1
|
Galkin F, Pulous FE, Fu Y, Zhang M, Pun FW, Ren F, Zhavoronkov A. Roles of hypoxia-inducible factor-prolyl hydroxylases in aging and disease. Ageing Res Rev 2024; 102:102551. [PMID: 39447706 DOI: 10.1016/j.arr.2024.102551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 10/07/2024] [Accepted: 10/15/2024] [Indexed: 10/26/2024]
Abstract
The prolyl hydroxylase domain-containing (PHD or EGL9-homologs) enzyme family is mainly known for its role in the cellular response to hypoxia. HIF-PH inhibitors can stabilize hypoxia-inducible factors (HIFs), activating transcriptional programs that promote processes such as angiogenesis and erythropoiesis to adapt to changes in oxygen levels. HIF-PH inhibitors have been clinically approved for treating several types of anaemia. While most discussions of the HIF-PH signalling axis focus on hypoxia, there is a growing recognition of its importance under normoxic conditions. Recent advances in PHD biology have highlighted the potential of targeting this pathway therapeutically for a range of aging-related diseases. In this article, we review these recent discoveries, situate them within the broader context of aging and disease, and explore current therapeutic strategies that target PHD enzymes for these indications.
Collapse
Affiliation(s)
- Fedor Galkin
- Insilico Medicine AI Ltd., Level 6, Unit 08, Block A, IRENA HQ Building, Masdar City, Abu Dhabi, UAE
| | - Fadi E Pulous
- Insilico Medicine US Inc., 1000 Massachusetts Avenue, Suite 126, Cambridge, MA 02138, United States
| | - Yanyun Fu
- Insilico Medicine Shanghai Ltd., Suite 902, Tower C, Changtai Plaza, 2889 Jinke Road, Pudong, Shanghai 201203, China
| | - Man Zhang
- Insilico Medicine Shanghai Ltd., Suite 902, Tower C, Changtai Plaza, 2889 Jinke Road, Pudong, Shanghai 201203, China
| | - Frank W Pun
- Insilico Medicine Hong Kong Ltd., Unit 310, 3/F, Building 8W, Hong Kong Science and Technology Park, Hong Kong SAR
| | - Feng Ren
- Insilico Medicine AI Ltd., Level 6, Unit 08, Block A, IRENA HQ Building, Masdar City, Abu Dhabi, UAE; Insilico Medicine Shanghai Ltd., Suite 902, Tower C, Changtai Plaza, 2889 Jinke Road, Pudong, Shanghai 201203, China; Insilico Medicine Hong Kong Ltd., Unit 310, 3/F, Building 8W, Hong Kong Science and Technology Park, Hong Kong SAR
| | - Alex Zhavoronkov
- Insilico Medicine AI Ltd., Level 6, Unit 08, Block A, IRENA HQ Building, Masdar City, Abu Dhabi, UAE; Insilico Medicine US Inc., 1000 Massachusetts Avenue, Suite 126, Cambridge, MA 02138, United States; Insilico Medicine Hong Kong Ltd., Unit 310, 3/F, Building 8W, Hong Kong Science and Technology Park, Hong Kong SAR; Insilico Medicine Canada Inc., 1250 René-Lévesque Ouest, Suite 3710, Montréal, Québec H3B 4W8, Canada; Buck Institute for Research on Aging, Novato, CA, United States.
| |
Collapse
|
2
|
Trusen S, Zimmermann JSA, Fries FN, Li Z, Chai N, Seitz B, Suiwal S, Amini M, Szentmáry N, Stachon T. Increased susceptibility of human limbal aniridia fibroblasts to oxidative stress. Exp Eye Res 2024; 248:110105. [PMID: 39303843 DOI: 10.1016/j.exer.2024.110105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 08/19/2024] [Accepted: 09/18/2024] [Indexed: 09/22/2024]
Abstract
Aniridia-associated keratopathy originates from a haploinsufficiency of the transcription factor PAX6 (PAX6+/-). In the corneal epithelium of PAX6+/- mice, a significant increase in oxidized proteins was observed, accompanied by impaired compensation for elevated oxidative stress (OS). The extent to which limbal fibroblast cells (LFCs) are affected by an increased susceptibility to OS in cases of congenital aniridia (AN) has not been determined, yet. Our aim was to examine the impact of OS on antioxidant enzyme expression in normal and AN-LFCs. Following isolation and culture of primary LFCs (n = 8) and AN-LFCs (n = 8), cells were treated with cobalt chloride for 48 h to chemically induce hypoxic conditions and OS. Subsequently, HIF-1α/-2α, PHD1/2, Nrf2, CAT, SOD1, PRDX6, and GPX1 gene expression was examined by qPCR. SOD1, PRDX6, and GPX1 protein levels were assessed from the cell lysate by Western blot. The induction of hypoxia led to reduced HIF-1α gene expression in both fibroblast groups (p≤0.008), while the decrease in PHD1 was limited to AN-LFCs (p = 0.0007). On the other hand, under hypoxic conditions, PHD2 showed higher mRNA expression in AN-LFCs compared to normal LFCs (p = 0.013). As a result of OS, the mRNA levels of Nrf2 (p<0.0001) and the antioxidant enzymes CAT (p = 0.005), SOD1 (p = 0.005), GPX1 (p = 0.002) decreased in AN-LFCs. This was accompanied by an increased protein expression of SOD1 (p = 0.019) and PRDX6 (p=0.0009). In the normal LFC group, the induced extent of OS had no impact on the gene (p≥0.151) and protein expression (p ≥ 0.629) of antioxidant enzymes, except for the GPX1 mRNA level (p = 0.027). AN-LFCs exhibit higher susceptibility to OS than normal LFCs. Therefore, in AN-LFCs, there are sustained alterations in gene and protein expression of antioxidative enzymes even after 48 h of CoCl2 treatment.
Collapse
Affiliation(s)
- Simon Trusen
- Dr. Rolf M. Schwiete Center for Limbal Stem Cell and Aniridia Research, Saarland University, Kirrberger Str. 100, 66424, Homburg/Saar, Germany.
| | - Julia Sarah Alexandra Zimmermann
- Dr. Rolf M. Schwiete Center for Limbal Stem Cell and Aniridia Research, Saarland University, Kirrberger Str. 100, 66424, Homburg/Saar, Germany
| | - Fabian Norbert Fries
- Dr. Rolf M. Schwiete Center for Limbal Stem Cell and Aniridia Research, Saarland University, Kirrberger Str. 100, 66424, Homburg/Saar, Germany; Department of Ophthalmology, Saarland University Medical Center, Kirrberger Str. 100, 66424, Homburg/Saar, Germany
| | - Zhen Li
- Dr. Rolf M. Schwiete Center for Limbal Stem Cell and Aniridia Research, Saarland University, Kirrberger Str. 100, 66424, Homburg/Saar, Germany
| | - Ning Chai
- Dr. Rolf M. Schwiete Center for Limbal Stem Cell and Aniridia Research, Saarland University, Kirrberger Str. 100, 66424, Homburg/Saar, Germany
| | - Berthold Seitz
- Department of Ophthalmology, Saarland University Medical Center, Kirrberger Str. 100, 66424, Homburg/Saar, Germany
| | - Shweta Suiwal
- Dr. Rolf M. Schwiete Center for Limbal Stem Cell and Aniridia Research, Saarland University, Kirrberger Str. 100, 66424, Homburg/Saar, Germany
| | - Maryam Amini
- Dr. Rolf M. Schwiete Center for Limbal Stem Cell and Aniridia Research, Saarland University, Kirrberger Str. 100, 66424, Homburg/Saar, Germany
| | - Nóra Szentmáry
- Dr. Rolf M. Schwiete Center for Limbal Stem Cell and Aniridia Research, Saarland University, Kirrberger Str. 100, 66424, Homburg/Saar, Germany
| | - Tanja Stachon
- Dr. Rolf M. Schwiete Center for Limbal Stem Cell and Aniridia Research, Saarland University, Kirrberger Str. 100, 66424, Homburg/Saar, Germany
| |
Collapse
|
3
|
Liu Y, Suhail Y, Novin A, Afzal J, Pant A, Kshitiz. Lactate in breast cancer cells is associated with evasion of hypoxia-induced cell cycle arrest and adverse patient outcome. Hum Cell 2024; 37:768-781. [PMID: 38478356 PMCID: PMC11256967 DOI: 10.1007/s13577-024-01046-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 02/14/2024] [Indexed: 04/15/2024]
Abstract
Tumor hypoxia is a common microenvironmental factor in breast cancers, resulting in stabilization of Hypoxia-Inducible Factor 1 (HIF-1), the master regulator of hypoxic response in cells. Metabolic adaptation by HIF-1 results in inhibition of citric acid cycle, causing accumulation of lactate in large concentrations in hypoxic cancers. Lactate can therefore serve as a secondary microenvironmental factor influencing cellular response to hypoxia. Presence of lactate can alter the hypoxic response of breast cancers in many ways, sometimes in opposite manners. Lactate stabilizes HIF-1 in oxidative condition, as well as destabilizes HIF-1 in hypoxia, increases cellular acidification, and mitigates HIF-1-driven inhibition of cellular respiration. We therefore tested the effect of lactate in MDA-MB-231 under hypoxia, finding that lactate can activate pathways associated with DNA replication, and cell cycling, as well as tissue morphogenesis associated with invasive processes. Using a bioengineered nano-patterned stromal invasion assay, we also confirmed that high lactate and induced HIF-1α gene overexpression can synergistically promote MDA-MB-231 dissemination and stromal trespass. Furthermore, using The Cancer Genome Atlas, we also surprisingly found that lactate in hypoxia promotes gene expression signatures prognosticating low survival in breast cancer patients. Our work documents that lactate accumulation contributes to increased heterogeneity in breast cancer gene expression promoting cancer growth and reducing patient survival.
Collapse
Affiliation(s)
- Yamin Liu
- Department of Biomedical Engineering, University of Connecticut Health, Farmington, CT, USA
| | - Yasir Suhail
- Department of Biomedical Engineering, University of Connecticut Health, Farmington, CT, USA
- Center for Cell Analysis and Modeling, University of Connecticut Health, Farmington, CT, USA
| | - Ashkan Novin
- Department of Biomedical Engineering, University of Connecticut Health, Farmington, CT, USA
| | - Junaid Afzal
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Aditya Pant
- Department of Biomedical Engineering, University of Connecticut Health, Farmington, CT, USA
- NEAG Comprehensive Cancer Center, University of Connecticut Health, Farmington, CT, USA
| | - Kshitiz
- Department of Biomedical Engineering, University of Connecticut Health, Farmington, CT, USA.
- Center for Cell Analysis and Modeling, University of Connecticut Health, Farmington, CT, USA.
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA.
- NEAG Comprehensive Cancer Center, University of Connecticut Health, Farmington, CT, USA.
| |
Collapse
|
4
|
Fu X, Luo X, Xiao P, Guo N. Yin Yang 1 facilitates the activation, inflammation, and extracellular matrix deposition of hepatic stellate cells in hepatic fibrosis. Pathol Int 2024; 74:197-209. [PMID: 38353379 DOI: 10.1111/pin.13410] [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: 08/28/2023] [Revised: 01/02/2024] [Accepted: 01/13/2024] [Indexed: 04/11/2024]
Abstract
Chronic hepatic diseases often involve fibrosis as a pivotal factor in their progression. This study investigates the regulatory mechanisms of Yin Yang 1 (YY1) in hepatic fibrosis. Our data reveal that YY1 binds to the prolyl hydroxylase domain 1 (PHD1) promoter. Rats treated with carbon tetrachloride (CCl4) display heightened fibrosis in liver tissues, accompanied by increased levels of YY1, PHD1, and the fibrosis marker alpha-smooth muscle actin (α-SMA). Elevated levels of YY1, PHD1, and α-SMA are observed in the liver tissues of CCl4-treated rats, primary hepatic stellate cells (HSCs) isolated from fibrotic liver tissues, and transforming growth factor beta-1 (TGF-β1)-induced HSCs. The human HSC cell line LX-2, upon YY1 overexpression, exhibits enhanced TGF-β1-induced activation, leading to increased expression of extracellular matrix (ECM)-related proteins and inflammatory cytokines. YY1 silencing produces the opposite effect. YY1 exerts a positive regulatory effect on the activation of the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway and PHD1 expression. PHD1 silencing rescues the promotion of YY1 in cell activation, ECM-related protein expression, and inflammatory cytokine production in TGF-β1-treated LX-2 cells. Overall, our findings propose a model wherein YY1 facilitates TGF-β1-induced HSC activation, ECM-related protein expression, and inflammatory cytokine production by promoting PHD1 expression and activating the PI3K/AKT signaling pathway. This study positions YY1 as a promising therapeutic target for hepatic fibrosis.
Collapse
Affiliation(s)
- Xiao Fu
- General Medicine Department, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, People's Republic of China
| | - Xin Luo
- General Medicine Department, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, People's Republic of China
| | - Ping Xiao
- General Medicine Department, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, People's Republic of China
| | - Ninghong Guo
- Clinical Trial Center, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, People's Republic of China
| |
Collapse
|
5
|
Stojkovic L, Jovanovic I, Dincic E, Djordjevic A, Kuveljic J, Djuric T, Stankovic A, Vojinovic S, Zivkovic M. Targeted RNAseq Revealed the Gene Expression Signature of Ferroptosis-Related Processes Associated with Disease Severity in Patients with Multiple Sclerosis. Int J Mol Sci 2024; 25:3016. [PMID: 38474262 DOI: 10.3390/ijms25053016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 02/15/2024] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
Detrimental molecular processes in multiple sclerosis (MS) lead to the cellular accumulation of lipid peroxidation products and iron in the CNS, which represents the main driving force for ferroptosis. Ferroptosis is an iron-dependent form of regulated cell death, with proposed roles in neurodegeneration, oligodendrocyte loss and neuroinflammation in the pathogenesis of MS. Ferroptosis-related gene expression signature and molecular markers, which could reflect MS severity and progression, are currently understudied in humans. To tackle these challenges, we have applied a curated approach to create and experimentally analyze a comprehensive panel of ferroptosis-related genes covering a wide range of biological processes associated with ferroptosis. We performed the first ferroptosis-related targeted RNAseq on PBMCs from highly distinctive MS phenotype groups: mild relapsing-remitting (RR) (n = 24) and severe secondary progressive (SP) (n = 24), along with protein detection of GPX4 and products of lipid peroxidation (MDA and 4-HNE). Out of 138 genes, 26 were differentially expressed genes (DEGs), indicating changes in both pro- and anti-ferroptotic genes, representing a molecular signature associated with MS severity. The top three DEGs, as non-core ferroptosis genes, CDKN1A, MAP1B and EGLN2, were replicated by qPCR to validate findings in independent patient groups (16 RR and 16 SP MS). Co-expression and interactions of DEGs were presented as additional valuable assets for deeper understanding of molecular mechanisms and key targets related to MS severity. Our study integrates a wide genetic signature and biochemical markers related to ferroptosis in easily obtainable PBMCs of MS patients with clinical data and disease severity, thus providing novel molecular markers which can complement disease-related changes in the brain and undergo further research as potential therapeutic targets.
Collapse
Affiliation(s)
- Ljiljana Stojkovic
- Laboratory for Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, P.O. Box 522, 11000 Belgrade, Serbia
| | - Ivan Jovanovic
- Laboratory for Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, P.O. Box 522, 11000 Belgrade, Serbia
| | - Evica Dincic
- Clinic for Neurology, Military Medical Academy, 11000 Belgrade, Serbia
- Medical Faculty, University of Defense in Belgrade, 11042 Belgrade, Serbia
| | - Ana Djordjevic
- Laboratory for Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, P.O. Box 522, 11000 Belgrade, Serbia
| | - Jovana Kuveljic
- Laboratory for Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, P.O. Box 522, 11000 Belgrade, Serbia
| | - Tamara Djuric
- Laboratory for Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, P.O. Box 522, 11000 Belgrade, Serbia
| | - Aleksandra Stankovic
- Laboratory for Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, P.O. Box 522, 11000 Belgrade, Serbia
| | - Slobodan Vojinovic
- Department of Neurology, Medical Faculty, University of Nis, 18000 Nis, Serbia
| | - Maja Zivkovic
- Laboratory for Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, P.O. Box 522, 11000 Belgrade, Serbia
| |
Collapse
|
6
|
Figarella K, Kim J, Ruan W, Mills T, Eltzschig HK, Yuan X. Hypoxia-adenosine axis as therapeutic targets for acute respiratory distress syndrome. Front Immunol 2024; 15:1328565. [PMID: 38312838 PMCID: PMC10835146 DOI: 10.3389/fimmu.2024.1328565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 01/03/2024] [Indexed: 02/06/2024] Open
Abstract
The human respiratory and circulatory systems collaborate intricately to ensure oxygen delivery to all cells, which is vital for ATP production and maintaining physiological functions and structures. During limited oxygen availability, hypoxia-inducible factors (HIFs) are stabilized and play a fundamental role in maintaining cellular processes for hypoxia adaptation. First discovered during investigations of erythropoietin production regulation, HIFs influence physiological and pathological processes, including development, inflammation, wound healing, and cancer. HIFs promote extracellular adenosine signaling by enhancing adenosine generation and receptor signaling, representing an endogenous feedback mechanism that curbs excessive inflammation, supports injury resolution, and enhances hypoxia tolerance. This is especially important for conditions that involve tissue hypoxia, such as acute respiratory distress syndrome (ARDS), which globally poses significant health challenges without specific treatment options. Consequently, pharmacological strategies to amplify HIF-mediated adenosine production and receptor signaling are of great importance.
Collapse
Affiliation(s)
- Katherine Figarella
- Department of Anesthesiology, Critical Care and Pain Medicine, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Jieun Kim
- Department of Anesthesiology, Critical Care and Pain Medicine, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Wei Ruan
- Department of Anesthesiology, Critical Care and Pain Medicine, University of Texas Health Science Center at Houston, Houston, TX, United States
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Tingting Mills
- Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Holger Klaus Eltzschig
- Department of Anesthesiology, Critical Care and Pain Medicine, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Xiaoyi Yuan
- Department of Anesthesiology, Critical Care and Pain Medicine, University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
7
|
Ma F, Zou Y, Chen X, Ma L, Ma R. Evolution, characterization, and expression profile of Egl-9 family hypoxia-inducible factor ( egln) in rainbow trout ( Oncorhynchus mykiss) under hypoxia stress. Anim Biotechnol 2023; 34:1753-1762. [PMID: 35289728 DOI: 10.1080/10495398.2022.2047994] [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: 11/01/2022]
Abstract
Egl-9 family hypoxia-inducible factor (egln), an oxygen-sensing enzyme family, has been thoroughly characterized in mammals and certain fishes, but there is few research on its involvement in reproductive development and hypoxic stress in rainbow trout. In this study, we investigated the gene structure, physicochemical properties, and evolutionary connection of the egln gene family. The expression profile of egln gene family and their regulatory mechanism were explored using bioinformatics analysis and hypoxia treatment experiments. Five egln genes were discovered in the rainbow trout genome in this investigation (egln1, egln2a, egln2b, egln3a, and egln3b). Domain prediction revealed that all egln proteins have p4hc conserved domains, and phylogenetic analysis revealed that rainbow trout egln2 and egln3 were closely related to Atlantic salmon. The results of real-time quantitative PCR (RT-qPCR) showed that egln genes were generally expressed in all detected tissues, and higher in the ovary, testis, and brain in normoxia. Under hypoxia, the expression level of eglns was significantly down-regulated in most tissues except the liver. Our research contributes to future research on the functional properties of egln genes, as well as the evolution of teleosts and the impact of hypoxia on biological immunity.
Collapse
Affiliation(s)
- Fang Ma
- Key Laboratory of Resource Utilization of Agricultural Solid Waste in Gansu Province, Tianshui Normal University, Tianshui, Gansu, China
| | - Yali Zou
- Key Laboratory of Resource Utilization of Agricultural Solid Waste in Gansu Province, Tianshui Normal University, Tianshui, Gansu, China
| | - Xin Chen
- Key Laboratory of Resource Utilization of Agricultural Solid Waste in Gansu Province, Tianshui Normal University, Tianshui, Gansu, China
| | - Lanfang Ma
- Key Laboratory of Resource Utilization of Agricultural Solid Waste in Gansu Province, Tianshui Normal University, Tianshui, Gansu, China
| | - Ruilin Ma
- Key Laboratory of Resource Utilization of Agricultural Solid Waste in Gansu Province, Tianshui Normal University, Tianshui, Gansu, China
| |
Collapse
|
8
|
Cross-Regulation of the Cellular Redox System, Oxygen, and Sphingolipid Signalling. Metabolites 2023; 13:metabo13030426. [PMID: 36984866 PMCID: PMC10054022 DOI: 10.3390/metabo13030426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/09/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023] Open
Abstract
Redox-active mediators are now appreciated as powerful molecules to regulate cellular dynamics such as viability, proliferation, migration, cell contraction, and relaxation, as well as gene expression under physiological and pathophysiological conditions. These molecules include the various reactive oxygen species (ROS), and the gasotransmitters nitric oxide (NO∙), carbon monoxide (CO), and hydrogen sulfide (H2S). For each of these molecules, direct targets have been identified which transmit the signal from the cellular redox state to a cellular response. Besides these redox mediators, various sphingolipid species have turned out as highly bioactive with strong signalling potential. Recent data suggest that there is a cross-regulation existing between the redox mediators and sphingolipid molecules that have a fundamental impact on a cell’s fate and organ function. This review will summarize the effects of the different redox-active mediators on sphingolipid signalling and metabolism, and the impact of this cross-talk on pathophysiological processes. The relevance of therapeutic approaches will be highlighted.
Collapse
|
9
|
Chan YY, Mbenza NM, Chan MC, Leung IKH. Assays to Study Hypoxia-Inducible Factor Prolyl Hydroxylase Domain 2 (PHD2), a Key Human Oxygen Sensing Protein. Methods Mol Biol 2023; 2648:187-206. [PMID: 37039992 DOI: 10.1007/978-1-0716-3080-8_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2023]
Abstract
Molecular oxygen is essential for all multicellular life forms. In humans, the hypoxia-inducible factor (HIF) prolyl hydroxylase domain-containing enzymes (PHDs) serve as important oxygen sensors by regulating the activity of HIF, the master regulator that mediates cellular oxygen homeostasis, in an oxygen-dependent manner. In normoxia, PHDs catalyze the prolyl hydroxylation of HIF, which leads to its degradation and prevents cellular hypoxic response to be triggered. PHDs are current inhibition targets for the potential treatments of a number of diseases. In this chapter, we discuss in vitro and cell-based methods to study the modulation of PHD2, the most important human PHD isoform in normoxia and mild hypoxia. These include the production and purification of recombinant PHD2, the use of mass spectrometry to follow PHD2-catalyzed reactions and the studies of HIF stabilization in cells by immunoblotting.
Collapse
Affiliation(s)
- Yan Ying Chan
- Department of Molecular Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Naasson M Mbenza
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Mun Chiang Chan
- Department of Molecular Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
- GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Ivanhoe K H Leung
- School of Chemistry and the Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
10
|
Molecular Classification of Genes Associated with Hypoxic Lipid Metabolism in Pancreatic Cancer. Biomolecules 2022; 12:biom12101533. [PMID: 36291742 PMCID: PMC9599075 DOI: 10.3390/biom12101533] [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: 09/13/2022] [Revised: 10/17/2022] [Accepted: 10/19/2022] [Indexed: 11/29/2022] Open
Abstract
Abnormal lipid metabolism often occurs under hypoxic microenvironment, which is an important energy supplement for cancer cell proliferation and metastasis. We aimed to explore the lipid metabolism characteristics and gene expression features of pancreatic ductal adenocarcinoma (PDAC) related to hypoxia and identify biomarkers for molecular classification based on hypoxic lipid metabolism that are evaluable for PDAC prognosis and therapy. The multiple datasets were analyzed integratively, including corresponding clinical information of samples. PDAC possesses a distinct metabolic profile and oxygen level compared with normal pancreatic tissues, according to the bioinformatics methods. In addition, a study on untargeted metabolomics using Ultra Performance Liquid Chromatography Tandem Mass Spectrometry(UPLC-MS) revealed lipid metabolites differences affected by oxygen. Analysis of PDAC gene expression profiling in The Cancer Genome Atlas (TCGA) revealed that the sphingolipid process correlates closely with HIF1α. According to the characters of HIF-1 and sphingolipid, samples can be clustered into three subgroups using non-negative matrix factorization clustering. In cluster2, patients had an increased survival time. Relatively high MUC16 mutation arises in cluster2 and may positively influence the cancer survival rates. This study explored the expression pattern of lipid metabolism under hypoxia microenvironment in PDAC. On the basis of metabolic signatures, we identified the prognosis subtypes linking lipid metabolism to hypoxia. The classifications may be conducive to developing personalized treatment programs targeting metabolic profiles.
Collapse
|
11
|
Ruan W, Ma X, Bang IH, Liang Y, Muehlschlegel JD, Tsai KL, Mills TW, Yuan X, Eltzschig HK. The Hypoxia-Adenosine Link during Myocardial Ischemia-Reperfusion Injury. Biomedicines 2022; 10:1939. [PMID: 36009485 PMCID: PMC9405579 DOI: 10.3390/biomedicines10081939] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/28/2022] [Accepted: 08/01/2022] [Indexed: 11/16/2022] Open
Abstract
Despite increasing availability and more successful interventional approaches to restore coronary reperfusion, myocardial ischemia-reperfusion injury is a substantial cause of morbidity and mortality worldwide. During myocardial ischemia, the myocardium becomes profoundly hypoxic, thus causing stabilization of hypoxia-inducible transcription factors (HIF). Stabilization of HIF leads to a transcriptional program that promotes adaptation to hypoxia and cellular survival. Transcriptional consequences of HIF stabilization include increases in extracellular production and signaling effects of adenosine. Extracellular adenosine functions as a signaling molecule via the activation of adenosine receptors. Several studies implicated adenosine signaling in cardioprotection, particularly through the activation of the Adora2a and Adora2b receptors. Adenosine receptor activation can lead to metabolic adaptation to enhance ischemia tolerance or dampen myocardial reperfusion injury via signaling events on immune cells. Many studies highlight that clinical strategies to target the hypoxia-adenosine link could be considered for clinical trials. This could be achieved by using pharmacologic HIF activators or by directly enhancing extracellular adenosine production or signaling as a therapy for patients with acute myocardial infarction, or undergoing cardiac surgery.
Collapse
Affiliation(s)
- Wei Ruan
- Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Department of Anesthesiology, Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Xinxin Ma
- Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - In Hyuk Bang
- Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Yafen Liang
- Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Jochen Daniel Muehlschlegel
- Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Kuang-Lei Tsai
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Tingting W. Mills
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Xiaoyi Yuan
- Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Holger K. Eltzschig
- Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
12
|
Votava JA, Reese SR, Deck KM, Nizzi CP, Anderson SA, Djamali A, Eisenstein RS. Dysregulation of the sensory and regulatory pathways controlling cellular iron metabolism in unilateral obstructive nephropathy. Am J Physiol Renal Physiol 2022; 322:F89-F103. [PMID: 34843656 PMCID: PMC8742730 DOI: 10.1152/ajprenal.00537.2020] [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: 10/09/2020] [Revised: 11/16/2021] [Accepted: 11/23/2021] [Indexed: 01/03/2023] Open
Abstract
Chronic kidney disease involves disturbances in iron metabolism including anemia caused by insufficient erythropoietin (EPO) production. However, underlying mechanisms responsible for the dysregulation of cellular iron metabolism are incompletely defined. Using the unilateral ureteral obstruction (UUO) model in Irp1+/+ and Irp1-/- mice, we asked if iron regulatory proteins (IRPs), the central regulators of cellular iron metabolism and suppressors of EPO production, contribute to the etiology of anemia in kidney failure. We identified a significant reduction in IRP protein level and RNA binding activity that associates with a loss of the iron uptake protein transferrin receptor 1 (TfR1), increased expression of the iron storage protein subunits H- and L-ferritin, and a low but overall variable level of stainable iron in the obstructed kidney. This reduction in IRP RNA binding activity and ferritin RNA levels suggests the concomitant rise in ferritin expression and iron content in kidney failure is IRP dependent. In contrast, the reduction in the Epo mRNA level in the obstructed kidney was not rescued by genetic ablation of IRP1, suggesting disruption of normal hypoxia-inducible factor (HIF)-2α regulation. Furthermore, reduced expression of some HIF-α target genes in UUO occurred in the face of increased expression of HIF-α proteins and prolyl hydroxylases 2 and 1, the latter of which is not known to be HIF-α mediated. Our results suggest that the IRP system drives changes in cellular iron metabolism that are associated with kidney failure in UUO but that the impact of IRPs on EPO production is overridden by disrupted hypoxia signaling.NEW & NOTEWORTHY This study demonstrates that iron metabolism and hypoxia signaling are dysregulated in unilateral obstructive nephropathy. Expression of iron regulatory proteins (IRPs), central regulators of cellular iron metabolism, and the iron uptake (transferrin receptor 1) and storage (ferritins) proteins they target is strongly altered. This suggests a role of IRPs in previously observed changes in iron metabolism in progressive renal disease. Hypoxia signaling is disrupted and appeared to dominate the action of IRP1 in controlling erythropoietin expression.
Collapse
Affiliation(s)
- James A Votava
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, Wisconsin
| | - Shannon R Reese
- Division of Nephrology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Kathryn M Deck
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, Wisconsin
| | - Christopher P Nizzi
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, Wisconsin
| | - Sheila A Anderson
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, Wisconsin
| | - Arjang Djamali
- Division of Nephrology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin
- Division of Transplant, Department of Surgery, University of Wisconsin-Madison, Madison, Wisconsin
| | - Richard S Eisenstein
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, Wisconsin
- Morgridge Institute for Research, University of Wisconsin-Madison, Madison, Wisconsin
| |
Collapse
|
13
|
Chen HY, Hsu M, Lio CWJ. Micro but mighty-Micronutrients in the epigenetic regulation of adaptive immune responses. Immunol Rev 2022; 305:152-164. [PMID: 34820863 PMCID: PMC8766944 DOI: 10.1111/imr.13045] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 11/11/2021] [Accepted: 11/14/2021] [Indexed: 01/03/2023]
Abstract
Micronutrients are essential small molecules required by organisms in minute quantity for survival. For instance, vitamins and minerals, the two major categories of micronutrients, are central for biological processes such as metabolism, cell replication, differentiation, and immune response. Studies estimated that around two billion humans worldwide suffer from micronutrient deficiencies, also known as "hidden hunger," linked to weakened immune responses. While micronutrients affect the immune system at multiple levels, recent studies showed that micronutrients potentially impact the differentiation and function of immune cells as cofactors for epigenetic enzymes, including the 2-oxoglutarate-dependent dioxygenase (2OGDD) family involved in histone and DNA demethylation. Here, we will first provide an overview of the role of DNA methylation in T cells and B cells, followed by the micronutrients ascorbate (vitamin C) and iron, two critical cofactors for 2OGDD. We will discuss the emerging evidence of these micronutrients could regulate adaptive immune response by influencing epigenetic remodeling.
Collapse
Affiliation(s)
| | | | - Chan-Wang Jerry Lio
- Corresponding author: Chan-Wang Jerry Lio (), Address: 460 W 12 Ave, Columbus, Ohio, USA 43064, Tel: (614)-247-5337
| |
Collapse
|
14
|
Li S, Fu J, Wang Y, Hu C, Xu F. LncRNA MIAT enhances cerebral ischaemia/reperfusion injury in rat model via interacting with EGLN2 and reduces its ubiquitin-mediated degradation. J Cell Mol Med 2021; 25:10140-10151. [PMID: 34687132 PMCID: PMC8572800 DOI: 10.1111/jcmm.16950] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 08/26/2021] [Accepted: 09/19/2021] [Indexed: 11/30/2022] Open
Abstract
Long non-coding RNA (lncRNA) MIAT (myocardial infarction associated transcript) has been characterized as a functional lncRNA modulating cerebral ischaemic/reperfusion (I/R) injury. However, the underlying mechanisms remain poorly understood. This study explored the functional partners of MIAT in primary rat neurons and their regulation on I/R injury. Sprague-Dawley rats were used to construct middle cerebral artery occlusion (MCAO) models. Their cerebral cortical neurons were used for in vitro oxygen-glucose deprivation/reoxygenation (OGD/R) models. Results showed that MIAT interacted with EGLN2 in rat cortical neurons. MIAT overexpression or knockdown did not alter EGLN2 transcription. In contrast, MIAT overexpression increased EGLN2 stability after I/R injury via reducing its ubiquitin-mediated degradation. EGLN2 was a substrate of MDM2, a ubiquitin E3 ligase. MDM2 interacted with the N-terminal of EGLN2 and mediated its K48-linked poly-ubiquitination, thereby facilitating its proteasomal degradation. MIAT knockdown enhanced the interaction and reduced EGLN2 stability. MIAT overexpression enhanced infarct volume and induced a higher ratio of neuronal apoptosis. EGLN2 knockdown significantly reversed the injury. MIAT overexpression reduced oxidative pentose phosphate pathway flux and increased oxidized/reduced glutathione ratio, the effects of which were abrogated by EGLN2 knockdown. In conclusion, MIAT might act as a stabilizer of EGLN2 via reducing MDM2 mediated K48 poly-ubiquitination. MIAT-EGLN2 axis exacerbates I/R injury via altering redox homeostasis in neurons.
Collapse
Affiliation(s)
- Suping Li
- Department of Neurology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China
| | - Jing Fu
- Department of Rehabilitation, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China
| | - Yi Wang
- Department of Specialty of Geriatric Endocrinology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China
| | - Chunmei Hu
- Department of Otolaryngology-Head and Neck Surgery, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China
| | - Fei Xu
- Department of Neurology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China
| |
Collapse
|
15
|
Olson KR. A Case for Hydrogen Sulfide Metabolism as an Oxygen Sensing Mechanism. Antioxidants (Basel) 2021; 10:antiox10111650. [PMID: 34829521 PMCID: PMC8615108 DOI: 10.3390/antiox10111650] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/08/2021] [Accepted: 10/13/2021] [Indexed: 12/30/2022] Open
Abstract
The ability to detect oxygen availability is a ubiquitous attribute of aerobic organisms. However, the mechanism(s) that transduce oxygen concentration or availability into appropriate physiological responses is less clear and often controversial. This review will make the case for oxygen-dependent metabolism of hydrogen sulfide (H2S) and polysulfides, collectively referred to as reactive sulfur species (RSS) as a physiologically relevant O2 sensing mechanism. This hypothesis is based on observations that H2S and RSS metabolism is inversely correlated with O2 tension, exogenous H2S elicits physiological responses identical to those produced by hypoxia, factors that affect H2S production or catabolism also affect tissue responses to hypoxia, and that RSS efficiently regulate downstream effectors of the hypoxic response in a manner consistent with a decrease in O2. H2S-mediated O2 sensing is then compared to the more generally accepted reactive oxygen species (ROS) mediated O2 sensing mechanism and a number of reasons are offered to resolve some of the confusion between the two.
Collapse
Affiliation(s)
- Kenneth R Olson
- Department of Physiology, Indiana University School of Medicine-South Bend, South Bend, IN 46617, USA
| |
Collapse
|
16
|
Gonzaga de França Lopes L, Gouveia Júnior FS, Karine Medeiros Holanda A, Maria Moreira de Carvalho I, Longhinotti E, Paulo TF, Abreu DS, Bernhardt PV, Gilles-Gonzalez MA, Cirino Nogueira Diógenes I, Henrique Silva Sousa E. Bioinorganic systems responsive to the diatomic gases O2, NO, and CO: From biological sensors to therapy. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.214096] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
17
|
Xu X, Shen X, Wang J, Feng W, Wang M, Miao X, Wu Q, Wu L, Wang X, Ma Y, Wu S, Bao X, Wang W, Wang Y, Huang Z. YAP prevents premature senescence of astrocytes and cognitive decline of Alzheimer's disease through regulating CDK6 signaling. Aging Cell 2021; 20:e13465. [PMID: 34415667 PMCID: PMC8441453 DOI: 10.1111/acel.13465] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 07/15/2021] [Accepted: 08/07/2021] [Indexed: 12/12/2022] Open
Abstract
Senescent astrocytes accumulate with aging and contribute to brain dysfunction and diseases such as Alzheimer's disease (AD), however, the mechanisms underlying the senescence of astrocytes during aging remain unclear. In the present study, we found that Yes‐associated Protein (YAP) was downregulated and inactivated in hippocampal astrocytes of aging mice and AD model mice, as well as in D‐galactose and paraquat‐induced senescent astrocytes, in a Hippo pathway‐dependent manner. Conditional knockout of YAP in astrocytes significantly promoted premature senescence of astrocytes, including reduction of cell proliferation, hypertrophic morphology, increase in senescence‐associated β‐galactosidase activity, and upregulation of several senescence‐associated genes such as p16, p53 and NF‐κB, and downregulation of Lamin B1. Further exploration of the underlying mechanism revealed that the expression of cyclin‐dependent kinase 6 (CDK6) was decreased in YAP knockout astrocytes in vivo and in vitro, and ectopic overexpression of CDK6 partially rescued YAP knockout‐induced senescence of astrocytes. Finally, activation of YAP signaling by XMU‐MP‐1 (an inhibitor of Hippo kinase MST1/2) partially rescued the senescence of astrocytes and improved the cognitive function of AD model mice and aging mice. Taken together, our studies identified unrecognized functions of YAP‐CDK6 pathway in preventing astrocytic senescence in vitro and in vivo, which may provide further insights and new targets for delaying brain aging and aging‐related neurodegenerative diseases such as AD.
Collapse
Affiliation(s)
- Xingxing Xu
- School of Basic Medical Sciences Wenzhou Medical University Wenzhou China
| | - Xiya Shen
- School of Basic Medical Sciences Wenzhou Medical University Wenzhou China
| | - Jiaojiao Wang
- School of Basic Medical Sciences Wenzhou Medical University Wenzhou China
| | - Wenjin Feng
- Zhejiang Sinogen Medical Equipment Co., Ltd Wenzhou China
| | - Mianxian Wang
- School of Basic Medical Sciences Wenzhou Medical University Wenzhou China
| | - Xuemeng Miao
- School of Mental Health Wenzhou Medical University Wenzhou China
| | - Qian Wu
- School of Mental Health Wenzhou Medical University Wenzhou China
| | - Lihao Wu
- School of the First Clinical Medical Sciences School of Information and Engineering Wenzhou Medical University Wenzhou China
| | - Xiaoning Wang
- School of the First Clinical Medical Sciences School of Information and Engineering Wenzhou Medical University Wenzhou China
| | - Yimin Ma
- School of Mental Health Wenzhou Medical University Wenzhou China
| | - Shuang Wu
- School of the First Clinical Medical Sciences School of Information and Engineering Wenzhou Medical University Wenzhou China
| | - Xiaomei Bao
- School of Basic Medical Sciences Wenzhou Medical University Wenzhou China
- Department of Obstetrics and Gynecology Wenzhou People's Hospital Wenzhou China
| | - Wei Wang
- School of Mental Health Wenzhou Medical University Wenzhou China
| | - Ying Wang
- Phase I Clinical Research Center Zhejiang Provincial People's Hospital of Hangzhou Medical College Hangzhou China
| | - Zhihui Huang
- School of Basic Medical Sciences Wenzhou Medical University Wenzhou China
- School of Mental Health Wenzhou Medical University Wenzhou China
- College of Pharmacy Hangzhou Normal University Hangzhou China
| |
Collapse
|
18
|
Xu L, Song H, Qiu Q, Jiang T, Ge P, Su Z, Ma W, Zhang R, Huang C, Li S, Lin D, Zhang J. Different Expressions of HIF-1α and Metabolism in Brain and Major Visceral Organs of Acute Hypoxic Mice. Int J Mol Sci 2021; 22:6705. [PMID: 34201416 PMCID: PMC8268807 DOI: 10.3390/ijms22136705] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/27/2021] [Accepted: 05/25/2021] [Indexed: 12/18/2022] Open
Abstract
Hypoxia is associated with clinical diseases. Extreme hypoxia leads to multiple organs failure. However, the different effects of hypoxia on brain and visceral organs still need to be clarified, and moreover, characteristics in vulnerable organs suffering from hypoxia remain elusive. In the present study, we first aimed to figure out the hypoxic sensitivity of organs. Adult male mice were exposed to 6% O2 or 8% O2 for 6 h. Control mice were raised under normoxic conditions. In vivo and in vitro imaging of anti-HIF-1α-NMs-cy5.5 nanocomposites showed that the expression level of hypoxia-inducible factor (HIF-1α) was the highest in the liver, followed by kidney and brain. HIF-1α was detected in the hepatocytes of liver, distal convoluted tubules of kidney and neurons of cerebral cortex. The liver, kidney and brain showed distinct metabolic profiles but an identical change in glutamate. Compared with kidney and brain, the liver had more characteristic metabolites and more disturbed metabolic pathways related to glutaminolysis and glycolysis. The level of O-phosphocholine, GTP, NAD and aspartate were upregulated in hypoxic mice brain, which displayed significant positive correlations with the locomotor activity in control mice, but not in hypoxic mice with impaired locomotor activities. Taken together, the liver, kidney and brain are the three main organs of the body that are strongly respond to acute hypoxia, and the liver exhibited the highest hypoxic sensitivity. The metabolic disorders appear to underlie the physiological function changes.
Collapse
Affiliation(s)
- Lu Xu
- Institute of Brain Diseases and Cognition, Medical College of Xiamen University, Xiamen 361102, China; (L.X.); (Q.Q.); (W.M.); (R.Z.)
| | - Hua Song
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China; (H.S.); (P.G.); (Z.S.)
| | - Qi Qiu
- Institute of Brain Diseases and Cognition, Medical College of Xiamen University, Xiamen 361102, China; (L.X.); (Q.Q.); (W.M.); (R.Z.)
| | - Ting Jiang
- Key Laboratory for Chemical Biology of Fujian Province, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China;
| | - Pingyun Ge
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China; (H.S.); (P.G.); (Z.S.)
| | - Zaiji Su
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China; (H.S.); (P.G.); (Z.S.)
| | - Wenhui Ma
- Institute of Brain Diseases and Cognition, Medical College of Xiamen University, Xiamen 361102, China; (L.X.); (Q.Q.); (W.M.); (R.Z.)
| | - Ran Zhang
- Institute of Brain Diseases and Cognition, Medical College of Xiamen University, Xiamen 361102, China; (L.X.); (Q.Q.); (W.M.); (R.Z.)
| | - Caihua Huang
- Research and Communication Center of Exercise and Health, Xiamen University of Technology, Xiamen 361024, China;
| | - Shanhua Li
- Institute of Brain Diseases and Cognition, Medical College of Xiamen University, Xiamen 361102, China; (L.X.); (Q.Q.); (W.M.); (R.Z.)
| | - Donghai Lin
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China; (H.S.); (P.G.); (Z.S.)
| | - Jiaxing Zhang
- Institute of Brain Diseases and Cognition, Medical College of Xiamen University, Xiamen 361102, China; (L.X.); (Q.Q.); (W.M.); (R.Z.)
| |
Collapse
|
19
|
Dengler F, Sova S, Salo AM, Mäki JM, Koivunen P, Myllyharju J. Expression and Roles of Individual HIF Prolyl 4-Hydroxylase Isoenzymes in the Regulation of the Hypoxia Response Pathway along the Murine Gastrointestinal Epithelium. Int J Mol Sci 2021; 22:4038. [PMID: 33919829 PMCID: PMC8070794 DOI: 10.3390/ijms22084038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/07/2021] [Accepted: 04/11/2021] [Indexed: 12/11/2022] Open
Abstract
The HIF prolyl 4-hydroxylases (HIF-P4H) control hypoxia-inducible factor (HIF), a powerful mechanism regulating cellular adaptation to decreased oxygenation. The gastrointestinal epithelium subsists in "physiological hypoxia" and should therefore have an especially well-designed control over this adaptation. Thus, we assessed the absolute mRNA expression levels of the HIF pathway components, Hif1a, HIF2a, Hif-p4h-1, 2 and 3 and factor inhibiting HIF (Fih1) in murine jejunum, caecum and colon epithelium using droplet digital PCR. We found a higher expression of all these genes towards the distal end of the gastrointestinal tract. We detected mRNA for Hif-p4h-1, 2 and 3 in all parts of the gastrointestinal tract. Hif-p4h-2 had significantly higher expression levels compared to Hif-p4h-1 and 3 in colon and caecum epithelium. To test the roles each HIF-P4H isoform plays in the gut epithelium, we measured the gene expression of classical HIF target genes in Hif-p4h-1-/-, Hif-p4h-2 hypomorph and Hif-p4h-3-/- mice. Only Hif-p4h-2 hypomorphism led to an upregulation of HIF target genes, confirming a predominant role of HIF-P4H-2. However, the abundance of Hif-p4h-1 and 3 expression in the gastrointestinal epithelium implies that these isoforms may have specific functions as well. Thus, the development of selective inhibitors might be useful for diverging therapeutic needs.
Collapse
Affiliation(s)
- Franziska Dengler
- Unit of Physiology, Pathophysiology and Experimental Endocrinology, University of Veterinary Medicine, 1210 Vienna, Austria
- Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu, 90220 Oulu, Finland; (S.S.); (A.M.S.); (J.M.M.); (P.K.); (J.M.)
- Institute of Veterinary Physiology, University of Leipzig, 04103 Leipzig, Germany
| | - Sofia Sova
- Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu, 90220 Oulu, Finland; (S.S.); (A.M.S.); (J.M.M.); (P.K.); (J.M.)
| | - Antti M. Salo
- Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu, 90220 Oulu, Finland; (S.S.); (A.M.S.); (J.M.M.); (P.K.); (J.M.)
| | - Joni M. Mäki
- Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu, 90220 Oulu, Finland; (S.S.); (A.M.S.); (J.M.M.); (P.K.); (J.M.)
| | - Peppi Koivunen
- Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu, 90220 Oulu, Finland; (S.S.); (A.M.S.); (J.M.M.); (P.K.); (J.M.)
| | - Johanna Myllyharju
- Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu, 90220 Oulu, Finland; (S.S.); (A.M.S.); (J.M.M.); (P.K.); (J.M.)
| |
Collapse
|
20
|
Losso JN. Food Processing, Dysbiosis, Gastrointestinal Inflammatory Diseases, and Antiangiogenic Functional Foods or Beverages. Annu Rev Food Sci Technol 2021; 12:235-258. [PMID: 33467906 DOI: 10.1146/annurev-food-062520-090235] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Foods and beverages provide nutrients and alter the gut microbiota, resulting in eubiosis or dysbiosis. Chronic consumption of a diet that is high in saturated or trans fats, meat proteins, reducing sugars, and salt and low in fiber induces dysbiosis. Dysbiosis, loss of redox homeostasis, mast cells, hypoxia, angiogenesis, the kynurenine pathway, transglutaminase 2, and/or the Janus kinase pathway are implicated in the pathogenesis and development of inflammatory bowel disease, celiac disease, and gastrointestinal malignancy. This review discusses the effects of oxidative, carbonyl, or glycative stress-inducing dietary ingredients or food processing-derived compounds on gut microbiota and gastrointestinal epithelial and mast cells as well as on the development of associated angiogenic diseases, including key signaling pathways. The preventive or therapeutic potential and the biochemical pathways of antiangiogenic or proangiogenic foods or beverages are also described. The outcomes of the interactions between disease pathways and components of food are critical for the design of foods and beverages for healthy lives.
Collapse
Affiliation(s)
- Jack N Losso
- School of Nutrition and Food Sciences, Louisiana State University, Baton Rouge, Louisiana 70803, USA;
| |
Collapse
|
21
|
Olson KR, Gao Y, DeLeon ER, Markel TA, Drucker N, Boone D, Whiteman M, Steiger AK, Pluth MD, Tessier CR, Stahelin RV. Extended hypoxia-mediated H 2 S production provides for long-term oxygen sensing. Acta Physiol (Oxf) 2020; 228:e13368. [PMID: 31442361 DOI: 10.1111/apha.13368] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 08/16/2019] [Accepted: 08/18/2019] [Indexed: 12/23/2022]
Abstract
AIM Numerous studies have shown that H2 S serves as an acute oxygen sensor in a variety of cells. We hypothesize that H2 S also serves in extended oxygen sensing. METHODS Here, we compare the effects of extended exposure (24-48 hours) to varying O2 tensions on H2 S and polysulphide metabolism in human embryonic kidney (HEK 293), human adenocarcinomic alveolar basal epithelial (A549), human colon cancer (HTC116), bovine pulmonary artery smooth muscle, human umbilical-derived mesenchymal stromal (stem) cells and porcine tracheal epithelium (PTE) using sulphur-specific fluorophores and fluorometry or confocal microscopy. RESULTS All cells continuously produced H2 S in 21% O2 and H2 S production was increased at lower O2 tensions. Decreasing O2 from 21% to 10%, 5% and 1% O2 progressively increased H2 S production in HEK293 cells and this was partially inhibited by a combination of inhibitors of H2 S biosynthesis, aminooxyacetate, propargyl glycine and compound 3. Mitochondria appeared to be the source of much of this increase in HEK 293 cells. H2 S production in all other cells and PTE increased when O2 was lowered from 21% to 5% except for HTC116 cells where 1% O2 was necessary to increase H2 S, presumably reflecting the hypoxic environment in vivo. Polysulphides (H2 Sn , where n = 2-7), the key signalling metabolite of H2 S also appeared to increase in many cells although this was often masked by high endogenous polysulphide concentrations. CONCLUSION These results show that cellular H2 S is increased during extended hypoxia and they suggest this is a continuously active O2 -sensing mechanism in a variety of cells.
Collapse
Affiliation(s)
- Kenneth R. Olson
- Indiana University School of Medicine‐South Bend South Bend Indiana
| | - Yan Gao
- Indiana University School of Medicine‐South Bend South Bend Indiana
| | - Eric R. DeLeon
- Indiana University School of Medicine‐South Bend South Bend Indiana
- Department of Biological Sciences University of Notre Dame Notre Dame Indiana
| | - Troy A. Markel
- Indiana University School of Medicine Riley Hospital for Children at IU Health Indianapolis Indiana
| | - Natalie Drucker
- Indiana University School of Medicine Riley Hospital for Children at IU Health Indianapolis Indiana
| | - David Boone
- Indiana University School of Medicine‐South Bend South Bend Indiana
| | | | - Andrea K. Steiger
- Department of Chemistry and Biochemistry University of Oregon Eugene Oregon
| | - Michael D. Pluth
- Department of Chemistry and Biochemistry University of Oregon Eugene Oregon
| | | | - Robert V. Stahelin
- Department of Medicinal Chemistry and Molecular Pharmacology Purdue University West Lafayette Indiana
| |
Collapse
|
22
|
Strowitzki MJ, Cummins EP, Taylor CT. Protein Hydroxylation by Hypoxia-Inducible Factor (HIF) Hydroxylases: Unique or Ubiquitous? Cells 2019; 8:cells8050384. [PMID: 31035491 PMCID: PMC6562979 DOI: 10.3390/cells8050384] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 04/24/2019] [Accepted: 04/24/2019] [Indexed: 02/07/2023] Open
Abstract
All metazoans that utilize molecular oxygen (O2) for metabolic purposes have the capacity to adapt to hypoxia, the condition that arises when O2 demand exceeds supply. This is mediated through activation of the hypoxia-inducible factor (HIF) pathway. At physiological oxygen levels (normoxia), HIF-prolyl hydroxylases (PHDs) hydroxylate proline residues on HIF-α subunits leading to their destabilization by promoting ubiquitination by the von-Hippel Lindau (VHL) ubiquitin ligase and subsequent proteasomal degradation. HIF-α transactivation is also repressed in an O2-dependent way due to asparaginyl hydroxylation by the factor-inhibiting HIF (FIH). In hypoxia, the O2-dependent hydroxylation of HIF-α subunits by PHDs and FIH is reduced, resulting in HIF-α accumulation, dimerization with HIF-β and migration into the nucleus to induce an adaptive transcriptional response. Although HIFs are the canonical substrates for PHD- and FIH-mediated protein hydroxylation, increasing evidence indicates that these hydroxylases may also have alternative targets. In addition to PHD-conferred alterations in protein stability, there is now evidence that hydroxylation can affect protein activity and protein/protein interactions for alternative substrates. PHDs can be pharmacologically inhibited by a new class of drugs termed prolyl hydroxylase inhibitors which have recently been approved for the treatment of anemia associated with chronic kidney disease. The identification of alternative targets of HIF hydroxylases is important in order to fully elucidate the pharmacology of hydroxylase inhibitors (PHI). Despite significant technical advances, screening, detection and verification of alternative functional targets for PHDs and FIH remain challenging. In this review, we discuss recently proposed non-HIF targets for PHDs and FIH and provide an overview of the techniques used to identify these.
Collapse
Affiliation(s)
- Moritz J Strowitzki
- UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland.
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Eoin P Cummins
- UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland.
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Cormac T Taylor
- UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland.
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland.
- Systems Biology Ireland, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
23
|
Li A, Zhang Y, Wang Z, Dong H, Fu N, Han X. The roles and signaling pathways of prolyl-4-hydroxylase 2 in the tumor microenvironment. Chem Biol Interact 2019; 303:40-49. [PMID: 30817904 DOI: 10.1016/j.cbi.2019.02.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 02/16/2019] [Accepted: 02/19/2019] [Indexed: 02/06/2023]
Abstract
Tumor hypoxia is a well-known microenvironmental factor that causes cancer progression and resistance to cancer treatment. Proline hydroxylases (PHDs), a small protein family, belong to an evolutionarily conserved superfamily of dioxygenases, considered the central regulator of the molecular hypoxia response. Prolyl-4-hydroxylase 2 (PHD2), one member of PHDs family, regulates the stability of the hypoxia-inducible factor-1 alpha (HIF-1α) in response to oxygen availability. During hypoxia, the inhibition of PHD2 permits the accumulation of HIF-1α, allowing the cellular adaptation to oxygen limitation, causing activation of numerous genes, which enhances the angiogenesis, metastasis and invasiveness. Accurate regulation of oxygen homeostasis is essential, and which implies PHD2 may have a regulatory role in the pathogenesis of cancer. Although ample evidence exists for a positive correlation between HIFs and tumor formation, metastasis and poor prognosis, the function of the PHD2 in carcinogenesis is less well understood. Despite their original role as the oxygen sensors of the cell and many of the its functions are clearly conveyed through the HIF system, PHD2 is currently known to display HIF-independent and hydroxylase-independent functions in cancer cells and stroma in the control of different cellular pathways. In this review, we summarize the recent advances in the structure, regulation and functions of PHD2 in cancer microenvironment.
Collapse
Affiliation(s)
- Anqi Li
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan, 250012, China
| | - Yu Zhang
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan, 250012, China
| | - Zuojun Wang
- Department of Pharmacy, Linqu Country People's Hospital, 438 Shanwang Road, Linqu, 262600, China
| | - Hailing Dong
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan, 250012, China
| | - Nange Fu
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan, 250012, China
| | - Xiuzhen Han
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan, 250012, China.
| |
Collapse
|
24
|
The molecular characterization, expression pattern and alternative initiation of Megalobrama amblycephala Hif prolyl hydroxylase Phd1. Gene 2018; 678:219-225. [DOI: 10.1016/j.gene.2018.08.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 07/27/2018] [Accepted: 08/03/2018] [Indexed: 12/11/2022]
|
25
|
Strowitzki MJ, Kirchberg J, Tuffs C, Schiedeck M, Ritter AS, Biller M, Harnoss JM, Lasitschka F, Schmidt T, Radhakrishnan P, Ulrich A, Schneider M. Loss of Prolyl-Hydroxylase 1 Protects against Biliary Fibrosis via Attenuated Activation of Hepatic Stellate Cells. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:2826-2838. [PMID: 30248340 DOI: 10.1016/j.ajpath.2018.08.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 08/10/2018] [Accepted: 08/14/2018] [Indexed: 12/12/2022]
Abstract
Liver fibrosis, eventually progressing to cirrhosis necessitating liver transplantation, poses a significant clinical problem. Oxygen shortage (hypoxia) and hypoxia-inducible transcription factors (HIFs) have been acknowledged as important drivers of liver fibrosis. The significance of oxygen-sensing HIF prolyl-hydroxylase (PHD) enzymes in this context has, however, remained elusive. In this study, we demonstrate that loss of PHD1 (PHD1-/-) attenuates the development of liver fibrosis in mice subjected to chronic bile duct injury, induced by 3,5-diethoxycarbonyl-1,4-dihydrocollidine. This effect was accompanied with reduced recruitment of inflammatory leukocytes and attenuated occurrence of profibrotic myofibroblasts in PHD1-/- livers. Further analyses focused on the significance of PHD1 in the activation of hepatic stellate cells (HSCs), which represent the driving force in liver fibrosis. Primary HSCs isolated from PHD1-/- mice displayed significantly attenuated myofibroblast differentiation and profibrogenic properties compared with HSCs isolated from wild-type mice. Consistently, the expression of various profibrogenic and promitogenic factors was reduced in PHD1-/- HSCs, without alterations in HIF-1α protein levels. Of importance, PHD1 protein was expressed in HSCs within human livers, and PHD1 transcript expression was significantly increased with disease severity in hepatic tissue from patients with liver fibrosis. Collectively, these findings indicate that PHD1 deficiency protects against liver fibrosis and that these effects are partly due to attenuated activation of HSCs. PHD1 may represent a therapeutic target to alleviate liver fibrosis.
Collapse
Affiliation(s)
- Moritz J Strowitzki
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Johanna Kirchberg
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Christopher Tuffs
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Maximilian Schiedeck
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Alina S Ritter
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Marvin Biller
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Jonathan M Harnoss
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Felix Lasitschka
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Thomas Schmidt
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Praveen Radhakrishnan
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Alexis Ulrich
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Martin Schneider
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
26
|
Barrett KE. New frontiers in gastrointestinal physiology and pathophysiology. J Physiol 2018; 596:3859-3860. [PMID: 29863278 PMCID: PMC6117559 DOI: 10.1113/jp276454] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Accepted: 05/21/2018] [Indexed: 12/14/2022] Open
Affiliation(s)
- Kim E. Barrett
- Department of MedicineUniversity of California San DiegoSchool of MedicineLa JollaCAUSA
| |
Collapse
|