1
|
Verouti S, Aeschlimann G, Wang Q, Del Olmo DA, Peyter AC, Menétrey S, Winter DV, Odermatt A, Pearce D, Hummler E, Vanderriele PE. Salt-sensitive hypertension in GR mutant rats is associated with altered plasma polyunsaturated fatty acid levels and aortic vascular reactivity. Pflugers Arch 2025; 477:37-53. [PMID: 39256246 PMCID: PMC11711871 DOI: 10.1007/s00424-024-03014-y] [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: 04/30/2024] [Revised: 08/22/2024] [Accepted: 08/23/2024] [Indexed: 09/12/2024]
Abstract
In humans, glucocorticoid resistance is attributed to mutations in the glucocorticoid receptor (GR). Most of these mutations result in decreased ligand binding, transactivation, and/or translocation, albeit with normal protein abundances. However, there is no clear genotype‒phenotype relationship between the severity or age at disease presentation and the degree of functional loss of the receptor. Previously, we documented that a GR+/- rat line developed clinical features of glucocorticoid resistance, namely, hypercortisolemia, adrenal hyperplasia, and salt-sensitive hypertension. In this study, we analyzed the GR+/em4 rat model heterozygously mutant for the deletion of exon 3, which encompasses the second zinc finger, including the domains of DNA binding, dimerization, and nuclear localization signals. On a standard diet, mutant rats exhibited a trend toward increased corticosterone levels and a normal systolic blood pressure and heart rate but presented with adrenal hyperplasia. They exhibited increased adrenal soluble epoxide hydroxylase (sEH), favoring an increase in less active polyunsaturated fatty acids. Indeed, a significant increase in nonactive omega-3 and omega-6 polyunsaturated fatty acids, such as 5(6)-DiHETrE or 9(10)-DiHOME, was observed with advanced age (10 versus 5 weeks old) and following a switch to a high-salt diet accompanied by salt-sensitive hypertension. In thoracic aortas, a reduced soluble epoxide hydrolase (sEH) protein abundance resulted in altered vascular reactivity upon a standard diet, which was blunted upon a high-salt diet. In conclusion, mutations in the GR affecting the ligand-binding domain as well as the dimerization domain resulted in deregulated GR signaling, favoring salt-sensitive hypertension in the absence of obvious mineralocorticoid excess.
Collapse
Affiliation(s)
- S Verouti
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
- National Center of Competence in Research, Kidney.CH, Lausanne, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - G Aeschlimann
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Q Wang
- Division of Nephrology and Hypertension, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - D Ancin Del Olmo
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - A C Peyter
- Neonatal Research Laboratory, Clinic of Neonatology, Department Woman-Mother-Child, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - S Menétrey
- Neonatal Research Laboratory, Clinic of Neonatology, Department Woman-Mother-Child, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - D V Winter
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - A Odermatt
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - D Pearce
- Department of Medicine and Cellular & Molecular Pharmacology, University of California, San Francisco, USA
| | - E Hummler
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
- National Center of Competence in Research, Kidney.CH, Lausanne, Switzerland
| | - P E Vanderriele
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland.
- National Center of Competence in Research, Kidney.CH, Lausanne, Switzerland.
| |
Collapse
|
2
|
Leow JWH, Chan ECY. CYP2J2-mediated metabolism of arachidonic acid in heart: A review of its kinetics, inhibition and role in heart rhythm control. Pharmacol Ther 2024; 258:108637. [PMID: 38521247 DOI: 10.1016/j.pharmthera.2024.108637] [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: 06/18/2023] [Revised: 02/06/2024] [Accepted: 03/11/2024] [Indexed: 03/25/2024]
Abstract
Cytochrome P450 2 J2 (CYP2J2) is primarily expressed extrahepatically and is the predominant epoxygenase in human cardiac tissues. This highlights its key role in the metabolism of endogenous substrates. Significant scientific interest lies in cardiac CYP2J2 metabolism of arachidonic acid (AA), an omega-6 polyunsaturated fatty acid, to regioisomeric bioactive epoxyeicosatrienoic acid (EET) metabolites that show cardioprotective effects including regulation of cardiac electrophysiology. From an in vitro perspective, the accurate characterization of the kinetics of CYP2J2 metabolism of AA including its inhibition and inactivation by drugs could be useful in facilitating in vitro-in vivo extrapolations to predict drug-AA interactions in drug discovery and development. In this review, background information on the structure, regulation and expression of CYP2J2 in human heart is presented alongside AA and EETs as its endogenous substrate and metabolites. The in vitro and in vivo implications of the kinetics of this endogenous metabolic pathway as well as its perturbation via inhibition and inactivation by drugs are elaborated. Additionally, the role of CYP2J2-mediated metabolism of AA to EETs in cardiac electrophysiology will be expounded.
Collapse
Affiliation(s)
- Jacqueline Wen Hui Leow
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore 117543, Singapore
| | - Eric Chun Yong Chan
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore 117543, Singapore.
| |
Collapse
|
3
|
Fragner ML, Parikh MA, Jackson KA, Schwartzman ML, Frishman WH, Peterson SJ. GPR75: A Newly Identified Receptor for Targeted Intervention in the Treatment of Obesity and Metabolic Syndrome. Cardiol Rev 2024:00045415-990000000-00259. [PMID: 38695569 PMCID: PMC11808825 DOI: 10.1097/crd.0000000000000711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Metabolic syndrome increases the risk of stroke, cardiovascular disease, and diabetes. The morbidity and mortality associated with this constellation of risk factors are equally alarming when considering the economic and global significance that this epidemic has on an institutional and patient level. Despite several current treatments available, there needs to be a continuous effort to explore more specific and effective druggable entities for preventative and therapeutic interventions. Within this context, the G-protein coupled receptor, GPR75, is an attractive pharmacological target. GPR75 and its association with its ligand, 20-hydroxyeicosatetraenoic acid, have been shown to promote hypertension, inflammation, obesity, and insulin resistance. This review will help shed light on this novel signaling pathway and offer a perspective on a promising new direction of targeting different aspects of the metabolic syndrome involving GPR75. Gene targeting of GPR75 is more effective than current pharmacologic therapies without the known side effects.
Collapse
Affiliation(s)
- Michael L. Fragner
- Department of Medicine, New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, NY
| | - Manish A. Parikh
- Department of Medicine, New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, NY
- Weill Department of Medicine, Weill Cornell Medicine, New York, NY
| | - Kaedrea A. Jackson
- Department of Emergency Medicine, New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, NY
| | | | | | - Stephen J. Peterson
- Department of Medicine, New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, NY
- Weill Department of Medicine, Weill Cornell Medicine, New York, NY
| |
Collapse
|
4
|
Davis JT, Elliott JE, Duke JW, Cristobal A, Lovering AT. Hyperoxia-induced stepwise reduction in blood flow through intrapulmonary, but not intracardiac, shunt during exercise. Am J Physiol Regul Integr Comp Physiol 2023; 325:R96-R105. [PMID: 37184225 PMCID: PMC10292968 DOI: 10.1152/ajpregu.00014.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 04/25/2023] [Accepted: 05/11/2023] [Indexed: 05/16/2023]
Abstract
Blood flow through intrapulmonary arteriovenous anastomoses (IPAVA) (QIPAVA) increases during exercise breathing air, but it has been proposed that QIPAVA is reduced during exercise while breathing a fraction of inspired oxygen ([Formula: see text]) of 1.00. It has been argued that the reduction in saline contrast bubbles through IPAVA is due to altered in vivo microbubble dynamics with hyperoxia reducing bubble stability, rather than closure of IPAVA. To definitively determine whether breathing hyperoxia decreases saline contrast bubble stability in vivo, the present study included individuals with and without patent foramen ovale (PFO) to determine if hyperoxia also eliminates left heart contrast in people with an intracardiac right-to-left shunt. Thirty-two participants consisted of 16 without a PFO; 8 females, 8 with a PFO; 4 females, and 8 with late-appearing left-sided contrast (4 females) completed five, 4-min bouts of constant-load cycle ergometer exercise (males: 250 W, females: 175 W), breathing an [Formula: see text] = 0.21, 0.40, 0.60, 0.80, and 1.00 in a balanced Latin Squares design. QIPAVA was assessed at rest and 3 min into each exercise bout via transthoracic saline contrast echocardiography and our previously used bubble scoring system. Bubble scores at [Formula: see text]= 0.21, 0.40, and 0.60 were unchanged and significantly greater than at [Formula: see text]= 0.80 and 1.00 in those without a PFO. Participants with a PFO had greater bubble scores at [Formula: see text]= 1.00 than those without a PFO. These data suggest that hyperoxia-induced decreases in QIPAVA during exercise occur when [Formula: see text] ≥ 0.80 and is not a result of altered in vivo microbubble dynamics supporting the idea that hyperoxia closes QIPAVA.
Collapse
Affiliation(s)
- James T Davis
- Indiana University School of Medicine, Department of Anatomy, Cell Biology and Physiology Bloomington, Indiana, United States
| | - Jonathan E Elliott
- Veterans Affairs Portland Health Care Systeme, Research Servic, Portland, Oregon, United States
- Department of Neurology, Oregon Health & Science University, Portland, Oregon, United States
| | - Joseph W Duke
- Department of Biological Sciences, Northern Arizona University, Flagstaff, Arizona, United States
| | - Alberto Cristobal
- Department of Human Physiology, University of Oregon, Eugene, Oregon, United States
| | - Andrew T Lovering
- Department of Human Physiology, University of Oregon, Eugene, Oregon, United States
| |
Collapse
|
5
|
Association of CYP2C19 Polymorphic Markers with Cardiovascular Disease Risk Factors in Gas Industry Workers Undergoing Periodic Medical Examinations. High Blood Press Cardiovasc Prev 2023; 30:151-165. [PMID: 36840850 DOI: 10.1007/s40292-023-00567-4] [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: 10/24/2022] [Accepted: 02/08/2023] [Indexed: 02/26/2023] Open
Abstract
INTRODUCTION Human cytochrome P450 (CYP) enzymes have a wide range of endogenous substrates and play a crucial role in cardiovascular physiology as well as in metabolic processes, so the issue of cytochrome P450 genes investigation has received considerable critical attention in the prevention of cardiovascular diseases (CVDs). AIM Comprehensive assessment of relationship between CYP2C19*2, CYP2C19*3 polymorphisms and CVD risk factors in gas industry workers undergoing periodic medical examination (PME). MATERIALS AND METHODS The study included 193 gas industry workers aged 30-55 years without acute diseases as well as exacerbations of chronic diseases, diabetes mellitus, and CVD history. CYP2C19 (rs4244285 and rs4986893) genotyping and analysis of the relationship between CYP2C19*2 and CYP2C19*3 and CVD risk factors were performed. RESULTS The CYP2C19*2 (A) and CYP2C19*3 (A) loss-of-function alleles frequencies were 20% and 2%, respectively. The frequency of high-normal blood pressure (BP) (130-139 and/or 85-89 mm Hg) detection was higher in the CYP2C19*2 (A) subgroup compared with wild-type GG allele carriers (26.7% vs. 5.2%, p = 0.03) in individuals without arterial hypertension (AH) and BP ≥ 140 and/or 90 mm Hg on PME. The median systolic BP levels were 5 mm Hg higher in CYP2C19*2 (A) group than in CYP2C19*2 (GG) group (125 vs. 120 mm Hg, p = 0.01). There was a similar trend for diastolic BP (85 vs. 80 mmHg, p = 0.08). CYP2C19*2 (A) was associated with higher mean levels of both systolic and diastolic BP (p = 0.015 and p = 0.044, respectively) in patients with AH. CYP2C19*2 was not associated with the other CVD risk factors analyzed. CONCLUSION The association of CYP2C19*2 with BP level suggests a possible role of this factor in AH development, which requires further research.
Collapse
|
6
|
Pascale JV, Wolf A, Kadish Y, Diegisser D, Kulaprathazhe MM, Yemane D, Ali S, Kim N, Baruch DE, Yahaya MAF, Dirice E, Adebesin AM, Falck JR, Schwartzman ML, Garcia V. 20-Hydroxyeicosatetraenoic acid (20-HETE): Bioactions, receptors, vascular function, cardiometabolic disease and beyond. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2023; 97:229-255. [PMID: 37236760 PMCID: PMC10683332 DOI: 10.1016/bs.apha.2023.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2023]
Abstract
Vascular function is dynamically regulated and dependent on a bevy of cell types and factors that work in concert across the vasculature. The vasoactive eicosanoid, 20-Hydroxyeicosatetraenoic acid (20-HETE) is a key player in this system influencing the sensitivity of the vasculature to constrictor stimuli, regulating endothelial function, and influencing the renin angiotensin system (RAS), as well as being a driver of vascular remodeling independent of blood pressure elevations. Several of these bioactions are accomplished through the ligand-receptor pairing between 20-HETE and its high-affinity receptor, GPR75. This 20-HETE axis is at the root of various vascular pathologies and processes including ischemia induced angiogenesis, arteriogenesis, septic shock, hypertension, atherosclerosis, myocardial infarction and cardiometabolic diseases including diabetes and insulin resistance. Pharmacologically, several preclinical tools have been developed to disrupt the 20-HETE axis including 20-HETE synthesis inhibitors (DDMS and HET0016), synthetic 20-HETE agonist analogues (20-5,14-HEDE and 20-5,14-HEDGE) and 20-HETE receptor blockers (AAA and 20-SOLA). Systemic or cell-specific therapeutic targeting of the 20-HETE-GPR75 axis continues to be an invaluable approach as studies examine the molecular underpinnings activated by 20-HETE under various physiological settings. In particular, the development and characterization of 20-HETE receptor blockers look to be a promising new class of compounds that can provide a considerable benefit to patients suffering from these cardiovascular pathologies.
Collapse
Affiliation(s)
- Jonathan V Pascale
- Department of Pharmacology, New York Medical College, Valhalla, NY, United States
| | - Alexandra Wolf
- Department of Pharmacology, New York Medical College, Valhalla, NY, United States
| | - Yonaton Kadish
- School of Medicine, New York Medical College, Valhalla, NY, United States
| | - Danielle Diegisser
- Department of Pharmacology, New York Medical College, Valhalla, NY, United States
| | | | - Danait Yemane
- Department of Pharmacology, New York Medical College, Valhalla, NY, United States
| | - Samir Ali
- School of Medicine, New York Medical College, Valhalla, NY, United States
| | - Namhee Kim
- School of Medicine, New York Medical College, Valhalla, NY, United States
| | - David E Baruch
- School of Medicine, New York Medical College, Valhalla, NY, United States
| | - Muhamad Afiq Faisal Yahaya
- Department of Basic Sciences, MAHSA University, Selangor Darul Ehsan, Malaysia; Department of Human Anatomy, Universiti Putra Malaysia (UPM), Selangor Darul Ehsan, Malaysia
| | - Ercument Dirice
- Department of Pharmacology, New York Medical College, Valhalla, NY, United States
| | - Adeniyi M Adebesin
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - John R Falck
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Michal L Schwartzman
- Department of Pharmacology, New York Medical College, Valhalla, NY, United States
| | - Victor Garcia
- Department of Pharmacology, New York Medical College, Valhalla, NY, United States.
| |
Collapse
|
7
|
Houeiss P, Njeim R, Tamim H, Hamdy AF, Azar TS, Azar WS, Noureldein M, Zeidan YH, Rashid A, Azar ST, Eid AA. Urinary 20-HETE: A prospective Non-Invasive prognostic and diagnostic marker for diabetic kidney disease. J Adv Res 2023; 44:109-117. [PMID: 36725183 PMCID: PMC9936418 DOI: 10.1016/j.jare.2022.04.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/25/2021] [Accepted: 04/22/2022] [Indexed: 02/04/2023] Open
Abstract
INTRODUCTION The identification and validation of a non-invasive prognostic marker for early detection of diabetic kidney disease (DKD) can lead to substantial improvement in therapeutic decision-making. OBJECTIVES The main objective of this study is to assess the potential role of the arachidonic acid (AA) metabolite 20-hydroxyeicosatetraenoic (20-HETE) in predicting the incidence and progression of DKD. METHODS Healthy patients and patients with diabetes were recruited from the Hamad General Hospital in Qatar, and urinary 20-HETE levels were measured. Data analysis was done using the Statistical Package for Social Sciences (SPSS). RESULTS Our results show that urinary 20-HETE-to-creatinine (20-HETE/Cr) ratios were significantly elevated in patients with DKD when compared to patients with diabetes who did not exhibit clinical signs of kidney injury (p < 0.001). This correlation was preserved in the multivariate linear regression accounting for age, diabetes, family history of kidney disease, hypertension, dyslipidemia, stroke and metabolic syndrome. Urinary 20-HETE/Cr ratios were also positively correlated with the severity of kidney injury as indicated by albuminuria levels (p < 0.001). A urinary 20-HETE/Cr ratio of 4.6 pmol/mg discriminated between the presence and absence of kidney disease with a sensitivity of 82.2 % and a specificity of 67.1%. More importantly, a 10-unit increase in urinary 20-HETE/Cr ratio was tied to a 10-fold increase in the risk of developing DKD, suggesting a 20-HETE prognostic efficiency. CONCLUSION Taken together, our results suggest that urinary 20-HETE levels can potentially be used as non-invasive diagnostic and prognostic markers for DKD.
Collapse
Affiliation(s)
- Pamela Houeiss
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Lebanon; AUB Diabetes Program, Faculty of Medicine, American University of Beirut, Lebanon
| | - Rachel Njeim
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Lebanon; AUB Diabetes Program, Faculty of Medicine, American University of Beirut, Lebanon
| | - Hani Tamim
- Department of Internal Medicine, Faculty of Medicine, American University of Beirut, Lebanon
| | - Ahmed F Hamdy
- Department of Nephrology, Hamad Medical Corporation, Doha, Qatar
| | - Tanya S Azar
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Lebanon; AUB Diabetes Program, Faculty of Medicine, American University of Beirut, Lebanon
| | - William S Azar
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Lebanon; AUB Diabetes Program, Faculty of Medicine, American University of Beirut, Lebanon; Department of Physiology and Biophysics, Georgetown University School of Medicine, Washington, DC, USA
| | - Mohamed Noureldein
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Lebanon; AUB Diabetes Program, Faculty of Medicine, American University of Beirut, Lebanon
| | - Youssef H Zeidan
- Department of Radiation Oncology, Faculty of Medicine, American University of Beirut, Lebanon
| | - Awad Rashid
- Department of Nephrology, Hamad Medical Corporation, Doha, Qatar
| | - Sami T Azar
- AUB Diabetes Program, Faculty of Medicine, American University of Beirut, Lebanon; Department of Internal Medicine, Faculty of Medicine, American University of Beirut, Lebanon
| | - Assaad A Eid
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Lebanon; AUB Diabetes Program, Faculty of Medicine, American University of Beirut, Lebanon.
| |
Collapse
|
8
|
Balarastaghi S, Rezaee R, Hayes AW, Yarmohammadi F, Karimi G. Mechanisms of Arsenic Exposure-Induced Hypertension and Atherosclerosis: an Updated Overview. Biol Trace Elem Res 2023; 201:98-113. [PMID: 35167029 DOI: 10.1007/s12011-022-03153-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 02/08/2022] [Indexed: 01/11/2023]
Abstract
Arsenic is an abundant element in the earth's crust. In the environment and within the human body, this toxic element can be found in both organic and inorganic forms. Chronic exposure to arsenic can predispose humans to cardiovascular diseases including hypertension, stroke, atherosclerosis, and blackfoot disease. Oxidative damage induced by reactive oxygen species is a major player in arsenic-induced toxicity, and it can affect genes expression, inflammatory responses, and/or nitric oxide homeostasis. Exposure to arsenic in drinking water can lead to vascular endothelial dysfunction which is reflected by an imbalance between vascular relaxation and contraction. Arsenic has been shown to inactivate endothelial nitric oxide synthase leading to a reduction of the generation and bioavailability of nitric oxide. Ultimately, these effects increase the risk of vascular diseases such as hypertension and atherosclerosis. The present article reviews how arsenic exposure contributes to hypertension and atherosclerosis development.
Collapse
Affiliation(s)
- Soudabeh Balarastaghi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ramin Rezaee
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - A Wallace Hayes
- Center for Environmental Occupational Risk Analysis and Management, College of Public Health, University of South Florida, Tampa, FL, USA
| | - Fatemeh Yarmohammadi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gholamreza Karimi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
9
|
Kotlyarov S, Kotlyarova A. Clinical significance of polyunsaturated fatty acids in the prevention of cardiovascular diseases. Front Nutr 2022; 9:998291. [PMID: 36276836 PMCID: PMC9582942 DOI: 10.3389/fnut.2022.998291] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Cardiovascular diseases are one of the most important problems of modern medicine. They are associated with a large number of health care visits, hospitalizations and mortality. Prevention of atherosclerosis is one of the most effective strategies and should start as early as possible. Correction of lipid metabolism disorders is associated with definite clinical successes, both in primary prevention and in the prevention of complications of many cardiovascular diseases. A growing body of evidence suggests a multifaceted role for polyunsaturated fatty acids. They demonstrate a variety of functions in inflammation, both participating directly in a number of cellular processes and acting as a precursor for subsequent biosynthesis of lipid mediators. Extensive clinical data also support the importance of polyunsaturated fatty acids, but all questions have not been answered to date, indicating the need for further research.
Collapse
Affiliation(s)
| | - Anna Kotlyarova
- Department of Pharmacy Management and Economics, Ryazan State Medical University, Ryazan, Russia
| |
Collapse
|
10
|
Yamaguchi A, Botta E, Holinstat M. Eicosanoids in inflammation in the blood and the vessel. Front Pharmacol 2022; 13:997403. [PMID: 36238558 PMCID: PMC9551235 DOI: 10.3389/fphar.2022.997403] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 09/05/2022] [Indexed: 01/14/2023] Open
Abstract
Polyunsaturated fatty acids (PUFAs) are structural components of membrane phospholipids in cells. PUFAs regulate cellular function through the formation of derived lipid mediators termed eicosanoids. The oxygenation of 20-carbon PUFAs via the oxygenases cyclooxygenases, lipoxygenases, or cytochrome P450, generates a class of classical eicosanoids including prostaglandins, thromboxanes and leukotrienes, and also the more recently identified hydroxy-, hydroperoxy-, epoxy- and oxo-eicosanoids, and the specialized pro-resolving (lipid) mediators. These eicosanoids play a critical role in the regulation of inflammation in the blood and the vessel. While arachidonic acid-derived eicosanoids are extensively studied due to their pro-inflammatory effects and therefore involvement in the pathogenesis of inflammatory diseases such as atherosclerosis, diabetes mellitus, hypertension, and the coronavirus disease 2019; in recent years, several eicosanoids have been reported to attenuate exacerbated inflammatory responses and participate in the resolution of inflammation. This review focused on elucidating the biosynthesis and the mechanistic signaling of eicosanoids in inflammation, as well as the pro-inflammatory and anti-inflammatory effects of these eicosanoids in the blood and the vascular wall.
Collapse
Affiliation(s)
- Adriana Yamaguchi
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Eliana Botta
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Michael Holinstat
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, United States,Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor, MI, United States,*Correspondence: Michael Holinstat,
| |
Collapse
|
11
|
Deme P, Moniruzzaman M, Moore D, Heaton R, Ellis R, Letendre S, Haughey N. Association of Plasma Eicosanoid Levels With Immune, Viral, and Cognitive Outcomes in People With HIV. Neurology 2022; 99:e1251-e1264. [PMID: 35851253 PMCID: PMC9576290 DOI: 10.1212/wnl.0000000000200945] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 05/20/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES To determine whether plasma eicosanoid levels are associated with immune, viral, and cognitive outcomes in people with HIV (PWH). METHODS We measured 42 eicosanoids in a longitudinal study of 95 PWH and 25 demographically comparable uninfected participants. Routine clinical chemistry, virologic, immune markers, and a neuropsychological test battery assessing 7 cognitive domains were administered to all participants at 2 study visits over an average of 6.5 months. RESULTS Plasma eicosanoid concentrations were elevated in PWH (n = 95) compared with seronegative controls (n = 25) (100% prediction power at 5% false discovery rate [FDR], α = 0.0531) and were negatively associated with lower current and nadir CD4 lymphocyte counts. Higher levels of eicosanoids were associated with impairments in working memory, verbal fluency, and executive function. Higher plasma viral load was associated with elevated proinflammatory eicosanoids (24% prediction power at 5% FDR and 42.4% prediction power at 10% FDR, α = 0.10). Longitudinal analyses showed that eicosanoid levels were correlated with viral load and with plasma creatinine. Despite associations of eicosanoids with viral loads, elevated plasma eicosanoids were similar in virally suppressed and not fully suppressed PWH. DISCUSSION These data show that HIV infection is associated with a robust production of eicosanoids that are not substantially reduced by antiretroviral therapy (ART). The sustained elevation of these oxylipins in PWH despite ART may contribute to an accelerated aging phenotype that includes earlier than expected brain and peripheral organ damage.
Collapse
Affiliation(s)
- Pragney Deme
- From the Department of Neurology (P.D., M.M., N.H.), Johns Hopkins University School of Medicine, Baltimore, MD; HIV Neurobehavioral Research Program and Departments of Neurosciences and Psychiatry (D.M., R.H., R.E., S.L.), School of Medicine, University of California, San Diego, La Jolla; and the Department of Psychiatry (N.H.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Mohammed Moniruzzaman
- From the Department of Neurology (P.D., M.M., N.H.), Johns Hopkins University School of Medicine, Baltimore, MD; HIV Neurobehavioral Research Program and Departments of Neurosciences and Psychiatry (D.M., R.H., R.E., S.L.), School of Medicine, University of California, San Diego, La Jolla; and the Department of Psychiatry (N.H.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - David Moore
- From the Department of Neurology (P.D., M.M., N.H.), Johns Hopkins University School of Medicine, Baltimore, MD; HIV Neurobehavioral Research Program and Departments of Neurosciences and Psychiatry (D.M., R.H., R.E., S.L.), School of Medicine, University of California, San Diego, La Jolla; and the Department of Psychiatry (N.H.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Robert Heaton
- From the Department of Neurology (P.D., M.M., N.H.), Johns Hopkins University School of Medicine, Baltimore, MD; HIV Neurobehavioral Research Program and Departments of Neurosciences and Psychiatry (D.M., R.H., R.E., S.L.), School of Medicine, University of California, San Diego, La Jolla; and the Department of Psychiatry (N.H.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Ronald Ellis
- From the Department of Neurology (P.D., M.M., N.H.), Johns Hopkins University School of Medicine, Baltimore, MD; HIV Neurobehavioral Research Program and Departments of Neurosciences and Psychiatry (D.M., R.H., R.E., S.L.), School of Medicine, University of California, San Diego, La Jolla; and the Department of Psychiatry (N.H.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Scott Letendre
- From the Department of Neurology (P.D., M.M., N.H.), Johns Hopkins University School of Medicine, Baltimore, MD; HIV Neurobehavioral Research Program and Departments of Neurosciences and Psychiatry (D.M., R.H., R.E., S.L.), School of Medicine, University of California, San Diego, La Jolla; and the Department of Psychiatry (N.H.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Norman Haughey
- From the Department of Neurology (P.D., M.M., N.H.), Johns Hopkins University School of Medicine, Baltimore, MD; HIV Neurobehavioral Research Program and Departments of Neurosciences and Psychiatry (D.M., R.H., R.E., S.L.), School of Medicine, University of California, San Diego, La Jolla; and the Department of Psychiatry (N.H.), Johns Hopkins University School of Medicine, Baltimore, MD.
| |
Collapse
|
12
|
Ciampi F, Gandy J, Ciliberti MG, Sevi A, Albenzio M, Santillo A. Pomegranate (Punica granatum) By-Product Extract Influences the Oxylipids Profile in Primary Bovine Aortic Endothelial Cells in a Model of Oxidative Stress. FRONTIERS IN ANIMAL SCIENCE 2022. [DOI: 10.3389/fanim.2022.837279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Aerobic metabolism produces reactive oxygen species (ROS) as a natural by-product that can play a significant role in cell signaling and homeostasis. Excessive and uncontrolled production of ROS, however, can lead to oxidative stress that causes damage to immune cells and is related to several diseases in dairy cattle. Endothelial cells are essential for optimal immune and inflammatory responses but are especially sensitive to the damaging effects of ROS. Accordingly, investigating antioxidant strategies that can mitigate the detrimental impact of ROS on endothelial functions could impact compromised host defenses that lead to increased disease susceptibility. The objective of this study was to test the antioxidant effect of different concentrations (20, 40, 60, 80 μg/ml) of pomegranate by-product extract (PBE) on bovine aortic endothelial cells (BAECs). A model of oxidative stress was developed using in vitro exposure of BAEC to 2,2′-azobis (2-amidinopropane) dihydrochloride (AAPH) to induce the formation of ROS. The BAEC were then analyzed for cell viability, ROS production, fatty acids profile, and oxylipids formation. The BAECs viability did not change after different concentrations of PBE and remained up to 80% over control; whereas, intracellular ROS showed a reduction passing from 20 to 50% with increasing PBE concentration from 20 to 80 μg/ml, respectively. The PBE extract clearly demonstrated efficacy in reducing the concentrations of pro-inflammatory oxylipids with a concomitant enhancement of anti-inflammatory oxylipids. In particular, the pro-inflammatory 13-hydroxyoctadecadienoic acid and its derived anti-inflammatory 13-hydroperoxoctadecaienoic acid were found lower and higher, respectively, in PBE+AAPH treated cells than AAPH treatment. Data from the present study support in vivo future experimental use of pomegranate by-product extract to study its potential beneficial effect against oxidative stress conditions in dairy cattle.
Collapse
|
13
|
Azcona JA, Tang S, Berry E, Zhang FF, Garvey R, Falck JR, Schwartzman ML, Yi T, Jeitner TM, Guo AM. Neutrophil-Derived Myeloperoxidase and Hypochlorous Acid Critically Contribute to 20-Hydroxyeicosatetraenoic Acid Increases that Drive Postischemic Angiogenesis. J Pharmacol Exp Ther 2022; 381:204-216. [PMID: 35306474 PMCID: PMC9190235 DOI: 10.1124/jpet.121.001036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 03/08/2022] [Indexed: 03/08/2025] Open
Abstract
Compensatory angiogenesis is an important adaptation for recovery from critical ischemia. We recently identified 20-hydroxyeicosatetraenoic acid (20-HETE) as a novel contributor of ischemia-induced angiogenesis. However, the precise mechanisms by which ischemia promotes 20-HETE increases that drive angiogenesis are unknown. This study aims to address the hypothesis that inflammatory neutrophil-derived myeloperoxidase (MPO) and hypochlorous acid (HOCl) critically contribute to 20-HETE increases leading to ischemic angiogenesis. Using Liquid Chromatography-Mass Spectrometry/Mass Spectrometry, Laser Doppler Perfusion Imaging, and Microvascular Density analysis, we found that neutrophil depletion and MPO knockout mitigate angiogenesis and 20-HETE production in the gracilis muscles of mice subjected to hindlimb ischemia. Furthermore, we found MPO and HOCl to be elevated in these tissues postischemia as assessed by immunofluorescence microscopy and in vivo live imaging of HOCl. Next, we demonstrated that the additions of either HOCl or an enzymatic system for generating HOCl to endothelial cells increase the expression of CYP4A11 and its product, 20-HETE. Finally, pharmacological interference of hypoxia inducible factor (HIF) signaling results in ablation of HOCl-induced CYP4A11 transcript and significant reductions in CYP4A11 protein. Collectively, we conclude that neutrophil-derived MPO and its product HOCl activate HIF-1α and CYP4A11 leading to increased 20-HETE production that drives postischemic compensatory angiogenesis. SIGNIFICANCE STATEMENT: Traditionally, neutrophil derived MPO and HOCl are exclusively associated in the innate immunity as potent bactericidal/virucidal factors. The present study establishes a novel paradigm by proposing a unique function for MPO/HOCl as signaling agents that drive critical physiological angiogenesis by activating the CYP4A11-20-HETE signaling axis via a HIF-1α-dependent mechanism. The findings from this study potentially identify novel therapeutic targets for the treatment of ischemia and other diseases associated with abnormal angiogenesis.
Collapse
Affiliation(s)
- Juan A Azcona
- Department of Pharmacology (J.A.A., F.F.Z., M.L.S., A.M.G.), Department of Biochemistry and Molecular Biology (J.A.A., T.M.J., A.M.G.), Department of Pathology (S.T.), and Department of Physiology (E.B.), New York Medical College, Valhalla, New York; Department of Radiology, Weill Cornell Medicine, New York, New York (J.A.A., T.M.J.); Tufts University, Medford, Massachusetts (R.G.); University of Texas Southwestern Medical Center, Dallas, Texas (J.R.F.); and Department of Chemistry, Fudan University, Shanghai, PR China (T.Y.)
| | - Samantha Tang
- Department of Pharmacology (J.A.A., F.F.Z., M.L.S., A.M.G.), Department of Biochemistry and Molecular Biology (J.A.A., T.M.J., A.M.G.), Department of Pathology (S.T.), and Department of Physiology (E.B.), New York Medical College, Valhalla, New York; Department of Radiology, Weill Cornell Medicine, New York, New York (J.A.A., T.M.J.); Tufts University, Medford, Massachusetts (R.G.); University of Texas Southwestern Medical Center, Dallas, Texas (J.R.F.); and Department of Chemistry, Fudan University, Shanghai, PR China (T.Y.)
| | - Elizabeth Berry
- Department of Pharmacology (J.A.A., F.F.Z., M.L.S., A.M.G.), Department of Biochemistry and Molecular Biology (J.A.A., T.M.J., A.M.G.), Department of Pathology (S.T.), and Department of Physiology (E.B.), New York Medical College, Valhalla, New York; Department of Radiology, Weill Cornell Medicine, New York, New York (J.A.A., T.M.J.); Tufts University, Medford, Massachusetts (R.G.); University of Texas Southwestern Medical Center, Dallas, Texas (J.R.F.); and Department of Chemistry, Fudan University, Shanghai, PR China (T.Y.)
| | - Frank F Zhang
- Department of Pharmacology (J.A.A., F.F.Z., M.L.S., A.M.G.), Department of Biochemistry and Molecular Biology (J.A.A., T.M.J., A.M.G.), Department of Pathology (S.T.), and Department of Physiology (E.B.), New York Medical College, Valhalla, New York; Department of Radiology, Weill Cornell Medicine, New York, New York (J.A.A., T.M.J.); Tufts University, Medford, Massachusetts (R.G.); University of Texas Southwestern Medical Center, Dallas, Texas (J.R.F.); and Department of Chemistry, Fudan University, Shanghai, PR China (T.Y.)
| | - Radha Garvey
- Department of Pharmacology (J.A.A., F.F.Z., M.L.S., A.M.G.), Department of Biochemistry and Molecular Biology (J.A.A., T.M.J., A.M.G.), Department of Pathology (S.T.), and Department of Physiology (E.B.), New York Medical College, Valhalla, New York; Department of Radiology, Weill Cornell Medicine, New York, New York (J.A.A., T.M.J.); Tufts University, Medford, Massachusetts (R.G.); University of Texas Southwestern Medical Center, Dallas, Texas (J.R.F.); and Department of Chemistry, Fudan University, Shanghai, PR China (T.Y.)
| | - John R Falck
- Department of Pharmacology (J.A.A., F.F.Z., M.L.S., A.M.G.), Department of Biochemistry and Molecular Biology (J.A.A., T.M.J., A.M.G.), Department of Pathology (S.T.), and Department of Physiology (E.B.), New York Medical College, Valhalla, New York; Department of Radiology, Weill Cornell Medicine, New York, New York (J.A.A., T.M.J.); Tufts University, Medford, Massachusetts (R.G.); University of Texas Southwestern Medical Center, Dallas, Texas (J.R.F.); and Department of Chemistry, Fudan University, Shanghai, PR China (T.Y.)
| | - Michal Laniado Schwartzman
- Department of Pharmacology (J.A.A., F.F.Z., M.L.S., A.M.G.), Department of Biochemistry and Molecular Biology (J.A.A., T.M.J., A.M.G.), Department of Pathology (S.T.), and Department of Physiology (E.B.), New York Medical College, Valhalla, New York; Department of Radiology, Weill Cornell Medicine, New York, New York (J.A.A., T.M.J.); Tufts University, Medford, Massachusetts (R.G.); University of Texas Southwestern Medical Center, Dallas, Texas (J.R.F.); and Department of Chemistry, Fudan University, Shanghai, PR China (T.Y.)
| | - Tao Yi
- Department of Pharmacology (J.A.A., F.F.Z., M.L.S., A.M.G.), Department of Biochemistry and Molecular Biology (J.A.A., T.M.J., A.M.G.), Department of Pathology (S.T.), and Department of Physiology (E.B.), New York Medical College, Valhalla, New York; Department of Radiology, Weill Cornell Medicine, New York, New York (J.A.A., T.M.J.); Tufts University, Medford, Massachusetts (R.G.); University of Texas Southwestern Medical Center, Dallas, Texas (J.R.F.); and Department of Chemistry, Fudan University, Shanghai, PR China (T.Y.)
| | - Thomas M Jeitner
- Department of Pharmacology (J.A.A., F.F.Z., M.L.S., A.M.G.), Department of Biochemistry and Molecular Biology (J.A.A., T.M.J., A.M.G.), Department of Pathology (S.T.), and Department of Physiology (E.B.), New York Medical College, Valhalla, New York; Department of Radiology, Weill Cornell Medicine, New York, New York (J.A.A., T.M.J.); Tufts University, Medford, Massachusetts (R.G.); University of Texas Southwestern Medical Center, Dallas, Texas (J.R.F.); and Department of Chemistry, Fudan University, Shanghai, PR China (T.Y.)
| | - Austin M Guo
- Department of Pharmacology (J.A.A., F.F.Z., M.L.S., A.M.G.), Department of Biochemistry and Molecular Biology (J.A.A., T.M.J., A.M.G.), Department of Pathology (S.T.), and Department of Physiology (E.B.), New York Medical College, Valhalla, New York; Department of Radiology, Weill Cornell Medicine, New York, New York (J.A.A., T.M.J.); Tufts University, Medford, Massachusetts (R.G.); University of Texas Southwestern Medical Center, Dallas, Texas (J.R.F.); and Department of Chemistry, Fudan University, Shanghai, PR China (T.Y.)
| |
Collapse
|
14
|
Lipidomics in Understanding Pathophysiology and Pharmacologic Effects in Inflammatory Diseases: Considerations for Drug Development. Metabolites 2022; 12:metabo12040333. [PMID: 35448520 PMCID: PMC9030008 DOI: 10.3390/metabo12040333] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 03/29/2022] [Accepted: 04/04/2022] [Indexed: 01/26/2023] Open
Abstract
The lipidome has a broad range of biological and signaling functions, including serving as a structural scaffold for membranes and initiating and resolving inflammation. To investigate the biological activity of phospholipids and their bioactive metabolites, precise analytical techniques are necessary to identify specific lipids and quantify their levels. Simultaneous quantification of a set of lipids can be achieved using high sensitivity mass spectrometry (MS) techniques, whose technological advancements have significantly improved over the last decade. This has unlocked the power of metabolomics/lipidomics allowing the dynamic characterization of metabolic systems. Lipidomics is a subset of metabolomics for multianalyte identification and quantification of endogenous lipids and their metabolites. Lipidomics-based technology has the potential to drive novel biomarker discovery and therapeutic development programs; however, appropriate standards have not been established for the field. Standardization would improve lipidomic analyses and accelerate the development of innovative therapies. This review aims to summarize considerations for lipidomic study designs including instrumentation, sample stabilization, data validation, and data analysis. In addition, this review highlights how lipidomics can be applied to biomarker discovery and drug mechanism dissection in various inflammatory diseases including cardiovascular disease, neurodegeneration, lung disease, and autoimmune disease.
Collapse
|
15
|
Isse FA, El-Sherbeni AA, El-Kadi AOS. The multifaceted role of cytochrome P450-Derived arachidonic acid metabolites in diabetes and diabetic cardiomyopathy. Drug Metab Rev 2022; 54:141-160. [PMID: 35306928 DOI: 10.1080/03602532.2022.2051045] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Understanding lipid metabolism is a critical key to understanding the pathogenesis of Diabetes Mellitus (DM). It is known that 60-90% of DM patients are obese or used to be obese. The incidence of obesity is rising owing to the modern sedentary lifestyle that leads to insulin resistance and increased levels of free fatty acids, predisposing tissues to utilize more lipids with less glucose uptake. However, the exact mechanism is not yet fully elucidated. Diabetic cardiomyopathy seems to be associated with these alterations in lipid metabolism. Arachidonic acid (AA) is an important fatty acid that is metabolized to several bioactive compounds by cyclooxygenases, lipoxygenases, and the more recently discovered, cytochrome P450 (P450) enzymes. P450 metabolizes AA to either epoxy-AA (EETs) or hydroxy-AA (HETEs). Studies showed that EETs could have cardioprotective effects and beneficial effects in reversing abnormalities in glucose and insulin homeostasis. Conversely, HETEs, most importantly 12-HETE and 20-HETE, were found to interfere with normal glucose and insulin homeostasis and thus, might be involved in diabetic cardiomyopathy. In this review, we highlight the role of P450-derived AA metabolites in the context of DM and diabetic cardiomyopathy and their potential use as a target for developing new treatments for DM and diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Fadumo Ahmed Isse
- Departmet of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
| | - Ahmed A El-Sherbeni
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Ayman O S El-Kadi
- Departmet of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
| |
Collapse
|
16
|
Kotlyarov S, Kotlyarova A. Involvement of Fatty Acids and Their Metabolites in the Development of Inflammation in Atherosclerosis. Int J Mol Sci 2022; 23:1308. [PMID: 35163232 PMCID: PMC8835729 DOI: 10.3390/ijms23031308] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/14/2022] [Accepted: 01/21/2022] [Indexed: 02/06/2023] Open
Abstract
Despite all the advances of modern medicine, atherosclerosis continues to be one of the most important medical and social problems. Atherosclerosis is the cause of several cardiovascular diseases, which are associated with high rates of disability and mortality. The development of atherosclerosis is associated with the accumulation of lipids in the arterial intima and the disruption of mechanisms that maintain the balance between the development and resolution of inflammation. Fatty acids are involved in many mechanisms of inflammation development and maintenance. Endothelial cells demonstrate multiple cross-linkages between lipid metabolism and innate immunity. In addition, these processes are linked to hemodynamics and the function of other cells in the vascular wall, highlighting the central role of the endothelium in vascular biology.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University, 390026 Ryazan, Russia
| | - Anna Kotlyarova
- Department of Pharmacology and Pharmacy, Ryazan State Medical University, 390026 Ryazan, Russia;
| |
Collapse
|
17
|
Froogh G, Garcia V, Laniado Schwartzman M. The CYP/20-HETE/GPR75 axis in hypertension. ADVANCES IN PHARMACOLOGY 2022; 94:1-25. [PMID: 35659370 PMCID: PMC10123763 DOI: 10.1016/bs.apha.2022.02.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
20-Hydroxyeicosatetraenoic acid (20-HETE) is a bioactive lipid generated from the ω-hydroxylation of arachidonic acid (AA) by enzymes of the cytochrome P450 (CYP) family, primarily the CYP4A and CYP4F subfamilies. 20-HETE is most notably identified as a modulator of vascular tone, regulator of renal function, and a contributor to the onset and development of hypertension and cardiovascular disease. 20-HETE-mediated signaling promotes hypertension by sensitizing the vasculature to constrictor stimuli, inducing endothelial dysfunction, and potentiating vascular inflammation. These bioactions are driven by the activation of the G-protein coupled receptor 75 (GPR75), a 20-HETE receptor (20HR). Given the capacity of 20-HETE signaling to drive pro-hypertensive mechanisms, the CYP/20-HETE/GPR75 axis has the potential to be a significant therapeutic target for the treatment of hypertension and cardiovascular diseases associated with increases in blood pressure. In this chapter, we review 20-HETE-mediated cellular mechanisms that promote hypertension, highlight important data in humans such as genetic variants in the CYP genes that potentiate 20-HETE production and describe recent findings in humans with 20HR/GPR75 mutations. Special emphasis is given to the 20HR and respective receptor blockers that have the potential to pave a path to translational and clinical studies for the treatment of 20-HETE-driven hypertension, and obesity/metabolic syndrome.
Collapse
|
18
|
Gerges SH, El-Kadi AOS. Sex differences in eicosanoid formation and metabolism: A possible mediator of sex discrepancies in cardiovascular diseases. Pharmacol Ther 2021; 234:108046. [PMID: 34808133 DOI: 10.1016/j.pharmthera.2021.108046] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/07/2021] [Accepted: 11/16/2021] [Indexed: 12/14/2022]
Abstract
Arachidonic acid is metabolized by cyclooxygenase, lipoxygenase, and cytochrome P450 enzymes to produce prostaglandins, leukotrienes, epoxyeicosatrienoic acids (EETs), and hydroxyeicosatetraenoic acids (HETEs), along with other eicosanoids. Eicosanoids have important physiological and pathological roles in the body, including the cardiovascular system. Evidence from several experimental and clinical studies indicates differences in eicosanoid levels, as well as in the activity or expression levels of their synthesizing and metabolizing enzymes between males and females. In addition, there is a clear state of gender specificity in cardiovascular diseases (CVD), which tend to be more common in men compared to women, and their risk increases significantly in postmenopausal women compared to younger women. This could be largely attributed to sex hormones, as androgens exert detrimental effects on the heart and blood vessels, whereas estrogen exhibits cardioprotective effects. Many of androgen and estrogen effects on the cardiovascular system are mediated by eicosanoids. For example, androgens increase the levels of cardiotoxic eicosanoids like 20-HETE, while estrogens increase the levels of cardioprotective EETs. Thus, sex differences in eicosanoid levels in the cardiovascular system could be an important underlying mechanism for the different effects of sex hormones and the differences in CVD between males and females. Understanding the role of eicosanoids in these differences can help improve the management of CVD.
Collapse
Affiliation(s)
- Samar H Gerges
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Ayman O S El-Kadi
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
19
|
Zhou Y, Khan H, Xiao J, Cheang WS. Effects of Arachidonic Acid Metabolites on Cardiovascular Health and Disease. Int J Mol Sci 2021; 22:12029. [PMID: 34769460 PMCID: PMC8584625 DOI: 10.3390/ijms222112029] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 10/29/2021] [Accepted: 11/04/2021] [Indexed: 02/06/2023] Open
Abstract
Arachidonic acid (AA) is an essential fatty acid that is released by phospholipids in cell membranes and metabolized by cyclooxygenase (COX), cytochrome P450 (CYP) enzymes, and lipid oxygenase (LOX) pathways to regulate complex cardiovascular function under physiological and pathological conditions. Various AA metabolites include prostaglandins, prostacyclin, thromboxanes, hydroxyeicosatetraenoic acids, leukotrienes, lipoxins, and epoxyeicosatrienoic acids. The AA metabolites play important and differential roles in the modulation of vascular tone, and cardiovascular complications including atherosclerosis, hypertension, and myocardial infarction upon actions to different receptors and vascular beds. This article reviews the roles of AA metabolism in cardiovascular health and disease as well as their potential therapeutic implication.
Collapse
Affiliation(s)
- Yan Zhou
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China;
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan 23200, Pakistan;
| | - Jianbo Xiao
- Department of Analytical Chemistry and Food Science, Faculty of Food Science and Technology, University of Vigo, 36310 Vigo, Spain;
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China
| | - Wai San Cheang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China;
| |
Collapse
|
20
|
Ma F, Luo L, Gao X. Metabolite and transcriptome analyses revealed the modulation of fructo-oligosaccharide on ileum metabolism of Taiping chickens. J Appl Microbiol 2021; 132:2249-2261. [PMID: 34608718 DOI: 10.1111/jam.15319] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 09/29/2021] [Accepted: 10/01/2021] [Indexed: 01/08/2023]
Abstract
AIM The metabolic markers and differentially expressed genes (DEGs) related to fructo-oligosaccharide (FOS) were screened, and the response of FOS to the ileum metabolic pathway of Taiping chickens was analysed. METHODS AND RESULTS Prebiotic are widely used in agricultural breeding for care and maintenance of animal health, especially FOS. Metabonomics evaluation of ileum of Taiping chicken ultra-performance liquid chromatography-quadruple time of-flight high-sensitivity mass spectrometry showed that 93 differentially altered metabolites were identified and divided into eight categories, of which organic acids and derivatives was the most important one. Transcriptomic analysis showed that DEGs were mainly enriched in drug metabolism-cytochrome p450, metabolism of xenobiotics by cytochrome p450, retinol metabolism and fat digestion and absorption. Integrated analysis of metabolite profiles and gene expression revealed that the significantly up-regulated GSTT1 was significantly correlated with most of the different lipid metabolites, suggesting that GSTT1 may play an important role in FOS regulation of lipid metabolism. CONCLUSIONS The results of this study suggest that supplementation of FOS can have a positive effect on gut metabolites, which may contribute to the overall health with indigenous chickens. SIGNIFICANCE AND IMPACT OF THE STUDY Insight into the responses of intestinal prebiotics of Taiping chicken is helpful to understand the role of prebiotics in maintaining intestinal microflora balance and improving immune response and productivity of poultry from the molecular and metabolic levels.
Collapse
Affiliation(s)
- Fang Ma
- Key Laboratory of Resource Utilization of Agricultural Solid Waste in Gansu Province, Tianshui Normal University, Tianshui, China
| | - Lintong Luo
- Key Laboratory of Resource Utilization of Agricultural Solid Waste in Gansu Province, Tianshui Normal University, Tianshui, China
| | - Xiang Gao
- Key Laboratory of Resource Utilization of Agricultural Solid Waste in Gansu Province, Tianshui Normal University, Tianshui, China
| |
Collapse
|
21
|
Shraim BA, Moursi MO, Benter IF, Habib AM, Akhtar S. The Role of Epidermal Growth Factor Receptor Family of Receptor Tyrosine Kinases in Mediating Diabetes-Induced Cardiovascular Complications. Front Pharmacol 2021; 12:701390. [PMID: 34408653 PMCID: PMC8365470 DOI: 10.3389/fphar.2021.701390] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 07/14/2021] [Indexed: 12/15/2022] Open
Abstract
Diabetes mellitus is a major debilitating disease whose global incidence is progressively increasing with currently over 463 million adult sufferers and this figure will likely reach over 700 million by the year 2045. It is the complications of diabetes such as cardiovascular, renal, neuronal and ocular dysfunction that lead to increased patient morbidity and mortality. Of these, cardiovascular complications that can result in stroke and cardiomyopathies are 2- to 5-fold more likely in diabetes but the underlying mechanisms involved in their development are not fully understood. Emerging research suggests that members of the Epidermal Growth Factor Receptor (EGFR/ErbB/HER) family of tyrosine kinases can have a dual role in that they are beneficially required for normal development and physiological functioning of the cardiovascular system (CVS) as well as in salvage pathways following acute cardiac ischemia/reperfusion injury but their chronic dysregulation may also be intricately involved in mediating diabetes-induced cardiovascular pathologies. Here we review the evidence for EGFR/ErbB/HER receptors in mediating these dual roles in the CVS and also discuss their potential interplay with the Renin-Angiotensin-Aldosterone System heptapeptide, Angiotensin-(1-7), as well the arachidonic acid metabolite, 20-HETE (20-hydroxy-5, 8, 11, 14-eicosatetraenoic acid). A greater understanding of the multi-faceted roles of EGFR/ErbB/HER family of tyrosine kinases and their interplay with other key modulators of cardiovascular function could facilitate the development of novel therapeutic strategies for treating diabetes-induced cardiovascular complications.
Collapse
Affiliation(s)
- Bara A Shraim
- College of Medicine, QU Health, Qatar University, Doha, Qatar.,Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| | - Moaz O Moursi
- College of Medicine, QU Health, Qatar University, Doha, Qatar.,Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| | - Ibrahim F Benter
- Faculty of Medicine, Eastern Mediterranean University, Famagusta, North Cyprus
| | - Abdella M Habib
- College of Medicine, QU Health, Qatar University, Doha, Qatar.,Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| | - Saghir Akhtar
- College of Medicine, QU Health, Qatar University, Doha, Qatar.,Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
22
|
Chhonker YS, Kanvinde S, Ahmad R, Singh AB, Oupický D, Murry DJ. Simultaneous Quantitation of Lipid Biomarkers for Inflammatory Bowel Disease Using LC-MS/MS. Metabolites 2021; 11:106. [PMID: 33673198 PMCID: PMC7918109 DOI: 10.3390/metabo11020106] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 01/31/2021] [Accepted: 02/01/2021] [Indexed: 12/30/2022] Open
Abstract
Eicosanoids are key mediators and regulators of inflammation and oxidative stress that are often used as biomarkers for severity and therapeutic responses in various diseases. We here report a highly sensitive LC-MS/MS method for the simultaneous quantification of at least 66 key eicosanoids in a widely used murine model of colitis. Chromatographic separation was achieved with Shim-Pack XR-ODSIII, 150 × 2.00 mm, 2.2 µm. The mobile phase was operated in gradient conditions and consisted of acetonitrile and 0.1% acetic acid in water with a total flow of 0.37 mL/min. This method is sensitive, with a limit of quantification ranging from 0.01 to 1 ng/mL for the various analytes, has a large dynamic range (200 ng/mL), and a total run time of 25 min. The inter- and intraday accuracy (85-115%), precision (≥85%), and recovery (40-90%) met the acceptance criteria per the US Food and Drug Administration guidelines. This method was successfully applied to evaluate eicosanoid metabolites in mice subjected to colitis versus untreated, healthy control mice. In summary, we developed a highly sensitive and fast LC-MS/MS method that can be used to identify biomarkers for inflammation and potentially help in prognosis of the disease in inflammatory bowel disease (IBD) patients, including the response to therapy.
Collapse
Affiliation(s)
- Yashpal S. Chhonker
- Clinical Pharmacology Laboratory, Department of Pharmacy Practice and Science, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Shrey Kanvinde
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198, USA; (S.K.); (D.O.)
| | - Rizwan Ahmad
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (R.A.); (A.B.S.)
| | - Amar B. Singh
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (R.A.); (A.B.S.)
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
- VA Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
| | - David Oupický
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198, USA; (S.K.); (D.O.)
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Daryl J. Murry
- Clinical Pharmacology Laboratory, Department of Pharmacy Practice and Science, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198, USA;
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
23
|
Zong L, Gao R, Guo Z, Shao Z, Wang Y, Eser BE. Characterization and modification of two self-sufficient CYP102 family enzymes from Bacillus amyloliquefaciens DSM 7 with distinct regioselectivity towards fatty acid hydroxylation. Biochem Eng J 2021. [DOI: 10.1016/j.bej.2020.107871] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
24
|
Hariharan A, Weir N, Robertson C, He L, Betsholtz C, Longden TA. The Ion Channel and GPCR Toolkit of Brain Capillary Pericytes. Front Cell Neurosci 2020; 14:601324. [PMID: 33390906 PMCID: PMC7775489 DOI: 10.3389/fncel.2020.601324] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/13/2020] [Indexed: 12/14/2022] Open
Abstract
Brain pericytes reside on the abluminal surface of capillaries, and their processes cover ~90% of the length of the capillary bed. These cells were first described almost 150 years ago (Eberth, 1871; Rouget, 1873) and have been the subject of intense experimental scrutiny in recent years, but their physiological roles remain uncertain and little is known of the complement of signaling elements that they employ to carry out their functions. In this review, we synthesize functional data with single-cell RNAseq screens to explore the ion channel and G protein-coupled receptor (GPCR) toolkit of mesh and thin-strand pericytes of the brain, with the aim of providing a framework for deeper explorations of the molecular mechanisms that govern pericyte physiology. We argue that their complement of channels and receptors ideally positions capillary pericytes to play a central role in adapting blood flow to meet the challenge of satisfying neuronal energy requirements from deep within the capillary bed, by enabling dynamic regulation of their membrane potential to influence the electrical output of the cell. In particular, we outline how genetic and functional evidence suggest an important role for Gs-coupled GPCRs and ATP-sensitive potassium (KATP) channels in this context. We put forth a predictive model for long-range hyperpolarizing electrical signaling from pericytes to upstream arterioles, and detail the TRP and Ca2+ channels and Gq, Gi/o, and G12/13 signaling processes that counterbalance this. We underscore critical questions that need to be addressed to further advance our understanding of the signaling topology of capillary pericytes, and how this contributes to their physiological roles and their dysfunction in disease.
Collapse
Affiliation(s)
- Ashwini Hariharan
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Nick Weir
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Colin Robertson
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Liqun He
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Christer Betsholtz
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.,Department of Medicine Huddinge (MedH), Karolinska Institutet & Integrated Cardio Metabolic Centre, Huddinge, Sweden
| | - Thomas A Longden
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, United States
| |
Collapse
|
25
|
Tian S, Zhou X, Phuntsok T, Zhao N, Zhang D, Ning C, Li D, Zhao H. Genomic Analyses Reveal Genetic Adaptations to Tropical Climates in Chickens. iScience 2020; 23:101644. [PMID: 33103083 PMCID: PMC7578744 DOI: 10.1016/j.isci.2020.101644] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 08/19/2020] [Accepted: 09/30/2020] [Indexed: 12/05/2022] Open
Abstract
The genetic footprints of adaptations to naturally occurring tropical stress along with domestication are poorly reported in chickens. Here, by conducting population genomic analyses of 67 chickens inhabiting distinct climates, we found signals of gene flow from Tibetan chickens to Sri Lankan and Saudi Arabian breeds and identified 12 positively selected genes that are likely involved in genetic adaptations to both tropical desert and tropical monsoon island climates. Notably, in tropical desert climate, advantageous alleles of TLR7 and ZC3HAV1, which could inhibit replication of viruses in cells, suggest immune adaptation to the defense against zoonotic diseases in chickens. Furthermore, comparative genomic analysis showed that four genes (OC90, PLA2G12B, GPR17 and TNFRSF11A) involved in arachidonic acid metabolism have undergone convergent adaptation to tropical desert climate between birds and mammals. Our study offers insights into the genetic mechanisms of adaptations to tropical climates in birds and other animals and provides practical value for breeding design and medical research on avian viruses.
Collapse
Affiliation(s)
- Shilin Tian
- Department of Ecology, Tibetan Centre for Ecology and Conservation at WHU-TU, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, 299 Bayi Road, Wuhan, Hubei 430072, China
| | - Xuming Zhou
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Beijing 100101, China
| | - Tashi Phuntsok
- Laboratory of Extreme Environmental Biological Resources and Adaptive Evolution, Research Center for Ecology, College of Science, Tibet University, Lhasa 850000, China
| | - Ning Zhao
- Laboratory of Extreme Environmental Biological Resources and Adaptive Evolution, Research Center for Ecology, College of Science, Tibet University, Lhasa 850000, China
| | - Dejing Zhang
- Novogene Bioinformatics Institute, Beijing 100015, China
| | - Chunyou Ning
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Diyan Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Huabin Zhao
- Department of Ecology, Tibetan Centre for Ecology and Conservation at WHU-TU, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, 299 Bayi Road, Wuhan, Hubei 430072, China
- Laboratory of Extreme Environmental Biological Resources and Adaptive Evolution, Research Center for Ecology, College of Science, Tibet University, Lhasa 850000, China
| |
Collapse
|
26
|
Investigation of the content differences of arachidonic acid metabolites in a mouse model of breast cancer by using LC-MS/MS. J Pharm Biomed Anal 2020; 194:113763. [PMID: 33279296 DOI: 10.1016/j.jpba.2020.113763] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 11/02/2020] [Accepted: 11/09/2020] [Indexed: 11/21/2022]
Abstract
Arachidonic acid (AA) is closely associated with breast cancer. In addition to the two metabolic pathways regulated by cyclooxygenase and lipoxygenase, AA has a third metabolic pathway through which cytochrome P450 (CYP) enzymes produce hydroxyeicosatetraenoic acids (HETEs) and epoxyeicosatrienoic acids (EETs). The targeted CYP-mediated pathway of AA can not only kill cancer cells but also inhibit the interstitial microenvironment around a tumor. Therefore, it makes sense to identify potential biomarkers from the AA metabolome for the diagnosis and treatment of breast cancer. This study established a liquid chromatography-tandem mass spectrometry (LC-MS/MS) method for the analysis of AA and its main metabolites, EETs and HETEs, in MMTV-PyMT mice, a spontaneous breast cancer mouse model. The results showed that there were significant differences in the concentrations of AA, 12-HETE, 19-HETE and 8,9-EET in plasma and tumor tissues between normal and MMTV-PyMT mice. Therefore, the eicosanoids mentioned above may be used as new biomarkers for breast cancer diagnosis. This study provides a new perspective for the recognition and diagnosis of breast cancer.
Collapse
|
27
|
Ababaikeri B, Abduriyim S, Tohetahong Y, Mamat T, Ahmat A, Halik M. Whole-genome sequencing of Tarim red deer ( Cervus elaphus yarkandensis) reveals demographic history and adaptations to an arid-desert environment. Front Zool 2020; 17:31. [PMID: 33072165 PMCID: PMC7565370 DOI: 10.1186/s12983-020-00379-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 10/02/2020] [Indexed: 01/08/2023] Open
Abstract
Background The initiation of desert conditions in the Tarim Basin in China since the late Miocene has led to the significant genetic structuring of local organisms. Tarim Red Deer (Cervus elaphus yarkandensis, TRD) have adapted to the harsh environmental conditions in this basin, including high solar radiation and temperature, aridity, and poor nutritional conditions. However, the underlying genetic basis of this adaptation is poorly understood. Results We sequenced the whole genomes of 13 TRD individuals, conducted comparative genomic analyses, and estimated demographic fluctuation. The ∂a∂i model estimated that the TRD and Tule elk (Cervus canadensis nannodes) populations diverged approximately 0.98 Mya. Analyses revealed a substantial influence of the Earth’s climate on the effective population size of TRD, associated with glacial advances and retreat, and human activities likely underlie a recent serious decline in population. A marked bottleneck may have profoundly affected the genetic diversity of TRD populations. We detected a set of candidate genes, pathways, and GO categories related to oxidative stress, water reabsorption, immune regulation, energy metabolism, eye protection, heat stress, respiratory system adaptation, prevention of high blood pressure, and DNA damage and repair that may directly or indirectly be involved in the adaptation of TRD to an arid-desert environment. Conclusions Our analyses highlight the role of historical global climates in the population dynamics of TRD. In light of ongoing global warming and the increasing incidence of droughts, our study offers insights into the genomic adaptations of animals, especially TRD, to extreme arid-desert environments and provides a valuable resource for future research on conservation design and biological adaptations to environmental change.
Collapse
Affiliation(s)
- Buweihailiqiemu Ababaikeri
- College of Life Sciences and Technology, Xinjiang University, Urumqi, 830046 Xinjiang China.,College of Xinjiang Uyghur Medicine, Hoten, 848000 Xinjiang China
| | - Shamshidin Abduriyim
- College of Life Science, Shihezi University, Shihezi, 832003 Xinjiang China.,Department of Ecology, Hubei Key Laboratory of Cell Homeostasis, College of Life Science, Wuhan University, Wuhan, 430072 Hubei China
| | - Yilamujiang Tohetahong
- College of Life Sciences and Technology, Xinjiang University, Urumqi, 830046 Xinjiang China
| | - Tayerjan Mamat
- College of Life Sciences and Technology, Xinjiang University, Urumqi, 830046 Xinjiang China
| | - Adil Ahmat
- College of Life Sciences and Technology, Xinjiang University, Urumqi, 830046 Xinjiang China
| | - Mahmut Halik
- College of Life Sciences and Technology, Xinjiang University, Urumqi, 830046 Xinjiang China
| |
Collapse
|
28
|
Gilani A, Agostinucci K, Pascale JV, Hossain S, Kandhi S, Pandey V, Garcia V, Nasjletti A, Laniado Schwartzman M. Proximal tubular-targeted overexpression of the Cyp4a12-20-HETE synthase promotes salt-sensitive hypertension in male mice. Am J Physiol Regul Integr Comp Physiol 2020; 319:R87-R95. [PMID: 32633545 PMCID: PMC7468799 DOI: 10.1152/ajpregu.00089.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/01/2020] [Accepted: 06/09/2020] [Indexed: 12/23/2022]
Abstract
20-Hydroxyeicosatetraenoic acid (20-HETE) has been linked to blood pressure (BP) regulation via actions on the renal microvasculature and tubules. We assessed tubular 20-HETE contribution to hypertension by generating transgenic mice overexpressing the CYP4A12-20-HETE synthase (PT-4a12 mice) under the control of the proximal tubule (PT)-specific promoter, phosphoenolpyruvate carboxykinase (PEPCK). 20-HETE levels in the kidney cortex of male (967±210 vs. 249±69 pg/mg protein), but not female (121±15 vs. 92±11 pg/mg protein) PT-4a12 mice, showed a 2.5-fold increase compared to WT. Renal cortical Cyp4a12 mRNA and CYP4A12 protein in male, but not female PT-4a12 mice increased by 2-3-fold compared to WT. Male PT-4a12 mice displayed elevated BP (142±1 vs. 111±4 mmHg, p<0.0001), whereas BP in females PT-4a12 mice was not significantly different from WT (118±2 vs. 117±2 mmHg; p=0.98). In male PT-4a12 mice, BP decreased when transitioned from a control salt (0.4%) to a low-salt diet (0.075%) from 135±4 to 120±6 mmHg (p<0.01) and increased to 153±5 mmHg (p<0.05) when placed on a high-salt diet (4%). Female PT-4a12 mice did not show changes in BP on either low- or high-salt diet. In conclusion, the expression of Cyp4a12 driven by the PEPCK promoter is sex-specific probably due to its X-linkage. The salt-sensitive hypertension seen in PT-4a12 male mice suggests a potential anti-natriuretic activity of 20-HETE that needs to be further explored.
Collapse
Affiliation(s)
- Ankit Gilani
- Pharmacology, New York Medical College, United States
| | | | | | - Sakib Hossain
- Pharmacology, New York Medical College, United States
| | | | | | | | | | | |
Collapse
|
29
|
The effect of ω-3 polyunsaturated fatty acids on the liver lipidome, proteome and bile acid profile: parenteral versus enteral administration. Sci Rep 2019; 9:19097. [PMID: 31836843 PMCID: PMC6910966 DOI: 10.1038/s41598-019-54225-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 11/10/2019] [Indexed: 12/19/2022] Open
Abstract
Parenteral nutrition (PN) is often associated with the deterioration of liver functions (PNALD). Omega-3 polyunsaturated fatty acids (PUFA) were reported to alleviate PNALD but the underlying mechanisms have not been fully unraveled yet. Using omics´ approach, we determined serum and liver lipidome, liver proteome, and liver bile acid profile as well as markers of inflammation and oxidative stress in rats administered either ω-6 PUFA based lipid emulsion (Intralipid) or ω-6/ω-3 PUFA blend (Intralipid/Omegaven) via the enteral or parenteral route. In general, we found that enteral administration of both lipid emulsions has less impact on the liver than the parenteral route. Compared with parenterally administered Intralipid, PN administration of ω-3 PUFA was associated with 1. increased content of eicosapentaenoic (EPA)- and docosahexaenoic (DHA) acids-containing lipid species; 2. higher abundance of CYP4A isoenzymes capable of bioactive lipid synthesis and the increased content of their potential products (oxidized EPA and DHA); 3. downregulation of enzymes involved CYP450 drug metabolism what may represent an adaptive mechanism counteracting the potential negative effects (enhanced ROS production) of PUFA metabolism; 4. normalized anti-oxidative capacity and 5. physiological BAs spectrum. All these findings may contribute to the explanation of ω-3 PUFA protective effects in the context of PN.
Collapse
|
30
|
Guan S, Jia B, Chao K, Zhu X, Tang J, Li M, Wu L, Xing L, Liu K, Zhang L, Wang X, Gao X, Huang M. UPLC–QTOF-MS-Based Plasma Lipidomic Profiling Reveals Biomarkers for Inflammatory Bowel Disease Diagnosis. J Proteome Res 2019; 19:600-609. [PMID: 31821004 DOI: 10.1021/acs.jproteome.9b00440] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Su Guan
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, P. R. China
| | - Bingjie Jia
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, P. R. China
| | - Kang Chao
- Department of Gastroenterology, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, P. R. China
| | - Xia Zhu
- Institute of Clinical Pharmacology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, P. R. China
| | - Jian Tang
- Department of Gastroenterology, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, P. R. China
| | - Miao Li
- Department of Gastroenterology, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, P. R. China
| | - Lvying Wu
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, P. R. China
| | - Lei Xing
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, P. R. China
| | - Kun Liu
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, P. R. China
| | - Lei Zhang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, P. R. China
| | - Xueding Wang
- Institute of Clinical Pharmacology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, P. R. China
| | - Xiang Gao
- Department of Gastroenterology, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, P. R. China
| | - Min Huang
- Institute of Clinical Pharmacology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, P. R. China
| |
Collapse
|
31
|
Multiplex profiling of inflammation-related bioactive lipid mediators in Toxocara canis- and Toxocara cati-induced neurotoxocarosis. PLoS Negl Trop Dis 2019; 13:e0007706. [PMID: 31557153 PMCID: PMC6762062 DOI: 10.1371/journal.pntd.0007706] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 08/14/2019] [Indexed: 12/18/2022] Open
Abstract
Background Somatic migration of Toxocara canis- and T. cati-larvae in humans may cause neurotoxocarosis (NT) when larvae accumulate and persist in the central nervous system (CNS). Host- or parasite-induced immunoregulatory processes contribute to the pathogenesis; however, detailed data on involvement of bioactive lipid mediators, e.g. oxylipins or eico-/docosanoids, which are involved in the complex molecular signalling network during infection and inflammation, are lacking. Methodology/Principal findings To elucidate if T. canis- and T. cati-induced NT affects the homeostasis of oxylipins during the course of infection, a comprehensive lipidomic profiling in brains (cerebra and cerebella) of experimentally infected C57BL/6J mice was conducted at six different time points post infection (pi) by liquid-chromatography coupled to electrospray tandem mass spectrometry (LC-ESI-MS/MS). Only minor changes were detected regarding pro-inflammatory prostaglandins (cyclooxygenase pathway). In contrast, a significant increase of metabolites resulting from lipoxygenase pathways was observed for both infection groups and brain regions, implicating a predominantly anti-inflammatory driven immune response. This observation was supported by a significantly increased 13-hydroxyoctadecadienoic acid (HODE)/9-HODE ratio during the subacute phase of infection, indicating an anti-inflammatory response to neuroinfection. Except for the specialised pro-resolving mediator (SPM) neuroprotectin D1 (NPD1), which was detected in mice infected with both pathogens during the subacute phase of infection, no other SPMs were detected. Conclusions/Significance The obtained results demonstrate the influence of Toxocara spp. on oxylipins as part of the immune response of the paratenic hosts. Furthermore, this study shows differences in the alteration of the oxylipin composition between T. canis- and T. cati-brain infection. Results contribute to a further understanding of the largely unknown pathogenesis and mechanisms of host-parasite interactions during NT. Neurotoxocarosis (NT) is induced by larvae of the zoonotic roundworms Toxocara canis and T. cati migrating and persisting in the central nervous system of paratenic hosts, and may be accompanied by severe neurological symptoms. Toxocara spp. are known to modulate the hosts’ immune response, but data concerning involvement of signalling molecules are lacking. An important class of mediators participating in the complex molecular signalling network during infection and inflammation are bioactive regulatory lipids, derived from arachidonic acid and other polyunsaturated fatty acids. For a better understanding of inflammatory processes in the brain during an infection with Toxocara spp., a comprehensive analysis of regulatory lipids was conducted. The infection was predominantly characterised by only minor changes in the pattern of pro-inflammatory oxylipins, while anti-inflammatory metabolites, derived from lipoxygenase pathways, were significantly elevated in the subacute phase as well as in the beginning of the chronic phase of infection. This trend was also reflected in the 13-HODE/9-HODE ratio, a biomarker for the immunological status of an active infection. Obtained data provide a valuable insight in the host’s immune reaction as response against neuroinvasive Toxocara spp.-larvae, contributing to the characterisation of the mostly unknown pathogenesis of NT.
Collapse
|
32
|
Chen L, Tang S, Zhang FF, Garcia V, Falck JR, Schwartzman ML, Arbab AS, Guo AM. CYP4A/20-HETE regulates ischemia-induced neovascularization via its actions on endothelial progenitor and preexisting endothelial cells. Am J Physiol Heart Circ Physiol 2019; 316:H1468-H1479. [PMID: 30951365 PMCID: PMC6620690 DOI: 10.1152/ajpheart.00690.2018] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 03/29/2019] [Accepted: 03/29/2019] [Indexed: 11/22/2022]
Abstract
20-Hydroxyeicosatetraenoic acid (20-HETE) was recently identified as a novel contributor of ischemia-induced neovascularization based on the key observation that pharmacological interferences of CYP4A/20-HETE decrease ischemic neovascularization. The objective of the present study is to examine whether the underlying cellular mechanisms involve endothelial progenitor cells (EPCs) and preexisting endothelial cells (ECs). We found that ischemia leads to a time-dependent increase of cyp4a12 expression and 20-HETE production, which are endothelial in origin, using immunofluorescent microscopy, Western blot analysis, and LC-MS/MS. This is accompanied by increases in the tissue stromal cell-derived factor-1α (SDF-1α) expressions as well as SDF-1α plasma levels, EPC mobilization from bone marrow, and subsequent homing to ischemic tissues. Pharmacological interferences of CYP4A/20-HETE with a 20-HETE synthesis inhibitor, dibromo-dodecenyl-methylsulfimide (DDMS), or a 20-HETE antagonist, N-(20-hydroxyeicosa-6(Z), 15(Z)-dienoyl) glycine (6, 15-20-HEDGE), significantly attenuated these increases. Importantly, we also determined that 20-HETE plays a novel role in maintaining EPC functions and increasing the expression of Oct4, Sox2, and Nanog, which are indicative of increased progenitor cell stemness. Flow cytometric analysis revealed that pharmacological interferences of CYP4A/20-HETE decrease the EPC population in culture, whereas 20-HETE increases the cultured EPC population. Furthermore, ischemia also markedly increased the proliferation, oxidative stress, and ICAM-1 expression in the preexisting EC in the hindlimb gracilis muscles. We found that these increases were markedly negated by DDMS and 6, 15-20-HEDGE. Taken together, CYP4A/20-HETE regulates ischemia-induced compensatory neovascularization via its combined actions on promoting EPC and local preexisting EC responses that are associated with increased neovascularization. NEW & NOTEWORTHY CYP4A/20-hydroxyeicosatetraenoic acid (20-HETE) was recently discovered as a novel contributor of ischemia-induced neovascularization. However, the underlying molecular and cellular mechanisms are completely unknown. Here, we show that CYP4A/20-HETE regulates the ischemic neovascularization process via its combined actions on both endothelial progenitor cells (EPCs) and preexisting endothelial cells. Moreover, this is the first study, to the best of our knowledge, that associates CYP4A/20-HETE with EPC differentiation and stemness.
Collapse
Affiliation(s)
- Li Chen
- State Key Laboratory of Oncology, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center , Guangzhou , People's Republic of China
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | - Samantha Tang
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | - Frank F Zhang
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | - Victor Garcia
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | - John R Falck
- University of Texas Southwestern Medical Center , Dallas, Texas
| | | | - Ali S Arbab
- Cancer Center, Augusta University , Augusta, Georgia
| | - Austin M Guo
- Department of Pharmacology, New York Medical College, Valhalla, New York
| |
Collapse
|
33
|
Isli F, Yildirim S, Ozturk Fincan GS, Ercan S, Sarioglu Y. Effects of epoxygenases on the nonadrenergic noncholinergic relaxant responses induced by electrical field stimulation in rabbit corpus cavernosum. Andrologia 2019; 51:e13317. [PMID: 31107569 DOI: 10.1111/and.13317] [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: 01/31/2019] [Revised: 04/15/2019] [Accepted: 04/28/2019] [Indexed: 11/29/2022] Open
Abstract
We aimed to investigate the effects of epoxygenases on electrical field stimulation (EFS)-mediated nitric oxide (NO)-dependent and NO-independent nonadrenergic noncholinergic (NANC) relaxations in isolated rabbit corpus cavernosum. The tissues of 20 male adult albino rabbits (2.5-3 kg) were suspended in organ baths containing aerated Krebs solution, and isometric contractions were recorded. EFS-mediated NANC relaxations were obtained on phenylephrin (3 × 10-5 M)-contracted tissues in the presence of guanethidine (10-6 M) and atropine (10-6 M). Miconazole (10-9 -10-4 M), 17-octadecynoic acid (ODYA) (10-10 -10-5 M), 14,15-epoxyeicosatrienoic acid (EET) (10-11 -10-8 M), 11,12-EET (10-12 -3 × 10-8 M) and 20-hydroxyeicosatetraenoic acid (HETE) (10-11 -3 × 10-8 M) were added cumulatively (n = 5-7 for each set of experiments). For NO-independent relaxations, Nω -nitro-l-arginine methyl ester (l-NAME) (10-4 M) was added before a group of experiments. Depending on the concentration, miconazole, 17-ODYA, 14,15-EET, 11,12-EET, and 20-HETE significantly enhanced both NO-dependent and NO-independent EFS-mediated relaxations (p < 0.05). Epoxygenases showed similar effect on NO-dependent and NO-independent relaxant responses except 20-HETE which caused significantly more enhanced relaxation on NO-dependent responses (p < 0.05). No drug caused a significant relaxation response on tissues contracted with phenylephrine. Epoxygenases contribute to EFS-mediated NO-dependent and NO-independent NANC relaxations by presynaptic mechanisms, offering a new treatment alternative for erectile dysfunction which needs to be explored in further in vivo, molecular and clinical studies.
Collapse
Affiliation(s)
- Fatma Isli
- Department of Rational Drug Use, Turkish Medicines and Medical Devices Agency, Ministry of Health, Ankara, Turkey
| | - Seniz Yildirim
- Ankara Numune Health Application and Research Centre, University of Health Sciences, Ankara, Turkey
| | | | - Sevim Ercan
- Member of Turkish Academy of Science, Ankara, Turkey
| | - Yusuf Sarioglu
- Department of Medical Pharmacology, Istinye University School of Medicine, Istanbul, Turkey
| |
Collapse
|
34
|
Joerk A, Ritter M, Langguth N, Seidel RA, Freitag D, Herrmann KH, Schaefgen A, Ritter M, Günther M, Sommer C, Braemer D, Walter J, Ewald C, Kalff R, Reichenbach JR, Westerhausen M, Pohnert G, Witte OW, Holthoff K. Propentdyopents as Heme Degradation Intermediates Constrict Mouse Cerebral Arterioles and Are Present in the Cerebrospinal Fluid of Patients With Subarachnoid Hemorrhage. Circ Res 2019; 124:e101-e114. [PMID: 30947629 DOI: 10.1161/circresaha.118.314160] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
RATIONALE Delayed ischemic neurological deficit is the most common cause of neurological impairment and unfavorable prognosis in patients with subarachnoid hemorrhage (SAH). Despite the existence of neuroimaging modalities that depict the onset of the accompanying cerebral vasospasm, preventive and therapeutic options are limited and fail to improve outcome owing to an insufficient pathomechanistic understanding of the delayed perfusion deficit. Previous studies have suggested that BOXes (bilirubin oxidation end products), originating from released heme surrounding ruptured blood vessels, are involved in arterial vasoconstriction. Recently, isolated intermediates of oxidative bilirubin degradation, known as PDPs (propentdyopents), have been considered as potential additional effectors in the development of arterial vasoconstriction. OBJECTIVE To investigate whether PDPs and BOXes are present in hemorrhagic cerebrospinal fluid and involved in the vasoconstriction of cerebral arterioles. METHODS AND RESULTS Via liquid chromatography/mass spectrometry, we measured increased PDP and BOX concentrations in cerebrospinal fluid of SAH patients compared with control subjects. Using differential interference contrast microscopy, we analyzed the vasoactivity of PDP isomers in vitro by monitoring the arteriolar diameter in mouse acute brain slices. We found an arteriolar constriction on application of PDPs in the concentration range that occurs in the cerebrospinal fluid of patients with SAH. By imaging arteriolar diameter changes using 2-photon microscopy in vivo, we demonstrated a short-onset vasoconstriction after intrathecal injection of either PDPs or BOXes. Using magnetic resonance imaging, we observed a long-term PDP-induced delay in cerebral perfusion. For all conditions, the arteriolar narrowing was dependent on functional big conductance potassium channels and was absent in big conductance potassium channels knockout mice. CONCLUSIONS For the first time, we have quantified significantly higher concentrations of PDP and BOX isomers in the cerebrospinal fluid of patients with SAH compared to controls. The vasoconstrictive effect caused by PDPs in vitro and in vivo suggests a hitherto unrecognized pathway contributing to the pathogenesis of delayed ischemic deficit in patients with SAH.
Collapse
Affiliation(s)
- Alexander Joerk
- From the Hans Berger Department of Neurology (A.J., N.L., A.S., Marvin Ritter, M.G., C.S., D.B., O.W.W., K.H.), Jena University Hospital, Germany.,Research Program Else Kröner-Forschungskolleg AntiAge (A.J.), Jena University Hospital, Germany
| | | | - Niklas Langguth
- From the Hans Berger Department of Neurology (A.J., N.L., A.S., Marvin Ritter, M.G., C.S., D.B., O.W.W., K.H.), Jena University Hospital, Germany
| | - Raphael Andreas Seidel
- Department of Anesthesiology and Intensive Care Medicine / Center for Sepsis Control and Care (R.A.S.), Jena University Hospital, Germany.,Institute of Inorganic and Analytical Chemistry, Friedrich Schiller University Jena, Germany (Marcel Ritter, R.A.S., M.W., G.P.)
| | - Diana Freitag
- Department of Neurosurgery (D.F., J.W., R.K.), Jena University Hospital, Germany
| | - Karl-Heinz Herrmann
- Medical Physics Group, Institute for Diagnostic and Interventional Radiology, Jena University Hospital, Germany (K.-H.H., J.R.R.)
| | - Anna Schaefgen
- From the Hans Berger Department of Neurology (A.J., N.L., A.S., Marvin Ritter, M.G., C.S., D.B., O.W.W., K.H.), Jena University Hospital, Germany
| | - Marvin Ritter
- From the Hans Berger Department of Neurology (A.J., N.L., A.S., Marvin Ritter, M.G., C.S., D.B., O.W.W., K.H.), Jena University Hospital, Germany
| | - Milena Günther
- From the Hans Berger Department of Neurology (A.J., N.L., A.S., Marvin Ritter, M.G., C.S., D.B., O.W.W., K.H.), Jena University Hospital, Germany
| | - Charline Sommer
- From the Hans Berger Department of Neurology (A.J., N.L., A.S., Marvin Ritter, M.G., C.S., D.B., O.W.W., K.H.), Jena University Hospital, Germany
| | - Dirk Braemer
- From the Hans Berger Department of Neurology (A.J., N.L., A.S., Marvin Ritter, M.G., C.S., D.B., O.W.W., K.H.), Jena University Hospital, Germany
| | - Jan Walter
- Department of Neurosurgery (D.F., J.W., R.K.), Jena University Hospital, Germany
| | - Christian Ewald
- Department of Neurosurgery, Brandenburg Medical School, Campus Brandenburg an der Havel, Germany (C.E.)
| | - Rolf Kalff
- Department of Neurosurgery (D.F., J.W., R.K.), Jena University Hospital, Germany
| | - Jürgen Rainer Reichenbach
- Medical Physics Group, Institute for Diagnostic and Interventional Radiology, Jena University Hospital, Germany (K.-H.H., J.R.R.)
| | - Matthias Westerhausen
- Institute of Inorganic and Analytical Chemistry, Friedrich Schiller University Jena, Germany (Marcel Ritter, R.A.S., M.W., G.P.)
| | - Georg Pohnert
- Institute of Inorganic and Analytical Chemistry, Friedrich Schiller University Jena, Germany (Marcel Ritter, R.A.S., M.W., G.P.)
| | - Otto Wilhelm Witte
- From the Hans Berger Department of Neurology (A.J., N.L., A.S., Marvin Ritter, M.G., C.S., D.B., O.W.W., K.H.), Jena University Hospital, Germany
| | - Knut Holthoff
- From the Hans Berger Department of Neurology (A.J., N.L., A.S., Marvin Ritter, M.G., C.S., D.B., O.W.W., K.H.), Jena University Hospital, Germany
| |
Collapse
|
35
|
Jarrar YB, Jarrar Q, abed A, Abu-Shalhoob M. Effects of nonsteroidal anti-inflammatory drugs on the expression of arachidonic acid-metabolizing Cyp450 genes in mouse hearts, kidneys and livers. Prostaglandins Other Lipid Mediat 2019; 141:14-21. [DOI: 10.1016/j.prostaglandins.2019.02.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 01/31/2019] [Accepted: 02/05/2019] [Indexed: 02/04/2023]
|
36
|
Angiotensin II upregulates CYP4A isoform expression in the rat kidney through angiotensin II type 1 receptor. Prostaglandins Other Lipid Mediat 2018; 139:80-86. [DOI: 10.1016/j.prostaglandins.2018.09.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 08/02/2018] [Accepted: 09/12/2018] [Indexed: 11/21/2022]
|
37
|
Zhang C, Booz GW, Yu Q, He X, Wang S, Fan F. Conflicting roles of 20-HETE in hypertension and renal end organ damage. Eur J Pharmacol 2018; 833:190-200. [PMID: 29886242 PMCID: PMC6057804 DOI: 10.1016/j.ejphar.2018.06.010] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Revised: 06/05/2018] [Accepted: 06/06/2018] [Indexed: 12/12/2022]
Abstract
20-HETE is a cytochrome P450-derived metabolite of arachidonic acid that has both pro- and anti-hypertensive actions that result from modulation of vascular and kidney function. In the vasculature, 20-HETE sensitizes vascular smooth muscle cells to constrictor stimuli and increases myogenic tone. By promoting smooth muscle cell migration and proliferation, as well as by acting on the vascular endothelium to cause endothelial dysfunction, angiotensin converting enzyme (ACE) expression, and inflammation, 20-HETE contributes to adverse vascular remodeling and increased blood pressure. A G protein-coupled receptor was recently identified as the effector for the vascular actions of 20-HETE. In addition, evidence suggests that 20-HETE contributes to hypertension via positive regulation of the renin-angiotensin-aldosterone system, as well as by causing renal fibrosis. On the other hand, 20-HETE exerts anti-hypertensive actions by inhibiting sodium reabsorption by the kidney in both the proximal tubule and thick ascending limb of Henle. This review discusses the pro- and anti-hypertensive roles of 20-HETE in the pathogenesis of hypertension-associated renal disease, the association of gene polymorphisms of cytochrome P450 enzymes with the development of hypertension and renal end organ damage in humans, and 20-HETE related pharmaceutical agents.
Collapse
MESH Headings
- Animals
- Antihypertensive Agents/metabolism
- Antihypertensive Agents/pharmacology
- Arachidonic Acid/metabolism
- Cytochrome P-450 Enzyme System/genetics
- Cytochrome P-450 Enzyme System/metabolism
- Endothelium, Vascular/cytology
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/physiopathology
- Fibrosis
- Humans
- Hydroxyeicosatetraenoic Acids/pharmacology
- Hydroxyeicosatetraenoic Acids/physiology
- Hypertension/complications
- Hypertension/drug therapy
- Hypertension/metabolism
- Hypertension/physiopathology
- Kidney/metabolism
- Kidney/pathology
- Kidney/physiopathology
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/physiopathology
- Peptidyl-Dipeptidase A/metabolism
- Polymorphism, Genetic
- Receptors, G-Protein-Coupled/metabolism
- Renal Elimination/physiology
- Renal Insufficiency/drug therapy
- Renal Insufficiency/etiology
- Renal Insufficiency/metabolism
- Renal Insufficiency/physiopathology
- Renin-Angiotensin System/physiology
- Sodium/metabolism
- Vascular Remodeling/physiology
Collapse
Affiliation(s)
- Chao Zhang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS 39216, USA; Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - George W Booz
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS 39216, USA
| | - Qing Yu
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiaochen He
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS 39216, USA
| | - Shaoxun Wang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS 39216, USA
| | - Fan Fan
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS 39216, USA.
| |
Collapse
|
38
|
Abstract
20-HETE, the ω-hydroxylation product of arachidonic acid catalyzed by enzymes of the cytochrome P450 (CYP) 4A and 4F gene families, is a bioactive lipid mediator with potent effects on the vasculature including stimulation of smooth muscle cell contractility, migration and proliferation as well as activation of endothelial cell dysfunction and inflammation. Clinical studies have shown elevated levels of plasma and urinary 20-HETE in human diseases and conditions such as hypertension, obesity and metabolic syndrome, myocardial infarction, stroke, and chronic kidney diseases. Studies of polymorphic associations also suggest an important role for 20-HETE in hypertension, stroke and myocardial infarction. Animal models of increased 20-HETE production are hypertensive and are more susceptible to cardiovascular injury. The current review summarizes recent findings that focus on the role of 20-HETE in the regulation of vascular and cardiac function and its contribution to the pathology of vascular and cardiac diseases.
Collapse
Affiliation(s)
- Petra Rocic
- Department of Pharmacology, New York Medical College School of Medicine, Valhalla, NY, United States
| | | |
Collapse
|
39
|
Costa TJ, Ceravolo GS, Echem C, Hashimoto CM, Costa BP, Santos-Eichler RA, Oliveira MA, Jiménez-Altayó F, Akamine EH, Dantas AP, Carvalho MHC. Detrimental Effects of Testosterone Addition to Estrogen Therapy Involve Cytochrome P-450-Induced 20-HETE Synthesis in Aorta of Ovariectomized Spontaneously Hypertensive Rat (SHR), a Model of Postmenopausal Hypertension. Front Physiol 2018; 9:490. [PMID: 29867542 PMCID: PMC5952044 DOI: 10.3389/fphys.2018.00490] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 04/17/2018] [Indexed: 12/02/2022] Open
Abstract
Postmenopausal period has been associated to different symptoms such as hot flashes, vulvovaginal atrophy, hypoactive sexual desire disorder (HSDD) and others. Clinical studies have described postmenopausal women presenting HSDD can benefit from the association of testosterone to conventional hormonal therapy. Testosterone has been linked to development of cardiovascular diseases including hypertension and it also increases cytochrome P-450-induced 20-HETE synthesis which in turn results in vascular dysfunction. However, the effect of testosterone plus estrogen in the cardiovascular system is still very poorly studied. The aim of the present study is to evaluate the role of cytochrome P-450 pathway in a postmenopausal hypertensive female treated with testosterone plus estrogen. For that, hypertensive ovariectomized rats (OVX-SHR) were used as a model of postmenopausal hypertension and four groups were created: SHAM-operated (SHAM), ovariectomized SHR (OVX), OVX treated for 15 days with conjugated equine estrogens [(CEE) 9.6 μg/Kg/day/po] or CEE associated to testosterone [(CEE+T) 2.85 mg/kg/weekly/im]. Phenylephrine-induced contraction and generation of reactive oxygen species (ROS) were markedly increased in aortic rings from OVX-SHR compared to SHAM rats which were restored by CEE treatment. On the other hand, CEE+T abolished vascular effects by CEE and augmented both systolic and diastolic blood pressure of SHR. Treatment of aortic rings with the CYP/20-HETE synthesis inhibitor HET0016 (1 μM) reduced phenylephrine hyperreactivity and the augmented ROS generation in the CEE+T group. These results are paralleled by the increased CYP4F3 protein expression and activity in aortas of CEE+T. In conclusion, we showed that association of testosterone to estrogen therapy produces detrimental effects in cardiovascular system of ovariectomized hypertensive females via CYP4F3/20-HETE pathway. Therefore, our findings support the standpoint that the CYP/20-HETE pathway is an important therapeutic target for the prevention of cardiovascular disease in menopausal women in the presence of high levels of testosterone.
Collapse
Affiliation(s)
- Tiago J Costa
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.,Facultat de Medicina, Departament de Farmacologia, Terapèutica i Toxicologia, Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Spain.,Group of Atherosclerosis and Coronary Disease, Institut Clinic del Torax, Institut d'Investigacions Biomédiques August Pi I Sunyer, Barcelona, Spain
| | - Graziela S Ceravolo
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.,Department of Physiological Sciences, State University of Londrina, Londrina, Brazil
| | - Cinthya Echem
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Carolina M Hashimoto
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Beatriz P Costa
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Rosangela A Santos-Eichler
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Maria Aparecida Oliveira
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Francesc Jiménez-Altayó
- Facultat de Medicina, Departament de Farmacologia, Terapèutica i Toxicologia, Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Eliana H Akamine
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Ana Paula Dantas
- Group of Atherosclerosis and Coronary Disease, Institut Clinic del Torax, Institut d'Investigacions Biomédiques August Pi I Sunyer, Barcelona, Spain
| | - Maria Helena C Carvalho
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
40
|
Plenty NL, Faulkner JL, Cotton J, Spencer SK, Wallace K, LaMarca B, Murphy SR. Arachidonic acid metabolites of CYP4A and CYP4F are altered in women with preeclampsia. Prostaglandins Other Lipid Mediat 2018; 136:15-22. [DOI: 10.1016/j.prostaglandins.2018.03.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 02/24/2018] [Accepted: 03/01/2018] [Indexed: 01/25/2023]
|
41
|
Racine ML, Crecelius AR, Luckasen GJ, Larson DG, Dinenno FA. Inhibition of Na + /K + -ATPase and K IR channels abolishes hypoxic hyperaemia in resting but not contracting skeletal muscle of humans. J Physiol 2018; 596:3371-3389. [PMID: 29603743 DOI: 10.1113/jp275913] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Accepted: 03/27/2018] [Indexed: 12/16/2022] Open
Abstract
KEY POINTS Increasing blood flow (hyperaemia) to exercising muscle helps match oxygen delivery and metabolic demand. During exercise in hypoxia, there is a compensatory increase in muscle hyperaemia that maintains oxygen delivery and tissue oxygen consumption. Nitric oxide (NO) and prostaglandins (PGs) contribute to around half of the augmented hyperaemia during hypoxic exercise, although the contributors to the remaining response are unknown. In the present study, inhibiting NO, PGs, Na+ /K+ -ATPase and inwardly rectifying potassium (KIR ) channels did not blunt augmented hyperaemia during hypoxic exercise beyond previous observations with NO/PG block alone. Furthermore, although inhibition of only Na+ /K+ -ATPase and KIR channels abolished hyperaemia during hypoxia at rest, it had no effect on augmented hyperaemia during hypoxic exercise. This is the first study in humans to demonstrate that Na+ /K+ -ATPase and KIR channel activation is required for augmented muscle hyperaemia during hypoxia at rest but not during hypoxic exercise, thus providing new insight into vascular control. ABSTRACT Exercise hyperaemia in hypoxia is augmented relative to the same exercise intensity in normoxia. During moderate-intensity handgrip exercise, endothelium-derived nitric oxide (NO) and vasodilating prostaglandins (PGs) contribute to ∼50% of the augmented forearm blood flow (FBF) response to hypoxic exercise (HypEx), although the mechanism(s) underlying the remaining response are unclear. We hypothesized that combined inhibition of NO, PGs, Na+ /K+ -ATPase and inwardly rectifying potassium (KIR ) channels would abolish the augmented hyperaemic response in HypEx. In healthy young adults, FBF responses were measured (Doppler ultrasound) and forearm vascular conductance was calculated during 5 min of rhythmic handgrip exercise at 20% maximum voluntary contraction under regional sympathoadrenal inhibition in normoxia and isocapnic HypEx (O2 saturation ∼80%). Compared to control, combined inhibition of NO, PGs, Na+ /K+ -ATPase and KIR channels (l-NMMA + ketorolac + ouabain + BaCl2; Protocol 1; n = 10) blunted the compensatory increase in FBF during HypEx by ∼50% (29 ± 6 mL min-1 vs. 62 ± 8 mL min-1 , respectively, P < 0.05). By contrast, ouabain + BaCl2 alone (Protocol 2; n = 10) did not affect this augmented hyperaemic response (50 ± 11 mL min-1 vs. 60 ± 13 mL min-1 , respectively, P > 0.05). However, the blocked condition in both protocols abolished the hyperaemic response to hypoxia at rest (P < 0.05). We conclude that activation of Na+ /K+ -ATPase and KIR channels is involved in the hyperaemic response to hypoxia at rest, although it does not contribute to the augmented exercise hyperaemia during hypoxia in humans.
Collapse
Affiliation(s)
- Matthew L Racine
- Human Cardiovascular Physiology Laboratory, Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, USA
| | - Anne R Crecelius
- Human Cardiovascular Physiology Laboratory, Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, USA
| | - Gary J Luckasen
- Cardiovascular Research Center, Colorado State University, Fort Collins, CO, USA.,Medical Center of the Rockies Foundation, University of Colorado Health System, Loveland, CO, USA
| | - Dennis G Larson
- Medical Center of the Rockies Foundation, University of Colorado Health System, Loveland, CO, USA
| | - Frank A Dinenno
- Human Cardiovascular Physiology Laboratory, Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, USA.,Cardiovascular Research Center, Colorado State University, Fort Collins, CO, USA
| |
Collapse
|
42
|
Thakare R, Chhonker YS, Gautam N, Nelson A, Casaburi R, Criner G, Dransfield MT, Make B, Schmid KK, Rennard SI, Alnouti Y. Simultaneous LC-MS/MS analysis of eicosanoids and related metabolites in human serum, sputum and BALF. Biomed Chromatogr 2018; 32:10.1002/bmc.4102. [PMID: 28975688 PMCID: PMC6003856 DOI: 10.1002/bmc.4102] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 09/05/2017] [Accepted: 09/24/2017] [Indexed: 01/09/2023]
Abstract
The differences among individual eicosanoids in eliciting different physiological and pathological responses are largely unknown because of the lack of valid and simple analytical methods for the quantification of individual eicosanoids and their metabolites in serum, sputum and bronchial alveolar lavage fluid (BALF). Therefore, a simple and sensitive LC-MS/MS method for the simultaneous quantification of 34 eicosanoids in human serum, sputum and BALF was developed and validated. This method is valid and sensitive with a limit of quantification ranging from 0.2 to 3 ng/mL for the various analytes, and has a large dynamic range (500 ng/mL) and a short run time (25 min). The intra- and inter-day accuracy and precision values met the acceptance criteria according to US Food and Drug Administration guidelines. Using this method, detailed eicosanoid profiles were quantified in serum, sputum and BALF from a pilot human study. In summary, a reliable and simple LC-MS/MS method to quantify major eicosanoids and their metabolites was developed and applied to quantify eicosanoids in human various fluids, demonstrating its suitability to assess eicosanoid biomarkers in human clinical trials.
Collapse
Affiliation(s)
- Rhishikesh Thakare
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Yashpal S. Chhonker
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Nagsen Gautam
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Amy Nelson
- Pulmonary and Critical Care Medicine Section, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Richard Casaburi
- Rehabilitation Clinical Trials Center, Los Angeles Biomedical Research Institute at Harbor UCLA Medical Center, Torrance, CA, USA
| | - Gerard Criner
- Division of Pulmonary and Critical Care Medicine, Temple University, Philadelphia, PA, USA
| | - Mark T. Dransfield
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama Birmingham, AL, USA
- Lung Health Center University of Alabama Birmingham, Birmingham, AL, USA
- Birmingham VA Medical Center, Birmingham, AL, USA
| | - Barry Make
- Division of Pulmonary, Critical Care, and Sleep Medicine, National Jewish Health, Denver, CO, USA
| | - Kendra K. Schmid
- College of Public Health, University of Nebraska Medical Center, Omaha, NE, USA
| | - Stephen I. Rennard
- Pulmonary and Critical Care Medicine Section, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
- Clinical Development Unit, Early Clinical Development, AstraZeneca, Cambridge, UK
| | - Yazen Alnouti
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
43
|
Cytochrome P450 4A11 inhibition assays based on characterization of lauric acid metabolites. Food Chem Toxicol 2018; 112:205-215. [DOI: 10.1016/j.fct.2017.12.063] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 12/19/2017] [Accepted: 12/29/2017] [Indexed: 01/08/2023]
|
44
|
Pandey V, Garcia V, Gilani A, Mishra P, Zhang FF, Paudyal MP, Falck JR, Nasjletti A, Wang WH, Schwartzman ML. The Blood Pressure-Lowering Effect of 20-HETE Blockade in Cyp4a14(-/-) Mice Is Associated with Natriuresis. J Pharmacol Exp Ther 2017; 363:412-418. [PMID: 28912346 PMCID: PMC5698946 DOI: 10.1124/jpet.117.243618] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 09/13/2017] [Indexed: 01/22/2023] Open
Abstract
20-Hydroxy-5,8,11,14-eicosatetraenoic acid (20-HETE) has been linked to pro-hypertensive and anti-hypertensive actions through its ability to promote vasoconstriction and inhibit Na transport in the ascending limb of the loop of Henle, respectively. In this study, we assessed the effects of 20-HETE blockade on blood pressure, renal hemodynamics, and urinary sodium excretion in Cyp4a14(-/-) male mice, which display androgen-driven 20-HETE-dependent hypertension. Administration of 2,5,8,11,14,17-hexaoxanonadecan-19-yl 20-hydroxyicosa-6(Z),15(Z)-dienoate (20-SOLA), a water-soluble 20-HETE antagonist, in the drinking water normalized the blood pressure of male Cyp4a14(-/-) hypertensive mice (±124 vs. ±153 mmHg) while having no effect on age-matched normotensive wild-type (WT) male mice. Hypertension in Cyp4a14(-/-) male mice was accompanied by decreased renal perfusion and reduced glomerular filtration rates, which were corrected by treatment with 20-SOLA. Interestingly, Cyp4a14(-/-) male mice treated with 20-SOLA displayed increased urinary sodium excretion that was paralleled by the reduction of blood pressure suggestive of an antinatriuretic activity of endogenous 20-HETE in the hypertensive mice. This interpretation is in line with the observation that the natriuretic response to acute isotonic saline loading in hypertensive Cyp4a14(-/-) male mice was significantly impaired relative to that in WT mice; this impairment was corrected by 20-SOLA treatment. Hence, endogenous 20-HETE appears to promote sodium conservation in hypertensive Cyp4a14(-/-) male mice, presumably, as a result of associated changes in renal hemodynamics and/or direct stimulatory action on tubular sodium reabsorption.
Collapse
Affiliation(s)
- Varunkumar Pandey
- Department of Pharmacology, New York Medical College School of Medicine, Valhalla, New York (V.P., V.G., A.G., P.M., F.F.Z., A.N., W.-H.W., M.L.S.); and Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas (M.P.P., J.R.F.)
| | - Victor Garcia
- Department of Pharmacology, New York Medical College School of Medicine, Valhalla, New York (V.P., V.G., A.G., P.M., F.F.Z., A.N., W.-H.W., M.L.S.); and Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas (M.P.P., J.R.F.)
| | - Ankit Gilani
- Department of Pharmacology, New York Medical College School of Medicine, Valhalla, New York (V.P., V.G., A.G., P.M., F.F.Z., A.N., W.-H.W., M.L.S.); and Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas (M.P.P., J.R.F.)
| | - Priyanka Mishra
- Department of Pharmacology, New York Medical College School of Medicine, Valhalla, New York (V.P., V.G., A.G., P.M., F.F.Z., A.N., W.-H.W., M.L.S.); and Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas (M.P.P., J.R.F.)
| | - Frank Fan Zhang
- Department of Pharmacology, New York Medical College School of Medicine, Valhalla, New York (V.P., V.G., A.G., P.M., F.F.Z., A.N., W.-H.W., M.L.S.); and Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas (M.P.P., J.R.F.)
| | - Mahesh P Paudyal
- Department of Pharmacology, New York Medical College School of Medicine, Valhalla, New York (V.P., V.G., A.G., P.M., F.F.Z., A.N., W.-H.W., M.L.S.); and Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas (M.P.P., J.R.F.)
| | - John R Falck
- Department of Pharmacology, New York Medical College School of Medicine, Valhalla, New York (V.P., V.G., A.G., P.M., F.F.Z., A.N., W.-H.W., M.L.S.); and Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas (M.P.P., J.R.F.)
| | - Alberto Nasjletti
- Department of Pharmacology, New York Medical College School of Medicine, Valhalla, New York (V.P., V.G., A.G., P.M., F.F.Z., A.N., W.-H.W., M.L.S.); and Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas (M.P.P., J.R.F.)
| | - Wen-Hui Wang
- Department of Pharmacology, New York Medical College School of Medicine, Valhalla, New York (V.P., V.G., A.G., P.M., F.F.Z., A.N., W.-H.W., M.L.S.); and Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas (M.P.P., J.R.F.)
| | - Michal Laniado Schwartzman
- Department of Pharmacology, New York Medical College School of Medicine, Valhalla, New York (V.P., V.G., A.G., P.M., F.F.Z., A.N., W.-H.W., M.L.S.); and Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas (M.P.P., J.R.F.)
| |
Collapse
|
45
|
Jamieson KL, Endo T, Darwesh AM, Samokhvalov V, Seubert JM. Cytochrome P450-derived eicosanoids and heart function. Pharmacol Ther 2017; 179:47-83. [PMID: 28551025 DOI: 10.1016/j.pharmthera.2017.05.005] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
46
|
Özen N, Nasırcılar Ülker S, Ülker P, Özcan F, Aslan M, Şentürk ÜK, Basralı F. Effect of 20-HETE inhibition on L-NAME-induced hypertension in rats. Clin Exp Hypertens 2017; 40:292-302. [DOI: 10.1080/10641963.2017.1368540] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Nur Özen
- Medical Faculty, Department of Physiology, Akdeniz University, Antalya, Turkey
| | | | - Pınar Ülker
- Medical Faculty, Department of Physiology, Akdeniz University, Antalya, Turkey
| | - Filiz Özcan
- Medical Faculty, Department of Biochemistry, Akdeniz University, Antalya, Turkey
| | - Mutay Aslan
- Medical Faculty, Department of Biochemistry, Akdeniz University, Antalya, Turkey
| | - Ümit Kemal Şentürk
- Medical Faculty, Department of Physiology, Akdeniz University, Antalya, Turkey
| | - Filiz Basralı
- Medical Faculty, Department of Physiology, Akdeniz University, Antalya, Turkey
| |
Collapse
|
47
|
Bioactive lipids derived from arachidonic acid metabolism in different types of renal replacement therapy. Chem Phys Lipids 2017; 206:71-77. [PMID: 28533146 DOI: 10.1016/j.chemphyslip.2017.05.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 05/11/2017] [Accepted: 05/16/2017] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Metabolism and plasma concentration of lipids and lipid-derived compounds play an important role in kidney physiology and pathological processes. The component of membrane phospholipids - arachidonic acid (AA) and its active derivatives - eicosanoids are involved in the development of hypertension, diabetes, inflammation and may contribute to progression of chronic kidney disease (CKD). The purpose of the study was to determine, whether the type of renal replacement therapy has an effect on eicosanoids metabolism. MATERIALS AND METHODS The study included 145 patients with CKD: on conservative treatment (n=68), on peritoneal dialysis (PD) (n=23) and undergoing chronic haemodialysis (HD) (n=54). The concentrations of TXB2, 20-HETE, 8-epi-PGF2α in platelet poor plasma (PPP) were determined using the ELISA method and 5-HETE, 12-HETE, 15-HETE were measured using the RP-HPLC. RESULTS The concentrations of TXB2 in HD group, both before (2.28±0.72ng/mL) and after (1.49±0.63ng/mL) haemodialysis treatment differed significantly from PD group (57.76±6.13ng/mL). Haemodialysis session led to the significant decrease in TXB2 plasma concentration (p=0.046). 20-HETE concentrations in HD group (113.55±107.54pg/mL and 199.54±142.98pg/mL before and after haemodialysis, respectively) were significantly higher than in CKD 3-5 group (8.96±12.66pg/mL) and PD group (47.78±34.07pg/mL). The highest concentration of 12-HETE was obtained in PD patients (3.58±3.99ng/mL) and differed significantly from HD group after haemodialysis (0.97±0.28ng/mL) and CKD3-5 group (1.06±0.52ng/mL). The concentrations of 5-HETE, 15-HETE and 8-epi-PGF2α-III did not differ significantly among examined groups. CONCLUSIONS The concentrations of active AA metabolites depend on the mode of renal replacement therapy and are associated with intensity of oxidative stress. They might be considered as potential indicators of kidney damage.
Collapse
|
48
|
Garcia V, Gilani A, Shkolnik B, Pandey V, Zhang FF, Dakarapu R, Gandham SK, Reddy NR, Graves JP, Gruzdev A, Zeldin DC, Capdevila JH, Falck JR, Schwartzman ML. 20-HETE Signals Through G-Protein-Coupled Receptor GPR75 (G q) to Affect Vascular Function and Trigger Hypertension. Circ Res 2017; 120:1776-1788. [PMID: 28325781 DOI: 10.1161/circresaha.116.310525] [Citation(s) in RCA: 133] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 03/03/2017] [Accepted: 03/20/2017] [Indexed: 12/31/2022]
Abstract
RATIONALE 20-Hydroxyeicosatetraenoic acid (20-HETE), one of the principle cytochrome P450 eicosanoids, is a potent vasoactive lipid whose vascular effects include stimulation of smooth muscle contractility, migration, and proliferation, as well as endothelial cell dysfunction and inflammation. Increased levels of 20-HETE in experimental animals and in humans are associated with hypertension, stroke, myocardial infarction, and vascular diseases. OBJECTIVE To date, a receptor/binding site for 20-HETE has been implicated based on the use of specific agonists and antagonists. The present study was undertaken to identify a receptor to which 20-HETE binds and through which it activates a signaling cascade that culminates in many of the functional outcomes attributed to 20-HETE in vitro and in vivo. METHODS AND RESULTS Using crosslinking analogs, click chemistry, binding assays, and functional assays, we identified G-protein receptor 75 (GPR75), currently an orphan G-protein-coupled receptor (GPCR), as a specific target of 20-HETE. In cultured human endothelial cells, 20-HETE binding to GPR75 stimulated Gαq/11 protein dissociation and increased inositol phosphate accumulation and GPCR-kinase interacting protein-1-GPR75 binding, which further facilitated the c-Src-mediated transactivation of epidermal growth factor receptor. This results in downstream signaling pathways that induce angiotensin-converting enzyme expression and endothelial dysfunction. Knockdown of GPR75 or GPCR-kinase interacting protein-1 prevented 20-HETE-mediated endothelial growth factor receptor phosphorylation and angiotensin-converting enzyme induction. In vascular smooth muscle cells, GPR75-20-HETE pairing is associated with Gαq/11- and GPCR-kinase interacting protein-1-mediated protein kinase C-stimulated phosphorylation of MaxiKβ, linking GPR75 activation to 20-HETE-mediated vasoconstriction. GPR75 knockdown in a mouse model of 20-HETE-dependent hypertension prevented blood pressure elevation and 20-HETE-mediated increases in angiotensin-converting enzyme expression, endothelial dysfunction, smooth muscle contractility, and vascular remodeling. CONCLUSIONS This is the first report to identify a GPCR target for an eicosanoid of this class. The discovery of 20-HETE-GPR75 pairing presented here provides the molecular basis for the signaling and pathophysiological functions mediated by 20-HETE in hypertension and cardiovascular diseases.
Collapse
Affiliation(s)
- Victor Garcia
- From the Department of Pharmacology, New York Medical College School of Medicine, Valhalla (V.G., A.G., B.S., V.P., F.F.Z., M.L.S.); Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas (R.D., S.K.G., N.R.R., J.R.F.); Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC (J.P.G., A.G., D.C.Z.); and Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (J.H.C.)
| | - Ankit Gilani
- From the Department of Pharmacology, New York Medical College School of Medicine, Valhalla (V.G., A.G., B.S., V.P., F.F.Z., M.L.S.); Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas (R.D., S.K.G., N.R.R., J.R.F.); Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC (J.P.G., A.G., D.C.Z.); and Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (J.H.C.)
| | - Brian Shkolnik
- From the Department of Pharmacology, New York Medical College School of Medicine, Valhalla (V.G., A.G., B.S., V.P., F.F.Z., M.L.S.); Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas (R.D., S.K.G., N.R.R., J.R.F.); Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC (J.P.G., A.G., D.C.Z.); and Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (J.H.C.)
| | - Varunkumar Pandey
- From the Department of Pharmacology, New York Medical College School of Medicine, Valhalla (V.G., A.G., B.S., V.P., F.F.Z., M.L.S.); Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas (R.D., S.K.G., N.R.R., J.R.F.); Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC (J.P.G., A.G., D.C.Z.); and Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (J.H.C.)
| | - Frank Fan Zhang
- From the Department of Pharmacology, New York Medical College School of Medicine, Valhalla (V.G., A.G., B.S., V.P., F.F.Z., M.L.S.); Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas (R.D., S.K.G., N.R.R., J.R.F.); Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC (J.P.G., A.G., D.C.Z.); and Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (J.H.C.)
| | - Rambabu Dakarapu
- From the Department of Pharmacology, New York Medical College School of Medicine, Valhalla (V.G., A.G., B.S., V.P., F.F.Z., M.L.S.); Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas (R.D., S.K.G., N.R.R., J.R.F.); Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC (J.P.G., A.G., D.C.Z.); and Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (J.H.C.)
| | - Shyam K Gandham
- From the Department of Pharmacology, New York Medical College School of Medicine, Valhalla (V.G., A.G., B.S., V.P., F.F.Z., M.L.S.); Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas (R.D., S.K.G., N.R.R., J.R.F.); Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC (J.P.G., A.G., D.C.Z.); and Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (J.H.C.)
| | - N Rami Reddy
- From the Department of Pharmacology, New York Medical College School of Medicine, Valhalla (V.G., A.G., B.S., V.P., F.F.Z., M.L.S.); Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas (R.D., S.K.G., N.R.R., J.R.F.); Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC (J.P.G., A.G., D.C.Z.); and Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (J.H.C.)
| | - Joan P Graves
- From the Department of Pharmacology, New York Medical College School of Medicine, Valhalla (V.G., A.G., B.S., V.P., F.F.Z., M.L.S.); Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas (R.D., S.K.G., N.R.R., J.R.F.); Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC (J.P.G., A.G., D.C.Z.); and Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (J.H.C.)
| | - Artiom Gruzdev
- From the Department of Pharmacology, New York Medical College School of Medicine, Valhalla (V.G., A.G., B.S., V.P., F.F.Z., M.L.S.); Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas (R.D., S.K.G., N.R.R., J.R.F.); Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC (J.P.G., A.G., D.C.Z.); and Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (J.H.C.)
| | - Darryl C Zeldin
- From the Department of Pharmacology, New York Medical College School of Medicine, Valhalla (V.G., A.G., B.S., V.P., F.F.Z., M.L.S.); Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas (R.D., S.K.G., N.R.R., J.R.F.); Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC (J.P.G., A.G., D.C.Z.); and Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (J.H.C.)
| | - Jorge H Capdevila
- From the Department of Pharmacology, New York Medical College School of Medicine, Valhalla (V.G., A.G., B.S., V.P., F.F.Z., M.L.S.); Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas (R.D., S.K.G., N.R.R., J.R.F.); Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC (J.P.G., A.G., D.C.Z.); and Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (J.H.C.)
| | - John R Falck
- From the Department of Pharmacology, New York Medical College School of Medicine, Valhalla (V.G., A.G., B.S., V.P., F.F.Z., M.L.S.); Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas (R.D., S.K.G., N.R.R., J.R.F.); Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC (J.P.G., A.G., D.C.Z.); and Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (J.H.C.)
| | - Michal Laniado Schwartzman
- From the Department of Pharmacology, New York Medical College School of Medicine, Valhalla (V.G., A.G., B.S., V.P., F.F.Z., M.L.S.); Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas (R.D., S.K.G., N.R.R., J.R.F.); Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC (J.P.G., A.G., D.C.Z.); and Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (J.H.C.).
| |
Collapse
|
49
|
Ungvari Z, Tarantini S, Kirkpatrick AC, Csiszar A, Prodan CI. Cerebral microhemorrhages: mechanisms, consequences, and prevention. Am J Physiol Heart Circ Physiol 2017; 312:H1128-H1143. [PMID: 28314762 PMCID: PMC5495931 DOI: 10.1152/ajpheart.00780.2016] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 02/22/2017] [Accepted: 03/09/2017] [Indexed: 12/20/2022]
Abstract
The increasing prevalence of multifocal cerebral microhemorrhages (CMHs, also known as "cerebral microbleeds") is a significant, newly recognized problem in the aging population of the Western world. CMHs are associated with rupture of small intracerebral vessels and are thought to progressively impair neuronal function, potentially contributing to cognitive decline, geriatric psychiatric syndromes, and gait disorders. Clinical studies show that aging and hypertension significantly increase prevalence of CMHs. CMHs are also now recognized by the National Institutes of Health as a major factor in Alzheimer's disease pathology. Moreover, the presence of CMHs is an independent risk factor for subsequent larger intracerebral hemorrhages. In this article, we review the epidemiology, detection, risk factors, clinical significance, and pathogenesis of CMHs. The potential age-related cellular mechanisms underlying the development of CMHs are discussed, with a focus on the structural determinants of microvascular fragility, age-related alterations in cerebrovascular adaptation to hypertension, the role of oxidative stress and matrix metalloproteinase activation, and the deleterious effects of arterial stiffening, increased pulse pressure, and impaired myogenic autoregulatory protection on the brain microvasculature. Finally, we examine potential treatments for the prevention of CMHs based on the proposed model of aging- and hypertension-dependent activation of the reactive oxygen species-matrix metalloproteinases axis, and we discuss critical questions to be addressed by future studies.
Collapse
Affiliation(s)
- Zoltan Ungvari
- Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; .,Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Stefano Tarantini
- Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma.,Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Angelia C Kirkpatrick
- Veterans Affairs Medical Center, Oklahoma City, Oklahoma.,Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; and
| | - Anna Csiszar
- Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma.,Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Calin I Prodan
- Department of Neurology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| |
Collapse
|
50
|
Wei Y, Xu M, Ren Y, Lu G, Xu Y, Song Y, Ji H. The cardioprotection of dihydrotanshinone I against myocardial ischemia–reperfusion injury via inhibition of arachidonic acid ω-hydroxylase. Can J Physiol Pharmacol 2016; 94:1267-1275. [DOI: 10.1139/cjpp-2016-0036] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Arachidonic acid (AA) is a precursor that is metabolized by several enzymes to many biological eicosanoids. Accumulating data indicate that the ω-hydroxylation metabolite of AA, 20-hydroxyeicosatetraenoic acid (20-HETE), is considered to be involved in the myocardial ischemia–reperfusion injury (MIRI). The inhibitors of AA ω-hydroxylase, however, are demonstrated to exhibit protective effects on MIRI. Dihydrotanshinone I (DI), a bioactive constituent of danshen, is proven to be a potent inhibitor of AA ω-hydroxylase by our preliminary study in vitro. The purpose of the present study was to investigate the cardioprotection of DI against MIRI and its effects on the concentrations of 20-HETE in vivo. Rats subjected to 30 min of ischemia followed by 24 h of reperfusion were assigned to intravenously receive vehicle (sham and ischemia–reperfusion), low (1 mg/kg), middle (2 mg/kg), or high (4 mg/kg) doses of DI before reperfusion. The results demonstrated that DI treatment could improve cardiac function, reduce infarct size, ameliorate the variations in myocardial zymogram and histopathological disorders, decrease 20-HETE generation, and regulate apoptosis-related protein in myocardial ischemia–reperfusion rats. These findings suggested DI could exert considerable cardioprotective action on MIRI by the attenuation of 20-HETE generation, subsequent myocardial injury, and apoptosis through inhibition on AA ω-hydroxylase.
Collapse
Affiliation(s)
- Yidan Wei
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, Jiangsu, China
| | - Meijuan Xu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, Jiangsu, China
- Department of Clinical Pharmacology, Affiliated Hospital of Nanjing University of Chinese Medicine, No. 155 Hanzhong Road, Nanjing, Jiangsu, China
| | - Yi Ren
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, Jiangsu, China
| | - Guo Lu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, Jiangsu, China
| | - Yangmei Xu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, Jiangsu, China
| | - Yangyang Song
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, Jiangsu, China
| | - Hui Ji
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, Jiangsu, China
| |
Collapse
|