1
|
Flores J, Pena C, Nugent K. Salt Sensitivity of Blood Pressure and the Role of the Immune System in Hypertension. Cardiol Rev 2024:00045415-990000000-00381. [PMID: 39679725 DOI: 10.1097/crd.0000000000000834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Salt-sensitive blood pressure is a clinical phenotype defined as exaggerated blood pressure responses to salt loading and salt depletion. This characteristic occurs in 25% of the general population and 50% of patients with hypertension and contributes to the pathogenesis of hypertension in some patients. Hypertension is associated with chronic inflammatory responses and has immune cell accumulation in several hypertensive target organs, including the brain, kidneys, heart, blood vessels, and the perivascular adipose tissue, and these cellular responses likely exacerbate hypertension. The different factors implicated in the pathogenesis of salt-sensitive hypertension include renin-angiotensin-aldosterone system dysfunction, aldosterone-dependent and aldosterone-independent mineralocorticoid receptor signaling, and the sympathetic nervous system dysfunction. Experimental studies have shown an important role of both innate and adaptive immune cells, especially lymphocytes, in angiotensin II-induced hypertension. The epithelial sodium channel (ENaC) allows entry of sodium into dendritic cells, and this leads to a sequence of events, including the production of reactive oxygen species, which activates the NLRP3 inflammasome and contributes to salt-sensitive hypertension through the amiloride-sensitive ENaC and isolevuglandin-adduct formation. This review summarizes the general aspects of salt sensitivity, focuses on the immunological/inflammatory factors involved in its development, considers general changes in microvasculature, and discusses management.
Collapse
Affiliation(s)
- Jackeline Flores
- From the Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX
| | | | | |
Collapse
|
2
|
Albritton CF, Demirci M, Neikirk K, Ertuglu LA, Ishimwe JA, Mutchler AL, Sheng Q, Laffer CL, Wanjalla CN, Ahmed T, Haynes AP, Saleem M, Beasley HK, Marshall AG, Vue Z, Ikizler AT, Kleyman TR, Kon V, Hinton A, Kirabo A. Myeloid Cell Glucocorticoid, Not Mineralocorticoid Receptor Signaling, Contributes to Salt-Sensitive Hypertension in Humans via Cortisol. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.10.598374. [PMID: 38915603 PMCID: PMC11195113 DOI: 10.1101/2024.06.10.598374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
BACKGROUND Salt sensitivity of blood pressure (SSBP) is an independent risk factor for cardiovascular morbidity and mortality, yet the etiology is poorly understood. We previously found that serum/glucocorticoid-regulated kinase 1 (SGK1) and epoxyeicosatrienoic acids (EETs) regulate epithelial sodium channel (ENaC)-dependent sodium entry into monocyte-derived antigen-presenting cells (APCs) and activation of NADPH oxidase, leading to the formation of isolevuglandins (IsoLGs) in SSBP. Whereas aldosterone via the mineralocorticoid receptor (MR) activates SGK1 leading to hypertension, our past findings indicate that levels of plasma aldosterone do not correlate with SSBP, and there is little to no MR expression in APCs. Thus, we hypothesized that cortisol acting via the glucocorticoid receptor (GR), not the MR in APCs mediates SGK1 actions to induce SSBP. METHODS We performed cellular indexing of transcriptomes and epitopes by sequencing (CITE-Seq) analysis on peripheral blood mononuclear cells of humans rigorously phenotyped for SSBP using an inpatient salt loading/depletion protocol to determine expression of MR, GR, and SGK1 in immune cells. In additional experiments, we performed bulk transcriptomic analysis on isolated human monocytes following in vitro treatment with high salt from a separate cohort. We then measured urine and plasma cortisol, cortisone, renin, and aldosterone. Subsequently, we measured the association of these hormones with changes in systolic, diastolic, mean arterial pressure and pulse pressure as well as immune cell activation via IsoLG formation. RESULTS We found that myeloid APCs predominantly express the GR and SGK1 with no expression of the MR. Expression of the GR in APCs increased after salt loading and decreased with salt depletion in salt-sensitive but not salt-resistant people and was associated with increased expression of SGK1. Moreover, we found that plasma and urine cortisol/cortisone but not aldosterone/renin correlated with SSBP and APCs activation via IsoLGs. We also found that cortisol negatively correlates with EETs. CONCLUSION Our findings suggest that renal cortisol signaling via the GR but not the MR in APCs contributes to SSBP via cortisol. Urine and plasma cortisol may provide an important currently unavailable feasible diagnostic tool for SSBP. Moreover, cortisol-GR-SGK1-ENaC signaling pathway may provide treatment options for SSBP.
Collapse
Affiliation(s)
- Claude F. Albritton
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37212-8802, USA
- Department of Biomedical Sciences, School of Graduate Studies, Meharry Medical College, Nashville, TN 37208-3501, USA
| | - Mert Demirci
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Kit Neikirk
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37212-8802, USA
| | - Lale A. Ertuglu
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Jeanne A Ishimwe
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37212-8802, USA
| | - Ashley L Mutchler
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37212-8802, USA
| | - Quanhu Sheng
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37212-8802, USA
| | - Cheryl L Laffer
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37212-8802, USA
| | - Celestine N. Wanjalla
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37212-8802, USA
| | - Taseer Ahmed
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37212-8802, USA
- Department of Pharmacology, College of Pharmacy, University of Sargodha, University Road, Sargodha, Punjab, 40100, Pakistan
| | - Alexandria Porcia Haynes
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37212-8802, USA
| | - Mohammad Saleem
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37212-8802, USA
| | - Heather K. Beasley
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37212-8802, USA
| | - Andrea G. Marshall
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37212-8802, USA
| | - Zer Vue
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37212-8802, USA
| | - Alp T Ikizler
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Thomas R. Kleyman
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Valentina Kon
- Division of Nephrology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN
| | - Antentor Hinton
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37212-8802, USA
| | - Annet Kirabo
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37212-8802, USA
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37212-8802, USA
- Vanderbilt Center for Immunobiology
- Vanderbilt Institute for Infection, Immunology and Inflammation
- Vanderbilt Institute for Global Health
| |
Collapse
|
3
|
Ertuglu LA, Mutchler AP, Jamison S, Laffer CL, Saleem M, Blackwell DJ, Kryshtal DO, Sahinoz M, Sheng Q, Wanjalla CN, Pakala S, Justin Y, Gutierrez OM, Kleyman TR, Knollmann BC, Ikizler TA, Kirabo A. Eicosanoid-Regulated Myeloid ENaC and Isolevuglandin Formation in Human Salt-Sensitive Hypertension. Hypertension 2024; 81:516-529. [PMID: 37675576 PMCID: PMC10918035 DOI: 10.1161/hypertensionaha.123.21285] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 08/15/2023] [Indexed: 09/08/2023]
Abstract
BACKGROUND The mechanisms by which salt increases blood pressure in people with salt sensitivity remain unclear. Our previous studies found that high sodium enters antigen-presenting cells (APCs) via the epithelial sodium channel and leads to the production of isolevuglandins and hypertension. In the current mechanistic clinical study, we hypothesized that epithelial sodium channel-dependent isolevuglandin-adduct formation in APCs is regulated by epoxyeicosatrienoic acids (EETs) and leads to salt-sensitive hypertension in humans. METHODS Salt sensitivity was assessed in 19 hypertensive subjects using an inpatient salt loading and depletion protocol. Isolevuglandin-adduct accumulation in APCs was analyzed using flow cytometry. Gene expression in APCs was analyzed using cellular indexing of transcriptomes and epitopes by sequencing analysis of blood mononuclear cells. Plasma and urine EETs were measured using liquid chromatography-mass spectrometry. RESULTS Baseline isolevuglandin+ APCs correlated with higher salt-sensitivity index. Isolevuglandin+ APCs significantly decreased from salt loading to depletion with an increasing salt-sensitivity index. We observed that human APCs express the epithelial sodium channel δ subunit, SGK1 (salt-sensing kinase serum/glucocorticoid kinase 1), and cytochrome P450 2S1. We found a direct correlation between baseline urinary 14,15 EET and salt-sensitivity index, whereas changes in urinary 14,15 EET negatively correlated with isolevuglandin+ monocytes from salt loading to depletion. Coincubation with 14,15 EET inhibited high-salt-induced increase in isolevuglandin+ APC. CONCLUSIONS Isolevuglandin formation in APCs responds to acute changes in salt intake in salt-sensitive but not salt-resistant people with hypertension, and this may be regulated by renal 14,15 EET. Baseline levels of isolevuglandin+ APCs or urinary 14,15 EET may provide diagnostic tools for salt sensitivity without a protocol of salt loading.
Collapse
Affiliation(s)
- Lale A. Ertuglu
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ashley Pitzer Mutchler
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center Nashville, TN, USA
| | - S Jamison
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center Nashville, TN, USA
- Meharry Medical College Nashville, Nashville, TN, United States
| | - Cheryl L. Laffer
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center Nashville, TN, USA
| | - Mohammad Saleem
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center Nashville, TN, USA
| | - Daniel J. Blackwell
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center Nashville, TN, USA
| | - Dmytro O. Kryshtal
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center Nashville, TN, USA
| | - Melis Sahinoz
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Quanhu Sheng
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Celestine N. Wanjalla
- Department of Internal Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center Nashville, TN, USA
| | - Suman Pakala
- Department of Internal Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center Nashville, TN, USA
| | - Yu Justin
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Orlando M Gutierrez
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Thomas R. Kleyman
- Departments of Medicine, Cell Biology, Pharmacology, and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Björn C. Knollmann
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center Nashville, TN, USA
| | - T. Alp Ikizler
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Annet Kirabo
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center Nashville, TN, USA
- Vanderbilt Center for Immunobiology (VCI)
- Vanderbilt Institute for Infection, Immunology, and Inflammation (VI4)
- Vanderbilt Institute for Global Health (VIGH)
| |
Collapse
|
4
|
Ertuglu LA, Laffer CL, Kirabo A. In Memoriam: Fernando Elijovich. Hypertension 2023; 80:e1-e3. [PMID: 36475860 DOI: 10.1161/hypertensionaha.122.20541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Lale A Ertuglu
- Division of Nephrology, Department of Medicine (L.A.E.), Vanderbilt University Medical Center, Nashville, TN
| | - Cheryl L Laffer
- Division of Clinical Pharmacology, Department of Medicine (C.L.L., A.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine (C.L.L., A.K.), Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
5
|
Imig JD. Orally active epoxyeicosatrienoic acid analogs in hypertension and renal injury. ADVANCES IN PHARMACOLOGY 2022; 94:27-55. [PMID: 35659375 PMCID: PMC10105514 DOI: 10.1016/bs.apha.2022.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Epoxyeicosatrienoic acids (EETs) are arachidonic acid metabolites synthesized by cytochrome P450 epoxygenases. Biological activities for EETs include vasodilation, decreasing inflammation, opposing apoptosis, and inhibiting renal sodium reabsorption. These actions are beneficial in lowering blood pressure and slowing kidney disease progression. Furthermore, evidence in human and experimental animal studies have found that decreased EET levels contribute to hypertension and kidney diseases. Consequently, EET mimics/analogs have been developed as a potential therapeutic for hypertension and acute and chronic kidney diseases. Their development has resulted in EET analogs that are orally active with favorable pharmacological profiles. Analogs for 8,9-EET, 11,12-EET, and 14,15-EET have been tested in several hypertension and kidney disease animal models. More recently, kidney targeted EET analogs have been synthesized and tested against drug-induced nephrotoxicity. Experimental evidence has demonstrated compelling therapeutic potential for EET analogs to oppose cardiovascular and kidney diseases. These EET analogs lower blood pressure, decrease kidney inflammation, improve vascular endothelial function, and decrease kidney fibrosis and apoptosis. Overall, these preclinical studies support the likelihood that EET analogs will advance to clinical trials for hypertension and associated comorbidities or acute and chronic kidney diseases.
Collapse
Affiliation(s)
- John D Imig
- Drug Discovery Center, Medical College of Wisconsin, Milwaukee, WI, United States.
| |
Collapse
|
6
|
Elijovich F, Kleyman TR, Laffer CL, Kirabo A. Immune Mechanisms of Dietary Salt-Induced Hypertension and Kidney Disease: Harry Goldblatt Award for Early Career Investigators 2020. Hypertension 2021; 78:252-260. [PMID: 34232678 DOI: 10.1161/hypertensionaha.121.16495] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Salt sensitivity of blood pressure is an independent risk factor for cardiovascular mortality not only in hypertensive but also in normotensive adults. The diagnosis of salt sensitivity of blood pressure is not feasible in the clinic due to lack of a simple diagnostic test, making it difficult to investigate therapeutic strategies. Most research efforts to understand the mechanisms of salt sensitivity of blood pressure have focused on renal regulation of sodium. However, salt retention or plasma volume expansion is not different between salt-sensitive and salt-resistant individuals. In addition, over 70% of extracellular fluid is interstitial and, therefore, not directly controlled by renal salt and water excretion. We discuss in this review how the seminal work by Harry Goldblatt paved the way for our attempts at understanding the mechanisms that underlie immune activation by salt in hypertension. We describe our findings that sodium, entering antigen-presenting cells via an epithelial sodium channel, triggers a PKC (protein kinase C)- and SGK1 (serum/glucocorticoid kinase 1)-stimulated activation of nicotinamide adenine dinucleotide phosphate oxidase, which, in turn, enhances lipid oxidation with generation of highly reactive isolevuglandins. Isolevuglandins adduct to proteins, with the potential to generate degraded peptide neoantigens. Activated antigen-presenting cells increase production of the TH17 polarizing cytokines, IL (interleukin)-6, IL-1β, and IL-23, which leads to differentiation and proliferation of IL-17A producing T cells. Our laboratory and others have shown that this cytokine contributes to hypertension. We also discuss where this sodium activation of antigen-presenting cells may occur in vivo and describe the multiple experiments, with pharmacological antagonists and knockout mice that we used to unravel this sequence of events in rodents. Finally, we describe experiments in mononuclear cells obtained from normotensive or hypertensive volunteers, which confirm that analogous processes of salt-induced immunity take place in humans.
Collapse
Affiliation(s)
- Fernando Elijovich
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (F.E., C.L.L., A.K.)
| | - Thomas R Kleyman
- Departments of Medicine, Cell Biology, Pharmacology, and Chemical Biology, University of Pittsburgh, PA (T.R.K.)
| | - Cheryl L Laffer
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (F.E., C.L.L., A.K.)
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (F.E., C.L.L., A.K.)
| |
Collapse
|
7
|
Imig JD. Diabetes risk associated with plasma epoxylipid levels. EBioMedicine 2021; 66:103331. [PMID: 33857907 PMCID: PMC8050865 DOI: 10.1016/j.ebiom.2021.103331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 03/22/2021] [Indexed: 11/20/2022] Open
Affiliation(s)
- John D Imig
- Drug Discovery Center, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
8
|
Luther JM, Wei DS, Ghoshal K, Peng D, Adler GK, Turcu AF, Nian H, Yu C, Solorzano CC, Pozzi A, Brown NJ. Treatment of Primary Aldosteronism Increases Plasma Epoxyeicosatrienoic Acids. Hypertension 2021; 77:1323-1331. [PMID: 33583202 PMCID: PMC8320355 DOI: 10.1161/hypertensionaha.120.14808] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 01/17/2021] [Indexed: 12/31/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- James M. Luther
- Vanderbilt University Medical Center Department of Medicine, Division of Clinical Pharmacology
| | - Dawei S. Wei
- Vanderbilt University Medical Center Department of Medicine, Division of Clinical Pharmacology
| | - Kakali Ghoshal
- Vanderbilt University Medical Center Department of Medicine, Division of Nephrology and Hypertension
| | - Dungeng Peng
- Vanderbilt University Medical Center Department of Medicine, Division of Clinical Pharmacology
| | - Gail K. Adler
- Brigham and Women’s Hospital, Division of Endocrinology and Hypertension, Department of Medicine, Harvard Medical School
| | - Adina F. Turcu
- University of Michigan, Division of Endocrinology, Department of Medicine
| | - Hui Nian
- Vanderbilt University Department of Biostatistics
| | - Chang Yu
- Vanderbilt University Department of Biostatistics
| | | | - Ambra Pozzi
- Vanderbilt University Medical Center Department of Medicine, Division of Nephrology and Hypertension
- Department of Veterans Affairs, Nashville, TN
| | - Nancy J. Brown
- Vanderbilt University Medical Center Department of Medicine, Division of Clinical Pharmacology
- Yale School of Medicine
| |
Collapse
|
9
|
Elijovich F, Kirabo A, Laffer CL. Hypothesis: Unrecognized actions of ENaC blockade in improving refractory-resistant hypertension and residual cardiovascular risk. Int J Cardiol Hypertens 2020; 7:100048. [PMID: 33447773 PMCID: PMC7803029 DOI: 10.1016/j.ijchy.2020.100048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 09/04/2020] [Indexed: 11/16/2022] Open
|
10
|
Gladine C, Ostermann AI, Newman JW, Schebb NH. MS-based targeted metabolomics of eicosanoids and other oxylipins: Analytical and inter-individual variabilities. Free Radic Biol Med 2019; 144:72-89. [PMID: 31085232 DOI: 10.1016/j.freeradbiomed.2019.05.012] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 04/19/2019] [Accepted: 05/10/2019] [Indexed: 02/07/2023]
Abstract
Oxylipins, including the well-known eicosanoids, are potent lipid mediators involved in numerous physiological and pathological processes. Therefore, their quantitative profiling has gained a lot of attention during the last years notably in the active field of health biomarker discovery. Oxylipins include hundreds of structurally and stereochemically distinct lipid species which today are most commonly analyzed by (ultra) high performance liquid chromatography-mass spectrometry based ((U)HPLC-MS) methods. To maximize the utility of oxylipin profiling in clinical research, it is crucial to understand and assess the factors contributing to the analytical and biological variability of oxylipin profiles in humans. In this review, these factors and their impacts are summarized and discussed, providing a framework for recommendations expected to enhance the interlaboratory comparability and biological interpretation of oxylipin profiling in clinical research.
Collapse
Affiliation(s)
- Cécile Gladine
- Université Clermont Auvergne, INRA, UNH, Unité de Nutrition Humaine, CRNH Auvergne, Clermont-Ferrand, France.
| | - Annika I Ostermann
- Chair of Food Chemistry, Faculty of Mathematics and Natural Sciences, Gaußstraße 20, University of Wuppertal, 42119, Wuppertal, Germany
| | - John W Newman
- United States Department of Agriculture, Agricultural Research Service, Western Human Nutrition Research Center, Davis, CA, USA; University of California Davis, Department of Nutrition, Davis, CA, USA
| | - Nils Helge Schebb
- Chair of Food Chemistry, Faculty of Mathematics and Natural Sciences, Gaußstraße 20, University of Wuppertal, 42119, Wuppertal, Germany
| |
Collapse
|
11
|
Abstract
Therapeutics for arachidonic acid pathways began with the development of non-steroidal anti-inflammatory drugs that inhibit cyclooxygenase (COX). The enzymatic pathways and arachidonic acid metabolites and respective receptors have been successfully targeted and therapeutics developed for pain, inflammation, pulmonary and cardiovascular diseases. These drugs target the COX and lipoxygenase pathways but not the third branch for arachidonic acid metabolism, the cytochrome P450 (CYP) pathway. Small molecule compounds targeting enzymes and CYP epoxy-fatty acid metabolites have evolved rapidly over the last two decades. These therapeutics have primarily focused on inhibiting soluble epoxide hydrolase (sEH) or agonist mimetics for epoxyeicosatrienoic acids (EET). Based on preclinical animal model studies and human studies, major therapeutic indications for these sEH inhibitors and EET mimics/analogs are renal and cardiovascular diseases. Novel small molecules that inhibit sEH have advanced to human clinical trials and demonstrate promise for cardiovascular diseases. Challenges remain for sEH inhibitor and EET analog drug development; however, there is a high likelihood that a drug that acts on this third branch of arachidonic acid metabolism will be utilized to treat a cardiovascular or kidney disease in the next decade.
Collapse
Affiliation(s)
- John D Imig
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA.
| |
Collapse
|