51
|
Lozano J, Rai A, Lees JG, Fang H, Claridge B, Lim SY, Greening DW. Scalable Generation of Nanovesicles from Human-Induced Pluripotent Stem Cells for Cardiac Repair. Int J Mol Sci 2022; 23:14334. [PMID: 36430812 PMCID: PMC9696585 DOI: 10.3390/ijms232214334] [Citation(s) in RCA: 18] [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: 09/26/2022] [Revised: 11/03/2022] [Accepted: 11/15/2022] [Indexed: 11/22/2022] Open
Abstract
Extracellular vesicles (EVs) from stem cells have shown significant therapeutic potential to repair injured cardiac tissues and regulate pathological fibrosis. However, scalable generation of stem cells and derived EVs for clinical utility remains a huge technical challenge. Here, we report a rapid size-based extrusion strategy to generate EV-like membranous nanovesicles (NVs) from easily sourced human iPSCs in large quantities (yield 900× natural EVs). NVs isolated using density-gradient separation (buoyant density 1.13 g/mL) are spherical in shape and morphologically intact and readily internalised by human cardiomyocytes, primary cardiac fibroblasts, and endothelial cells. NVs captured the dynamic proteome of parental cells and include pluripotency markers (LIN28A, OCT4) and regulators of cardiac repair processes, including tissue repair (GJA1, HSP20/27/70, HMGB1), wound healing (FLNA, MYH9, ACTC1, ILK), stress response/translation initiation (eIF2S1/S2/S3/B4), hypoxia response (HMOX2, HSP90, GNB1), and extracellular matrix organization (ITGA6, MFGE8, ITGB1). Functionally, NVs significantly promoted tubule formation of endothelial cells (angiogenesis) (p < 0.05) and survival of cardiomyocytes exposed to low oxygen conditions (hypoxia) (p < 0.0001), as well as attenuated TGF-β mediated activation of cardiac fibroblasts (p < 0.0001). Quantitative proteome profiling of target cell proteome following NV treatments revealed upregulation of angiogenic proteins (MFGE8, MYH10, VDAC2) in endothelial cells and pro-survival proteins (CNN2, THBS1, IGF2R) in cardiomyocytes. In contrast, NVs attenuated TGF-β-driven extracellular matrix remodelling capacity in cardiac fibroblasts (ACTN1, COL1A1/2/4A2/12A1, ITGA1/11, THBS1). This study presents a scalable approach to generating functional NVs for cardiac repair.
Collapse
Affiliation(s)
- Jonathan Lozano
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
- Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, VIC 3086, Australia
- Department of Microbiology, Anatomy, Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC 3086, Australia
| | - Alin Rai
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
- Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, VIC 3086, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC 3010, Australia
- Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Jarmon G. Lees
- O’Brien Institute Department, St Vincent’s Institute of Medical Research, Melbourne, VIC 3065, Australia
- Department of Surgery and Medicine, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Haoyun Fang
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Bethany Claridge
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC 3086, Australia
| | - Shiang Y. Lim
- O’Brien Institute Department, St Vincent’s Institute of Medical Research, Melbourne, VIC 3065, Australia
- Department of Surgery and Medicine, University of Melbourne, Melbourne, VIC 3010, Australia
- National Heart Research Institute Singapore, National Heart Centre, Singapore 169609, Singapore
- Drug Discovery Biology, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Melbourne, VIC 3800, Australia
| | - David W. Greening
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
- Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, VIC 3086, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC 3010, Australia
- Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC 3086, Australia
| |
Collapse
|
52
|
Zhang Q, Bai X, Lin T, Wang X, Zhang B, Dai L, Shi J, Zhang Y, Zhao X. HMOX1 Promotes Ferroptosis in Mammary Epithelial Cells via FTH1 and Is Involved in the Development of Clinical Mastitis in Dairy Cows. Antioxidants (Basel) 2022; 11:2221. [PMID: 36421410 PMCID: PMC9686786 DOI: 10.3390/antiox11112221] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 11/04/2022] [Accepted: 11/07/2022] [Indexed: 08/26/2023] Open
Abstract
Ferroptosis is associated with inflammatory diseases as a lethal iron-dependent lipid peroxidation; its role in the development of clinical mastitis (CM) in dairy cows is not well understood. The aim of this study was to identify differentially expressed proteins (DEPs) associated with iron homeostasis and apoptosis, and to investigate further their roles in dairy cows with CM. The results suggested that ferroptosis occurs in the mammary glands of Holstein cows with CM. Using data-independent acquisition proteomics, 302 DEPs included in 11 GO terms related to iron homeostasis and apoptosis were identified. In particular, heme oxygenase-1 (HMOX1) was identified and involved in nine pathways. In addition, ferritin heavy chain 1 (FTH1) was identified and involved in the ferroptosis pathway. HMOX1 and FTH1 were located primarily in mammary epithelial cells (MECs), and displayed significantly up-regulated expression patterns compared to the control group (healthy cows). The expression levels of HMOX1 and FTH1 were up-regulated in a dose-dependent manner in LPS induced MAC-T cells with increased iron accumulation. The expression levels of HMOX1 and FTH1 and iron accumulation levels in the MAC-T cells were significantly up-regulated by using LPS, but were lower than the levels seen with Erastin (ERA). Finally, we deduced the mechanism of ferroptosis in the MECs of Holstein cows with CM. These results provide new insights for the prevention and treatment of ferroptosis-mediated clinical mastitis in dairy animals.
Collapse
Affiliation(s)
- Quanwei Zhang
- College of Life Science and Technology, Gansu Agriculture University, Lanzhou 730070, China
- College of Veterinary Medicine, Gansu Agriculture University, Lanzhou 730070, China
- Gansu Key Laboratory of Animal Reproductive Physiology and Reproductive Regulation, Lanzhou 730070, China
| | - Xu Bai
- College of Life Science and Technology, Gansu Agriculture University, Lanzhou 730070, China
- Gansu Key Laboratory of Animal Reproductive Physiology and Reproductive Regulation, Lanzhou 730070, China
| | - Ting Lin
- College of Life Science and Technology, Gansu Agriculture University, Lanzhou 730070, China
- Gansu Key Laboratory of Animal Reproductive Physiology and Reproductive Regulation, Lanzhou 730070, China
| | - Xueying Wang
- College of Veterinary Medicine, Gansu Agriculture University, Lanzhou 730070, China
- Gansu Key Laboratory of Animal Reproductive Physiology and Reproductive Regulation, Lanzhou 730070, China
| | - Bohao Zhang
- College of Veterinary Medicine, Gansu Agriculture University, Lanzhou 730070, China
- Gansu Key Laboratory of Animal Reproductive Physiology and Reproductive Regulation, Lanzhou 730070, China
| | - Lijun Dai
- College of Life Science and Technology, Gansu Agriculture University, Lanzhou 730070, China
- Gansu Key Laboratory of Animal Reproductive Physiology and Reproductive Regulation, Lanzhou 730070, China
| | - Jun Shi
- College of Life Science and Technology, Gansu Agriculture University, Lanzhou 730070, China
- Gansu Key Laboratory of Animal Reproductive Physiology and Reproductive Regulation, Lanzhou 730070, China
| | - Yong Zhang
- College of Life Science and Technology, Gansu Agriculture University, Lanzhou 730070, China
- College of Veterinary Medicine, Gansu Agriculture University, Lanzhou 730070, China
- Gansu Key Laboratory of Animal Reproductive Physiology and Reproductive Regulation, Lanzhou 730070, China
| | - Xingxu Zhao
- College of Life Science and Technology, Gansu Agriculture University, Lanzhou 730070, China
- College of Veterinary Medicine, Gansu Agriculture University, Lanzhou 730070, China
- Gansu Key Laboratory of Animal Reproductive Physiology and Reproductive Regulation, Lanzhou 730070, China
| |
Collapse
|
53
|
Mo Q, Li S, You S, Wang D, Zhang J, Li M, Wang C. Puerarin Reduces Oxidative Damage and Photoaging Caused by UVA Radiation in Human Fibroblasts by Regulating Nrf2 and MAPK Signaling Pathways. Nutrients 2022; 14:nu14224724. [PMID: 36432411 PMCID: PMC9694396 DOI: 10.3390/nu14224724] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/04/2022] [Accepted: 11/05/2022] [Indexed: 11/11/2022] Open
Abstract
Fibroblasts account for more than 95% of dermal cells maintaining dermal structure and function. However, UVA penetrates the dermis and causes oxidative stress that damages the dermis and accelerates skin aging. Puerarin, the main active ingredient of Puerariae lobata, has been demonstrated to withstand oxidative stress caused by a variety of factors. However, there are limited findings on whether puerarin protects fibroblasts from UVA-induced oxidative stress damage. The effects of puerarin on human skin fibroblasts (HSF) under UVA-induced oxidative stress were investigated in this study. It is found that puerarin upregulates antioxidant enzymes' mRNA expression level and their content through modulating the KEAP1-Nrf2/ARE signaling pathway, thus improving cell antioxidant capacity and successfully eliminating UVA-induced reactive oxygen species (ROS) and lipid oxidation product malondialdehyde (MDA). Additionally, puerarin blocks the overexpression of human extracellular signal-regulated kinase (ERK), human c-Jun amino-terminal kinase (JNK), and P38, which downregulates matrix metalloproteinase 1 (MMP-1) expression and increases type I collagen (COL-1) expression. Moreover, preliminary research on mouse skin suggests that puerarin can hydrate, moisturize, and increase the antioxidant capacity of skin tissue. These findings suggest that puerarin can protect the skin against photoaging.
Collapse
Affiliation(s)
- Qiuting Mo
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University, Fucheng Road, Beijing 100048, China
- Institute of Cosmetic Regulatory Science, College of Chemistry and Materials Engineering, Beijing Technology & Business University, Fucheng Road, Haidian District, Beijing 100048, China
- Beijing Key Laboratory of Plant Resource Research and Development, College of Chemistry and Materials Engineering, Beijing Technology and Business University, Fucheng Road, Haidian District, Beijing 100048, China
| | - Shuping Li
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University, Fucheng Road, Beijing 100048, China
- Institute of Cosmetic Regulatory Science, College of Chemistry and Materials Engineering, Beijing Technology & Business University, Fucheng Road, Haidian District, Beijing 100048, China
- Beijing Key Laboratory of Plant Resource Research and Development, College of Chemistry and Materials Engineering, Beijing Technology and Business University, Fucheng Road, Haidian District, Beijing 100048, China
| | - Shiquan You
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University, Fucheng Road, Beijing 100048, China
- Institute of Cosmetic Regulatory Science, College of Chemistry and Materials Engineering, Beijing Technology & Business University, Fucheng Road, Haidian District, Beijing 100048, China
- Beijing Key Laboratory of Plant Resource Research and Development, College of Chemistry and Materials Engineering, Beijing Technology and Business University, Fucheng Road, Haidian District, Beijing 100048, China
| | - Dongdong Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University, Fucheng Road, Beijing 100048, China
- Institute of Cosmetic Regulatory Science, College of Chemistry and Materials Engineering, Beijing Technology & Business University, Fucheng Road, Haidian District, Beijing 100048, China
- Beijing Key Laboratory of Plant Resource Research and Development, College of Chemistry and Materials Engineering, Beijing Technology and Business University, Fucheng Road, Haidian District, Beijing 100048, China
| | - Jiachan Zhang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University, Fucheng Road, Beijing 100048, China
- Institute of Cosmetic Regulatory Science, College of Chemistry and Materials Engineering, Beijing Technology & Business University, Fucheng Road, Haidian District, Beijing 100048, China
- Beijing Key Laboratory of Plant Resource Research and Development, College of Chemistry and Materials Engineering, Beijing Technology and Business University, Fucheng Road, Haidian District, Beijing 100048, China
| | - Meng Li
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University, Fucheng Road, Beijing 100048, China
- Institute of Cosmetic Regulatory Science, College of Chemistry and Materials Engineering, Beijing Technology & Business University, Fucheng Road, Haidian District, Beijing 100048, China
- Beijing Key Laboratory of Plant Resource Research and Development, College of Chemistry and Materials Engineering, Beijing Technology and Business University, Fucheng Road, Haidian District, Beijing 100048, China
- Correspondence:
| | - Changtao Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University, Fucheng Road, Beijing 100048, China
- Institute of Cosmetic Regulatory Science, College of Chemistry and Materials Engineering, Beijing Technology & Business University, Fucheng Road, Haidian District, Beijing 100048, China
- Beijing Key Laboratory of Plant Resource Research and Development, College of Chemistry and Materials Engineering, Beijing Technology and Business University, Fucheng Road, Haidian District, Beijing 100048, China
| |
Collapse
|
54
|
Yu FF, Yuan Y, Ao Y, Hua L, Wang W, Cao Y, Xi J, Luan Y, Hou S, Zhang XY. A New Product of Bilirubin Degradation by H 2O 2 and Its Formation in Activated Neutrophils and in an Inflammatory Mouse Model. Biomolecules 2022; 12:biom12091237. [PMID: 36139076 PMCID: PMC9496627 DOI: 10.3390/biom12091237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 08/26/2022] [Accepted: 09/01/2022] [Indexed: 11/17/2022] Open
Abstract
Bilirubin (BR) is a tetrapyrrolic compound stemming from heme catabolism with diverse physiological functions. It can be oxidized by H2O2 to form several degradation products, some of which have been detected in vivo and may contribute to the pathogenesis of certain diseases. However, the oxidative degradation of BR is complex and the conditions that BR degradation occurs pathophysiologically remain obscure. Neutrophils are known to generate large amounts of reactive oxygen species, including H2O2, upon activation and they are mobilized to inflammatory sites; therefore, we hypothesized that activated neutrophils could cause BR degradation, which could occur at inflammatory sites. In the present study, we investigated BR degradation by H2O2 and identified hematinic acid (BHP1) and a new product BHP2, whose structure was characterized as 2,5-diformyl-4-methyl-1H-pyrrole-3-propanoic acid. An LC-MS/MS method for the quantitation of the two compounds was then established. Using the LC-MS/MS method, we observed the concentration-dependent formation of BHP1 and BHP2 in mouse neutrophils incubated with 10 and 30 μM of BR with the yields being 16 ± 3.2 and 31 ± 5.9 pmol/106 cells for BHP1, and 25 ± 4.4 and 71 ± 26 pmol/106 cells for BHP2, respectively. After adding phorbol 12-myristate 13-acetate, a neutrophil agonist, to 30 μM of BR-treated cells, the BHP1 yield increased to 43 ± 6.6 pmol/106 cells, whereas the BHP2 one decreased to 47 ± 9.2 pmol/106 cells. The two products were also detected in hemorrhagic skins of mice with dermal inflammation and hemorrhage at levels of 4.5 ± 1.9 and 0.18 ± 0.10 nmol/g tissue, respectively, which were significantly higher than those in the non-hemorrhagic skins. BHP2 was neurotoxic starting at 0.10 μM but BHP1 was not, as assessed using Caenorhabditis elegans as the animal model. Neutrophil-mediated BR degradation may be a universally pathophysiological process in inflammation and can be particularly important under pathological conditions concerning hemorrhage.
Collapse
Affiliation(s)
- Fei-Fei Yu
- Institute of Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Yao Yuan
- Shanghai Jiao Tong University-Hangzhou Future Sci-Tech City Joint Research Center for Tumor Immunotherapy, Hangzhou Innovation Institute for Systems Oncology, Hangzhou 311121, China
| | - Yan Ao
- Ministry of Education-Shanghai Key Laboratory of Children’s Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Li Hua
- School of Public Health, Hongqiao International Institute of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Wu Wang
- Institute of Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
- Correspondence: (W.W.); (S.H.); (X.-Y.Z.)
| | - Yiyi Cao
- School of Public Health, Hongqiao International Institute of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jing Xi
- School of Public Health, Hongqiao International Institute of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yang Luan
- School of Public Health, Hongqiao International Institute of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Shangwei Hou
- Shanghai Jiao Tong University-Hangzhou Future Sci-Tech City Joint Research Center for Tumor Immunotherapy, Hangzhou Innovation Institute for Systems Oncology, Hangzhou 311121, China
- Correspondence: (W.W.); (S.H.); (X.-Y.Z.)
| | - Xin-Yu Zhang
- School of Public Health, Hongqiao International Institute of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Correspondence: (W.W.); (S.H.); (X.-Y.Z.)
| |
Collapse
|
55
|
Mauerhofer C, Afonyushkin T, Oskolkova OV, Hellauer K, Gesslbauer B, Schmerda J, Ke Y, Zimmer A, Birukova AA, Birukov KG, Bochkov V. Low Concentrations of Oxidized Phospholipids Increase Stress Tolerance of Endothelial Cells. Antioxidants (Basel) 2022; 11:1741. [PMID: 36139816 PMCID: PMC9495896 DOI: 10.3390/antiox11091741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/19/2022] [Accepted: 08/24/2022] [Indexed: 12/15/2022] Open
Abstract
Oxidized phospholipids (OxPLs) are generated by enzymatic or autooxidation of esterified polyunsaturated fatty acids (PUFAs) residues. OxPLs are present in circulation and atherosclerotic plaques where they are thought to induce predominantly proinflammatory and toxic changes in endothelial (ECs) and other cell types. Unexpectedly, we found that low concentrations of OxPLs were not toxic but protected ECs from stress induced by serum deprivation or cytostatic drugs. The protective effect was observed in ECs obtained from different vessels and was monitored using a variety of readouts based on different biological and chemical principles. Analysis of the structure−activity relationship identified oxidized or missing fatty acid residue (OxPLs or Lyso-PLs, respectively) as a prerequisite for the protective action of a PL. Protective OxPLs or Lyso-PLs acquired detergent-like properties and formed in solution aggregates <10 nm in diameter (likely micelles), which were in striking contrast with large aggregates (>1000 nm, likely multilayer liposomes) produced by nonoxidized precursor PLs. Because surfactants, OxPLs, and Lyso-PLs are known to extract membrane cholesterol, we tested if this effect might trigger the protection of endothelial cells. The protective action of OxPLs and Lyso-PLs was inhibited by cotreatment with cholesterol and mimicked by cholesterol-binding beta-cyclodextrin but not inactive α-cyclodextrin. Wide-scale mRNA expression analysis in four types of ECs showed the induction of genes encoding for heat shock proteins (HSPs) and secreted prosurvival peptides and proteins. Inducers of HSPs, chemical chaperones, and pure prosurvival factors mimicked the protective action of OxPLs/Lyso-PLs. We hypothesize that oxidation changes the physicochemical properties of PLs, thus promoting membrane cholesterol redistribution or extraction leading to the expression of intra- and extracellular prosurvival factors.
Collapse
Affiliation(s)
- Christina Mauerhofer
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, University of Graz, Humboldtstrasse 46/III, 8010 Graz, Austria
| | - Taras Afonyushkin
- Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse 17, 1090 Vienna, Austria
| | - Olga V. Oskolkova
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, University of Graz, Humboldtstrasse 46/III, 8010 Graz, Austria
| | - Klara Hellauer
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, University of Graz, Humboldtstrasse 46/III, 8010 Graz, Austria
| | - Bernd Gesslbauer
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, University of Graz, Humboldtstrasse 46/III, 8010 Graz, Austria
| | - Jasmin Schmerda
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, University of Graz, Humboldtstrasse 46/III, 8010 Graz, Austria
| | - Yunbo Ke
- Department of Anesthesiology, University of Maryland School of Medicine, 20 Penn. Street, HSF-2, Room 145, Baltimore, MD 21201, USA
| | - Andreas Zimmer
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical Technology and Biopharmacy, University of Graz, Universitätsplatz 1/EG, 8010 Graz, Austria
| | - Anna A. Birukova
- Department of Anesthesiology, University of Maryland School of Medicine, 20 Penn. Street, HSF-2, Room 145, Baltimore, MD 21201, USA
| | - Konstantin G. Birukov
- Department of Anesthesiology, University of Maryland School of Medicine, 20 Penn. Street, HSF-2, Room 145, Baltimore, MD 21201, USA
| | - Valery Bochkov
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, University of Graz, Humboldtstrasse 46/III, 8010 Graz, Austria
- Field of Excellence BioHealth, University of Graz, 8010 Graz, Austria
| |
Collapse
|
56
|
Canagliflozin Inhibits Human Endothelial Cell Inflammation through the Induction of Heme Oxygenase-1. Int J Mol Sci 2022; 23:ijms23158777. [PMID: 35955910 PMCID: PMC9369341 DOI: 10.3390/ijms23158777] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/02/2022] [Accepted: 08/05/2022] [Indexed: 11/16/2022] Open
Abstract
Sodium-glucose co-transporter 2 (SGLT2) inhibitors improve cardiovascular outcomes in patients with type 2 diabetes mellitus (T2DM). Studies have also shown that canagliflozin directly acts on endothelial cells (ECs). Since heme oxygenase-1 (HO-1) is an established modulator of EC function, we investigated if canagliflozin regulates the endothelial expression of HO-1, and if this enzyme influences the biological actions of canagliflozin in these cells. Treatment of human ECs with canagliflozin stimulated a concentration- and time-dependent increase in HO-1 that was associated with a significant increase in HO activity. Canagliflozin also evoked a concentration-dependent blockade of EC proliferation, DNA synthesis, and migration that was unaffected by inhibition of HO-1 activity and/or expression. Exposure of ECs to a diabetic environment increased the adhesion of monocytes to ECs, and this was attenuated by canagliflozin. Knockdown of HO-1 reduced the anti-inflammatory effect of canagliflozin which was restored by bilirubin but not carbon monoxide. In conclusion, this study identified canagliflozin as a novel inducer of HO-1 in human ECs. It also found that HO-1-derived bilirubin contributed to the anti-inflammatory action of canagliflozin, but not the anti-proliferative and antimigratory effects of the drug. The ability of canagliflozin to regulate HO-1 expression and EC function may contribute to the clinical profile of the drug.
Collapse
|
57
|
Machado SE, Spangler D, Stacks DA, Darley-Usmar V, Benavides GA, Xie M, Balla J, Zarjou A. Counteraction of Myocardial Ferritin Heavy Chain Deficiency by Heme Oxygenase-1. Int J Mol Sci 2022; 23:8300. [PMID: 35955444 PMCID: PMC9368247 DOI: 10.3390/ijms23158300] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/12/2022] [Accepted: 07/25/2022] [Indexed: 12/13/2022] Open
Abstract
Given the abundance of heme proteins (cytochromes) in the mitochondrion, it is evident that a meticulously orchestrated iron metabolism is essential for cardiac health. Here, we examined the functional significance of myocardial ferritin heavy chain (FtH) in a model of acute myocardial infarction. We report that FtH deletion did not alter either the mitochondrial regulatory and surveillance pathways (fission and fusion) or mitochondrial bioenergetics in response to injury. Furthermore, deletion of myocardial FtH did not affect cardiac function, assessed by measurement of left ventricular ejection fraction, on days 1, 7, and 21 post injury. To identify the modulated pathways providing cardiomyocyte protection coincident with FtH deletion, we performed unbiased transcriptomic analysis. We found that following injury, FtH deletion was associated with upregulation of several genes with anti-ferroptotic properties, including heme oxygenase-1 (HO-1) and the cystine/glutamate anti-porter (Slc7a11). These results suggested that HO-1 overexpression mitigates ferroptosis via upregulation of Slc7a11. Indeed, using transgenic mice with HO-1 overexpression, we demonstrate that overexpressed HO-1 is coupled with increased Slc7a11 expression. In conclusion, we demonstrate that following injury, myocardial FtH deletion leads to a compensatory upregulation in a number of anti-ferroptotic genes, including HO-1. Such HO-1 induction leads to overexpression of Slc7a11 and protects the heart against ischemia-reperfusion-mediated ferroptosis, preserves mitochondrial function, and overall function of the myocardium.
Collapse
Affiliation(s)
- Sarah E. Machado
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (S.E.M.); (D.S.); (D.A.S.)
| | - Daryll Spangler
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (S.E.M.); (D.S.); (D.A.S.)
| | - Delores A. Stacks
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (S.E.M.); (D.S.); (D.A.S.)
| | - Victor Darley-Usmar
- Mitochondrial Medicine Laboratory, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (V.D.-U.); (G.A.B.)
| | - Gloria A. Benavides
- Mitochondrial Medicine Laboratory, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (V.D.-U.); (G.A.B.)
| | - Min Xie
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA;
| | - József Balla
- ELKH-UD Vascular Pathophysiology Research Group 11003, Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary;
| | - Abolfazl Zarjou
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (S.E.M.); (D.S.); (D.A.S.)
| |
Collapse
|
58
|
Clinical and Laboratory Biomarkers in Paroxysmal Atrial Fibrillation: A Single Center Cross-Sectional Study. CONTRAST MEDIA & MOLECULAR IMAGING 2022; 2022:7012377. [PMID: 35845733 PMCID: PMC9259273 DOI: 10.1155/2022/7012377] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 06/03/2022] [Accepted: 06/07/2022] [Indexed: 11/18/2022]
Abstract
The clinical risk profile of paroxysmal atrial fibrillation (pAF) patients is inconclusive. We aimed to identify clinical and laboratory biomarkers in patients with pAF and the differences in biomarkers among genders. A cross-sectional study was conducted with a total of 181 participants in a single center in Beijing Anzhen Hospital. The participants were grouped according to the presence of pAF and sex differences, and clinical and laboratory results were collected and compared. The 181 participants had a mean age of 52.9 ± 15.1 years (pAF group, 60.4 ± 9.9 years, SR group, 48.3 ± 15.9 years, P < 0.05). Patients with pAF had significantly higher rates of age, left atrial (LA) diameter, haemoglobin (Hb) levels, tissue inhibitor of metalloproteinase-1 (TIMP-1), soluble tumour suppressor-2 (sST2), B-type natriuretic peptide (BNP) and indirect bilirubin (Ibil), mean haemoglobin concentration (MCHC), and hypertension (HTN) and smoking (P < 0.05). Multivariable logistic regression analysis revealed that age (OR = 1.075, 95% CI: 1.035–1.118, P < 0.0001), smoking (OR = 4.538, 95% CI: 1.559–13.205, P = 0.006), and MCHC (OR = 1.062, 95% CI: 1.019–1.106, P = 0.004) were independent predictive factors for pAF. Multivariable logistic regression analysis found that age (OR = 1.107, 95% CI: 1.016–1.206, P = 0.02) and Ibil level (OR = 2.303, 95% CI: 1.158–4.582, P = 0.017) were independent predictive factors of the occurrence of pAF in females; BNP (OR = 1.015, 95% CI: 1.002–1.029, P = 0.029) was an independent predictive variable of pAF in males. Age, smoking, and MCHC were independent predictive factors of pAF. BNP was an independent predictive biomarker of pAF in males, while in females, age and Ibil were independent predictive factors.
Collapse
|
59
|
Bell NT, Payne CM, Sammut IA, Larsen DS. Mechanistic Studies of Carbon Monoxide Release from Norborn‐2‐en‐7‐one CORMs. ASIAN J ORG CHEM 2022. [DOI: 10.1002/ajoc.202200350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Nathan T. Bell
- University of Otago - Dunedin Campus: University of Otago Chemistry NEW ZEALAND
| | | | - Ivan A Sammut
- University of Otago Pharmacology and Toxicology NEW ZEALAND
| | - David S Larsen
- University of Otago Chemistry Union Place WestPO Box 56 Dunedin 9054 9016 Dunedin NEW ZEALAND
| |
Collapse
|
60
|
Role of Heme Oxygenase in Gastrointestinal Epithelial Cells. Antioxidants (Basel) 2022; 11:antiox11071323. [PMID: 35883814 PMCID: PMC9311893 DOI: 10.3390/antiox11071323] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 02/04/2023] Open
Abstract
The gastrointestinal tract is a unique organ containing both vascular and luminal routes lined by epithelial cells forming the mucosa, which play an important role in the entry of nutrients and act as a selective barrier, excluding potentially harmful agents. Mucosal surfaces establish a selective barrier between hostile external environments and the internal milieu. Heme is a major nutritional source of iron and is a pro-oxidant that causes oxidative stress. Heme oxygenases (HOs) catalyze the rate-limiting step in heme degradation, resulting in the formation of iron, carbon monoxide, and biliverdin, which are subsequently converted to bilirubin by biliverdin reductase. In gastrointestinal pathogenesis, HO-1, an inducible isoform of HO, is markedly induced in epithelial cells and plays an important role in protecting mucosal cells. Recent studies have focused on the biological effects of the products of this enzymatic reaction, which have antioxidant, anti-inflammatory, and cytoprotective functions. In this review, the essential roles of HO in the gastrointestinal tract are summarized, focusing on nutrient absorption, protection against cellular stresses, and the maintenance and regulation of tight junction proteins, emphasizing the potential therapeutic implications. The biochemical basis of the potential therapeutic implications of glutamine for HO-1 induction in gastrointestinal injury is also discussed.
Collapse
|
61
|
Examining the expression levels of ferroptosis-related genes in angiographically determined coronary artery disease patients. Mol Biol Rep 2022; 49:7677-7686. [PMID: 35622307 DOI: 10.1007/s11033-022-07583-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 05/06/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Cardiovascular diseases are the leading cause of death worldwide, with several conditions being affected by oxidative stress. Ferroptosis, recently identified programmed cell death mechanism, is relies on oxidative stress. This study aimed to determine the expressions of the genes involved in the molecular pathways of oxidative stress and ferroptosis and the association of these genes with CAD risk factors in CAD and non-CAD individuals. METHODS AND RESULTS The blood samples of individuals who underwent coronary angiography were collected and divided according to CAD status. Total RNA isolation was performed using the PAXgene RNA isolation kit from the whole blood samples. The mRNA expression levels of RTN3, GPX4, CAT, HMOX1, ELOVL5, SLC25A1, SLC7A11, and ACSL4 genes were determined using Real-Time PCR. Biochemical analyses were done before coronary angiography, and the results were evaluated statistically. The expression levels of the CAT gene are significantly lower in the CAD group when compared to non-CAD. HMOX1 expression levels are positively correlated with stenosis percentage, Gensini, and SYNTAX scores in the CAD group. RTN3, SLC25A1, and GPX4 mRNA expressions are correlated with HDL-C levels. Moreover, HbA1c levels and BMI, correlate negatively with ACSL4 expression in non-CAD controls. Also, ELOVL5 expression is negatively correlated with total bilirubin and direct bilirubin levels in the CAD group. CONCLUSIONS In this study, the genes related to oxidative stress and ferroptosis were found associated with biochemical parameters associated with CAD risk. These preliminary results may provide a new perspective to further studies investigating the reasons behind the identified associations.
Collapse
|
62
|
Abdel Sater AH, Bouferraa Y, Amhaz G, Haibe Y, Lakkiss AE, Shamseddine A. From Tumor Cells to Endothelium and Gut Microbiome: A Complex Interaction Favoring the Metastasis Cascade. Front Oncol 2022; 12:804983. [PMID: 35600385 PMCID: PMC9117727 DOI: 10.3389/fonc.2022.804983] [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: 10/29/2021] [Accepted: 04/12/2022] [Indexed: 11/30/2022] Open
Abstract
Metastasis is a complicated process through which tumor cells disseminate to distant organs and adapt to novel tumor microenvironments. This multi-step cascade relies on the accumulation of genetic and epigenetic alterations within the tumor cells as well as the surrounding non-tumor stromal cells. Endothelial cells constitute a major player in promoting metastasis formation either by inducing the growth of tumor cells or by directing them towards dissemination in the blood or lymph. In fact, the direct and indirect interactions between tumor and endothelial cells were shown to activate several mechanisms allowing cancer cells’ invasion and extravasation. On the other side, gastrointestinal cancer development was shown to be associated with the disruption of the gut microbiome. While several proposed mechanisms have been investigated in this regard, gut and tumor-associated microbiota were shown to impact the gut endothelial barrier, increasing the dissemination of bacteria through the systemic circulation. This bacterial dislocation allows the formation of an inflammatory premetastatic niche in the distant organs promoting the metastatic cascade of primary tumors. In this review, we discuss the role of the endothelial cells in the metastatic cascade of tumors. We will focus on the role of the gut vascular barrier in the regulation metastasis. We will also discuss the interaction between this vascular barrier and the gut microbiota enhancing the process of metastasis. In addition, we will try to elucidate the different mechanisms through which this bacterial dislocation prepares the favorable metastatic niche at distant organs allowing the dissemination and successful deposition of tumor cells in the new microenvironments. Finally, and given the promising results of the studies combining immune checkpoint inhibitors with either microbiota alterations or anti-angiogenic therapy in many types of cancer, we will elaborate in this review the complex interaction between these 3 factors and their possible therapeutic combination to optimize response to treatment.
Collapse
Affiliation(s)
- Ali H Abdel Sater
- Department of Internal Medicine, Division of Hematology/Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Youssef Bouferraa
- Department of Internal Medicine, Division of Hematology/Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Ghid Amhaz
- Department of Internal Medicine, Division of Hematology/Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Yolla Haibe
- Department of Internal Medicine, Division of Hematology/Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Ahmed El Lakkiss
- Department of Internal Medicine, Division of Hematology/Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Ali Shamseddine
- Department of Internal Medicine, Division of Hematology/Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
63
|
Wang M, Yu J, Yang Q, Guo C, Zhang W, Li W, Weng Y, Ding Y, Wang J. Beta-Boswellic Acid Protects Against Cerebral Ischemia/Reperfusion Injury via the Protein Kinase C Epsilon/Nuclear Factor Erythroid 2-like 2/Heme Oxygenase-1 Pathway. Mol Neurobiol 2022; 59:4242-4256. [PMID: 35505050 DOI: 10.1007/s12035-022-02848-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 04/23/2022] [Indexed: 12/25/2022]
Abstract
Ischemic strokes are associated with a high rate of disability and death globally. Cerebral ischemia/reperfusion (I/R) injury is a type of brain damage associated with oxidative stress after an ischemic stroke. Beta-boswellic acid (β-BA) reportedly exerts antioxidant and neuroprotective effects, but its role in cerebral I/R injury is unclear. The aim of this research was to investigate the neuroprotective effects, as well as the mechanisms of β-BA in cerebral I/R injury. In vivo experiments were conducted using a rat middle cerebral artery occlusion and reperfusion (MCAO/R) model, and in vitro experiments were performed using a rat neuronal oxygen-glucose deprivation and reoxygenation (OGD/R) model. Triphenyltetrazolium chloride staining, neurological function scores, terminal deoxynucleotidyl transferase deoxyuridine triphosphate nick end labeling, hematoxylin and eosin staining, and antioxidant levels in the brain were used to assess the effects of β-BA. Flow cytometry was used to detect reactive oxygen species and apoptotic cells. Western blotting and immunofluorescence staining were used to measure protein levels. The results showed that β-BA markedly improved neurological deficits and decreased infarct volume and necrotic neurons in rats. The in vitro results showed that β-BA protected neurons against OGD/R-induced injury. Additionally, β-BA significantly increased the phosphorylation of protein kinase C epsilon (PRKCE) at S729, the translocation of nuclear factor erythroid 2-like 2 (NFE2L2), and expression of heme oxygenase-1 (HMOX1). This study demonstrates that β-BA exerts neuroprotective effects against cerebral I/R via the activation of the PRKCE/NFE2L2/HMOX1 pathway and is a potential therapeutic candidate for ischemic stroke.
Collapse
Affiliation(s)
- Mingming Wang
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Jiaoyan Yu
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Qi Yang
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Chao Guo
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Wei Zhang
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Weiwei Li
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Yan Weng
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Yi Ding
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| | - Jingwen Wang
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
64
|
Heme Oxygenase-1: An Anti-Inflammatory Effector in Cardiovascular, Lung, and Related Metabolic Disorders. Antioxidants (Basel) 2022; 11:antiox11030555. [PMID: 35326205 PMCID: PMC8944973 DOI: 10.3390/antiox11030555] [Citation(s) in RCA: 97] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 02/24/2022] [Accepted: 03/10/2022] [Indexed: 12/12/2022] Open
Abstract
The heme oxygenase (HO) enzyme system catabolizes heme to carbon monoxide (CO), ferrous iron, and biliverdin-IXα (BV), which is reduced to bilirubin-IXα (BR) by biliverdin reductase (BVR). HO activity is represented by two distinct isozymes, the inducible form, HO-1, and a constitutive form, HO-2, encoded by distinct genes (HMOX1, HMOX2, respectively). HO-1 responds to transcriptional activation in response to a wide variety of chemical and physical stimuli, including its natural substrate heme, oxidants, and phytochemical antioxidants. The expression of HO-1 is regulated by NF-E2-related factor-2 and counter-regulated by Bach-1, in a heme-sensitive manner. Additionally, HMOX1 promoter polymorphisms have been associated with human disease. The induction of HO-1 can confer protection in inflammatory conditions through removal of heme, a pro-oxidant and potential catalyst of lipid peroxidation, whereas iron released from HO activity may trigger ferritin synthesis or ferroptosis. The production of heme-derived reaction products (i.e., BV, BR) may contribute to HO-dependent cytoprotection via antioxidant and immunomodulatory effects. Additionally, BVR and BR have newly recognized roles in lipid regulation. CO may alter mitochondrial function leading to modulation of downstream signaling pathways that culminate in anti-apoptotic, anti-inflammatory, anti-proliferative and immunomodulatory effects. This review will present evidence for beneficial effects of HO-1 and its reaction products in human diseases, including cardiovascular disease (CVD), metabolic conditions, including diabetes and obesity, as well as acute and chronic diseases of the liver, kidney, or lung. Strategies targeting the HO-1 pathway, including genetic or chemical modulation of HO-1 expression, or application of BR, CO gas, or CO donor compounds show therapeutic potential in inflammatory conditions, including organ ischemia/reperfusion injury. Evidence from human studies indicate that HO-1 expression may represent a biomarker of oxidative stress in various clinical conditions, while increases in serum BR levels have been correlated inversely to risk of CVD and metabolic disease. Ongoing human clinical trials investigate the potential of CO as a therapeutic in human disease.
Collapse
|
65
|
Liu C, Liu Y, Yu Y, Zhao Y, Yu A. Comprehensive analysis of ferroptosis-related genes and prognosis of cutaneous melanoma. BMC Med Genomics 2022; 15:39. [PMID: 35232428 PMCID: PMC8886785 DOI: 10.1186/s12920-022-01194-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 02/24/2022] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Cutaneous Melanoma (CM) is a malignant disease with increasing incidence and high mortality. Ferroptosis is a new kind of cell death and related to tumor blood and lymphatic metastasis. This study aims at using bioinformatics technology to construct a prognostic signature and identify ferroptosis-related biomarkers to improve the prognosis and treatment of cutaneous melanoma. METHODS We used bioinformatics tools to analyze RNA sequencing expression data with clinical information from multiple databases, utilized varieties of statistical methods to construct a ferroptosis-related prognostic signature of cutaneous melanoma and screened out specific genes with independent prognostic ability. RESULTS We obtained 22 ferroptosis-related (P < 0.05) prognostic DEGs in the uniCox regression analysis, among which 10 high-expressed genes (ATG5, CHAC1, FANCD2, FBXL5, HMOX2, HSPB1, NQO1, PEBP1, PRNP, SLC3A2) were screened out by LASSO regression analysis to establish a predictive model. Meanwhile, the ferroptosis-related signature and the nomogram we drew performed an excellent performance based on Kaplan-Meier (K-M), Receiver operating characteristic (ROC) and calibration curves. Univariate and multivariable cox analyses displayed that our model was greater than other prognostic features. GO and KEGG analyses revealed that 10-biomarker signature was mainly related to epidermis differentiation and immunity. ssGSEA analysis indicated that the immune status between the two risk groups was highly different. Besides, we found that two genes (CP, ZEB1) had independent prognostic ability and can be applied for drug research. Both genes were highly related to immunity. GSEA illustrated that ZEB1 may be involved in cellular functions such as proliferation, apoptosis, and migration, while CP was closely connected to immune cell related functions. CONCLUSION The present study suggested a 10-biomarker signature can be clinically used to predict the prognosis of cutaneous melanoma, which was better than conventional factors. CP and ZEB1 were independent prognostic genes and can be applied to guide treatment. In addition, ZEB1 mutation was highly related to overall survival in cutaneous melanoma, while CP may be associated with tumor progression. Our study comprehensively analyzed the relationship between iron metabolism, ferroptosis-related genes, and the prognosis of cutaneous melanoma, provided new insight for molecular mechanisms and treatment of ferroptosis and cutaneous melanoma.
Collapse
Affiliation(s)
- Changjiang Liu
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, People's Republic of China
| | - Yuhang Liu
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, People's Republic of China
| | - Yifeng Yu
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, People's Republic of China
| | - Yong Zhao
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, People's Republic of China
| | - Aixi Yu
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, People's Republic of China.
| |
Collapse
|
66
|
Yang R, Shen YJ, Chen M, Zhao JY, Chen SH, Zhang W, Song JK, Li L, Du GH. Quercetin attenuates ischemia reperfusion injury by protecting the blood-brain barrier through Sirt1 in MCAO rats. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2022; 24:278-289. [PMID: 34292112 DOI: 10.1080/10286020.2021.1949302] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 06/25/2021] [Indexed: 05/25/2023]
Abstract
The purpose of the present study was to examine the protective action and mechanisms of quercetin on the blood-brain barrier (BBB) in rats subjected to transient middle cerebral artery occlusion (tMCAO) and reperfusion. Quercetin (10, 30, 50 mg/kg) was intraperitoneally administered at the onset of reperfusion. The results showed that quercetin significantly reduced cerebral infarct volume, neurological deficit, BBB permeability and ROS generation via Sirt1/Nrf2/HO-1 signaling pathway. Moreover, EX527, a selective inhibitor of Sirt1, reversed these neuroprotective effects. Our findings indicate that quercetin has neuroprotective effects against cerebral ischemia-reperfusion injury by protecting BBB through Sirt1 signaling pathway in MCAO rats.
Collapse
Affiliation(s)
- Ran Yang
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yan-Jia Shen
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Miao Chen
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jia-Ying Zhao
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Shu-Han Chen
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Wen Zhang
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jun-Ke Song
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Li Li
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Guan-Hua Du
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
67
|
Yu J, Fu J, Zhang X, Cui X, Cheng M. The Integration of Metabolomic and Proteomic Analyses Revealed Alterations in Inflammatory-Related Protein Metabolites in Endothelial Progenitor Cells Subjected to Oscillatory Shear Stress. Front Physiol 2022; 13:825966. [PMID: 35250628 PMCID: PMC8889118 DOI: 10.3389/fphys.2022.825966] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/17/2022] [Indexed: 01/13/2023] Open
Abstract
Background Endothelial progenitor cells (EPCs) play essential roles in vascular repair. Our previous study suggests OSS would lead EPCs transdifferention into the mesenchymal cell that aggravates pathological vascular remodeling. The primary purpose of this study was to apply OSS in vitro in EPCs and then explore proteins, metabolites, and the protein-metabolite network of EPCs. Methods Endothelial progenitor cells were kept in static or treated with OSS. For OSS treatment, the Flexcell STR-4000 parallel plate flow system was used to simulate OSS for 12 h. Subsequently, an untargeted metabolomic LC/MS analysis and a TMT-labeled quantitative proteomic analysis were performed. Results A total of 4,699 differentially expressed proteins (DEPs) were identified, among which 73 differentially expressed proteins were potentially meaningful (P < 0.05), with 66 upregulated and 7 downregulated expressions. There were 5,664 differential metabolites (DEMs), of which 401 DEMs with biologically potential marker significance (VIP > 1, P < 0.05), of which 137 were upregulated and 264 were downregulated. The Prison correlation analysis of DEPs and DEMs was performed, and the combined DEPs–DEMs pathway analyses of the KGLM database show 39 pathways. Among the DEPs, including the Phosphoserine phosphatase (PSPH), Prostaglandin E synthase 3 (PTGES3), Glutamate–cysteine ligase regulatory subunit (GCLM), Transaldolase (TALDO1), Isocitrate dehydrogenase 1 (IDH1) and Glutathione S-transferase omega-1 (GSTO1), which are significantly enriched in the citric acid cycle (TCA cycle) and fatty acid metabolic pathways, promoting glycolysis and upregulation of fatty acid synthesis. Moreover, we screened the 6 DEPs with the highest correlation with DEMs for predicting the onset of early AS and performed qPCR to validate them. Conclusion The comprehensive analysis reveals the following main changes in EPCs after the OSS treatment: dysregulation of glutamate and glycine metabolism and their transport/catabolic related proteins. Disorders of fatty acid and glycerophospholipid metabolism accompanied by alterations in the corresponding metabolic enzymes. Elevated expression of glucose metabolism.
Collapse
|
68
|
Luo K, Ogawa M, Ayer A, Britton WJ, Stocker R, Kikuchi K, Oehlers SH. Zebrafish Heme Oxygenase 1a Is Necessary for Normal Development and Macrophage Migration. Zebrafish 2022; 19:7-17. [PMID: 35108124 DOI: 10.1089/zeb.2021.0058] [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] [Indexed: 01/02/2023] Open
Abstract
Heme oxygenase function is highly conserved between vertebrates where it plays important roles in normal embryonic development and controls oxidative stress. Expression of the zebrafish heme oxygenase 1 genes is known to be responsive to oxidative stress suggesting a conserved physiological function. In this study, we generate a knockout allele of zebrafish hmox1a and characterize the effects of hmox1a and hmox1b loss on embryonic development. We find that loss of hmox1a or hmox1b causes developmental defects in only a minority of embryos, in contrast to Hmox1 gene deletions in mice that cause loss of most embryos. Using a tail wound inflammation assay we find a conserved role for hmox1a, but not hmox1b, in normal macrophage migration to the wound site. Together our results indicate that zebrafish hmox1a has clearly a partitioned role from hmox1b that is more consistent with conserved functions of mammalian Heme oxygenase 1.
Collapse
Affiliation(s)
- Kaiming Luo
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, Australia.,Victor Chang Cardiac Research Institute, Darlinghurst, Australia
| | - Masahito Ogawa
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia
| | - Anita Ayer
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia.,The Heart Research Institute, Newtown, Australia
| | - Warwick J Britton
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, Australia.,Department of Clinical Immunology, Royal Prince Alfred Hospital, Camperdown, Australia
| | - Roland Stocker
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia.,The Heart Research Institute, Newtown, Australia
| | - Kazu Kikuchi
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia.,National Cerebral and Cardiovascular Center, Suita, Japan
| | - Stefan H Oehlers
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, Australia.,Sydney Institute for Infectious Diseases, The University of Sydney, Camperdown, Australia
| |
Collapse
|
69
|
Fleischhacker AS, Sarkar A, Liu L, Ragsdale SW. Regulation of protein function and degradation by heme, heme responsive motifs, and CO. Crit Rev Biochem Mol Biol 2022; 57:16-47. [PMID: 34517731 PMCID: PMC8966953 DOI: 10.1080/10409238.2021.1961674] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Heme is an essential biomolecule and cofactor involved in a myriad of biological processes. In this review, we focus on how heme binding to heme regulatory motifs (HRMs), catalytic sites, and gas signaling molecules as well as how changes in the heme redox state regulate protein structure, function, and degradation. We also relate these heme-dependent changes to the affected metabolic processes. We center our discussion on two HRM-containing proteins: human heme oxygenase-2, a protein that binds and degrades heme (releasing Fe2+ and CO) in its catalytic core and binds Fe3+-heme at HRMs located within an unstructured region of the enzyme, and the transcriptional regulator Rev-erbβ, a protein that binds Fe3+-heme at an HRM and is involved in CO sensing. We will discuss these and other proteins as they relate to cellular heme composition, homeostasis, and trafficking. In addition, we will discuss the HRM-containing family of proteins and how the stability and activity of these proteins are regulated in a dependent manner through the HRMs. Then, after reviewing CO-mediated protein regulation of heme proteins, we turn our attention to the involvement of heme, HRMs, and CO in circadian rhythms. In sum, we stress the importance of understanding the various roles of heme and the distribution of the different heme pools as they relate to the heme redox state, CO, and heme binding affinities.
Collapse
Affiliation(s)
- Angela S. Fleischhacker
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Anindita Sarkar
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Liu Liu
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Stephen W. Ragsdale
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
70
|
Heme oxygenase 1 gene single nucleotide polymorphism (rs2071746) as a predictor of esophageal varices development in cirrhotic patients. Meta Gene 2022. [DOI: 10.1016/j.mgene.2022.101013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
71
|
Hanna DA, Moore CM, Liu L, Yuan X, Dominic IM, Fleischhacker AS, Hamza I, Ragsdale SW, Reddi AR. Heme oxygenase-2 (HO-2) binds and buffers labile ferric heme in human embryonic kidney cells. J Biol Chem 2021; 298:101549. [PMID: 34973332 PMCID: PMC8808069 DOI: 10.1016/j.jbc.2021.101549] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/22/2021] [Accepted: 12/23/2021] [Indexed: 01/13/2023] Open
Abstract
Heme oxygenases (HOs) detoxify heme by oxidatively degrading it into carbon monoxide, iron, and biliverdin, which is reduced to bilirubin and excreted. Humans express two isoforms of HO: the inducible HO-1, which is upregulated in response to excess heme and other stressors, and the constitutive HO-2. Much is known about the regulation and physiological function of HO-1, whereas comparatively little is known about the role of HO-2 in regulating heme homeostasis. The biochemical necessity for expressing constitutive HO-2 is dependent on whether heme is sufficiently abundant and accessible as a substrate under conditions in which HO-1 is not induced. By measuring labile heme, total heme, and bilirubin in human embryonic kidney HEK293 cells with silenced or overexpressed HO-2, as well as various HO-2 mutant alleles, we found that endogenous heme is too limiting a substrate to observe HO-2-dependent heme degradation. Rather, we discovered a novel role for HO-2 in the binding and buffering of heme. Taken together, in the absence of excess heme, we propose that HO-2 regulates heme homeostasis by acting as a heme buffering factor that controls heme bioavailability. When heme is in excess, HO-1 is induced, and both HO-2 and HO-1 can provide protection from heme toxicity via enzymatic degradation. Our results explain why catalytically inactive mutants of HO-2 are cytoprotective against oxidative stress. Moreover, the change in bioavailable heme due to HO-2 overexpression, which selectively binds ferric over ferrous heme, is consistent with labile heme being oxidized, thereby providing new insights into heme trafficking and signaling.
Collapse
Affiliation(s)
- David A. Hanna
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Courtney M. Moore
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Liu Liu
- Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan, USA
| | - Xiaojing Yuan
- Department of Animal and Avian Sciences, Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, USA
| | - Iramofu M. Dominic
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia, USA
| | | | - Iqbal Hamza
- Department of Animal and Avian Sciences, Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, USA
| | - Stephen W. Ragsdale
- Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan, USA
| | - Amit R. Reddi
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia, USA,School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia, USA,Parker Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, Georgia, USA,For correspondence: Amit R. Reddi
| |
Collapse
|
72
|
Sharifi-Rad J, Quispe C, Ayatollahi SA, Kobarfard F, Staniak M, Stępień A, Czopek K, Sen S, Acharya K, Matthews KR, Sener B, Devkota HP, Kırkın C, Özçelik B, Victoriano M, Martorell M, Rasul Suleria HA, Alshehri MM, Chandran D, Kumar M, Cruz-Martins N, Cho WC. Chemical Composition, Biological Activity, and Health-Promoting Effects of Withania somnifera for Pharma-Food Industry Applications. J FOOD QUALITY 2021; 2021:1-14. [DOI: 10.1155/2021/8985179] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2023] Open
Abstract
The Withania genus comes from the Solanaceae family and includes around 23 species, spread over some areas of the Mediterranean, Asia, and East Africa. Widely used in traditional medicine for thousands of years, these plants are rich in secondary metabolites, with special emphasis on steroidal lactones, named withanolides which are used as ingredients in numerous formulations for a plethora of diseases, such as asthma, diabetes, arthritis, impotence, amnesia, hypertension, anxiety, stress, cancer, neurodegenerative, and cardiovascular diseases, and many others. Among them, Withania somnifera (L.) Dunal is the most widely addressed species from a pharmacological and agroindustrial point of view. In this sense, this review provides an overview of the folk uses, phytochemical composition, and biological activity, such as antioxidant, antimicrobial, anti-inflammatory, and cytotoxic activity of W. somnifera, although more recently other species have also been increasingly investigated. In addition, their health-promoting effects, i.e., antistress, anxiolytic, adaptogenic, antirheumatoid arthritis, chemoprotective, and cardiorespiratory-enhancing abilities, along with safety and adverse effects are also discussed.
Collapse
Affiliation(s)
- Javad Sharifi-Rad
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Cristina Quispe
- Facultad de Ciencias de la Salud, Universidad Arturo Prat, Avda. Arturo Prat 2120, Iquique 1110939, Chile
| | - Seyed Abdulmajid Ayatollahi
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Pharmacognosy and Biotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farzad Kobarfard
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Medicinal Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mariola Staniak
- Institute of Soil Science and Plant Cultivation–State Research Institute, Czartoryskich 8, Puławy 24-100, Poland
| | - Anna Stępień
- Institute of Soil Science and Plant Cultivation–State Research Institute, Czartoryskich 8, Puławy 24-100, Poland
| | - Katarzyna Czopek
- Institute of Soil Science and Plant Cultivation–State Research Institute, Czartoryskich 8, Puławy 24-100, Poland
| | - Surjit Sen
- Molecular and Applied Mycology and Plant Pathology Laboratory, Department of Botany, University of Calcutta, Kolkata 700019, India
- Department of Botany, Fakir Chand College, Diamond Harbour, West Bengal 743331, India
| | - Krishnendu Acharya
- Molecular and Applied Mycology and Plant Pathology Laboratory, Department of Botany, University of Calcutta, Kolkata 700019, India
| | - Karl R. Matthews
- Department of Food Science, Rutgers University, New Brunswick, New Jersey, USA
| | - Bilge Sener
- Gazi University, Faculty of Pharmacy, Department of Pharmacognosy, Ankara 06330, Turkey
| | - Hari Prasad Devkota
- Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Kumamoto 862-0973, Japan
| | - Celale Kırkın
- Department Food Engineering, Faculty of Chemical and Metallurgical Engineering, Istanbul Technical University, Maslak, Istanbul 34469, Turkey
| | - Beraat Özçelik
- Department Food Engineering, Faculty of Chemical and Metallurgical Engineering, Istanbul Technical University, Maslak, Istanbul 34469, Turkey
- Bioactive Research & Innovation Food Manufacturing Industry Trade Ltd. Co., Maslak, Istanbul 34469, Turkey
| | - Montserrat Victoriano
- Department of Nutrition and Dietetics, Faculty of Pharmacy, University of Concepción, Concepción 4070386, Chile
| | - Miquel Martorell
- Department of Nutrition and Dietetics, Faculty of Pharmacy, University of Concepción, Concepción 4070386, Chile
- Centre for Healthy Living, University of Concepción, Concepción 4070386, Chile
| | | | - Mohammed M. Alshehri
- Pharmaceutical Care Department, Ministry of National Guard-Health Affairs, Riyadh, Saudi Arabia
| | - Deepak Chandran
- Department of Veterinary Sciences and Animal Husbandry, Amrita School of Agricultural Sciences, Amrita Vishwa Vidyapeetham University, Coimbatore 642109, India
| | - Manoj Kumar
- Chemical and Biochemical Processing Division, ICAR–Central Institute for Research on Cotton Technology, Mumbai 400019, India
| | - Natália Cruz-Martins
- Faculty of Medicine, University of Porto, Porto, Portugal
- Institute for Research and Innovation in Health (i3S), University of Porto, Porto, Portugal
- Institute of Research and Advanced Training in Health Sciences and Technologies (CESPU), Rua Central de Gandra, 1317, Gandra 4585-116, Portugal
- TOXRUN–Toxicology Research Unit, University Institute of Health Sciences, CESPU, CRL, 4585-116 Gandra, Portugal
| | - William C. Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong
| |
Collapse
|
73
|
Rodriguez-Gonzalez JC, Hernández-Balmaseda I, Declerck K, Pérez-Novo C, Logie E, Theys C, Jakubek P, Quiñones-Maza OL, Dantas-Cassali G, Carlos Dos Reis D, Van Camp G, Lopes Paz MT, Rodeiro-Guerra I, Delgado-Hernández R, Vanden Berghe W. Antiproliferative, Antiangiogenic, and Antimetastatic Therapy Response by Mangiferin in a Syngeneic Immunocompetent Colorectal Cancer Mouse Model Involves Changes in Mitochondrial Energy Metabolism. Front Pharmacol 2021; 12:670167. [PMID: 34924998 PMCID: PMC8678272 DOI: 10.3389/fphar.2021.670167] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 11/03/2021] [Indexed: 12/24/2022] Open
Abstract
In spite of the current advances and achievements in cancer treatments, colorectal cancer (CRC) persists as one of the most prevalent and deadly tumor types in both men and women worldwide. Drug resistance, adverse side effects and high rate of angiogenesis, metastasis and tumor relapse remain one of the greatest challenges in long-term management of CRC and urges need for new leads of anticancer drugs. We demonstrate that CRC treatment with the phytopharmaceutical mangiferin (MGF), a glucosylxanthone present in Mango tree stem bark and leaves (Mangifera Indica L.), induces dose-dependent tumor regression and decreases lung metastasis in a syngeneic immunocompetent allograft mouse model of murine CT26 colon carcinoma, which increases overall survival of mice. Antimetastatic and antiangiogenic MGF effects could be further validated in a wound healing in vitro model in human HT29 cells and in a matrigel plug implant mouse model. Interestingly, transcriptome pathway enrichment analysis demonstrates that MGF inhibits tumor growth, metastasis and angiogenesis by multi-targeting of mitochondrial oxidoreductase and fatty acid β-oxidation metabolism, PPAR, SIRT, NFκB, Stat3, HIF, Wnt and GP6 signaling pathways. MGF effects on fatty acid β-oxidation metabolism and carnitine palmitoyltransferase 1 (CPT1) protein expression could be further confirmed in vitro in human HT29 colon cells. In conclusion, antitumor, antiangiogenic and antimetastatic effects of MGF treatment hold promise to reduce adverse toxicity and to mitigate therapeutic outcome of colorectal cancer treatment by targeting mitochondrial energy metabolism in the tumor microenvironment.
Collapse
Affiliation(s)
| | | | - Ken Declerck
- Laboratory of Protein Science, Proteomics and Epigenetic Signaling (PPES) and Integrated Personalized and Precision Oncology Network (IPPON), Department of Biomedical Sciences, University of Antwerp, Campus Drie Eiken, Antwerp, Belgium
| | - Claudina Pérez-Novo
- Laboratory of Protein Science, Proteomics and Epigenetic Signaling (PPES) and Integrated Personalized and Precision Oncology Network (IPPON), Department of Biomedical Sciences, University of Antwerp, Campus Drie Eiken, Antwerp, Belgium
| | - Emilie Logie
- Laboratory of Protein Science, Proteomics and Epigenetic Signaling (PPES) and Integrated Personalized and Precision Oncology Network (IPPON), Department of Biomedical Sciences, University of Antwerp, Campus Drie Eiken, Antwerp, Belgium
| | - Claudia Theys
- Laboratory of Protein Science, Proteomics and Epigenetic Signaling (PPES) and Integrated Personalized and Precision Oncology Network (IPPON), Department of Biomedical Sciences, University of Antwerp, Campus Drie Eiken, Antwerp, Belgium
| | - Patrycja Jakubek
- Laboratory of Protein Science, Proteomics and Epigenetic Signaling (PPES) and Integrated Personalized and Precision Oncology Network (IPPON), Department of Biomedical Sciences, University of Antwerp, Campus Drie Eiken, Antwerp, Belgium.,Department of Food Chemistry, Technology and Biotechnology, Faculty of Chemistry, Gdansk University of Technology, Gdansk, Poland
| | | | - Geovanni Dantas-Cassali
- Departamento de Farmacología, Instituto de Ciencias Biológicas (ICB), Universidad Federal de Minas Gerais (UFMG), Horizonte, Brazil
| | - Diego Carlos Dos Reis
- Departamento de Farmacología, Instituto de Ciencias Biológicas (ICB), Universidad Federal de Minas Gerais (UFMG), Horizonte, Brazil
| | - Guy Van Camp
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Miriam Teresa Lopes Paz
- Departamento de Farmacología, Instituto de Ciencias Biológicas (ICB), Universidad Federal de Minas Gerais (UFMG), Horizonte, Brazil
| | - Idania Rodeiro-Guerra
- Laboratorio de Farmacología, Instituto de Ciencias del Mar (ICIMAR), CITMA, La Habana, Cuba
| | - René Delgado-Hernández
- Centro de Estudios para las Investigaciones y Evaluaciones Biológicas (CEIEB), Instituto de Farmacia y Alimentos (IFAL), Universidad de La Habana, La Habana, Cuba.,Facultad de Ciencias Naturales y Agropecuarias, Universidat de Santander (UDES), Bucaramanga, Colombia
| | - Wim Vanden Berghe
- Laboratory of Protein Science, Proteomics and Epigenetic Signaling (PPES) and Integrated Personalized and Precision Oncology Network (IPPON), Department of Biomedical Sciences, University of Antwerp, Campus Drie Eiken, Antwerp, Belgium
| |
Collapse
|
74
|
Zhou X, Yuan W, Xiong X, Zhang Z, Liu J, Zheng Y, Wang J, Liu J. HO-1 in Bone Biology: Potential Therapeutic Strategies for Osteoporosis. Front Cell Dev Biol 2021; 9:791585. [PMID: 34917622 PMCID: PMC8669958 DOI: 10.3389/fcell.2021.791585] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 11/12/2021] [Indexed: 02/05/2023] Open
Abstract
Osteoporosis is a prevalent bone disorder characterized by bone mass reduction and deterioration of bone microarchitecture leading to bone fragility and fracture risk. In recent decades, knowledge regarding the etiological mechanisms emphasizes that inflammation, oxidative stress and senescence of bone cells contribute to the development of osteoporosis. Studies have demonstrated that heme oxygenase 1 (HO-1), an inducible enzyme catalyzing heme degradation, exhibits anti-inflammatory, anti-oxidative stress and anti-apoptosis properties. Emerging evidence has revealed that HO-1 is critical in the maintenance of bone homeostasis, making HO-1 a potential target for osteoporosis treatment. In this Review, we aim to provide an introduction to current knowledge of HO-1 biology and its regulation, focusing specifically on its roles in bone homeostasis and osteoporosis. We also examine the potential of HO-1-based pharmacological therapeutics for osteoporosis and issues faced during clinical translation.
Collapse
Affiliation(s)
- Xueman Zhou
- State Key Laboratory of Oral Diseases and National Clinical Research Center for West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Lab for Aging Research, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Wenxiu Yuan
- State Key Laboratory of Oral Diseases and National Clinical Research Center for West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Lab for Aging Research, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xin Xiong
- State Key Laboratory of Oral Diseases and National Clinical Research Center for West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zhenzhen Zhang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Lab for Aging Research, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Jiaqi Liu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Lab for Aging Research, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Yingcheng Zheng
- State Key Laboratory of Oral Diseases and National Clinical Research Center for West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Lab for Aging Research, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Jun Wang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jin Liu
- Lab for Aging Research, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
75
|
Ahmad I, Molyvdas A, Jian MY, Zhou T, Traylor AM, Cui H, Liu G, Song W, Agarwal A, Jilling T, Aggarwal S, Matalon S. AICAR decreases acute lung injury by phosphorylating AMPK and upregulating heme oxygenase-1. Eur Respir J 2021; 58:2003694. [PMID: 34049949 PMCID: PMC9144003 DOI: 10.1183/13993003.03694-2020] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 05/11/2021] [Indexed: 11/05/2022]
Abstract
AIM We investigated the mechanisms by which N1-(β-d-ribofuranosyl)-5-aminoimidazole-4-carboxamide ribonucleotide (AICAR), an activator of AMP-activated protein kinase (AMPK), decreases lung injury and mortality when administered to mice post exposure to bromine gas (Br2). METHODS We exposed male C57BL/6 mice and heme oxygenase-1 (HO-1)-deficient (HO-1-/-) and corresponding wild-type (WT) littermate mice to Br2 (600 ppm for 45 or 30 min, respectively) in environmental chambers and returned them to room air. AICAR was administered 6 h post exposure (10 mg·kg-1, intraperitoneal). We assessed survival, indices of lung injury, high mobility group box 1 (HMGB1) in the plasma, HO-1 levels in lung tissues and phosphorylation of AMPK and its upstream liver kinase B1 (LKB1). Rat alveolar type II epithelial (L2) cells and human club-like epithelial (H441) cells were also exposed to Br2 (100 ppm for 10 min). After 24 h we measured apoptosis and necrosis, AMPK and LKB1 phosphorylation, and HO-1 expression. RESULTS There was a marked downregulation of phosphorylated AMPK and LKB1 in lung tissues and in L2 and H441 cells post exposure. AICAR increased survival in C57BL/6 but not in HO-1-/- mice. In WT mice, AICAR decreased lung injury and restored phosphorylated AMPK and phosphorylated LKB1 to control levels and increased HO-1 levels in both lung tissues and cells exposed to Br2. Treatment of L2 and H441 cells with small interfering RNAs against nuclear factor erythroid 2-related factor 2 or HO-1 abrogated the protective effects of AICAR. CONCLUSIONS Our data indicate that the primary mechanism for the protective action of AICAR in toxic gas injury is the upregulation of lung HO-1 levels.
Collapse
Affiliation(s)
- Israr Ahmad
- Division of Molecular and Translational Biomedicine, Dept of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Center for Pulmonary Injury and Repair, University of Alabama at Birmingham, Birmingham, AL, USA
- These authors contributed equally to this study
| | - Adam Molyvdas
- Division of Molecular and Translational Biomedicine, Dept of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Center for Pulmonary Injury and Repair, University of Alabama at Birmingham, Birmingham, AL, USA
- These authors contributed equally to this study
| | - Ming-Yuan Jian
- Division of Molecular and Translational Biomedicine, Dept of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Center for Pulmonary Injury and Repair, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ting Zhou
- Division of Molecular and Translational Biomedicine, Dept of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Center for Pulmonary Injury and Repair, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Amie M Traylor
- Division of Nephrology, Dept of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Huachun Cui
- Division of Pulmonary, Allergy, and Critical Care Medicine, Dept of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Gang Liu
- Division of Pulmonary, Allergy, and Critical Care Medicine, Dept of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Weifeng Song
- Dept of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Anupam Agarwal
- Division of Nephrology, Dept of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Tamas Jilling
- Division of Neonatology, Dept of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Saurabh Aggarwal
- Division of Molecular and Translational Biomedicine, Dept of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Center for Pulmonary Injury and Repair, University of Alabama at Birmingham, Birmingham, AL, USA
- These authors contributed equally as senior authors
| | - Sadis Matalon
- Division of Molecular and Translational Biomedicine, Dept of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Center for Pulmonary Injury and Repair, University of Alabama at Birmingham, Birmingham, AL, USA
- These authors contributed equally as senior authors
| |
Collapse
|
76
|
Roberts JA, Varma VR, An Y, Varma S, Candia J, Fantoni G, Tiwari V, Anerillas C, Williamson A, Saito A, Loeffler T, Schilcher I, Moaddel R, Khadeer M, Lovett J, Tanaka T, Pletnikova O, Troncoso JC, Bennett DA, Albert MS, Yu K, Niu M, Haroutunian V, Zhang B, Peng J, Croteau DL, Resnick SM, Gorospe M, Bohr VA, Ferrucci L, Thambisetty M. A brain proteomic signature of incipient Alzheimer's disease in young APOE ε4 carriers identifies novel drug targets. SCIENCE ADVANCES 2021; 7:eabi8178. [PMID: 34757788 PMCID: PMC8580310 DOI: 10.1126/sciadv.abi8178] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 09/14/2021] [Indexed: 05/13/2023]
Abstract
Aptamer-based proteomics revealed differentially abundant proteins in Alzheimer’s disease (AD) brains in the Baltimore Longitudinal Study of Aging and Religious Orders Study (mean age, 89 ± 9 years). A subset of these proteins was also differentially abundant in the brains of young APOE ε4 carriers relative to noncarriers (mean age, 39 ± 6 years). Several of these proteins represent targets of approved and experimental drugs for other indications and were validated using orthogonal methods in independent human brain tissue samples as well as in transgenic AD models. Using cell culture–based phenotypic assays, we showed that drugs targeting the cytokine transducer STAT3 and the Src family tyrosine kinases, YES1 and FYN, rescued molecular phenotypes relevant to AD pathogenesis. Our findings may accelerate the development of effective interventions targeting the earliest molecular triggers of AD.
Collapse
Affiliation(s)
- Jackson A. Roberts
- Clinical and Translational Neuroscience Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032
| | - Vijay R. Varma
- Clinical and Translational Neuroscience Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Yang An
- Brain Aging and Behavior Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | | | - Julián Candia
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Giovanna Fantoni
- Clinical Research Core, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Vinod Tiwari
- Section on DNA Repair, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Carlos Anerillas
- Laboratory of Genetics and Genomics, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Andrew Williamson
- Clinical and Translational Neuroscience Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Atsushi Saito
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Tina Loeffler
- QPS Austria GmbH, Parkring 12, 8074 Grambach, Austria
| | | | - Ruin Moaddel
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Mohammed Khadeer
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Jacqueline Lovett
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Toshiko Tanaka
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Olga Pletnikova
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Juan C. Troncoso
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - David A. Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
| | - Marilyn S. Albert
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Kaiwen Yu
- Departments of Structural Biology and Developmental Neurobiology, Center for Proteomics and Metabolomics, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Mingming Niu
- Departments of Structural Biology and Developmental Neurobiology, Center for Proteomics and Metabolomics, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Vahram Haroutunian
- Departments of Psychiatry and Neuroscience, The Alzheimer’s Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Mental Illness Research, Education and Clinical Center (MIRECC), James J. Peters VA Medical Center, Bronx, NY 10468, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences and Department of Pharmacological Sciences, Mount Sinai Center for Transformative Disease Modeling, Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Junmin Peng
- Departments of Structural Biology and Developmental Neurobiology, Center for Proteomics and Metabolomics, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Deborah L. Croteau
- Section on DNA Repair, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Susan M. Resnick
- Brain Aging and Behavior Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Myriam Gorospe
- Laboratory of Genetics and Genomics, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Vilhelm A. Bohr
- Section on DNA Repair, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Luigi Ferrucci
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Madhav Thambisetty
- Clinical and Translational Neuroscience Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| |
Collapse
|
77
|
Ferulic acid: A review of its pharmacology, pharmacokinetics and derivatives. Life Sci 2021; 284:119921. [PMID: 34481866 DOI: 10.1016/j.lfs.2021.119921] [Citation(s) in RCA: 202] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/17/2021] [Accepted: 08/25/2021] [Indexed: 12/14/2022]
Abstract
Ferulic acid, a kind of phenolic substance widely existing in plants, is an important active component of many traditional Chinese medicines. So far, it has been proved that ferulic acid has a variety of biological activities, especially in oxidative stress, inflammation, vascular endothelial injury, fibrosis, apoptosis and platelet aggregation. Many studies have shown that ferulic acid can inhibit PI3K/AKT pathway, the production of ROS and the activity of aldose reductase. The anti-inflammatory effect of ferulic acid is mainly related to the levels of PPAR γ, CAM and NF-κ B and p38 MAPK signaling pathways. Ferulic acid not only protects vascular endothelium by ERK1/2 and NO/ET-1 signal, but also plays an anti-fibrosis role by TGF-β/Smad and MMPs/TIMPs system. Moreover, ferulic acid has ant-apoptotic and anti-platelet effects. In addition to the pharmacological effects of ferulic acid, its pharmacokinetics and derivatives were also discussed in this paper. This review provides the latest summary of the latest research on ferulic acid.
Collapse
|
78
|
Piotrowski ER, Tift MS, Crocker DE, Pearson AB, Vázquez-Medina JP, Keith AD, Khudyakov JI. Ontogeny of Carbon Monoxide-Related Gene Expression in a Deep-Diving Marine Mammal. Front Physiol 2021; 12:762102. [PMID: 34744798 PMCID: PMC8567018 DOI: 10.3389/fphys.2021.762102] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 09/28/2021] [Indexed: 11/13/2022] Open
Abstract
Marine mammals such as northern elephant seals (NES) routinely experience hypoxemia and ischemia-reperfusion events to many tissues during deep dives with no apparent adverse effects. Adaptations to diving include increased antioxidants and elevated oxygen storage capacity associated with high hemoprotein content in blood and muscle. The natural turnover of heme by heme oxygenase enzymes (encoded by HMOX1 and HMOX2) produces endogenous carbon monoxide (CO), which is present at high levels in NES blood and has been shown to have cytoprotective effects in laboratory systems exposed to hypoxia. To understand how pathways associated with endogenous CO production and signaling change across ontogeny in diving mammals, we measured muscle CO and baseline expression of 17 CO-related genes in skeletal muscle and whole blood of three age classes of NES. Muscle CO levels approached those of animals exposed to high exogenous CO, increased with age, and were significantly correlated with gene expression levels. Muscle expression of genes associated with CO production and antioxidant defenses (HMOX1, BVR, GPX3, PRDX1) increased with age and was highest in adult females, while that of genes associated with protection from lipid peroxidation (GPX4, PRDX6, PRDX1, SIRT1) was highest in adult males. In contrast, muscle expression of mitochondrial biogenesis regulators (PGC1A, ESRRA, ESRRG) was highest in pups, while genes associated with inflammation (HMOX2, NRF2, IL1B) did not vary with age or sex. Blood expression of genes involved in regulation of inflammation (IL1B, NRF2, BVR, IL10) was highest in pups, while HMOX1, HMOX2 and pro-inflammatory markers (TLR4, CCL4, PRDX1, TNFA) did not vary with age. We propose that ontogenetic upregulation of baseline HMOX1 expression in skeletal muscle of NES may, in part, underlie increases in CO levels and expression of genes encoding antioxidant enzymes. HMOX2, in turn, may play a role in regulating inflammation related to ischemia and reperfusion in muscle and circulating immune cells. Our data suggest putative ontogenetic mechanisms that may enable phocid pups to transition to a deep-diving lifestyle, including high baseline expression of genes associated with mitochondrial biogenesis and immune system activation during postnatal development and increased expression of genes associated with protection from lipid peroxidation in adulthood.
Collapse
Affiliation(s)
| | - Michael S. Tift
- Department of Biology and Marine Biology, University of North Carolina Wilmington, Wilmington, NC, United States
| | - Daniel E. Crocker
- Biology Department, Sonoma State University, Rohnert Park, CA, United States
| | - Anna B. Pearson
- Department of Biology and Marine Biology, University of North Carolina Wilmington, Wilmington, NC, United States
| | - José P. Vázquez-Medina
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA, United States
| | - Anna D. Keith
- Department of Biological Sciences, University of the Pacific, Stockton, CA, United States
| | - Jane I. Khudyakov
- Department of Biological Sciences, University of the Pacific, Stockton, CA, United States
| |
Collapse
|
79
|
Potor L, Hendrik Z, Patsalos A, Katona É, Méhes G, Póliska S, Csősz É, Kalló G, Komáromi I, Combi Z, Posta N, Sikura KÉ, Pethő D, Oros M, Vereb G, Tóth C, Gergely P, Nagy L, Balla G, Balla J. Oxidation of Hemoglobin Drives a Proatherogenic Polarization of Macrophages in Human Atherosclerosis. Antioxid Redox Signal 2021; 35:917-950. [PMID: 34269613 PMCID: PMC8905252 DOI: 10.1089/ars.2020.8234] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Aim: The aim of our study was to explore the pathophysiologic role of oxidation of hemoglobin (Hb) to ferrylHb in human atherosclerosis. Results: We observed a severe oxidation of Hb to ferrylHb in complicated atherosclerotic lesions of carotid arteries with oxidative changes of the globin moieties, detected previously described oxidation hotspots in Hb (β1Cys93; β1Cys112; β2Cys112) and identified a novel oxidation hotspot (α1Cys104). After producing a monoclonal anti-ferrylHb antibody, ferrylHb was revealed to be localized extracellularly and also internalized by macrophages in the human hemorrhagic complicated lesions. We demonstrated that ferrylHb is taken up via phagocytosis as well as CD163 receptor-mediated endocytosis and then transported to lysosomes involving actin polymerization. Internalization of ferrylHb was accompanied by upregulation of heme oxygenase-1 and H-ferritin and accumulation of iron within lysosomes as a result of heme/iron uptake. Importantly, macrophages exposed to ferrylHb in atherosclerotic plaques exhibited a proinflammatory phenotype, as reflected by elevated levels of IL-1β and TNF-α. To find further signatures of ferrylHb in complicated lesions, we performed RNA-seq analysis on biopsies from patients who underwent endarterectomies. RNA-seq analysis demonstrated that human complicated lesions had a unique transcriptomic profile different from arteries and atheromatous plaques. Pathways affected in complicated lesions included gene changes associated with phosphoinositide 3-kinase (PI3K) signaling, lipid transport, tissue remodeling, and vascularization. Targeted analysis of gene expression associated with calcification, apoptosis, and hemolytic-specific clusters indicated an increase in the severity of complicated lesions compared with atheroma. A 39% overlap in the differential gene expression profiles of human macrophages exposed to ferrylHb and the complicated lesion profiles was uncovered. Among these 547 genes, we found inflammatory, angiogenesis, and iron metabolism gene clusters regulated in macrophages. Innovation and Conclusion: We conclude that oxidation of Hb to ferrylHb contributes to the progression of atherosclerosis via polarizing macrophages into a proatherogenic phenotype. Antioxid. Redox Signal. 35, 917-950.
Collapse
Affiliation(s)
- László Potor
- ELKH-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, Debrecen, Hungary.,Department of Pediatrics, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Division of Nephrology, Department of Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hungary
| | - Zoltán Hendrik
- ELKH-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, Debrecen, Hungary.,Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hungary.,Department of Pathology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Andreas Patsalos
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Department of Medicine and Johns Hopkins University School of Medicine, St. Petersburg, Florida, USA.,Department of Biological Chemistry, Johns Hopkins University School of Medicine, St. Petersburg, Florida, USA.,Johns Hopkins All Children's Hospital, St. Petersburg, Florida, USA
| | - Éva Katona
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Gábor Méhes
- Department of Pathology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Szilárd Póliska
- Proteomics Core Facility, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Éva Csősz
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Gergő Kalló
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - István Komáromi
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zsolt Combi
- Division of Nephrology, Department of Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hungary
| | - Niké Posta
- Division of Nephrology, Department of Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hungary
| | - Katalin Éva Sikura
- ELKH-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, Debrecen, Hungary.,Department of Pediatrics, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Dávid Pethő
- Division of Nephrology, Department of Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hungary
| | - Melinda Oros
- Division of Nephrology, Department of Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hungary
| | - György Vereb
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Csaba Tóth
- Division of Vascular Surgery, Department of Surgery, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Péter Gergely
- Department of Forensic Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - László Nagy
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Department of Medicine and Johns Hopkins University School of Medicine, St. Petersburg, Florida, USA
| | - György Balla
- ELKH-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, Debrecen, Hungary.,Department of Pediatrics, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - József Balla
- ELKH-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, Debrecen, Hungary.,Division of Nephrology, Department of Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
80
|
Callan MB, Thawley VJ, Marryott KA, Shabro A, Fernando S, Kahn S, Hudson KE, Hod EA. Hemolytic anemia blunts the cytokine response to transfusion of older red blood cells in mice and dogs. Transfusion 2021; 61:3309-3319. [PMID: 34633666 DOI: 10.1111/trf.16690] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/25/2021] [Accepted: 09/16/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Transfusion of red blood cells (RBCs) stored for longer durations induces hemolysis and inflammatory cytokine production in murine and canine models. Despite immune system activation by stored RBCs, human randomized trials suggest that fresher RBC transfusions do not improve clinical outcomes. We hypothesized that underlying recipient hemolysis may affect cytokine responses to older RBC transfusions. STUDY DESIGN AND METHODS C57BL/6 mouse cohorts were infused with anti-TER119 antibody to induce hemolysis, rabbit anti-platelet antiserum to induce immune thrombocytopenia (ITP), or appropriate control antibodies. Two days later, mice were transfused with fresh or stored RBCs. Furthermore, in a prospective, randomized, blinded trial, 38 client-owned dogs with primary autoimmune hemolytic anemia (AIHA) and two dogs with ITP, requiring RBC transfusion, were enrolled and randomized to receive fresh (≤7 days) or old (≥21 days) stored RBC transfusions. Monocyte chemoattractant protein (MCP)-1 levels were assessed at defined times after transfusion. RESULTS Prior immune-mediated hemolysis blunted the MCP-1 response to stored RBC transfusion in mice (361 ± 111 pg/ml vs. 6836 ± 1528 pg/ml in mice with immune hemolysis vs. ITP, respectively; mean ± SD; p < .0001). Although hemolysis markers increased after transfusion of older RBCs, the cytokine response was also muted in dogs with AIHA. No differences in morbidity or mortality were evident comparing dogs randomized to fresh or old RBCs. CONCLUSION These data suggest that underlying hemolysis blunts inflammatory responses to old RBC transfusions. The canine data support randomized trial results suggesting a lack of clinical benefit with fresh RBC transfusions in subjects with underlying, baseline hemolysis.
Collapse
Affiliation(s)
- Mary Beth Callan
- Department of Clinical Sciences and Advanced Medicine, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, USA
| | - Vincent J Thawley
- Department of Clinical Sciences and Advanced Medicine, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, USA
| | - Kimberly A Marryott
- Department of Clinical Sciences and Advanced Medicine, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, USA
| | - Aidin Shabro
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center - New York Presbyterian Hospital, New York, New York, USA
| | - Sebastian Fernando
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center - New York Presbyterian Hospital, New York, New York, USA
| | - Stacie Kahn
- Department of Pediatrics, Columbia University Irving Medical Center - New York Presbyterian Hospital, New York, New York, USA
| | - Krystalyn E Hudson
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center - New York Presbyterian Hospital, New York, New York, USA
| | - Eldad A Hod
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center - New York Presbyterian Hospital, New York, New York, USA
| |
Collapse
|
81
|
Matalon S. In celebration of the 100th anniversary of Physiological Reviews. Physiol Rev 2021; 101:1981-1985. [PMID: 34459221 DOI: 10.1152/physrev.00032.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- Sadis Matalon
- University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
82
|
Abstract
Significance: As the central metabolic organ, the liver is exposed to a variety of potentially cytotoxic, proinflammatory, profibrotic, and carcinogenic stimuli. To protect the organism from these deleterious effects, the liver has evolved a number of defense systems, which include antioxidant substrates and enzymes, anti-inflammatory tools, enzymatic biotransformation systems, and metabolic pathways. Recent Advances: One of the pivotal systems that evolved during phylogenesis was the heme catabolic pathway. Comprising the important enzymes heme oxygenase and biliverdin reductase, this complex pathway has a number of key functions including enzymatic activities, but also cell signaling, and DNA transcription. It further generates two important bile pigments, biliverdin and bilirubin, as well as the gaseous molecule carbon monoxide. These heme degradation products have potent antioxidant, immunosuppressive, and cytoprotective effects. Recent data suggest that the pathway participates in the regulation of metabolic and hormonal processes implicated in the pathogenesis of hepatic and other diseases. Critical Issues: This review discusses the impact of the heme catabolic pathway on major liver diseases, with particular focus on the involvement of cellular targeting and signaling in the pathogenesis of these conditions. Future Directions: To utilize the biological consequences of the heme catabolic pathway, several unique therapeutic strategies have been developed. Research indicates that pharmaceutical, nutraceutical, and lifestyle modifications positively affect the pathway, delivering potentially long-term clinical benefits. However, further well-designed studies are needed to confirm the clinical benefits of these approaches. Antioxid. Redox Signal. 35, 734-752.
Collapse
Affiliation(s)
- Libor Vítek
- Fourth Department of Internal Medicine, and Institute of Medical Biochemistry and Laboratory Diagnostics, General University Hospital and First Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
83
|
Alonso-Piñeiro JA, Gonzalez-Rovira A, Sánchez-Gomar I, Moreno JA, Durán-Ruiz MC. Nrf2 and Heme Oxygenase-1 Involvement in Atherosclerosis Related Oxidative Stress. Antioxidants (Basel) 2021; 10:1463. [PMID: 34573095 PMCID: PMC8466960 DOI: 10.3390/antiox10091463] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/02/2021] [Accepted: 09/09/2021] [Indexed: 12/19/2022] Open
Abstract
Atherosclerosis remains the underlying process responsible for cardiovascular diseases and the high mortality rates associated. This chronic inflammatory disease progresses with the formation of occlusive atherosclerotic plaques over the inner walls of vascular vessels, with oxidative stress being an important element of this pathology. Oxidation of low-density lipoproteins (ox-LDL) induces endothelial dysfunction, foam cell activation, and inflammatory response, resulting in the formation of fatty streaks in the atherosclerotic wall. With this in mind, different approaches aim to reduce oxidative damage as a strategy to tackle the progression of atherosclerosis. Special attention has been paid in recent years to the transcription factor Nrf2 and its downstream-regulated protein heme oxygenase-1 (HO-1), both known to provide protection against atherosclerotic injury. In the current review, we summarize the involvement of oxidative stress in atherosclerosis, focusing on the role that these antioxidant molecules exert, as well as the potential therapeutic strategies applied to enhance their antioxidant and antiatherogenic properties.
Collapse
Affiliation(s)
- Jose Angel Alonso-Piñeiro
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, 11519 Puerto Real, Spain; (J.A.A.-P.); (A.G.-R.); (I.S.-G.)
- Institute of Research and Innovation in Biomedical Sciences of Cádiz (INiBICA), 11001 Cádiz, Spain
| | - Almudena Gonzalez-Rovira
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, 11519 Puerto Real, Spain; (J.A.A.-P.); (A.G.-R.); (I.S.-G.)
- Institute of Research and Innovation in Biomedical Sciences of Cádiz (INiBICA), 11001 Cádiz, Spain
| | - Ismael Sánchez-Gomar
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, 11519 Puerto Real, Spain; (J.A.A.-P.); (A.G.-R.); (I.S.-G.)
- Institute of Research and Innovation in Biomedical Sciences of Cádiz (INiBICA), 11001 Cádiz, Spain
| | - Juan Antonio Moreno
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), UGC Nephrology, Hospital Universitario Reina Sofia, 14004 Cordoba, Spain;
- Department of Cell Biology, Physiology, and Immunology, Agrifood Campus of International Excellence (ceiA3), University of Cordoba, 14014 Cordoba, Spain
| | - Ma Carmen Durán-Ruiz
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, 11519 Puerto Real, Spain; (J.A.A.-P.); (A.G.-R.); (I.S.-G.)
- Institute of Research and Innovation in Biomedical Sciences of Cádiz (INiBICA), 11001 Cádiz, Spain
| |
Collapse
|
84
|
Synchronized Cardiac Impulses Emerge From Heterogeneous Local Calcium Signals Within and Among Cells of Pacemaker Tissue. JACC Clin Electrophysiol 2021; 6:907-931. [PMID: 32819526 DOI: 10.1016/j.jacep.2020.06.022] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/19/2020] [Accepted: 06/24/2020] [Indexed: 12/11/2022]
Abstract
OBJECTIVES This study sought to identify subcellular Ca2+ signals within and among cells comprising the sinoatrial node (SAN) tissue. BACKGROUND The current paradigm of SAN impulse generation: 1) is that full-scale action potentials (APs) of a common frequency are initiated at 1 site and are conducted within the SAN along smooth isochrones; and 2) does not feature fine details of Ca2+ signaling present in isolated SAN cells, in which small subcellular, subthreshold local Ca2+ releases (LCRs) self-organize to generate cell-wide APs. METHODS Immunolabeling was combined with a novel technique to detect the occurrence of LCRs and AP-induced Ca2+ transients (APCTs) in individual pixels (chronopix) across the entire mouse SAN images. RESULTS At high magnification, Ca2+ signals appeared markedly heterogeneous in space, amplitude, frequency, and phase among cells comprising an HCN4+/CX43- cell meshwork. The signaling exhibited several distinguishable patterns of LCR/APCT interactions within and among cells. Rhythmic APCTs that were apparently conducted within the meshwork were transferred to a truly conducting HCN4-/CX43+ network of striated cells via narrow functional interfaces where different cell types intertwine, that is, the SAN anatomic/functional unit. At low magnification, the earliest APCT of each cycle occurred within a small area of the HCN4 meshwork, and subsequent APCT appearance throughout SAN pixels was discontinuous and asynchronous. CONCLUSIONS The study has discovered a novel, microscopic Ca2+ signaling paradigm of SAN operation that has escaped detection using low-resolution, macroscopic tissue isochrones employed in prior studies: synchronized APs emerge from heterogeneous subcellular subthreshold Ca2+ signals, resembling multiscale complex processes of impulse generation within clusters of neurons in neuronal networks.
Collapse
|
85
|
Wang T, Ashrafi A, Modareszadeh P, Deese AR, Chacon Castro MDC, Alemi PS, Zhang L. An Analysis of the Multifaceted Roles of Heme in the Pathogenesis of Cancer and Related Diseases. Cancers (Basel) 2021; 13:4142. [PMID: 34439295 PMCID: PMC8393563 DOI: 10.3390/cancers13164142] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/08/2021] [Accepted: 08/13/2021] [Indexed: 12/28/2022] Open
Abstract
Heme is an essential prosthetic group in proteins and enzymes involved in oxygen utilization and metabolism. Heme also plays versatile and fascinating roles in regulating fundamental biological processes, ranging from aerobic respiration to drug metabolism. Increasing experimental and epidemiological data have shown that altered heme homeostasis accelerates the development and progression of common diseases, including various cancers, diabetes, vascular diseases, and Alzheimer's disease. The effects of heme on the pathogenesis of these diseases may be mediated via its action on various cellular signaling and regulatory proteins, as well as its function in cellular bioenergetics, specifically, oxidative phosphorylation (OXPHOS). Elevated heme levels in cancer cells intensify OXPHOS, leading to higher ATP generation and fueling tumorigenic functions. In contrast, lowered heme levels in neurons may reduce OXPHOS, leading to defects in bioenergetics and causing neurological deficits. Further, heme has been shown to modulate the activities of diverse cellular proteins influencing disease pathogenesis. These include BTB and CNC homology 1 (BACH1), tumor suppressor P53 protein, progesterone receptor membrane component 1 protein (PGRMC1), cystathionine-β-synthase (CBS), soluble guanylate cyclase (sGC), and nitric oxide synthases (NOS). This review provides an in-depth analysis of heme function in influencing diverse molecular and cellular processes germane to disease pathogenesis and the modes by which heme modulates the activities of cellular proteins involved in the development of cancer and other common diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Li Zhang
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX 75080, USA; (T.W.); (A.A.); (P.M.); (A.R.D.); (M.D.C.C.C.); (P.S.A.)
| |
Collapse
|
86
|
Balla J, Zarjou A. Heme Burden and Ensuing Mechanisms That Protect the Kidney: Insights from Bench and Bedside. Int J Mol Sci 2021; 22:8174. [PMID: 34360940 PMCID: PMC8347331 DOI: 10.3390/ijms22158174] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 07/23/2021] [Accepted: 07/27/2021] [Indexed: 02/06/2023] Open
Abstract
With iron at its core, the tetrapyrrole heme ring is a cardinal prosthetic group made up of many proteins that participate in a wide array of cellular functions and metabolism. Once released, due to its pro-oxidant properties, free heme in sufficient amounts can result in injurious effects to the kidney and other organs. Heme oxygenase-1 (HO-1) has evolved to promptly attend to such injurious potential by facilitating degradation of heme into equimolar amounts of carbon monoxide, iron, and biliverdin. HO-1 induction is a beneficial response to tissue injury in diverse animal models of diseases, including those that affect the kidney. These protective attributes are mainly due to: (i) prompt degradation of heme leading to restraining potential hazardous effects of free heme, and (ii) generation of byproducts that along with induction of ferritin have proven beneficial in a number of pathological conditions. This review will focus on describing clinical aspects of some of the conditions with the unifying end-result of increased heme burden and will discuss the molecular mechanisms that ensue to protect the kidneys.
Collapse
Affiliation(s)
- József Balla
- ELKH-UD Vascular Biology and Myocardial Pathophysiology Research Group, Division of Nephrology, Department of Medicine, Faculty of Medicine, Hungarian Academy of Sciences, H-4032 Debrecen, Hungary;
| | - Abolfazl Zarjou
- Nephrology Research and Training Center, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, 618 Zeigler Research Building, 703 South 19th Street, Birmingham, AL 35294, USA
| |
Collapse
|
87
|
Gao H, Sun X, Liu Y, Liang S, Zhang B, Wang L, Ren J. Analysis of Hub Genes and the Mechanism of Immune Infiltration in Stanford Type a Aortic Dissection. Front Cardiovasc Med 2021; 8:680065. [PMID: 34277731 PMCID: PMC8284479 DOI: 10.3389/fcvm.2021.680065] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 06/04/2021] [Indexed: 12/11/2022] Open
Abstract
Background: Stanford type A aortic dissection (AAD) is a catastrophic disease. An immune infiltrate has been found within the aortic wall of dissected aortic specimens. The recall and activation of macrophages are key events in the early phases of AAD. Herein, the immune filtration profile of AAD was uncovered. Methods: Gene expression data from the GSE52093, GSE98770 and GSE153434 datasets were downloaded from the Gene Expression Omnibus (GEO). The differentially expressed genes (DEGs) of each dataset were calculated and then integrated. A protein-protein interaction (PPI) network was established with the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING), and the hub genes were identified in Cytoscape. Furthermore, gene ontology (GO) functional annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis of hub genes were performed. Finally, we set GSE52093 and GSE98770 as the training set and GSE153434 as the validation set to assess immune infiltration in AAD using CIBERSORTx and analyzed the correlations between immune cells and hub genes in both the training and validation sets. Results: Sixty-one integrated DEGs were identified. The top 10 hub genes were selected from the PPI network, and 140 biological process (BP) terms and 12 pathways were enriched among the top 10 hub genes. The proportions of monocytes and macrophages were significantly higher in AAD tissues than in normal tissues. Notably, this result was consistent in the training set and the validation set. In addition, we found that among the hub genes, CA9, CXCL5, GDF15, VEGFA, CCL20, HMOX1, and SPP1 were positively correlated with CD14, a cell marker of monocytes, while CA9, CXCL5, GDF15, and VEGFA were positively correlated with CD68, a cell marker of macrophages in the training set. Finally, according to the results of the GO and KEGG analysis of hub genes, we found that the monocyte/macrophage-related genes were involved in immune-inflammatory responses through degradation of the extracellular matrix, endothelial cell apoptosis, hypoxia and the interaction of cytokines and chemokines. Conclusion: The monocyte-macrophage system plays a major role in immune-inflammatory responses in the development of AAD. Several hub genes are involved in this process via diverse mechanisms.
Collapse
Affiliation(s)
- Haoyu Gao
- Department of Cardiovascular Surgery, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaogang Sun
- Department of Cardiovascular Surgery, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yanxiang Liu
- Department of Cardiovascular Surgery, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shenghua Liang
- Department of Cardiovascular Surgery, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bowen Zhang
- Department of Cardiovascular Surgery, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Luchen Wang
- Department of Cardiovascular Surgery, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jie Ren
- Department of Cardiovascular Surgery, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
88
|
Zhou Y, Liao J, Mei Z, Liu X, Ge J. Insight into Crosstalk between Ferroptosis and Necroptosis: Novel Therapeutics in Ischemic Stroke. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:9991001. [PMID: 34257829 PMCID: PMC8257382 DOI: 10.1155/2021/9991001] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/30/2021] [Accepted: 06/14/2021] [Indexed: 12/16/2022]
Abstract
Ferroptosis is a nonapoptotic form of cell death characterized by iron-dependent accumulation of lipid hydroperoxides to lethal levels. Necroptosis, an alternative form of programmed necrosis, is regulated by receptor-interacting protein (RIP) 1 activation and by RIP3 and mixed-lineage kinase domain-like (MLKL) phosphorylation. Ferroptosis and necroptosis both play important roles in the pathological progress in ischemic stroke, which is a complex brain disease regulated by several cell death pathways. In the past few years, increasing evidence has suggested that the crosstalk occurs between necroptosis and ferroptosis in ischemic stroke. However, the potential links between ferroptosis and necroptosis in ischemic stroke have not been elucidated yet. Hence, in this review, we overview and analyze the mechanism underlying the crosstalk between necroptosis and ferroptosis in ischemic stroke. And we find that iron overload, one mechanism of ferroptosis, leads to mitochondrial permeability transition pore (MPTP) opening, which aggravates RIP1 phosphorylation and contributes to necroptosis. In addition, heat shock protein 90 (HSP90) induces necroptosis and ferroptosis by promoting RIP1 phosphorylation and suppressing glutathione peroxidase 4 (GPX4) activation. In this work, we try to deliver a new perspective in the exploration of novel therapeutic targets for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Yue Zhou
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Jun Liao
- Medical School, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Zhigang Mei
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, Medical College of China Three Gorges University, Yichang, Hubei 443002, China
| | - Xun Liu
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Jinwen Ge
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
- School of Medicine, Shaoyang University, Shaoyang, Hunan 422000, China
| |
Collapse
|
89
|
Yang X, Lu W, Hopper CP, Ke B, Wang B. Nature's marvels endowed in gaseous molecules I: Carbon monoxide and its physiological and therapeutic roles. Acta Pharm Sin B 2021; 11:1434-1445. [PMID: 34221861 PMCID: PMC8245769 DOI: 10.1016/j.apsb.2020.10.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 08/03/2020] [Accepted: 09/07/2020] [Indexed: 02/08/2023] Open
Abstract
Nature has endowed gaseous molecules such as O2, CO2, CO, NO, H2S, and N2 with critical and diverse roles in sustaining life, from supplying energy needed to power life and building blocks for life's physical structure to mediating and coordinating cellular functions. In this article, we give a brief introduction of the complex functions of the various gaseous molecules in life and then focus on carbon monoxide as a specific example of an endogenously produced signaling molecule to highlight the importance of this class of molecules. The past twenty years have seen much progress in understanding CO's mechanism(s) of action and pharmacological effects as well as in developing delivery methods for easy administration. One remarkable trait of CO is its pleiotropic effects that have few parallels, except perhaps its sister gaseous signaling molecules such as nitric oxide and hydrogen sulfide. This review will delve into the sophistication of CO-mediated signaling as well as its validated pharmacological functions and possible therapeutic applications.
Collapse
Affiliation(s)
- Xiaoxiao Yang
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303, USA
| | - Wen Lu
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303, USA
| | - Christopher P. Hopper
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303, USA
- Institut für Experimentelle Biomedizin, Universitätsklinikum Würzburg, Würzburg, Bavaria 97080, Germany
| | - Bowen Ke
- Department of Anesthesiology, West China Hospital, Chengdu 610041, China
| | - Binghe Wang
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303, USA
| |
Collapse
|
90
|
Abstract
Nitric oxide, studied to evaluate its role in cardiovascular physiology, has cardioprotective and therapeutic effects in cellular signaling, mitochondrial function, and in regulating inflammatory processes. Heme oxygenase (major role in catabolism of heme into biliverdin, carbon monoxide (CO), and iron) has similar effects as well. CO has been suggested as the molecule that is responsible for many of the above mentioned cytoprotective and therapeutic pathways as CO is a signaling molecule in the control of physiological functions. This is counterintuitive as toxic effects are related to its binding to hemoglobin. However, CO is normally produced in the body. Experimental evidence indicates that this toxic gas, CO, exerts cytoprotective properties related to cellular stress including the heart and is being assessed for its cytoprotective and cytotherapeutic properties. While survival of adult cardiomyocytes depends on oxidative phosphorylation (survival and resulting cardiac function is impaired by mitochondrial damage), mitochondrial biogenesis is modified by the heme oxygenase-1/CO system and can result in promotion of mitochondrial biogenesis by associating mitochondrial redox status to the redox-active transcription factors. It has been suggested that the heme oxygenase-1/CO system is important in differentiation of embryonic stem cells and maturation of cardiomyocytes which is thought to mitigate progression of degenerative cardiovascular diseases. Effects on other cardiac cells are being studied. Acute exposure to air pollution (and, therefore, CO) is associated with cardiovascular mortality, myocardial infarction, and heart failure, but changes in the endogenous heme oxygenase-1 system (and, thereby, CO) positively affect cardiovascular health. We will review the effect of CO on heart health and function in this article.
Collapse
Affiliation(s)
- Vicki L Mahan
- Department of Surgery and Pediatrics, Drexel University College of Medicine, Philadelphia, PA, USA
| |
Collapse
|
91
|
Nitti M, Ivaldo C, Traverso N, Furfaro AL. Clinical Significance of Heme Oxygenase 1 in Tumor Progression. Antioxidants (Basel) 2021; 10:antiox10050789. [PMID: 34067625 PMCID: PMC8155918 DOI: 10.3390/antiox10050789] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/30/2021] [Accepted: 05/10/2021] [Indexed: 02/07/2023] Open
Abstract
Heme oxygenase 1 (HO-1) plays a key role in cell adaptation to stressors through the antioxidant, antiapoptotic, and anti-inflammatory properties of its metabolic products. For these reasons, in cancer cells, HO-1 can favor aggressiveness and resistance to therapies, leading to poor prognosis/outcome. Genetic polymorphisms of HO-1 promoter have been associated with an increased risk of cancer progression and a high degree of therapy failure. Moreover, evidence from cancer biopsies highlights the possible correlation between HO-1 expression, pathological features, and clinical outcome. Indeed, high levels of HO-1 in tumor specimens often correlate with reduced survival rates. Furthermore, HO-1 modulation has been proposed in order to improve the efficacy of antitumor therapies. However, contrasting evidence on the role of HO-1 in tumor biology has been reported. This review focuses on the role of HO-1 as a promising biomarker of cancer progression; understanding the correlation between HO-1 and clinical data might guide the therapeutic choice and improve the outcome of patients in terms of prognosis and life quality.
Collapse
|
92
|
Ma Y, Jia L, Wang Y, Ji Y, Chen J, Ma H, Lin X, Zhang Y, Li W, Ni H, Xie L, Xie Y, Xiang M. Heme Oxygenase-1 in Macrophages Impairs the Perfusion Recovery After Hindlimb Ischemia by Suppressing Autolysosome-Dependent Degradation of NLRP3. Arterioscler Thromb Vasc Biol 2021; 41:1710-1723. [PMID: 33761761 DOI: 10.1161/atvbaha.121.315805] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Databases, Genetic
- Disease Models, Animal
- Heme Oxygenase-1/genetics
- Heme Oxygenase-1/metabolism
- Hindlimb
- Humans
- Inflammasomes/genetics
- Inflammasomes/metabolism
- Inflammation Mediators/metabolism
- Ischemia/enzymology
- Ischemia/genetics
- Ischemia/physiopathology
- Lysosomes/enzymology
- Macrophages/enzymology
- Male
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Skeletal/blood supply
- Muscle, Skeletal/enzymology
- Muscle, Skeletal/physiopathology
- NLR Family, Pyrin Domain-Containing 3 Protein/genetics
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- Neovascularization, Physiologic
- Proteolysis
- Recovery of Function
- Regional Blood Flow
- Mice
Collapse
Affiliation(s)
- Yuankun Ma
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Liangliang Jia
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yidong Wang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yongli Ji
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jian Chen
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hong Ma
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoping Lin
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuhao Zhang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wudi Li
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hui Ni
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lan Xie
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yao Xie
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Meixiang Xiang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
93
|
Szade A, Szade K, Mahdi M, Józkowicz A. The role of heme oxygenase-1 in hematopoietic system and its microenvironment. Cell Mol Life Sci 2021; 78:4639-4651. [PMID: 33787980 PMCID: PMC8195762 DOI: 10.1007/s00018-021-03803-z] [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: 10/13/2020] [Revised: 02/09/2021] [Accepted: 02/24/2021] [Indexed: 12/22/2022]
Abstract
Hematopoietic system transports all necessary nutrients to the whole organism and provides the immunological protection. Blood cells have high turnover, therefore, this system must be dynamically controlled and must have broad regeneration potential. In this review, we summarize how this complex system is regulated by the heme oxygenase-1 (HO-1)-an enzyme, which degrades heme to biliverdin, ferrous ion and carbon monoxide. First, we discuss how HO-1 influences hematopoietic stem cells (HSC) self-renewal, aging and differentiation. We also describe a critical role of HO-1 in endothelial cells and mesenchymal stromal cells that constitute the specialized bone marrow niche of HSC. We further discuss the molecular and cellular mechanisms by which HO-1 modulates innate and adaptive immune responses. Finally, we highlight how modulation of HO-1 activity regulates the mobilization of bone marrow hematopoietic cells to peripheral blood. We critically discuss the issue of metalloporphyrins, commonly used pharmacological modulators of HO-1 activity, and raise the issue of their important HO-1-independent activities.
Collapse
Affiliation(s)
- Agata Szade
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland.
| | - Krzysztof Szade
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland
| | - Mahdi Mahdi
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland
| | - Alicja Józkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland
| |
Collapse
|
94
|
Yao ME, Su MY, Huang Y, Chen W. Physiologically increased total bilirubin is associated with reduced risk of first myocardial infarction: A meta-analysis and dose-response analysis. Nutr Metab Cardiovasc Dis 2021; 31:1016-1026. [PMID: 33612380 DOI: 10.1016/j.numecd.2021.01.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 12/30/2020] [Accepted: 01/04/2021] [Indexed: 01/10/2023]
Abstract
AIM Bilirubin has potential predictive and prognostic value for myocardial infarction (MI), but the clinical evidence remains controversial. We performed this meta-analysis to systematically quantify the relationships between circulating bilirubin levels and the incidence of MI and post-MI adverse events. DATA SYNTHESIS We searched the PubMed, Cochrane Library, Embase, and Web of Science databases for ad-hoc studies, published up to October 17, 2020, recording bilirubin before MI (predictive analyses) or adverse events (prognostic analyses). Relative risks (RR) were pooled by a random-effects model. The dose-response analysis was conducted by restricted cubic splines. In patients without previous MI, increased total bilirubin (TB) reduced the risk of long-term (>3 year) first MI by 22% (95% confidence interval [CI]: 0.69-0.88, n = 4). The dose-response analysis indicated that the RR for first MI decreased by 2.7% per each 2 μmol/L increment of TB (three studies, 95% CI: 1.3%-4.1%, P < 0.001), with a cut-off value of 12.60 μmol/L for RR > 1.00. Elevated bilirubin reduced the incidence of first and recurrent MI by 36% (95% CI: 0.42-0.98, n = 7). However, after suffering MI, higher TB concentrations could not decrease the risk of recurrent MI (RR: 1.02, 95% CI: 0.67-1.55, n = 5) and increased the incidence of short-term (<1 year) post-MI major adverse cardiovascular events, all-cause mortality, and cardiovascular mortality, but not long-term (≥1 year). CONCLUSION Higher TB levels within a physiological range reduced the incidence of long-term first MI, with a cut-off value of 12.60 μmol/L.
Collapse
Affiliation(s)
- Miao-En Yao
- Longhua Hospital Shanghai University of Traditional Chinese Medicine, NO.725, South Wanping Road, Xuhui District, Shanghai, 200032, China; Shanghai University of Traditional Chinese Medicine, 1200 Cai Lun Road, Zhangjiang Hi-TechPark, Pudong New Area, Shanghai, 201203, China
| | - Mei-Yi Su
- Second Clinical College of Guangzhou University of Chinese Medicine, NO.232, Outer-ring East Road, Guangzhou Higher Education Mega Center, Panyu District, Guangzhou, 510006, China; Department of Cardiovascular Medicine, Second Affiliated Hospital of Guangzhou University of Chinese Medicine, NO.111, Dade Road, Yue-Xiu District, Guangzhou, 510120, China
| | - Yi Huang
- Longhua Hospital Shanghai University of Traditional Chinese Medicine, NO.725, South Wanping Road, Xuhui District, Shanghai, 200032, China; Shanghai University of Traditional Chinese Medicine, 1200 Cai Lun Road, Zhangjiang Hi-TechPark, Pudong New Area, Shanghai, 201203, China
| | - Wei Chen
- Department of Critical Care Medicine, Longhua Hospital Shanghai University of Traditional Chinese Medicine, NO.725, South Wanping Road, Xuhui District, Shanghai, 200032, China.
| |
Collapse
|
95
|
Protective Effect of Lactobacillus rhamnosus GG on TiO 2 Nanoparticles-Induced Oxidative Stress Damage in the Liver of Young Rats. NANOMATERIALS 2021; 11:nano11030803. [PMID: 33801059 PMCID: PMC8004042 DOI: 10.3390/nano11030803] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/13/2021] [Accepted: 03/18/2021] [Indexed: 12/17/2022]
Abstract
The potential toxicity of titanium dioxide nanoparticles (TiO2 NPs) to mammals has become a widespread concern. Young individuals exposed to TiO2 NPs have a higher risk than adults. In this study, the protective effects of Lactobacillus rhamnosus GG (LGG) on liver toxicity in young rats induced by TiO2 NPs were explored. Results show that the four-week-old rats that underwent LGG after the oral intake of TiO2 NPs could prevent weight loss, reduce hematological indicators (WBC and NEUT) and serum biochemical indicators (AST, ALT, AST/ALT, and ALP). Moreover, it alleviated the pathological damage of the liver (as indicated by the disordered hepatocytes, more eosinophilic, ballooning degeneration, and accompany with blood cells), but it did not reduce the Ti contents in the liver. In addition, RT-qPCR results indicated that LGG restored the expression of anti-oxidative stress-related genes, such as SOD1, SOD2, CAT, HO-1, GSH, GCLC, and GCLM in the liver. In summary, the hepatotoxicity of TiO2 NPs in young rats is closely related to oxidative stress, and the antioxidant effect of LGG might protect the harmful effects caused by TiO2 NPs.
Collapse
|
96
|
Li H, Liu B, Lian L, Zhou J, Xiang S, Zhai Y, Chen Y, Ma X, Wu W, Hou L. High dose expression of heme oxigenase-1 induces retinal degeneration through ER stress-related DDIT3. Mol Neurodegener 2021; 16:16. [PMID: 33691741 PMCID: PMC7944639 DOI: 10.1186/s13024-021-00437-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 03/01/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Oxidative stress is a common cause of neurodegeneration and plays a central role in retinal degenerative diseases. Heme oxygenase-1 (HMOX1) is a redox-regulated enzyme that is induced in neurodegenerative diseases and acts against oxidative stress but can also promote cell death, a phenomenon that is still unexplained in molecular terms. Here, we test whether HMOX1 has opposing effects during retinal degeneration and investigate the molecular mechanisms behind its pro-apoptotic role. METHODS Basal and induced levels of HMOX1 in retinas are examined during light-induced retinal degeneration in mice. Light damage-independent HMOX1 induction at two different expression levels is achieved by intraocular injection of different doses of an adeno-associated virus vector expressing HMOX1. Activation of Müller glial cells, retinal morphology and photoreceptor cell death are examined using hematoxylin-eosin staining, TUNEL assays, immunostaining and retinal function are evaluated with electroretinograms. Downstream gene expression of HMOX1 is analyzed by RNA-seq, qPCR examination and western blotting. The role of one of these genes, the pro-apoptotic DNA damage inducible transcript 3 (Ddit3), is analyzed in a line of knockout mice. RESULTS Light-induced retinal degeneration leads to photoreceptor degeneration and concomitant HMOX1 induction. HMOX1 expression at low levels before light exposure prevents photoreceptor degeneration but expression at high levels directly induces photoreceptor degeneration even without light stress. Photoreceptor degeneration following high level expression of HMOX1 is associated with a mislocalization of rhodopsin in photoreceptors and an increase in the expression of DDIT3. Genetic deletion of Ddit3 in knockout mice prevents photoreceptor cell degeneration normally resulting from high level HMOX1 expression. CONCLUSION The results reveal that the expression levels determine whether HMOX1 is protective or deleterious in the retina. Furthermore, in contrast to the protective low dose of HMOX1, the deleterious high dose is associated with induction of DDIT3 and endoplasmic reticulum stress as manifested, for instance, in rhodopsin mislocalization. Hence, future applications of HMOX1 or its regulated targets in gene therapy approaches should carefully consider expression levels in order to avoid potentially devastating effects.
Collapse
Affiliation(s)
- Huirong Li
- Laboratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, and State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Bo Liu
- Laboratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, and State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Lili Lian
- Laboratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, and State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Jiajia Zhou
- Laboratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, and State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Shengjin Xiang
- Eye Hospital of Wenzhou Medical University, Wenzhou, 325003 China
| | - Yifan Zhai
- Laboratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, and State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Yu Chen
- Laboratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, and State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Xiaoyin Ma
- Laboratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, and State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Wencan Wu
- Eye Hospital of Wenzhou Medical University, Wenzhou, 325003 China
| | - Ling Hou
- Laboratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, and State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
97
|
Li R, Yu L, Qin Y, Zhou Y, Liu W, Li Y, Chen Y, Xu Y. Protective effects of rare earth lanthanum on acute ethanol-induced oxidative stress in mice via Keap 1/Nrf2/p62 activation. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 758:143626. [PMID: 33243512 DOI: 10.1016/j.scitotenv.2020.143626] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 10/29/2020] [Accepted: 11/07/2020] [Indexed: 06/11/2023]
Abstract
With the widespread application of rare earth elements (REEs) in environment safety, food and medicine, they accumulate in the ecosystem and different human organs where REEs exert certain biological effects. Low dose REEs are proved to perform antioxidant effects, while high concentration can cause oxidative stress. However, scant information about rational doses and underlying mechanism of REEs as oxidants/antioxidants were illustrated. To elucidate these problems, here we performed a study that the ICR mice were received 0.1, 0.2, 1.0, 2.0 and 20.0 mg/kg lanthanum nitrate (La(NO3)3) by gavage for 30 days, and then were given 12 mL/kg ethanol once to undergo acute ethanol-induced oxidative stress. The antioxidant enzymes, antioxidants, peroxides and related proteins in Keap 1/Nrf2/p62 signaling pathway were measured. The results showed that La(NO3)3 inhibited hepatic morphological alternations by histopathological examination. Meanwhile, elevated superoxide dismutase (SOD) and glutathione (GSH), coupled with decreased alanine aminotransferase (ALT), aspartate aminotransferase (AST), malondialdehyde (MDA) and protein carbonyl (PC) were observed in serum and liver tissues of mice by enzyme-linked immunosorbent assay test. Furthermore, western blot analysis demonstrated that oxidative stress was alleviated due to enhanced NF-E2-related factor 2 (Nrf2) and phosphorylated p62 expressions as well as lower Kelch-like ECH-associated protein-1 (Keap 1), followed by the activation of heme oxygenase 1 (HO-1), NAD(P)H quinone oxidoreductase 1 (NQO-1) and glutamate cysteine ligase, catalytic (GCLC) proteins. Our findings clearly highlighted that La(NO3)3 could restore the redox homeostasis disrupted by ethanol through provoking Keap 1/Nrf2/p62 signaling pathway, and the optimal dosages were 1.0 and 2.0 mg/kg.
Collapse
Affiliation(s)
- Ruijun Li
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100083, China
| | - Lanlan Yu
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100083, China
| | - Yong Qin
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100083, China
| | - Yalin Zhou
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100083, China
| | - Wei Liu
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100083, China
| | - Yong Li
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100083, China
| | - Yuhan Chen
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100083, China
| | - Yajun Xu
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100083, China; Toxicological Research and Risk Assessment for Food Safety, Beijing 100083, China.
| |
Collapse
|
98
|
Martínez-Casales M, Hernanz R, Alonso MJ. Vascular and Macrophage Heme Oxygenase-1 in Hypertension: A Mini-Review. Front Physiol 2021; 12:643435. [PMID: 33716792 PMCID: PMC7952647 DOI: 10.3389/fphys.2021.643435] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 02/01/2021] [Indexed: 11/13/2022] Open
Abstract
Hypertension is one predictive factor for stroke and heart ischemic disease. Nowadays, it is considered an inflammatory disease with elevated cytokine levels, oxidative stress, and infiltration of immune cells in several organs including heart, kidney, and vessels, which contribute to the hypertension-associated cardiovascular damage. Macrophages, the most abundant immune cells in tissues, have a high degree of plasticity that is manifested by polarization in different phenotypes, with the most well-known being M1 (proinflammatory) and M2 (anti-inflammatory). In hypertension, M1 phenotype predominates, producing inflammatory cytokines and oxidative stress, and mediating many mechanisms involved in the pathogenesis of this disease. The increase in the renin-angiotensin system and sympathetic activity contributes to the macrophage mobilization and to its polarization to the pro-inflammatory phenotype. Heme oxygenase-1 (HO-1), a phase II detoxification enzyme responsible for heme catabolism, is induced by oxidative stress, among others. HO-1 has been shown to protect against oxidative and inflammatory insults in hypertension, reducing end organ damage and blood pressure, not only by its expression at the vascular level, but also by shifting macrophages toward the anti-inflammatory phenotype. The regulatory role of heme availability for the synthesis of enzymes involved in hypertension development, such as cyclooxygenase or nitric oxide synthase, seems to be responsible for many of the beneficial HO-1 effects; additionally, the antioxidant, anti-inflammatory, antiapoptotic, and antiproliferative effects of the end products of its reaction, carbon monoxide, biliverdin/bilirubin, and Fe2+, would also contribute. In this review, we analyze the role of HO-1 in hypertensive pathology, focusing on its expression in macrophages.
Collapse
Affiliation(s)
- Marta Martínez-Casales
- Depto. de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Alcorcón, Spain
| | - Raquel Hernanz
- Depto. de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Alcorcón, Spain.,Centro de Investigación en Red en Enfermedades Cardiovasculares (CIBER-CV), Madrid, Spain
| | - María J Alonso
- Depto. de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Alcorcón, Spain.,Centro de Investigación en Red en Enfermedades Cardiovasculares (CIBER-CV), Madrid, Spain
| |
Collapse
|
99
|
Szabó R, Hoffmann A, Börzsei D, Kupai K, Veszelka M, Berkó AM, Pávó I, Gesztelyi R, Juhász B, Turcsán Z, Pósa A, Varga C. Hormone Replacement Therapy and Aging: A Potential Therapeutic Approach for Age-Related Oxidative Stress and Cardiac Remodeling. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8364297. [PMID: 33623635 PMCID: PMC7875635 DOI: 10.1155/2021/8364297] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 08/23/2020] [Accepted: 01/10/2021] [Indexed: 12/13/2022]
Abstract
Advanced age is an independent risk factor for cardiovascular diseases, which might be further exacerbated by estrogen deficiency. Hormone replacement therapy (HRT) decreases cardiovascular risks and events in postmenopausal women; however, its effects are not fully elucidated in older individuals. Thus, the aim of our study is to examine the impact of HRT on oxidant/antioxidant homeostasis and cardiac remodeling. In our experiment, control (fertile) and aging (~20-month-old) female Wistar rats were used. Aging rats were further divided into estrogen- (E2, 0.1 mg/kg/day per os) or raloxifene- (RAL, 1.0 mg/kg/day per os) treated subgroups. After 2 weeks of treatment, cardiac heme oxygenase (HO) activity, total glutathione (GSH) content, matrix metalloproteinase-2 (MMP-2) activity, and the concentrations of collagen type I and tissue inhibitor of metalloproteinase (TIMP-2), as well as the infarct size, were determined. The aging process significantly decreased the antioxidant HO activity and GSH content, altered the MMP-2/TIMP-2 signaling, and resulted in an excessive collagen accumulation, which culminated in cardiovascular injury. However, 2 weeks of either E2 or RAL treatment enhanced the antioxidant defense mechanisms and attenuated cardiac remodeling related to aging. Our findings clearly show that 2-week-long HRT is a potential intervention to bias successful cardiovascular aging via reducing oxidative damage and cardiovascular dysfunction.
Collapse
Affiliation(s)
- Renáta Szabó
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged H-6726, Hungary
- Interdisciplinary Excellence Centre, Department of Physiology, Anatomy and Neuroscience, University of Szeged, Szeged, Hungary
| | - Alexandra Hoffmann
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged H-6726, Hungary
| | - Denise Börzsei
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged H-6726, Hungary
| | - Krisztina Kupai
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged H-6726, Hungary
- 1st Department of Medicine, University of Szeged, Szeged H-6720, Hungary
| | - Médea Veszelka
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged H-6726, Hungary
| | - Anikó Magyariné Berkó
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged H-6726, Hungary
| | - Imre Pávó
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged H-6726, Hungary
| | - Rudolf Gesztelyi
- Department of Pharmacology and Pharmacotherapy, University of Debrecen, Debrecen H-4032, Hungary
| | - Béla Juhász
- Department of Pharmacology and Pharmacotherapy, University of Debrecen, Debrecen H-4032, Hungary
| | - Zsolt Turcsán
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged H-6726, Hungary
| | - Anikó Pósa
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged H-6726, Hungary
- Interdisciplinary Excellence Centre, Department of Physiology, Anatomy and Neuroscience, University of Szeged, Szeged, Hungary
| | - Csaba Varga
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged H-6726, Hungary
| |
Collapse
|
100
|
Li Y, Dong Y, Meng L, Yu P, Zhao P, Gong M, Gao Q, Shi H, Meng C, Gao Y. Effects of Exogenous Biliverdin Treatment on Neurobehaviors in Mice. Biol Pharm Bull 2021; 44:325-331. [PMID: 33642542 DOI: 10.1248/bpb.b20-00340] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The neuroprotective effects of heme oxygenase (HO) have been well investigated. The potential effects of exogenous supplementation of biliverdin (BVD), one of the main products catalyzed by HO, on neurobehaviors are still largely unknown. The present study aimed to investigate the effects of BVD treatment on depression, anxiety, and memory in adult mice. Mice were injected with BVD through tail vein daily for a total 5 d, and depression- and anxiety-like behaviors were conducted by using open field test (OFT), novelty suppressed feeding (NSF), forced swimming test (FST) and tail suspension test (TST) since the third day of BVD administration. Novel object recognition (NOR) paradigm was used for memory formation test. After the final test, serum and hippocampal levels of interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) of mice were analyzed by enzyme-linked immunosorbent assay (ELISA). The results showed that BVD treatment at low dose (2 mg/kg) induced depression-like behaviors, and high dose (8 mg/kg) BVD injection increased anxiety-like behaviors and impaired memory formation in mice. ELISA data showed that BVD treatment significantly increased hippocampal IL-6 and TNF-α level while only decreasing serum IL-6 level of mice. The present data suggest that exogenous BVD treatment induced depression- and anxiety-like phenotypes, which may be related to inflammatory factors, providing BVD may be a potential target for the prevention of mental disorders.
Collapse
Affiliation(s)
- Yueyi Li
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University
| | - Yan Dong
- Intensive Care Unit of Hebei Hospital of Traditional Chinese Medicine, Hebei University of Chinese Medicine
| | - Li Meng
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University
| | - Panpan Yu
- Department of State Assets and Laboratory Administrative, Hebei Medical University
| | - Penghui Zhao
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University
| | - Miao Gong
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University
| | - Qiang Gao
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University
| | - Haishui Shi
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medicinal University
- Hebei Key Laboratory of Neurophysiology, Hebei Medicinal University
| | - Cuili Meng
- Department of Biochemistry, School of Basic Medicine, Xingtai Medical College
| | - Yuan Gao
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medicinal University
- Hebei Key Laboratory of Neurophysiology, Hebei Medicinal University
| |
Collapse
|