1
|
Wang X, Wu Q, Zhong M, Chen Y, Wang Y, Li X, Zhao W, Ge C, Wang X, Yu Y, Yang S, Wang T, Xie E, Shi W, Min J, Wang F. Adipocyte-derived ferroptotic signaling mitigates obesity. Cell Metab 2024:S1550-4131(24)00456-X. [PMID: 39729998 DOI: 10.1016/j.cmet.2024.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 09/29/2024] [Accepted: 11/12/2024] [Indexed: 12/29/2024]
Abstract
Ferroptosis is characterized as an iron-dependent and lipophilic form of cell death. However, it remains unclear what role ferroptosis has in adipose tissue function and activity. Here, we find a lower ferroptotic signature in the adipose tissue of individuals and mice with obesity. We further find that activation of ferroptotic signaling by a non-lethal dose of ferroptosis agonists significantly reduces lipid accumulation in primary adipocytes and high-fat diet (HFD)-fed mice. Notably, adipocyte-specific overexpression of acyl-coenzyme A synthetase long-chain family member 4 (Acsl4) or deletion of ferritin heavy chain (Fth) protects mice from HFD-induced adipose expansion and metabolic disorders via activation of ferroptotic signaling. Mechanistically, we find that 5,15-dihydroxyeicosatetraenoic acid (5,15-DiHETE) activates ferroptotic signaling, resulting in the degradation of hypoxia-inducible factor-1α (HIF1α), thereby derepressing a thermogenic program regulated by the c-Myc-peroxisome proliferator-activated receptor gamma coactivator-1 beta (Pgc1β) pathway. Our findings suggest that activating ferroptosis signaling in adipose tissues might help to prevent and treat obesity and its related metabolic disorders.
Collapse
Affiliation(s)
- Xue Wang
- The Second Affiliated Hospital, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou 310058, China; The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; School of Public Health, Basic Medical Sciences, School of Pharmacology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China; School of Public Health, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China
| | - Qian Wu
- International Institutes of Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Meijuan Zhong
- School of Public Health, Basic Medical Sciences, School of Pharmacology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Ying Chen
- Ministry of Education Key Laboratory of Metabolism and Molecular Medicine, Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yudi Wang
- The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xin Li
- School of Public Health, Basic Medical Sciences, School of Pharmacology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Wenxi Zhao
- The Second Affiliated Hospital, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Chaodong Ge
- The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xinhui Wang
- The Second Affiliated Hospital, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yingying Yu
- The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Sisi Yang
- The Second Affiliated Hospital, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Tianyi Wang
- School of Public Health, Basic Medical Sciences, School of Pharmacology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Enjun Xie
- The Second Affiliated Hospital, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Wanting Shi
- The Second Affiliated Hospital, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Junxia Min
- The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China.
| | - Fudi Wang
- The Second Affiliated Hospital, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou 310058, China; School of Public Health, Basic Medical Sciences, School of Pharmacology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China; School of Public Health, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China.
| |
Collapse
|
2
|
Brendel H, Mittag J, Hofmann A, Hempel H, Giebe S, Diaba-Nuhoho P, Wolk S, Reeps C, Morawietz H, Brunssen C. NADPH Oxidase 4: Crucial for Endothelial Function under Hypoxia-Complementing Prostacyclin. Antioxidants (Basel) 2024; 13:1178. [PMID: 39456432 PMCID: PMC11504732 DOI: 10.3390/antiox13101178] [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: 07/29/2024] [Revised: 09/23/2024] [Accepted: 09/26/2024] [Indexed: 10/28/2024] Open
Abstract
Aim: The primary endothelial NADPH oxidase isoform 4 (NOX4) is notably induced during hypoxia, with emerging evidence suggesting its vasoprotective role through H2O2 production. Therefore, we aimed to elucidate NOX4's significance in endothelial function under hypoxia. Methods: Human vessels, in addition to murine vessels from Nox4-/- mice, were explored. On a functional level, Mulvany myograph experiments were performed. To obtain mechanistical insights, human endothelial cells were cultured under hypoxia with inhibitors of hypoxia-inducible factors. Additionally, endothelial cells were cultured under combined hypoxia and laminar shear stress conditions. Results: In human occluded vessels, NOX4 expression strongly correlated with prostaglandin I2 synthase (PTGIS). Hypoxia significantly elevated NOX4 and PTGIS expression and activity in human endothelial cells. Inhibition of prolyl hydroxylase domain (PHD) enzymes, which stabilize hypoxia-inducible factors (HIFs), increased NOX4 and PTGIS expression even under normoxic conditions. NOX4 mRNA expression was reduced by HIF1a inhibition, while PTGIS mRNA expression was only affected by the inhibition of HIF2a under hypoxia. Endothelial function assessments revealed hypoxia-induced endothelial dysfunction in mesenteric arteries from wild-type mice. Mesenteric arteries from Nox4-/- mice exhibited an altered endothelial function under hypoxia, most prominent in the presence of cyclooxygenase inhibitor diclofenac to exclude the impact of prostacyclin. Restored protective laminar shear stress, as it might occur after thrombolysis, angioplasty, or stenting, attenuated the hypoxic response in endothelial cells, reducing HIF1a expression and its target NOX4 while enhancing eNOS expression. Conclusions: Hypoxia strongly induces NOX4 and PTGIS, with a close correlation between both factors in occluded, hypoxic human vessels. This relationship ensured endothelium-dependent vasodilation under hypoxic conditions. Protective laminar blood flow restores eNOS expression and mitigates the hypoxic response on NOX4 and PTGIS.
Collapse
Affiliation(s)
- Heike Brendel
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, 01307 Dresden, Germany; (J.M.); (H.H.); (S.G.); (P.D.-N.); (C.B.)
| | - Jennifer Mittag
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, 01307 Dresden, Germany; (J.M.); (H.H.); (S.G.); (P.D.-N.); (C.B.)
| | - Anja Hofmann
- Division of Vascular and Endovascular Surgery, Department of Visceral, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, 01307 Dresden, Germany; (A.H.); (S.W.); (C.R.)
| | - Helene Hempel
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, 01307 Dresden, Germany; (J.M.); (H.H.); (S.G.); (P.D.-N.); (C.B.)
| | - Sindy Giebe
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, 01307 Dresden, Germany; (J.M.); (H.H.); (S.G.); (P.D.-N.); (C.B.)
| | - Patrick Diaba-Nuhoho
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, 01307 Dresden, Germany; (J.M.); (H.H.); (S.G.); (P.D.-N.); (C.B.)
- Department of Paediatric and Adolescent Medicine, Paediatric Haematology and Oncology, University Hospital Münster, 48149 Münster, Germany
| | - Steffen Wolk
- Division of Vascular and Endovascular Surgery, Department of Visceral, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, 01307 Dresden, Germany; (A.H.); (S.W.); (C.R.)
| | - Christian Reeps
- Division of Vascular and Endovascular Surgery, Department of Visceral, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, 01307 Dresden, Germany; (A.H.); (S.W.); (C.R.)
| | - Henning Morawietz
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, 01307 Dresden, Germany; (J.M.); (H.H.); (S.G.); (P.D.-N.); (C.B.)
| | - Coy Brunssen
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, 01307 Dresden, Germany; (J.M.); (H.H.); (S.G.); (P.D.-N.); (C.B.)
| |
Collapse
|
3
|
Germeys C, Vandoorne T, Davie K, Poovathingal S, Heeren K, Vermeire W, Nami F, Moisse M, Quaegebeur A, Sierksma A, Rué L, Sicart A, Eykens C, De Cock L, De Strooper B, Carmeliet P, Van Damme P, De Bock K, Van Den Bosch L. Targeting EGLN2/PHD1 protects motor neurons and normalizes the astrocytic interferon response. Cell Rep 2024; 43:114719. [PMID: 39255062 DOI: 10.1016/j.celrep.2024.114719] [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: 03/07/2024] [Revised: 07/08/2024] [Accepted: 08/20/2024] [Indexed: 09/12/2024] Open
Abstract
Neuroinflammation and dysregulated energy metabolism are linked to motor neuron degeneration in amyotrophic lateral sclerosis (ALS). The egl-9 family hypoxia-inducible factor (EGLN) enzymes, also known as prolyl hydroxylase domain (PHD) enzymes, are metabolic sensors regulating cellular inflammation and metabolism. Using an oligonucleotide-based and a genetic approach, we showed that the downregulation of Egln2 protected motor neurons and mitigated the ALS phenotype in two zebrafish models and a mouse model of ALS. Single-nucleus RNA sequencing of the murine spinal cord revealed that the loss of EGLN2 induced an astrocyte-specific downregulation of interferon-stimulated genes, mediated via the stimulator of interferon genes (STING) protein. In addition, we found that the genetic deletion of EGLN2 restored this interferon response in patient induced pluripotent stem cell (iPSC)-derived astrocytes, confirming the link between EGLN2 and astrocytic interferon signaling. In conclusion, we identified EGLN2 as a motor neuron protective target normalizing the astrocytic interferon-dependent inflammatory axis in vivo, as well as in patient-derived cells.
Collapse
Affiliation(s)
- Christine Germeys
- KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), 3000 Leuven, Belgium; VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, 3000 Leuven, Belgium
| | - Tijs Vandoorne
- KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), 3000 Leuven, Belgium; VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, 3000 Leuven, Belgium
| | - Kristofer Davie
- VIB-KU Leuven, Center for Brain & Disease Research Technologies, Single Cell Bioinformatics Unit, 3000 Leuven, Belgium
| | - Suresh Poovathingal
- VIB-KU Leuven, Center for Brain & Disease Research Technologies, Single Cell Microfluidics & Analytics Unit, 3000 Leuven, Belgium; VIB, Center for AI & Computational Biology (VIB.AI), 3000 Leuven, Belgium
| | - Kara Heeren
- KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), 3000 Leuven, Belgium; VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, 3000 Leuven, Belgium
| | - Wendy Vermeire
- KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), 3000 Leuven, Belgium; VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, 3000 Leuven, Belgium
| | - FatemehArefeh Nami
- KU Leuven - University of Leuven, Department of Development and Regeneration, Stem Cell Institute Leuven (SCIL), 3000 Leuven, Belgium
| | - Matthieu Moisse
- KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), 3000 Leuven, Belgium; VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, 3000 Leuven, Belgium
| | - Annelies Quaegebeur
- University of Cambridge, Department of Clinical Neurosciences, CB2 2PY Cambridge, UK; Cambridge University Hospitals, Department of Histopathology, CB2 0QQ Cambridge, UK
| | - Annerieke Sierksma
- KU Leuven - University of Leuven, Department of Neurosciences, Research Group Molecular Neurobiology and Leuven Brain Institute (LBI), 3000 Leuven, Belgium; VIB, Center for Brain & Disease Research, Laboratory for the Research of Neurodegenerative Diseases, 3000 Leuven, Belgium
| | - Laura Rué
- KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), 3000 Leuven, Belgium; VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, 3000 Leuven, Belgium
| | - Adrià Sicart
- KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), 3000 Leuven, Belgium; VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, 3000 Leuven, Belgium
| | - Caroline Eykens
- KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), 3000 Leuven, Belgium; VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, 3000 Leuven, Belgium
| | - Lenja De Cock
- KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), 3000 Leuven, Belgium; VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, 3000 Leuven, Belgium
| | - Bart De Strooper
- KU Leuven - University of Leuven, Department of Neurosciences, Research Group Molecular Neurobiology and Leuven Brain Institute (LBI), 3000 Leuven, Belgium; VIB, Center for Brain & Disease Research, Laboratory for the Research of Neurodegenerative Diseases, 3000 Leuven, Belgium; Dementia Research Institute, University College London, WC1E 6BT London, UK
| | - Peter Carmeliet
- KU Leuven - University of Leuven, Department of Oncology and Leuven Cancer Institute (LKI), Laboratory of Angiogenesis and Vascular Metabolism, 3000 Leuven, Belgium; VIB, Center for Cancer Biology, Laboratory of Angiogenesis and Vascular Metabolism, 3000 Leuven, Belgium; Khalifa University of Science and Technology, Center for Biotechnology, Abu Dhabi, United Arab Emirates
| | - Philip Van Damme
- KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), 3000 Leuven, Belgium; VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, 3000 Leuven, Belgium; University Hospitals Leuven, Department of Neurology, 3000 Leuven, Belgium
| | - Katrien De Bock
- ETH Zürich, Department of Health Sciences and Technology, 8092 Zürich, Switzerland
| | - Ludo Van Den Bosch
- KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), 3000 Leuven, Belgium; VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, 3000 Leuven, Belgium.
| |
Collapse
|
4
|
Yu G, Yuan L, Li X, Zuo M, Wang R, Chen M, Liu Y, Liu X, Xiao W. Zebrafish phd1 enhances mavs-mediated antiviral responses in a hydroxylation-independent manner. J Virol 2024; 98:e0103824. [PMID: 39162481 PMCID: PMC11406971 DOI: 10.1128/jvi.01038-24] [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: 06/14/2024] [Accepted: 07/08/2024] [Indexed: 08/21/2024] Open
Abstract
PHD1 is a member of the prolyl hydroxylase domain protein (PHD1-4) family, which plays a prominent role in the post-translational modification of its target proteins by hydroxylating proline residues. The best-characterized targets of PHD1 are hypoxia-inducible factor α (HIF-1α and HIF-2α), two master regulators of the hypoxia signaling pathway. In this study, we show that zebrafish phd1 positively regulates mavs-mediated antiviral innate immunity. Overexpression of phd1 enhances the cellular antiviral response. Consistently, zebrafish lacking phd1 are more susceptible to spring viremia of carp virus infection. Further assays indicate that phd1 interacts with mavs through the C-terminal transmembrane domain of mavs and promotes mavs aggregation. In addition, zebrafish phd1 attenuates K48-linked polyubiquitination of mavs, leading to stabilization of mavs. However, the enzymatic activity of phd1 is not required for phd1 to activate mavs. In conclusion, this study reveals a novel function of phd1 in the regulation of antiviral innate immunity.IMPORTANCEPHD1 is a key regulator of the hypoxia signaling pathway, but its role in antiviral innate immunity is largely unknown. In this study, we found that zebrafish phd1 enhances cellular antiviral responses in a hydroxylation-independent manner. Phd1 interacts with mavs through the C-terminal transmembrane domain of mavs and promotes mavs aggregation. In addition, phd1 attenuates K48-linked polyubiquitination of mavs, leading to stabilization of mavs. Zebrafish lacking phd1 are more susceptible to spring viremia of carp virus infection. These findings reveal a novel role for phd1 in the regulation of mavs-mediated antiviral innate immunity.
Collapse
Affiliation(s)
- Guangqing Yu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, People's Republic of China
| | - Le Yuan
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Chinese Academy of Sciences, Wuhan, People's Republic of China
| | - Xiong Li
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Chinese Academy of Sciences, Wuhan, People's Republic of China
| | - Mingzhong Zuo
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, People's Republic of China
| | - Rui Wang
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Chinese Academy of Sciences, Wuhan, People's Republic of China
| | - Mengjuan Chen
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, People's Republic of China
| | - Yuqing Liu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, People's Republic of China
| | - Xing Liu
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Chinese Academy of Sciences, Wuhan, People's Republic of China
- Hubei Hongshan Laboratory, Wuhan, People's Republic of China
- The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan, People's Republic of China
| | - Wuhan Xiao
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Chinese Academy of Sciences, Wuhan, People's Republic of China
- Hubei Hongshan Laboratory, Wuhan, People's Republic of China
- The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, People's Republic of China
| |
Collapse
|
5
|
Xie J, Zhang Z. Recent Advances and Therapeutic Implications of 2-Oxoglutarate-Dependent Dioxygenases in Ischemic Stroke. Mol Neurobiol 2024; 61:3949-3975. [PMID: 38041714 DOI: 10.1007/s12035-023-03790-1] [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: 08/04/2023] [Accepted: 11/08/2023] [Indexed: 12/03/2023]
Abstract
Ischemic stroke is a common disease with a high disability rate and mortality, which brings heavy pressure on families and medical insurance. Nowadays, the golden treatments for ischemic stroke in the acute phase mainly include endovascular therapy and intravenous thrombolysis. Some drugs are used to alleviate brain injury in patients with ischemic stroke, such as edaravone and 3-n-butylphthalide. However, no effective neuroprotective drug for ischemic stroke has been acknowledged. 2-Oxoglutarate-dependent dioxygenases (2OGDDs) are conserved and common dioxygenases whose activities depend on O2, Fe2+, and 2OG. Most 2OGDDs are expressed in the brain and are essential for the development and functions of the brain. Therefore, 2OGDDs likely play essential roles in ischemic brain injury. In this review, we briefly elucidate the functions of most 2OGDDs, particularly the effects of regulations of 2OGDDs on various cells in different phases after ischemic stroke. It would also provide promising potential therapeutic targets and directions of drug development for protecting the brain against ischemic injury and improving outcomes of ischemic stroke.
Collapse
Affiliation(s)
- Jian Xie
- Department of Neurology, Affiliated Zhongda Hospital, Research Institution of Neuropsychiatry, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Zhijun Zhang
- Department of Neurology, Affiliated Zhongda Hospital, Research Institution of Neuropsychiatry, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China.
- Shenzhen Key Laboratory of Precision Diagnosis and Treatment of Depression, Department of Mental Health and Public Health, Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, Guangdong, China.
| |
Collapse
|
6
|
Deng H, Wang Z, Zhu C, Chen Z. Prolyl hydroxylase domain enzyme PHD2 inhibits proliferation and metabolism in non-small cell lung cancer cells in HIF-dependent and HIF-independent manners. Front Oncol 2024; 14:1370393. [PMID: 39007099 PMCID: PMC11240288 DOI: 10.3389/fonc.2024.1370393] [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: 01/14/2024] [Accepted: 06/14/2024] [Indexed: 07/16/2024] Open
Abstract
Prolyl hydroxylase domain protein 2 (PHD2) is one of the intracellular oxygen sensors that mediates proteasomal degradation of hypoxia-inducible factor (HIF)-α via hydroxylation under normoxic conditions. Because of its canonical function in the hypoxia signaling pathway, PHD2 is generally regarded as a tumor suppressor. However, the effects of PHD2 in tumorigenesis are not entirely dependent on HIF-α. Based on analysis of data from the Cancer Genome Atlas (TCGA) database, we observed that the expression of PHD2 is upregulated in non-small cell lung cancer (NSCLC), which accounts for approximately 80-85% of lung cancers. This suggests that PHD2 may play an important role in NSCLC. However, the function of PHD2 in NSCLC remains largely unknown. In this study, we established PHD2-deficient H1299 cells and PHD2-knockdown A549 cells to investigate the function of PHD2 in NSCLC and found that PHD2 suppresses cell proliferation and metabolism but induces ROS levels in human NSCLC cells. Further results indicated that the function of PHD2 in NSCLC is dependent on its enzymatic activity and partially independent of HIF. Moreover, we performed RNA-sequencing and transcriptomic analysis to explore the underlying mechanisms and identified some potential targets and pathways regulated by PHD2, apart from the canonical HIF-mediated hypoxia signaling pathway. These results provide some clues to uncover novel roles of PHD2 in lung cancer progression.
Collapse
Affiliation(s)
- Hongyan Deng
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- College of Life Science, Wuhan University, Wuhan, China
| | - Zixuan Wang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Chunchun Zhu
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Zhu Chen
- Department of Reproduction, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
7
|
Ayub A, Hasan MK, Mahmud Z, Hossain MS, Kabir Y. Dissecting the multifaceted roles of autophagy in cancer initiation, growth, and metastasis: from molecular mechanisms to therapeutic applications. Med Oncol 2024; 41:183. [PMID: 38902544 DOI: 10.1007/s12032-024-02417-2] [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: 04/12/2024] [Accepted: 05/28/2024] [Indexed: 06/22/2024]
Abstract
Autophagy is a cytoplasmic defense mechanism that cells use to break and reprocess their intracellular components. This utilization of autophagy is regarded as a savior in nutrient-deficient and other stressful conditions. Hence, autophagy keeps contact with and responds to miscellaneous cellular tensions and diverse pathways of signal transductions, such as growth signaling and cellular death. Importantly, autophagy is regarded as an effective tumor suppressor because regular autophagic breakdown is essential for cellular maintenance and minimizing cellular damage. However, paradoxically, autophagy has also been observed to promote the events of malignancies. This review discussed the dual role of autophagy in cancer, emphasizing its influence on tumor survival and progression. Possessing such a dual contribution to the malignant establishment, the prevention of autophagy can potentially advocate for the advancement of malignant transformation. In contrast, for the context of the instituted tumor, the agents of preventing autophagy potently inhibit the advancement of the tumor. Key regulators, including calpain 1, mTORC1, and AMPK, modulate autophagy in response to nutritional conditions and stress. Oncogenic mutations like RAS and B-RAF underscore autophagy's pivotal role in cancer development. The review also delves into autophagy's context-dependent roles in tumorigenesis, metastasis, and the tumor microenvironment (TME). It also discusses the therapeutic effectiveness of autophagy for several cancers. The recent implication of autophagy in the control of both innate and antibody-mediated immune systems made it a center of attention to evaluating its role concerning tumor antigens and treatments of cancer.
Collapse
Affiliation(s)
- Afia Ayub
- Department of Biochemistry and Molecular Biology, Tejgaon College, National University, Gazipur, 1704, Bangladesh
| | - Md Kamrul Hasan
- Department of Biochemistry and Molecular Biology, Tejgaon College, National University, Gazipur, 1704, Bangladesh.
- Department of Health Research Methods, Evidence, and Impact, McMaster University, 1280 Main St. W., Hamilton, L8S 4K1, Canada.
- Department of Public Health, North South University, Dhaka, Bangladesh.
| | - Zimam Mahmud
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, 1000, Bangladesh.
| | - Md Sabbir Hossain
- Department of Biochemistry and Molecular Biology, Tejgaon College, National University, Gazipur, 1704, Bangladesh
| | - Yearul Kabir
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, 1000, Bangladesh.
| |
Collapse
|
8
|
Laird M, Ku JC, Raiten J, Sriram S, Moore M, Li Y. Mitochondrial metabolism regulation and epigenetics in hypoxia. Front Physiol 2024; 15:1393232. [PMID: 38915781 PMCID: PMC11194441 DOI: 10.3389/fphys.2024.1393232] [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: 02/28/2024] [Accepted: 05/13/2024] [Indexed: 06/26/2024] Open
Abstract
The complex and dynamic interaction between cellular energy control and gene expression modulation is shown by the intersection between mitochondrial metabolism and epigenetics in hypoxic environments. Poor oxygen delivery to tissues, or hypoxia, is a basic physiological stressor that sets off a series of reactions in cells to adapt and endure oxygen-starved environments. Often called the "powerhouse of the cell," mitochondria are essential to cellular metabolism, especially regarding producing energy through oxidative phosphorylation. The cellular response to hypoxia entails a change in mitochondrial metabolism to improve survival, including epigenetic modifications that control gene expression without altering the underlying genome. By altering the expression of genes involved in angiogenesis, cell survival, and metabolism, these epigenetic modifications help cells adapt to hypoxia. The sophisticated interplay between mitochondrial metabolism and epigenetics in hypoxia is highlighted by several important points, which have been summarized in the current article. Deciphering the relationship between mitochondrial metabolism and epigenetics during hypoxia is essential to understanding the molecular processes that regulate cellular adaptation to reduced oxygen concentrations.
Collapse
Affiliation(s)
- Madison Laird
- Western Michigan University Homer Stryker School of Medicine, Kalamazoo, MI, United States
| | - Jennifer C. Ku
- Western Michigan University Homer Stryker School of Medicine, Kalamazoo, MI, United States
| | - Jacob Raiten
- Western Michigan University Homer Stryker School of Medicine, Kalamazoo, MI, United States
| | - Sashwat Sriram
- Western Michigan University Homer Stryker School of Medicine, Kalamazoo, MI, United States
| | - Megan Moore
- Western Michigan University Homer Stryker School of Medicine, Kalamazoo, MI, United States
| | - Yong Li
- Department of Orthopaedic Surgery, Biomedical Engineering, Western Michigan University Homer Stryker School of Medicine, Kalamazoo, MI, United States
| |
Collapse
|
9
|
Wu Z, Wang Y, Gao R, Chen J, Chen Y, Li M, Gao Y. Potential therapeutic effects of traditional Chinese medicine in acute mountain sickness: pathogenesis, mechanisms and future directions. Front Pharmacol 2024; 15:1393209. [PMID: 38895636 PMCID: PMC11183292 DOI: 10.3389/fphar.2024.1393209] [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: 02/28/2024] [Accepted: 05/06/2024] [Indexed: 06/21/2024] Open
Abstract
Background and objectives Acute mountain sickness (AMS) is a pathology with different symptoms in which the organism is not adapted to the environment that occurs under the special environment of high altitude. Its main mechanism is the organism's tissue damage caused by acute hypobaric hypoxia. Traditional Chinese medicine (TCM) theory focuses on the holistic concept. TCM has made remarkable achievements in the treatment of many mountain sicknesses. This review outlines the pathogenesis of AMS in modern and traditional medicine, the progress of animal models of AMS, and summarizes the therapeutic effects of TCM on AMS. Methods Using the keywords "traditional Chinese medicine," "herbal medicine," "acute mountain sickness," "high-altitude pulmonary edema," "high-altitude cerebral edema," "acute hypobaric hypoxia," and "high-altitude," all relevant TCM literature published up to November 2023 were collected from Scopus, Web of Science, PubMed, and China National Knowledge Infrastructure databases, and the key information was analyzed. Results We systematically summarised the effects of acute hypobaric hypoxia on the tissues of the organism, the study of the methodology for the establishment of an animal model of AMS, and retrieved 18 proprietary Chinese medicines for the clinical treatment of AMS. The therapeutic principle of medicines is mainly invigorating qi, activating blood and removing stasis. The components of botanical drugs mainly include salidroside, ginsenoside Rg1, and tetrahydrocurcumin. The mechanism of action of TCM in the treatment of AMS is mainly through the regulation of HIF-1α/NF-κB signaling pathway, inhibition of inflammatory response and oxidative stress, and enhancement of energy metabolism. Conclusion The main pathogenesis of AMS is unclear. Still, TCM formulas and components have been used to treat AMS through multifaceted interventions, such as compound danshen drip pills, Huangqi Baihe granules, salidroside, and ginsenoside Rg1. These components generally exert anti-AMS pharmacological effects by inhibiting the expression of VEGF, concentration of MDA and pro-inflammatory factors, down-regulating NF-κB/NLRP3 pathway, and promoting SOD and Na + -K + -ATPase activities, which attenuates acute hypobaric hypoxia-induced tissue injury. This review comprehensively analyses the application of TCM in AMS and makes suggestions for more in-depth studies in the future, aiming to provide some ideas and insights for subsequent studies.
Collapse
Affiliation(s)
- Zhenhui Wu
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, China
- Department of Hematology, Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, China
| | - Yihao Wang
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Rong Gao
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Junru Chen
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Yingfan Chen
- Department of Traditional Chinese Medicine, The Sixth Medical Center of Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Maoxing Li
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Yue Gao
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, China
- Beijing Institute of Radiation Medicine, Beijing, China
| |
Collapse
|
10
|
Zhang YY, Jin PP, Guo DZ, Bian D. Modified Zhenwu Tang delays chronic renal failure progression by modulating oxidative stress and hypoxic responses in renal proximal tubular epithelial cells. Heliyon 2024; 10:e31265. [PMID: 38803876 PMCID: PMC11128522 DOI: 10.1016/j.heliyon.2024.e31265] [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: 11/14/2023] [Revised: 05/13/2024] [Accepted: 05/14/2024] [Indexed: 05/29/2024] Open
Abstract
Background Tubulointerstitial fibrosis (TIF) is a critical pathological feature of chronic renal failure (CRF), with oxidative stress (OS) and hypoxic responses in renal proximal tubular epithelial cells playing pivotal roles in disease progression. This study explores the effects of Modified Zhenwu Tang (MZWT) on these processes, aiming to uncover its potential mechanisms in slowing CRF progression. Methods We used adenine (Ade) to induce CRF in rats, which were then treated with benazepril hydrochloride (Lotensin) and MZWT for 8 weeks. Assessments included liver and renal function, electrolytes, blood lipids, renal tissue pathology, OS levels, the hypoxia-inducible factor (HIF) pathway, inflammatory markers, and other relevant indicators. In vitro, human renal cortical proximal tubular epithelial cells were subjected to hypoxia and lipopolysaccharide for 72 h, with concurrent treatment using MZWT, FM19G11, and N-acetyl-l-cysteine. Measurements taken included reactive oxygen species (ROS), HIF pathway activity, inflammatory markers, and other relevant indicators. Results Ade treatment induced significant disruptions in renal function, blood lipids, electrolytes, and tubulointerstitial architecture, alongside heightened OS, HIF pathway activation, and inflammatory responses in rats. In vivo, MZWT effectively ameliorated proteinuria, renal dysfunction, lipid and electrolyte imbalances, and renal tissue damage; it also suppressed OS, HIF pathway activation, epithelial-mesenchymal transition (EMT) in proximal tubular epithelial cells, and reduced the production of inflammatory cytokines and collagen fibers. In vitro findings demonstrated that MZWT decreased apoptosis, reduced ROS production, curbed OS, HIF pathway activation, and EMT in proximal tubular epithelial cells, and diminished the output of inflammatory cytokines and collagen. Conclusion OS and hypoxic responses significantly contribute to TIF development. MZWT mitigates these responses in renal proximal tubular epithelial cells, thereby delaying the progression of CRF.
Collapse
Affiliation(s)
- Yuan-yuan Zhang
- Graduate School, Hebei University of Chinese Medicine, Hebei, Shijiazhuang, 050000, China
| | - Pei-pei Jin
- Hebei Yiling Hospital, Hebei, Shijiazhuang, 050000, China
| | - Deng-zhou Guo
- The First Affiliated Hospital of Hebei University of Chinese Medicine, Hebei Provincial Hospital of Traditional Chinese Medicine, Shijiazhuang, Chang'an District, Zhongshan East Road 389, 050011, China
| | - Dong Bian
- The First Affiliated Hospital of Hebei University of Chinese Medicine, Hebei Provincial Hospital of Traditional Chinese Medicine, Shijiazhuang, Chang'an District, Zhongshan East Road 389, 050011, China
| |
Collapse
|
11
|
Deora N, Agrohi P, Mallick PK, Sinha A. Critical insights on "Association of the C allele of rs479200 in the EGLN1 gene with COVID‑19 severity in Indian population: a novel finding". Hum Genomics 2024; 18:52. [PMID: 38790075 PMCID: PMC11127436 DOI: 10.1186/s40246-024-00618-4] [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: 03/11/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
The recent article by Harit et al. in Human Genomics reported a novel association of the C allele of rs479200 in the human EGLN1 gene with severe COVID-19 in Indian patients. The gene in context is an oxygen-sensor gene whose T allele has been reported to contribute to the inability to cope with hypoxia due to increased expression of the EGLN1 gene and therefore persons with TT genotype of EGLN1 rs479200 are more susceptible to severe manifestations of hypoxia. In contrast to this dogma, Harit et al. showed that the C allele is associated with the worsening of COVID-19 hypoxia without suggesting or even discussing the scientific plausibility of the association. The article also suffers from certain epidemiological, statistical, and mathematical issues that need to be critically elaborated and discussed. In this context, the findings of Harit et al. may be re-evaluated.
Collapse
Affiliation(s)
- Nimita Deora
- Department of Epidemiology, ICMR-National Institute of Malaria Research, New Delhi, 110077, India
| | - Priya Agrohi
- Department of Molecular Biology, ICMR-National Institute of Malaria Research, New Delhi, 110077, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Prashant K Mallick
- Department of Molecular Biology, ICMR-National Institute of Malaria Research, New Delhi, 110077, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Abhinav Sinha
- Department of Epidemiology, ICMR-National Institute of Malaria Research, New Delhi, 110077, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India.
| |
Collapse
|
12
|
Duan M, Ru X, Zhou J, Li Y, Guo P, Kang W, Li W, Chen Z, Feng H, Chen Y. Endothelial EGLN3-PKM2 signaling induces the formation of acute astrocytic barrier to alleviate immune cell infiltration after subarachnoid hemorrhage. Fluids Barriers CNS 2024; 21:42. [PMID: 38755642 PMCID: PMC11100217 DOI: 10.1186/s12987-024-00550-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 05/09/2024] [Indexed: 05/18/2024] Open
Abstract
BACKGROUND Most subarachnoid hemorrhage (SAH) patients have no obvious hematoma lesions but exhibit blood-brain barrier dysfunction and vasogenic brain edema. However, there is a few days between blood‒brain barrier dysfunction and vasogenic brain edema. The present study sought to investigate whether this phenomenon is caused by endothelial injury induced by the acute astrocytic barrier, also known as the glial limitans. METHODS Bioinformatics analyses of human endothelial cells and astrocytes under hypoxia were performed based on the GEO database. Wild-type, EGLN3 and PKM2 conditional knock-in mice were used to confirm glial limitan formation after SAH. Then, the effect of endothelial EGLN3-PKM2 signaling on temporal and spatial changes in glial limitans was evaluated in both in vivo and in vitro models of SAH. RESULTS The data indicate that in the acute phase after SAH, astrocytes can form a temporary protective barrier, the glia limitans, around blood vessels that helps maintain barrier function and improve neurological prognosis. Molecular docking studies have shown that endothelial cells and astrocytes can promote glial limitans-based protection against early brain injury through EGLN3/PKM2 signaling and further activation of the PKC/ERK/MAPK signaling pathway in astrocytes after SAH. CONCLUSION Improving the ability to maintain glial limitans may be a new therapeutic strategy for improving the prognosis of SAH patients.
Collapse
Affiliation(s)
- Mingxu Duan
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
- Chongqing Key Laboratory of Intelligent Diagnosis, Treatment and Rehabilitation of Central Nervous System Injuries, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Xufang Ru
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
- Chongqing Key Laboratory of Intelligent Diagnosis, Treatment and Rehabilitation of Central Nervous System Injuries, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jiru Zhou
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
- Chongqing Key Laboratory of Intelligent Diagnosis, Treatment and Rehabilitation of Central Nervous System Injuries, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yuanshu Li
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
- Chongqing Key Laboratory of Intelligent Diagnosis, Treatment and Rehabilitation of Central Nervous System Injuries, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Peiwen Guo
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
- Chongqing Key Laboratory of Intelligent Diagnosis, Treatment and Rehabilitation of Central Nervous System Injuries, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Wenbo Kang
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
- Chongqing Key Laboratory of Intelligent Diagnosis, Treatment and Rehabilitation of Central Nervous System Injuries, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Wenyan Li
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
- Chongqing Key Laboratory of Intelligent Diagnosis, Treatment and Rehabilitation of Central Nervous System Injuries, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Zhi Chen
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
- Chongqing Key Laboratory of Intelligent Diagnosis, Treatment and Rehabilitation of Central Nervous System Injuries, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Hua Feng
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.
- Chongqing Key Laboratory of Intelligent Diagnosis, Treatment and Rehabilitation of Central Nervous System Injuries, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| | - Yujie Chen
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.
- Chongqing Key Laboratory of Intelligent Diagnosis, Treatment and Rehabilitation of Central Nervous System Injuries, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| |
Collapse
|
13
|
Zhang C, Meng Y, Han J. Emerging roles of mitochondrial functions and epigenetic changes in the modulation of stem cell fate. Cell Mol Life Sci 2024; 81:26. [PMID: 38212548 PMCID: PMC11072137 DOI: 10.1007/s00018-023-05070-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 11/27/2023] [Accepted: 11/28/2023] [Indexed: 01/13/2024]
Abstract
Mitochondria serve as essential organelles that play a key role in regulating stem cell fate. Mitochondrial dysfunction and stem cell exhaustion are two of the nine distinct hallmarks of aging. Emerging research suggests that epigenetic modification of mitochondria-encoded genes and the regulation of epigenetics by mitochondrial metabolites have an impact on stem cell aging or differentiation. Here, we review how key mitochondrial metabolites and behaviors regulate stem cell fate through an epigenetic approach. Gaining insight into how mitochondria regulate stem cell fate will help us manufacture and preserve clinical-grade stem cells under strict quality control standards, contributing to the development of aging-associated organ dysfunction and disease.
Collapse
Affiliation(s)
- Chensong Zhang
- State Key Laboratory of Biotherapy and Cancer Center, Frontiers Science Center for Disease-Related Molecular Network, and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yang Meng
- State Key Laboratory of Biotherapy and Cancer Center, Frontiers Science Center for Disease-Related Molecular Network, and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Junhong Han
- State Key Laboratory of Biotherapy and Cancer Center, Frontiers Science Center for Disease-Related Molecular Network, and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
14
|
Moreton N, Puzio M, McCormack J, O'Connor JJ. The effects of prolyl hydroxylase inhibition during and post, hypoxia, oxygen glucose deprivation and oxidative stress, in isolated rat hippocampal slices. Brain Res Bull 2023; 205:110822. [PMID: 37984622 DOI: 10.1016/j.brainresbull.2023.110822] [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: 09/25/2023] [Revised: 11/05/2023] [Accepted: 11/17/2023] [Indexed: 11/22/2023]
Abstract
The contributions of hypoxia and oxidative stress to the pathophysiology of acute ischemic stroke are well established and can lead to disruptions in synaptic signaling. Hypoxia and oxidative stress lead to the neurotoxic overproduction of reactive oxygen species (ROS) and the stabilization of hypoxia inducible factors (HIF). Compounds such as prolyl-4-hydroxylase domain enzyme inhibitors (PHDIs) have been shown to have a preconditioning and neuroprotective effect against ischemic insults such as hypoxia, anoxia, oxygen glucose deprivation (OGD) or H2O2. Therefore, this study explored the effects of two PHDIs, JNJ-42041935 (10 µM) and roxadustat (100 µM) on cell viability using organotypic hippocampal slice cultures. We also assessed the effects of these compounds on synaptic transmission during and post hypoxia, OGD and H2O2 application in isolated rat hippocampal slices using field recording electrophysiological techniques and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor subunit trafficking using immunohistochemistry. Our organotypic data demonstrated a protective role for both inhibitors, where slices had significantly less cell death post anoxia and OGD compared to controls. We also report a distinct modulatory role for both JNJ-42041935 and roxadustat on fEPSP slope post hypoxia and OGD but not H2O2. In addition, we report that application of roxadustat impaired long-term potentiation, but only when applied post-hypoxia. This inhibitory effect was not reversed with co-application of the cyclin-dependent kinase 5 (CDK-5) inhibitor, roscovitine (10 µM), suggesting a CDK-5 independent synaptic AMPAR trafficking mechanism. Both hypoxia and OGD induced a reduction in synaptic AMPA GluA2 subunits, the OGD effect being reversed by prior treatment with both JNJ-42041935 and roxadustat. These results suggest an important role for PHDs in synaptic signaling and plasticity during episodes of ischemic stress.
Collapse
Affiliation(s)
- Niamh Moreton
- UCD School of Biomolecular & Biomedical Science, UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Martina Puzio
- UCD School of Biomolecular & Biomedical Science, UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Janet McCormack
- UCD Research Pathology Core, UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - John J O'Connor
- UCD School of Biomolecular & Biomedical Science, UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
15
|
Li Y, Yuan T, Zhang H, Liu S, Lun J, Guo J, Wang Y, Zhang Y, Fang J. PHD3 inhibits colon cancer cell metastasis through the occludin-p38 pathway. Acta Biochim Biophys Sin (Shanghai) 2023; 55:1749-1757. [PMID: 37814811 PMCID: PMC10679873 DOI: 10.3724/abbs.2023103] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 05/06/2023] [Indexed: 10/11/2023] Open
Abstract
Prolyl hydroxylase 3 (PHD3) hydroxylates HIFα in the presence of oxygen, leading to HIFα degradation. PHD3 inhibits tumorigenesis. However, the underlying mechanism is not well understood. Herein, we demonstrate that PHD3 inhibits the metastasis of colon cancer cells through the occludin-p38 MAPK pathway independent of its hydroxylase activity. We find that PHD3 inhibits colon cancer cell metastasis in the presence of the PHD inhibitor DMOG, and prolyl hydroxylase-deficient PHD3(H196A) suppresses cell metastasis as well. PHD3 controls the stability of the tight junction protein occludin in a hydroxylase-independent manner. We further find that PHD3-inhibited colon cancer cell metastasis is rescued by knockdown of occludin and that occludin acts as a negative regulator of cell metastasis, implying that PHD3 suppresses metastasis through occludin. Furthermore, knockdown of occludin induces phosphorylation of p38 MAPK, and the p38 inhibitor SB203580 impedes cell migration and invasion induced by occludin knockdown, indicating that occludin functions through p38. Moreover, knockdown of occludin enhances the expression of MKK3/6, the upstream kinase of p38, while overexpression of occludin decreases its expression. Our results suggest that PHD3 inhibits the metastasis of colon cancer cells through the occludin-p38 pathway independent of its hydroxylase activity. These findings reveal a previously undiscovered mechanism underlying the regulation of cancer cell metastasis by PHD3 and highlight a noncanonical hydroxylase-independent function of PHD3 in the suppression of cancer cells.
Collapse
Affiliation(s)
- Yuyao Li
- Department of Oncologythe Affiliated Hospital of Qingdao UniversitySchool of Basic Medicine of Qingdao UniversityQingdao Cancer InstituteQingdao266071China
| | - Tanglong Yuan
- Shenzhen BranchGuangdong Laboratory for Lingnan Modern AgricultureKey Laboratory of Synthetic BiologyMinistry of Agriculture and Rural AffairsAgricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhen518000China
| | - Hongwei Zhang
- Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao UniversityJinan250014China
| | - Shuting Liu
- Department of Oncologythe Affiliated Hospital of Qingdao UniversitySchool of Basic Medicine of Qingdao UniversityQingdao Cancer InstituteQingdao266071China
| | - Jie Lun
- Department of Oncologythe Affiliated Hospital of Qingdao UniversitySchool of Basic Medicine of Qingdao UniversityQingdao Cancer InstituteQingdao266071China
| | - Jing Guo
- Department of Oncologythe Affiliated Hospital of Qingdao UniversitySchool of Basic Medicine of Qingdao UniversityQingdao Cancer InstituteQingdao266071China
| | - Yu Wang
- Department of Oncologythe Affiliated Hospital of Qingdao UniversitySchool of Basic Medicine of Qingdao UniversityQingdao Cancer InstituteQingdao266071China
| | - Yuying Zhang
- School of Public HealthQingdao UniversityQingdao266071China
| | - Jing Fang
- Department of Oncologythe Affiliated Hospital of Qingdao UniversitySchool of Basic Medicine of Qingdao UniversityQingdao Cancer InstituteQingdao266071China
| |
Collapse
|
16
|
Lombardi F, Augello FR, Palumbo P, Bonfili L, Artone S, Altamura S, Sheldon JM, Latella G, Cifone MG, Eleuteri AM, Cinque B. Bacterial Lysate from the Multi-Strain Probiotic SLAB51 Triggers Adaptative Responses to Hypoxia in Human Caco-2 Intestinal Epithelial Cells under Normoxic Conditions and Attenuates LPS-Induced Inflammatory Response. Int J Mol Sci 2023; 24:ijms24098134. [PMID: 37175841 PMCID: PMC10179068 DOI: 10.3390/ijms24098134] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 04/28/2023] [Accepted: 05/01/2023] [Indexed: 05/15/2023] Open
Abstract
Hypoxia-inducible factor-1α (HIF-1α), a central player in maintaining gut-microbiota homeostasis, plays a pivotal role in inducing adaptive mechanisms to hypoxia and is negatively regulated by prolyl hydroxylase 2 (PHD2). HIF-1α is stabilized through PI3K/AKT signaling regardless of oxygen levels. Considering the crucial role of the HIF pathway in intestinal mucosal physiology and its relationships with gut microbiota, this study aimed to evaluate the ability of the lysate from the multi-strain probiotic formulation SLAB51 to affect the HIF pathway in a model of in vitro human intestinal epithelium (intestinal epithelial cells, IECs) and to protect from lipopolysaccharide (LPS) challenge. The exposure of IECs to SLAB51 lysate under normoxic conditions led to a dose-dependent increase in HIF-1α protein levels, which was associated with higher glycolytic metabolism and L-lactate production. Probiotic lysate significantly reduced PHD2 levels and HIF-1α hydroxylation, thus leading to HIF-1α stabilization. The ability of SLAB51 lysate to increase HIF-1α levels was also associated with the activation of the PI3K/AKT pathway and with the inhibition of NF-κB, nitric oxide synthase 2 (NOS2), and IL-1β increase elicited by LPS treatment. Our results suggest that the probiotic treatment, by stabilizing HIF-1α, can protect from an LPS-induced inflammatory response through a mechanism involving PI3K/AKT signaling.
Collapse
Affiliation(s)
- Francesca Lombardi
- Department of Life, Health & Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | | | - Paola Palumbo
- Department of Life, Health & Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Laura Bonfili
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy
| | - Serena Artone
- Department of Life, Health & Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Serena Altamura
- Department of Life, Health & Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Jenna Marie Sheldon
- Dr. Kiran C Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33314-7796, USA
| | - Giovanni Latella
- Department of Life, Health & Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Maria Grazia Cifone
- Department of Life, Health & Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Anna Maria Eleuteri
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy
| | - Benedetta Cinque
- Department of Life, Health & Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| |
Collapse
|
17
|
Okuda C, Ueda Y, Muroi M, Sanada E, Osada H, Shiono Y, Kimura KI, Takeda K, Kawaguchi K, Kataoka T. Allantopyrone A interferes with the degradation of hypoxia-inducible factor 1α protein by reducing proteasome activity in human fibrosarcoma HT-1080 cells. J Antibiot (Tokyo) 2023; 76:324-334. [PMID: 36997727 DOI: 10.1038/s41429-023-00610-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 02/08/2023] [Accepted: 02/24/2023] [Indexed: 04/01/2023]
Abstract
Allantopyrone A is an α-pyrone metabolite that was originally isolated from the endophytic fungus Allantophomopsis lycopodina KS-97. We previously demonstrated that allantopyrone A exhibits anti-cancer, anti-inflammatory, and neuroprotective activities. In the present study, we showed that allantopyrone A up-regulated the protein expression of hypoxia-inducible factor (HIF)-1α in human fibrosarcoma HT-1080 cells. It also up-regulated the mRNA expression of BNIP3 and ENO1, but not other HIF target genes or HIF1A. Allantopyrone A did not inhibit the prolyl hydroxylation of HIF-1α, but enhanced the ubiquitination of cellular proteins. Consistent with this result, chymotrypsin-like and trypsin-like proteasome activities were reduced, but not completely inactivated by allantopyrone A. Allantopyrone A decreased the amount of proteasome catalytic subunits. Therefore, the present results showed that allantopyrone A interfered with the degradation of HIF-1α protein by reducing proteasome activity in human fibrosarcoma HT-1080 cells.
Collapse
|
18
|
Gong Y, Behera G, Erber L, Luo A, Chen Y. HypDB: A functionally annotated web-based database of the proline hydroxylation proteome. PLoS Biol 2022; 20:e3001757. [PMID: 36026437 PMCID: PMC9455854 DOI: 10.1371/journal.pbio.3001757] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 09/08/2022] [Accepted: 07/13/2022] [Indexed: 01/16/2023] Open
Abstract
Proline hydroxylation (Hyp) regulates protein structure, stability, and protein-protein interaction. It is widely involved in diverse metabolic and physiological pathways in cells and diseases. To reveal functional features of the Hyp proteome, we integrated various data sources for deep proteome profiling of the Hyp proteome in humans and developed HypDB (https://www.HypDB.site), an annotated database and web server for Hyp proteome. HypDB provides site-specific evidence of modification based on extensive LC-MS analysis and literature mining with 14,413 nonredundant Hyp sites on 5,165 human proteins including 3,383 Class I and 4,335 Class II sites. Annotation analysis revealed significant enrichment of Hyp on key functional domains and tissue-specific distribution of Hyp abundance across 26 types of human organs and fluids and 6 cell lines. The network connectivity analysis further revealed a critical role of Hyp in mediating protein-protein interactions. Moreover, the spectral library generated by HypDB enabled data-independent analysis (DIA) of clinical tissues and the identification of novel Hyp biomarkers in lung cancer and kidney cancer. Taken together, our integrated analysis of human proteome with publicly accessible HypDB revealed functional diversity of Hyp substrates and provides a quantitative data source to characterize Hyp in pathways and diseases.
Collapse
Affiliation(s)
- Yao Gong
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota at Twin Cities, Minneapolis, Minnesota, United States of America
- Bioinformatics and Computational Biology Program, University of Minnesota at Twin Cities, Minneapolis, Minnesota, United States of America
| | - Gaurav Behera
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota at Twin Cities, Minneapolis, Minnesota, United States of America
| | - Luke Erber
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota at Twin Cities, Minneapolis, Minnesota, United States of America
| | - Ang Luo
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota at Twin Cities, Minneapolis, Minnesota, United States of America
| | - Yue Chen
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota at Twin Cities, Minneapolis, Minnesota, United States of America
- Bioinformatics and Computational Biology Program, University of Minnesota at Twin Cities, Minneapolis, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
19
|
Tetrahydropyridin-4-ylpicolinoylglycines as novel and orally active prolyl hydroxylase 2 (PHD2) inhibitors for the treatment of renal anemia. Eur J Med Chem 2022; 238:114479. [PMID: 35675755 DOI: 10.1016/j.ejmech.2022.114479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/15/2022] [Accepted: 05/16/2022] [Indexed: 11/21/2022]
Abstract
Prolyl hydroxylase 2 (PHD2) is a key regulatory enzyme responsible for the degradation of hypoxia-inducible factor-α (HIF-α). Pharmacological inhibition of PHD2 stabilizes HIF-α and induces the production of endogenous erythropoietin (EPO), which is regarded as a promising strategy for the treatment of renal anemia. To date, a series of PHD2 inhibitors have been approved or advanced into clinical studies. In this study, we developed a new type of PHD2 inhibitors with the tetrahydropyridin-4-ylpicolinoylglycine scaffold by using a scaffold hopping strategy. Among them, compound 25 showed potent inhibition toward PHD2 with an IC50 of 6.55 ± 0.41 nM. Furthermore, compound 25 upregulated reticulocytes in C57BL/6 mice. The subacute toxicological assay demonstrated 25 has no obvious toxicity in vivo. Overall, compound 25 is a promising candidate for the treatment of renal anemia.
Collapse
|
20
|
Tang J, Deng H, Wang Z, Zha H, Liao Q, Zhu C, Chen X, Sun X, Jia S, Ouyang G, Liu X, Xiao W. EGLN1 prolyl hydroxylation of hypoxia-induced transcription factor HIF1α is repressed by SET7-catalyzed lysine methylation. J Biol Chem 2022; 298:101961. [PMID: 35452683 PMCID: PMC9123262 DOI: 10.1016/j.jbc.2022.101961] [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: 01/05/2022] [Revised: 04/13/2022] [Accepted: 04/15/2022] [Indexed: 11/30/2022] Open
Abstract
Egg laying defective nine 1 (EGLN1) functions as an oxygen sensor to catalyze prolyl hydroxylation of the transcription factor hypoxia-inducible factor-1 α (HIF1α) under normoxia conditions, leading to its proteasomal degradation. Thus, EGLN1 plays a central role in the HIF-mediated hypoxia signaling pathway; however, the post-translational modifications that control EGLN1 function remain largely unknown. Here, we identified that a lysine monomethylase, SET7, catalyzes EGLN1 methylation on lysine 297, resulting in the repression of EGLN1 activity in catalyzing prolyl hydroxylation of HIF1α. Notably, we demonstrate that the methylation mimic mutant of EGLN1 loses the capability to suppress the hypoxia signaling pathway, leading to the enhancement of cell proliferation and the oxygen consumption rate. Collectively, our data identify a novel modification of EGLN1 that is critical for inhibiting its enzymatic activity, and which may benefit cellular adaptation to conditions of hypoxia.
Collapse
Affiliation(s)
- Jinhua Tang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Hongyan Deng
- College of Life Science, Wuhan University, Wuhan, 430072, P. R. China
| | - Zixuan Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Huangyuan Zha
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences
| | - Qian Liao
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Chunchun Zhu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Xiaoyun Chen
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Xueyi Sun
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Shuke Jia
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Gang Ouyang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Xing Liu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Beijing, 100049, P. R. China; The Innovation of Seed Design, Chinese Academy of Sciences, Wuhan, 430072, P. R. China.
| | - Wuhan Xiao
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Beijing, 100049, P. R. China; The Innovation of Seed Design, Chinese Academy of Sciences, Wuhan, 430072, P. R. China; Hubei Hongshan Laboratory, Wuhan, 430070, P. R. China.
| |
Collapse
|
21
|
Miao M, Wu M, Li Y, Zhang L, Jin Q, Fan J, Xu X, Gu R, Hao H, Zhang A, Jia Z. Clinical Potential of Hypoxia Inducible Factors Prolyl Hydroxylase Inhibitors in Treating Nonanemic Diseases. Front Pharmacol 2022; 13:837249. [PMID: 35281917 PMCID: PMC8908211 DOI: 10.3389/fphar.2022.837249] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 01/19/2022] [Indexed: 12/19/2022] Open
Abstract
Hypoxia inducible factors (HIFs) and their regulatory hydroxylases the prolyl hydroxylase domain enzymes (PHDs) are the key mediators of the cellular response to hypoxia. HIFs are normally hydroxylated by PHDs and degraded, while under hypoxia, PHDs are suppressed, allowing HIF-α to accumulate and transactivate multiple target genes, including erythropoiesis, and genes participate in angiogenesis, iron metabolism, glycolysis, glucose transport, cell proliferation, survival, and so on. Aiming at stimulating HIFs, a group of small molecules antagonizing HIF-PHDs have been developed. Of these HIF-PHDs inhibitors (HIF-PHIs), roxadustat (FG-4592), daprodustat (GSK-1278863), vadadustat (AKB-6548), molidustat (BAY 85-3934) and enarodustat (JTZ-951) are approved for clinical usage or have progressed into clinical trials for chronic kidney disease (CKD) anemia treatment, based on their activation effect on erythropoiesis and iron metabolism. Since HIFs are involved in many physiological and pathological conditions, efforts have been made to extend the potential usage of HIF-PHIs beyond anemia. This paper reviewed the progress of preclinical and clinical research on clinically available HIF-PHIs in pathological conditions other than CKD anemia.
Collapse
Affiliation(s)
- Mengqiu Miao
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Mengqiu Wu
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Yuting Li
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Lingge Zhang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Qianqian Jin
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Jiaojiao Fan
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.,School of Medicine, Southeast University, Nanjing, China
| | - Xinyue Xu
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.,School of Medicine, Southeast University, Nanjing, China
| | - Ran Gu
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Haiping Hao
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism, China Pharmaceutical University, Nanjing, China
| | - Aihua Zhang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Zhanjun Jia
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| |
Collapse
|
22
|
Husak JF, Lailvaux SP. Conserved and convergent mechanisms underlying performance-life-history trade-offs. J Exp Biol 2022; 225:274252. [PMID: 35119073 DOI: 10.1242/jeb.243351] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Phenotypic trade-offs are inevitable in nature, but the mechanisms driving them are poorly understood. Movement and oxygen are essential to all animals, and as such, the common ancestor to all living animals passed on mechanisms to acquire oxygen and contract muscle, sometimes at the expense of other activities or expression of traits. Nevertheless, convergent pathways have also evolved to deal with critical trade-offs that are necessary to survive ubiquitous environmental challenges. We discuss how whole-animal performance traits, such as locomotion, are important to fitness, yet costly, resulting in trade-offs with other aspects of the phenotype via specific conserved and convergent mechanistic pathways across all animals. Specifically, we discuss conserved pathways involved in muscle structure and signaling, insulin/insulin-like signaling, sirtuins, mitochondria and hypoxia-inducible factors, as well as convergent pathways involved in energy regulation, development, reproductive investment and energy storage. The details of these mechanisms are only known from a few model systems, and more comparative studies are needed. We make two main recommendations as a framework for future studies of animal form and function. First, studies of performance should consider the broader life-history context of the organism, and vice versa, as performance expression can require a large portion of acquired resources. Second, studies of life histories or mechanistic pathways that measure performance should do so in meaningful and standardized ways. Understanding proximate mechanisms of phenotypic trade-offs will not only better explain the phenotypes of the organisms we study, but also allow predictions about phenotypic variation at the evolutionary scale.
Collapse
Affiliation(s)
- Jerry F Husak
- Department of Biology, University of St. Thomas, St. Paul, MN 55105, USA
| | - Simon P Lailvaux
- Department of Biological Sciences, University of New Orleans, New Orleans, LA 70148, USA
| |
Collapse
|
23
|
Li X, Zhang Q, Wang Z, Zhuang Q, Zhao M. Immune and Metabolic Alterations in Liver Fibrosis: A Disruption of Oxygen Homeostasis? Front Mol Biosci 2022; 8:802251. [PMID: 35187072 PMCID: PMC8850363 DOI: 10.3389/fmolb.2021.802251] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 12/17/2021] [Indexed: 12/06/2022] Open
Abstract
According to the WHO, “cirrhosis of the liver” was the 11th leading cause of death globally in 2019. Many kinds of liver diseases can develop into liver cirrhosis, and liver fibrosis is the main pathological presentation of different aetiologies, including toxic damage, viral infection, and metabolic and genetic diseases. It is characterized by excessive synthesis and decreased decomposition of extracellular matrix (ECM). Hepatocyte cell death, hepatic stellate cell (HSC) activation, and inflammation are crucial incidences of liver fibrosis. The process of fibrosis is also closely related to metabolic and immune disorders, which are usually induced by the destruction of oxygen homeostasis, including mitochondrial dysfunction, oxidative stress, and hypoxia pathway activation. Mitochondria are important organelles in energy generation and metabolism. Hypoxia-inducible factors (HIFs) are key factors activated when hypoxia occurs. Both are considered essential factors of liver fibrosis. In this review, the authors highlight the impact of oxygen imbalance on metabolism and immunity in liver fibrosis as well as potential novel targets for antifibrotic therapies.
Collapse
Affiliation(s)
- Xinyu Li
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Quyan Zhang
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Zeyu Wang
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Quan Zhuang
- Transplantation Center, The Third Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Quan Zhuang, ; Mingyi Zhao,
| | - Mingyi Zhao
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Quan Zhuang, ; Mingyi Zhao,
| |
Collapse
|
24
|
Ruan Y, Fang X, Guo T, Liu Y, Hu Y, Wang X, Hu Y, Gao L, Li Y, Pi J, Xu Y. Metabolic reprogramming in the arsenic carcinogenesis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 229:113098. [PMID: 34952379 DOI: 10.1016/j.ecoenv.2021.113098] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 12/06/2021] [Accepted: 12/14/2021] [Indexed: 06/14/2023]
Abstract
Chronic exposure to arsenic has been associated with a variety of cancers with the mechanisms undefined. Arsenic exposure causes alterations in metabolites in bio-samples. Recent research progress on cancer biology suggests that metabolic reprogramming contributes to tumorigenesis. Therefore, metabolic reprogramming provides a new clue for the mechanisms of arsenic carcinogenesis. In the present manuscript, we review the latest findings in reprogramming of glucose, lipids, and amino acids in response to arsenic exposure. Most studies focused on glucose reprogramming and found that arsenic exposure enhanced glycolysis. However, in vivo studies observed "reverse Warburg effect" in some cases due to the complexity of the disease evolution and microenvironment. Arsenic exposure has been reported to disturb lipid deposition by inhibiting lipolysis, and induce serine-glycine one-carbon pathway. As a dominant mechanism for arsenic toxicity, oxidative stress is considered to link with metabolism reprogramming. Few studies analyzed the causal relationship between metabolic reprogramming and arsenic-induced cancers. Metabolic alterations may vary with exposure doses and periods. Identifying metabolic alterations common among humans and experiment models with human-relevant exposure characteristics may guide future investigations.
Collapse
Affiliation(s)
- Yihui Ruan
- Group of Chronic Disease and Environmental Genomics, School of Public Health, China Medical University, P.R. China
| | - Xin Fang
- Group of Chronic Disease and Environmental Genomics, School of Public Health, China Medical University, P.R. China
| | - Tingyue Guo
- Group of Chronic Disease and Environmental Genomics, School of Public Health, China Medical University, P.R. China
| | - Yiting Liu
- Group of Chronic Disease and Environmental Genomics, School of Public Health, China Medical University, P.R. China
| | - Yu Hu
- Group of Chronic Disease and Environmental Genomics, School of Public Health, China Medical University, P.R. China
| | - Xuening Wang
- Group of Chronic Disease and Environmental Genomics, School of Public Health, China Medical University, P.R. China
| | - Yuxin Hu
- Experimental Teaching Center, School of Public Health, China Medical University, P.R. China
| | - Lanyue Gao
- Experimental Teaching Center, School of Public Health, China Medical University, P.R. China
| | - Yongfang Li
- The Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, China Medical University, P.R. China
| | - Jingbo Pi
- The Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, China Medical University, P.R. China; Program of Environmental Toxicology, School of Public Health, China Medical University, P.R. China
| | - Yuanyuan Xu
- Group of Chronic Disease and Environmental Genomics, School of Public Health, China Medical University, P.R. China; The Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, China Medical University, P.R. China.
| |
Collapse
|
25
|
Zhang Y, Wang H, Oliveira RHM, Zhao C, Popel AS. Systems biology of angiogenesis signaling: Computational models and omics. WIREs Mech Dis 2021; 14:e1550. [PMID: 34970866 PMCID: PMC9243197 DOI: 10.1002/wsbm.1550] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 12/03/2021] [Accepted: 12/06/2021] [Indexed: 01/10/2023]
Abstract
Angiogenesis is a highly regulated multiscale process that involves a plethora of cells, their cellular signal transduction, activation, proliferation, differentiation, as well as their intercellular communication. The coordinated execution and integration of such complex signaling programs is critical for physiological angiogenesis to take place in normal growth, development, exercise, and wound healing, while its dysregulation is critically linked to many major human diseases such as cancer, cardiovascular diseases, and ocular disorders; it is also crucial in regenerative medicine. Although huge efforts have been devoted to drug development for these diseases by investigation of angiogenesis‐targeted therapies, only a few therapeutics and targets have proved effective in humans due to the innate multiscale complexity and nonlinearity in the process of angiogenic signaling. As a promising approach that can help better address this challenge, systems biology modeling allows the integration of knowledge across studies and scales and provides a powerful means to mechanistically elucidate and connect the individual molecular and cellular signaling components that function in concert to regulate angiogenesis. In this review, we summarize and discuss how systems biology modeling studies, at the pathway‐, cell‐, tissue‐, and whole body‐levels, have advanced our understanding of signaling in angiogenesis and thereby delivered new translational insights for human diseases. This article is categorized under:Cardiovascular Diseases > Computational Models Cancer > Computational Models
Collapse
Affiliation(s)
- Yu Zhang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Hanwen Wang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Rebeca Hannah M Oliveira
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Chen Zhao
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Aleksander S Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
26
|
Chacon-Barahona JA, Salladay-Perez IA, Lanning NJ. Lung Adenocarcinoma Transcriptomic Analysis Predicts Adenylate Kinase Signatures Contributing to Tumor Progression and Negative Patient Prognosis. Metabolites 2021; 11:metabo11120859. [PMID: 34940617 PMCID: PMC8705281 DOI: 10.3390/metabo11120859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 11/30/2021] [Indexed: 11/16/2022] Open
Abstract
The ability to detect and respond to hypoxia within a developing tumor appears to be a common feature amongst most cancers. This hypoxic response has many molecular drivers, but none as widely studied as Hypoxia-Inducible Factor 1 (HIF-1). Recent evidence suggests that HIF-1 biology within lung adenocarcinoma (LUAD) may be associated with expression levels of adenylate kinases (AKs). Using LUAD patient transcriptome data, we sought to characterize AK gene signatures related to lung cancer hallmarks, such as hypoxia and metabolic reprogramming, to identify conserved biological themes across LUAD tumor progression. Transcriptomic analysis revealed perturbation of HIF-1 targets to correlate with altered expression of most AKs, with AK4 having the strongest correlation. Enrichment analysis of LUAD tumor AK4 gene signatures predicts signatures involved in pyrimidine, and by extension, nucleotide metabolism across all LUAD tumor stages. To further discriminate potential drivers of LUAD tumor progression within AK4 gene signatures, partial least squares discriminant analysis was used at LUAD stage-stage interfaces, identifying candidate genes that may promote LUAD tumor growth or regression. Collectively, these results characterize regulatory gene networks associated with the expression of all nine human AKs that may contribute to underlying metabolic perturbations within LUAD and reveal potential mechanistic insight into the complementary role of AK4 in LUAD tumor development.
Collapse
Affiliation(s)
- Jonathan A. Chacon-Barahona
- Department of Biological Sciences, California State University, Los Angeles, CA 90032, USA; (J.A.C.-B.); (I.A.S.-P.)
| | - Ivan A. Salladay-Perez
- Department of Biological Sciences, California State University, Los Angeles, CA 90032, USA; (J.A.C.-B.); (I.A.S.-P.)
- Molecular Biology Interdepartmental Program, University of California, Los Angeles, CA 94701, USA
| | - Nathan James Lanning
- Department of Biological Sciences, California State University, Los Angeles, CA 90032, USA; (J.A.C.-B.); (I.A.S.-P.)
- Correspondence: ; Tel.: +1-(323)-343-2092
| |
Collapse
|
27
|
Li S, Fu J, Wang Y, Hu C, Xu F. LncRNA MIAT enhances cerebral ischaemia/reperfusion injury in rat model via interacting with EGLN2 and reduces its ubiquitin-mediated degradation. J Cell Mol Med 2021; 25:10140-10151. [PMID: 34687132 PMCID: PMC8572800 DOI: 10.1111/jcmm.16950] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 08/26/2021] [Accepted: 09/19/2021] [Indexed: 11/30/2022] Open
Abstract
Long non-coding RNA (lncRNA) MIAT (myocardial infarction associated transcript) has been characterized as a functional lncRNA modulating cerebral ischaemic/reperfusion (I/R) injury. However, the underlying mechanisms remain poorly understood. This study explored the functional partners of MIAT in primary rat neurons and their regulation on I/R injury. Sprague-Dawley rats were used to construct middle cerebral artery occlusion (MCAO) models. Their cerebral cortical neurons were used for in vitro oxygen-glucose deprivation/reoxygenation (OGD/R) models. Results showed that MIAT interacted with EGLN2 in rat cortical neurons. MIAT overexpression or knockdown did not alter EGLN2 transcription. In contrast, MIAT overexpression increased EGLN2 stability after I/R injury via reducing its ubiquitin-mediated degradation. EGLN2 was a substrate of MDM2, a ubiquitin E3 ligase. MDM2 interacted with the N-terminal of EGLN2 and mediated its K48-linked poly-ubiquitination, thereby facilitating its proteasomal degradation. MIAT knockdown enhanced the interaction and reduced EGLN2 stability. MIAT overexpression enhanced infarct volume and induced a higher ratio of neuronal apoptosis. EGLN2 knockdown significantly reversed the injury. MIAT overexpression reduced oxidative pentose phosphate pathway flux and increased oxidized/reduced glutathione ratio, the effects of which were abrogated by EGLN2 knockdown. In conclusion, MIAT might act as a stabilizer of EGLN2 via reducing MDM2 mediated K48 poly-ubiquitination. MIAT-EGLN2 axis exacerbates I/R injury via altering redox homeostasis in neurons.
Collapse
Affiliation(s)
- Suping Li
- Department of Neurology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China
| | - Jing Fu
- Department of Rehabilitation, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China
| | - Yi Wang
- Department of Specialty of Geriatric Endocrinology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China
| | - Chunmei Hu
- Department of Otolaryngology-Head and Neck Surgery, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China
| | - Fei Xu
- Department of Neurology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China
| |
Collapse
|
28
|
Dzhalilova D, Kosyreva A, Vishnyakova P, Zolotova N, Tsvetkov I, Mkhitarov V, Mikhailova L, Kakturskiy L, Makarova O. Age-related differences in hypoxia-associated genes and cytokine profile in male Wistar rats. Heliyon 2021; 7:e08085. [PMID: 34632150 PMCID: PMC8488852 DOI: 10.1016/j.heliyon.2021.e08085] [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: 04/01/2021] [Revised: 08/25/2021] [Accepted: 09/26/2021] [Indexed: 11/29/2022] Open
Abstract
Hypoxia tolerance of the organism depends on many factors, including age. High newborn organisms tolerance and high level of oxidative stress throughout aging were demonstrated by many studies. However, there is lack of investigations reflecting the expression of key hypoxia-inducible factor HIF in different age organisms in correlation to levels of pro-inflammatory and anti-inflammatory cytokines. Liver is a sensitive to hypoxia organ, and is an important organ in providing an acute reaction to infections – it synthesizes acute inflammation phase proteins, in particular, C-reactive protein. The aim of study was to determine relationship between age-related tolerance to hypoxia and HIF-1 and PHD2 (prolyl hydroxylase domain protein) expression levels in the liver and the production of cytokines in the spleen in newborn, prepubertal and adult Wistar rats. Newborn rats are characterized by high mRNA Hif-1α expression level in the liver, accompanied by a low content of HIF-1 protein and high level of PHD2. The growth in HIF-1α protein level throughout age is accompanied by the growth of pro-inflammatory cytokines level. Prepubertal animals are the least hypoxia resistant and their HIF-1α mRNA expression level was higher than in adult animals. The PHD2 activity in prepubertal animals was significantly reduced in comparison to newborn rats, and the HIF-1α protein level did not change. Further studies require the identification of additional mechanisms, determining the regulation of the HIF-1α level in prepubertal animals.
Collapse
Affiliation(s)
- Dzhuliia Dzhalilova
- Department of Immunomorphology of Inflammation, Federal State Budgetary Institution Research Institute of Human Morphology, 117418, Tsurupa st., 3, Moscow, Russia
| | - Anna Kosyreva
- Department of Immunomorphology of Inflammation, Federal State Budgetary Institution Research Institute of Human Morphology, 117418, Tsurupa st., 3, Moscow, Russia.,Histology Department, Peoples Friendship University of Russia (RUDN University), 117198, Miklukho-Maklaya st. 6, Moscow, Russia
| | - Polina Vishnyakova
- Histology Department, Peoples Friendship University of Russia (RUDN University), 117198, Miklukho-Maklaya st. 6, Moscow, Russia.,National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, 117997, Oparina st., 4, Moscow, Russia
| | - Natalia Zolotova
- Department of Immunomorphology of Inflammation, Federal State Budgetary Institution Research Institute of Human Morphology, 117418, Tsurupa st., 3, Moscow, Russia
| | - Ivan Tsvetkov
- Department of Immunomorphology of Inflammation, Federal State Budgetary Institution Research Institute of Human Morphology, 117418, Tsurupa st., 3, Moscow, Russia
| | - Vladimir Mkhitarov
- Department of Immunomorphology of Inflammation, Federal State Budgetary Institution Research Institute of Human Morphology, 117418, Tsurupa st., 3, Moscow, Russia
| | - Liliya Mikhailova
- Department of Immunomorphology of Inflammation, Federal State Budgetary Institution Research Institute of Human Morphology, 117418, Tsurupa st., 3, Moscow, Russia
| | - Lev Kakturskiy
- Department of Immunomorphology of Inflammation, Federal State Budgetary Institution Research Institute of Human Morphology, 117418, Tsurupa st., 3, Moscow, Russia
| | - Olga Makarova
- Department of Immunomorphology of Inflammation, Federal State Budgetary Institution Research Institute of Human Morphology, 117418, Tsurupa st., 3, Moscow, Russia
| |
Collapse
|
29
|
Yang Y, Lee EH, Yang Z. Hypoxia conditioned mesenchymal stem cells in tissue regeneration application. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:966-977. [PMID: 34569290 DOI: 10.1089/ten.teb.2021.0145] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Mesenchymal stem cells (MSCs) have been demonstrated as promising cell sources for tissue regeneration due to their capability of self-regeneration, differentiation and immunomodulation. MSCs also exert extensive paracrine effects through release of trophic factors and extracellular vesicles. However, despite extended exploration of MSCs in pre-clinical studies, the results are far from satisfactory due to the poor engraftment and low level of survival after implantation. Hypoxia preconditioning has been proposed as an engineering approach to improve the therapeutic potential of MSCs. During in vitro culture, hypoxic conditions can promote MSC proliferation, survival and migration through various cellular responses to the reduction of oxygen tension. The multilineage differentiation potential of MSCs is altered under hypoxia, with consistent reports of enhanced chondrogenesis. Hypoxia also stimulates the paracrine activities of MSCs and increases the production of secretome both in terms of soluble factors as well as extracellular vesicles. The secretome from hypoxia preconditioned MSCs play important roles in promoting cell proliferation and migration, enhancing angiogenesis while inhibiting apoptosis and inflammation. In this review, we summarise current knowledge of hypoxia-induced changes in MSCs and discuss the application of hypoxia preconditioned MSCs as well as hypoxic secretome in different kinds of disease models.
Collapse
Affiliation(s)
- Yanmeng Yang
- National University of Singapore, 37580, Orthopaedic Surgery, 27 Medical Drive, Singapore, Singapore, 117510;
| | - Eng Hin Lee
- National University of Singapore, Department of Orthopaedic Surgery, 1E Kent Ridge Road, NUHS Tower Block, Level 11, Singapore, Singapore, 119228;
| | - Zheng Yang
- National University of Singapore, Life Sciences Institute, Singapore, Singapore;
| |
Collapse
|
30
|
Zheng S, Liu Q, Liu T, Lu X. Posttranslational modification of pyruvate kinase type M2 (PKM2): novel regulation of its biological roles to be further discovered. J Physiol Biochem 2021; 77:355-363. [PMID: 33835423 DOI: 10.1007/s13105-021-00813-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 04/01/2021] [Indexed: 11/28/2022]
Abstract
PKM2, pyruvate kinase type M2, has been shown to play a key role in aerobic glycolysis and to regulate the malignant behaviors of cancer cells. Recently, PKM2 has been revealed to hold dual metabolic and nonmetabolic roles. Working as both a pyruvate kinase with catalytic activity and a protein kinase that phosphorylates its substrates, PKM2 stands at the crossroads of glycolysis and tumor growth. Recently, it was revealed that the catalytic activity of PKM2 can be regulated by its posttranslational modification (PTM). Several PTM types, including phosphorylation, methylation, acetylation, oxidation, hydroxylation, succinylation, and glycylation, have been gradually identified on different amino acid residues of the PKM2 coding sequence. In this review, we highlight the recent advancements in understanding PKM2 PTMs and the regulatory roles conferred by PTMs during anaerobic glycolysis in tumors.
Collapse
Affiliation(s)
- Shutao Zheng
- Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, People's Republic of China
- State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Urumqi, Xinjiang Uygur Autonomous Region, People's Republic of China
| | - Qing Liu
- Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, People's Republic of China
- State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Urumqi, Xinjiang Uygur Autonomous Region, People's Republic of China
| | - Tao Liu
- Department of Clinical Laboratory, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, People's Republic of China
| | - Xiaomei Lu
- Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, People's Republic of China.
- State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Urumqi, Xinjiang Uygur Autonomous Region, People's Republic of China.
| |
Collapse
|
31
|
Feng D, Zhai Z, Shao Z, Zhang Y, Powell-Coffman JA. Crosstalk in oxygen homeostasis networks: SKN-1/NRF inhibits the HIF-1 hypoxia-inducible factor in Caenorhabditis elegans. PLoS One 2021; 16:e0249103. [PMID: 34242227 PMCID: PMC8270126 DOI: 10.1371/journal.pone.0249103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 06/24/2021] [Indexed: 11/30/2022] Open
Abstract
During development, homeostasis, and disease, organisms must balance responses that allow adaptation to low oxygen (hypoxia) with those that protect cells from oxidative stress. The evolutionarily conserved hypoxia-inducible factors are central to these processes, as they orchestrate transcriptional responses to oxygen deprivation. Here, we employ genetic strategies in C. elegans to identify stress-responsive genes and pathways that modulate the HIF-1 hypoxia-inducible factor and facilitate oxygen homeostasis. Through a genome-wide RNAi screen, we show that RNAi-mediated mitochondrial or proteasomal dysfunction increases the expression of hypoxia-responsive reporter Pnhr-57::GFP in C. elegans. Interestingly, only a subset of these effects requires hif-1. Of particular importance, we found that skn-1 RNAi increases the expression of hypoxia-responsive reporter Pnhr-57::GFP and elevates HIF-1 protein levels. The SKN-1/NRF transcription factor has been shown to promote oxidative stress resistance. We present evidence that the crosstalk between HIF-1 and SKN-1 is mediated by EGL-9, the prolyl hydroxylase that targets HIF-1 for oxygen-dependent degradation. Treatment that induces SKN-1, such as heat or gsk-3 RNAi, increases expression of a Pegl-9::GFP reporter, and this effect requires skn-1 function and a putative SKN-1 binding site in egl-9 regulatory sequences. Collectively, these data support a model in which SKN-1 promotes egl-9 transcription, thereby inhibiting HIF-1. We propose that this interaction enables animals to adapt quickly to changes in cellular oxygenation and to better survive accompanying oxidative stress.
Collapse
Affiliation(s)
- Dingxia Feng
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, United States of America
| | - Zhiwei Zhai
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, United States of America
| | - Zhiyong Shao
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, United States of America
| | - Yi Zhang
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, United States of America
| | - Jo Anne Powell-Coffman
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, United States of America
| |
Collapse
|
32
|
Khalaf K, Hana D, Chou JTT, Singh C, Mackiewicz A, Kaczmarek M. Aspects of the Tumor Microenvironment Involved in Immune Resistance and Drug Resistance. Front Immunol 2021; 12:656364. [PMID: 34122412 PMCID: PMC8190405 DOI: 10.3389/fimmu.2021.656364] [Citation(s) in RCA: 219] [Impact Index Per Article: 54.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 04/27/2021] [Indexed: 12/11/2022] Open
Abstract
The tumor microenvironment (TME) is a complex and ever-changing "rogue organ" composed of its own blood supply, lymphatic and nervous systems, stroma, immune cells and extracellular matrix (ECM). These complex components, utilizing both benign and malignant cells, nurture the harsh, immunosuppressive and nutrient-deficient environment necessary for tumor cell growth, proliferation and phenotypic flexibility and variation. An important aspect of the TME is cellular crosstalk and cell-to-ECM communication. This interaction induces the release of soluble factors responsible for immune evasion and ECM remodeling, which further contribute to therapy resistance. Other aspects are the presence of exosomes contributed by both malignant and benign cells, circulating deregulated microRNAs and TME-specific metabolic patterns which further potentiate the progression and/or resistance to therapy. In addition to biochemical signaling, specific TME characteristics such as the hypoxic environment, metabolic derangements, and abnormal mechanical forces have been implicated in the development of treatment resistance. In this review, we will provide an overview of tumor microenvironmental composition, structure, and features that influence immune suppression and contribute to treatment resistance.
Collapse
Affiliation(s)
- Khalil Khalaf
- Department of Cancer Diagnostics and Immunology, Greater Poland Cancer Center, Poznań, Poland
- Department of Cancer Immunology, Poznan University of Medical Sciences, Poznań, Poland
| | - Doris Hana
- Department of Cancer Diagnostics and Immunology, Greater Poland Cancer Center, Poznań, Poland
- Department of Cancer Immunology, Poznan University of Medical Sciences, Poznań, Poland
| | - Jadzia Tin-Tsen Chou
- Department of Cancer Diagnostics and Immunology, Greater Poland Cancer Center, Poznań, Poland
- Department of Cancer Immunology, Poznan University of Medical Sciences, Poznań, Poland
| | - Chandpreet Singh
- Department of Cancer Diagnostics and Immunology, Greater Poland Cancer Center, Poznań, Poland
- Department of Cancer Immunology, Poznan University of Medical Sciences, Poznań, Poland
| | - Andrzej Mackiewicz
- Department of Cancer Diagnostics and Immunology, Greater Poland Cancer Center, Poznań, Poland
- Department of Cancer Immunology, Poznan University of Medical Sciences, Poznań, Poland
| | - Mariusz Kaczmarek
- Department of Cancer Diagnostics and Immunology, Greater Poland Cancer Center, Poznań, Poland
- Department of Cancer Immunology, Poznan University of Medical Sciences, Poznań, Poland
| |
Collapse
|
33
|
Ma M, Qin F, Wu C, Xiong W, Yu B, Wei S, Huang C, Xu J, Yang X, Yuan J. Optimal vacuum erectile device therapy regimen for penile rehabilitation in a bilateral cavernous nerve crush rat model. Andrology 2021; 9:894-905. [PMID: 33420755 DOI: 10.1111/andr.12968] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 11/19/2020] [Accepted: 01/04/2021] [Indexed: 02/05/2023]
Abstract
BACKGROUND Vacuum erectile device (VED) therapy has been widely used in penile rehabilitation after radical prostatectomy; however, there is no consensus on the best regimen. OBJECTIVES To explore an optimal VED therapy regimen in bilateral cavernous nerve crush (BCNC) rat model. MATERIALS AND METHODS Adult male rats were used to measure the effects of different durations (1-30 min) of VED treatment on penile length, penile blood gas analysis, and adverse effects. Forty-eight adult male rats were randomly divided into Sham, BCNC, and VED treatment groups (2-3-2-3 min, 4-3-3 min, 5-5 min, and 10 min). Penile length, erectile function, and side effects were detected after VED treatment. Histopathological staining and Western blotting were performed to explore the cellular and molecular changes. RESULTS Prolongation of the duration of VED treatment significantly decreased the penile oxygen saturation, partial oxygen pressure, and arterial blood ratio (P < 0.05). Compared with the BCNC group, all VED treatment regimens partially reversed BCNC-induced penile shortening and erectile dysfunction (P < 0.0001), with the 4-3-3-min and 5-5-min treatment groups exhibiting more significant improvement than the 10-min and 2-3-2-3-min treatment groups (P < 0.0001). The mechanism may be related to the up-regulation of the smooth muscle cell/collagen ratio, endothelial nitric oxide synthase, and α-smooth muscle actin (all P < 0.0001); and the down-regulation of hypoxia-inducible factor-1α, transforming growth factor-β1, and apoptosis (all P < 0.0001). The incidence of adverse effects in the 2-3-2-3-min treatment group was the highest. DISCUSSION The commonly used VED therapy regimens maintained erectile function and penile length of BCNC rat by relieving hypoxia and fibrosis, and no further benefits were observed with increased treatment frequency or prolonged treatment duration. CONCLUSION Two consecutive 5-min treatments with a short interval is the optimal VED therapy regimen for penile rehabilitation in BCNC rat model.
Collapse
Affiliation(s)
- Ming Ma
- Andrology Laboratory, West China Hospital, Sichuan University, Chengdu, China
- Department of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Feng Qin
- Andrology Laboratory, West China Hospital, Sichuan University, Chengdu, China
| | - Changjing Wu
- Andrology Laboratory, West China Hospital, Sichuan University, Chengdu, China
| | - Wenfeng Xiong
- Andrology Laboratory, West China Hospital, Sichuan University, Chengdu, China
| | - Botao Yu
- Andrology Laboratory, West China Hospital, Sichuan University, Chengdu, China
- Department of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Shanzun Wei
- Andrology Laboratory, West China Hospital, Sichuan University, Chengdu, China
- Department of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Chunxu Huang
- Andrology Laboratory, West China Hospital, Sichuan University, Chengdu, China
| | - Jianrong Xu
- Andrology Laboratory, West China Hospital, Sichuan University, Chengdu, China
| | - Xinzong Yang
- Andrology Laboratory, West China Hospital, Sichuan University, Chengdu, China
| | - Jiuhong Yuan
- Andrology Laboratory, West China Hospital, Sichuan University, Chengdu, China
- Department of Urology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
34
|
Yu M, Lun J, Zhang H, Zhu L, Zhang G, Fang J. The non-canonical functions of HIF prolyl hydroxylases and their dual roles in cancer. Int J Biochem Cell Biol 2021; 135:105982. [PMID: 33894356 DOI: 10.1016/j.biocel.2021.105982] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 04/12/2021] [Accepted: 04/19/2021] [Indexed: 12/20/2022]
Abstract
The hypoxia-inducible factor (HIF) prolyl hydroxylases (PHDs) are dioxygenases using oxygen and 2-oxoglutarate as co-substrates. Under normoxia, PHDs hydroxylate the conserved prolyl residues of HIFα, leading to HIFα degradation. In hypoxia PHDs are inactivated, which results in HIFα accumulation. The accumulated HIFα enters nucleus and initiates gene transcription. Many studies have shown that PHDs have substrates other than HIFα, implying that they have HIF-independent non-canonical functions. Besides modulating protein stability, the PHDs-mediated prolyl hydroxylation affects protein-protein interaction and protein activity for alternative substrates. Increasing evidence indicates that PHDs also have hydroxylase-independent functions. They influence protein stability, enzyme activity, and protein-protein interaction in a hydroxylase-independent manner. These findings highlight the functional diversity and complexity of PHDs. Due to having inhibitory activity on HIFα, PHDs are proposed to act as tumor suppressors. However, research shows that PHDs exert either tumor-promoting or tumor-suppressing features. Here, we try to summarize the current understanding of PHDs hydroxylase-dependent and -independent functions and their roles in cancer.
Collapse
Affiliation(s)
- Mengchao Yu
- Cancer Institute, The Affiliated Hospital of Qingdao University, Cancer Institute, Qingdao University, Qingdao, 266061, China
| | - Jie Lun
- Cancer Institute, The Affiliated Hospital of Qingdao University, Cancer Institute, Qingdao University, Qingdao, 266061, China
| | - Hongwei Zhang
- Shandong Provincial Maternal and Child Health Care Hospital, Jinan, 250014, China
| | - Lei Zhu
- Cancer Institute, The Affiliated Hospital of Qingdao University, Cancer Institute, Qingdao University, Qingdao, 266061, China
| | - Gang Zhang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Cancer Institute, Qingdao University, Qingdao, 266061, China.
| | - Jing Fang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Cancer Institute, Qingdao University, Qingdao, 266061, China.
| |
Collapse
|
35
|
Paredes F, Williams HC, San Martin A. Metabolic adaptation in hypoxia and cancer. Cancer Lett 2021; 502:133-142. [PMID: 33444690 PMCID: PMC8158653 DOI: 10.1016/j.canlet.2020.12.020] [Citation(s) in RCA: 116] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 12/09/2020] [Accepted: 12/11/2020] [Indexed: 12/17/2022]
Abstract
The ability of tumor cells to adapt to changes in oxygen tension is essential for tumor development. Low oxygen concentration influences cellular metabolism and, thus, affects proliferation, migration, and invasion. A focal point of the cell's adaptation to hypoxia is the transcription factor HIF1α (hypoxia-inducible factor 1 alpha), which affects the expression of specific gene networks involved in cellular energetics and metabolism. This review illustrates the mechanisms by which HIF1α-induced metabolic adaptation promotes angiogenesis, participates in the escape from immune recognition, and increases cancer cell antioxidant capacity. In addition to hypoxia, metabolic inhibition of 2-oxoglutarate-dependent dioxygenases regulates HIF1α stability and transcriptional activity. This phenomenon, known as pseudohypoxia, is frequently used by cancer cells to promote glycolytic metabolism to support biomass synthesis for cell growth and proliferation. In this review, we highlight the role of the most important metabolic intermediaries that are at the center of cancer's biology, and in particular, the participation of these metabolites in HIF1α retrograde signaling during the establishment of pseudohypoxia. Finally, we will discuss how these changes affect both the development of cancers and their resistance to treatment.
Collapse
Affiliation(s)
- Felipe Paredes
- Department of Medicine, Division of Cardiology, Emory University, Atlanta, GA, 30322, USA
| | - Holly C Williams
- Department of Medicine, Division of Cardiology, Emory University, Atlanta, GA, 30322, USA
| | - Alejandra San Martin
- Department of Medicine, Division of Cardiology, Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
36
|
Chowdhury R, Abboud MI, McAllister TE, Banerji B, Bhushan B, Sorensen JL, Kawamura A, Schofield CJ. Use of cyclic peptides to induce crystallization: case study with prolyl hydroxylase domain 2. Sci Rep 2020; 10:21964. [PMID: 33319810 PMCID: PMC7738489 DOI: 10.1038/s41598-020-76307-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 10/27/2020] [Indexed: 12/18/2022] Open
Abstract
Crystallization is the bottleneck in macromolecular crystallography; even when a protein crystallises, crystal packing often influences ligand-binding and protein-protein interaction interfaces, which are the key points of interest for functional and drug discovery studies. The human hypoxia-inducible factor prolyl hydroxylase 2 (PHD2) readily crystallises as a homotrimer, but with a sterically blocked active site. We explored strategies aimed at altering PHD2 crystal packing by protein modification and molecules that bind at its active site and elsewhere. Following the observation that, despite weak inhibition/binding in solution, succinamic acid derivatives readily enable PHD2 crystallization, we explored methods to induce crystallization without active site binding. Cyclic peptides obtained via mRNA display bind PHD2 tightly away from the active site. They efficiently enable PHD2 crystallization in different forms, both with/without substrates, apparently by promoting oligomerization involving binding to the C-terminal region. Although our work involves a specific case study, together with those of others, the results suggest that mRNA display-derived cyclic peptides may be useful in challenging protein crystallization cases.
Collapse
Affiliation(s)
- Rasheduzzaman Chowdhury
- Chemistry Research Laboratory, Department of Chemistry, University of Oxford, Oxford, OX1 3TA, UK
| | - Martine I Abboud
- Chemistry Research Laboratory, Department of Chemistry, University of Oxford, Oxford, OX1 3TA, UK
| | - Tom E McAllister
- Chemistry Research Laboratory, Department of Chemistry, University of Oxford, Oxford, OX1 3TA, UK
- Chemistry - School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK
| | - Biswadip Banerji
- Chemistry Research Laboratory, Department of Chemistry, University of Oxford, Oxford, OX1 3TA, UK
| | - Bhaskar Bhushan
- Chemistry Research Laboratory, Department of Chemistry, University of Oxford, Oxford, OX1 3TA, UK
| | - John L Sorensen
- Chemistry Research Laboratory, Department of Chemistry, University of Oxford, Oxford, OX1 3TA, UK
| | - Akane Kawamura
- Chemistry Research Laboratory, Department of Chemistry, University of Oxford, Oxford, OX1 3TA, UK
- Chemistry - School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK
| | - Christopher J Schofield
- Chemistry Research Laboratory, Department of Chemistry, University of Oxford, Oxford, OX1 3TA, UK.
| |
Collapse
|
37
|
Hypoxia and Oxygen-Sensing Signaling in Gene Regulation and Cancer Progression. Int J Mol Sci 2020; 21:ijms21218162. [PMID: 33142830 PMCID: PMC7663541 DOI: 10.3390/ijms21218162] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 10/28/2020] [Accepted: 10/29/2020] [Indexed: 12/19/2022] Open
Abstract
Oxygen homeostasis regulation is the most fundamental cellular process for adjusting physiological oxygen variations, and its irregularity leads to various human diseases, including cancer. Hypoxia is closely associated with cancer development, and hypoxia/oxygen-sensing signaling plays critical roles in the modulation of cancer progression. The key molecules of the hypoxia/oxygen-sensing signaling include the transcriptional regulator hypoxia-inducible factor (HIF) which widely controls oxygen responsive genes, the central members of the 2-oxoglutarate (2-OG)-dependent dioxygenases, such as prolyl hydroxylase (PHD or EglN), and an E3 ubiquitin ligase component for HIF degeneration called von Hippel–Lindau (encoding protein pVHL). In this review, we summarize the current knowledge about the canonical hypoxia signaling, HIF transcription factors, and pVHL. In addition, the role of 2-OG-dependent enzymes, such as DNA/RNA-modifying enzymes, JmjC domain-containing enzymes, and prolyl hydroxylases, in gene regulation of cancer progression, is specifically reviewed. We also discuss the therapeutic advancement of targeting hypoxia and oxygen sensing pathways in cancer.
Collapse
|
38
|
Dzhalilova D, Makarova O. Differences in Tolerance to Hypoxia: Physiological, Biochemical, and Molecular-Biological Characteristics. Biomedicines 2020; 8:E428. [PMID: 33080959 PMCID: PMC7603118 DOI: 10.3390/biomedicines8100428] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/14/2020] [Accepted: 10/15/2020] [Indexed: 02/07/2023] Open
Abstract
Hypoxia plays an important role in the development of many infectious, inflammatory, and tumor diseases. The predisposition to such disorders is mostly provided by differences in basic tolerance to oxygen deficiency, which we discuss in this review. Except the direct exposure of different-severity hypoxia in decompression chambers or in highland conditions, there are no alternative methods for determining organism tolerance. Due to the variability of the detection methods, differences in many parameters between tolerant and susceptible organisms are still not well-characterized, but some of them can serve as biomarkers of susceptibility to hypoxia. At the moment, several potential biomarkers in conditions after hypoxic exposure have been identified both in experimental animals and humans. The main potential biomarkers are Hypoxia-Inducible Factor (HIF)-1, Heat-Shock Protein 70 (HSP70), and NO. Due to the different mechanisms of various high-altitude diseases, biomarkers may not be highly specific and universal. Therefore, it is extremely important to conduct research on hypoxia susceptibility biomarkers. Moreover, it is important to develop a method for the evaluation of organisms' basic hypoxia tolerance without the necessity of any oxygen deficiency exposure. This can contribute to new personalized medicine approaches' development for diagnostics and the treatment of inflammatory and tumor diseases, taking into account hypoxia tolerance differences.
Collapse
Affiliation(s)
- Dzhuliia Dzhalilova
- Department of Immunomorphology of Inflammation, Federal State Budgetary Institution ‘Research Institute of Human Morphology’, Moscow 117418, Russia;
| | | |
Collapse
|
39
|
Smaila BD, Holland SD, Babaeijandaghi F, Henderson HG, Rossi FMV, Ramer MS. Systemic hypoxia mimicry enhances axonal regeneration and functional recovery following peripheral nerve injury. Exp Neurol 2020; 334:113436. [PMID: 32814068 DOI: 10.1016/j.expneurol.2020.113436] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/28/2020] [Accepted: 08/13/2020] [Indexed: 01/15/2023]
Abstract
Despite the ability of peripheral nerves to regenerate after injury, failure occurs due to an inability of supporting cells to maintain growth, resulting in long-term consequences such as sensorimotor dysfunction and neuropathic pain. Here, we investigate the potential of engaging the cellular adaptive response to hypoxia, via inhibiting its negative regulators, to enhance the regenerative process. Under normoxic conditions, prolyl hydroxylase domain (PHD) proteins 1, 2, and 3 hydroxylate the key metabolic regulator hypoxia inducible factor 1α (HIF1α), marking it for subsequent proteasomal degradation. We inhibited PHD protein function systemically via either individual genetic deletion or pharmacological pan-PHD inhibition using dimethyloxalylglycine (DMOG). We show enhanced axonal regeneration after sciatic nerve crush injury in PHD1-/- mice, PHD3-/- mice, and in DMOG-treated mice, and in PHD1-/- and DMOG-treated mice a reduction in hypersensitivity to cooling after permanent sciatic ligation. Electromyographically, PHD1-/- and PHD3-/- mice showed an increased CMAP amplitude one-month post-injury, probably due to protection against denervation induced muscle atrophy, while DMOG-treated and PHD2+/- mice showed reduced latencies, indicating improved motor axon function. DMOG treatment did not affect the growth of dorsal root ganglion neurites in vitro, suggesting a lack of direct effects of DMOG on axonal regrowth. Enhanced regeneration in vivo was concurrent with an increase in macrophage density, and a shift in macrophage polarization state ratios (from M1-like toward M2-like) in DMOG-treated animals. These results indicate PHD proteins as a novel therapeutic target to improve regenerative and functional outcomes after peripheral nerve injury without manipulating molecular O2.
Collapse
Affiliation(s)
- Brittney D Smaila
- International Collaboration on Repair Discoveries (ICORD), The University of British Columbia, 818 West 10(th) Ave, Vancouver, BC V5Z1M9, Canada
| | - Seth D Holland
- International Collaboration on Repair Discoveries (ICORD), The University of British Columbia, 818 West 10(th) Ave, Vancouver, BC V5Z1M9, Canada
| | - Farshad Babaeijandaghi
- The Biomedical Research Centre, The University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T1Z3, Canada
| | - Holly G Henderson
- International Collaboration on Repair Discoveries (ICORD), The University of British Columbia, 818 West 10(th) Ave, Vancouver, BC V5Z1M9, Canada
| | - Fabio M V Rossi
- The Biomedical Research Centre, The University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T1Z3, Canada
| | - Matt S Ramer
- International Collaboration on Repair Discoveries (ICORD), The University of British Columbia, 818 West 10(th) Ave, Vancouver, BC V5Z1M9, Canada.
| |
Collapse
|
40
|
Li X, Zhang Q, Nasser MI, Xu L, Zhang X, Zhu P, He Q, Zhao M. Oxygen homeostasis and cardiovascular disease: A role for HIF? Biomed Pharmacother 2020; 128:110338. [PMID: 32526454 DOI: 10.1016/j.biopha.2020.110338] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 05/27/2020] [Accepted: 05/30/2020] [Indexed: 12/17/2022] Open
Abstract
Hypoxia, the decline of tissue oxygen stress, plays a role in mediating cellular processes. Cardiovascular disease, relatively widespread with increased mortality, is closely correlated with oxygen homeostasis regulation. Besides, hypoxia-inducible factor-1(HIF-1) is reported to be a crucial component in regulating systemic hypoxia-induced physiological and pathological modifications like oxidative stress, damage, angiogenesis, vascular remodeling, inflammatory reaction, and metabolic remodeling. In addition, HIF1 controls the movement, proliferation, apoptosis, differentiation and activity of numerous core cells, such as cardiomyocytes, endothelial cells (ECs), smooth muscle cells (SMCs), and macrophages. Here we review the molecular regulation of HIF-1 in cardiovascular diseases, intended to improve therapeutic approaches for clinical diagnoses. Better knowledge of the oxygen balance control and the signal mechanisms involved is important to advance the development of hypoxia-related diseases.
Collapse
Affiliation(s)
- Xinyu Li
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province 410013, China; Xiangya School of Medicine, Central South University, Changsha, Hunan Province 410013, China
| | - Quyan Zhang
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province 410013, China; Xiangya School of Medicine, Central South University, Changsha, Hunan Province 410013, China
| | - M I Nasser
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510100, China
| | - Linyong Xu
- Xiangya School of Life Science, Central South University, Changsha, Hunan Province 410013, China
| | - Xueyan Zhang
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province 410013, China; Xiangya School of Medicine, Central South University, Changsha, Hunan Province 410013, China
| | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510100, China.
| | - Qingnan He
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province 410013, China.
| | - Mingyi Zhao
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province 410013, China.
| |
Collapse
|
41
|
Zhang Q, Guo D, Wang Y, Wang X, Wang Q, Wu Y, Li C, Wang W, Wang Y. Danqi Pill Protects Against Heart Failure Post-Acute Myocardial Infarction via HIF-1α/PGC-1α Mediated Glucose Metabolism Pathway. Front Pharmacol 2020; 11:458. [PMID: 32372956 PMCID: PMC7187888 DOI: 10.3389/fphar.2020.00458] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 03/24/2020] [Indexed: 12/28/2022] Open
Abstract
AIM Heart failure (HF) post-acute myocardial infarction (AMI) leads to a large number of hospitalizations and deaths worldwide. Danqi pill (DQP) is included in the 2015 national pharmacopoeia and widely applied in the treatment of HF in clinics in China. We examined whether DQP acted on glucose metabolism to protect against HF post-AMI via hypoxia inducible factor-1 alpha (HIF-1α)/peroxisome proliferator-activated receptor α co-activator (PGC-1α) pathway. METHODS AND RESULTS In this study, left anterior descending (LAD) artery ligation induced HF post-AMI rats and oxygen-glucose deprivation-reperfusion (OGD/R)-induced H9C2 cell model were structured to explore the efficacy and mechanism of DQP. Here we showed that DQP protected the heart against ischemic damage as evidenced by improved cardiac functions and attenuated inflammatory infiltration. The expressions of critical proteins involved in glucose intake and transportation such as GLUT4 and PKM2 were up-regulated, while negative regulatory proteins involved in oxidative phosphorylation were attenuated in the treatment of DQP. Moreover, DQP up-regulated NRF1 and TFAM, promoted mitochondrial biogenesis and increased myocardial adenosine triphosphate (ATP) level. The protection effects of DQP were significantly compromised by HIF-1α siRNA, suggesting that HIF-1α signaling pathway was the potential target of DQP on HF post-AMI. CONCLUSIONS DQP exhibits the efficacy to improve myocardial glucose metabolism, mitochondrial oxidative phosphorylation and biogenesis by regulating HIF-1α/PGC-1α signaling pathway in HF post-AMI rats.
Collapse
Affiliation(s)
- Qian Zhang
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Dongqing Guo
- The School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Yuanyuan Wang
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoping Wang
- The School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Qiyan Wang
- The School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Yan Wu
- Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Chun Li
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Wei Wang
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yong Wang
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- The School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
42
|
Rajendran G, Schonfeld MP, Tiwari R, Huang S, Torosyan R, Fields T, Park J, Susztak K, Kapitsinou PP. Inhibition of Endothelial PHD2 Suppresses Post-Ischemic Kidney Inflammation through Hypoxia-Inducible Factor-1. J Am Soc Nephrol 2020; 31:501-516. [PMID: 31996410 PMCID: PMC7062211 DOI: 10.1681/asn.2019050523] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 11/15/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Prolyl-4-hydroxylase domain-containing proteins 1-3 (PHD1 to PHD3) regulate the activity of the hypoxia-inducible factors (HIFs) HIF-1 and HIF-2, transcription factors that are key regulators of hypoxic vascular responses. We previously reported that deficiency of endothelial HIF-2 exacerbated renal ischemia-reperfusion injury, whereas inactivation of endothelial PHD2, the main oxygen sensor, provided renoprotection. Nevertheless, the molecular mechanisms by which endothelial PHD2 dictates AKI outcomes remain undefined. METHODS To investigate the function of the endothelial PHD2/HIF axis in ischemic AKI, we examined the effects of endothelial-specific ablation of PHD2 in a mouse model of renal ischemia-reperfusion injury. We also interrogated the contribution of each HIF isoform by concurrent endothelial deletion of both PHD2 and HIF-1 or both PHD2 and HIF-2. RESULTS Endothelial deletion of Phd2 preserved kidney function and limited transition to CKD. Mechanistically, we found that endothelial Phd2 ablation protected against renal ischemia-reperfusion injury by suppressing the expression of proinflammatory genes and recruitment of inflammatory cells in a manner that was dependent on HIF-1 but not HIF-2. Persistence of renoprotective responses after acute inducible endothelial-specific loss of Phd2 in adult mice ruled out a requirement for PHD2 signaling in hematopoietic cells. Although Phd2 inhibition was not sufficient to induce detectable HIF activity in the kidney endothelium, in vitro experiments implicated a humoral factor in the anti-inflammatory effects generated by endothelial PHD2/HIF-1 signaling. CONCLUSIONS Our findings suggest that activation of endothelial HIF-1 signaling through PHD2 inhibition may offer a novel therapeutic approach against ischemic AKI.
Collapse
Affiliation(s)
- Ganeshkumar Rajendran
- Department of Medicine, Anatomy and Cell Biology and
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas; and
| | - Michael P Schonfeld
- Department of Medicine, Anatomy and Cell Biology and
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas; and
| | - Ratnakar Tiwari
- Department of Medicine, Anatomy and Cell Biology and
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas; and
| | - Shengping Huang
- Department of Medicine, Anatomy and Cell Biology and
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas; and
| | - Rafael Torosyan
- Department of Medicine, Anatomy and Cell Biology and
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas; and
| | - Timothy Fields
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas; and
| | - Jihwan Park
- Renal Electrolyte and Hypertension Division, Department of Medicine and Genetics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Katalin Susztak
- Renal Electrolyte and Hypertension Division, Department of Medicine and Genetics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Pinelopi P Kapitsinou
- Department of Medicine, Anatomy and Cell Biology and
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas; and
| |
Collapse
|
43
|
Romanelli-Credrez L, Doitsidou M, Alkema MJ, Salinas G. HIF-1 Has a Central Role in Caenorhabditis elegans Organismal Response to Selenium. Front Genet 2020; 11:63. [PMID: 32161616 PMCID: PMC7052493 DOI: 10.3389/fgene.2020.00063] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 01/17/2020] [Indexed: 11/13/2022] Open
Abstract
Selenium is a trace element for most organisms; its deficiency and excess are detrimental. Selenium beneficial effects are mainly due to the role of the 21st genetically encoded amino acid selenocysteine (Sec). Selenium also exerts Sec-independent beneficial effects. Its harmful effects are thought to be mainly due to non-specific incorporation in protein synthesis. Yet the selenium response in animals is poorly understood. In Caenorhabditis elegans, Sec is genetically incorporated into a single selenoprotein. Similar to mammals, a 20-fold excess of the optimal selenium requirement is harmful. Sodium selenite (Na2SeO3) excess causes development retardation, impaired growth, and neurodegeneration of motor neurons. To study the organismal response to selenium we performed a genetic screen for C. elegans mutants that are resistant to selenite. We isolated non-sense and missense egl-9/EGLN mutants that confer robust resistance to selenium. In contrast, hif-1/HIF null mutant was highly sensitive to selenium, establishing a role for this transcription factor in the selenium response. We showed that EGL-9 regulates HIF-1 activity through VHL-1, and identified CYSL-1 as a key sensor that transduces the selenium signal. Finally, we showed that the key enzymes involved in sulfide and sulfite stress (sulfide quinone oxidoreductase and sulfite oxidase) are not required for selenium resistance. In contrast, knockout strains in the persulfide dioxygenase ETHE-1 and the sulfurtransferase MPST-7 affect the organismal response to selenium. In sum, our results identified a transcriptional pathway as well as enzymes possibly involved in the organismal selenium response.
Collapse
Affiliation(s)
- Laura Romanelli-Credrez
- Laboratorio de Biología de Gusanos. Unidad Mixta, Departamento de Biociencias, Facultad de Química, Universidad de la República-Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Maria Doitsidou
- Centre for Discovery Brain Sciences (CDBS), University of Edinburgh, Edinburgh, United Kingdom
| | - Mark J Alkema
- Neurobiology Department, University of Massachusetts Medical School, Worcester, MA, United States
| | - Gustavo Salinas
- Laboratorio de Biología de Gusanos. Unidad Mixta, Departamento de Biociencias, Facultad de Química, Universidad de la República-Institut Pasteur de Montevideo, Montevideo, Uruguay
| |
Collapse
|
44
|
Bartman CM, Eckle T. Circadian-Hypoxia Link and its Potential for Treatment of Cardiovascular Disease. Curr Pharm Des 2020; 25:1075-1090. [PMID: 31096895 DOI: 10.2174/1381612825666190516081612] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 05/03/2019] [Indexed: 12/29/2022]
Abstract
Throughout the evolutionary time, all organisms and species on Earth evolved with an adaptation to consistent oscillations of sunlight and darkness, now recognized as 'circadian rhythm.' Single-cellular to multisystem organisms use circadian biology to synchronize to the external environment and provide predictive adaptation to changes in cellular homeostasis. Dysregulation of circadian biology has been implicated in numerous prevalent human diseases, and subsequently targeting the circadian machinery may provide innovative preventative or treatment strategies. Discovery of 'peripheral circadian clocks' unleashed widespread investigations into the potential roles of clock biology in cellular, tissue, and organ function in healthy and diseased states. Particularly, oxygen-sensing pathways (e.g. hypoxia inducible factor, HIF1), are critical for adaptation to changes in oxygen availability in diseases such as myocardial ischemia. Recent investigations have identified a connection between the circadian rhythm protein Period 2 (PER2) and HIF1A that may elucidate an evolutionarily conserved cellular network that can be targeted to manipulate metabolic function in stressed conditions like hypoxia or ischemia. Understanding the link between circadian and hypoxia pathways may provide insights and subsequent innovative therapeutic strategies for patients with myocardial ischemia. This review addresses our current understanding of the connection between light-sensing pathways (PER2), and oxygen-sensing pathways (HIF1A), in the context of myocardial ischemia and lays the groundwork for future studies to take advantage of these two evolutionarily conserved pathways in the treatment of myocardial ischemia.
Collapse
Affiliation(s)
- Colleen Marie Bartman
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, Graduate Training Program in Cell Biology, Stem Cells, and Development, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Tobias Eckle
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, Graduate Training Program in Cell Biology, Stem Cells, and Development, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
45
|
Vanderhaeghen T, Vandewalle J, Libert C. Hypoxia-inducible factors in metabolic reprogramming during sepsis. FEBS J 2020; 287:1478-1495. [PMID: 31970890 DOI: 10.1111/febs.15222] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 12/20/2019] [Accepted: 01/20/2020] [Indexed: 12/15/2022]
Abstract
Sepsis is a highly heterogeneous syndrome that is caused by an imbalanced host response to infection. Despite huge investments, sepsis remains a contemporary threat with significant burden on health systems. Vascular dysfunction and elevated oxygen consumption by highly metabolically active immune cells result in tissue hypoxia during inflammation. The transcription factor hypoxia-inducible factor-1a (HIF1α), and its family members, plays an important role in cellular metabolism and adaptation to cellular stress caused by hypoxia. In this review, we discuss the role of HIF in sepsis. We show possible mechanisms by which the inflammatory response activated during sepsis affects the HIF pathway. The activated HIF pathway in turn changes the metabolism of both innate and adaptive immune cells. As HIF expression in leukocytes of septic patients can be directly linked with mortality, we discuss multiple ways of interfering with the HIF signaling pathway.
Collapse
Affiliation(s)
- Tineke Vanderhaeghen
- Center for Inflammation Research, VIB, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Belgium
| | - Jolien Vandewalle
- Center for Inflammation Research, VIB, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Belgium
| | - Claude Libert
- Center for Inflammation Research, VIB, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Belgium
| |
Collapse
|
46
|
D'Hulst G, Soro-Arnaiz I, Masschelein E, Veys K, Fitzgerald G, Smeuninx B, Kim S, Deldicque L, Blaauw B, Carmeliet P, Breen L, Koivunen P, Zhao SM, De Bock K. PHD1 controls muscle mTORC1 in a hydroxylation-independent manner by stabilizing leucyl tRNA synthetase. Nat Commun 2020; 11:174. [PMID: 31924757 PMCID: PMC6954236 DOI: 10.1038/s41467-019-13889-6] [Citation(s) in RCA: 297] [Impact Index Per Article: 59.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 12/06/2019] [Indexed: 02/08/2023] Open
Abstract
mTORC1 is an important regulator of muscle mass but how it is modulated by oxygen and nutrients is not completely understood. We show that loss of the prolyl hydroxylase domain isoform 1 oxygen sensor in mice (PHD1KO) reduces muscle mass. PHD1KO muscles show impaired mTORC1 activation in response to leucine whereas mTORC1 activation by growth factors or eccentric contractions was preserved. The ability of PHD1 to promote mTORC1 activity is independent of its hydroxylation activity but is caused by decreased protein content of the leucyl tRNA synthetase (LRS) leucine sensor. Mechanistically, PHD1 interacts with and stabilizes LRS. This interaction is promoted during oxygen and amino acid depletion and protects LRS from degradation. Finally, elderly subjects have lower PHD1 levels and LRS activity in muscle from aged versus young human subjects. In conclusion, PHD1 ensures an optimal mTORC1 response to leucine after episodes of metabolic scarcity. mTORC1 is an important regulator of muscle mass. Here, the authors show that the PHD1 controls muscle mass in a hydroxylation-independent manner. PHD1 prevents the degradation of leucine sensor LRS during oxygen and amino acid depletion to ensure effective mTORC1 activation in response to leucine.
Collapse
Affiliation(s)
- Gommaar D'Hulst
- Department Health Sciences and Technology, Laboratory of Exercise and Health, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Inés Soro-Arnaiz
- Department Health Sciences and Technology, Laboratory of Exercise and Health, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Evi Masschelein
- Department Health Sciences and Technology, Laboratory of Exercise and Health, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Koen Veys
- Laboratory of Angiogenesis and Vascular Metabolism, VIB Center for Cancer Biology (CCB), VIB, Leuven, Belgium.,Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Gillian Fitzgerald
- Department Health Sciences and Technology, Laboratory of Exercise and Health, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Benoit Smeuninx
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.,MRC Arthritis Research UK Centre for Musculoskeletal Ageing Research, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Sunghoon Kim
- Medicinal Bioconvergence Research Center, College of Pharmacy, Seoul National University, Gwanak-gu, Seoul, South Korea
| | - Louise Deldicque
- Institute of Neuroscience, Université catholique de Louvain, Louvain-La-Neuve, Belgium
| | - Bert Blaauw
- Department of Biomedical Sciences, Venetian Institute of Molecular Medicine, University of Padova, Padova, Italy
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, VIB Center for Cancer Biology (CCB), VIB, Leuven, Belgium.,Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Leigh Breen
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.,MRC Arthritis Research UK Centre for Musculoskeletal Ageing Research, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Peppi Koivunen
- Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine, Oulu Center for Cell-Matrix Research, University of Oulu, Oulu, Finland
| | - Shi-Min Zhao
- Obstetrics and Gynaecology Hospital of Fudan University, State Key Lab of Genetic Engineering, School of Life Sciences and Institutes of Biomedical Sciences, Shanghai, P. R. China.,Institute of Biomedical Science, Fudan University, Shanghai, P. R. China.,Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, P. R. China
| | - Katrien De Bock
- Department Health Sciences and Technology, Laboratory of Exercise and Health, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland.
| |
Collapse
|
47
|
Luo A, Chen Y. Label-Free Interactome Analysis Revealed an Essential Role of CUL3-KEAP1 Complex in Mediating the Ubiquitination and Degradation of PHD2. J Proteome Res 2020; 19:260-268. [PMID: 31763849 DOI: 10.1021/acs.jproteome.9b00513] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Prolyl hydroxylase domain-containing protein 2 (PHD2/EGLN1) is a key regulatory enzyme that plays a fundamental role in the cellular hypoxic response pathway, mediating proline hydroxylation-dependent protein degradation of selected target proteins. However, the regulation of PHD2 homeostasis at the protein level is not well understood. Here, we perform label-free quantitative interactome analysis through immunoprecipitation coupled with mass spectrometry analysis. To minimize the side effects caused by ectopic overexpression, in HeLa cells, we stably overexpressed Flag-tagged PHD2 while suppressing the endogenous PHD2 by using an shRNA targeting its 3' UTR region. We identified and validated Cullin 3 as a novel PHD2 interactor in vivo. Through candidate screening, we further identified CUL3-KEAP1 E3 ubiquitin ligase complex as the major enzyme that regulates PHD2 degradation. Overexpression of either CUL3, KEAP1, or both significantly increases PHD2 ubiquitination and reduces PHD2 protein abundance. The knockdown of CUL3 or KEAP1 decreased PHD2 ubiquitination and inhibited PHD2 degradation. Accordingly, loss of the CUL3-KEAP1 complex under hypoxia promoted PHD2 stabilization and led to significantly reduced abundance of the PHD2 target, hypoxia-inducible factor 1A (HIF1A). Thus, CUL3-KEAP1 is an essential pathway that regulates PHD2 ubiquitination and degradation in cells.
Collapse
Affiliation(s)
- Ang Luo
- Department of Biochemistry, Molecular Biology and Biophysics , The University of Minnesota at Twin Cities , Minneapolis , Minnesota 55455 , United States
| | - Yue Chen
- Department of Biochemistry, Molecular Biology and Biophysics , The University of Minnesota at Twin Cities , Minneapolis , Minnesota 55455 , United States
| |
Collapse
|
48
|
Erber L, Luo A, Chen Y. Targeted and Interactome Proteomics Revealed the Role of PHD2 in Regulating BRD4 Proline Hydroxylation. Mol Cell Proteomics 2019; 18:1772-1781. [PMID: 31239290 PMCID: PMC6731074 DOI: 10.1074/mcp.ra119.001535] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 06/19/2019] [Indexed: 12/18/2022] Open
Abstract
Proline hydroxylation is a critical cellular mechanism regulating energy homeostasis and development. Our previous study identified and validated Bromodomain-containing protein 4 (BRD4) as a proline hydroxylation substrate in cancer cells. Yet, the regulatory mechanism and the functional significance of the modification remain unknown. In this study, we developed targeted quantification assays using parallel-reaction monitoring and biochemical analysis to identify the major regulatory enzyme of BRD4 proline hydroxylation. We further performed quantitative interactome analysis to determine the functional significance of the modification pathway in BRD4-mediated protein-protein interactions and gene transcription. Our findings revealed that PHD2 is the key regulatory enzyme of BRD4 proline hydroxylation and the modification significantly affects BRD4 interactions with key transcription factors as well as BRD4-mediated transcriptional activation. Taken together, this study provided mechanistic insights into the oxygen-dependent modification of BRD4 and revealed new roles of the pathway in regulating BRD4-dependent gene expression.
Collapse
Affiliation(s)
- Luke Erber
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455
| | - Ang Luo
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455
| | - Yue Chen
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455.
| |
Collapse
|
49
|
Strowitzki MJ, Ritter AS, Kimmer G, Schneider M. Hypoxia-adaptive pathways: A pharmacological target in fibrotic disease? Pharmacol Res 2019; 147:104364. [PMID: 31376431 DOI: 10.1016/j.phrs.2019.104364] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 07/19/2019] [Accepted: 07/19/2019] [Indexed: 02/07/2023]
Abstract
Wound healing responses are physiological reactions to injuries and share common characteristics and phases independently of the injured organ or tissue. A major hallmark of wound healing responses is the formation of extra-cellular matrix (ECM), mainly consisting of collagen fibers, to restore the initial organ architecture and function. Overshooting wound healing responses result in unphysiological accumulation of ECM and collagen deposition, a process called fibrosis. Importantly, hypoxia (oxygen demand exceeds supply) plays a significant role during wound healing responses and fibrotic diseases. Under hypoxic conditions, cells activate a gene program, including the stabilization of hypoxia-inducible factors (HIFs), which induces the expression of HIF target genes counteracting hypoxia. In contrast, in normoxia, so-called HIF-prolyl hydroxylases (PHDs) oxygen-dependently hydroxylate HIF-α, which marks it for proteasomal degradation. Importantly, PHDs can be pharmacologically inhibited (PHI) by so-called PHD inhibitors. There is mounting evidence that the HIF-pathway is continuously up-regulated during the development of tissue fibrosis, and that pharmacological (HIFI) or genetic inhibition of HIF can prevent organ fibrosis. By contrast, initial (short-term) activation of the HIF pathway via PHI during wound healing seems to be beneficial in several models of inflammation or acute organ injury. Thus, timing and duration of PHI and HIFI treatment seem to be crucial. In this review, we will highlight the role of hypoxia-adaptive pathways during wound healing responses and development of fibrotic disease. Moreover, we will discuss whether PHI and HIFI might be a promising treatment option in fibrotic disease, and consider putative pitfalls that might result from this approach.
Collapse
Affiliation(s)
- Moritz J Strowitzki
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Alina S Ritter
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Gwendolyn Kimmer
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Martin Schneider
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany.
| |
Collapse
|
50
|
Roy IM, Biswas A, Verfaillie C, Khurana S. Energy Producing Metabolic Pathways in Functional Regulation of the Hematopoietic Stem Cells. IUBMB Life 2019; 70:612-624. [PMID: 29999238 DOI: 10.1002/iub.1870] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 04/20/2018] [Indexed: 02/06/2023]
Abstract
The hematopoietic system has a very well-studied hierarchy with the long-term (LT) hematopoietic stem cells (HSCs) taking the top position. The pool of quiescent adult LT-HSCs generated during the fetal and early postnatal life acts as a reservoir to supply all the blood cells. Therefore, the maintenance of this stem cell pool is pivotal to maintaining homeostasis in hematopoietic system. It has long been known that external cues, along with the internal genetic factors influence the status of HSCs in the bone marrow (BM). Hypoxia is one such factor that regulates the vascular as well as hematopoietic ontogeny from a very early time point in development. The metabolic outcomes of a hypoxic microenvironment play important roles in functional regulation of HSCs, especially in case of adult BM HSCs. Anaerobic metabolic pathways therefore perform prominent role in meeting energy demands. Increased oxidative pathways on the other hand result in loss of stemness. Recent studies have attributed the functional differences in HSCs across different life stages to their metabolic phenotypes regulated by respective niches. Indicating thus, that various energy production pathways could play distinct role in regulating HSC function at different developmental/physiological states. Here, we review the current status of our understanding over the role that energy production pathways play in regulating HSC stemness. © 2018 IUBMB Life, 70(7):612-624, 2018.
Collapse
Affiliation(s)
- Irene M Roy
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, Kerala, India
| | - Atreyi Biswas
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, Kerala, India
| | | | - Satish Khurana
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, Kerala, India
| |
Collapse
|