1
|
Liu Y, Wu Z, Li Y, Chen Y, Zhao X, Wu M, Xia Y. Metabolic reprogramming and interventions in angiogenesis. J Adv Res 2025; 70:323-338. [PMID: 38704087 PMCID: PMC11976431 DOI: 10.1016/j.jare.2024.05.001] [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: 03/15/2024] [Revised: 04/30/2024] [Accepted: 05/01/2024] [Indexed: 05/06/2024] Open
Abstract
BACKGROUND Endothelial cell (EC) metabolism plays a crucial role in the process of angiogenesis. Intrinsic metabolic events such as glycolysis, fatty acid oxidation, and glutamine metabolism, support secure vascular migration and proliferation, energy and biomass production, as well as redox homeostasis maintenance during vessel formation. Nevertheless, perturbation of EC metabolism instigates vascular dysregulation-associated diseases, especially cancer. AIM OF REVIEW In this review, we aim to discuss the metabolic regulation of angiogenesis by EC metabolites and metabolic enzymes, as well as prospect the possible therapeutic opportunities and strategies targeting EC metabolism. KEY SCIENTIFIC CONCEPTS OF REVIEW In this work, we discuss various aspects of EC metabolism considering normal and diseased vasculature. Of relevance, we highlight that the implications of EC metabolism-targeted intervention (chiefly by metabolic enzymes or metabolites) could be harnessed in orchestrating a spectrum of pathological angiogenesis-associated diseases.
Collapse
Affiliation(s)
- Yun Liu
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Zifang Wu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yikun Li
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China; College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Yating Chen
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Xuan Zhao
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China.
| | - Miaomiao Wu
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan 410128, China.
| | - Yaoyao Xia
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China.
| |
Collapse
|
2
|
Luo Z, Wang Q, Fan X, Koh XQ, Loh XJ, Wu C, Li Z, Wu YL. ROS-Driven Nanoventilator for MRSA-Induced Acute Lung Injury Treatment via In Situ Oxygen Supply, Anti-Inflammation and Immunomodulation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2406060. [PMID: 40106334 DOI: 10.1002/advs.202406060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 08/06/2024] [Indexed: 03/22/2025]
Abstract
Hypoxia, inflammatory response and pathogen (bacterial or viral) infection are the three main factors that lead to death in patients with acute lung injury (ALI). Among them, hypoxia activates the expression of HIF-1α, further exacerbating the production of ROS and inflammatory response. Currently, anti-inflammatory and pathogen elimination treatment strategies have effectively alleviated infectious pneumonia, but improving lung hypoxia still faces challenges. Here, a vancomycin-loaded nanoventilator (SCVN) containing superoxide dismutase (SOD) and catalase (CAT) is developed, which is prepared by encapsulating SOD, CAT and vancomycin into a nanocapsule by in situ polymerization. This nanocapsule can effectively improve the stability and loading rate of enzymes, and enhance their enzyme cascade efficiency, thereby efficiently consuming •O2 - and H2O2 to generate O2 in situ and reducing ROS level. More interestingly, in situ O2 supply can effectively relieve lung hypoxia to reduce HIF-1α expression and balance the number of M1/M2 macrophages to reduce the levels of TNF-α, IL-1β and IL-6, thereby alleviating the inflammatory response. Meanwhile, vancomycin can target and kill MRSA, fundamentally solving the cause of pneumonia. This nanoventilator with antibacterial, anti-inflammatory, ROS scavenging and in situ O2 supply functions will provide a universal clinical treatment strategy for ALI caused by pathogens.
Collapse
Affiliation(s)
- Zheng Luo
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore, 138634, Republic of Singapore
| | - Qi Wang
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Xiaotong Fan
- Institute of Sustainability for Chemicals, Energy and Environment (ISCE2), Agency for Science, Technology and Research (A*STAR), 1 Pesek Road, Jurong Island, Singapore, 627833, Republic of Singapore
| | - Xue Qi Koh
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore, 138634, Republic of Singapore
| | - Xian Jun Loh
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore, 138634, Republic of Singapore
| | - Caisheng Wu
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Zibiao Li
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore, 138634, Republic of Singapore
- Institute of Sustainability for Chemicals, Energy and Environment (ISCE2), Agency for Science, Technology and Research (A*STAR), 1 Pesek Road, Jurong Island, Singapore, 627833, Republic of Singapore
- Department of Materials Science and Engineering, National University of Singapore, Singapore, 117576, Republic of Singapore
| | - Yun-Long Wu
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| |
Collapse
|
3
|
Jin Y, Liu Z, Yang Z, Fang L, Zhao FQ, Liu H. Effects of hypoxia stress on the milk synthesis in bovine mammary epithelial cells. J Anim Sci Biotechnol 2025; 16:37. [PMID: 40050971 PMCID: PMC11887346 DOI: 10.1186/s40104-025-01174-0] [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: 10/05/2024] [Accepted: 02/05/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Milk synthesis is an energy-intensive process influenced by oxygen availability. This study investigates how hypoxia affects milk synthesis in BMECs, focusing on key genes involved in lactation and energy metabolism. METHODS BMECs were cultured in a normoxic environment and then transferred to a hypoxia chamber with 1% O2 for specified durations. The study evaluated cellular responses through various molecular experiments and RNA sequencing. Small interfering RNA was employed to knock down HIF-1α to investigate whether the lactation-related phenotype alteration depends on HIF-1α. RESULTS Hypoxia disrupted milk protein production by reducing mTOR/P70S6K/4EBP1 signaling and downregulating genes critical for amino acid transport and protein synthesis. Triglyceride synthesis increased due to enhanced fatty acid uptake and the upregulation of regulatory proteins, including FASN and PPARγ. Although glucose uptake was elevated under hypoxia, key enzymes for lactose synthesis were downregulated, suggesting a redirection of glucose toward energy production. Mitochondrial function was impaired under hypoxia, with reduced gene expression in TCA cycle, ETC, cytosol-mitochondrial transport, decreased ATP levels, increased ROS levels, and structural alterations. Additionally, lipid synthesis and glucose uptake depend on HIF-1α, while milk protein synthesis alterations occurred independently of HIF-1α. CONCLUSIONS Hypoxia alters milk synthesis in BMECs by disrupting milk protein synthesis, enhancing lipid metabolism, and impairing energy production. These findings provide valuable insights into the molecular mechanisms underlying the effect of oxygen deprivation on lactation efficiency, offering potential targets for mitigating hypoxic stress in the mammary glands of dairy animals.
Collapse
Affiliation(s)
- Yanshan Jin
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Zhuolin Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Ziyan Yang
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Lizhu Fang
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Feng-Qi Zhao
- Department of Animal and Veterinary Sciences, University of Vermont, Burlington, VT, 05405, USA
| | - Hongyun Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
4
|
Tidma S, Sutton GD, Dang LTM, Jiang C, Teets TS. Dual-emitting cyclometalated Ir(III) complexes with salicylaldimine-bound fluorophores for ratiometric oxygen sensing. Chem Commun (Camb) 2024; 60:11572-11575. [PMID: 39315650 DOI: 10.1039/d4cc03220a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
A new class of oxygen-sensing dual-emitting cyclometalated Ir(III) complexes is described. They function as ratiometric sensors that combine the blue fluorescence from coumarin as a self-referenced internal standard with yellow to red phosphorescence from bis-cyclometalated iridium complexes. The compounds have phosphorescence quantum yields up to 15%, lifetimes ranging from 0.23 to 9.4 μs, and are capable of sensing a wide range of O2 partial pressures.
Collapse
Affiliation(s)
- Sirawit Tidma
- Department of Chemistry, University of Houston, 3585 Cullen Blvd. Room 112, Houston, Tx, 77204-5003, USA.
| | - Gregory D Sutton
- Department of Chemistry, University of Houston, 3585 Cullen Blvd. Room 112, Houston, Tx, 77204-5003, USA.
| | - Linh T M Dang
- Department of Chemistry, University of Houston, 3585 Cullen Blvd. Room 112, Houston, Tx, 77204-5003, USA.
| | - Chenggang Jiang
- Department of Chemistry, University of Houston, 3585 Cullen Blvd. Room 112, Houston, Tx, 77204-5003, USA.
| | - Thomas S Teets
- Department of Chemistry, University of Houston, 3585 Cullen Blvd. Room 112, Houston, Tx, 77204-5003, USA.
| |
Collapse
|
5
|
Zheng S, Jiang J, Shu Z, Qiu C, Jiang L, Zhao N, Lin X, Qian Y, Liang B, Qiu L. Fine particulate matter (PM 2.5) induces testosterone disruption by triggering ferroptosis through SIRT1/HIF-1α signaling pathway in male mice. Free Radic Biol Med 2024; 221:40-51. [PMID: 38759901 DOI: 10.1016/j.freeradbiomed.2024.05.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 05/10/2024] [Accepted: 05/13/2024] [Indexed: 05/19/2024]
Abstract
Fine particulate matter (PM2.5), a significant component of air pollution particulate matter, is inevitable and closely associated with increasing male reproductive disorder. However, the testicular targets of PM2.5 and its toxicity related molecular mechanisms are still not fully understood. In this study, the conditional knockout (cKO) mice and primary Leydig cells were used to explore the testicular targets of PM2.5 and the related underlying mechanisms. First, apparent the structure impairment of seminiferous tubules, Leydig cells vacuolization, decline of serum testosterone and sperm quality reduction were found in male wild-type (WT) and Sirt1 knockout mice after exposure to PM2.5. Enrichment analyses revealed that differentially expressed genes (DEGs) were enriched in steroid hormone biosynthesis, ferroptosis, and HIF-1 signaling pathway in the mice testes after exposure to PM2.5, which were subsequently verified by the molecular biological analyses. Notably, similar enrichment analyses results were also observed in primary Leydig cells after treatment with PM2.5. In addition, Knockdown of Sirt1 significantly increased PM2.5-induced expression and activation of HIF-1α, which was in parallel to the changes of cellular iron levels, oxidative stress indicators and the ferroptosis markers. In conclusion, this highlights that PM2.5 triggers ferroptosis via SIRT1/HIF-1α signaling pathway to inhibit testosterone synthesis in males. These findings provide a novel research support for the study that PM2.5 causes male reproductive injury.
Collapse
Affiliation(s)
- Shaokai Zheng
- School of Public Health, Nantong University, 9 Seyuan Rd, Nantong, 226019, PR China
| | - Jinchen Jiang
- School of Public Health, Nantong University, 9 Seyuan Rd, Nantong, 226019, PR China
| | - Zhenhao Shu
- School of Public Health, Nantong University, 9 Seyuan Rd, Nantong, 226019, PR China
| | - Chong Qiu
- Medical School, Nantong University, 19 Qixiu Rd, Nantong, 226001, PR China
| | - Lianlian Jiang
- School of Public Health, Nantong University, 9 Seyuan Rd, Nantong, 226019, PR China
| | - Nannan Zhao
- School of Public Health, Nantong University, 9 Seyuan Rd, Nantong, 226019, PR China
| | - Xiaojun Lin
- School of Public Health, Nantong University, 9 Seyuan Rd, Nantong, 226019, PR China
| | - Yingyun Qian
- Graduate School, Nantong University, 9 Seyuan Rd, Nantong, 226019, PR China
| | - Bo Liang
- Department of Ultrasound, Affiliated Hospital 2 of Nantong University, 6 Hai'er Lane North Rd, Nantong, 226019, PR China.
| | - Lianglin Qiu
- School of Public Health, Nantong University, 9 Seyuan Rd, Nantong, 226019, PR China.
| |
Collapse
|
6
|
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
|
7
|
Huynh GT, Tunny SS, Frith JE, Meagher L, Corrie SR. Organosilica Nanosensors for Monitoring Spatiotemporal Changes in Oxygen Levels in Bacterial Cultures. ACS Sens 2024; 9:2383-2394. [PMID: 38687178 DOI: 10.1021/acssensors.3c02747] [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] [Indexed: 05/02/2024]
Abstract
Oxygen plays a central role in aerobic metabolism, and while many approaches have been developed to measure oxygen concentration in biological environments over time, monitoring spatiotemporal changes in dissolved oxygen levels remains challenging. To address this, we developed a ratiometric core-shell organosilica nanosensor for continuous, real-time optical monitoring of oxygen levels in biological environments. The nanosensors demonstrate good steady state characteristics (KpSV = 0.40 L/mg, R2 = 0.95) and respond reversibly to changes in oxygen concentration in buffered solutions and report similar oxygen level changes in response to bacterial cell growth (Escherichia coli) in comparison to a commercial bulk optode-based sensing film. We further demonstrated that the oxygen nanosensors could be distributed within a growing culture of E. coli and used to record oxygen levels over time and in different locations within a static culture, opening the possibility of spatiotemporal monitoring in complex biological systems.
Collapse
Affiliation(s)
- Gabriel T Huynh
- Department of Chemical and Biological Engineering, Monash University, Clayton, VIC 3800, Australia
- Commonwealth Scientific and Industrial Research Organization (CSIRO) Manufacturing, Clayton, VIC 3168, Australia
| | - Salma S Tunny
- Department of Chemical and Biological Engineering, Monash University, Clayton, VIC 3800, Australia
| | - Jessica E Frith
- Department of Materials Science and Engineering, Monash University, Clayton, VIC 3800, Australia
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia
- ARC Training Centre for Cell and Tissue Engineering Technologies, Monash University, Clayton, VIC 3800, Australia
| | - Laurence Meagher
- Department of Materials Science and Engineering, Monash University, Clayton, VIC 3800, Australia
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia
- ARC Training Centre for Cell and Tissue Engineering Technologies, Monash University, Clayton, VIC 3800, Australia
| | - Simon R Corrie
- Department of Chemical and Biological Engineering, Monash University, Clayton, VIC 3800, Australia
- ARC Training Centre for Cell and Tissue Engineering Technologies, Monash University, Clayton, VIC 3800, Australia
| |
Collapse
|
8
|
Liu X, Tang J, Wang Z, Zhu C, Deng H, Sun X, Yu G, Rong F, Chen X, Liao Q, Jia S, Liu W, Zha H, Fan S, Cai X, Gui JF, Xiao W. Oxygen enhances antiviral innate immunity through maintenance of EGLN1-catalyzed proline hydroxylation of IRF3. Nat Commun 2024; 15:3533. [PMID: 38670937 PMCID: PMC11053110 DOI: 10.1038/s41467-024-47814-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
Oxygen is essential for aerobic organisms, but little is known about its role in antiviral immunity. Here, we report that during responses to viral infection, hypoxic conditions repress antiviral-responsive genes independently of HIF signaling. EGLN1 is identified as a key mediator of the oxygen enhancement of antiviral innate immune responses. Under sufficient oxygen conditions, EGLN1 retains its prolyl hydroxylase activity to catalyze the hydroxylation of IRF3 at proline 10. This modification enhances IRF3 phosphorylation, dimerization and nuclear translocation, leading to subsequent IRF3 activation. Furthermore, mice and zebrafish with Egln1 deletion, treatment with the EGLN inhibitor FG4592, or mice carrying an Irf3 P10A mutation are more susceptible to viral infections. These findings not only reveal a direct link between oxygen and antiviral responses, but also provide insight into the mechanisms by which oxygen regulates innate immunity.
Collapse
Affiliation(s)
- Xing Liu
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, P. R. China
- Hubei Hongshan Laboratory, Wuhan, P. R. China
- University of Chinese Academy of Sciences, Beijing, P. R. China
- The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan, P. R. China
| | - Jinhua Tang
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, P. R. China
- Department of Pharmacy, Women and Children's Hospital of Chongqing Medical University, Chongqing, P. R. China
| | - Zixuan Wang
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, P. R. China
- University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Chunchun Zhu
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, P. R. China
- University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Hongyan Deng
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, P. R. China
| | - Xueyi Sun
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, P. R. China
- University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Guangqing Yu
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, P. R. China
- University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Fangjing Rong
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, P. R. China
- University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Xiaoyun Chen
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, P. R. China
- University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Qian Liao
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, P. R. China
- University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Shuke Jia
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, P. R. China
- University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Wen Liu
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, P. R. China
- University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Huangyuan Zha
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, P. R. China
| | - Sijia Fan
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, P. R. China
| | - Xiaolian Cai
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, P. R. China
| | - Jian-Fang Gui
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, P. R. China
- Hubei Hongshan Laboratory, Wuhan, P. R. China
- University of Chinese Academy of Sciences, Beijing, P. R. China
- The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan, P. R. China
| | - Wuhan Xiao
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, P. R. China.
- Hubei Hongshan Laboratory, Wuhan, P. R. China.
- University of Chinese Academy of Sciences, Beijing, P. R. China.
- The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan, P. R. China.
| |
Collapse
|
9
|
Urrutia AA, Mesa-Ciller C, Guajardo-Grence A, Alkan HF, Soro-Arnáiz I, Vandekeere A, Ferreira Campos AM, Igelmann S, Fernández-Arroyo L, Rinaldi G, Lorendeau D, De Bock K, Fendt SM, Aragonés J. HIF1α-dependent uncoupling of glycolysis suppresses tumor cell proliferation. Cell Rep 2024; 43:114103. [PMID: 38607920 PMCID: PMC11063627 DOI: 10.1016/j.celrep.2024.114103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 11/20/2023] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
Hypoxia-inducible factor-1α (HIF1α) attenuates mitochondrial activity while promoting glycolysis. However, lower glycolysis is compromised in human clear cell renal cell carcinomas, in which HIF1α acts as a tumor suppressor by inhibiting cell-autonomous proliferation. Here, we find that, unexpectedly, HIF1α suppresses lower glycolysis after the glyceraldehyde 3-phosphate dehydrogenase (GAPDH) step, leading to reduced lactate secretion in different tumor cell types when cells encounter a limited pyruvate supply such as that typically found in the tumor microenvironment in vivo. This is because HIF1α-dependent attenuation of mitochondrial oxygen consumption increases the NADH/NAD+ ratio that suppresses the activity of the NADH-sensitive GAPDH glycolytic enzyme. This is manifested when pyruvate supply is limited, since pyruvate acts as an electron acceptor that prevents the increment of the NADH/NAD+ ratio. Furthermore, this anti-glycolytic function provides a molecular basis to explain how HIF1α can suppress tumor cell proliferation by increasing the NADH/NAD+ ratio.
Collapse
Affiliation(s)
- Andrés A Urrutia
- Research Unit, Hospital of Santa Cristina, Research Institute Princesa (IIS IP), Autonomous University of Madrid, 28009 Madrid, Spain
| | - Claudia Mesa-Ciller
- Research Unit, Hospital of Santa Cristina, Research Institute Princesa (IIS IP), Autonomous University of Madrid, 28009 Madrid, Spain
| | - Andrea Guajardo-Grence
- Research Unit, Hospital of Santa Cristina, Research Institute Princesa (IIS IP), Autonomous University of Madrid, 28009 Madrid, Spain
| | - H Furkan Alkan
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB Center for Cancer Biology, VIB, Herestraat 49, 3000 Leuven, Belgium; Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Herestraat 49, 3000 Leuven, Belgium
| | - Inés Soro-Arnáiz
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH), Zurich, Switzerland
| | - Anke Vandekeere
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB Center for Cancer Biology, VIB, Herestraat 49, 3000 Leuven, Belgium; Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Herestraat 49, 3000 Leuven, Belgium
| | - Ana Margarida Ferreira Campos
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB Center for Cancer Biology, VIB, Herestraat 49, 3000 Leuven, Belgium; Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Herestraat 49, 3000 Leuven, Belgium
| | - Sebastian Igelmann
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB Center for Cancer Biology, VIB, Herestraat 49, 3000 Leuven, Belgium; Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Herestraat 49, 3000 Leuven, Belgium
| | - Lucía Fernández-Arroyo
- Research Unit, Hospital of Santa Cristina, Research Institute Princesa (IIS IP), Autonomous University of Madrid, 28009 Madrid, Spain
| | - Gianmarco Rinaldi
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB Center for Cancer Biology, VIB, Herestraat 49, 3000 Leuven, Belgium; Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Herestraat 49, 3000 Leuven, Belgium
| | - Doriane Lorendeau
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB Center for Cancer Biology, VIB, Herestraat 49, 3000 Leuven, Belgium; Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Herestraat 49, 3000 Leuven, Belgium
| | - Katrien De Bock
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH), Zurich, Switzerland
| | - Sarah-Maria Fendt
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB Center for Cancer Biology, VIB, Herestraat 49, 3000 Leuven, Belgium; Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Herestraat 49, 3000 Leuven, Belgium
| | - Julián Aragonés
- Research Unit, Hospital of Santa Cristina, Research Institute Princesa (IIS IP), Autonomous University of Madrid, 28009 Madrid, Spain; CIBER de Enfermedades Cardiovasculares (CIBERCV), Carlos III Health Institute, Madrid, Spain.
| |
Collapse
|
10
|
Yamagata K, Mizumoto T, Yoshizawa T. The Emerging Role of SIRT7 in Glucose and Lipid Metabolism. Cells 2023; 13:48. [PMID: 38201252 PMCID: PMC10778536 DOI: 10.3390/cells13010048] [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: 11/21/2023] [Revised: 12/13/2023] [Accepted: 12/24/2023] [Indexed: 01/12/2024] Open
Abstract
Sirtuins (SIRT1-7 in mammals) are a family of NAD+-dependent lysine deacetylases and deacylases that regulate diverse biological processes, including metabolism, stress responses, and aging. SIRT7 is the least well-studied member of the sirtuins, but accumulating evidence has shown that SIRT7 plays critical roles in the regulation of glucose and lipid metabolism by modulating many target proteins in white adipose tissue, brown adipose tissue, and liver tissue. This review focuses on the emerging roles of SIRT7 in glucose and lipid metabolism in comparison with SIRT1 and SIRT6. We also discuss the possible implications of SIRT7 inhibition in the treatment of metabolic diseases such as type 2 diabetes and obesity.
Collapse
Affiliation(s)
- Kazuya Yamagata
- Department of Medical Biochemistry, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan; (T.M.); (T.Y.)
- Center for Metabolic Regulation of Healthy Aging, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Tomoya Mizumoto
- Department of Medical Biochemistry, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan; (T.M.); (T.Y.)
| | - Tatsuya Yoshizawa
- Department of Medical Biochemistry, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan; (T.M.); (T.Y.)
| |
Collapse
|
11
|
Wang Y, Liu T, Cai Y, Liu W, Guo J. SIRT6's function in controlling the metabolism of lipids and glucose in diabetic nephropathy. Front Endocrinol (Lausanne) 2023; 14:1244705. [PMID: 37876546 PMCID: PMC10591331 DOI: 10.3389/fendo.2023.1244705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 09/21/2023] [Indexed: 10/26/2023] Open
Abstract
Diabetic nephropathy (DN) is a complication of diabetes mellitus (DM) and the main cause of excess mortality in patients with type 2 DM. The pathogenesis and progression of DN are closely associated with disorders of glucose and lipid metabolism. As a member of the sirtuin family, SIRT6 has deacetylation, defatty-acylation, and adenosine diphosphate-ribosylation enzyme activities as well as anti-aging and anticancer activities. SIRT6 plays an important role in glucose and lipid metabolism and signaling, especially in DN. SIRT6 improves glucose and lipid metabolism by controlling glycolysis and gluconeogenesis, affecting insulin secretion and transmission and regulating lipid decomposition, transport, and synthesis. Targeting SIRT6 may provide a new therapeutic strategy for DN by improving glucose and lipid metabolism. This review elaborates on the important role of SIRT6 in glucose and lipid metabolism, discusses the potential of SIRT6 as a therapeutic target to improve glucose and lipid metabolism and alleviate DN occurrence and progression of DN, and describes the prospects for future research.
Collapse
Affiliation(s)
- Ying Wang
- Country Renal Research Institution of Beijing University of Chinese Medicine, Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Tongtong Liu
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuzi Cai
- Country Renal Research Institution of Beijing University of Chinese Medicine, Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Weijing Liu
- Country Renal Research Institution of Beijing University of Chinese Medicine, Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Jing Guo
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
12
|
Liao Q, Zhu C, Sun X, Wang Z, Chen X, Deng H, Tang J, Jia S, Liu W, Xiao W, Liu X. Disruption of sirtuin 7 in zebrafish facilitates hypoxia tolerance. J Biol Chem 2023; 299:105074. [PMID: 37481210 PMCID: PMC10448219 DOI: 10.1016/j.jbc.2023.105074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 07/09/2023] [Accepted: 07/11/2023] [Indexed: 07/24/2023] Open
Abstract
SIRT7 is a member of the sirtuin family proteins with nicotinamide adenine dinucleotide (NAD+)-dependent histone deacetylase activity, which can inhibit the activity of hypoxia-inducible factors independently of its enzymatic activity. However, the role of SIRT7 in affecting hypoxia signaling in vivo is still elusive. Here, we find that sirt7-null zebrafish are more resistant to hypoxic conditions, along with an increase of hypoxia-responsive gene expression and erythrocyte numbers, compared with their wildtype siblings. Overexpression of sirt7 suppresses the expression of hypoxia-responsive genes. Further assays indicate that sirt7 interacts with zebrafish hif-1αa, hif-1αb, hif-2αa, and hif-2αb to inhibit their transcriptional activity and mediate their protein degradation. In addition, sirt7 not only binds to the hypoxia responsive element of hypoxia-inducible gene promoters but also causes a reduction of H3K18Ac on these promoters. Sirt7 may regulate hypoxia-responsive gene expression through its enzymatic and nonenzymatic activities. This study provides novel insights into sirt7 function and sheds new light on the regulation of hypoxia signaling by sirt7.
Collapse
Affiliation(s)
- Qian Liao
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; University of Chinese Academy of Sciences, Beijing, China
| | - Chunchun Zhu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; University of Chinese Academy of Sciences, Beijing, China
| | - Xueyi Sun
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; University of Chinese Academy of Sciences, Beijing, China
| | - Zixuan Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; University of Chinese Academy of Sciences, Beijing, China
| | - Xiaoyun Chen
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; University of Chinese Academy of Sciences, Beijing, China
| | - Hongyan Deng
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Jinhua Tang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Shuke Jia
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; University of Chinese Academy of Sciences, Beijing, China
| | - Wen Liu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; University of Chinese Academy of Sciences, Beijing, China
| | - Wuhan Xiao
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; University of Chinese Academy of Sciences, Beijing, China; Hubei Hongshan Laboratory, Wuhan, China; The Innovation of Seed Design, Chinese Academy of Sciences, Wuhan, China.
| | - Xing Liu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; University of Chinese Academy of Sciences, Beijing, China; The Innovation of Seed Design, Chinese Academy of Sciences, Wuhan, China.
| |
Collapse
|
13
|
Yue Y, Tang Y, Huang H, Zheng D, Liu C, Zhang H, Liu Y, Li Y, Sun X, Lu L. VBP1 negatively regulates CHIP and selectively inhibits the activity of hypoxia-inducible factor (HIF)-1α but not HIF-2α. J Biol Chem 2023:104829. [PMID: 37201586 DOI: 10.1016/j.jbc.2023.104829] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 05/07/2023] [Accepted: 05/09/2023] [Indexed: 05/20/2023] Open
Abstract
Hypoxia-inducible factor-1 (HIF-1) is a critical transcription factor that regulates expression of genes involved in cellular adaptation to low oxygen levels. Aberrant regulation of the HIF-1 signaling pathway is linked to various human diseases. Previous studies have established that HIF-1α is rapidly degraded in a von Hippel-Lindau protien (pVHL)-dependent manner under normoxic conditions. In this study, we find that pVHL binding protein 1 (VBP1) is a negative regulator of HIF-1α but not HIF-2α using zebrafish as an in vivo model and in vitro cell culture models. Deletion of vbp1 in zebrafish caused Hif-1α accumulation and upregulation of Hif target genes. Moreover, vbp1 was involved in induction of hematopoietic stem cells (HSCs) under hypoxic conditions. However, VBP1 interacted with and promoted the degradation of HIF-1α in a pVHL-independent manner. Mechanistically, we identify the ubiquitin ligase CHIP and HSP70 as new VBP1 binding partners, and demonstrate that VBP1 negatively regulated CHIP and facilitated CHIP-mediated degradation of HIF-1α. In patients with clear cell renal cell carcinoma (ccRCC), lower VBP1 expression was associated with worse survival outcomes. In conclusion, our results link VBP1 with CHIP stability and provide insights into underlying molecular mechanisms of HIF-1α-driven pathological processes.
Collapse
Affiliation(s)
- Yiming Yue
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Biological Products, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Yanfei Tang
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Biological Products, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Hao Huang
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Biological Products, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Dongdong Zheng
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Biological Products, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Cong Liu
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Biological Products, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Haifeng Zhang
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Biological Products, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Yunzhang Liu
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Biological Products, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Yun Li
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Biological Products, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Xiangrong Sun
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Ling Lu
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Biological Products, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.
| |
Collapse
|
14
|
Hagizawa H, Koyamatsu S, Okada S, Kaito T, Tsumaki N. Chondrocyte-like cells in nucleus pulposus and articular chondrocytes have similar transcriptomic profiles and are paracrine-regulated by hedgehog from notochordal cells and subchondral bone. Front Cell Dev Biol 2023; 11:1151947. [PMID: 37255604 PMCID: PMC10225674 DOI: 10.3389/fcell.2023.1151947] [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: 01/27/2023] [Accepted: 05/02/2023] [Indexed: 06/01/2023] Open
Abstract
Objective: The nucleus pulposus (NP) comprises notochordal NP cells (NCs) and chondrocyte-like NP cells (CLCs). Although morphological similarities between CLCs and chondrocytes have been reported, interactions between CLCs and NCs remain unclear. In this study, we aimed to clarify regulatory mechanisms of cells in the NP and chondrocytes. Design: We performed single-cell RNA sequencing (scRNA-seq) analysis of the articular cartilage (AC) and NP of three-year-old cynomolgus monkeys in which NCs were present. We then performed immunohistochemical analysis of NP and distal femur. We added sonic hedgehog (SHH) to primary chondrocyte culture. Results: The scRNA-seq analysis revealed that CLCs and some articular chondrocytes had similar gene expression profiles, particularly related to GLI1, the nuclear mediator of the hedgehog pathway. In the NP, cell-cell interaction analysis revealed SHH expression in NCs, resulting in hedgehog signaling to CLCs. In contrast, no hedgehog ligands were expressed by chondrocytes in AC samples. Immunohistochemical analysis of the distal end of femur indicated that SHH and Indian hedgehog (IHH) were expressed around the subchondral bone that was excluded from our scRNA-seq sample. scRNA-seq data analysis and treatment of primary chondrocytes with SHH revealed that hedgehog proteins mediated an increase in hypoxia-inducible factor 1-alpha (HIF-1α) levels. Conclusion: CLCs and some articular chondrocytes have similar transcriptional profiles, regulated by paracrine hedgehog proteins secreted from NCs in the NP and from the subchondral bone in the AC to promote the HIF-1α pathway.
Collapse
Affiliation(s)
- Hiroki Hagizawa
- Department of Tissue Biochemistry, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan
- Department of Orthopaedic Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Saeko Koyamatsu
- Department of Tissue Biochemistry, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan
| | - Seiji Okada
- Department of Orthopaedic Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Takashi Kaito
- Department of Orthopaedic Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Noriyuki Tsumaki
- Department of Tissue Biochemistry, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan
- Department of Clinical Application, Center for IPS Cell Research and Application, Kyoto University, Kyoto, Japan
- Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Osaka University, Osaka, Japan
| |
Collapse
|
15
|
Yu Y, He J, Liu W, Li Z, Weng S, He J, Guo C. Molecular Characterization and Functional Analysis of Hypoxia-Responsive Factor Prolyl Hydroxylase Domain 2 in Mandarin Fish ( Siniperca chuatsi). Animals (Basel) 2023; 13:ani13091556. [PMID: 37174593 PMCID: PMC10177477 DOI: 10.3390/ani13091556] [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: 03/22/2023] [Revised: 04/18/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023] Open
Abstract
With increased breeding density, the phenomenon of hypoxia gradually increases in aquaculture. Hypoxia is primarily mediated by the hypoxia-inducible factor 1 (HIF-1) signaling pathway. Prolyl hydroxylase domain proteins (PHD) are cellular oxygen-sensing molecules that regulate the stability of HIF-1α through hydroxylation. In this study, the characterization of the PHD2 from mandarin fish Siniperca chuatsi (scPHD2) and its roles in the HIF-1 signaling pathway were investigated. Bioinformation analysis showed that scPHD2 had the conserved prolyl 4-hydroxylase alpha subunit homolog domains at its C-terminal and was more closely related to other Perciformes PHD2 than other PHD2. Tissue-distribution results revealed that scphd2 gene was expressed in all tissues tested and more highly expressed in blood and liver than in other tested tissues. Dual-luciferase reporter gene and RT-qPCR assays showed that scPHD2 overexpression could significantly inhibit the HIF-1 signaling pathway. Co-immunoprecipitation analysis showed that scPHD2 could interact with scHIF-1α. Protein degradation experiment results suggested that scPHD2 could promote scHIF-1α degradation through the proteasome degradation pathway. This study advances our understanding of how the HIF-1 signaling pathway is regulated by scPHD2 and will help in understanding the molecular mechanisms underlying hypoxia adaptation in teleost fish.
Collapse
Affiliation(s)
- Yang Yu
- State Key Laboratory for Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Guangdong Provincial Observation and Research Station for Marine Ranching of the Lingdingyang Bay, School of Marine Sciences, Sun Yat-sen University, 135 Xingang Road West, Guangzhou 510275, China
| | - Jian He
- State Key Laboratory for Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Guangdong Provincial Observation and Research Station for Marine Ranching of the Lingdingyang Bay, School of Marine Sciences, Sun Yat-sen University, 135 Xingang Road West, Guangzhou 510275, China
| | - Wenhui Liu
- State Key Laboratory for Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Guangdong Provincial Observation and Research Station for Marine Ranching of the Lingdingyang Bay, School of Marine Sciences, Sun Yat-sen University, 135 Xingang Road West, Guangzhou 510275, China
| | - Zhimin Li
- State Key Laboratory for Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Guangdong Provincial Observation and Research Station for Marine Ranching of the Lingdingyang Bay, School of Marine Sciences, Sun Yat-sen University, 135 Xingang Road West, Guangzhou 510275, China
| | - Shaoping Weng
- Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-sen University, 135 Xingang Road West, Guangzhou 510275, China
| | - Jianguo He
- State Key Laboratory for Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Guangdong Provincial Observation and Research Station for Marine Ranching of the Lingdingyang Bay, School of Marine Sciences, Sun Yat-sen University, 135 Xingang Road West, Guangzhou 510275, China
| | - Changjun Guo
- State Key Laboratory for Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Guangdong Provincial Observation and Research Station for Marine Ranching of the Lingdingyang Bay, School of Marine Sciences, Sun Yat-sen University, 135 Xingang Road West, Guangzhou 510275, China
- Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-sen University, 135 Xingang Road West, Guangzhou 510275, China
| |
Collapse
|
16
|
Chen JX, Li L, Cantrell AC, Williams QA, Zeng H. High Glucose Activates Prolyl Hydroxylases and Disrupts HIF-α Signaling via the P53/TIGAR Pathway in Cardiomyocyte. Cells 2023; 12:1060. [PMID: 37048134 PMCID: PMC10093703 DOI: 10.3390/cells12071060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 03/24/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023] Open
Abstract
The induction of hypoxia tolerance has emerged as a novel therapeutic strategy for the treatment of ischemic diseases. The disruption of hypoxic signaling by hyperglycemia has been shown to contribute to diabetic cardiomyopathy. In this study, we explored the potential molecular mechanisms by which high glucose (HG) impairs hypoxia-inducible factor-α (HIF-α) signaling in cardiomyocytes. The exposure of H9c2 cell lines to HG resulted in time- and concentration-dependent decreases in HIF-1α and HIF-2α expression together with an increase in prolyl hydroxylase-1,2 (PHD1 and PHD2) expression, the main regulators of HIF-α destabilization in the heart. The exposure of H9c2 cells to normal glucose (5.5 mM) and high glucose (15, 30, and 45 mM) led to dose-dependent increases in p53 and TIGAR and a decrease in SIRT3 expression. The pretreatment of H9c2 with p53 siRNA to knockdown p53 attenuated PHD1 and PHD2 expression, thus significantly enhancing HIF-1α and HIF-2α expression in H9c2 cells under HG conditions. Interestingly, pretreatment with p53 siRNA altered H9c2 cell metabolism by reducing oxygen consumption rate and increasing glycolysis. Similarly, pretreatment with TIGAR siRNA blunted HG-induced PHD1 and PHD2 expression. This was accompanied by an increase in HIF-1α and HIF-2α expression with a reduction in oxygen consumption rate in H9c2 cells. Furthermore, pretreatment with adenovirus-SIRT3 (Ad-SIRT3) significantly reduced the HG-induced expression of p53 and PHDs and increased HIF-1α levels in H9c2 cells. Ad-SIRT3 treatment also regulated PHDs-HIF-1α levels in the hearts of diabetic db/db mice. Our study revealed a novel role of the HG-induced disruption of PHDs-HIF-α signaling via upregulating p53 and TIGAR expression. Therefore, the p53/TIGAR signaling pathway may be a novel target for diabetic cardiomyopathy.
Collapse
Affiliation(s)
| | | | | | | | - Heng Zeng
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA; (J.-X.C.)
| |
Collapse
|
17
|
Sutton GD, Jiang C, Liu G, Teets TS. Ratiometric oxygen sensors of cyclometalated iridium(III) with enhanced quantum yields and variable dynamic ranges. Dalton Trans 2023; 52:3195-3202. [PMID: 36794766 DOI: 10.1039/d3dt00350g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
Four luminescent ratiometric oxygen sensors, pairing phosphorescent cyclometalated iridium with coumarin or BODIPY fluorophores, are presented here. These compounds realize three key improvements over our previous designs, namely higher phosphorescence quantum yields, the ability to access intermediate dynamic ranges better suited for typical atmospheric O2 levels, and the possibility of using visible excitation instead of UV. These ratiometric sensors are accessed via very simple, 1-step syntheses involving direct reaction of the chloro-bridged cyclometalated iridium dimer with the pyridyl-substituted fluorophore. They have phosphorescent quantum yields up to 29% with short to intermediate phosphoresence lifetimes ranging from 1.7 to 5.3 μs in three of the sensors, with the fourth having a long lifetime of 440 μs that is very responsive to oxygen. In one case, visible excitation of 430 nm is used to provide dual emission instead of UV excitation.
Collapse
Affiliation(s)
- Gregory D Sutton
- Department of Chemistry, University of Houston, 3585 Cullen Blvd., Room 112, Houston, TX, 77204-5003, USA.
| | - Chenggang Jiang
- Department of Chemistry, University of Houston, 3585 Cullen Blvd., Room 112, Houston, TX, 77204-5003, USA.
| | - Gardenia Liu
- Department of Chemistry, University of Houston, 3585 Cullen Blvd., Room 112, Houston, TX, 77204-5003, USA.
| | - Thomas S Teets
- Department of Chemistry, University of Houston, 3585 Cullen Blvd., Room 112, Houston, TX, 77204-5003, USA.
| |
Collapse
|
18
|
Lee HY, Kim DS, Hwang GY, Lee JK, Lee HL, Jung JW, Hwang SY, Baek SW, Yoon SL, Ha Y, Kim KN, Han I, Han DK, Lee CK. Multi-modulation of immune-inflammatory response using bioactive molecule-integrated PLGA composite for spinal fusion. Mater Today Bio 2023; 19:100611. [PMID: 36969699 PMCID: PMC10034518 DOI: 10.1016/j.mtbio.2023.100611] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/03/2023] [Accepted: 03/11/2023] [Indexed: 03/15/2023] Open
Abstract
Despite current developments in bone substitute technology for spinal fusion, there is a lack of adequate materials for bone regeneration in clinical applications. Recombinant human bone morphogenetic protein-2 (rhBMP-2) is commercially available, but a severe inflammatory response is a known side effect. Bone graft substitutes that enhance osteogenesis without adverse effects are needed. We developed a bioactive molecule-laden PLGA composite with multi-modulation for bone fusion. This bioresorbable composite scaffold was considered for bone tissue engineering. Among the main components, magnesium hydroxide (MH) aids in reduction of acute inflammation affecting disruption of new bone formation. Decellularized bone extracellular matrix (bECM) and demineralized bone matrix (DBM) composites were used for osteoconductive and osteoinductive activities. A bioactive molecule, polydeoxyribonucleotide (PDRN, PN), derived from trout was used for angiogenesis during bone regeneration. A nano-emulsion method that included Span 80 was used to fabricate bioactive PLGA-MH-bECM/DBM-PDRN (PME2/PN) composite to obtain a highly effective and safe scaffold. The synergistic effect provided by PME2/PN improved not only osteogenic and angiogenic gene expression for bone fusion but also improved immunosuppression and polarization of macrophages that were important for bone tissue repair, using a rat model of posterolateral spinal fusion (PLF). It thus had sufficient biocompatibility and bioactivity for spinal fusion.
Collapse
Affiliation(s)
- Hye Yeong Lee
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
| | - Da-Seul Kim
- Department of Biomedical Science, CHA University, Gyeonggi-do, 13488, Republic of Korea
- School of Integrative Engineering, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Gwang Yong Hwang
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
| | - Jun-Kyu Lee
- Department of Biomedical Science, CHA University, Gyeonggi-do, 13488, Republic of Korea
| | - Hye-Lan Lee
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
| | - Ji-Won Jung
- Department of Biomedical Science, CHA University, Gyeonggi-do, 13488, Republic of Korea
| | - Sae Yeon Hwang
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
- Graduate Program in Bioindustrial Engineering, Yonsei University, Seoul, 03722, Republic of Korea
| | - Seung-Woon Baek
- Department of Biomedical Science, CHA University, Gyeonggi-do, 13488, Republic of Korea
| | - Sol lip Yoon
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
| | - Yoon Ha
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
| | - Keung Nyun Kim
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
| | - Inbo Han
- Department of Neurosurgery, CHA University School of Medicine, CHA Bungdang Medical Center, Gyeonggi-do, 13496, Republic of Korea
| | - Dong Keun Han
- Department of Biomedical Science, CHA University, Gyeonggi-do, 13488, Republic of Korea
- Corresponding author.
| | - Chang Kyu Lee
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
- Corresponding author.
| |
Collapse
|
19
|
Downstream Targets of VHL/HIF-α Signaling in Renal Clear Cell Carcinoma Progression: Mechanisms and Therapeutic Relevance. Cancers (Basel) 2023; 15:cancers15041316. [PMID: 36831657 PMCID: PMC9953937 DOI: 10.3390/cancers15041316] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/09/2023] [Accepted: 02/11/2023] [Indexed: 02/22/2023] Open
Abstract
The clear cell variant of renal cell carcinoma (ccRCC) is the most common renal epithelial malignancy and responsible for most of the deaths from kidney cancer. Patients carrying inactivating mutations in the Von Hippel-Lindau (VHL) gene have an increased proclivity to develop several types of tumors including ccRCC. Normally, the Hypoxia Inducible Factor alpha (HIF-α) subunits of the HIF heterodimeric transcription factor complex are regulated by oxygen-dependent prolyl-hydroxylation, VHL-mediated ubiquitination and proteasomal degradation. Loss of pVHL function results in elevated levels of HIF-α due to increased stability, leading to RCC progression. While HIF-1α acts as a tumor suppressor, HIF-2α promotes oncogenic potential by driving tumor progression and metastasis through activation of hypoxia-sensitive signaling pathways and overexpression of HIF-2α target genes. One strategy to suppress ccRCC aggressiveness is directed at inhibition of HIF-2α and the associated molecular pathways leading to cell proliferation, angiogenesis, and metastasis. Indeed, clinical and pre-clinical data demonstrated the effectiveness of HIF-2α targeted therapy in attenuating ccRCC progression. This review focuses on the signaling pathways and the involved genes (cyclin D, c-Myc, VEGF-a, EGFR, TGF-α, GLUT-1) that confer oncogenic potential downstream of the VHL-HIF-2α signaling axis in ccRCC. Discussed as well are current treatment options (including receptor tyrosine kinase inhibitors such as sunitinib), the medical challenges (high prevalence of metastasis at the time of diagnosis, refractory nature of advanced disease to current treatment options), scientific challenges and future directions.
Collapse
|
20
|
Wang R, Liang L, Matsumoto M, Iwata K, Umemura A, He F. Reactive Oxygen Species and NRF2 Signaling, Friends or Foes in Cancer? Biomolecules 2023; 13:biom13020353. [PMID: 36830722 PMCID: PMC9953152 DOI: 10.3390/biom13020353] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 02/03/2023] [Accepted: 02/09/2023] [Indexed: 02/17/2023] Open
Abstract
The imbalance between reactive oxygen species (ROS) production and clearance causes oxidative stress and ROS, which play a central role in regulating cell and tissue physiology and pathology. Contingent upon concentration, ROS influence cancer development in contradictory ways, either stimulating cancer survival and growth or causing cell death. Cells developed evolutionarily conserved programs to sense and adapt redox the fluctuations to regulate ROS as either signaling molecules or toxic insults. The transcription factor nuclear factor erythroid 2-related factor 2 (NRF2)-KEAP1 system is the master regulator of cellular redox and metabolic homeostasis. NRF2 has Janus-like roles in carcinogenesis and cancer development. Short-term NRF2 activation suppresses tissue injury, inflammation, and cancer initiation. However, cancer cells often exhibit constitutive NRF2 activation due to genetic mutations or oncogenic signaling, conferring advantages for cancer cells' survival and growth. Emerging evidence suggests that NRF2 hyperactivation, as an adaptive cancer phenotype under stressful tumor environments, regulates all hallmarks of cancer. In this review, we summarized the source of ROS, regulation of ROS signaling, and cellular sensors for ROS and oxygen (O2), we reviewed recent progress on the regulation of ROS generation and NRF2 signaling with a focus on the new functions of NRF2 in cancer development that reach beyond what we originally envisioned, including regulation of cancer metabolism, autophagy, macropinocytosis, unfolded protein response, proteostasis, and circadian rhythm, which, together with anti-oxidant and drug detoxification enzymes, contributes to cancer development, metastasis, and anticancer therapy resistance.
Collapse
Affiliation(s)
- Ruolei Wang
- The Center for Cancer Research, Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Lirong Liang
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Misaki Matsumoto
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Kazumi Iwata
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Atsushi Umemura
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
- Correspondence: (A.U.); (F.H.); Tel.: +75-251-5332 (A.U.); +86-21-5132-2501 (F.H.)
| | - Feng He
- The Center for Cancer Research, Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Correspondence: (A.U.); (F.H.); Tel.: +75-251-5332 (A.U.); +86-21-5132-2501 (F.H.)
| |
Collapse
|
21
|
Guette-Marquet S, Roques C, Bergel A. Direct electrochemical detection of trans-plasma membrane electron transfer: A possible alternative pathway for cell respiration. Biosens Bioelectron 2022; 220:114896. [DOI: 10.1016/j.bios.2022.114896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/04/2022] [Accepted: 11/06/2022] [Indexed: 11/09/2022]
|
22
|
Small extracellular vesicles of hypoxic endothelial cells regulate the therapeutic potential of adipose-derived mesenchymal stem cells via miR-486-5p/PTEN in a limb ischemia model. J Nanobiotechnology 2022; 20:422. [PMID: 36153544 PMCID: PMC9509557 DOI: 10.1186/s12951-022-01632-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 09/12/2022] [Indexed: 11/29/2022] Open
Abstract
Background Patients with critical limb ischemia (CLI) are at great risk of major amputation and cardiovascular events. Adipose-derived mesenchymal stem cell (ADSC) therapy is a promising therapeutic strategy for CLI, but the poor engraftment and insufficient angiogenic ability of ADSCs limit their regenerative potential. Herein, we explored the potential of human umbilical vein endothelial cells (HUVECs)-derived small extracellular vesicles (sEVs) for enhancing the therapeutic efficacy of ADSCs in CLI. Results sEVs derived from hypoxic HUVECs enhanced the resistance of ADSCs to reactive oxygen species (ROS) and further improved the proangiogenic ability of ADSCs in vitro. We found that the hypoxic environment altered the composition of sEVs from HUVECs and that hypoxia increased the level of miR-486-5p in sEVs. Compared to normoxic sEVs (nsEVs), hypoxic sEVs (hsEVs) of HUVECs significantly downregulated the phosphatase and tensin homolog (PTEN) via direct targeting of miR-486-5p, therefore activating the AKT/MTOR/HIF-1α pathway and influencing the survival and pro-angiogenesis ability of ADSCs. In a hindlimb ischemia model, we discovered that hsEVs-primed ADSCs exhibited superior cell engraftment, and resulted in better angiogenesis and tissue repair. Conclusion hsEVs could be used as a therapeutic booster to improve the curative potential of ADSCs in a limb ischemia model. This finding offers new insight for CLI treatment. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01632-1.
Collapse
|
23
|
Torres-Soria AK, Romero Y, Balderas-Martínez YI, Velázquez-Cruz R, Torres-Espíndola LM, Camarena A, Flores-Soto E, Solís-Chagoyán H, Ruiz V, Carlos-Reyes Á, Salinas-Lara C, Luis-García ER, Chávez J, Castillejos-López M, Aquino-Gálvez A. Functional Repercussions of Hypoxia-Inducible Factor-2α in Idiopathic Pulmonary Fibrosis. Cells 2022; 11:cells11192938. [PMID: 36230900 PMCID: PMC9562026 DOI: 10.3390/cells11192938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/06/2022] [Accepted: 09/07/2022] [Indexed: 11/16/2022] Open
Abstract
Hypoxia and hypoxia-inducible factors (HIFs) are essential in regulating several cellular processes, such as survival, differentiation, and the cell cycle; this adaptation is orchestrated in a complex way. In this review, we focused on the impact of hypoxia in the physiopathology of idiopathic pulmonary fibrosis (IPF) related to lung development, regeneration, and repair. There is robust evidence that the responses of HIF-1α and -2α differ; HIF-1α participates mainly in the acute phase of the response to hypoxia, and HIF-2α in the chronic phase. The analysis of their structure and of different studies showed a high specificity according to the tissue and the process involved. We propose that hypoxia-inducible transcription factor 2a (HIF-2α) is part of the persistent aberrant regeneration associated with developing IPF.
Collapse
Affiliation(s)
- Ana Karen Torres-Soria
- Red MEDICI, Carrera de Médico Cirujano, Facultad de Estudios Superiores de Iztacala Universidad Nacional Autónoma de México, Mexico City 54090, Mexico
| | - Yair Romero
- Facultad de Ciencias, Universidad Nacional Autónoma México, Mexico City 04510, Mexico
| | - Yalbi I. Balderas-Martínez
- Laboratorio de Biología Computacional, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico
| | - Rafael Velázquez-Cruz
- Laboratorio de Genómica del Metabolismo Óseo, Instituto Nacional de Medicina Genómica, Mexico City 14610, Mexico
| | | | - Angel Camarena
- Laboratorio de HLA, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 04530, Mexico
| | - Edgar Flores-Soto
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Héctor Solís-Chagoyán
- Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico
| | - Víctor Ruiz
- Departamento de Fibrosis Pulmonar, Laboratorio de Biología Molecular, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico
| | - Ángeles Carlos-Reyes
- Laboratorio de Onco-Inmunobiología, Departamento de Enfermedades Crónico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico
| | - Citlaltepetl Salinas-Lara
- Red MEDICI, Carrera de Médico Cirujano, Facultad de Estudios Superiores de Iztacala Universidad Nacional Autónoma de México, Mexico City 54090, Mexico
| | - Erika Rubí Luis-García
- Departamento de Fibrosis Pulmonar, Laboratorio de Biología Celular, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico
| | - Jaime Chávez
- Departamento de Hiperreactividad Bronquial, Instituto Nacional de Enfermedades, Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico
| | - Manuel Castillejos-López
- Departamento de Epidemiología y Estadística, Instituto Nacional de Enfermedades, Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico
- Correspondence: (M.C.-L.); (A.A.-G.)
| | - Arnoldo Aquino-Gálvez
- Departamento de Fibrosis Pulmonar, Laboratorio de Biología Molecular, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico
- Correspondence: (M.C.-L.); (A.A.-G.)
| |
Collapse
|
24
|
Wu Y, Sutton GD, Halamicek MDS, Xing X, Bao J, Teets TS. Cyclometalated iridium-coumarin ratiometric oxygen sensors: improved signal resolution and tunable dynamic ranges. Chem Sci 2022; 13:8804-8812. [PMID: 35975154 PMCID: PMC9350586 DOI: 10.1039/d2sc02909j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 06/21/2022] [Indexed: 12/31/2022] Open
Abstract
In this work we introduce a new series of ratiometric oxygen sensors based on phosphorescent cyclometalated iridium centers partnered with organic coumarin fluorophores. Three different cyclometalating ligands and two different pyridyl-containing coumarin types were used to prepare six target complexes with tunable excited-state energies. Three of the complexes display dual emission, with fluorescence arising from the coumarin ligand, and phosphorescence from either the cyclometalated iridium center or the coumarin. These dual-emitting complexes function as ratiometric oxygen sensors, with the phosphorescence quenched under O2 while fluorescence is unaffected. The use of blue-fluorescent coumarins results in good signal resolution between fluorescence and phosphorescence. Moreover, the sensitivity and dynamic range, measured with Stern-Volmer analysis, can be tuned two orders of magnitude by virtue of our ability to synthetically control the triplet excited-state ordering. The complex with cyclometalated iridium 3MLCT phosphorescence operates under hyperoxic conditions, whereas the two complexes with coumarin-centered phosphorescence are sensitive to very low levels of O2 and function as hypoxic sensors.
Collapse
Affiliation(s)
- Yanyu Wu
- University of Houston, Department of Chemistry 3585 Cullen Blvd., Room 112 Houston TX 77204-5003 USA
| | - Gregory D Sutton
- University of Houston, Department of Chemistry 3585 Cullen Blvd., Room 112 Houston TX 77204-5003 USA
| | - Michael D S Halamicek
- University of Houston, Department of Chemistry 3585 Cullen Blvd., Room 112 Houston TX 77204-5003 USA
| | - Xinxin Xing
- University of Houston, Department of Electrical and Computer Engineering and Texas Center for Superconductivity (TcSUH) Houston TX 77204 USA
| | - Jiming Bao
- University of Houston, Department of Electrical and Computer Engineering and Texas Center for Superconductivity (TcSUH) Houston TX 77204 USA
| | - Thomas S Teets
- University of Houston, Department of Chemistry 3585 Cullen Blvd., Room 112 Houston TX 77204-5003 USA
| |
Collapse
|
25
|
Ferrante F, Giaimo BD, Friedrich T, Sugino T, Mertens D, Kugler S, Gahr BM, Just S, Pan L, Bartkuhn M, Potente M, Oswald F, Borggrefe T. Hydroxylation of the NOTCH1 intracellular domain regulates Notch signaling dynamics. Cell Death Dis 2022; 13:600. [PMID: 35821235 PMCID: PMC9276811 DOI: 10.1038/s41419-022-05052-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 06/22/2022] [Accepted: 06/28/2022] [Indexed: 01/21/2023]
Abstract
Notch signaling plays a pivotal role in the development and, when dysregulated, it contributes to tumorigenesis. The amplitude and duration of the Notch response depend on the posttranslational modifications (PTMs) of the activated NOTCH receptor - the NOTCH intracellular domain (NICD). In normoxic conditions, the hydroxylase FIH (factor inhibiting HIF) catalyzes the hydroxylation of two asparagine residues of the NICD. Here, we investigate how Notch-dependent gene transcription is regulated by hypoxia in progenitor T cells. We show that the majority of Notch target genes are downregulated upon hypoxia. Using a hydroxyl-specific NOTCH1 antibody we demonstrate that FIH-mediated NICD1 hydroxylation is reduced upon hypoxia or treatment with the hydroxylase inhibitor dimethyloxalylglycine (DMOG). We find that a hydroxylation-resistant NICD1 mutant is functionally impaired and more ubiquitinated. Interestingly, we also observe that the NICD1-deubiquitinating enzyme USP10 is downregulated upon hypoxia. Moreover, the interaction between the hydroxylation-defective NICD1 mutant and USP10 is significantly reduced compared to the NICD1 wild-type counterpart. Together, our data suggest that FIH hydroxylates NICD1 in normoxic conditions, leading to the recruitment of USP10 and subsequent NICD1 deubiquitination and stabilization. In hypoxia, this regulatory loop is disrupted, causing a dampened Notch response.
Collapse
Affiliation(s)
- Francesca Ferrante
- grid.8664.c0000 0001 2165 8627Institute of Biochemistry, University of Giessen, Friedrichstrasse 24, 35392 Giessen, Germany
| | - Benedetto Daniele Giaimo
- grid.8664.c0000 0001 2165 8627Institute of Biochemistry, University of Giessen, Friedrichstrasse 24, 35392 Giessen, Germany
| | - Tobias Friedrich
- grid.8664.c0000 0001 2165 8627Institute of Biochemistry, University of Giessen, Friedrichstrasse 24, 35392 Giessen, Germany ,Biomedical Informatics and Systems Medicine, Science Unit for Basic and Clinical Medicine, Aulweg 128, 35392 Giessen, Germany
| | - Toshiya Sugino
- grid.418032.c0000 0004 0491 220XMax Planck Institute for Heart and Lung Research, Angiogenesis and Metabolism Laboratory, Ludwigstr. 43, 61231 Bad Nauheim, Germany
| | - Daniel Mertens
- grid.410712.10000 0004 0473 882XUniversity Medical Center Ulm, Center for Internal Medicine, Department of Internal Medicine III, Albert-Einstein-Allee 23, 89081 Ulm, Germany ,grid.7497.d0000 0004 0492 0584German Cancer Research Center (DKFZ), Bridging Group Mechanisms of Leukemogenesis, B061, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Sabrina Kugler
- grid.410712.10000 0004 0473 882XUniversity Medical Center Ulm, Center for Internal Medicine, Department of Internal Medicine III, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Bernd Martin Gahr
- grid.410712.10000 0004 0473 882XUniversity Medical Center Ulm, Center for Internal Medicine, Molecular Cardiology, Department of Internal Medicine II, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Steffen Just
- grid.410712.10000 0004 0473 882XUniversity Medical Center Ulm, Center for Internal Medicine, Molecular Cardiology, Department of Internal Medicine II, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Leiling Pan
- grid.410712.10000 0004 0473 882XUniversity Medical Center Ulm, Center for Internal Medicine, Department of Internal Medicine I, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Marek Bartkuhn
- Biomedical Informatics and Systems Medicine, Science Unit for Basic and Clinical Medicine, Aulweg 128, 35392 Giessen, Germany ,Institute for Lung Health (ILH), Aulweg 132, 35392 Giessen, Germany
| | - Michael Potente
- grid.418032.c0000 0004 0491 220XMax Planck Institute for Heart and Lung Research, Angiogenesis and Metabolism Laboratory, Ludwigstr. 43, 61231 Bad Nauheim, Germany ,grid.484013.a0000 0004 6879 971XBerlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Berlin, Germany ,grid.419491.00000 0001 1014 0849Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Franz Oswald
- grid.410712.10000 0004 0473 882XUniversity Medical Center Ulm, Center for Internal Medicine, Department of Internal Medicine I, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Tilman Borggrefe
- grid.8664.c0000 0001 2165 8627Institute of Biochemistry, University of Giessen, Friedrichstrasse 24, 35392 Giessen, Germany
| |
Collapse
|
26
|
Activating Parkin-dependent mitophagy alleviates oxidative stress, apoptosis, and promotes random-pattern skin flaps survival. Commun Biol 2022; 5:616. [PMID: 35732814 PMCID: PMC9217959 DOI: 10.1038/s42003-022-03556-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 06/01/2022] [Indexed: 11/09/2022] Open
Abstract
The random-pattern skin flap is a crucial technique in reconstructive surgery and flap necrosis caused by ischemia/reperfusion injury is a major postoperative complication. Herein, we investigated the mechanism of mitophagy induced by Melatonin (ML) and its effect on the survival of skin flaps. Our results demonstrated that ML could activate mitophagy, ameliorate oxidative stress and alleviate apoptosis in Tert-Butyl hydroperoxide solution (TBHP)-stimulated human umbilical vein endothelial cells in vitro. Inhibiting ML-induced mitophagy considerably abolished its protective effects. Moreover, knockdown of Parkin by siRNA inhibited ML-induced mitophagy, and subsequently exacerbated oxidative stress and apoptosis. Further study demonstrated that inhibition of AMPK reversed these protective effects of ML and downregulated the expression of TFEB. In the vivo study, ML effectively promoted flap survival by activating mitophagy and subsequently ameliorating oxidative stress and mitigating apoptosis. These results established that ML is a potent agent capable for increasing random-pattern skin flap survival by activating Parkin-dependent mitophagy through the AMPK-TFEB signaling pathway.
Collapse
|
27
|
Peters MC, Di Martino S, Boelens T, Qin J, van Mil A, Doevendans PA, Chamuleau SAJ, Sluijter JPG, Neef K. Follistatin-like 1 promotes proliferation of matured human hypoxic iPSC-cardiomyocytes and is secreted by cardiac fibroblasts. Mol Ther Methods Clin Dev 2022; 25:3-16. [PMID: 35317048 PMCID: PMC8917270 DOI: 10.1016/j.omtm.2022.02.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 02/19/2022] [Indexed: 12/12/2022]
Abstract
The human heart has limited regenerative capacity. Therefore, patients often progress to heart failure after ischemic injury, despite advances in reperfusion therapies generally decreasing mortality. Depending on its glycosylation state, Follistatin-like 1 (FSTL1) has been shown to increase cardiomyocyte (CM) proliferation, decrease CM apoptosis, and prevent cardiac rupture in animal models of ischemic heart disease. To explore its therapeutic potential, we used a human in vitro model of cardiac ischemic injury with human induced pluripotent stem cell-derived CMs (iPSC-CMs) and assessed regenerative effects of two differently glycosylated variants of human FSTL1. Furthermore, we investigated the FSTL1-mediated interplay between human cardiac fibroblasts (cFBs) and iPSC-CMs in hypoxia. Both FSTL1 variants increased viability, while only hypo-glycosylated FSTL1 increased CM proliferation post-hypoxia. Human fetal cardiac fibroblasts (fcFBs) expressed and secreted FSTL1 under normoxic conditions, while FSTL1 secretion increased by iPSC-cFBs upon hypoxia but decreased in iPSC-CMs. Co-culture of iPSC-CMs and cFBs increased FSTL1 secretion compared with cFB mono-culture. Taken together, we confirm that FSTL1 induces iPSC-CM proliferation in a human cardiac in vitro hypoxia damage model. Furthermore, we show hypoxia-related FSTL1 secretion by human cFBs and indications for FSTL1-mediated intercellular communication between cardiac cell types in response to hypoxic conditions.
Collapse
Affiliation(s)
- Marijn C Peters
- Department of Cardiology, Laboratory of Experimental Cardiology, Regenerative Medicine Centre Utrecht, University Medical Centre Utrecht, University Utrecht, 3584 CX Utrecht, the Netherlands
| | - Sofia Di Martino
- Department of Cardiology, Laboratory of Experimental Cardiology, Regenerative Medicine Centre Utrecht, University Medical Centre Utrecht, University Utrecht, 3584 CX Utrecht, the Netherlands
| | - Thomas Boelens
- Department of Cardiology, Laboratory of Experimental Cardiology, Regenerative Medicine Centre Utrecht, University Medical Centre Utrecht, University Utrecht, 3584 CX Utrecht, the Netherlands
| | - Jiabin Qin
- Department of Cardiology, Laboratory of Experimental Cardiology, Regenerative Medicine Centre Utrecht, University Medical Centre Utrecht, University Utrecht, 3584 CX Utrecht, the Netherlands
| | - Alain van Mil
- Department of Cardiology, Laboratory of Experimental Cardiology, Regenerative Medicine Centre Utrecht, University Medical Centre Utrecht, University Utrecht, 3584 CX Utrecht, the Netherlands
| | - Pieter A Doevendans
- Department of Cardiology, Laboratory of Experimental Cardiology, Regenerative Medicine Centre Utrecht, University Medical Centre Utrecht, University Utrecht, 3584 CX Utrecht, the Netherlands
| | - Steven A J Chamuleau
- Department of Cardiology, Laboratory of Experimental Cardiology, Regenerative Medicine Centre Utrecht, University Medical Centre Utrecht, University Utrecht, 3584 CX Utrecht, the Netherlands.,Department of Cardiology, Amsterdam Medical Centre, 1105 AZ Amsterdam, the Netherlands
| | - Joost P G Sluijter
- Department of Cardiology, Laboratory of Experimental Cardiology, Regenerative Medicine Centre Utrecht, University Medical Centre Utrecht, University Utrecht, 3584 CX Utrecht, the Netherlands
| | - Klaus Neef
- Department of Cardiology, Laboratory of Experimental Cardiology, Regenerative Medicine Centre Utrecht, University Medical Centre Utrecht, University Utrecht, 3584 CX Utrecht, the Netherlands
| |
Collapse
|
28
|
Yuan Z, Zeng Y, Tian Y, Wang S, Hong B, Yang M. SIRT6 serves as a polyhedron in glycolytic metabolism and ageing-related diseases. Exp Gerontol 2022; 162:111765. [DOI: 10.1016/j.exger.2022.111765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/24/2022] [Accepted: 03/07/2022] [Indexed: 11/04/2022]
|
29
|
Jin W, Zhao J, Yang E, Wang Y, Wang Q, Wu Y, Tong F, Tan Y, Zhou J, Kang C. Neuronal STAT3/HIF-1α/PTRF axis-mediated bioenergetic disturbance exacerbates cerebral ischemia-reperfusion injury via PLA2G4A. Theranostics 2022; 12:3196-3216. [PMID: 35547748 PMCID: PMC9065197 DOI: 10.7150/thno.71029] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 03/24/2022] [Indexed: 11/05/2022] Open
Abstract
Ischemic stroke is an acute and severe neurological disease with high mortality and disability rates worldwide. Polymerase I and transcript release factor (PTRF) plays a pivotal role in regulating cellular senescence, glucose intolerance, lipid metabolism, and mitochondrial bioenergetics, but its mechanism, characteristics, and functions in neuronal cells following the cerebral ischemia-reperfusion (I/R) injury remain to be determined. Methods: Transcription factor motif analysis, chromatin immunoprecipitation (ChIP), luciferase and co-Immunoprecipitation (co-IP) assays were performed to investigate the mechanisms of PTRF in neuronal cells after I/R injury. Lentiviral-sgRNA against PTRF gene was introduced to HT22 cells, and adeno-associated virus (AAV) encoding a human synapsin (hSyn) promoter-driven construct was transduced a short hairpin RNA (shRNA) against PTRF mRNA in primary neuronal cells and the cortex of the cerebral I/R mice for investigating the role of PTRF in neuronal damage and PLA2G4A change induced by the cerebral I/R injury. Results: Here, we reported that neuronal PTRF was remarkably increased in the cerebral penumbra after I/R injury, and HIF-1α and STAT3 regulated the I/R-dependent expression of PTRF via binding to its promoter in neuronal cells. Moreover, overexpression of neuronal PTRF enhanced the activity and stability of PLA2G4A by decreasing its proteasome-mediated degradation pathway. Subsequently, PTRF promoted reprogramming of lipid metabolism and altered mitochondrial bioenergetics, which could lead to oxidative damage, involving autophagy, lipid peroxidation, and ferroptosis via PLA2G4A in neuronal cells. Furthermore, inhibition of neuronal PTRF/PLA2G4A-axis markedly reduced the neurological deficits, cerebral infarct volumes, and mortality rates in the mice following cerebral I/R injury. Conclusion: Our results thus identify that the STAT3/HIF-1α/PTRF-axis in neurons, aggravating cerebral I/R injury by regulating the activity and stability of PLA2G4A, might be a novel therapeutic target for ischemic stroke.
Collapse
Affiliation(s)
- Weili Jin
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China.,Tianjin Neurological Institute, Key Laboratory of Post-neurotrauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin City, Tianjin 300052, China
| | - Jixing Zhao
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China.,Tianjin Neurological Institute, Key Laboratory of Post-neurotrauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin City, Tianjin 300052, China
| | - Eryan Yang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China.,Tianjin Neurological Institute, Key Laboratory of Post-neurotrauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin City, Tianjin 300052, China
| | - Yunfei Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China.,Tianjin Neurological Institute, Key Laboratory of Post-neurotrauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin City, Tianjin 300052, China
| | - Qixue Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China.,Tianjin Neurological Institute, Key Laboratory of Post-neurotrauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin City, Tianjin 300052, China
| | - Ye Wu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China.,Tianjin Neurological Institute, Key Laboratory of Post-neurotrauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin City, Tianjin 300052, China
| | - Fei Tong
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China.,Tianjin Neurological Institute, Key Laboratory of Post-neurotrauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin City, Tianjin 300052, China
| | - Yanli Tan
- Department of Pathology, Affiliated Hospital of Hebei University, Baoding 071000, China
| | - Junhu Zhou
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China.,Tianjin Neurological Institute, Key Laboratory of Post-neurotrauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin City, Tianjin 300052, China
| | - Chunsheng Kang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China.,Tianjin Neurological Institute, Key Laboratory of Post-neurotrauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin City, Tianjin 300052, China
| |
Collapse
|
30
|
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
|
31
|
Caliani I, De Marco G, Cappello T, Giannetto A, Mancini G, Ancora S, Maisano M, Parrino V, Cappello S, Bianchi N, Oliva S, Luciano A, Mauceri A, Leonzio C, Fasulo S. Assessment of the effectiveness of a novel BioFilm-Membrane BioReactor oil-polluted wastewater treatment technology by applying biomarkers in the mussel Mytilus galloprovincialis. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2022; 243:106059. [PMID: 34991045 DOI: 10.1016/j.aquatox.2021.106059] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 10/26/2021] [Accepted: 12/18/2021] [Indexed: 06/14/2023]
Abstract
Petrochemical industries and oil refineries are sources of hazardous chemicals into the aquatic environments, and often a leading cause of reduced oxygen availability, thus resulting in adverse effects in biota. This study is an expansion of our previous work on the assessment of the BioFilm-Membrane Bioreactor (BF-MBR) to mitigate the impact of oil-polluted wastewater on marine environments. Specifically, this study evaluated the reduction of selected chemical constituents (hydrocarbons and trace metals) and toxicity related to hypoxia and DNA damage to mussels Mytilus galloprovincialis, before and after treatment of oil-polluted wastewater with the BF-MBR. The application of a multidisciplinary approach provided evidence of the efficiency of BF-MBR to significantly reducing the pollutants load from oily contaminated seawaters. As result, the health status of mussels was preserved by a hypoxic condition due to oily pollutants, as evidenced by the modulation in the gene expression of HIF-1α and PHD and changes in the level of hypotaurine and taurine. Moreover, ameliorative effects in the energy metabolism were also found in mussel gills showing increased levels of glycogen, glucose and ATP, as well as a mitigated genotoxicity was revealed by the Micronucleus and Comet assays. Overall, findings from this study support the use of the BF-MBR as a promising treatment biotechnology to avoid or limiting the compromise of marine environments from oil pollution.
Collapse
Affiliation(s)
- Ilaria Caliani
- Department of Physics, Earth and Environmental Sciences, University of Siena, Siena, Italy
| | - Giuseppe De Marco
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d'Alcontres 31, Messina 98166, Italy
| | - Tiziana Cappello
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d'Alcontres 31, Messina 98166, Italy
| | - Alessia Giannetto
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d'Alcontres 31, Messina 98166, Italy
| | - Giuseppe Mancini
- Electric, Electronics and Computer Engineering Department, University of Catania, Catania, Italy
| | - Stefania Ancora
- Department of Physics, Earth and Environmental Sciences, University of Siena, Siena, Italy
| | - Maria Maisano
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d'Alcontres 31, Messina 98166, Italy.
| | - Vincenzo Parrino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d'Alcontres 31, Messina 98166, Italy
| | - Simone Cappello
- Institute for Biological Resources and Marine Biotechnology (IRBIM)-National Research Center, Messina, Italy
| | - Nicola Bianchi
- Department of Physics, Earth and Environmental Sciences, University of Siena, Siena, Italy
| | - Sabrina Oliva
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d'Alcontres 31, Messina 98166, Italy
| | - Antonella Luciano
- Energy and Sustainable Economic Development - Department for Sustainability, ENEA - Italian National Agency for the New Technologies, Casaccia Research Centre, Rome, Italy
| | - Angela Mauceri
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d'Alcontres 31, Messina 98166, Italy
| | - Claudio Leonzio
- Department of Physics, Earth and Environmental Sciences, University of Siena, Siena, Italy
| | - Salvatore Fasulo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d'Alcontres 31, Messina 98166, Italy
| |
Collapse
|
32
|
Molecular Characterization and Response of Prolyl Hydroxylase Domain (PHD) Genes to Hypoxia Stress in Hypophthalmichthys molitrix. Animals (Basel) 2022; 12:ani12020131. [PMID: 35049755 PMCID: PMC8772553 DOI: 10.3390/ani12020131] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 01/01/2022] [Accepted: 01/04/2022] [Indexed: 12/16/2022] Open
Abstract
Simple Summary Hypoxia is a common challenge for aquatic organisms, and prolyl hydroxylase domain (PHD) proteins play important roles in hypoxic adaptation by regulating the stability of the hypoxia-inducible factor 1 alpha subunit (HIF-1α). In this study, the full-length cDNAs of three PHD genes were obtained from Hypophthalmichthys molitrix, which is an important freshwater fish and sensitive to low oxygen tension. The amino acid sequence analysis and phylogenetic analysis of PHDs were performed among various species. Furthermore, the expression patterns and the transcriptional responses of H. molitrix PHD genes to acute hypoxia, continued hypoxia, and reoxygenation were explored in different tissues. Our study preliminarily explored the physiological regulation functions of PHD genes at the transcriptional level when addressing the hypoxic challenge and provided a foundation for future systematic explorations of the molecular mechanisms underlying hypoxia adaptation in silver carp. Abstract As an economically and ecologically important freshwater fish, silver carp (Hypophthalmichthys molitrix) is sensitive to low oxygen tension. Prolyl hydroxylase domain (PHD) proteins are critical regulators of adaptive responses to hypoxia for their function of regulating the hypoxia inducible factor-1 alpha subunit (HIF-1α) stability via hydroxylation reaction. In the present study, three PHD genes were cloned from H. molitrix by rapid amplification of cDNA ends (RACE). The total length of HmPHD1, HmPHD2, and HmPHD3 were 2981, 1954, and 1847 base pair (bp), and contained 1449, 1080, and 738 bp open reading frames (ORFs) that encoded 482, 359, and 245 amino acids (aa), respectively. Amino acid sequence analysis showed that HmPHD1, HmPHD2, and HmPHD3 had the conserved prolyl 4-hydroxylase alpha subunit homolog domains at their C-termini. Meanwhile, the evaluation of phylogeny revealed PHD2 and PHD3 of H. molitrix were more closely related as they belonged to sister clades, whereas the clade of PHD1 was relatively distant from these two. The transcripts of PHD genes are ubiquitously distributed in H. molitrix tissues, with the highest expressional level of HmPHD1 and HmPHD3 in liver, and HmPHD2 in muscle. After acute hypoxic treatment for 0.5 h, PHD genes of H. molitrix were induced mainly in liver and brain, and different from HmPHD1 and HmPHD2, the expression of HmPHD3 showed no overt tissue specificity. Furthermore, under continued hypoxic condition, PHD genes exhibited an obviously rapid but gradually attenuated response from 3 h to 24 h, and upon reoxygenation, the transcriptional expression of PHD genes showed a decreasing trend in most of the tissues. These results indicate that the PHD genes of H. molitrix are involved in the early response to hypoxic stress, and they show tissue-specific transcript expression when performing physiological regulation functions. This study is of great relevance for advancing our understanding of how PHD genes are regulated when addressing the hypoxic challenge and provides a reference for the subsequent research of the molecular mechanisms underlying hypoxia adaptation in silver carp.
Collapse
|
33
|
Tombor LS, Dimmeler S. Why is endothelial resilience key to maintain cardiac health? Basic Res Cardiol 2022; 117:35. [PMID: 35834003 PMCID: PMC9283358 DOI: 10.1007/s00395-022-00941-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 02/01/2023]
Abstract
Myocardial injury as induced by myocardial infarction results in tissue ischemia, which critically incepts cardiomyocyte death. Endothelial cells play a crucial role in restoring oxygen and nutrient supply to the heart. Latest advances in single-cell multi-omics, together with genetic lineage tracing, reveal a transcriptional and phenotypical adaptation to the injured microenvironment, which includes alterations in metabolic, mesenchymal, hematopoietic and pro-inflammatory signatures. The extent of transition in mesenchymal or hematopoietic cell lineages is still debated, but it is clear that several of the adaptive phenotypical changes are transient and endothelial cells revert back to a naïve cell state after resolution of injury responses. This resilience of endothelial cells to acute stress responses is important for preventing chronic dysfunction. Here, we summarize how endothelial cells adjust to injury and how this dynamic response contributes to repair and regeneration. We will highlight intrinsic and microenvironmental factors that contribute to endothelial cell resilience and may be targetable to maintain a functionally active, healthy microcirculation.
Collapse
Affiliation(s)
- Lukas S. Tombor
- Institute of Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany ,Faculty for Biological Sciences, Goethe University Frankfurt, Frankfurt, Germany
| | - Stefanie Dimmeler
- Institute of Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany ,Faculty for Biological Sciences, Goethe University Frankfurt, Frankfurt, Germany
| |
Collapse
|
34
|
Abstract
Significance: Vitamin C (ascorbate), in regard to its effectiveness against malignancies, has had a controversial history in cancer treatment. It has been shown that in vitro and in vivo anticancer efficacy of ascorbate relies on its pro-oxidant effect mainly from an increased generation of reactive oxygen species (ROS). A growing understanding of its anticancer activities and pharmacokinetic properties has prompted scientists to re-evaluate the significance of ascorbate in cancer treatment. Recent Advances: A recent resurge in ascorbate research emerged after discovering that, at high doses, ascorbate preferentially kills Kirsten-Ras (K-ras)- and B-raf oncogene (BRAF)-mutant cancer cells. In addition, some of the main hallmarks of cancer cells, such as redox homeostasis and oxygen-sensing regulation (through inhibition of hypoxia-inducible factor-1 alpha [HIF-1α] activity), are affected by vitamin C. Critical Issues: Currently, there is no clear consensus from the literature in regard to the beneficial effects of antioxidants. Results from both human and animal studies provide no clear evidence about the benefit of antioxidant treatment in preventing or suppressing cancer development. Since pro-oxidants may affect both normal and tumor cells, the extremely low toxicity of ascorbate represents a main advantage. This guarantees the safe inclusion of ascorbate in clinical protocols to treat cancer patients. Future Directions: Current research could focus on elucidating the wide array of reactions between ascorbate and reactive species, namely ROS, reactive nitrogen species as well as reactive sulfide species, and their intracellular molecular targets. Unraveling these mechanisms could allow researchers to assess what could be the optimal combination of ascorbate with standard treatments.
Collapse
Affiliation(s)
- Christophe Glorieux
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, P. R. China
| | - Pedro Buc Calderon
- Química y Farmacia, Facultad de Ciencias de la Salud, Universidad Arturo Prat, Iquique, Chile.,Research Group in Metabolism and Nutrition, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
35
|
Qiu Q, Zou H, Zou H, Jing T, Li X, Yan G, Geng N, Zhang B, Zhang Z, Zhang S, Yao B, Zhang G, Zou C. 3-Bromopyruvate-induced glycolysis inhibition impacts larval growth and development and carbohydrate homeostasis in fall webworm, Hyphantria cunea Drury. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2021; 179:104961. [PMID: 34802511 DOI: 10.1016/j.pestbp.2021.104961] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 08/26/2021] [Accepted: 08/27/2021] [Indexed: 06/13/2023]
Abstract
As a typical glycolytic inhibitor, 3-bromopyruvate (3-BrPA) has been extensively studied in cancer therapy in recent decades. However, few studies focused on 3-BrPA in regulating the growth and development of insects, and the relationship and regulatory mechanism between glycolysis and chitin biosynthesis remain largely unknown. The Hyphantria cunea, named fall webworm, is a notorious defoliator, which caused a huge economic loss to agriculture and forestry. Here, we investigated the effects of 3-BrPA on the growth and development, glycolysis, carbohydrate homeostasis, as well as chitin synthesis in H. cunea larvae. To elucidate the action mechanism of 3-BrPA on H. cunea will provide a new insight for the control of this pest. The results showed that 3-BrPA dramatically restrained the growth and development of H. cunea larvae and resulted in larval lethality. Meanwhile, we confirmed that 3-BrPA caused a significant decrease in carbohydrate, adenosine triphosphate (ATP), pyruvic acid (PA), and triglyceride (TG) levels by inhibiting glycolysis in H. cunea larvae. Further studies indicated that 3-BrPA significantly affected the activities of hexokinase (HK), phosphofructokinase (PFK), pyruvate kinase (PK), glucose 6-phosphate dehydrogenase (G6PDH) and trehalase, as well as expressions of the genes related to glycolysis, resulting in carbohydrate homeostasis disorder. Moreover, it was found that 3-BrPA enhanced 20-hydroxyecdysone (20E) signaling by upregulating HcCYP306A1 and HcCYP314A1, two critical genes in 20E synthesis pathway, and accelerated chitin synthesis by upregulating transcriptional levels of genes in the chitin synthesis pathway in H. cunea larvae. Taken together, our findings provide a novel insight into the mechanism of glycolytic inhibitor in regulating the growth and development of insects, and lay a foundation for the potential application of glycolytic inhibitors in pest control as well.
Collapse
Affiliation(s)
- Qian Qiu
- School of Forestry, Northeast Forestry University, Harbin 150040, PR China
| | - Haifeng Zou
- School of Forestry, Northeast Forestry University, Harbin 150040, PR China
| | - Hang Zou
- School of Forestry, Northeast Forestry University, Harbin 150040, PR China
| | - Tianzhong Jing
- School of Forestry, Northeast Forestry University, Harbin 150040, PR China
| | - XingPeng Li
- School of Forestry, Beihua University, Jilin 132013, PR China
| | - Gaige Yan
- School of Forestry, Northeast Forestry University, Harbin 150040, PR China
| | - Nannan Geng
- School of Forestry, Northeast Forestry University, Harbin 150040, PR China
| | - Bihan Zhang
- School of Forestry, Northeast Forestry University, Harbin 150040, PR China
| | - Zhidong Zhang
- School of Forestry, Northeast Forestry University, Harbin 150040, PR China
| | - Shengyu Zhang
- School of Forestry, Northeast Forestry University, Harbin 150040, PR China
| | - Bin Yao
- School of Forestry, Northeast Forestry University, Harbin 150040, PR China
| | - Guocai Zhang
- School of Forestry, Northeast Forestry University, Harbin 150040, PR China.
| | - Chuanshan Zou
- School of Forestry, Northeast Forestry University, Harbin 150040, PR China.
| |
Collapse
|
36
|
Jones AK, Rozance PJ, Brown LD, Lorca RA, Julian CG, Moore LG, Limesand SW, Wesolowski SR. Uteroplacental nutrient flux and evidence for metabolic reprogramming during sustained hypoxemia. Physiol Rep 2021; 9:e15033. [PMID: 34558219 PMCID: PMC8461030 DOI: 10.14814/phy2.15033] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 08/23/2021] [Indexed: 01/14/2023] Open
Abstract
Gestational hypoxemia is often associated with reduced birth weight, yet how hypoxemia controls uteroplacental nutrient metabolism and supply to the fetus is unclear. This study tested the effects of maternal hypoxemia (HOX) between 0.8 and 0.9 gestation on uteroplacental nutrient metabolism and flux to the fetus in pregnant sheep. Despite hypoxemia, uteroplacental and fetal oxygen utilization and net glucose and lactate uptake rates were similar in HOX (n = 11) compared to CON (n = 7) groups. HOX fetuses had increased lactate and pyruvate concentrations and increased net pyruvate output to the utero-placenta. In the HOX group, uteroplacental flux of alanine to the fetus was decreased, as was glutamate flux from the fetus. HOX fetuses had increased alanine and decreased aspartate, serine, and glutamate concentrations. In HOX placental tissue, we identified hypoxic responses that should increase mitochondrial efficiency (decreased SDHB, increased COX4I2) and increase lactate production from pyruvate (increased LDHA protein and LDH activity, decreased LDHB and MPC2), both resembling metabolic reprogramming, but with evidence for decreased (PFK1, PKM2), rather than increased, glycolysis and AMPK phosphorylation. This supports a fetal-uteroplacental shuttle during sustained hypoxemia whereby uteroplacental tissues produce lactate as fuel for the fetus using pyruvate released from the fetus, rather than pyruvate produced from glucose in the placenta, given the absence of increased uteroplacental glucose uptake and glycolytic gene activation. Together, these results provide new mechanisms for how hypoxemia, independent of AMPK activation, regulates uteroplacental metabolism and nutrient allocation to the fetus, which allow the fetus to defend its oxidative metabolism and growth.
Collapse
Affiliation(s)
- Amanda K. Jones
- Perinatal Research Center, Department of PediatricsUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Paul J. Rozance
- Perinatal Research Center, Department of PediatricsUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Laura D. Brown
- Perinatal Research Center, Department of PediatricsUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Ramón A. Lorca
- Department of Obstetrics and GynecologyUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Colleen G. Julian
- Department of MedicineUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Lorna G. Moore
- Department of Obstetrics and GynecologyUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Sean W. Limesand
- School of Animal and Comparative Biomedical SciencesUniversity of ArizonaTucsonArizonaUSA
| | - Stephanie R. Wesolowski
- Perinatal Research Center, Department of PediatricsUniversity of Colorado School of MedicineAuroraColoradoUSA
| |
Collapse
|
37
|
Oyama Y, Walker LA, Eckle T. Targeting circadian PER2 as therapy in myocardial ischemia and reperfusion injury. Chronobiol Int 2021; 38:1262-1273. [PMID: 34034593 PMCID: PMC8355134 DOI: 10.1080/07420528.2021.1928160] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/30/2021] [Accepted: 05/04/2021] [Indexed: 01/22/2023]
Abstract
The cycle of day and night dominates life on earth. Therefore, almost all living organisms adopted a molecular clock linked to the light-dark cycles. It is now well established that this molecular clock is crucial for human health and wellbeing. Disruption of the molecular clockwork directly results in a myriad of disorders, including cardiovascular diseases. Further, the onset of many cardiovascular diseases such as acute myocardial infarction exhibits a circadian periodicity with worse outcomes in the early morning hours. Based on these observations, the research community became interested in manipulating the molecular clock to treat cardiovascular diseases. In recent years, several exciting discoveries of pharmacological agents or molecular mechanisms targeting the molecular clockwork have paved the way for circadian medicine's arrival in cardiovascular diseases. The current review will outline the most recent circadian therapeutic advances related to the circadian rhythm protein Period2 (PER2) to treat myocardial ischemia and summarize future research in the respective field.
Collapse
Affiliation(s)
- Yoshimasa Oyama
- Department of Anesthesiology, University of Colorado Denver School of Medicine, Aurora, Colorado, USA
- Department of Anesthesiology and Intensive Care Medicine, Oita University Faculty of Medicine, Oita, Japan
| | - Lori A Walker
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Tobias Eckle
- Department of Anesthesiology, University of Colorado Denver School of Medicine, Aurora, Colorado, USA
- Department of Cell and Developmental Biology, University of Colorado Denver School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
38
|
Ullah K, Wu R. Hypoxia-Inducible Factor Regulates Endothelial Metabolism in Cardiovascular Disease. Front Physiol 2021; 12:670653. [PMID: 34290616 PMCID: PMC8287728 DOI: 10.3389/fphys.2021.670653] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 05/13/2021] [Indexed: 12/30/2022] Open
Abstract
Endothelial cells (ECs) form a physical barrier between the lumens and vascular walls of arteries, veins, capillaries, and lymph vessels; thus, they regulate the extravasation of nutrients and oxygen from the circulation into the perivascular space and participate in mechanisms that maintain cardiovascular homeostasis and promote tissue growth and repair. Notably, their role in tissue repair is facilitated, at least in part, by their dependence on glycolysis for energy production, which enables them to resist hypoxic damage and promote angiogenesis in ischemic regions. ECs are also equipped with a network of oxygen-sensitive molecules that collectively activate the response to hypoxic injury, and the master regulators of the hypoxia response pathway are hypoxia-inducible factors (HIFs). HIFs reinforce the glycolytic dependence of ECs under hypoxic conditions, but whether HIF activity attenuates or exacerbates the progression and severity of cardiovascular dysfunction varies depending on the disease setting. This review summarizes how HIF regulates the metabolic and angiogenic activity of ECs under both normal and hypoxic conditions and in a variety of diseases that are associated with cardiovascular complications.
Collapse
Affiliation(s)
- Karim Ullah
- Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Rongxue Wu
- Biological Sciences Division, Department of Medicine, University of Chicago, Chicago, IL, United States
| |
Collapse
|
39
|
Zamora-Briseño JA, Améndola-Pimenta M, Ortega-Rosas DA, Pereira-Santana A, Hernández-Velázquez IM, González-Penagos CE, Pérez-Vega JA, Del Río-García M, Árcega-Cabrera F, Rodríguez-Canul R. Gill and liver transcriptomic responses of Achirus lineatus (Neopterygii: Achiridae) exposed to water-accommodated fraction (WAF) of light crude oil reveal an onset of hypoxia-like condition. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:34309-34327. [PMID: 33646544 DOI: 10.1007/s11356-021-12909-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 02/08/2021] [Indexed: 06/12/2023]
Abstract
Crude oil is one of the most widespread pollutants released into the marine environment, and native species have provided useful information about the effect of crude oil pollution in marine ecosystems. We consider that the lined sole Achirus lineatus can be a useful monitor of the effect of crude oil in the Gulf of Mexico (GoM) because this flounder species has a wide distribution along the GoM, and its response to oil components is relevant. The objective of this study was to compare the transcriptomic changes in liver and gill of adults lined sole fish (Achirus lineatus) exposed to a sublethal acute concentration of water-accommodated fraction (WAF) of light crude oil for 48 h. RNA-Seq was performed to assess the transcriptional changes in both organs. A total of 1073 differentially expressed genes (DEGs) were detected in gills; 662 (61.69%) were upregulated, and 411 (38.30%) were downregulated whereas in liver, 515 DEGs; 306 (59.42%) were upregulated, and 209 (40.58%) were downregulated. Xenobiotic metabolism and redox metabolism, along with DNA repair mechanisms, were activated. The induction of hypoxia-regulated genes and the generalized regulation of multiple signaling pathways support the hypothesis that WAF exposition causes a hypoxia-like condition.
Collapse
Affiliation(s)
- Jesús Alejandro Zamora-Briseño
- Departamento de Recursos del Mar, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional-Unidad Mérida, Km 6 Antigua Carretera a Progreso, CORDEMEX, CP 97310, Mérida, Yucatán, Mexico
| | - Monica Améndola-Pimenta
- Departamento de Recursos del Mar, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional-Unidad Mérida, Km 6 Antigua Carretera a Progreso, CORDEMEX, CP 97310, Mérida, Yucatán, Mexico
| | | | - Alejandro Pereira-Santana
- División de Biotecnología Industrial, CONACYT-Centro de Investigación y Asistencia en Tecnología y Diseño del estado de Jalisco, Camino Arenero 1227, El Bajío, C.P. 45019, Zapopan, Jalisco, Mexico
| | - Ioreni Margarita Hernández-Velázquez
- Departamento de Recursos del Mar, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional-Unidad Mérida, Km 6 Antigua Carretera a Progreso, CORDEMEX, CP 97310, Mérida, Yucatán, Mexico
| | - Carlos Eduardo González-Penagos
- Departamento de Recursos del Mar, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional-Unidad Mérida, Km 6 Antigua Carretera a Progreso, CORDEMEX, CP 97310, Mérida, Yucatán, Mexico
| | - Juan Antonio Pérez-Vega
- Departamento de Recursos del Mar, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional-Unidad Mérida, Km 6 Antigua Carretera a Progreso, CORDEMEX, CP 97310, Mérida, Yucatán, Mexico
| | - Marcela Del Río-García
- Departamento de Recursos del Mar, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional-Unidad Mérida, Km 6 Antigua Carretera a Progreso, CORDEMEX, CP 97310, Mérida, Yucatán, Mexico
| | - Flor Árcega-Cabrera
- Unidad de Química Sisal, Facultad de Química, Universidad Nacional Autónoma de México, Puerto de Abrigo S/N, 97356, Sisal, Yucatán, Mexico
| | - Rossanna Rodríguez-Canul
- Departamento de Recursos del Mar, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional-Unidad Mérida, Km 6 Antigua Carretera a Progreso, CORDEMEX, CP 97310, Mérida, Yucatán, Mexico.
- Laboratorio de Inmunología y Biología Molecular, CINVESTAV-IPN Unidad Mérida, Antigua carretera a Progreso Km 6., CP 97310, Mérida, Yucatán, Mexico.
| |
Collapse
|
40
|
Yu W, Yang X, Zhang Q, Sun L, Yuan S, Xin Y. Targeting GLS1 to cancer therapy through glutamine metabolism. Clin Transl Oncol 2021; 23:2253-2268. [PMID: 34023970 DOI: 10.1007/s12094-021-02645-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 05/12/2021] [Indexed: 12/22/2022]
Abstract
Glutamine metabolism is one of the hallmarks of cancers which is described as an essential role in serving as a major energy and building blocks supply to cell proliferation in cancer cells. Many malignant tumor cells always display glutamine addiction. The "kidney-type" glutaminase (GLS1) is a metabolism enzyme which plays a significant part in glutaminolysis. Interestingly, GLS1 is often overexpressed in highly proliferative cancer cells to fulfill enhanced glutamine demand. So far, GLS1 has been proved to be a significant target during the carcinogenesis process, and emerging evidence reveals that its inhibitors could provide a benefit strategy for cancer therapy. Herein, we summarize the prognostic value of GLS1 in multiple cancer type and its related regulatory factors which are associated with antitumor activity. Moreover, this review article highlights the remarkable reform of discovery and development for GLS1 inhibitors. On the basis of case studies, our perspectives for targeting GLS1 and development of GLS1 antagonist are discussed in the final part.
Collapse
Affiliation(s)
- Wei Yu
- China Pharmaceutical University, Nanjing, 21000, Jiangsu, China
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital, Zhuhai Hospital Affiliated With Jinan University, Jinan University, Zhuhai, 519000, Guangdong, China
| | - XiangYu Yang
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital, Zhuhai Hospital Affiliated With Jinan University, Jinan University, Zhuhai, 519000, Guangdong, China
| | - Qian Zhang
- China Pharmaceutical University, Nanjing, 21000, Jiangsu, China
| | - Li Sun
- China Pharmaceutical University, Nanjing, 21000, Jiangsu, China
| | - ShengTao Yuan
- China Pharmaceutical University, Nanjing, 21000, Jiangsu, China.
| | - YongJie Xin
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital, Zhuhai Hospital Affiliated With Jinan University, Jinan University, Zhuhai, 519000, Guangdong, China.
| |
Collapse
|
41
|
Heinemann-Yerushalmi L, Bentovim L, Felsenthal N, Vinestock RC, Michaeli N, Krief S, Silberman A, Cohen M, Ben-Dor S, Brenner O, Haffner-Krausz R, Itkin M, Malitsky S, Erez A, Zelzer E. BCKDK regulates the TCA cycle through PDC in the absence of PDK family during embryonic development. Dev Cell 2021; 56:1182-1194.e6. [PMID: 33773101 DOI: 10.1016/j.devcel.2021.03.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 12/10/2020] [Accepted: 03/01/2021] [Indexed: 12/15/2022]
Abstract
Pyruvate dehydrogenase kinases (PDK1-4) inhibit the TCA cycle by phosphorylating pyruvate dehydrogenase complex (PDC). Here, we show that PDK family is dispensable for murine embryonic development and that BCKDK serves as a compensatory mechanism by inactivating PDC. First, we knocked out all four Pdk genes one by one. Surprisingly, Pdk total KO embryos developed and were born in expected ratios but died by postnatal day 4 because of hypoglycemia or ketoacidosis. Moreover, PDC was phosphorylated in these embryos, suggesting that another kinase compensates for PDK family. Bioinformatic analysis implicated branched-chain ketoacid dehydrogenase kinase (Bckdk), a key regulator of branched-chain amino acids (BCAAs) catabolism. Indeed, knockout of Bckdk and Pdk family led to the loss of PDC phosphorylation, an increase in PDC activity and pyruvate entry into the TCA cycle, and embryonic lethality. These findings reveal a regulatory crosstalk hardwiring BCAA and glucose catabolic pathways, which feed the TCA cycle.
Collapse
Affiliation(s)
| | - Lital Bentovim
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Neta Felsenthal
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ron Carmel Vinestock
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Nofar Michaeli
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Sharon Krief
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Alon Silberman
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Marina Cohen
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Shifra Ben-Dor
- Bioinformatics and Biological Computing Unit, Biological Services, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Ori Brenner
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Rebecca Haffner-Krausz
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Maxim Itkin
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Sergey Malitsky
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ayelet Erez
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Elazar Zelzer
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel.
| |
Collapse
|
42
|
Application of Metabolomics in Pediatric Asthma: Prediction, Diagnosis and Personalized Treatment. Metabolites 2021; 11:metabo11040251. [PMID: 33919626 PMCID: PMC8072856 DOI: 10.3390/metabo11040251] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/07/2021] [Accepted: 04/15/2021] [Indexed: 12/16/2022] Open
Abstract
Asthma in children remains a significant public health challenge affecting 5–20% of children in Europe and is associated with increased morbidity and societal healthcare costs. The high variation in asthma incidence among countries may be attributed to differences in genetic susceptibility and environmental factors. This respiratory disorder is described as a heterogeneous syndrome of multiple clinical manifestations (phenotypes) with varying degrees of severity and airway hyper-responsiveness, which is based on patient symptoms, lung function and response to pharmacotherapy. However, an accurate diagnosis is often difficult due to diversities in clinical presentation. Therefore, identifying early diagnostic biomarkers and improving the monitoring of airway dysfunction and inflammatory through non-invasive methods are key goals in successful pediatric asthma management. Given that asthma is caused by the interaction between genes and environmental factors, an emerging approach, metabolomics—the systematic analysis of small molecules—can provide more insight into asthma pathophysiological mechanisms, enable the identification of early biomarkers and targeted personalized therapies, thus reducing disease burden and societal cost. The purpose of this review is to present evidence on the utility of metabolomics in pediatric asthma through the analysis of intermediate metabolites of biochemical pathways that involve carbohydrates, amino acids, lipids, organic acids and nucleotides and discuss their potential application in clinical practice. Also, current challenges on the integration of metabolomics in pediatric asthma management and needed next steps are critically discussed.
Collapse
|
43
|
Novais A, Chatzopoulou E, Chaussain C, Gorin C. The Potential of FGF-2 in Craniofacial Bone Tissue Engineering: A Review. Cells 2021; 10:932. [PMID: 33920587 PMCID: PMC8073160 DOI: 10.3390/cells10040932] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/10/2021] [Accepted: 04/15/2021] [Indexed: 12/21/2022] Open
Abstract
Bone is a hard-vascularized tissue, which renews itself continuously to adapt to the mechanical and metabolic demands of the body. The craniofacial area is prone to trauma and pathologies that often result in large bone damage, these leading to both aesthetic and functional complications for patients. The "gold standard" for treating these large defects is autologous bone grafting, which has some drawbacks including the requirement for a second surgical site with quantity of bone limitations, pain and other surgical complications. Indeed, tissue engineering combining a biomaterial with the appropriate cells and molecules of interest would allow a new therapeutic approach to treat large bone defects while avoiding complications associated with a second surgical site. This review first outlines the current knowledge of bone remodeling and the different signaling pathways involved seeking to improve our understanding of the roles of each to be able to stimulate or inhibit them. Secondly, it highlights the interesting characteristics of one growth factor in particular, FGF-2, and its role in bone homeostasis, before then analyzing its potential usefulness in craniofacial bone tissue engineering because of its proliferative, pro-angiogenic and pro-osteogenic effects depending on its spatial-temporal use, dose and mode of administration.
Collapse
Affiliation(s)
- Anita Novais
- Pathologies, Imagerie et Biothérapies Orofaciales, Université de Paris, URP2496, 1 rue Maurice Arnoux, 92120 Montrouge, France; (A.N.); (E.C.); (C.C.)
- AP-HP Département d’Odontologie, Services d’odontologie, GH Pitié Salpêtrière, Henri Mondor, Paris Nord, Hôpital Rothschild, Paris, France
| | - Eirini Chatzopoulou
- Pathologies, Imagerie et Biothérapies Orofaciales, Université de Paris, URP2496, 1 rue Maurice Arnoux, 92120 Montrouge, France; (A.N.); (E.C.); (C.C.)
- AP-HP Département d’Odontologie, Services d’odontologie, GH Pitié Salpêtrière, Henri Mondor, Paris Nord, Hôpital Rothschild, Paris, France
- Département de Parodontologie, Université de Paris, UFR Odontologie-Garancière, 75006 Paris, France
| | - Catherine Chaussain
- Pathologies, Imagerie et Biothérapies Orofaciales, Université de Paris, URP2496, 1 rue Maurice Arnoux, 92120 Montrouge, France; (A.N.); (E.C.); (C.C.)
- AP-HP Département d’Odontologie, Services d’odontologie, GH Pitié Salpêtrière, Henri Mondor, Paris Nord, Hôpital Rothschild, Paris, France
| | - Caroline Gorin
- Pathologies, Imagerie et Biothérapies Orofaciales, Université de Paris, URP2496, 1 rue Maurice Arnoux, 92120 Montrouge, France; (A.N.); (E.C.); (C.C.)
- AP-HP Département d’Odontologie, Services d’odontologie, GH Pitié Salpêtrière, Henri Mondor, Paris Nord, Hôpital Rothschild, Paris, France
| |
Collapse
|
44
|
Ju C, Wang M, Tak E, Kim B, Emontzpohl C, Yang Y, Yuan X, Kutay H, Liang Y, Hall DR, Dar WA, Bynon JS, Carmeliet P, Ghoshal K, Eltzschig HK. Hypoxia-inducible factor-1α-dependent induction of miR122 enhances hepatic ischemia tolerance. J Clin Invest 2021; 131:140300. [PMID: 33792566 PMCID: PMC8011886 DOI: 10.1172/jci140300] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 02/10/2021] [Indexed: 12/29/2022] Open
Abstract
Hepatic ischemia and reperfusion (IR) injury contributes to the morbidity and mortality associated with liver transplantation. microRNAs (miRNAs) constitute a family of noncoding RNAs that regulate gene expression at the posttranslational level through the repression of specific target genes. Here, we hypothesized that miRNAs could be targeted to enhance hepatic ischemia tolerance. A miRNA screen in a murine model of hepatic IR injury pointed us toward the liver-specific miRNA miR122. Subsequent studies in mice with hepatocyte-specific deletion of miR122 (miR122loxP/loxP Alb-Cre+ mice) during hepatic ischemia and reperfusion revealed exacerbated liver injury. Transcriptional studies implicated hypoxia-inducible factor-1α (HIF1α) in the induction of miR122 and identified the oxygen-sensing prolyl hydroxylase domain 1 (PHD1) as a miR122 target. Further studies indicated that HIF1α-dependent induction of miR122 participated in a feed-forward pathway for liver protection via the enhancement of hepatic HIF responses through PHD1 repression. Moreover, pharmacologic studies utilizing nanoparticle-mediated miR122 overexpression demonstrated attenuated liver injury. Finally, proof-of-principle studies in patients undergoing orthotopic liver transplantation showed elevated miR122 levels in conjunction with the repression of PHD1 in post-ischemic liver biopsies. Taken together, the present findings provide molecular insight into the functional role of miR122 in enhancing hepatic ischemia tolerance and suggest the potential utility of pharmacologic interventions targeting miR122 to dampen hepatic injury during liver transplantation.
Collapse
Affiliation(s)
- Cynthia Ju
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Texas, USA
| | - Meng Wang
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Texas, USA
| | - Eunyoung Tak
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Boyun Kim
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Texas, USA
| | - Christoph Emontzpohl
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Texas, USA
| | - Yang Yang
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Texas, USA
| | - Xiaoyi Yuan
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Texas, USA
| | - Huban Kutay
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Yafen Liang
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Texas, USA
| | - David R. Hall
- Department of Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Wasim A. Dar
- Department of Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - J. Steve Bynon
- Department of Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, and
- Center for Cancer Biology, Department of Oncology, Katholieke University Leuven, Leuven, Belgium
| | - Kalpana Ghoshal
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
- Department of Pathology, The Ohio State University, Columbus, Ohio, USA
| | - Holger K. Eltzschig
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Texas, USA
| |
Collapse
|
45
|
Derry PJ, Vo ATT, Gnanansekaran A, Mitra J, Liopo AV, Hegde ML, Tsai AL, Tour JM, Kent TA. The Chemical Basis of Intracerebral Hemorrhage and Cell Toxicity With Contributions From Eryptosis and Ferroptosis. Front Cell Neurosci 2020; 14:603043. [PMID: 33363457 PMCID: PMC7755086 DOI: 10.3389/fncel.2020.603043] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 10/06/2020] [Indexed: 12/12/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a particularly devastating event both because of the direct injury from space-occupying blood to the sequelae of the brain exposed to free blood components from which it is normally protected. Not surprisingly, the usual metabolic and energy pathways are overwhelmed in this situation. In this review article, we detail the complexity of red blood cell degradation, the contribution of eryptosis leading to hemoglobin breakdown into its constituents, the participants in that process, and the points at which injury can be propagated such as elaboration of toxic radicals through the metabolism of the breakdown products. Two prominent products of this breakdown sequence, hemin, and iron, induce a variety of pathologies including free radical damage and DNA breakage, which appear to include events independent from typical oxidative DNA injury. As a result of this confluence of damaging elements, multiple pathways of injury, cell death, and survival are likely engaged including ferroptosis (which may be the same as oxytosis but viewed from a different perspective) and senescence, suggesting that targeting any single cause will likely not be a sufficient strategy to maximally improve outcome. Combination therapies in addition to safe methods to reduce blood burden should be pursued.
Collapse
Affiliation(s)
- Paul J Derry
- Center for Genomics and Precision Medicine, Department of Translational Medical Sciences, Institute of Biosciences and Technology, College of Medicine, Texas A&M Health Science Center, Houston, TX, United States
| | - Anh Tran Tram Vo
- Center for Genomics and Precision Medicine, Department of Translational Medical Sciences, Institute of Biosciences and Technology, College of Medicine, Texas A&M Health Science Center, Houston, TX, United States
| | - Aswini Gnanansekaran
- Center for Genomics and Precision Medicine, Department of Translational Medical Sciences, Institute of Biosciences and Technology, College of Medicine, Texas A&M Health Science Center, Houston, TX, United States
| | - Joy Mitra
- Department of Neurosurgery, Center for Neuroregeneration, The Houston Methodist Research Institute, Houston, TX, United States
| | - Anton V Liopo
- Center for Genomics and Precision Medicine, Department of Translational Medical Sciences, Institute of Biosciences and Technology, College of Medicine, Texas A&M Health Science Center, Houston, TX, United States
| | - Muralidhar L Hegde
- Department of Neurosurgery, Center for Neuroregeneration, The Houston Methodist Research Institute, Houston, TX, United States
| | - Ah-Lim Tsai
- Division of Hematology, Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - James M Tour
- Department of Chemistry, Rice University, Houston, TX, United States.,Department of Computer Science, George R. Brown School of Engineering, Rice University, Houston, TX, United States.,Department of Materials Science and NanoEngineering, George R. Brown School of Engineering, Rice University, Houston, TX, United States
| | - Thomas A Kent
- Center for Genomics and Precision Medicine, Department of Translational Medical Sciences, Institute of Biosciences and Technology, College of Medicine, Texas A&M Health Science Center, Houston, TX, United States.,Department of Chemistry, Rice University, Houston, TX, United States.,Stanley H. Appel Department of Neurology, Institute for Academic Medicine, Houston Methodist Hospital, Houston, TX, United States
| |
Collapse
|
46
|
Cai X, Zhou Z, Zhu J, Liao Q, Zhang D, Liu X, Wang J, Ouyang G, Xiao W. Zebrafish Hif3α modulates erythropoiesis via regulation of gata1 to facilitate hypoxia tolerance. Development 2020; 147:226111. [PMID: 33037038 DOI: 10.1242/dev.185116] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 09/24/2020] [Indexed: 12/18/2022]
Abstract
The hypoxia-inducible factors 1α and 2α (HIF1α and HIF2α) are master regulators of the cellular response to O2. In addition to HIF1α and HIF2α, HIF3α is another identified member of the HIFα family. Even though the question of whether some HIF3α isoforms have transcriptional activity or repressive activity is still under debate, it is evident that the full length of HIF3α acts as a transcription factor. However, its function in hypoxia signaling is largely unknown. Here, we show that loss of hif3 a in zebrafish reduced hypoxia tolerance. Further assays indicated that erythrocyte number was decreased because red blood cell maturation was impeded by hif3 a disruption. We found that gata1 expression was downregulated in hif3 a null zebrafish, as were several hematopoietic marker genes, including alas2, band3, hbae1, hbae3 and hbbe1 Hif3α recognized the hypoxia response element located in the promoter of gata1 and directly bound to the promoter to transactivate gata1 expression. Our results suggested that hif3 a facilities hypoxia tolerance by modulating erythropoiesis via gata1 regulation.
Collapse
Affiliation(s)
- Xiaolian Cai
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ziwen Zhou
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Junji Zhu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qian Liao
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Dawei Zhang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Xing Liu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China.,The Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China.,The Innovation of Seed Design, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Jing Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China.,The Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China.,The Innovation of Seed Design, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Gang Ouyang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China.,The Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China.,The Innovation of Seed Design, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Wuhan Xiao
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China .,The Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China.,The Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China.,University of Chinese Academy of Sciences, Beijing, 100049, China.,The Innovation of Seed Design, Chinese Academy of Sciences, Wuhan, 430072, China
| |
Collapse
|
47
|
Rey E, Meléndez‐Rodríguez F, Marañón P, Gil‐Valle M, Carrasco AG, Torres‐Capelli M, Chávez S, del Pozo‐Maroto E, Rodríguez de Cía J, Aragonés J, García‐Monzón C, González‐Rodríguez Á. Hypoxia-inducible factor 2α drives hepatosteatosis through the fatty acid translocase CD36. Liver Int 2020; 40:2553-2567. [PMID: 32432822 PMCID: PMC7539965 DOI: 10.1111/liv.14519] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 05/11/2020] [Accepted: 05/12/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS Molecular mechanisms by which hypoxia might contribute to hepatosteatosis, the earliest stage in non-alcoholic fatty liver disease (NAFLD) pathogenesis, remain still to be elucidated. We aimed to assess the impact of hypoxia-inducible factor 2α (HIF2α) on the fatty acid translocase CD36 expression and function in vivo and in vitro. METHODS CD36 expression and intracellular lipid content were determined in hypoxic hepatocytes, and in hypoxic CD36- or HIF2α -silenced human liver cells. Histological analysis, and HIF2α and CD36 expression were evaluated in livers from animals in which von Hippel-Lindau (Vhl) gene is inactivated (Vhlf/f -deficient mice), or both Vhl and Hif2a are simultaneously inactivated (Vhlf/f Hif2α/f -deficient mice), and from 33 biopsy-proven NAFLD patients and 18 subjects with histologically normal liver. RESULTS In hypoxic hepatocytes, CD36 expression and intracellular lipid content were augmented. Noteworthy, CD36 knockdown significantly reduced lipid accumulation, and HIF2A gene silencing markedly reverted both hypoxia-induced events in hypoxic liver cells. Moreover livers from Vhlf/f -deficient mice showed histologic characteristics of non-alcoholic steatohepatitis (NASH) and increased CD36 mRNA and protein amounts, whereas both significantly decreased and NASH features markedly ameliorated in Vhlf/f Hif2αf/f -deficient mice. In addition, both HIF2α and CD36 were significantly overexpressed within the liver of NAFLD patients and, interestingly, a significant positive correlation between hepatic transcript levels of CD36 and erythropoietin (EPO), a HIF2α -dependent gene target, was observed in NAFLD patients. CONCLUSIONS This study provides evidence that HIF2α drives lipid accumulation in human hepatocytes by upregulating CD36 expression and function, and could contribute to hepatosteatosis setup.
Collapse
Affiliation(s)
- Esther Rey
- Unidad de InvestigaciónHospital Universitario Santa CristinaInstituto de Investigación Sanitaria del Hospital Universitario de La PrincesaMadridSpain
| | - Florinda Meléndez‐Rodríguez
- Unidad de InvestigaciónHospital Universitario Santa CristinaInstituto de Investigación Sanitaria del Hospital Universitario de La PrincesaUniversidad Autónoma de MadridMadridSpain,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV)MadridSpain
| | - Patricia Marañón
- Unidad de InvestigaciónHospital Universitario Santa CristinaInstituto de Investigación Sanitaria del Hospital Universitario de La PrincesaMadridSpain
| | - Miriam Gil‐Valle
- Unidad de InvestigaciónHospital Universitario Santa CristinaInstituto de Investigación Sanitaria del Hospital Universitario de La PrincesaMadridSpain
| | - Almudena G. Carrasco
- Unidad de InvestigaciónHospital Universitario Santa CristinaInstituto de Investigación Sanitaria del Hospital Universitario de La PrincesaMadridSpain,Present address:
Dpto. Ciencias Básicas de la SaludUniversidad Rey Juan CarlosAlcorcónSpain
| | - Mar Torres‐Capelli
- Unidad de InvestigaciónHospital Universitario Santa CristinaInstituto de Investigación Sanitaria del Hospital Universitario de La PrincesaUniversidad Autónoma de MadridMadridSpain
| | - Stephania Chávez
- Unidad de InvestigaciónHospital Universitario Santa CristinaInstituto de Investigación Sanitaria del Hospital Universitario de La PrincesaMadridSpain
| | - Elvira del Pozo‐Maroto
- Unidad de InvestigaciónHospital Universitario Santa CristinaInstituto de Investigación Sanitaria del Hospital Universitario de La PrincesaMadridSpain,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD)MadridSpain
| | - Javier Rodríguez de Cía
- Unidad de InvestigaciónHospital Universitario Santa CristinaInstituto de Investigación Sanitaria del Hospital Universitario de La PrincesaMadridSpain,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD)MadridSpain
| | - Julián Aragonés
- Unidad de InvestigaciónHospital Universitario Santa CristinaInstituto de Investigación Sanitaria del Hospital Universitario de La PrincesaUniversidad Autónoma de MadridMadridSpain,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV)MadridSpain
| | - Carmelo García‐Monzón
- Unidad de InvestigaciónHospital Universitario Santa CristinaInstituto de Investigación Sanitaria del Hospital Universitario de La PrincesaMadridSpain,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD)MadridSpain
| | - Águeda González‐Rodríguez
- Unidad de InvestigaciónHospital Universitario Santa CristinaInstituto de Investigación Sanitaria del Hospital Universitario de La PrincesaMadridSpain,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD)MadridSpain
| |
Collapse
|
48
|
Oyama Y, Bartman CM, Bonney S, Lee JS, Walker LA, Han J, Borchers CH, Buttrick PM, Aherne CM, Clendenen N, Colgan SP, Eckle T. Intense Light-Mediated Circadian Cardioprotection via Transcriptional Reprogramming of the Endothelium. Cell Rep 2020; 28:1471-1484.e11. [PMID: 31390562 PMCID: PMC6708043 DOI: 10.1016/j.celrep.2019.07.020] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 05/15/2019] [Accepted: 07/08/2019] [Indexed: 01/10/2023] Open
Abstract
Consistent daylight oscillations and abundant oxygen availability are fundamental to human health. Here, we investigate the intersection between light-sensing (Period 2 [PER2]) and oxygen-sensing (hypoxia-inducible factor [HIF1A]) pathways in cellular adaptation to myocardial ischemia. We demonstrate that intense light is cardioprotective via circadian PER2 amplitude enhancement, mimicking hypoxia-elicited adenosine- and HIF1A-metabolic adaptation to myocardial ischemia under normoxic conditions. Whole-genome array from intense light-exposed wild-type or Per2-/- mice and myocardial ischemia in endothelial-specific PER2-deficient mice uncover a critical role for intense light in maintaining endothelial barrier function via light-enhanced HIF1A transcription. A proteomics screen in human endothelia reveals a dominant role for PER2 in metabolic reprogramming to hypoxia via mitochondrial translocation, tricarboxylic acid (TCA) cycle enzyme activity regulation, and HIF1A transcriptional adaption to hypoxia. Translational investigation of intense light in human subjects identifies similar PER2 mechanisms, implicating the use of intense light for the treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Yoshimasa Oyama
- Mucosal Inflammation Program, Departments of Medicine and Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Colleen M Bartman
- Mucosal Inflammation Program, Departments of Medicine and Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Graduate Training Program in Cell Biology, Stem Cells, and Development, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Stephanie Bonney
- Mucosal Inflammation Program, Departments of Medicine and Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Graduate Training Program in Cell Biology, Stem Cells, and Development, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - J Scott Lee
- Mucosal Inflammation Program, Departments of Medicine and Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Lori A Walker
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jun Han
- Department of Biochemistry and Microbiology, Genome BC Proteomics Centre, University of Victoria, Victoria, BC, Canada
| | - Christoph H Borchers
- Department of Biochemistry and Microbiology, Genome BC Proteomics Centre, University of Victoria, Victoria, BC, Canada; Segal Cancer Proteomics Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, Quebec, Canada; Gerald Bronfman Department of Oncology, Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - Peter M Buttrick
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Carol M Aherne
- Mucosal Inflammation Program, Departments of Medicine and Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Nathan Clendenen
- Mucosal Inflammation Program, Departments of Medicine and Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Sean P Colgan
- Mucosal Inflammation Program, Departments of Medicine and Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Tobias Eckle
- Mucosal Inflammation Program, Departments of Medicine and Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Graduate Training Program in Cell Biology, Stem Cells, and Development, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
49
|
Yu G, Li X, Zhou Z, Tang J, Wang J, Liu X, Fan S, Ouyang G, Xiao W. Zebrafish phd3 Negatively Regulates Antiviral Responses via Suppression of Irf7 Transactivity Independent of Its Prolyl Hydroxylase Activity. THE JOURNAL OF IMMUNOLOGY 2020; 205:1135-1146. [PMID: 32669312 DOI: 10.4049/jimmunol.1900902] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 06/14/2020] [Indexed: 12/30/2022]
Abstract
Prolyl hydroxylase domain (PHD)-containing enzyme 3 belongs to the Caenorhabditis elegans gene egl-9 family of prolyl hydroxylases, which has initially been revealed to hydroxylate hypoxia-inducible factor α (HIF-α) and mediate HIF-α degradation. In addition to modulating its target function by hydroxylation, PHD3 has been also shown to influence its binding partners' function independent of its prolyl hydroxylase activity. In this study, we report that overexpression of zebrafish phd3 suppresses cellular antiviral response. Moreover, disruption of phd3 in zebrafish increases the survival rate upon spring viremia of carp virus exposure. Further assays indicate that phd3 interacts with irf7 through the C-terminal IRF association domain of irf7 and diminishes K63-linked ubiquitination of irf7. However, the enzymatic activity of phd3 is not required for phd3 to inhibit irf7 transactivity. This study provides novel insights into phd3 function and sheds new light on the regulation of irf7 in retinoic acid-inducible gene I-like receptor signaling.
Collapse
Affiliation(s)
- Guangqing Yu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China; The Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Wuhan 430072, People's Republic of China; The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China; and University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Xiong Li
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China; The Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Wuhan 430072, People's Republic of China; The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China; and University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Ziwen Zhou
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China; The Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Wuhan 430072, People's Republic of China; The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China; and University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Jinhua Tang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China; The Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Wuhan 430072, People's Republic of China; The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China; and University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Jing Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China; The Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Wuhan 430072, People's Republic of China; The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China; and University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Xing Liu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China; The Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Wuhan 430072, People's Republic of China; The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China; and University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Sijia Fan
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China; The Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Wuhan 430072, People's Republic of China; The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China; and University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Gang Ouyang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China; The Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Wuhan 430072, People's Republic of China; The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China; and University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Wuhan Xiao
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China; The Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Wuhan 430072, People's Republic of China; The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China; and University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| |
Collapse
|
50
|
Tretter V, Zach ML, Böhme S, Ullrich R, Markstaller K, Klein KU. Investigating Disturbances of Oxygen Homeostasis: From Cellular Mechanisms to the Clinical Practice. Front Physiol 2020; 11:947. [PMID: 32848874 PMCID: PMC7417655 DOI: 10.3389/fphys.2020.00947] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 07/14/2020] [Indexed: 12/22/2022] Open
Abstract
Soon after its discovery in the 18th century, oxygen was applied as a therapeutic agent to treat severely ill patients. Lack of oxygen, commonly termed as hypoxia, is frequently encountered in different disease states and is detrimental to human life. However, at the end of the 19th century, Paul Bert and James Lorrain Smith identified what is known as oxygen toxicity. The molecular basis of this phenomenon is oxygen's readiness to accept electrons and to form different variants of aggressive radicals that interfere with normal cell functions. The human body has evolved to maintain oxygen homeostasis by different molecular systems that are either activated in the case of oxygen under-supply, or to scavenge and to transform oxygen radicals when excess amounts are encountered. Research has provided insights into cellular mechanisms of oxygen homeostasis and is still called upon in order to better understand related diseases. Oxygen therapy is one of the prime clinical interventions, as it is life saving, readily available, easy to apply and economically affordable. However, the current state of research also implicates a reconsidering of the liberal application of oxygen causing hyperoxia. Increasing evidence from preclinical and clinical studies suggest detrimental outcomes as a consequence of liberal oxygen therapy. In this review, we summarize concepts of cellular mechanisms regarding different forms of disturbed cellular oxygen homeostasis that may help to better define safe clinical application of oxygen therapy.
Collapse
Affiliation(s)
- Verena Tretter
- Department of Anaesthesia, General Intensive Care and Pain Therapy, Medical University Vienna, Vienna, Austria
| | | | | | | | | | | |
Collapse
|