signaling, which broadly defines the conversion of extracellular signals into intracellular biochemical changes, can also contribute to the acquisition of diseased cell states. Metal ions exhibit unique chemical properties that can be exploited by the cell. For instance, metal ions maintain the ionic balance within the cell, coordinate amino acid residues or nucleobases altering folding and function of biomolecules, or directly catalyze specific chemical reactions. Thus, metals are essential cell signaling effectors in normal physiology and disease. Deciphering metal ion signaling is a challenging endeavor that can illuminate pathways to be targeted for therapeutic intervention. Here, we review key cellular processes where metal ions play essential roles and describe how targeting metal ion signaling pathways has been instrumental to dissecting the biochemistry of the cell and how this has led to the development of effective therapeutic strategies.
Collapse
Affiliation(s)
- Raphaël Rodriguez
- Institut
Curie, CNRS, INSERM, PSL Research University, 75005 Paris, France
| | - Sebastian Müller
- Institut
Curie, CNRS, INSERM, PSL Research University, 75005 Paris, France
| | - Ludovic Colombeau
- Institut
Curie, CNRS, INSERM, PSL Research University, 75005 Paris, France
| | - Stéphanie Solier
- Institut
Curie, CNRS, INSERM, PSL Research University, 75005 Paris, France
- Université
Paris-Saclay, UVSQ, 78180 Montigny-le-Bretonneux, France
| | | | - Tatiana Cañeque
- Institut
Curie, CNRS, INSERM, PSL Research University, 75005 Paris, France
| |
Collapse
|
21
|
Shi Y, Gilkes DM. HIF-1 and HIF-2 in cancer: structure, regulation, and therapeutic prospects. Cell Mol Life Sci 2025; 82:44. [PMID: 39825916 PMCID: PMC11741981 DOI: 10.1007/s00018-024-05537-0] [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: 08/05/2024] [Revised: 10/27/2024] [Accepted: 12/01/2024] [Indexed: 01/20/2025]
Abstract
Hypoxia, or a state of low tissue oxygenation, has been characterized as an important feature of solid tumors that is related to aggressive phenotypes. The cellular response to hypoxia is controlled by Hypoxia-inducible factors (HIFs), a family of transcription factors. HIFs promote the transcription of gene products that play a role in tumor progression including proliferation, angiogenesis, metastasis, and drug resistance. HIF-1 and HIF-2 are well known and widely described. Although these proteins share a high degree of homology, HIF-1 and HIF-2 have non-redundant roles in cancer. In this review, we summarize the similarities and differences between HIF-1α and HIF-2α in their structure, expression, and DNA binding. We also discuss the canonical and non-canonical regulation of HIF-1α and HIF-2α under hypoxic and normal conditions. Finally, we outline recent strategies aimed at targeting HIF-1α and/or HIF-2α.
Collapse
Affiliation(s)
- Yi Shi
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Daniele M Gilkes
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
22
|
Caca J, Bartelt A, Egea V. Hypoxia Regulates Brown Adipocyte Differentiation and Stimulates miR-210 by HIF-1α. Int J Mol Sci 2024; 26:117. [PMID: 39795975 PMCID: PMC11720532 DOI: 10.3390/ijms26010117] [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: 12/06/2024] [Revised: 12/19/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
MicroRNAs (miRNAs) are short sequences of single-stranded non-coding RNAs that target messenger RNAs, leading to their repression or decay. Interestingly, miRNAs play a role in the cellular response to low oxygen levels, known as hypoxia, which is associated with reactive oxygen species and oxidative stress. However, the physiological implications of hypoxia-induced miRNAs ("hypoxamiRs") remain largely unclear. Here, we investigate the role of miR-210 in brown adipocyte differentiation and thermogenesis. We treated the cells under sympathetic stimulation with hypoxia, CoCl2, or IOX2. To manipulate miR-210, we performed reverse transfection with antagomiRs. Adipocyte markers expression, lipid accumulation, lipolysis, and oxygen consumption were measured. Hypoxia hindered BAT differentiation and suppressed sympathetic stimulation. Hypoxia-induced HIF-1α stabilization increased miR-210 in brown adipocytes. Interestingly, miR-210-5p enhanced differentiation under normoxic conditions but was insufficient to rescue the inhibition of brown adipocyte differentiation under hypoxic conditions. Although adrenergic stimulation activated HIF-1α signaling and upregulated miR-210 expression, inhibition of miR-210-5p did not significantly influence UCP1 expression or oxygen consumption. In summary, hypoxia and adrenergic stimulation upregulated miR-210, which impacted brown adipocyte differentiation and thermogenesis. These findings offer new insights for the physiological role of hypoxamiRs in brown adipose tissue, which could aid in understanding oxidative stress and treatment of metabolic disorders.
Collapse
Affiliation(s)
- Jan Caca
- Institute for Cardiovascular Prevention (IPEK), Faculty of Medicine, Ludwig-Maximilians-Universität München, 81377 Munich, Germany;
| | - Alexander Bartelt
- Institute for Cardiovascular Prevention (IPEK), Faculty of Medicine, Ludwig-Maximilians-Universität München, 81377 Munich, Germany;
- German Center for Cardiovascular Research, Partner Site Munich Heart Alliance, Ludwig-Maximilians-Universität München, 80336 Munich, Germany
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Neuherberg, Germany
- German Center for Diabetes Research, 85764 Neuherberg, Germany
- Chair of Translational Nutritional Medicine, Department of Molecular Life Sciences, TUM School of Life Sciences, Technical University of Munich, 85354 Freising, Germany
- Else Kröner Fresenius Center for Nutritional Medicine, Technical University of Munich, 85354 Munich, Germany
| | - Virginia Egea
- Institute for Cardiovascular Prevention (IPEK), Faculty of Medicine, Ludwig-Maximilians-Universität München, 81377 Munich, Germany;
- German Center for Cardiovascular Research, Partner Site Munich Heart Alliance, Ludwig-Maximilians-Universität München, 80336 Munich, Germany
| |
Collapse
|
23
|
Chen CH, Hsu WL, Tsai PSJ, Lai CF, Wu MT, Lee YJ. Evaluation of hypoxia-inducible factor-1α and urine non-transferrin-bound iron concentrations in cats with chronic kidney disease. Front Vet Sci 2024; 11:1482998. [PMID: 39748872 PMCID: PMC11694447 DOI: 10.3389/fvets.2024.1482998] [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: 08/19/2024] [Accepted: 12/09/2024] [Indexed: 01/04/2025] Open
Abstract
Introduction Hypoxia-inducible factors (HIF) regulate gene transcription, which aids hypoxia adaptation while promoting renal fibrosis. Non-transferrin-bound iron (NTBI) is a catalytic form of iron that can lead to oxidative damage. However, NTBI in cat biofluids has rarely been evaluated. Aims We assessed cat plasma and urine HIF-1α (pHIF-1α/uHIF-1α) concentrations and urine NTBI (uNTBI) concentrations to investigate their relationship with chronic kidney disease (CKD) severity. Methods pHIF-1α and uHIF-1α concentrations were measured using commercial ELISA kits, while uNTBI concentrations were detected by inductively coupled plasma mass spectrometry. Results Healthy cats (n = 35) and cats with CKD (n = 84) formed the study cohorts. pHIF-1α concentrations increased from 9.48 pg./mL (median) in the healthy cohort to 11.42 pg./mL in early-stage CKD cats but decreased to 8.50 pg./mL in late-stage CKD cats. uHIF-1α concentrations gradually decreased with a significant difference between the control group (44.61 pg./mL) and the late-stage CKD group (36.79 pg./mL, p < 0.001). Cats with proteinuria had significantly higher uNTBI concentrations (35.61 ppb) than non-proteinuric cats (25.13 ppb, p = 0.019). Finally, the concentrations of pHIF-1α and uHIF-1α were positively correlated independent of renal function. Conclusion and clinical importance Overall, pHIF-1α and uHIF-1α concentrations are lower in advanced CKD cats, while uNTBI concentrations are significantly higher in proteinuric cats.
Collapse
Affiliation(s)
- Chien-Hui Chen
- School of Veterinary Medicine, College of Bio-Resources and Agriculture, Institute of Veterinary Clinical Science, National Taiwan University, Taipei, Taiwan
- Veterinary Hospital, College of Bio-Resources and Agriculture, National Taiwan University, Taipei, Taiwan
| | - Wei-Li Hsu
- College of Veterinary Medicine, Graduate Institute of Microbiology and Public Health, National Chung-Hsing University, Taichung, Taiwan
| | - Pei-Shiue Jason Tsai
- School of Veterinary Medicine, College of Bio-Resources and Agriculture, Institute of Veterinary Medicine, National Taiwan University, Taipei, Taiwan
| | - Chun-Fu Lai
- National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Meng-Ting Wu
- Department of Chemistry, College of Science, National Taiwan University, Taipei, Taiwan
| | - Ya-Jane Lee
- School of Veterinary Medicine, College of Bio-Resources and Agriculture, Institute of Veterinary Clinical Science, National Taiwan University, Taipei, Taiwan
- Veterinary Hospital, College of Bio-Resources and Agriculture, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
24
|
Pauzaite T, Nathan JA. A closer look at the role of deubiquitinating enzymes in the Hypoxia Inducible Factor pathway. Biochem Soc Trans 2024; 52:2253-2265. [PMID: 39584532 PMCID: PMC11668284 DOI: 10.1042/bst20230861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 10/31/2024] [Accepted: 11/01/2024] [Indexed: 11/26/2024]
Abstract
Hypoxia Inducible transcription Factors (HIFs) are central to the metazoan oxygen-sensing response. Under low oxygen conditions (hypoxia), HIFs are stabilised and govern an adaptive transcriptional programme to cope with prolonged oxygen starvation. However, when oxygen is present, HIFs are continuously degraded by the proteasome in a process involving prolyl hydroxylation and subsequent ubiquitination by the Von Hippel Lindau (VHL) E3 ligase. The essential nature of VHL in the HIF response is well established but the role of other enzymes involved in ubiquitination is less clear. Deubiquitinating enzymes (DUBs) counteract ubiquitination and provide an important regulatory aspect to many signalling pathways involving ubiquitination. In this review, we look at the complex network of ubiquitination and deubiquitination in controlling HIF signalling in normal and low oxygen tensions. We discuss the relative importance of DUBs in opposing VHL, and explore roles of DUBs more broadly in hypoxia, in both VHL and HIF independent contexts. We also consider the catalytic and non-catalytic roles of DUBs, and elaborate on the potential benefits and challenges of inhibiting these enzymes for therapeutic use.
Collapse
Affiliation(s)
- Tekle Pauzaite
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah, Biomedical Centre, Department of Medicine, University of Cambridge, Cambridge CB2 0AW, U.K
| | - James A. Nathan
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah, Biomedical Centre, Department of Medicine, University of Cambridge, Cambridge CB2 0AW, U.K
| |
Collapse
|
25
|
Wang X, Zhai H, Guo J, Wang X, Gu L, Li T, Liu Q. siRNA silencing and hypoxia challenge indicate that the function of common carp (Cyprinus carpio) hif-1αb genes are tightly linked to hif-1αa and hif-3α genes. BMC Genomics 2024; 25:1203. [PMID: 39695393 DOI: 10.1186/s12864-024-11141-0] [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: 06/13/2024] [Accepted: 12/09/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Fishes are susceptible to hypoxia stress, while the common carp is known for its high tolerance to hypoxia. The hypoxia-inducible factor (HIF) pathway directly regulates the cell's response to hypoxia. Still, it is currently unknown which members of the hif-α genes are present in common carp and their specific functions. RESULTS In this study, we found that the hif-1α, hif-2α, and hif-3α genes of common carp all contained twice the number of copies of their orthologs in zebrafish. Common carp has four copies of the hif-1α gene, of which the two hif-1αa genes were expressed at low levels in the vast majority of tissues, while the two hif-1αb genes were expressed at high levels in multiple tissues. We silenced the two hif-1αb genes using chitosan nanoparticles (CSNPs) carrying siRNA and subjected two groups to hypoxic stress. Transcriptome sequencing results show that whether under normoxia or hypoxia, the number of differentially expressed genes (DEGs) caused by silencing the hif-1αb genes in the heart exceeds 1,000, far more than the number of DEGs in the gills or brain. GO enrichment and KEGG enrichment showed that DEGs in the heart were mainly related to immune function and myocardial contraction. DEGs in the gills and brain also enriched many immune-related terms, and some DEGs in the gills were related to iron metabolism and erythropoiesis. Among the paralogs, the two hif-1αa genes were most obviously up-regulated under normoxia, while the hif-3α genes were most obviously up-regulated under hypoxia. We did not find any downstream genes of the HIF pathway that were specifically regulated by the hif-1αb genes. CONCLUSIONS The main effect site of the common carp hif-1αb genes is the heart, and their main functions are to regulate immune response and myocardial contraction. Their functions are partially redundant with the hif-1αa genes and hif-3α genes. When their expressions are inhibited, the expression of hif-1αa genes or hif-3α genes would be up-regulated in specific contexts, thereby compensating for their loss of function. The downstream genes of the HIF pathway in common carp may be generally regulated by multiple hif-α genes.
Collapse
Grants
- [20210302124494] the Department of Science and Technology of Shanxi Province
- [20210302124494] the Department of Science and Technology of Shanxi Province
- [20210302124494] the Department of Science and Technology of Shanxi Province
- [20210302124494] the Department of Science and Technology of Shanxi Province
- [20210302124494] the Department of Science and Technology of Shanxi Province
- [20210302124494] the Department of Science and Technology of Shanxi Province
- [20210302124494] the Department of Science and Technology of Shanxi Province
- [J201911301, J202111303, 2020L0158, SXYBKY201713] the Department of Education of Shanxi Province
- [J201911301, J202111303, 2020L0158, SXYBKY201713] the Department of Education of Shanxi Province
- [J201911301, J202111303, 2020L0158, SXYBKY201713] the Department of Education of Shanxi Province
- [J201911301, J202111303, 2020L0158, SXYBKY201713] the Department of Education of Shanxi Province
- [J201911301, J202111303, 2020L0158, SXYBKY201713] the Department of Education of Shanxi Province
- [J201911301, J202111303, 2020L0158, SXYBKY201713] the Department of Education of Shanxi Province
- [J201911301, J202111303, 2020L0158, SXYBKY201713] the Department of Education of Shanxi Province
- [2017YJ05] Shanxi Agricultural University
- [2017YJ05] Shanxi Agricultural University
- [2017YJ05] Shanxi Agricultural University
- [2017YJ05] Shanxi Agricultural University
- [2017YJ05] Shanxi Agricultural University
- [2017YJ05] Shanxi Agricultural University
- [2017YJ05] Shanxi Agricultural University
Collapse
Affiliation(s)
- Xianzong Wang
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China.
- Shanxi Key Laboratory of Animal Genetics Resource Utilization and Breeding, Shanxi Agricultural University, Taigu, 030801, China.
| | - Huili Zhai
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Jiali Guo
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Xueyi Wang
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Libo Gu
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Tongyao Li
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Qing Liu
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China.
- Shanxi Key Laboratory of Animal Genetics Resource Utilization and Breeding, Shanxi Agricultural University, Taigu, 030801, China.
| |
Collapse
|
26
|
Carullo N, Sorbo D, Faga T, Pugliese S, Zicarelli MT, Costa D, Ielapi N, Battaglia Y, Pisani A, Coppolino G, Bolignano D, Michael A, Serra R, Andreucci M. Anemia and Mineral Bone Disorder in Kidney Disease Patients: The Role of FGF-23 and Other Related Factors. Int J Mol Sci 2024; 25:12838. [PMID: 39684548 DOI: 10.3390/ijms252312838] [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: 10/27/2024] [Revised: 11/20/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Anemia and mineral and bone disorder (MBD) are significant complications of chronic kidney disease (CKD). The erythropoietin (Epo) pathway plays a key role in both of these processes in CKD. Another molecule that plays an important role in CKD-MBD is fibroblast growth factor (FGF)-23, whose main role is to maintain serum phosphate levels in the normal range, acting via its co-receptor Klotho; however, its activity may also be related to anemia and inflammation. In this review, the regulation of Epo and FGF-23 and the molecular mechanisms of their action are outlined. Furthermore, the complex interaction between EPO and FGF-23 is discussed, as well as their association with other anemia-related factors and processes such as Klotho, vitamin D, and iron deficiency. Together, these may be part of a "kidney-bone marrow-bone axis" that promotes CKD-MBD.
Collapse
Affiliation(s)
- Nazareno Carullo
- "G. Jazzolino" Hospital, A.S.P. Vibo Valentia, I89900 Vibo Valentia, Italy
| | - David Sorbo
- San Bortolo Hospital, ULSS 8 Berica, I36100 Vicenza, Italy
| | - Teresa Faga
- Department of Health Sciences, "Magna Graecia" University, I88100 Catanzaro, Italy
| | - Sara Pugliese
- Department of Health Sciences, "Magna Graecia" University, I88100 Catanzaro, Italy
| | - Maria Teresa Zicarelli
- Amantea Outpatient Clinic, A.S.P. Cosenza, I87032 Amantea, Italy
- Department of Medical and Surgical Sciences, "Magna Graecia" University, I88100 Catanzaro, Italy
| | - Davide Costa
- Department of Medical and Surgical Sciences, "Magna Graecia" University, I88100 Catanzaro, Italy
- Interuniversity Center of Phlebolymphology (CIFL), "Magna Graecia" University, I88100 Catanzaro, Italy
| | - Nicola Ielapi
- Interuniversity Center of Phlebolymphology (CIFL), "Magna Graecia" University, I88100 Catanzaro, Italy
- Department of Public Health and Infectious Disease, "Sapienza" University of Rome, I00185 Rome, Italy
| | - Yuri Battaglia
- Department of Medicine, University of Verona, I37129 Verona, Italy
| | - Antonio Pisani
- Department of Public Health, University of Naples Federico II, I80131 Naples, Italy
| | - Giuseppe Coppolino
- Department of Health Sciences, "Magna Graecia" University, I88100 Catanzaro, Italy
| | - Davide Bolignano
- Department of Medical and Surgical Sciences, "Magna Graecia" University, I88100 Catanzaro, Italy
| | - Ashour Michael
- Department of Health Sciences, "Magna Graecia" University, I88100 Catanzaro, Italy
| | - Raffaele Serra
- Department of Medical and Surgical Sciences, "Magna Graecia" University, I88100 Catanzaro, Italy
- Interuniversity Center of Phlebolymphology (CIFL), "Magna Graecia" University, I88100 Catanzaro, Italy
| | - Michele Andreucci
- Department of Health Sciences, "Magna Graecia" University, I88100 Catanzaro, Italy
| |
Collapse
|
27
|
周 志, 孙 凡, 江 秉. [Research Progress in the Role of Hypoxia-Inducible Factor 1 in Altitude Sickness and the Mechanisms Involved]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2024; 55:1424-1435. [PMID: 39990820 PMCID: PMC11839359 DOI: 10.12182/20241160303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Indexed: 02/25/2025]
Abstract
Individuals who reside at high altitudes for extended periods or those who visit these regions briefly frequently experience high-altitude response, which triggers a series of physiological and pathological changes in the body, ultimately causing altitude sickness. One of the most critical features of high-altitude environments is hypoxia. Recent studies have demonstrated that hypoxia-inducible factor 1 (HIF-1) plays a central role in mediating the body's response to hypoxic conditions at high altitudes. HIF-1, a heterodimeric transcription factor composed of an oxygen-sensitive subunit α (HIF-1α) and a constitutively expressed subunit β (HIF-1β), directly regulates the expression of multiple target genes, thereby modulating various physiological processes essential for cellular adaptation to hypoxia. According to a substantial body of research, aberrant expression of HIF-1 is implicated in the pathogenesis and progression of various diseases, including altitude sickness, cardiovascular disorders, neurological conditions, inflammatory diseases, cognitive impairment, immune dysregulation, and cancer. In this review, we provided an in-depth examination of the structural characteristics and regulatory mechanisms governing HIF-1 expression, discussed its downstream target genes, and highlighted the inhibitors currently under development. Additionally, we summarized the pivotal role and underlying mechanisms of HIF-1 in the development of altitude sickness, particularly its regulatory role in the pathophysiological processes of high-altitude pulmonary edema (HAPE), high-altitude cerebral edema (HACE), and high-altitude pulmonary hypertension (HAPH). Through a thorough examination of the role of HIF-1, we aim to provide a theoretical foundation and potential therapeutic targets for the prevention and treatment of altitude sickness.
Collapse
Affiliation(s)
- 志豪 周
- 天健先进生物医学实验室 郑州大学医学科学院(河南 450000)Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450000, China
| | - 凡丽 孙
- 天健先进生物医学实验室 郑州大学医学科学院(河南 450000)Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450000, China
| | - 秉华 江
- 天健先进生物医学实验室 郑州大学医学科学院(河南 450000)Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450000, China
| |
Collapse
|
28
|
Lin TK, Huang CR, Lin KJ, Hsieh YH, Chen SD, Lin YC, Chao AC, Yang DI. Potential Roles of Hypoxia-Inducible Factor-1 in Alzheimer's Disease: Beneficial or Detrimental? Antioxidants (Basel) 2024; 13:1378. [PMID: 39594520 PMCID: PMC11591038 DOI: 10.3390/antiox13111378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/04/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
The major pathological characteristics of Alzheimer's disease (AD) include senile plaques and neurofibrillary tangles (NFTs), which are mainly composed of aggregated amyloid-beta (Aβ) peptide and hyperphosphorylated tau protein, respectively. The excessive production of reactive oxygen species (ROS) and neuroinflammation are crucial contributing factors to the pathological mechanisms of AD. Hypoxia-inducible factor-1 (HIF-1) is a transcription factor critical for tissue adaption to low-oxygen tension. Growing evidence has suggested HIF-1 as a potential therapeutic target for AD; conversely, other experimental findings indicate that HIF-1 induction contributes to AD pathogenesis. These previous findings thus point to the complex, even contradictory, roles of HIF-1 in AD. In this review, we first introduce the general pathogenic mechanisms of AD as well as the potential pathophysiological roles of HIF-1 in cancer, immunity, and oxidative stress. Based on current experimental evidence in the literature, we then discuss the possible beneficial as well as detrimental mechanisms of HIF-1 in AD; these sections also include the summaries of multiple chemical reagents and proteins that have been shown to exert beneficial effects in AD via either the induction or inhibition of HIF-1.
Collapse
Affiliation(s)
- Tsu-Kung Lin
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833401, Taiwan; (T.-K.L.); (C.-R.H.); (S.-D.C.)
- College of Medicine, Chang Gung University, Taoyuan 333323, Taiwan
| | - Chi-Ren Huang
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833401, Taiwan; (T.-K.L.); (C.-R.H.); (S.-D.C.)
- College of Medicine, Chang Gung University, Taoyuan 333323, Taiwan
| | - Kai-Jung Lin
- Department of Family Medicine, National Taiwan University Hospital, Taipei 100225, Taiwan;
| | - Yi-Heng Hsieh
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan;
| | - Shang-Der Chen
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833401, Taiwan; (T.-K.L.); (C.-R.H.); (S.-D.C.)
| | - Yi-Chun Lin
- Department of Neurology, Taipei City Hospital Renai Branch, Taipei 106243, Taiwan;
| | - A-Ching Chao
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung 807377, Taiwan
- Department of Neurology, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
- Department of Sports Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
| | - Ding-I Yang
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan;
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| |
Collapse
|
29
|
Pruitt L, Abbott RK. Hypoxia-adenosinergic regulation of B cell responses. Front Immunol 2024; 15:1478506. [PMID: 39559353 PMCID: PMC11570280 DOI: 10.3389/fimmu.2024.1478506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 10/15/2024] [Indexed: 11/20/2024] Open
Abstract
Hypoxic microenvironments induce widespread metabolic changes that have been shown to be critical in regulating innate and adaptive immune responses. Hypoxia-induced changes include the generation of extracellular adenosine followed by subsequent signaling through adenosine receptors on immune cells. This evolutionarily conserved "hypoxia-adenosinergic" pathway of hypoxia → extracellular adenosine → adenosine receptor signaling has been shown to be critical in limiting and redirecting T cell responses including in tumor microenvironments and the gut mucosa. However, the question of whether hypoxic microenvironments are involved in the development of B cell responses has remained unexplored until recently. The discovery that germinal centers (GC), the anatomic site in which B cells undergo secondary diversification and affinity maturation, develop a hypoxic microenvironment has sparked new interest in how this evolutionarily conserved pathway affects antibody responses. In this review we will summarize what is known about hypoxia-adenosinergic microenvironments in lymphocyte development and ongoing immune responses. Specific focus will be placed on new developments regarding the role of the hypoxia-adenosinergic pathway in regulating GC development and humoral immunity.
Collapse
Affiliation(s)
| | - Robert K. Abbott
- Department of Pathology, University of Texas Medical Branch,
Galveston, TX, United States
| |
Collapse
|
30
|
Liao C, Hu L, Zhang Q. Von Hippel-Lindau protein signalling in clear cell renal cell carcinoma. Nat Rev Urol 2024; 21:662-675. [PMID: 38698165 DOI: 10.1038/s41585-024-00876-w] [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] [Accepted: 03/26/2024] [Indexed: 05/05/2024]
Abstract
The distinct pathological and molecular features of kidney cancer in adaptation to oxygen homeostasis render this malignancy an attractive model for investigating hypoxia signalling and potentially developing potent targeted therapies. Hypoxia signalling has a pivotal role in kidney cancer, particularly within the most prevalent subtype, known as renal cell carcinoma (RCC). Hypoxia promotes various crucial pathological processes, such as hypoxia-inducible factor (HIF) activation, angiogenesis, proliferation, metabolic reprogramming and drug resistance, all of which contribute to kidney cancer development, growth or metastasis formation. A substantial portion of kidney cancers, in particular clear cell RCC (ccRCC), are characterized by a loss of function of Von Hippel-Lindau tumour suppressor (VHL), leading to the accumulation of HIF proteins, especially HIF2α, a crucial driver of ccRCC. Thus, therapeutic strategies targeting pVHL-HIF signalling have been explored in ccRCC, culminating in the successful development of HIF2α-specific antagonists such as belzutifan (PT2977), an FDA-approved drug to treat VHL-associated diseases including advanced-stage ccRCC. An increased understanding of hypoxia signalling in kidney cancer came from the discovery of novel VHL protein (pVHL) targets, and mechanisms of synthetic lethality with VHL mutations. These breakthroughs can pave the way for the development of innovative and potent combination therapies in kidney cancer.
Collapse
Affiliation(s)
- Chengheng Liao
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Lianxin Hu
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Qing Zhang
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
31
|
Kling L, Eulenberg-Gustavus C, Jerke U, Rousselle A, Eckardt KU, Schreiber A, Kettritz R. β 2-integrins control HIF1α activation in human neutrophils. Front Immunol 2024; 15:1406967. [PMID: 39469705 PMCID: PMC11513320 DOI: 10.3389/fimmu.2024.1406967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 09/12/2024] [Indexed: 10/30/2024] Open
Abstract
During inflammation, human neutrophils engage β2-integrins to migrate from the blood circulation to inflammatory sites with high cytokine but low oxygen concentrations. We tested the hypothesis that the inhibition of prolyl hydroxylase domain-containing enzymes (PHDs), cytokines, and β2-integrins cooperates in HIF pathway activation in neutrophils. Using either the PHD inhibitor roxadustat (ROX) (pseudohypoxia) or normobaric hypoxia to stabilize HIF, we observed HIF1α protein accumulation in adherent neutrophils. Several inflammatory mediators did not induce HIF1α protein but provided additive or even synergistic signals (e.g., GM-CSF) under pseudohypoxic and hypoxic conditions. Importantly, and in contrast to adherent neutrophils, HIF1α protein expression was not detected in strictly suspended neutrophils despite PHD enzyme inhibition and the presence of inflammatory mediators. Blocking β2-integrins in adherent and activating β2-integrins in suspension neutrophils established the indispensability of β2-integrins for increasing HIF1α protein. Using GM-CSF as an example, increased HIF1α mRNA transcription via JAK2-STAT3 was necessary but not sufficient for HIF1α protein upregulation. Importantly, we found that β2-integrins led to HIF1α mRNA translation through the phosphorylation of the essential translation initiation factors eIF4E and 4EBP1. Finally, pseudohypoxic and hypoxic conditions inducing HIF1α consistently delayed apoptosis in adherent neutrophils on fibronectin under low serum concentrations. Pharmacological HIF1α inhibition reversed delayed apoptosis, supporting the importance of this pathway for neutrophil survival under conditions mimicking extravascular sites. We describe a novel β2-integrin-controlled mechanism of HIF1α stabilization in human neutrophils. Conceivably, this mechanism restricts HIF1α activation in response to hypoxia and pharmacological PHD enzyme inhibitors to neutrophils migrating toward inflammatory sites.
Collapse
Affiliation(s)
- Lovis Kling
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité – Universitätsmedizin Berlin, Berlin, Germany
- Department of Nephrology and Medical Intensive Care, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Claudia Eulenberg-Gustavus
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Uwe Jerke
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Anthony Rousselle
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Kai-Uwe Eckardt
- Department of Nephrology and Medical Intensive Care, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Adrian Schreiber
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité – Universitätsmedizin Berlin, Berlin, Germany
- Department of Nephrology and Medical Intensive Care, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ralph Kettritz
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité – Universitätsmedizin Berlin, Berlin, Germany
- Department of Nephrology and Medical Intensive Care, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
32
|
Bjørgen H, Brimsholm M, Asserson CF, Skaar K, Knutsen GM, Oaland Ø, Haldorsen R, Fjelldal PG, Hansen T, Rimstad E, Kleist BA, Lund-Iversen M, Kowalewski MP, Koppang EO. Deciphering the pathogenesis of melanized focal changes in the white skeletal muscle of farmed Atlantic salmon (Salmo salar). JOURNAL OF FISH DISEASES 2024; 47:e13988. [PMID: 38943363 DOI: 10.1111/jfd.13988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 07/01/2024]
Abstract
Melanized focal changes (MFCs) in the fillet of farmed Atlantic salmon is a major quality concern. The changes are thought to initially appear as acute red focal changes (RFCs) that progress into chronic MFCs. Recent findings have indicated that hypoxia may be important in their development, possibly leading to necrosis affecting not only myocytes but also adipocytes. Thus, the aim of this study was to investigate possible hypoxic conditions in RFCs and the subsequent inflammatory responses and lesions in the adipose tissue in RFCs and MFCs. A collection of RFCs, MFCs and control muscle samples from several groups of farmed salmon was studied. Using immunohistochemistry, we found induction of the hypoxia-inducible factor 1 pathway in RFCs. Histological investigations of RFCs and MFCs revealed different stages of fat necrosis, including necrotic adipocytes, a myospherulosis-like reaction and the formation of pseudocystic spaces. Accumulations of foamy macrophages were detected in MFCs, indicating degradation and phagocytosis of lipids. Using in situ hybridization, we showed the presence of tyrosinase- and tyrosinase-related protein-1-expressing amelanotic cells in RFCs, which in turn became melanized in MFCs. In conclusion, we propose a sequence of events leading to the formation of MFCs, highlighting the pivotal role of adiposity, hypoxia and fat necrosis.
Collapse
Affiliation(s)
- Håvard Bjørgen
- Unit of Anatomy, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway
| | - Malin Brimsholm
- Unit of Anatomy, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway
| | | | - Kirstin Skaar
- Institute of Veterinary Anatomy, Vetsuisse Faculty, University of Zurich (UZH), Zurich, Switzerland
| | | | | | | | | | - Tom Hansen
- Matre Research Station, Institute of Marine Research, Matredal, Norway
| | - Espen Rimstad
- Unit of Virology, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway
| | | | | | - Mariusz Pawel Kowalewski
- Institute of Veterinary Anatomy, Vetsuisse Faculty, University of Zurich (UZH), Zurich, Switzerland
| | - Erling Olaf Koppang
- Unit of Anatomy, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway
| |
Collapse
|
33
|
Tian L, Liu Q, Wang X, Chen S, Li Y. Fighting ferroptosis: Protective effects of dexmedetomidine on vital organ injuries. Life Sci 2024; 354:122949. [PMID: 39127318 DOI: 10.1016/j.lfs.2024.122949] [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: 05/08/2024] [Revised: 07/25/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
Vital organ injury is one of the leading causes of global mortality and socio-economic burdens. Current treatments have limited efficacy, and new strategies are needed. Dexmedetomidine (DEX) is a highly selective α2-adrenergic receptor that protects multiple organs by reducing inflammation and preventing cell death. However, its exact mechanism is not yet fully understood. Understanding the underlying molecular mechanisms of its protective effects is crucial as it could provide a basis for designing highly targeted and more effective drugs. Ferroptosis is the primary mode of cell death during organ injury, and recent studies have shown that DEX can protect vital organs from this process. This review provides a detailed analysis of preclinical in vitro and in vivo studies and gains a better understanding of how DEX protects against vital organ injuries by inhibiting ferroptosis. Our findings suggest that DEX can potentially protect vital organs mainly by regulating iron metabolism and the antioxidant defense system. This is the first review that summarizes all evidence of ferroptosis's role in DEX's protective effects against vital organ injuries. Our work aims to provide new insights into organ therapy with DEX and accelerate its translation from the laboratory to clinical settings.
Collapse
Affiliation(s)
- Lei Tian
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Qian Liu
- Department of Anesthesiology, Zigong First People's Hospital, Zigong, China
| | - Xing Wang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Suheng Chen
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Yulan Li
- Department of Anesthesiology, The First Hospital of Lanzhou University, Lanzhou, China.
| |
Collapse
|
34
|
Lee D, Tomita Y, Miwa Y, Kunimi H, Nakai A, Shoda C, Negishi K, Kurihara T. Recent Insights into Roles of Hypoxia-Inducible Factors in Retinal Diseases. Int J Mol Sci 2024; 25:10140. [PMID: 39337623 PMCID: PMC11432567 DOI: 10.3390/ijms251810140] [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: 07/04/2024] [Revised: 09/19/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024] Open
Abstract
Hypoxia-inducible factors (HIFs) are transcriptional factors that function as strong regulators of oxygen homeostasis and cellular metabolisms. The maintenance of cellular oxygen levels is critical as either insufficient or excessive oxygen affects development and physiologic and pathologic conditions. In the eye, retinas have a high metabolic demand for oxygen. Retinal ischemia can cause visual impairment in various sight-threating disorders including age-related macular degeneration, diabetic retinopathy, and some types of glaucoma. Therefore, understanding the potential roles of HIFs in the retina is highly important for managing disease development and progression. This review focuses on the physiologic and pathologic roles of HIFs as regulators of oxygen homeostasis and cellular metabolism in the retina, drawing on recent evidence. Our summary will promote comprehensive approaches to targeting HIFs for therapeutic purposes in retinal diseases.
Collapse
Affiliation(s)
- Deokho Lee
- Laboratory of Photobiology, Keio University School of Medicine, Tokyo 160-8582, Japan
- Laboratory of Chorioretinal Biology, Keio University School of Medicine, Tokyo 160-8582, Japan
- Department of Ophthalmology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Yohei Tomita
- Laboratory of Photobiology, Keio University School of Medicine, Tokyo 160-8582, Japan
- Laboratory of Chorioretinal Biology, Keio University School of Medicine, Tokyo 160-8582, Japan
- Department of Ophthalmology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Yukihiro Miwa
- Laboratory of Photobiology, Keio University School of Medicine, Tokyo 160-8582, Japan
- Department of Ophthalmology, Keio University School of Medicine, Tokyo 160-8582, Japan
- Aichi Animal Eye Clinic, Aichi 464-0027, Japan
| | - Hiromitsu Kunimi
- Laboratory of Photobiology, Keio University School of Medicine, Tokyo 160-8582, Japan
- Department of Ophthalmology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Ayaka Nakai
- Laboratory of Photobiology, Keio University School of Medicine, Tokyo 160-8582, Japan
- Department of Ophthalmology, Keio University School of Medicine, Tokyo 160-8582, Japan
- Department of Ophthalmology, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Chiho Shoda
- Laboratory of Photobiology, Keio University School of Medicine, Tokyo 160-8582, Japan
- Department of Ophthalmology, Keio University School of Medicine, Tokyo 160-8582, Japan
- Department of Ophthalmology, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Kazuno Negishi
- Department of Ophthalmology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Toshihide Kurihara
- Laboratory of Photobiology, Keio University School of Medicine, Tokyo 160-8582, Japan
- Department of Ophthalmology, Keio University School of Medicine, Tokyo 160-8582, Japan
| |
Collapse
|
35
|
Peters K, Staehlke S, Rebl H, Jonitz-Heincke A, Hahn O. Impact of Metal Ions on Cellular Functions: A Focus on Mesenchymal Stem/Stromal Cell Differentiation. Int J Mol Sci 2024; 25:10127. [PMID: 39337612 PMCID: PMC11432215 DOI: 10.3390/ijms251810127] [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: 08/02/2024] [Revised: 09/06/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Metals play a crucial role in the human body, especially as ions in metalloproteins. Essential metals, such as calcium, iron, and zinc are crucial for various physiological functions, but their interactions within biological networks are complex and not fully understood. Mesenchymal stem/stromal cells (MSCs) are essential for tissue regeneration due to their ability to differentiate into various cell types. This review article addresses the effects of physiological and unphysiological, but not directly toxic, metal ion concentrations, particularly concerning MSCs. Overloading or unbalancing of metal ion concentrations can significantly impair the function and differentiation capacity of MSCs. In addition, excessive or unbalanced metal ion concentrations can lead to oxidative stress, which can affect viability or inflammation. Data on the effects of metal ions on MSC differentiation are limited and often contradictory. Future research should, therefore, aim to clarify the mechanisms by which metal ions affect MSC differentiation, focusing on aspects such as metal ion interactions, ion concentrations, exposure duration, and other environmental conditions. Understanding these interactions could ultimately improve the design of biomaterials and implants to promote MSC-mediated tissue regeneration. It could also lead to the development of innovative therapeutic strategies in regenerative medicine.
Collapse
Affiliation(s)
- Kirsten Peters
- Institute of Cell Biology, Rostock University Medical Center Rostock, Schillingallee 69, 18057 Rostock, Germany; (S.S.); (H.R.); (O.H.)
| | - Susanne Staehlke
- Institute of Cell Biology, Rostock University Medical Center Rostock, Schillingallee 69, 18057 Rostock, Germany; (S.S.); (H.R.); (O.H.)
| | - Henrike Rebl
- Institute of Cell Biology, Rostock University Medical Center Rostock, Schillingallee 69, 18057 Rostock, Germany; (S.S.); (H.R.); (O.H.)
| | - Anika Jonitz-Heincke
- Research Laboratory for Biomechanics and Implant Technology, Department of Orthopaedics, Rostock University Medical Center, Doberaner Strasse 142, 18057 Rostock, Germany;
| | - Olga Hahn
- Institute of Cell Biology, Rostock University Medical Center Rostock, Schillingallee 69, 18057 Rostock, Germany; (S.S.); (H.R.); (O.H.)
| |
Collapse
|
36
|
Islam MS, Jesmin. Exploring the Correlation Between Hypoxia, HIF1A Variants, and Breast Cancer in Different Ethnicities, and Bangladeshi Women: Through ELISA and Integrative Multi-Omics Analysis. Biomark Insights 2024; 19:11772719241278176. [PMID: 39314258 PMCID: PMC11418304 DOI: 10.1177/11772719241278176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 08/09/2024] [Indexed: 09/25/2024] Open
Abstract
Background Hypoxia, a condition where there is a lack of oxygen, is known to play a role in cancer progression. Objective This study investigates the correlation between HIF1A gene-altered expression and hypoxia in Bangladeshi breast cancer (BC) cases and TCGA_BC datasets. Design This case-control study compares BC cases to healthy controls to understand the relationship between gene changes and cancer. Method This study used advanced analysis methods to examine the transcriptional landscape of BC, and quantitatively assessed its correlation using integrated multi-omics analysis. Results In Bangladeshi BC cases, the T allele of HIF1A rs1154946 correlates notably (P-value < .001) with BC incidence. ELISA results confirmed a significant association (P-value < .005) between elevated HIF1A expression and BC-related hypoxia. Bioinformatics eQTL analysis validated the correlation between increased HIF1A expression and rs11549465 T allele (P-value < .01). Structural analyses suggested that rs11549465 (P582S) mutation may decrease protein stability (ΔΔG-value: -1.24 kcal/mole), potentially affecting HIF1A function. HIF1A enrichment analysis in BC underscores strong associations with oxygen levels, hypoxia, metabolic processes, apoptosis, and programed cell death (P-value < .001). Transcriptomic data demonstrated a robust correlation (P-value < .0001) between HIF1A expression and copy-number alterations, mutations, and abnormal methylation. Altered HIF1A expression showed strong negative correlations (P-value < .00001) with methylation and the expression of the ER (ESR1), in Whites. Survival analysis revealed marked differences in overall survival linked to high and low HIF1A expression (P-value < .00001). Furthermore, HIF1A expression significantly correlated (P-value < .000001) with hypoxia, TMB, MSI, and immune infiltration by CD8+ T cells, neutrophils, dendritic, and macrophages, providing deeper insights into the BC microenvironment. Conclusion Thus, the HIF1A gene could serve as a promising biomarker for breast cancer progression, control, and survival across ethnicities, emphasizing its role in disease development and regulation.
Collapse
Affiliation(s)
- Md. Shihabul Islam
- Department of Genetic Engineering & Biotechnology, University of Rajshahi, Rajshahi, Bangladesh
| | - Jesmin
- Department of Genetic Engineering & Biotechnology, University of Dhaka, Dhaka, Bangladesh
| |
Collapse
|
37
|
Mirabelli M, Misiti R, Sicilia L, Brunetti FS, Chiefari E, Brunetti A, Foti DP. Hypoxia in Human Obesity: New Insights from Inflammation towards Insulin Resistance-A Narrative Review. Int J Mol Sci 2024; 25:9802. [PMID: 39337290 PMCID: PMC11432683 DOI: 10.3390/ijms25189802] [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: 08/02/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
Insulin resistance (IR), marked by reduced cellular responsiveness to insulin, and obesity, defined by the excessive accumulation of adipose tissue, are two intertwined conditions that significantly contribute to the global burden of cardiometabolic diseases. Adipose tissue, beyond merely storing triglycerides, acts as an active producer of biomolecules. In obesity, as adipose tissue undergoes hypertrophy, it becomes dysfunctional, altering the release of adipocyte-derived factors, known as adipokines. This dysfunction promotes low-grade chronic inflammation, exacerbates IR, and creates a hyperglycemic, proatherogenic, and prothrombotic environment. However, the fundamental cause of these phenomena remains unclear. This narrative review points to hypoxia as a critical trigger for the molecular changes associated with fat accumulation, particularly within visceral adipose tissue (VAT). The activation of hypoxia-inducible factor-1 (HIF-1), a transcription factor that regulates homeostatic responses to low oxygen levels, initiates a series of molecular events in VAT, leading to the aberrant release of adipokines, many of which are still unexplored, and potentially affecting peripheral insulin sensitivity. Recent discoveries have highlighted the role of hypoxia and miRNA-128 in regulating the insulin receptor in visceral adipocytes, contributing to their dysfunctional behavior, including impaired glucose uptake. Understanding the complex interplay between adipose tissue hypoxia, dysfunction, inflammation, and IR in obesity is essential for developing innovative, targeted therapeutic strategies.
Collapse
Affiliation(s)
- Maria Mirabelli
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (M.M.)
- Operative Unit of Endocrinology, “Renato Dulbecco” University Hospital, 88100 Catanzaro, Italy
| | - Roberta Misiti
- Department of Experimental and Clinical Medicine, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy;
- Operative Unit of Clinical Pathology, “Renato Dulbecco” Hospital, 88100 Catanzaro, Italy
| | - Luciana Sicilia
- Operative Unit of Endocrinology, “Renato Dulbecco” University Hospital, 88100 Catanzaro, Italy
| | - Francesco S. Brunetti
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (M.M.)
| | - Eusebio Chiefari
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (M.M.)
- Operative Unit of Endocrinology, “Renato Dulbecco” University Hospital, 88100 Catanzaro, Italy
| | - Antonio Brunetti
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (M.M.)
- Operative Unit of Endocrinology, “Renato Dulbecco” University Hospital, 88100 Catanzaro, Italy
| | - Daniela P. Foti
- Department of Experimental and Clinical Medicine, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy;
- Operative Unit of Clinical Pathology, “Renato Dulbecco” Hospital, 88100 Catanzaro, Italy
| |
Collapse
|
38
|
Mäkinen S, Sree S, Ala-Nisula T, Kultalahti H, Koivunen P, Koistinen HA. Activation of the hypoxia-inducible factor pathway by roxadustat improves glucose metabolism in human primary myotubes from men. Diabetologia 2024; 67:1943-1954. [PMID: 38814443 PMCID: PMC11410918 DOI: 10.1007/s00125-024-06185-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 04/19/2024] [Indexed: 05/31/2024]
Abstract
AIMS/HYPOTHESIS Hypoxia-inducible factor prolyl 4-hydroxylase (HIF-P4H) enzymes regulate adaptive cellular responses to low oxygen concentrations. Inhibition of HIF-P4Hs leads to stabilisation of hypoxia-inducible factors (HIFs) and activation of the HIF pathway affecting multiple biological processes to rescue cells from hypoxia. As evidence from animal models suggests that HIF-P4H inhibitors could be used to treat metabolic disorders associated with insulin resistance, we examined whether roxadustat, an HIF-P4H inhibitor approved for the treatment of renal anaemia, would have an effect on glucose metabolism in primary human myotubes. METHODS Primary skeletal muscle cell cultures, established from biopsies of vastus lateralis muscle from men with normal glucose tolerance (NGT) (n=5) or type 2 diabetes (n=8), were treated with roxadustat. Induction of HIF target gene expression was detected with quantitative real-time PCR. Glucose uptake and glycogen synthesis were investigated with radioactive tracers. Glycolysis and mitochondrial respiration rates were measured with a Seahorse analyser. RESULTS Exposure to roxadustat stabilised nuclear HIF1α protein expression in human myotubes. Treatment with roxadustat led to induction of HIF target gene mRNAs for GLUT1 (also known as SLC2A1), HK2, MCT4 (also known as SLC16A4) and HIF-P4H-2 (also known as PHD2 or EGLN1) in myotubes from donors with NGT, with a blunted response in myotubes from donors with type 2 diabetes. mRNAs for LDHA, PDK1 and GBE1 were induced to a similar degree in myotubes from donors with NGT or type 2 diabetes. Exposure of myotubes to roxadustat led to a 1.4-fold increase in glycolytic rate in myotubes from men with NGT (p=0.0370) and a 1.7-fold increase in myotubes from donors with type 2 diabetes (p=0.0044), with no difference between the groups (p=0.1391). Exposure to roxadustat led to a reduction in basal mitochondrial respiration in both groups (p<0.01). Basal glucose uptake rates were similar in myotubes from donors with NGT (20.2 ± 2.7 pmol mg-1 min-1) and type 2 diabetes (25.3 ± 4.4 pmol mg-1 min-1, p=0.4205). Treatment with roxadustat enhanced insulin-stimulated glucose uptake in myotubes from donors with NGT (1.4-fold vs insulin-only condition, p=0.0023). The basal rate of glucose incorporation into glycogen was lower in myotubes from donors with NGT (233 ± 12.4 nmol g-1 h-1) than in myotubes from donors with type 2 diabetes (360 ± 40.3 nmol g-1 h-1, p=0.0344). Insulin increased glycogen synthesis by 1.9-fold (p=0.0025) in myotubes from donors with NGT, whereas roxadustat did not affect their basal or insulin-stimulated glycogen synthesis. Insulin increased glycogen synthesis by 1.7-fold (p=0.0031) in myotubes from donors with type 2 diabetes. While basal glycogen synthesis was unaffected by roxadustat, pretreatment with roxadustat enhanced insulin-stimulated glycogen synthesis in myotubes from donors with type 2 diabetes (p=0.0345). CONCLUSIONS/INTERPRETATION Roxadustat increases glycolysis and inhibits mitochondrial respiration in primary human myotubes regardless of diabetes status. Roxadustat may also improve insulin action on glycogen synthesis in myotubes from donors with type 2 diabetes.
Collapse
Affiliation(s)
- Selina Mäkinen
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Sreesha Sree
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Tuulia Ala-Nisula
- Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine, Oulu Center for Cell-Matrix Research, University of Oulu, Oulu, Finland
| | - Henric Kultalahti
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Peppi Koivunen
- Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine, Oulu Center for Cell-Matrix Research, University of Oulu, Oulu, Finland
| | - Heikki A Koistinen
- Minerva Foundation Institute for Medical Research, Helsinki, Finland.
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.
| |
Collapse
|
39
|
Capatina AL, Malcolm JR, Stenning J, Moore RL, Bridge KS, Brackenbury WJ, Holding AN. Hypoxia-induced epigenetic regulation of breast cancer progression and the tumour microenvironment. Front Cell Dev Biol 2024; 12:1421629. [PMID: 39282472 PMCID: PMC11392762 DOI: 10.3389/fcell.2024.1421629] [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: 04/22/2024] [Accepted: 08/19/2024] [Indexed: 09/19/2024] Open
Abstract
The events that control breast cancer progression and metastasis are complex and intertwined. Hypoxia plays a key role both in oncogenic transformation and in fueling the metastatic potential of breast cancer cells. Here we review the impact of hypoxia on epigenetic regulation of breast cancer, by interfering with multiple aspects of the tumour microenvironment. The co-dependent relationship between oxygen depletion and metabolic shift to aerobic glycolysis impacts on a range of enzymes and metabolites available in the cell, promoting posttranslational modifications of histones and chromatin, and changing the gene expression landscape to facilitate tumour development. Hormone signalling, particularly through ERα, is also tightly regulated by hypoxic exposure, with HIF-1α expression being a prognostic marker for therapeutic resistance in ER+ breast cancers. This highlights the strong need to understand the hypoxia-endocrine signalling axis and exploit it as a therapeutic target. Furthermore, hypoxia has been shown to enhance metastasis in TNBC cells, as well as promoting resistance to taxanes, radiotherapy and even immunotherapy through microRNA regulation and changes in histone packaging. Finally, several other mediators of the hypoxic response are discussed. We highlight a link between ionic dysregulation and hypoxia signalling, indicating a potential connection between HIF-1α and tumoural Na+ accumulation which would be worth further exploration; we present the role of Ca2+ in mediating hypoxic adaptation via chromatin remodelling, transcription factor recruitment and changes in signalling pathways; and we briefly summarise some of the findings regarding vesicle secretion and paracrine induced epigenetic reprogramming upon hypoxic exposure in breast cancer. By summarising these observations, this article highlights the heterogeneity of breast cancers, presenting a series of pathways with potential for therapeutic applications.
Collapse
Affiliation(s)
| | - Jodie R Malcolm
- Department of Biology, University of York, York, United Kingdom
| | - Jack Stenning
- Department of Biology, University of York, York, United Kingdom
| | - Rachael L Moore
- York Biomedical Research Institute, University of York, York, United Kingdom
| | - Katherine S Bridge
- Department of Biology, University of York, York, United Kingdom
- York Biomedical Research Institute, University of York, York, United Kingdom
| | - William J Brackenbury
- Department of Biology, University of York, York, United Kingdom
- York Biomedical Research Institute, University of York, York, United Kingdom
| | - Andrew N Holding
- Department of Biology, University of York, York, United Kingdom
- York Biomedical Research Institute, University of York, York, United Kingdom
| |
Collapse
|
40
|
Arai Y, Cha R, Nakagawa S, Inoue A, Nakamura K, Takahashi K. Cartilage Homeostasis under Physioxia. Int J Mol Sci 2024; 25:9398. [PMID: 39273346 PMCID: PMC11395513 DOI: 10.3390/ijms25179398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/16/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024] Open
Abstract
Articular cartilage receives nutrients and oxygen from the synovial fluid to maintain homeostasis. However, compared to tissues with abundant blood flow, articular cartilage is exposed to a hypoxic environment (i.e., physioxia) and has an enhanced hypoxic stress response. Hypoxia-inducible factors (HIFs) play a pivotal role in this physioxic environment. In normoxic conditions, HIFs are downregulated, whereas in physioxic conditions, they are upregulated. The HIF-α family comprises three members: HIF-1α, HIF-2α, and HIF-3α. Each member has a distinct function in articular cartilage. In osteoarthritis, which is primarily caused by degeneration of articular cartilage, HIF-1α is upregulated in chondrocytes and is believed to protect articular cartilage by acting anabolically on it. Conversely, in contrast to HIF-1α, HIF-2α exerts a catabolic influence on articular cartilage. It may therefore be possible to develop a new treatment for OA by controlling the expression of HIF-1α and HIF-2α with drugs or by altering the oxygen environment in the joints.
Collapse
Affiliation(s)
- Yuji Arai
- Department of Sports and Para-Sports Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Ryota Cha
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Shuji Nakagawa
- Department of Sports and Para-Sports Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Atsuo Inoue
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Kei Nakamura
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Kenji Takahashi
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| |
Collapse
|
41
|
Ohgaki R, Hirase Y, Xu M, Okanishi H, Kanai Y. LAT1 expression in colorectal cancer cells is unresponsive to HIF-1/2α accumulation under experimental hypoxia. Sci Rep 2024; 14:19635. [PMID: 39179631 PMCID: PMC11343765 DOI: 10.1038/s41598-024-70603-3] [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: 01/26/2024] [Accepted: 08/19/2024] [Indexed: 08/26/2024] Open
Abstract
L-type amino acid transporter 1 (LAT1) is upregulated in various cancer types and contributes to disease progression. Previous studies have demonstrated or suggested that hypoxia-inducible factors (HIFs), the key transcription factors in hypoxic responses, control the expression of LAT1 gene in several types of cancer cells. However, this regulatory relationship has not been investigated yet in colorectal cancer (CRC), one of the cancer types in which the increased LAT1 expression holds prognostic significance. In this study, we found that neither LAT1 mRNA nor protein is induced under hypoxic condition (1% O2) in CRC HT-29 cells in vitro, regardless of the prominent HIF-1/2α accumulation and HIFs-dependent upregulation of glucose transporter 1. The hypoxic treatment generally did not increase either the mRNA or protein expression of LAT1 in eight CRC cell lines tested, in contrast to the pronounced upregulation by amino acid restriction. Interestingly, knockdown of von Hippel-Lindau ubiquitin ligase to inhibit the proteasomal degradation of HIFs caused an accumulation of HIF-2α and increased the LAT1 expression in certain CRC cell lines. This study contributes to delineating the molecular mechanisms responsible for the pathological expression of LAT1 in CRC cells, emphasizing the ambiguity of HIFs-dependent transcriptional upregulation of LAT1 across cancer cells.
Collapse
Affiliation(s)
- Ryuichi Ohgaki
- Department of Bio-System Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Osaka, 565-0871, Japan.
| | - Yuma Hirase
- Department of Bio-System Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Minhui Xu
- Department of Bio-System Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hiroki Okanishi
- Department of Bio-System Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yoshikatsu Kanai
- Department of Bio-System Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Osaka, 565-0871, Japan.
- Department of Metabolic Reprogramming and Signal Regulation, Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
42
|
Putnová I, Putnová BM, Hurník P, Štembírek J, Buchtová M, Kolísková P. Primary cilia-associated signalling in squamous cell carcinoma of head and neck region. Front Oncol 2024; 14:1413255. [PMID: 39234399 PMCID: PMC11372790 DOI: 10.3389/fonc.2024.1413255] [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: 04/06/2024] [Accepted: 07/29/2024] [Indexed: 09/06/2024] Open
Abstract
Squamous cell carcinoma (SCC) of the head and neck originates from the mucosal lining of the upper aerodigestive tract, including the lip, tongue, nasopharynx, oropharynx, larynx and hypopharynx. In this review, we summarise what is currently known about the potential function of primary cilia in the pathogenesis of this disease. As primary cilia represent a key cellular structure for signal transduction and are related to cell proliferation, an understanding of their role in carcinogenesis is necessary for the design of new treatment approaches. Here, we introduce cilia-related signalling in head and neck squamous cell carcinoma (HNSCC) and its possible association with HNSCC tumorigenesis. From this point of view, PDGF, EGF, Wnt and Hh signalling are discussed as all these pathways were found to be dysregulated in HNSCC. Moreover, we review the clinical potential of small molecules affecting primary cilia signalling to target squamous cell carcinoma of the head and neck area.
Collapse
Affiliation(s)
- Iveta Putnová
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czechia
- Department of Anatomy, Histology and Embryology, University of Veterinary Sciences Brno, Brno, Czechia
| | - Barbora Moldovan Putnová
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czechia
- Department of Pathological Morphology and Parasitology, University of Veterinary Sciences Brno, Brno, Czechia
| | - Pavel Hurník
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czechia
- Institute of Molecular and Clinical Pathology and Medical Genetics, University Hospital Ostrava, Ostrava, Czechia
- Institute of Molecular and Clinical Pathology and Medical Genetics, Faculty of Medicine, University of Ostrava, Ostrava, Czechia
| | - Jan Štembírek
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czechia
- Department of Maxillofacial Surgery, University Hospital Ostrava, Ostrava, Czechia
| | - Marcela Buchtová
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czechia
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
| | - Petra Kolísková
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czechia
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
| |
Collapse
|
43
|
Ting KKY. Revisiting the role of hypoxia-inducible factors and nuclear factor erythroid 2-related factor 2 in regulating macrophage inflammation and metabolism. Front Cell Infect Microbiol 2024; 14:1403915. [PMID: 39119289 PMCID: PMC11306205 DOI: 10.3389/fcimb.2024.1403915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 07/11/2024] [Indexed: 08/10/2024] Open
Abstract
The recent birth of the immunometabolism field has comprehensively demonstrated how the rewiring of intracellular metabolism is critical for supporting the effector functions of many immune cell types, such as myeloid cells. Among all, the transcriptional regulation mediated by Hypoxia-Inducible Factors (HIFs) and Nuclear factor erythroid 2-related factor 2 (NRF2) have been consistently shown to play critical roles in regulating the glycolytic metabolism, redox homeostasis and inflammatory responses of macrophages (Mφs). Although both of these transcription factors were first discovered back in the 1990s, new advances in understanding their function and regulations have been continuously made in the context of immunometabolism. Therefore, this review attempts to summarize the traditionally and newly identified functions of these transcription factors, including their roles in orchestrating the key events that take place during glycolytic reprogramming in activated myeloid cells, as well as their roles in mediating Mφ inflammatory responses in various bacterial infection models.
Collapse
Affiliation(s)
- Kenneth K. Y. Ting
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| |
Collapse
|
44
|
Jiao M, Hu M, Pan D, Liu X, Bao X, Kim J, Li F, Li CY. VHL loss enhances antitumor immunity by activating the anti-viral DNA-sensing pathway. iScience 2024; 27:110285. [PMID: 39050705 PMCID: PMC11267025 DOI: 10.1016/j.isci.2024.110285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 05/14/2024] [Accepted: 06/13/2024] [Indexed: 07/27/2024] Open
Abstract
von Hippel-Lindau (VHL), known as a tumor suppressor gene, is frequently mutated in clear cell renal cell carcinoma (ccRCC). However, VHL mutation is not sufficient to promote tumor formation. In most cases other than ccRCC, VHL loss alters cellular homeostasis and causes cell stress and metabolic changes by stabilizing hypoxia-inducible factor (HIF) levels, resulting in a fitness disadvantage. In addition, the function of VHL in regulating immune response is still not well established. In this study, we demonstrate that VHL loss enhances the efficacy of anti-programmed death 1 (PD1) treatment in multiple murine tumor models in a T cell-dependent manner. Mechanistically, we discovered that upregulation of HIF1α/2α induced by VHL loss decreased mitochondrial outer membrane potential and caused the cytoplasmic leakage of mitochondrial DNA, which triggered cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) activation and induced type I interferons. Our study thus provided mechanistic insights into the role of VHL gene loss in boosting antitumor immunity.
Collapse
Affiliation(s)
- Meng Jiao
- Department of Dermatology, Duke University Medical Center, Durham, NC 27710, USA
| | - Mengjie Hu
- Department of Dermatology, Duke University Medical Center, Durham, NC 27710, USA
| | - Dong Pan
- Department of Dermatology, Duke University Medical Center, Durham, NC 27710, USA
| | - Xinjian Liu
- Department of Biochemistry, Molecular Cancer Research Center, School of Medicine, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Xuhui Bao
- Department of Dermatology, Duke University Medical Center, Durham, NC 27710, USA
| | - Jonathan Kim
- School of Medicine, Duke University, Durham, NC 27710, USA
| | - Fang Li
- Department of Dermatology, Duke University Medical Center, Durham, NC 27710, USA
| | - Chuan-Yuan Li
- Department of Dermatology, Duke University Medical Center, Durham, NC 27710, USA
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
45
|
Lee PWT, Suwa T, Kobayashi M, Yang H, Koseki LR, Takeuchi S, Chow CCT, Yasuhara T, Harada H. Hypoxia- and Postirradiation reoxygenation-induced HMHA1/ARHGAP45 expression contributes to cancer cell invasion in a HIF-dependent manner. Br J Cancer 2024; 131:37-48. [PMID: 38740970 PMCID: PMC11231347 DOI: 10.1038/s41416-024-02691-x] [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: 07/19/2023] [Revised: 04/05/2024] [Accepted: 04/10/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Cancer cells in severely hypoxic regions have been reported to invade towards tumour blood vessels after surviving radiotherapy in a postirradiation reoxygenation- and hypoxia-inducible factor (HIF)-dependent manner and cause recurrence. However, how HIF induces invasiveness of irradiated and reoxygenated cancer cells remains unclear. METHODS Here, we identified human minor histocompatibility antigen 1 (HMHA1), which has been suggested to function in cytoskeleton dynamics and cellular motility, as a responsible factor and elucidated its mechanism of action using molecular and cellular biology techniques. RESULTS HMHA1 expression was found to be induced at the transcription initiation level in a HIF-dependent manner under hypoxia. Boyden chamber invasion assay revealed that the induction of HMHA1 expression is required for the increase in invasion of hypoxic cancer cells. Reoxygenation treatment after ionising radiation in vitro that mimics dynamic changes of a microenvironment in hypoxic regions of tumour tissues after radiation therapy further enhanced HMHA1 expression and invasive potential of HMHA1 wildtype cancer cells in ROS- and HIF-dependent manners, but not of HMHA1 knockout cells. CONCLUSION These results together provide insights into a potential molecular mechanism of the acquisition of invasiveness by hypoxic cancer cells after radiotherapy via the activation of the ROS/HIF/HMHA1 axis.
Collapse
Affiliation(s)
- Peter W T Lee
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan
- Department of Genome Repair Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan
| | - Tatsuya Suwa
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan
- Department of Genome Repair Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan
- Department of Oncology, University of Oxford, Oxford, OX3 7DQ, UK
| | - Minoru Kobayashi
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan
- Department of Genome Repair Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan
| | - Hui Yang
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan
- Department of Genome Repair Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan
| | - Lina R Koseki
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan
| | - Satoshi Takeuchi
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan
| | - Christalle C T Chow
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan
| | - Takaaki Yasuhara
- Laboratory of Genome Stress Response, Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan
- Department of Late Effects Studies, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan
| | - Hiroshi Harada
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan.
- Department of Genome Repair Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan.
| |
Collapse
|
46
|
Ortmann BM, Taylor CT, Rocha S. Hypoxia research, where to now? Trends Biochem Sci 2024; 49:573-582. [PMID: 38599898 DOI: 10.1016/j.tibs.2024.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 03/01/2024] [Accepted: 03/18/2024] [Indexed: 04/12/2024]
Abstract
Investigating how cells and organisms sense and respond to O2 levels is essential to our understanding of physiology and pathology. This field has advanced considerably since the discovery of the major transcription factor family, hypoxia-inducible factor (HIF), and the enzymes that control its levels: prolyl hydroxylases (PHDs). However, with its expansion, new complexities have emerged. Herein we highlight three main areas where, in our opinion, the research community could direct some of their attention. These include non-transcriptional roles of HIFs, specificity and O2 sensitivity of 2-oxoglutarate-dependent dioxygenases (2-OGDDs), and new tools and methods to detect O2 concentrations in cells and organs. A greater understanding of these areas would answer big questions and help drive our knowledge of cellular responses to hypoxia forward.
Collapse
Affiliation(s)
- Brian M Ortmann
- Wolfson Childhood Cancer Research Centre, Newcastle University, Newcastle upon Tyne, UK.
| | - Cormac T Taylor
- School of Medicine and Medical Sciences, University College Dublin, Dublin, Ireland.
| | - Sonia Rocha
- Institute of Systems Molecular and Integrative Biology, University of Liverpool, Liverpool, UK.
| |
Collapse
|
47
|
Nishikiori N, Sato T, Ogawa T, Higashide M, Umetsu A, Suzuki S, Furuhashi M, Ohguro H, Watanabe M. TGF-β Isoforms and Local Environments Greatly Modulate Biological Nature of Human Retinal Pigment Epithelium Cells. Bioengineering (Basel) 2024; 11:581. [PMID: 38927817 PMCID: PMC11201039 DOI: 10.3390/bioengineering11060581] [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: 04/30/2024] [Revised: 06/02/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
To characterize transforming growth factor-β (TGF-β) isoform (TGF-β1~3)-b's biological effects on the human retinal pigment epithelium (RPE) under normoxia and hypoxia conditions, ARPE19 cells cultured by 2D (two-dimensional) and 3D (three-dimensional) conditions were subjected to various analyses, including (1) an analysis of barrier function by trans-epithelial electrical resistance (TEER) measurements; (2) qPCR analysis of major ECM molecules including collagen 1 (COL1), COL4, and COL6; α-smooth muscle actin (αSMA); hypoxia-inducible factor 1α (HIF1α); and peroxisome proliferator-activated receptor-gamma coactivator (PGC1α), a master regulator for mitochondrial respiration;, tight junction-related molecules, Zonula occludens-1 (ZO1) and E-cadherin; and vascular endothelial growth factor (VEGF); (3) physical property measurements of 3D spheroids; and (4) cellular metabolic analysis. Diverse effects among TGF-β isoforms were observed, and those effects were also different between normoxia and hypoxia conditions: (1) TGF-β1 and TGF-β3 caused a marked increase in TEER values, and TGF-β2 caused a substantial increase in TEER values under normoxia conditions and hypoxia conditions, respectively; (2) the results of qPCR analysis supported data obtained by TEER; (3) 3D spheroid sizes were decreased by TGF-β isoforms, among which TGF-β1 had the most potent effect under both oxygen conditions; (4) 3D spheroid stiffness was increased by TGF-β2 and TGF-β3 or by TGF-β1 and TGF-β3 under normoxia conditions and hypoxia conditions, respectively; and (5) the TGF-β isoform altered mitochondrial and glycolytic functions differently under oxygen conditions and/or culture conditions. These collective findings indicate that the TGF-β-induced biological effects of 2D and 3D cultures of ARPE19 cells were substantially diverse depending on the three TGF-β isoforms and oxygen levels, suggesting that pathological conditions including epithelial-mesenchymal transition (EMT) of the RPE may be exclusively modulated by both factors.
Collapse
Affiliation(s)
- Nami Nishikiori
- Departments of Ophthalmology, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (N.N.); (M.H.); (A.U.); (S.S.); (H.O.)
| | - Tatsuya Sato
- Departments of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (T.S.); (T.O.); (M.F.)
- Departments of Cellular Physiology and Signal Transduction, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan
| | - Toshifumi Ogawa
- Departments of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (T.S.); (T.O.); (M.F.)
- Departments of Cellular Physiology and Signal Transduction, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan
| | - Megumi Higashide
- Departments of Ophthalmology, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (N.N.); (M.H.); (A.U.); (S.S.); (H.O.)
| | - Araya Umetsu
- Departments of Ophthalmology, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (N.N.); (M.H.); (A.U.); (S.S.); (H.O.)
| | - Soma Suzuki
- Departments of Ophthalmology, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (N.N.); (M.H.); (A.U.); (S.S.); (H.O.)
| | - Masato Furuhashi
- Departments of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (T.S.); (T.O.); (M.F.)
| | - Hiroshi Ohguro
- Departments of Ophthalmology, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (N.N.); (M.H.); (A.U.); (S.S.); (H.O.)
| | - Megumi Watanabe
- Departments of Ophthalmology, Sapporo Medical University, S1W17, Chuo-ku, Sapporo 060-8556, Japan; (N.N.); (M.H.); (A.U.); (S.S.); (H.O.)
| |
Collapse
|
48
|
O'Brien KM, Rix AS, Jasmin A, Lavelle E. The hypoxia response pathway in the Antarctic fish Notothenia coriiceps is functional despite a poly Q/E insertion mutation in HIF-1α. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2024; 50:101218. [PMID: 38412701 PMCID: PMC11128347 DOI: 10.1016/j.cbd.2024.101218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 02/12/2024] [Accepted: 02/12/2024] [Indexed: 02/29/2024]
Abstract
Antarctic notothenioid fishes, inhabiting the oxygen-rich Southern Ocean, possess a polyglutamine and glutamic acid (poly Q/E) insertion mutation in the master transcriptional regulator of oxygen homeostasis, hypoxia- inducible factor-1α (HIF-1α). To determine if this mutation impairs the ability of HIF-1 to regulate gene expression in response to hypoxia, we exposed Notothenia coriiceps, with a poly Q/E insertion mutation in HIF-1α that is 9 amino acids long, to hypoxia (2.3 mg L-1 O2) or normoxia (10 mg L -1 O2) for 12 h. Heart ventricles, brain, liver, and gill tissue were harvested and changes in gene expression quantified using RNA sequencing. Levels of glycogen and lactate were also quantified to determine if anaerobic metabolism increases in response to hypoxia. Exposure to hypoxia resulted in 818 unique differentially expressed genes (DEGs) in liver tissue of N. coriiceps. Many hypoxic genes were induced, including ones involved in the MAP kinase and FoxO pathways, glycolytic metabolism, and vascular remodeling. In contrast, there were fewer than 104 unique DEGs in each of the other tissues sampled. Lactate levels significantly increased in liver in response to hypoxia, indicating that anaerobic metabolism increases in response to hypoxia in this tissue. Overall, our results indicate that the hypoxia response pathway is functional in N. coriiceps despite a poly Q/E mutation in HIF-1α, and confirm that Antarctic fishes are capable of altering gene expression in response to hypoxia.
Collapse
Affiliation(s)
- K M O'Brien
- University of Alaska Fairbanks, Institute of Arctic Biology and Department of Biology & Wildlife, Fairbanks, AK 99775, USA.
| | - A S Rix
- University of Alaska Fairbanks, Institute of Arctic Biology and Department of Biology & Wildlife, Fairbanks, AK 99775, USA.
| | - A Jasmin
- University of Alaska Fairbanks, Institute of Arctic Biology and Department of Biology & Wildlife, Fairbanks, AK 99775, USA
| | - E Lavelle
- National Center for Genome Resources, Santa Fe, NM 87505, USA.
| |
Collapse
|
49
|
Xu H, Zhang J, Zhuang J, Chen Y, Chen L, Wang J, Cao R, Liu F, Wang K, Zhang X, Wang L, Chen G. 2,2- dimethylbenzopyran derivatives containing pyridone structural fragments as selective dual-targeting inhibitors of HIF-1α and EZH2 for the treatment of lung cancer. Bioorg Chem 2024; 147:107419. [PMID: 38703440 DOI: 10.1016/j.bioorg.2024.107419] [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: 01/16/2024] [Revised: 04/16/2024] [Accepted: 04/29/2024] [Indexed: 05/06/2024]
Abstract
We formerly reported that EZH2 inhibitors sensitized HIF-1 inhibitor-resistant cells and inhibited HIF-1α to promote SUZ12 transcription, leading to enhanced EZH2 enzyme activity and elevated H3K27me3 levels, and conversely, inhibition of EZH2 promoted HIF-1α transcription. HIF-1α and EZH2 interacted to form a negative feedback loop that reinforced each other's activity. In this paper, a series of 2,2- dimethylbenzopyran derivatives containing pyridone structural fragments were designed and synthesized with DYB-03, a HIF-1α inhibitor previously reported by our group, and Tazemetostat, an EZH2 inhibitor approved by FDA, as lead compounds. Among these compounds, D-01 had significant inhibitory activities on HIF-1α and EZH2. In vitro experiments showed that D-01 significantly inhibited the migration of A549 cells, clone, invasion and angiogenesis. Moreover, D-01 had good pharmacokinetic profiles. All the results about compound D-01 could lay a foundation for the research and development of HIF-1α and EZH2 dual-targeting compounds.
Collapse
Affiliation(s)
- Huashen Xu
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Jie Zhang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, PR China; Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Benxi 117004, PR China
| | - Junning Zhuang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Yuanguang Chen
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Lu Chen
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Jianmin Wang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, PR China; Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Benxi 117004, PR China
| | - Ruolin Cao
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Fuqin Liu
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Kaibo Wang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Xiaoyu Zhang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Lihui Wang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, PR China; Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Benxi 117004, PR China.
| | - Guoliang Chen
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China.
| |
Collapse
|
50
|
Duan LJ, Jiang Y, Fong GH. Endothelial HIF2α suppresses retinal angiogenesis in neonatal mice by upregulating NOTCH signaling. Development 2024; 151:dev202802. [PMID: 38770916 PMCID: PMC11190433 DOI: 10.1242/dev.202802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 05/01/2024] [Indexed: 05/22/2024]
Abstract
Prolyl hydroxylase domain (PHD) proteins are oxygen sensors that use intracellular oxygen as a substrate to hydroxylate hypoxia-inducible factor (HIF) α proteins, routing them for polyubiquitylation and proteasomal degradation. Typically, HIFα accumulation in hypoxic or PHD-deficient tissues leads to upregulated angiogenesis. Here, we report unexpected retinal phenotypes associated with endothelial cell (EC)-specific gene targeting of Phd2 (Egln1) and Hif2alpha (Epas1). EC-specific Phd2 disruption suppressed retinal angiogenesis, despite HIFα accumulation and VEGFA upregulation. Suppressed retinal angiogenesis was observed both in development and in the oxygen-induced retinopathy (OIR) model. On the other hand, EC-specific deletion of Hif1alpha (Hif1a), Hif2alpha, or both did not affect retinal vascular morphogenesis. Strikingly, retinal angiogenesis appeared normal in mice double-deficient for endothelial PHD2 and HIF2α. In PHD2-deficient retinal vasculature, delta-like 4 (DLL4, a NOTCH ligand) and HEY2 (a NOTCH target) were upregulated by HIF2α-dependent mechanisms. Inhibition of NOTCH signaling by a chemical inhibitor or DLL4 antibody partially rescued retinal angiogenesis. Taken together, our data demonstrate that HIF2α accumulation in retinal ECs inhibits rather than stimulates retinal angiogenesis, in part by upregulating DLL4 expression and NOTCH signaling.
Collapse
Affiliation(s)
- Li-Juan Duan
- Center for Vascular Biology, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Yida Jiang
- Center for Vascular Biology, University of Connecticut Health Center, Farmington, CT 06030, USA
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Guo-Hua Fong
- Center for Vascular Biology, University of Connecticut Health Center, Farmington, CT 06030, USA
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT 06030, USA
| |
Collapse
|