1
|
McLeod K, Datta V, Fuller S. Adipokines as Cardioprotective Factors: BAT Steps Up to the Plate. Biomedicines 2025; 13:710. [PMID: 40149686 PMCID: PMC11940801 DOI: 10.3390/biomedicines13030710] [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: 01/10/2025] [Revised: 03/07/2025] [Accepted: 03/11/2025] [Indexed: 03/29/2025] Open
Abstract
Cardiovascular disease is the leading cause of death throughout most of the industrialized world. Metabolic syndrome (MetS) and its associated pathologies are underlying factors in the etiology of cardiovascular disease, as well as a plethora of other maladies which cause excess morbidity and mortality. Adipose tissue (AT) has come to be regarded as a bona fide endocrine organ which secretes specific molecular entities constituting part of a complex web of inter-organ crosstalk that functions as a key determinant of whole-body metabolic phenotype. Brown adipose tissue (BAT) has classically been regarded as a thermogenic tissue exerting its metabolic effects primarily through its capacity to oxidize substrates decoupled from ATP resynthesis, thereby resulting in increased energy expenditure (EE) and heat production. However, in recent years, BAT has begun to receive attention as a secretory organ in its own right. The molecules secreted specifically by BAT have been termed "batokines", and currently available evidence supports the notion that batokines exert favorable metabolic effects on multiple organ systems. While maintenance of healthy body composition by conferring resistance to excessive adiposity is a rather obvious mechanism by which BAT operates via increased EE, effects on critical organs such as the heart remain unclear. This narrative review focuses on four types of batokines (FGF21, neuregulin 4, 12,13-diHOME, and BAT-derived microRNAs) for which evidence of modulation of cardiovascular function exists in the context of pathological states such as hypertension, atherosclerosis, and ischemia/reperfusion injury. Given the overwhelming burden of cardiometabolic disease, further study of the functions of BAT and its secretome is warranted and will intensify in the future.
Collapse
Affiliation(s)
- Keely McLeod
- School of Kinesiology, University of Louisiana at Lafayette, Lafayette, LA 70506, USA; (K.M.); (V.D.)
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA
| | - Victoria Datta
- School of Kinesiology, University of Louisiana at Lafayette, Lafayette, LA 70506, USA; (K.M.); (V.D.)
| | - Scott Fuller
- School of Kinesiology, University of Louisiana at Lafayette, Lafayette, LA 70506, USA; (K.M.); (V.D.)
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA
| |
Collapse
|
2
|
Revol-Cavalier J, Quaranta A, Newman JW, Brash AR, Hamberg M, Wheelock CE. The Octadecanoids: Synthesis and Bioactivity of 18-Carbon Oxygenated Fatty Acids in Mammals, Bacteria, and Fungi. Chem Rev 2025; 125:1-90. [PMID: 39680864 PMCID: PMC11719350 DOI: 10.1021/acs.chemrev.3c00520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 11/06/2024] [Accepted: 11/15/2024] [Indexed: 12/18/2024]
Abstract
The octadecanoids are a broad class of lipids consisting of the oxygenated products of 18-carbon fatty acids. Originally referring to production of the phytohormone jasmonic acid, the octadecanoid pathway has been expanded to include products of all 18-carbon fatty acids. Octadecanoids are formed biosynthetically in mammals via cyclooxygenase (COX), lipoxygenase (LOX), and cytochrome P450 (CYP) activity, as well as nonenzymatically by photo- and autoxidation mechanisms. While octadecanoids are well-known mediators in plants, their role in the regulation of mammalian biological processes has been generally neglected. However, there have been significant advancements in recognizing the importance of these compounds in mammals and their involvement in the mediation of inflammation, nociception, and cell proliferation, as well as in immuno- and tissue modulation, coagulation processes, hormone regulation, and skin barrier formation. More recently, the gut microbiome has been shown to be a significant source of octadecanoid biosynthesis, providing additional biosynthetic routes including hydratase activity (e.g., CLA-HY, FA-HY1, FA-HY2). In this review, we summarize the current field of octadecanoids, propose standardized nomenclature, provide details of octadecanoid preparation and measurement, summarize the phase-I metabolic pathway of octadecanoid formation in mammals, bacteria, and fungi, and describe their biological activity in relation to mammalian pathophysiology as well as their potential use as biomarkers of health and disease.
Collapse
Affiliation(s)
- Johanna Revol-Cavalier
- Unit
of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm SE-171 77, Sweden
- Larodan
Research Laboratory, Karolinska Institutet, Stockholm SE-171 77, Sweden
| | - Alessandro Quaranta
- Unit
of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm SE-171 77, Sweden
| | - John W. Newman
- Western
Human Nutrition Research Center, Agricultural
Research Service, USDA, Davis, California 95616, United States
- Department
of Nutrition, University of California, Davis, Davis, California 95616, United States
- West
Coast Metabolomics Center, Genome Center, University of California, Davis, Davis, California 95616, United States
| | - Alan R. Brash
- Department
of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Mats Hamberg
- Unit
of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm SE-171 77, Sweden
- Larodan
Research Laboratory, Karolinska Institutet, Stockholm SE-171 77, Sweden
| | - Craig E. Wheelock
- Unit
of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm SE-171 77, Sweden
- Department
of Respiratory Medicine and Allergy, Karolinska
University Hospital, Stockholm SE-141-86, Sweden
| |
Collapse
|
3
|
Matsumura S, Shida A, Tsuchii M, Wada M, Charneau J, Tsuji M, Hasumi K, Suzuki E. Intradomain Allosteric Regulation of Soluble Epoxide Hydrolase by Its Substrates. Int J Mol Sci 2024; 25:13496. [PMID: 39769258 PMCID: PMC11676941 DOI: 10.3390/ijms252413496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 12/13/2024] [Accepted: 12/15/2024] [Indexed: 01/11/2025] Open
Abstract
Soluble epoxide hydrolase (sEH) is a bifunctional enzyme with epoxide hydrolase activity in the C-terminal domain (C-EH) and lipid phosphate phosphatase activity in the N-terminal domain (N-phos). The C-EH hydrolyzes bioactive epoxy fatty acids such as epoxyeicosatrienoic acid (EET). The N-phos hydrolyzes lipid phosphomonesters, including the signaling molecules of lysophosphatidic acid (LPA). Here, we report that the C-EH and N-phos are reciprocally regulated by their respective substrates. Full-length sEH (sEH-FL) showed positive cooperativity toward the substrate for each domain. Similar cooperativity was found when truncated enzymes having only C- and N-terminal domains, sEH-C and sEH-N, respectively, were used, suggesting an intra-domain nature of the cooperativity. In addition, the N-phos substrate LPA inhibited C-EH activity in sEH-FL and sEH-C equally. Similarly, the C-EH substrate EET inhibited N-phos activity. Structural and kinetic data suggest the presence of allosteric sites in each domain of the sEH enzyme, which share the binding of LPA and EET. Thus, each of the two sEH activities is regulated by a substrate of its own and by that of the other domain. This mechanism may explain why sEH has evolved to have two different enzyme activities, which possibly allows sEH to balance the metabolism of bioactive lipids.
Collapse
Affiliation(s)
- Shin Matsumura
- Department of Applied Biological Science, Tokyo University of Agriculture and Technology, Fuchu 183-8509, Japan; (S.M.); (A.S.); (M.T.); (K.H.)
| | - Ayano Shida
- Department of Applied Biological Science, Tokyo University of Agriculture and Technology, Fuchu 183-8509, Japan; (S.M.); (A.S.); (M.T.); (K.H.)
| | - Moeno Tsuchii
- Department of Applied Biological Science, Tokyo University of Agriculture and Technology, Fuchu 183-8509, Japan; (S.M.); (A.S.); (M.T.); (K.H.)
| | - Mika Wada
- Department of Research and Development, TMS Co., Fuchu 183-0055, Japan; (M.W.); (J.C.)
| | - Jimmy Charneau
- Department of Research and Development, TMS Co., Fuchu 183-0055, Japan; (M.W.); (J.C.)
| | - Motonori Tsuji
- Institute of Molecular Function, Saitama 341-0037, Japan;
| | - Keiji Hasumi
- Department of Applied Biological Science, Tokyo University of Agriculture and Technology, Fuchu 183-8509, Japan; (S.M.); (A.S.); (M.T.); (K.H.)
- Department of Research and Development, TMS Co., Fuchu 183-0055, Japan; (M.W.); (J.C.)
| | - Eriko Suzuki
- Department of Applied Biological Science, Tokyo University of Agriculture and Technology, Fuchu 183-8509, Japan; (S.M.); (A.S.); (M.T.); (K.H.)
| |
Collapse
|
4
|
Yousef A, Fang L, Heidari M, Kranrod J, Seubert JM. The role of CYP-sEH derived lipid mediators in regulating mitochondrial biology and cellular senescence: implications for the aging heart. Front Pharmacol 2024; 15:1486717. [PMID: 39703395 PMCID: PMC11655241 DOI: 10.3389/fphar.2024.1486717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 10/28/2024] [Indexed: 12/21/2024] Open
Abstract
Cellular senescence is a condition characterized by stable, irreversible cell cycle arrest linked to the aging process. The accumulation of senescent cells in the cardiac muscle can contribute to various cardiovascular diseases (CVD). Telomere shortening, epigenetic modifications, DNA damage, mitochondrial dysfunction, and oxidative stress are known contributors to the onset of cellular senescence in the heart. The link between mitochondrial processes and cellular senescence contributed to the age-related decline in cardiac function. These include changes in mitochondrial functions and behaviours that arise from various factors, including impaired dynamics, dysregulated biogenesis, mitophagy, mitochondrial DNA (mtDNA), reduced respiratory capacity, and mitochondrial structural changes. Thus, regulation of mitochondrial biology has a role in cellular senescence and cardiac function in aging hearts. Targeting senescent cells may provide a novel therapeutic approach for treating and preventing CVD associated with aging. CYP epoxygenases metabolize N-3 and N-6 polyunsaturated fatty acids (PUFA) into epoxylipids that are readily hydrolyzed to diol products by soluble epoxide hydrolase (sEH). Increasing epoxylipids levels or inhibition of sEH has demonstrated protective effects in the aging heart. Evidence suggests they may play a role in cellular senescence by regulating mitochondria, thus reducing adverse effects of aging in the heart. In this review, we discuss how mitochondria induce cellular senescence and how epoxylipids affect the senescence process in the aged heart.
Collapse
Affiliation(s)
- Ala Yousef
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Liye Fang
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Mobina Heidari
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Joshua Kranrod
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - John M. Seubert
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
5
|
Ağagündüz D, Yeşildemir Ö, Koçyiğit E, Koçak T, Özen Ünaldı B, Ayakdaş G, Budán F. Oxylipins Derived from PUFAs in Cardiometabolic Diseases: Mechanism of Actions and Possible Nutritional Interactions. Nutrients 2024; 16:3812. [PMID: 39599599 PMCID: PMC11597274 DOI: 10.3390/nu16223812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 10/31/2024] [Accepted: 11/05/2024] [Indexed: 11/29/2024] Open
Abstract
Oxylipins are oxidized fatty acids, both saturated and unsaturated, formed through pathways that involve singlet oxygen or dioxygen-mediated oxygenation reactions and are primarily produced by enzyme families such as cyclooxygenases, lipoxygenases, and cytochrome P450. These lipid-based complex bioactive molecules are pivotal signal mediators, acting in a hormone-like manner in the pathophysiology of numerous diseases, especially cardiometabolic diseases via modulating plenty of mechanisms. It has been reported that omega-6 and omega-3 oxylipins are important novel biomarkers of cardiometabolic diseases. Moreover, collected literature has noted that diet and dietary components, especially fatty acids, can modulate these oxygenated lipid products since they are mainly derived from dietary omega-3 and omega-6 polyunsaturated fatty acids (PUFAs) or linoleic acid and α-linolenic by elongation and desaturation pathways. This comprehensive review aims to examine their correlations to cardiometabolic diseases and how diets modulate oxylipins. Also, some aspects of developing new biomarkers and therapeutical utilization are detailed in this review.
Collapse
Affiliation(s)
- Duygu Ağagündüz
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Gazi University, 06490 Ankara, Türkiye
| | - Özge Yeşildemir
- Department of Nutrition and Dietetics, Bursa Uludag University, Görükle Campus, 16059 Bursa, Türkiye;
| | - Emine Koçyiğit
- Department of Nutrition and Dietetics, Ordu University, Cumhuriyet Yerleşkesi, 52200 Ordu, Türkiye;
| | - Tevfik Koçak
- Department of Nutrition and Dietetics, Gümüşhane University, Gümüşhanevî Kampüsü, 29100 Gümüşhane, Türkiye;
| | - Buket Özen Ünaldı
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Afyonkarahisar Health Sciences University, 03030 Afyonkarahisar, Türkiye;
| | - Gamze Ayakdaş
- Department of Nutrition and Dietetics, Acıbadem University, Kerem Aydınlar Campus, 34752 İstanbul, Türkiye;
| | - Ferenc Budán
- Institute of Physiology, Medical School, University of Pécs, H-7624 Pécs, Hungary
| |
Collapse
|
6
|
Almestica-Roberts M, Nguyen ND, Sun L, Serna SN, Rapp E, Burrell-Gerbers KL, Memon TA, Stone BL, Nkoy FL, Lamb JG, Deering-Rice CE, Rower JE, Reilly CA. The Cytochrome P450 2C8*3 Variant (rs11572080) Is Associated with Improved Asthma Symptom Control in Children and Altered Lipid Mediator Production and Inflammatory Response in Human Bronchial Epithelial Cells. Drug Metab Dispos 2024; 52:836-846. [PMID: 38772712 PMCID: PMC11257687 DOI: 10.1124/dmd.124.001684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 04/16/2024] [Accepted: 05/14/2024] [Indexed: 05/23/2024] Open
Abstract
This study investigated an association between the cytochrome P450 (CYP) 2C8*3 polymorphism with asthma symptom control in children and changes in lipid metabolism and pro-inflammatory signaling by human bronchial epithelial cells (HBECs) treated with cigarette smoke condensate (CSC). CYP genes are inherently variable in sequence, and while such variations are known to produce clinically relevant effects on drug pharmacokinetics and pharmacodynamics, the effects on endogenous substrate metabolism and associated physiologic processes are less understood. In this study, CYP2C8*3 was associated with improved asthma symptom control among children: Mean asthma control scores were 3.68 (n = 207) for patients with one or more copies of the CYP2C8*3 allele versus 4.42 (n = 965) for CYP2C8*1/*1 (P = 0.0133). In vitro, CYP2C8*3 was associated with an increase in montelukast 36-hydroxylation and a decrease in linoleic acid metabolism despite lower mRNA and protein expression. Additionally, CYP2C8*3 was associated with reduced mRNA expression of interleukin-6 (IL-6) and C-X-C motif chemokine ligand 8 (CXCL-8) by HBECs in response to CSC, which was replicated using the soluble epoxide hydrolase inhibitor, 12-[[(tricyclo[3.3.1.13,7]dec-1-ylamino)carbonyl]amino]-dodecanoic acid. Interestingly, 9(10)- and 12(13)- dihydroxyoctadecenoic acid, the hydrolyzed metabolites of 9(10)- and 12(13)- epoxyoctadecenoic acid, increased the expression of IL-6 and CXCL-8 mRNA by HBECs. This study reveals previously undocumented effects of the CYP2C8*3 variant on the response of HBECs to exogenous stimuli. SIGNIFICANCE STATEMENT: These findings suggest a role for CYP2C8 in regulating the epoxyoctadecenoic acid:dihydroxyoctadecenoic acid ratio leading to a change in cellular inflammatory responses elicited by environmental stimuli that exacerbate asthma.
Collapse
Affiliation(s)
- Marysol Almestica-Roberts
- Department of Pharmacology and Toxicology, Center for Human Toxicology (M.A.-R., N.D.N., L.S., S.N.S., E.R., K.L.B.-G., T.A.M., J.G.L., C.E.D.-R., J.E.R., C.A.R.) and Department of Pediatrics, School of Medicine (B.L.S., F.L.N.), University of Utah, Salt Lake City, Utah
| | - Nam D Nguyen
- Department of Pharmacology and Toxicology, Center for Human Toxicology (M.A.-R., N.D.N., L.S., S.N.S., E.R., K.L.B.-G., T.A.M., J.G.L., C.E.D.-R., J.E.R., C.A.R.) and Department of Pediatrics, School of Medicine (B.L.S., F.L.N.), University of Utah, Salt Lake City, Utah
| | - Lili Sun
- Department of Pharmacology and Toxicology, Center for Human Toxicology (M.A.-R., N.D.N., L.S., S.N.S., E.R., K.L.B.-G., T.A.M., J.G.L., C.E.D.-R., J.E.R., C.A.R.) and Department of Pediatrics, School of Medicine (B.L.S., F.L.N.), University of Utah, Salt Lake City, Utah
| | - Samantha N Serna
- Department of Pharmacology and Toxicology, Center for Human Toxicology (M.A.-R., N.D.N., L.S., S.N.S., E.R., K.L.B.-G., T.A.M., J.G.L., C.E.D.-R., J.E.R., C.A.R.) and Department of Pediatrics, School of Medicine (B.L.S., F.L.N.), University of Utah, Salt Lake City, Utah
| | - Emmanuel Rapp
- Department of Pharmacology and Toxicology, Center for Human Toxicology (M.A.-R., N.D.N., L.S., S.N.S., E.R., K.L.B.-G., T.A.M., J.G.L., C.E.D.-R., J.E.R., C.A.R.) and Department of Pediatrics, School of Medicine (B.L.S., F.L.N.), University of Utah, Salt Lake City, Utah
| | - Katherine L Burrell-Gerbers
- Department of Pharmacology and Toxicology, Center for Human Toxicology (M.A.-R., N.D.N., L.S., S.N.S., E.R., K.L.B.-G., T.A.M., J.G.L., C.E.D.-R., J.E.R., C.A.R.) and Department of Pediatrics, School of Medicine (B.L.S., F.L.N.), University of Utah, Salt Lake City, Utah
| | - Tosifa A Memon
- Department of Pharmacology and Toxicology, Center for Human Toxicology (M.A.-R., N.D.N., L.S., S.N.S., E.R., K.L.B.-G., T.A.M., J.G.L., C.E.D.-R., J.E.R., C.A.R.) and Department of Pediatrics, School of Medicine (B.L.S., F.L.N.), University of Utah, Salt Lake City, Utah
| | - Bryan L Stone
- Department of Pharmacology and Toxicology, Center for Human Toxicology (M.A.-R., N.D.N., L.S., S.N.S., E.R., K.L.B.-G., T.A.M., J.G.L., C.E.D.-R., J.E.R., C.A.R.) and Department of Pediatrics, School of Medicine (B.L.S., F.L.N.), University of Utah, Salt Lake City, Utah
| | - Flory L Nkoy
- Department of Pharmacology and Toxicology, Center for Human Toxicology (M.A.-R., N.D.N., L.S., S.N.S., E.R., K.L.B.-G., T.A.M., J.G.L., C.E.D.-R., J.E.R., C.A.R.) and Department of Pediatrics, School of Medicine (B.L.S., F.L.N.), University of Utah, Salt Lake City, Utah
| | - John G Lamb
- Department of Pharmacology and Toxicology, Center for Human Toxicology (M.A.-R., N.D.N., L.S., S.N.S., E.R., K.L.B.-G., T.A.M., J.G.L., C.E.D.-R., J.E.R., C.A.R.) and Department of Pediatrics, School of Medicine (B.L.S., F.L.N.), University of Utah, Salt Lake City, Utah
| | - Cassandra E Deering-Rice
- Department of Pharmacology and Toxicology, Center for Human Toxicology (M.A.-R., N.D.N., L.S., S.N.S., E.R., K.L.B.-G., T.A.M., J.G.L., C.E.D.-R., J.E.R., C.A.R.) and Department of Pediatrics, School of Medicine (B.L.S., F.L.N.), University of Utah, Salt Lake City, Utah
| | - Joseph E Rower
- Department of Pharmacology and Toxicology, Center for Human Toxicology (M.A.-R., N.D.N., L.S., S.N.S., E.R., K.L.B.-G., T.A.M., J.G.L., C.E.D.-R., J.E.R., C.A.R.) and Department of Pediatrics, School of Medicine (B.L.S., F.L.N.), University of Utah, Salt Lake City, Utah
| | - Christopher A Reilly
- Department of Pharmacology and Toxicology, Center for Human Toxicology (M.A.-R., N.D.N., L.S., S.N.S., E.R., K.L.B.-G., T.A.M., J.G.L., C.E.D.-R., J.E.R., C.A.R.) and Department of Pediatrics, School of Medicine (B.L.S., F.L.N.), University of Utah, Salt Lake City, Utah
| |
Collapse
|
7
|
Valencia R, Kranrod JW, Fang L, Soliman AM, Azer B, Clemente-Casares X, Seubert JM. Linoleic acid-derived diol 12,13-DiHOME enhances NLRP3 inflammasome activation in macrophages. FASEB J 2024; 38:e23748. [PMID: 38940767 DOI: 10.1096/fj.202301640rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 05/30/2024] [Accepted: 06/05/2024] [Indexed: 06/29/2024]
Abstract
12,13-dihydroxy-9z-octadecenoic acid (12,13-DiHOME) is a linoleic acid diol derived from cytochrome P-450 (CYP) epoxygenase and epoxide hydrolase (EH) metabolism. 12,13-DiHOME is associated with inflammation and mitochondrial damage in the innate immune response, but how 12,13-DiHOME contributes to these effects is unclear. We hypothesized that 12,13-DiHOME enhances macrophage inflammation through effects on NOD-like receptor protein 3 (NLRP3) inflammasome activation. To test this hypothesis, we utilized human monocytic THP1 cells differentiated into macrophage-like cells with phorbol myristate acetate (PMA). 12,13-DiHOME present during lipopolysaccharide (LPS)-priming of THP1 macrophages exacerbated nigericin-induced NLRP3 inflammasome activation. Using high-resolution respirometry, we observed that priming with LPS+12,13-DiHOME altered mitochondrial respiratory function. Mitophagy, measured using mito-Keima, was also modulated by 12,13-DiHOME present during priming. These mitochondrial effects were associated with increased sensitivity to nigericin-induced mitochondrial depolarization and reactive oxygen species production in LPS+12,13-DiHOME-primed macrophages. Nigericin-induced mitochondrial damage and NLRP3 inflammasome activation in LPS+12,13-DiHOME-primed macrophages were ablated by the mitochondrial calcium uniporter (MCU) inhibitor, Ru265. 12,13-DiHOME present during LPS-priming also enhanced nigericin-induced NLRP3 inflammasome activation in primary murine bone marrow-derived macrophages. In summary, these data demonstrate a pro-inflammatory role for 12,13-DiHOME by enhancing NLRP3 inflammasome activation in macrophages.
Collapse
Affiliation(s)
- Robert Valencia
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Cardiovascular Research Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Joshua W Kranrod
- Cardiovascular Research Institute, University of Alberta, Edmonton, Alberta, Canada
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Liye Fang
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Cardiovascular Research Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Amro M Soliman
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Brandon Azer
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Xavier Clemente-Casares
- Cardiovascular Research Institute, University of Alberta, Edmonton, Alberta, Canada
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, Alberta, Canada
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
| | - John M Seubert
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Cardiovascular Research Institute, University of Alberta, Edmonton, Alberta, Canada
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
8
|
Yousef A, Sosnowski DK, Fang L, Legaspi RJ, Korodimas J, Lee A, Magor KE, Seubert JM. Cardioprotective response and senescence in aged sEH null female mice exposed to LPS. Am J Physiol Heart Circ Physiol 2024; 326:H1366-H1385. [PMID: 38578240 DOI: 10.1152/ajpheart.00706.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 03/20/2024] [Accepted: 03/27/2024] [Indexed: 04/06/2024]
Abstract
Deterioration of physiological systems, like the cardiovascular system, occurs progressively with age impacting an individual's health and increasing susceptibility to injury and disease. Cellular senescence has an underlying role in age-related alterations and can be triggered by natural aging or prematurely by stressors such as the bacterial toxin lipopolysaccharide (LPS). The metabolism of polyunsaturated fatty acids by CYP450 enzymes produces numerous bioactive lipid mediators that can be further metabolized by soluble epoxide hydrolase (sEH) into diol metabolites, often with reduced biological effects. In our study, we observed age-related cardiac differences in female mice, where young mice demonstrated resistance to LPS injury, and genetic deletion or pharmacological inhibition of sEH using trans-4-[4-(3-adamantan-1-yl-ureido)-cyclohexyloxy]-benzoic acid attenuated LPS-induced cardiac dysfunction in aged female mice. Bulk RNA-sequencing analyses revealed transcriptomics differences in aged female hearts. The confirmatory analysis demonstrated changes to inflammatory and senescence gene markers such as Il-6, Mcp1, Il-1β, Nlrp3, p21, p16, SA-β-gal, and Gdf15 were attenuated in the hearts of aged female mice where sEH was deleted or inhibited. Collectively, these findings highlight the role of sEH in modulating the aging process of the heart, whereby targeting sEH is cardioprotective.NEW & NOTEWORTHY Soluble epoxide hydrolase (sEH) is an essential enzyme for converting epoxy fatty acids to their less bioactive diols. Our study suggests deletion or inhibition of sEH impacts the aging process in the hearts of female mice resulting in cardioprotection. Data indicate targeting sEH limits inflammation, preserves mitochondria, and alters cellular senescence in the aged female heart.
Collapse
Affiliation(s)
- Ala Yousef
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Deanna K Sosnowski
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Liye Fang
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Renald James Legaspi
- Department of Biological Sciences, Faculty of Science, University of Alberta, Edmonton, Alberta, Canada
| | - Jacob Korodimas
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Andy Lee
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Katharine E Magor
- Department of Biological Sciences, Faculty of Science, University of Alberta, Edmonton, Alberta, Canada
| | - John M Seubert
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
9
|
Solati Z, Surendran A, Aukema HM, Ravandi A. Impact of Reperfusion on Plasma Oxylipins in ST-Segment Elevation Myocardial Infarction. Metabolites 2023; 14:19. [PMID: 38248822 PMCID: PMC10821107 DOI: 10.3390/metabo14010019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/21/2023] [Accepted: 12/22/2023] [Indexed: 01/23/2024] Open
Abstract
ST-segment elevation myocardial infarction (STEMI) occurs as a result of acute occlusion of the coronary artery. Despite successful reperfusion using primary percutaneous coronary intervention (PPCI), a large percentage of myocardial cells die after reperfusion, which is recognized as ischemia/reperfusion injury (I/R). There are rapid changes in plasma lipidome during myocardial reperfusion injury. However, the impact of coronary artery reperfusion on plasma oxylipins is unknown. This study aimed to investigate alterations in the oxylipin profiles of STEMI patients during ischemia and at various reperfusion time points following PPCI. Blood samples were collected from patients presenting with STEMI prior to PPCI (Isch, n = 45) and subsequently 2 h following successful reperfusion by PPCI (R-2 h, n = 42), after 24 h (R-24 h, n = 44), after 48 h (R-48 h, n = 43), and then 30 days post PPCI (R-30 d, n = 29). As controls, blood samples were collected from age- and sex-matched patients with non-obstructive coronary artery disease after diagnostic coronary angiography. High-performance liquid chromatography-mass spectrometry (HPLC-MS/MS) using deuterated standards was used to identify and quantify oxylipins. In patients presenting with STEMI prior to reperfusion (Isch group), the levels of docosahexaenoic acid (DHA)-derived oxylipins were significantly higher when compared with controls. Their levels were also significantly correlated with the peak levels of creatine kinase (CK) and troponin T(TnT) before reperfusion (CK: r = 0.33, p = 0.046, TnT: r = 0.50, p = 1.00 × 10-3). The total concentrations of oxylipins directly produced by 5-lipoxygenase (5-LOX) were also significantly elevated in the Isch group compared with controls. The ratio of epoxides (generated through epoxygenase) to diols (generated by soluble epoxide hydrolysis (sEH)) was significantly lower in the Isch group compared with the controls. Following reperfusion, there was an overall reduction in plasma oxylipins in STEMI patients starting at 24 h post PPCI until 30 days. Univariate receiver operating characteristic (ROC) curve analysis also showed that an elevated ratio of epoxides to diols during ischemia is a predictor of smaller infarct size in patients with STEMI. This study revealed a large alteration in plasma oxylipins in patients presenting with STEMI when compared with controls. Total oxylipin levels rapidly reduced post reperfusion with stable levels reached 24 h post reperfusion and maintained for up to 30 days post infarct. Given the shifts in plasma oxylipins following coronary artery reperfusion, further research is needed to delineate their clinical impact in STEMI patients.
Collapse
Affiliation(s)
- Zahra Solati
- Precision Cardiovascular Medicine Group, St. Boniface Hospital, Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada (H.M.A.)
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - Arun Surendran
- Precision Cardiovascular Medicine Group, St. Boniface Hospital, Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada (H.M.A.)
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - Harold M. Aukema
- Precision Cardiovascular Medicine Group, St. Boniface Hospital, Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada (H.M.A.)
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Research Centre, Winnipeg, MB R2H 2A6, Canada
| | - Amir Ravandi
- Precision Cardiovascular Medicine Group, St. Boniface Hospital, Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada (H.M.A.)
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
- Section of Cardiology, University of Manitoba, Winnipeg, MB R3T 5V6, Canada
| |
Collapse
|
10
|
Tepebaşi MY, Aşci H, Coşan S, Sevük MA, Karakuyu NF, Özmen Ö. Irbesartan has a curative effect on lipopolysaccharide-induced cardiotoxicity by antioxidant and antiapoptotic pathways. Rev Port Cardiol 2023; 42:895-903. [PMID: 37385588 DOI: 10.1016/j.repc.2023.03.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 02/23/2023] [Accepted: 03/01/2023] [Indexed: 07/01/2023] Open
Abstract
INTRODUCTION AND OBJECTIVE Lipopolysaccharide (LPS) has been associated with myocardial inflammation, oxidative stress, apoptosis, and cardiac dysfunction, as well as death by causing sepsis. In this study, we investigated the effect of irbesartan (IRB), an angiotensin receptor antagonist, on cardiotoxicity caused by LPS. METHODS The experiment involved 24 Wistar albino rats divided into three groups of eight: control, LPS (5 mg/kg), and LPS (5 mg/kg)+IRB (3 mg/kg). Parameters including total oxidative status, total antioxidant status, oxidative stress index, and ischemia-modified albumin were measured to assess oxidative stress in heart tissues and serum. Serum CK, CK-MB, and LDH levels were measured spectrophotometrically. RT-qPCR was used to detect the mRNA expression levels of Bcl-2, BAX, p53, caspase-3, and sirtuin 1. Tissues taken from the heart and aorta were examined by immunohistochemistry and histopathology. RESULTS While there was an increase in the parameters indicating heart damage, oxidative stress, and apoptosis in the group given LPS, there was an improvement in all parameters and heart damage in the group treated with IRB. CONCLUSION As a result of our study, we determined that IRB has an ameliorating effect on myocardial damage caused by oxidative stress and apoptosis developed by the LPS-induced sepsis model.
Collapse
Affiliation(s)
| | - Halil Aşci
- Department of Medical Pharmacology, University of Süleyman Demirel, Isparta, TR, Turkey
| | - Samet Coşan
- Department of Medical Pharmacology, University of Süleyman Demirel, Isparta, TR, Turkey
| | | | - Nasıf Fatih Karakuyu
- Department of Medical Pharmacology, University of Süleyman Demirel, Isparta, TR, Turkey
| | - Özlem Özmen
- Department of Pathology, University of Mehmet Akif Ersoy, Burdur, TR, Turkey
| |
Collapse
|
11
|
Edin ML, Gruzdev A, Bradbury JA, Graves JP, Lih FB, DeGraff LM, Fleming I, Zeldin DC. Disruption of Ephx2 in cardiomyocytes but not endothelial cells improves functional recovery after ischemia-reperfusion in isolated mouse hearts. J Biol Chem 2023; 299:103049. [PMID: 36822325 PMCID: PMC10040734 DOI: 10.1016/j.jbc.2023.103049] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 02/10/2023] [Accepted: 02/11/2023] [Indexed: 02/25/2023] Open
Abstract
Cytochromes P450 metabolize arachidonic acid to epoxyeicosatrienoic acids (EETs) which have numerous effects. After cardiac ischemia, EET-induced coronary vasodilation increases delivery of oxygen/nutrients to the myocardium, and EET-induced signaling protects cardiomyocytes against postischemic mitochondrial damage. Soluble epoxide hydrolase 2 (EPHX2) diminishes the benefits of EETs through hydrolysis to less active dihydroxyeicosatrienoic acids. EPHX2 inhibition or genetic disruption improves recovery of cardiac function after ischemia. Immunohistochemical staining revealed EPHX2 expression in cardiomyocytes and some endothelial cells but little expression in cardiac smooth muscle cells or fibroblasts. To determine specific roles of EPHX2 in cardiac cell types, we generated mice with cell-specific disruption of Ephx2 in endothelial cells (Ephx2fx/fx/Tek-cre) or cardiomyocytes (Ephx2fx/fx/Myh6-cre) to compare to global Ephx2-deficient mice (global Ephx2-/-) and WT (Ephx2fx/fx) mice in expression, EET hydrolase activity, and heart function studies. Most cardiac EPHX2 expression and activity is in cardiomyocytes with substantially less activity in endothelial cells. Ephx2fx/fx/Tek-cre hearts have similar EPHX2 expression, hydrolase activity, and postischemic cardiac function as control Ephx2fx/fx hearts. However, Ephx2fx/fx/Myh6-cre hearts were similar to global Ephx2-/- hearts with significantly diminished EPHX2 expression, decreased hydrolase activity, and enhanced postischemic cardiac function compared to Ephx2fx/fx hearts. During reperfusion, Ephx2fx/fx/Myh6-cre hearts displayed increased ERK activation compared to Ephx2fx/fx hearts, which could be reversed by EEZE treatment. EPHX2 did not regulate coronary vasodilation in this model. We conclude that EPHX2 is primarily expressed in cardiomyocytes where it regulates EET hydrolysis and postischemic cardiac function, whereas endothelial EPHX2 does not play a significant role in these processes.
Collapse
Affiliation(s)
- Matthew L Edin
- Division of Intramural Research, National Institute for Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Artiom Gruzdev
- Division of Intramural Research, National Institute for Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - J Alyce Bradbury
- Division of Intramural Research, National Institute for Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Joan P Graves
- Division of Intramural Research, National Institute for Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Fred B Lih
- Division of Intramural Research, National Institute for Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Laura M DeGraff
- Division of Intramural Research, National Institute for Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Ingrid Fleming
- Institute for Vascular Signaling, Centre of Molecular Medicine, Goethe University, Frankfurt am Main, Germany
| | - Darryl C Zeldin
- Division of Intramural Research, National Institute for Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA.
| |
Collapse
|
12
|
ElKhatib MAW, Isse FA, El-Kadi AOS. Effect of inflammation on cytochrome P450-mediated arachidonic acid metabolism and the consequences on cardiac hypertrophy. Drug Metab Rev 2022; 55:50-74. [PMID: 36573379 DOI: 10.1080/03602532.2022.2162075] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The incidence of heart failure (HF) is generally preceded by cardiac hypertrophy (CH), which is the enlargement of cardiac myocytes in response to stress. During CH, the metabolism of arachidonic acid (AA), which is present in the cell membrane phospholipids, is modulated. Metabolism of AA gives rise to hydroxyeicosatetraenoic acids (HETEs) and epoxyeicosatrienoic acids (EETs) via cytochrome P450 (CYP) ω-hydroxylases and CYP epoxygenases, respectively. A plethora of studies demonstrated the involvement of CYP-mediated AA metabolites in the pathogenesis of CH. Also, inflammation is known to be a characteristic hallmark of CH. In this review, our aim is to highlight the impact of inflammation on CYP-derived AA metabolites and CH. Inflammation is shown to modulate the expression of various CYP ω-hydroxylases and CYP epoxygenases and their respective metabolites in the heart. In general, HETEs such as 20-HETE and mid-chain HETEs are pro-inflammatory, while EETs are characterized by their anti-inflammatory and cardioprotective properties. Several mechanisms are implicated in inflammation-induced CH, including the modulation of NF-κB and MAPK. This review demonstrated the inflammatory modulation of cardiac CYPs and their metabolites in the context of CH and the anti-inflammatory strategies that can be employed in the treatment of CH and HF.
Collapse
Affiliation(s)
| | - Fadumo Ahmed Isse
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
| | | |
Collapse
|
13
|
Nummela A, Laaksonen L, Scheinin A, Kaisti K, Vahlberg T, Neuvonen M, Valli K, Revonsuo A, Perola M, Niemi M, Scheinin H, Laitio T. Circulating oxylipin and bile acid profiles of dexmedetomidine, propofol, sevoflurane, and S-ketamine: a randomised controlled trial using tandem mass spectrometry. BJA OPEN 2022; 4:100114. [PMID: 37588789 PMCID: PMC10430865 DOI: 10.1016/j.bjao.2022.100114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 11/11/2022] [Indexed: 08/18/2023]
Abstract
Background This exploratory study aimed to investigate whether dexmedetomidine, propofol, sevoflurane, and S-ketamine affect oxylipins and bile acids, which are functionally diverse molecules with possible connections to cellular bioenergetics, immune modulation, and organ protection. Methods In this randomised, open-label, controlled, parallel group, Phase IV clinical drug trial, healthy male subjects (n=160) received equipotent doses (EC50 for verbal command) of dexmedetomidine (1.5 ng ml-1; n=40), propofol (1.7 μg ml-1; n=40), sevoflurane (0.9% end-tidal; n=40), S-ketamine (0.75 μg ml-1; n=20), or placebo (n=20). Blood samples for tandem mass spectrometry were obtained at baseline, after study drug administration at 60 and 130 min from baseline; 40 metabolites were analysed. Results Statistically significant changes vs placebo were observed in 62.5%, 12.5%, 5.0%, and 2.5% of analytes in dexmedetomidine, propofol, sevoflurane, and S-ketamine groups, respectively. Data are presented as standard deviation score, 95% confidence interval, and P-value. Dexmedetomidine induced wide-ranging decreases in oxylipins and bile acids. Amongst others, 9,10-dihydroxyoctadecenoic acid (DiHOME) -1.19 (-1.6; -0.78), P<0.001 and 12,13-DiHOME -1.22 (-1.66; -0.77), P<0.001 were affected. Propofol elevated 9,10-DiHOME 2.29 (1.62; 2.96), P<0.001 and 12,13-DiHOME 2.13 (1.42; 2.84), P<0.001. Analytes were mostly unaffected by S-ketamine. Sevoflurane decreased tauroursodeoxycholic acid (TUDCA) -2.7 (-3.84; -1.55), P=0.015. Conclusions Dexmedetomidine-induced oxylipin alterations may be connected to pathways associated with organ protection. In contrast to dexmedetomidine, propofol emulsion elevated DiHOMEs, oxylipins associated with acute respiratory distress syndrome, and mitochondrial dysfunction in high concentrations. Further research is needed to establish the behaviour of DIHOMEs during prolonged propofol/dexmedetomidine infusions and to verify the sevoflurane-induced reduction in TUDCA, a suggested neuroprotective agent. Clinical trial registration NCT02624401.
Collapse
Affiliation(s)
- Aleksi Nummela
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland
- Department of Internal Medicine, Turku University Hospital, Turku, Finland
| | - Lauri Laaksonen
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland
- Department of Peri-operative Services, University of Turku and Turku University Hospital, Turku, Finland
| | - Annalotta Scheinin
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland
- Department of Peri-operative Services, University of Turku and Turku University Hospital, Turku, Finland
| | - Kaike Kaisti
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland
- Department of Peri-operative Services, University of Turku and Turku University Hospital, Turku, Finland
| | - Tero Vahlberg
- Department of Clinical Medicine, Biostatistics, Intensive Care and Pain Medicine, University of Turku and Turku University Hospital, Turku, Finland
| | - Mikko Neuvonen
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Katja Valli
- Department of Peri-operative Services, University of Turku and Turku University Hospital, Turku, Finland
- Department of Psychology and Speech-Language Pathology, and Turku Brain and Mind Center, University of Turku, Turku, Finland
- Department of Cognitive Neuroscience and Philosophy, School of Bioscience, University of Skövde, Skövde, Sweden
| | - Antti Revonsuo
- Department of Psychology and Speech-Language Pathology, and Turku Brain and Mind Center, University of Turku, Turku, Finland
- Department of Cognitive Neuroscience and Philosophy, School of Bioscience, University of Skövde, Skövde, Sweden
| | - Markus Perola
- Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Mikko Niemi
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Harry Scheinin
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland
- Department of Peri-operative Services, University of Turku and Turku University Hospital, Turku, Finland
- Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Timo Laitio
- Department of Peri-operative Services, University of Turku and Turku University Hospital, Turku, Finland
| |
Collapse
|
14
|
Sosnowski DK, Jamieson KL, Gruzdev A, Li Y, Valencia R, Yousef A, Kassiri Z, Zeldin DC, Seubert JM. Cardiomyocyte-specific disruption of soluble epoxide hydrolase limits inflammation to preserve cardiac function. Am J Physiol Heart Circ Physiol 2022; 323:H670-H687. [PMID: 35985007 PMCID: PMC9512117 DOI: 10.1152/ajpheart.00217.2022] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/11/2022] [Accepted: 08/12/2022] [Indexed: 11/22/2022]
Abstract
Endotoxemia elicits a multiorgan inflammatory response that results in cardiac dysfunction and often leads to death. Inflammation-induced metabolism of endogenous N-3 and N-6 polyunsaturated fatty acids generates numerous lipid mediators, such as epoxy fatty acids (EpFAs), which protect the heart. However, EpFAs are hydrolyzed by soluble epoxide hydrolase (sEH), which attenuates their cardioprotective actions. Global genetic disruption of sEH preserves EpFA levels and attenuates cardiac dysfunction in mice following acute lipopolysaccharide (LPS)-induced inflammatory injury. In leukocytes, EpFAs modulate the innate immune system through the NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome. However, the mechanisms by which both EpFAs and sEH inhibition exert their protective effects in the cardiomyocyte are still elusive. This study investigated whether cardiomyocyte-specific sEH disruption attenuates inflammation and cardiac dysfunction in acute LPS inflammatory injury via modulation of the NLRP3 inflammasome. We use tamoxifen-inducible CreER recombinase technology to target sEH genetic disruption to the cardiomyocyte. Primary cardiomyocyte studies provide mechanistic insight into inflammasome signaling. For the first time, we demonstrate that cardiomyocyte-specific sEH disruption preserves cardiac function and attenuates inflammatory responses by limiting local cardiac inflammation and activation of the systemic immune response. Mechanistically, inhibition of cardiomyocyte-specific sEH activity or exogenous EpFA treatment do not prevent upregulation of NLRP3 inflammasome machinery in neonatal rat cardiomyocytes. Rather, they limit downstream activation of the pathway leading to release of fewer chemoattractant factors and recruitment of immune cells to the heart. These data emphasize that cardiomyocyte sEH is vital for mediating detrimental systemic inflammation.NEW & NOTEWORTHY The cardioprotective effects of genetic disruption and pharmacological inhibition of sEH have been demonstrated in a variety of cardiac disease models, including acute LPS inflammatory injury. For the first time, it has been demonstrated that sEH genetic disruption limited to the cardiomyocyte profoundly preserves cardiac function and limits local and systemic inflammation following acute LPS exposure. Hence, cardiomyocytes serve a critical role in the innate immune response that can be modulated to protect the heart.
Collapse
Affiliation(s)
- Deanna K Sosnowski
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - K Lockhart Jamieson
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Artiom Gruzdev
- National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Yingxi Li
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Robert Valencia
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Ala Yousef
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Zamaneh Kassiri
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Darryl C Zeldin
- National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - John M Seubert
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
15
|
Angelotti A, Snoke DB, Ormiston K, Cole RM, Borkowski K, Newman JW, Orchard TS, Belury MA. Potential Cardioprotective Effects and Lipid Mediator Differences in Long-Chain Omega-3 Polyunsaturated Fatty Acid Supplemented Mice Given Chemotherapy. Metabolites 2022; 12:metabo12090782. [PMID: 36144189 PMCID: PMC9505633 DOI: 10.3390/metabo12090782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/04/2022] [Accepted: 08/22/2022] [Indexed: 11/25/2022] Open
Abstract
Many commonly used chemotherapies induce mitochondrial dysfunction in cardiac muscle, which leads to cardiotoxicity and heart failure later in life. Dietary long-chain omega-3 polyunsaturated fatty acids (LC n-3 PUFA) have demonstrated cardioprotective function in non-chemotherapy models of heart failure, potentially through the formation of LC n-3 PUFA-derived bioactive lipid metabolites. However, it is unknown whether dietary supplementation with LC n-3 PUFA can protect against chemotherapy-induced cardiotoxicity. To test this, 36 female ovariectomized C57BL/6J mice were randomized in a two-by-two factorial design to either a low (0 g/kg EPA + DHA) or high (12.2 g/kg EPA + DHA) LC n-3 PUFA diet, and received either two vehicle or two chemotherapy (9 mg/kg anthracycline + 90 mg/kg cyclophosphamide) tail vein injections separated by two weeks. Body weight and food intake were measured as well as heart gene expression and fatty acid composition. Heart mitochondria were isolated using differential centrifugation. Mitochondrial isolate oxylipin and N-acylethanolamide levels were measured by mass spectrometry after alkaline hydrolysis. LC n-3 PUFA supplementation attenuated some chemotherapy-induced differences (Myh7, Col3a1) in heart gene expression, and significantly altered various lipid species in cardiac mitochondrial preparations including several epoxy fatty acids [17(18)-EpETE] and N-acylethanolamines (arachidonoylethanolamine, AEA), suggesting a possible functional link between heart lipids and cardiotoxicity.
Collapse
Affiliation(s)
- Austin Angelotti
- Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Deena B. Snoke
- Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA
- Department of Medicine, University of Vermont Larner College of Medicine, Burlington, VT 05405, USA
| | - Kate Ormiston
- Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Rachel M. Cole
- Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Kamil Borkowski
- West Coast Metabolomics Center, Genome Center, University of California Davis, Davis, CA 95616, USA
| | - John W. Newman
- West Coast Metabolomics Center, Genome Center, University of California Davis, Davis, CA 95616, USA
- Western Human Nutrition Research Center, United States Department of Agriculture-Agriculture Research Service, Davis, CA 95616, USA
- Department of Nutrition, University of California-Davis, Davis, CA 95616, USA
| | - Tonya S. Orchard
- Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Martha A. Belury
- Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA
- Correspondence:
| |
Collapse
|
16
|
Abstract
INTRODUCTION Cytochrome P450s (CYPs) are a superfamily of monooxygenases with diverse biological roles. CYP2J2 is an isozyme highly expressed in the heart where it metabolizes endogenous substrates such as N-3/N-6 polyunsaturated fatty acids (PUFA) to produce lipid mediators involved in homeostasis and cardioprotective responses. Expanding our knowledge of the role CYP2J2 has within the heart is important for understanding its impact on cardiac health and disease. AREAS COVERED The objective of this review was to assess the state of knowledge regarding cardiac CYP2J2. A literature search was conducted using PubMed-MEDLINE (from 2022 and earlier) to evaluate relevant studies regarding CYP2J2 mediated cardioprotection, small molecule modulators, effects of CYP2J2 substrates toward biologically relevant effects and implications of CYP2J2 polymorphisms and sexual dimorphism in the heart. EXPERT OPINION Cardiac CYP2J2-mediated metabolism of endogenous and exogenous substrates have been shown to impact cardiac function. Identifying individual factors, like sex and age, that affect CYP2J2 require further elucidation to better understand CYP2J2's clinical relevance. Resolving the biological targets and activities of CYP2J2-derived PUFA metabolites will be necessary to safely target CYP2J2 and design novel analogues. Targeting CYP2J2 for therapeutic aims offers a potential novel approach to regulating cardiac homeostasis, drug metabolism and cardioprotection.
Collapse
|
17
|
Mohammed A, Kalle AM, Reddanna P. Managing SARS-CoV2 Infections Through Resolution of Inflammation by Eicosanoids: A Review. J Inflamm Res 2022; 15:4349-4358. [PMID: 35937919 PMCID: PMC9346295 DOI: 10.2147/jir.s355568] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 04/22/2022] [Indexed: 12/15/2022] Open
Abstract
Severe Corona Virus Disease is characterized by angiocentric inflammation of lungs and cytokine storm leading to potentially fatal multiple organ failure. Several studies have shown the high levels of pro-inflammatory cytokines, indicative of a poor prognosis in COVID-19. Eicosanoids play an important role in the induction of inflammation and cytokine production, while anti-inflammatory and pro-resolving properties of some eicosanoic acid derivatives enable inflamed tissues to return to homeostasis through the resolution of inflammation by aiding the clearance of cell debris and downregulation of pro-inflammatory stimulants. This review attempts to provide an overall insight on the eicosanoids synthesis and their role in the resolution of inflammation in the context of Corona Virus infection.
Collapse
Affiliation(s)
- Ameena Mohammed
- Department of Biological Science, Indian Institute of Science Education and Research Kolkata, Nadia, West Bengal, 741246, India
| | - Arunasree M Kalle
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, 500046, India
| | - Pallu Reddanna
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, 500046, India
| |
Collapse
|
18
|
Sosnowski DK, Jamieson KL, Darwesh AM, Zhang H, Keshavarz-Bahaghighat H, Valencia R, Viveiros A, Edin ML, Zeldin DC, Oudit GY, Seubert JM. Changes in the Left Ventricular Eicosanoid Profile in Human Dilated Cardiomyopathy. Front Cardiovasc Med 2022; 9:879209. [PMID: 35665247 PMCID: PMC9160304 DOI: 10.3389/fcvm.2022.879209] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 04/20/2022] [Indexed: 11/13/2022] Open
Abstract
Objective Metabolites derived from N−3 and N−6 polyunsaturated fatty acids (PUFAs) have both beneficial and detrimental effects on the heart. However, contribution of these lipid mediators to dilated cardiomyopathy (DCM)-associated mitochondrial dysfunction remains unknown. This study aimed to characterize DCM-specific alterations in the PUFA metabolome in conjunction with cardiac mitochondrial quality in human explanted heart tissues. Methods Left ventricular tissues obtained from non-failing control (NFC) or DCM explanted hearts, were assessed for N−3 and N−6 PUFA metabolite levels using LC-MS/MS. mRNA and protein expression of CYP2J2, CYP2C8 and epoxide hydrolase enzymes involved in N−3 and N−6 PUFA metabolism were quantified. Cardiac mitochondrial quality was assessed by transmission electron microscopy, measurement of respiratory chain complex activities and oxygen consumption (respiratory control ratio, RCR) during ADP-stimulated ATP production. Results Formation of cardioprotective CYP-derived lipid mediators, epoxy fatty acids (EpFAs), and their corresponding diols were enhanced in DCM hearts. These findings were corroborated by increased expression of CYP2J2 and CYP2C8 enzymes, as well as microsomal and soluble epoxide hydrolase enzymes, suggesting enhanced metabolic flux and EpFA substrate turnover. DCM hearts demonstrated marked damage to mitochondrial ultrastructure and attenuated mitochondrial function. Incubation of fresh DCM cardiac fibers with the protective EpFA, 19,20-EDP, significantly improved mitochondrial function. Conclusions The current study demonstrates that increased expressions of CYP-epoxygenase enzymes and epoxide hydrolases in the DCM heart correspond with enhanced PUFA-derived EpFA turnover. This is accompanied by severe mitochondrial functional impairment which can be rescued by the administration of exogenous EpFAs.
Collapse
Affiliation(s)
- Deanna K. Sosnowski
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - K. Lockhart Jamieson
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Ahmed M. Darwesh
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Hao Zhang
- Department of Medicine, Mazankowski Alberta Heart Institute, Edmonton, AB, Canada
- Mazankowski Alberta Heart Institute, Edmonton, AB, Canada
| | | | - Robert Valencia
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Anissa Viveiros
- Department of Medicine, Mazankowski Alberta Heart Institute, Edmonton, AB, Canada
- Mazankowski Alberta Heart Institute, Edmonton, AB, Canada
| | - Matthew L. Edin
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health (NIH), Durham, NC, United States
| | - Darryl C. Zeldin
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health (NIH), Durham, NC, United States
| | - Gavin Y. Oudit
- Department of Medicine, Mazankowski Alberta Heart Institute, Edmonton, AB, Canada
- Mazankowski Alberta Heart Institute, Edmonton, AB, Canada
| | - John M. Seubert
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
- Mazankowski Alberta Heart Institute, Edmonton, AB, Canada
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- *Correspondence: John M. Seubert
| |
Collapse
|
19
|
Singh N, Li D, McReynolds CB, Morisseau C, Hammock BD. Improved ELISA for linoleate-derived diols in human plasma utilizing a polyHRP-based secondary tracer. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2022; 14:1810-1819. [PMID: 35481804 PMCID: PMC9116231 DOI: 10.1039/d2ay00272h] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Dihydroxyoctadecenoic acids (DiHOMEs) are cytochrome P450 pathway-derived metabolites of linoleic acid, a highly abundant dietary fatty acid. They serve thermogenic functions at low concentrations but, at high concentrations, are involved in proinflammatory and deleterious outcomes in a wide range of pathologies. Hence, the development of a reliable analytical method is critical to elucidate their potential as biomarkers of health, and enzyme-linked immunoassay (ELISA)-based approaches offer unique benefits as alternatives to traditional liquid chromatography-tandem mass spectrometry (LC-MS/MS) systems. Accordingly, an earlier ELISA for DiHOMEs was dramatically improved employing new secondary tracers and geared towards use in human plasma, a universal matrix in biomedical applications, as well as urine. Three ELISA formats, two utilizing polyHRP-based secondary labels for signal amplification, were compared. The best format involved a biotinylated detection antibody and a polyHRP-conjugated streptavidin tracer. Assay detectability was enhanced 20-fold, relative to the original immunoassay, and performance assessments validated precision, selectivity, and robustness. Fast and easy extraction-clean up steps yielded high analytical recovery and permitted the assay to operate in moderate concentrations (up to 20%) of plasma, expanding its practical relevance. Finally, the ELISA was applied towards detection of DiHOMEs in clinical samples and authenticated with complementary LC-MS/MS analysis. Hence, the method provides a valuable analytical tool to investigate the diverse and extensive roles of DiHOMEs in regulatory biology.
Collapse
Affiliation(s)
- Nalin Singh
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer Center, University of California Davis, Davis, California, 95616, USA.
| | - Dongyang Li
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer Center, University of California Davis, Davis, California, 95616, USA.
| | - Cindy B McReynolds
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer Center, University of California Davis, Davis, California, 95616, USA.
| | - Christophe Morisseau
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer Center, University of California Davis, Davis, California, 95616, USA.
| | - Bruce D Hammock
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer Center, University of California Davis, Davis, California, 95616, USA.
| |
Collapse
|
20
|
Shikuma A, Kami D, Maeda R, Suzuki Y, Sano A, Taya T, Ogata T, Konkel A, Matoba S, Schunck WH, Gojo S. Amelioration of Endotoxemia by a Synthetic Analog of Omega-3 Epoxyeicosanoids. Front Immunol 2022; 13:825171. [PMID: 35281027 PMCID: PMC8908263 DOI: 10.3389/fimmu.2022.825171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 02/07/2022] [Indexed: 11/13/2022] Open
Abstract
Sepsis, a systemic inflammatory response to pathogenic factors, is a difficult to treat life-threatening condition associated with cytokine and eicosanoid storms and multi-organ damage. Omega-3 polyunsaturated fatty acids, such as eicosapentaenoic (EPA) and docosahexaenoic acid, are the precursors of potent anti-inflammatory lipid mediators, including 17,18-epoxyeicosatetraenoic acid (17,18-EEQ), the main metabolite of EPA generated by cytochrome P450 epoxygenases. Searching for novel therapeutic or preventative agents in sepsis, we tested a metabolically robust synthetic analog of 17,18-EEQ (EEQ-A) for its ability to reduce mortality, organ damage, and pro-inflammatory cytokine transcript level in a mouse model of lipopolysaccharide (LPS)-induced endotoxemia, which is closely related to sepsis. Overall survival significantly improved following preventative EEQ-A administration along with decreased transcript level of pro-inflammatory cytokines. On the other hand, the therapeutic protocol was effective in improving survival at 48 hours but insignificant at 72 hours. Histopathological analyses showed significant reductions in hemorrhagic and necrotic damage and infiltration in the liver. In vitro studies with THP-1 and U937 cells showed EEQ-A mediated repression of LPS-induced M1 polarization and enhancement of IL-4-induced M2 polarization of macrophages. Moreover, EEQ-A attenuated the LPS-induced decline of mitochondrial function in THP-1 cells, as indicated by increased basal respiration and ATP production as well as reduction of the metabolic shift to glycolysis. Taken together, these data demonstrate that EEQ-A has potent anti-inflammatory and immunomodulatory properties that may support therapeutic strategies for ameliorating the endotoxemia.
Collapse
Affiliation(s)
- Akira Shikuma
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Daisuke Kami
- Department of Regenerative Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Ryotaro Maeda
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yosuke Suzuki
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Arata Sano
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Toshihiko Taya
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takehiro Ogata
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan.,Department of Pathology and Cell Regulation, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | | | - Satoaki Matoba
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | | | - Satoshi Gojo
- Department of Regenerative Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
21
|
Abstract
Supplemental Digital Content is available in the text. This is the largest study describing the role of P450 epoxygenase metabolites in septic shock in humans and suggests a novel therapeutic target.
Collapse
|
22
|
Wang L, Wang Z, Liu X, Zhang Y, Wang M, Liang X, Li G. Effects of extracellular histones on left ventricular diastolic function and potential mechanisms in mice with sepsis. Am J Transl Res 2022; 14:150-165. [PMID: 35173835 PMCID: PMC8829607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 10/14/2021] [Indexed: 06/14/2023]
Abstract
OBJECTIVE Extracellular histone (EH) is involved in the development of septic myocardial injury (SMI). In this study, we explored whether EH could induce left ventricular diastolic dysfunction (LVDD) in sepsis, and investigated the potential mechanisms through in vivo and in vitro experiments using animal models. METHODS The ratio between E-wave and A-wave (E/A ratio), left ventricular end diastolic volume, and isovolumic relaxation time (IVRT) were measured in cecal ligation and perforation (CLP)- and EH-treated male C57BL/6J mice using echocardiography. The protein and mRNA levels of apoptosis-related proteins (cleaved caspase-3, Bcl-2, and Bax) and cardiac troponin T (cTnT) in the left ventricular tissue/cardiomyocytes were measured using enzyme-linked immunosorbent assay, qRT-PCR, and western blotting. Cardiomyocyte apoptosis was detected by flow cytometry. RESULTS CLP mice presented with LVDD, which was accompanied by increased circulating histones, cTnT and Bax protein levels. Circulating histones were correlated with cTnT, Bax, IVRT, and E/A ratio in CLP mice. Intraperitoneal injection of EH resulted in LVDD in mice. EH induced cardiomyocyte apoptosis, and histone neutralizing agents improved SMI and protected mice against CLP- and EH-induced death. CONCLUSION EH is involved in septic LVDD, and this alteration might be associated with EH-induced apoptosis. EH may serve as a potential therapeutic target for SMI.
Collapse
Affiliation(s)
- Lijun Wang
- Department of Cardiology, Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical UniversityTianjin 300211, People’s Republic of China
- Department of Emergency Medicine, Tianjin Medical University General HospitalTianjin 300052, People’s Republic of China
| | - Ziyi Wang
- Department of Emergency Medicine, Tianjin Medical University General HospitalTianjin 300052, People’s Republic of China
- School of Clinical Medicine, Tsinghua UniversityBeijing 100084, People’s Republic of China
| | - Xing Liu
- Department of Cardiology, Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical UniversityTianjin 300211, People’s Republic of China
| | - Yue Zhang
- Department of Cardiology, Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical UniversityTianjin 300211, People’s Republic of China
| | - Manman Wang
- Department of Cardiology, Affiliated Hospital of Jining Medical UniversityJining 272000, Shandong, People’s Republic of China
| | - Xue Liang
- Department of Cardiology, Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical UniversityTianjin 300211, People’s Republic of China
| | - Guangping Li
- Department of Cardiology, Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical UniversityTianjin 300211, People’s Republic of China
| |
Collapse
|
23
|
Lai J, Chen C. The Role of Epoxyeicosatrienoic Acids in Cardiac Remodeling. Front Physiol 2021; 12:642470. [PMID: 33716791 PMCID: PMC7943617 DOI: 10.3389/fphys.2021.642470] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 01/25/2021] [Indexed: 12/12/2022] Open
Abstract
Epoxyeicosatrienoic acids (EETs) are metabolites of arachidonic acid by cytochrome P450 (CYP) epoxygenases, which include four regioisomers: 5,6-EET, 8,9-EET, 11,12-EET, and 14,15-EET. Each of them possesses beneficial effects against inflammation, fibrosis, and apoptosis, which could combat cardiovascular diseases. Numerous studies have demonstrated that elevation of EETs by overexpression of CYP2J2, inhibition of sEH, or treatment with EET analogs showed protective effects in various cardiovascular diseases, including hypertension, myocardial infarction, and heart failure. As is known to all, cardiac remodeling is the major pathogenesis of cardiovascular diseases. This review will begin with the introduction of EETs and their protective effects in cardiovascular diseases. In the following, the roles of EETs in cardiac remodeling, with a particular emphasis on myocardial hypertrophy, apoptosis, fibrosis, inflammation, and angiogenesis, will be summarized. Finally, it is suggested that upregulation of EETs is a potential therapeutic strategy for cardiovascular diseases. The EET-related drug development against cardiac remodeling is also discussed, including the overexpression of CYP2J2, inhibition of sEH, and the analogs of EET.
Collapse
Affiliation(s)
- Jinsheng Lai
- Division of Cardiology, Tongji Hospital, Tongji Medical College and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Chen Chen
- Division of Cardiology, Tongji Hospital, Tongji Medical College and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
24
|
Darwesh AM, Bassiouni W, Sosnowski DK, Seubert JM. Can N-3 polyunsaturated fatty acids be considered a potential adjuvant therapy for COVID-19-associated cardiovascular complications? Pharmacol Ther 2021; 219:107703. [PMID: 33031856 PMCID: PMC7534795 DOI: 10.1016/j.pharmthera.2020.107703] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 09/28/2020] [Indexed: 02/06/2023]
Abstract
Coronavirus disease 2019 (COVID-19), caused by the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), has currently led to a global pandemic with millions of confirmed and increasing cases around the world. The novel SARS-CoV-2 not only affects the lungs causing severe acute respiratory dysfunction but also leads to significant dysfunction in multiple organs and physiological systems including the cardiovascular system. A plethora of studies have shown the viral infection triggers an exaggerated immune response, hypercoagulation and oxidative stress, which contribute significantly to poor cardiovascular outcomes observed in COVID-19 patients. To date, there are no approved vaccines or therapies for COVID-19. Accordingly, cardiovascular protective and supportive therapies are urgent and necessary to the overall prognosis of COVID-19 patients. Accumulating literature has demonstrated the beneficial effects of n-3 polyunsaturated fatty acids (n-3 PUFA) toward the cardiovascular system, which include ameliorating uncontrolled inflammatory reactions, reduced oxidative stress and mitigating coagulopathy. Moreover, it has been demonstrated the n-3 PUFAs, eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), are precursors to a group of potent bioactive lipid mediators, generated endogenously, which mediate many of the beneficial effects attributed to their parent compounds. Considering the favorable safety profile for n-3 PUFAs and their metabolites, it is reasonable to consider n-3 PUFAs as potential adjuvant therapies for the clinical management of COVID-19 patients. In this article, we provide an overview of the pathogenesis of cardiovascular complications secondary to COVID-19 and focus on the mechanisms that may contribute to the likely benefits of n-3 PUFAs and their metabolites.
Collapse
Affiliation(s)
- Ahmed M Darwesh
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Wesam Bassiouni
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Deanna K Sosnowski
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - John M Seubert
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada; Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
25
|
Von Bank H, Hurtado-Thiele M, Oshimura N, Simcox J. Mitochondrial Lipid Signaling and Adaptive Thermogenesis. Metabolites 2021; 11:124. [PMID: 33671745 PMCID: PMC7926967 DOI: 10.3390/metabo11020124] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/10/2021] [Accepted: 02/12/2021] [Indexed: 12/22/2022] Open
Abstract
Thermogenesis is an energy demanding process by which endotherms produce heat to maintain their body temperature in response to cold exposure. Mitochondria in the brown and beige adipocytes play a key role in thermogenesis, as the site for uncoupling protein 1 (UCP1), which allows for the diffusion of protons through the mitochondrial inner membrane to produce heat. To support this energy demanding process, the mitochondria in brown and beige adipocytes increase oxidation of glucose, amino acids, and lipids. This review article explores the various mitochondria-produced and processed lipids that regulate thermogenesis including cardiolipins, free fatty acids, and acylcarnitines. These lipids play a number of roles in thermogenic adipose tissue including structural support of UCP1, transcriptional regulation, fuel source, and activation of cell signaling cascades.
Collapse
Affiliation(s)
| | | | | | - Judith Simcox
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; (H.V.B.); (M.H.-T.); (N.O.)
| |
Collapse
|
26
|
Soluble Epoxide Hydrolase in Aged Female Mice and Human Explanted Hearts Following Ischemic Injury. Int J Mol Sci 2021; 22:ijms22041691. [PMID: 33567578 PMCID: PMC7915306 DOI: 10.3390/ijms22041691] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 02/02/2021] [Indexed: 12/14/2022] Open
Abstract
Myocardial infarction (MI) accounts for a significant proportion of death and morbidity in aged individuals. The risk for MI in females increases as they enter the peri-menopausal period, generally occurring in middle-age. Cytochrome (CYP) 450 metabolizes N-3 and N-6 polyunsaturated fatty acids (PUFA) into numerous lipid mediators, oxylipids, which are further metabolised by soluble epoxide hydrolase (sEH), reducing their activity. The objective of this study was to characterize oxylipid metabolism in the left ventricle (LV) following ischemic injury in females. Human LV specimens were procured from female patients with ischemic cardiomyopathy (ICM) or non-failing controls (NFC). Female C57BL6 (WT) and sEH null mice averaging 13–16 months old underwent permanent occlusion of the left anterior descending coronary artery (LAD) to induce myocardial infarction. WT (wild type) mice received vehicle or sEH inhibitor, trans-4-[4-(3-adamantan-1-yl-ureido)-cyclohexyloxy]-benzoic acid (tAUCB), in their drinking water ad libitum for 28 days. Cardiac function was assessed using echocardiography and electrocardiogram. Protein expression was determined using immunoblotting, mitochondrial activity by spectrophotometry, and cardiac fibre respiration was measured using a Clark-type electrode. A full metabolite profile was determined by LC–MS/MS. sEH was significantly elevated in ischemic LV specimens from patients, associated with fundamental changes in oxylipid metabolite formation and significant decreases in mitochondrial enzymatic function. In mice, pre-treatment with tAUCB or genetic deletion of sEH significantly improved survival, preserved cardiac function, and maintained mitochondrial quality following MI in female mice. These data indicate that sEH may be a relevant pharmacologic target for women with MI. Although future studies are needed to determine the mechanisms, in this pilot study we suggest targeting sEH may be an effective strategy for reducing ischemic injury and mortality in middle-aged females.
Collapse
|
27
|
Pinckard KM, Shettigar VK, Wright KR, Abay E, Baer LA, Vidal P, Dewal RS, Das D, Duarte-Sanmiguel S, Hernández-Saavedra D, Arts PJ, Lehnig AC, Bussberg V, Narain NR, Kiebish MA, Yi F, Sparks LM, Goodpaster BH, Smith SR, Pratley RE, Lewandowski ED, Raman SV, Wold LE, Gallego-Perez D, Coen PM, Ziolo MT, Stanford KI. A Novel Endocrine Role for the BAT-Released Lipokine 12,13-diHOME to Mediate Cardiac Function. Circulation 2020; 143:145-159. [PMID: 33106031 DOI: 10.1161/circulationaha.120.049813] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Brown adipose tissue (BAT) is an important tissue for thermogenesis, making it a potential target to decrease the risks of obesity, type 2 diabetes, and cardiovascular disease, and recent studies have also identified BAT as an endocrine organ. Although BAT has been implicated to be protective in cardiovascular disease, to this point there are no studies that identify a direct role for BAT to mediate cardiac function. METHODS To determine the role of BAT on cardiac function, we utilized a model of BAT transplantation. We then performed lipidomics and identified an increase in the lipokine 12,13-dihydroxy-9Z-octadecenoic acid (12,13-diHOME). We utilized a mouse model with sustained overexpression of 12,13-diHOME and investigated the role of 12,13-diHOME in a nitric oxide synthase type 1 deficient (NOS1-/-) mouse and in isolated cardiomyocytes to determine effects on function and respiration. We also investigated 12,13-diHOME in a cohort of human patients with heart disease. RESULTS Here, we determined that transplantation of BAT (+BAT) improves cardiac function via the release of the lipokine 12,13-diHOME. Sustained overexpression of 12,13-diHOME using tissue nanotransfection negated the deleterious effects of a high-fat diet on cardiac function and remodeling, and acute injection of 12,13-diHOME increased cardiac hemodynamics via direct effects on the cardiomyocyte. Furthermore, incubation of cardiomyocytes with 12,13-diHOME increased mitochondrial respiration. The effects of 12,13-diHOME were absent in NOS1-/- mice and cardiomyocytes. We also provide the first evidence that 12,13-diHOME is decreased in human patients with heart disease. CONCLUSIONS Our results identify an endocrine role for BAT to enhance cardiac function that is mediated by regulation of calcium cycling via 12,13-diHOME and NOS1.
Collapse
Affiliation(s)
- Kelsey M Pinckard
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (K.M.P., V.K.S., K.R.W., E.A., L.A.B., P.V., R.S.D., D.H.-S., P.J.A., A.C.L., E.D.L., S.V.R., L.E.W., D.G.P., M.T.Z., K.I.S.).,Department of Physiology and Cell Biology (K.M.P., V.K.S., K.R.W., E.A., L.A.B., P.V., R.S.D., D.H.-S., P.J.A., A.C.L., L.E.W., M.T.Z., K.I.S.), The Ohio State University College of Medicine, Columbus
| | - Vikram K Shettigar
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (K.M.P., V.K.S., K.R.W., E.A., L.A.B., P.V., R.S.D., D.H.-S., P.J.A., A.C.L., E.D.L., S.V.R., L.E.W., D.G.P., M.T.Z., K.I.S.).,Department of Physiology and Cell Biology (K.M.P., V.K.S., K.R.W., E.A., L.A.B., P.V., R.S.D., D.H.-S., P.J.A., A.C.L., L.E.W., M.T.Z., K.I.S.), The Ohio State University College of Medicine, Columbus
| | - Katherine R Wright
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (K.M.P., V.K.S., K.R.W., E.A., L.A.B., P.V., R.S.D., D.H.-S., P.J.A., A.C.L., E.D.L., S.V.R., L.E.W., D.G.P., M.T.Z., K.I.S.).,Department of Physiology and Cell Biology (K.M.P., V.K.S., K.R.W., E.A., L.A.B., P.V., R.S.D., D.H.-S., P.J.A., A.C.L., L.E.W., M.T.Z., K.I.S.), The Ohio State University College of Medicine, Columbus
| | - Eaman Abay
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (K.M.P., V.K.S., K.R.W., E.A., L.A.B., P.V., R.S.D., D.H.-S., P.J.A., A.C.L., E.D.L., S.V.R., L.E.W., D.G.P., M.T.Z., K.I.S.).,Department of Physiology and Cell Biology (K.M.P., V.K.S., K.R.W., E.A., L.A.B., P.V., R.S.D., D.H.-S., P.J.A., A.C.L., L.E.W., M.T.Z., K.I.S.), The Ohio State University College of Medicine, Columbus
| | - Lisa A Baer
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (K.M.P., V.K.S., K.R.W., E.A., L.A.B., P.V., R.S.D., D.H.-S., P.J.A., A.C.L., E.D.L., S.V.R., L.E.W., D.G.P., M.T.Z., K.I.S.).,Department of Physiology and Cell Biology (K.M.P., V.K.S., K.R.W., E.A., L.A.B., P.V., R.S.D., D.H.-S., P.J.A., A.C.L., L.E.W., M.T.Z., K.I.S.), The Ohio State University College of Medicine, Columbus
| | - Pablo Vidal
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (K.M.P., V.K.S., K.R.W., E.A., L.A.B., P.V., R.S.D., D.H.-S., P.J.A., A.C.L., E.D.L., S.V.R., L.E.W., D.G.P., M.T.Z., K.I.S.).,Department of Physiology and Cell Biology (K.M.P., V.K.S., K.R.W., E.A., L.A.B., P.V., R.S.D., D.H.-S., P.J.A., A.C.L., L.E.W., M.T.Z., K.I.S.), The Ohio State University College of Medicine, Columbus
| | - Revati S Dewal
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (K.M.P., V.K.S., K.R.W., E.A., L.A.B., P.V., R.S.D., D.H.-S., P.J.A., A.C.L., E.D.L., S.V.R., L.E.W., D.G.P., M.T.Z., K.I.S.).,Department of Physiology and Cell Biology (K.M.P., V.K.S., K.R.W., E.A., L.A.B., P.V., R.S.D., D.H.-S., P.J.A., A.C.L., L.E.W., M.T.Z., K.I.S.), The Ohio State University College of Medicine, Columbus
| | - Devleena Das
- Department of Biomedical Engineering (D.D., S.D.-S., D.G.P.), The Ohio State University, Columbus
| | - Silvia Duarte-Sanmiguel
- Department of Biomedical Engineering (D.D., S.D.-S., D.G.P.), The Ohio State University, Columbus.,Department of Nutrition (S.D.-S.), The Ohio State University, Columbus
| | - Diego Hernández-Saavedra
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (K.M.P., V.K.S., K.R.W., E.A., L.A.B., P.V., R.S.D., D.H.-S., P.J.A., A.C.L., E.D.L., S.V.R., L.E.W., D.G.P., M.T.Z., K.I.S.).,Department of Physiology and Cell Biology (K.M.P., V.K.S., K.R.W., E.A., L.A.B., P.V., R.S.D., D.H.-S., P.J.A., A.C.L., L.E.W., M.T.Z., K.I.S.), The Ohio State University College of Medicine, Columbus
| | - Peter J Arts
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (K.M.P., V.K.S., K.R.W., E.A., L.A.B., P.V., R.S.D., D.H.-S., P.J.A., A.C.L., E.D.L., S.V.R., L.E.W., D.G.P., M.T.Z., K.I.S.).,Department of Physiology and Cell Biology (K.M.P., V.K.S., K.R.W., E.A., L.A.B., P.V., R.S.D., D.H.-S., P.J.A., A.C.L., L.E.W., M.T.Z., K.I.S.), The Ohio State University College of Medicine, Columbus
| | - Adam C Lehnig
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (K.M.P., V.K.S., K.R.W., E.A., L.A.B., P.V., R.S.D., D.H.-S., P.J.A., A.C.L., E.D.L., S.V.R., L.E.W., D.G.P., M.T.Z., K.I.S.).,Department of Physiology and Cell Biology (K.M.P., V.K.S., K.R.W., E.A., L.A.B., P.V., R.S.D., D.H.-S., P.J.A., A.C.L., L.E.W., M.T.Z., K.I.S.), The Ohio State University College of Medicine, Columbus
| | | | | | | | - Fanchao Yi
- Translational Research Institute for Metabolism and Diabetes, AdventHealth, Orlando, FL (F.Y., L.M.S., B.H.G., S.R.S., R.E.P., E.D.L., P.M.C.)
| | - Lauren M Sparks
- Translational Research Institute for Metabolism and Diabetes, AdventHealth, Orlando, FL (F.Y., L.M.S., B.H.G., S.R.S., R.E.P., E.D.L., P.M.C.)
| | - Bret H Goodpaster
- Translational Research Institute for Metabolism and Diabetes, AdventHealth, Orlando, FL (F.Y., L.M.S., B.H.G., S.R.S., R.E.P., E.D.L., P.M.C.)
| | - Steven R Smith
- Translational Research Institute for Metabolism and Diabetes, AdventHealth, Orlando, FL (F.Y., L.M.S., B.H.G., S.R.S., R.E.P., E.D.L., P.M.C.)
| | - Richard E Pratley
- Translational Research Institute for Metabolism and Diabetes, AdventHealth, Orlando, FL (F.Y., L.M.S., B.H.G., S.R.S., R.E.P., E.D.L., P.M.C.)
| | - E Douglas Lewandowski
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (K.M.P., V.K.S., K.R.W., E.A., L.A.B., P.V., R.S.D., D.H.-S., P.J.A., A.C.L., E.D.L., S.V.R., L.E.W., D.G.P., M.T.Z., K.I.S.).,Department of Internal Medicine (E.D.L., S.V.R., M.T.Z., K.I.S.), The Ohio State University College of Medicine, Columbus.,Translational Research Institute for Metabolism and Diabetes, AdventHealth, Orlando, FL (F.Y., L.M.S., B.H.G., S.R.S., R.E.P., E.D.L., P.M.C.).,Sanford Burnham Prebys Medical Discovery Institute at Lake Nona, Orlando, FL (E.D.L.)
| | - Subha V Raman
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (K.M.P., V.K.S., K.R.W., E.A., L.A.B., P.V., R.S.D., D.H.-S., P.J.A., A.C.L., E.D.L., S.V.R., L.E.W., D.G.P., M.T.Z., K.I.S.).,Department of Internal Medicine (E.D.L., S.V.R., M.T.Z., K.I.S.), The Ohio State University College of Medicine, Columbus
| | - Loren E Wold
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (K.M.P., V.K.S., K.R.W., E.A., L.A.B., P.V., R.S.D., D.H.-S., P.J.A., A.C.L., E.D.L., S.V.R., L.E.W., D.G.P., M.T.Z., K.I.S.).,Department of Physiology and Cell Biology (K.M.P., V.K.S., K.R.W., E.A., L.A.B., P.V., R.S.D., D.H.-S., P.J.A., A.C.L., L.E.W., M.T.Z., K.I.S.), The Ohio State University College of Medicine, Columbus.,College of Nursing (L.E.W.), The Ohio State University, Columbus
| | - Daniel Gallego-Perez
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (K.M.P., V.K.S., K.R.W., E.A., L.A.B., P.V., R.S.D., D.H.-S., P.J.A., A.C.L., E.D.L., S.V.R., L.E.W., D.G.P., M.T.Z., K.I.S.).,Department of Surgery (D.G.P.), The Ohio State University College of Medicine, Columbus.,Department of Biomedical Engineering (D.D., S.D.-S., D.G.P.), The Ohio State University, Columbus
| | - Paul M Coen
- Translational Research Institute for Metabolism and Diabetes, AdventHealth, Orlando, FL (F.Y., L.M.S., B.H.G., S.R.S., R.E.P., E.D.L., P.M.C.)
| | - Mark T Ziolo
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (K.M.P., V.K.S., K.R.W., E.A., L.A.B., P.V., R.S.D., D.H.-S., P.J.A., A.C.L., E.D.L., S.V.R., L.E.W., D.G.P., M.T.Z., K.I.S.).,Department of Physiology and Cell Biology (K.M.P., V.K.S., K.R.W., E.A., L.A.B., P.V., R.S.D., D.H.-S., P.J.A., A.C.L., L.E.W., M.T.Z., K.I.S.), The Ohio State University College of Medicine, Columbus.,Department of Internal Medicine (E.D.L., S.V.R., M.T.Z., K.I.S.), The Ohio State University College of Medicine, Columbus
| | - Kristin I Stanford
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (K.M.P., V.K.S., K.R.W., E.A., L.A.B., P.V., R.S.D., D.H.-S., P.J.A., A.C.L., E.D.L., S.V.R., L.E.W., D.G.P., M.T.Z., K.I.S.).,Department of Physiology and Cell Biology (K.M.P., V.K.S., K.R.W., E.A., L.A.B., P.V., R.S.D., D.H.-S., P.J.A., A.C.L., L.E.W., M.T.Z., K.I.S.), The Ohio State University College of Medicine, Columbus.,Department of Internal Medicine (E.D.L., S.V.R., M.T.Z., K.I.S.), The Ohio State University College of Medicine, Columbus
| |
Collapse
|
28
|
Darwesh AM, Bassiouni W, Adebesin AM, Mohammad AS, Falck JR, Seubert JM. A Synthetic Epoxydocosapentaenoic Acid Analogue Ameliorates Cardiac Ischemia/Reperfusion Injury: The Involvement of the Sirtuin 3-NLRP3 Pathway. Int J Mol Sci 2020; 21:ijms21155261. [PMID: 32722183 PMCID: PMC7432620 DOI: 10.3390/ijms21155261] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 07/20/2020] [Accepted: 07/21/2020] [Indexed: 02/07/2023] Open
Abstract
While survival rates have markedly improved following cardiac ischemia-reperfusion (IR) injury, the resulting heart damage remains an important issue. Preserving mitochondrial quality and limiting NLRP3 inflammasome activation is an approach to limit IR injury, in which the mitochondrial deacetylase sirtuin 3 (SIRT3) has a role. Recent data demonstrate cytochrome P450 (CYP450)-derived epoxy metabolites, epoxydocosapentaenoic acids (EDPs), of docosahexaenoic acid (DHA), attenuate cardiac IR injury. EDPs undergo rapid removal and inactivation by enzymatic and non-enzymatic processes. The current study hypothesizes that the cardioprotective effects of the synthetic EDP surrogates AS-27, SA-26 and AA-4 against IR injury involve activation of SIRT3. Isolated hearts from wild type (WT) mice were perfused in the Langendorff mode with vehicle, AS-27, SA-26 or AA-4. Improved postischemic functional recovery, maintained cardiac ATP levels, reduced oxidative stress and attenuation of NLRP3 activation were observed in hearts perfused with the analogue SA-26. Assessment of cardiac mitochondria demonstrated SA-26 preserved SIRT3 activity and reduced acetylation of manganese superoxide dismutase (MnSOD) suggesting enhanced antioxidant capacity. Together, these data demonstrate that the cardioprotective effects of the EDP analogue SA-26 against IR injury involve preservation of mitochondrial SIRT3 activity, which attenuates a detrimental innate NLRP3 inflammasome response.
Collapse
Affiliation(s)
- Ahmed M. Darwesh
- Faculty of Pharmacy and Pharmaceutical Sciences, 2026-M Katz Group Centre for Pharmacy and Health Research, University of Alberta, 11361-97 Ave, Edmonton, AB T6G 2E1, Canada;
| | - Wesam Bassiouni
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2E1, Canada;
| | - Adeniyi Michael Adebesin
- Division of Chemistry, Departments of Biochemistry and Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (A.M.A.); (A.S.M.); (J.R.F.)
| | - Abdul Sattar Mohammad
- Division of Chemistry, Departments of Biochemistry and Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (A.M.A.); (A.S.M.); (J.R.F.)
| | - John R. Falck
- Division of Chemistry, Departments of Biochemistry and Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (A.M.A.); (A.S.M.); (J.R.F.)
| | - John M. Seubert
- Faculty of Pharmacy and Pharmaceutical Sciences, 2026-M Katz Group Centre for Pharmacy and Health Research, University of Alberta, 11361-97 Ave, Edmonton, AB T6G 2E1, Canada;
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2E1, Canada;
- Correspondence: ; Tel.: +1-780-492-0007; Fax: +1-780-492-1217
| |
Collapse
|
29
|
Keshavarz-Bahaghighat H, Darwesh AM, Sosnowski DK, Seubert JM. Mitochondrial Dysfunction and Inflammaging in Heart Failure: Novel Roles of CYP-Derived Epoxylipids. Cells 2020; 9:E1565. [PMID: 32604981 PMCID: PMC7408578 DOI: 10.3390/cells9071565] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 06/19/2020] [Accepted: 06/24/2020] [Indexed: 02/07/2023] Open
Abstract
Age-associated changes leading to a decline in cardiac structure and function contribute to the increased susceptibility and incidence of cardiovascular diseases (CVD) in elderly individuals. Indeed, age is considered a risk factor for heart failure and serves as an important predictor for poor prognosis in elderly individuals. Effects stemming from chronic, low-grade inflammation, inflammaging, are considered important determinants in cardiac health; however, our understanding of the mechanisms involved remains unresolved. A steady decline in mitochondrial function is recognized as an important biological consequence found in the aging heart which contributes to the development of heart failure. Dysfunctional mitochondria contribute to increased cellular stress and an innate immune response by activating the NLRP-3 inflammasomes, which have a role in inflammaging and age-related CVD pathogenesis. Emerging evidence suggests a protective role for CYP450 epoxygenase metabolites of N-3 and N-6 polyunsaturated fatty acids (PUFA), epoxylipids, which modulate various aspects of the immune system and protect mitochondria. In this article, we provide insight into the potential roles N-3 and N-6 PUFA have modulating mitochondria, inflammaging and heart failure.
Collapse
Affiliation(s)
- Hedieh Keshavarz-Bahaghighat
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 2E1, Canada; (H.K.-B.); (A.M.D.); (D.K.S.)
| | - Ahmed M. Darwesh
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 2E1, Canada; (H.K.-B.); (A.M.D.); (D.K.S.)
| | - Deanna K. Sosnowski
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 2E1, Canada; (H.K.-B.); (A.M.D.); (D.K.S.)
| | - John M. Seubert
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 2E1, Canada; (H.K.-B.); (A.M.D.); (D.K.S.)
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2E1, Canada
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta 2020-M Katz Group Centre for Pharmacy and Health Research 11361-87 Avenue, Edmonton, AB T6G 2E1, Canada
| |
Collapse
|
30
|
Bhagwati S, Siddiqi MI. Identification of potential soluble epoxide hydrolase (sEH) inhibitors by ligand-based pharmacophore model and biological evaluation. J Biomol Struct Dyn 2019; 38:4956-4966. [DOI: 10.1080/07391102.2019.1691659] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- Sudha Bhagwati
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute (CSIR-CDRI), Lucknow, India
| | - Mohammad Imran Siddiqi
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute (CSIR-CDRI), Lucknow, India
| |
Collapse
|
31
|
Osthues T, Sisignano M. Oxidized Lipids in Persistent Pain States. Front Pharmacol 2019; 10:1147. [PMID: 31680947 PMCID: PMC6803483 DOI: 10.3389/fphar.2019.01147] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 09/05/2019] [Indexed: 12/13/2022] Open
Abstract
Chemotherapy, nerve injuries, or diseases like multiple sclerosis can cause pathophysiological processes of persistent and neuropathic pain. Thereby, the activation threshold of ion channels is reduced in peripheral sensory neurons to normally noxious stimuli like heat, cold, acid, or mechanical due to sensitization processes. This leads to enhanced neuronal activity, which can result in mechanical allodynia, cold allodynia, thermal hyperalgesia, spontaneous pain, and may initiate persistent and neuropathic pain. The treatment options for persistent and neuropathic pain patients are limited; for about 50% of them, current medication is not efficient due to severe side effects or low response to the treatment. Therefore, it is of special interest to find additional treatment strategies. One approach is the control of neuronal sensitization processes. Herein, signaling lipids are crucial mediators and play an important role during the onset and maintenance of pain. As preclinical studies demonstrate, lipids may act as endogenous ligands or may sensitize transient receptor potential (TRP)-channels. Likewise, they can cause enhanced activity of sensory neurons by mechanisms involving G-protein coupled receptors and activation of intracellular protein kinases. In this regard, oxidized metabolites of the essential fatty acid linoleic acid, 9- and 13-hydroxyoctadecadienoic acid (HODE), their dihydroxy-metabolites (DiHOMEs), as well as epoxides of linoleic acid (EpOMEs) and of arachidonic acid (EETs), as well as lysophospholipids, sphingolipids, and specialized pro-resolving mediators (SPMs) have been reported to play distinct roles in pain transmission or inhibition. Here, we discuss the underlying molecular mechanisms of the oxidized linoleic acid metabolites and eicosanoids. Furthermore, we critically evaluate their role as potential targets for the development of novel analgesics and for the treatment of persistent or neuropathic pain.
Collapse
Affiliation(s)
- Tabea Osthues
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Branch for Translational Medicine and Pharmacology TMP, Frankfurt, Germany
| | - Marco Sisignano
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt/ZAFES, University Hospital, Goethe-University, Frankfurt, Germany
| |
Collapse
|
32
|
Genetic Deletion or Pharmacological Inhibition of Soluble Epoxide Hydrolase Ameliorates Cardiac Ischemia/Reperfusion Injury by Attenuating NLRP3 Inflammasome Activation. Int J Mol Sci 2019; 20:ijms20143502. [PMID: 31319469 PMCID: PMC6678157 DOI: 10.3390/ijms20143502] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 07/12/2019] [Accepted: 07/16/2019] [Indexed: 02/06/2023] Open
Abstract
Activation of the nucleotide-binding oligomerization domain-like receptor (NLR) family pyrin domain containing 3 (NLRP3) inflammasome cascade has a role in the pathogenesis of ischemia/reperfusion (IR) injury. There is growing evidence indicating cytochrome p450 (CYP450)-derived metabolites of n-3 and n-6 polyunsaturated fatty acids (PUFAs) possess both adverse and protective effects in the heart. CYP-derived epoxy metabolites are rapidly hydrolyzed by the soluble epoxide hydrolase (sEH). The current study hypothesized that the cardioprotective effects of inhibiting sEH involves limiting activation of the NLRP3 inflammasome. Isolated hearts from young wild-type (WT) and sEH null mice were perfused in the Langendorff mode with either vehicle or the specific sEH inhibitor t-AUCB. Improved post-ischemic functional recovery and better mitochondrial respiration were observed in both sEH null hearts or WT hearts perfused with t-AUCB. Inhibition of sEH markedly attenuated the activation of the NLRP3 inflammasome complex and limited the mitochondrial localization of the fission protein dynamin-related protein-1 (Drp-1) triggered by IR injury. Cardioprotective effects stemming from the inhibition of sEH included preserved activities of both cytosolic thioredoxin (Trx)-1 and mitochondrial Trx-2 antioxidant enzymes. Together, these data demonstrate that inhibiting sEH imparts cardioprotection against IR injury via maintaining post-ischemic mitochondrial function and attenuating a detrimental innate inflammatory response.
Collapse
|