1
|
Dienel A, Hong SH, Zeineddine HA, Thomas S, M SC, Jose DA, Torres K, Guzman J, Dunn A, T PK, Rao GN, Blackburn SL, McBride DW. 12/15-Lipooxygenase Inhibition Reduces Microvessel Constriction and Microthrombi After Subarachnoid Hemorrhage in Mice. Transl Stroke Res 2024:10.1007/s12975-024-01295-0. [PMID: 39294532 DOI: 10.1007/s12975-024-01295-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 08/28/2024] [Accepted: 09/02/2024] [Indexed: 09/20/2024]
Abstract
Impaired cerebral circulation, induced by blood vessel constrictions and microthrombi, leads to delayed cerebral ischemia after subarachnoid hemorrhage (SAH). 12/15-Lipooxygenase (12/15-LOX) overexpression has been implicated in worsening early brain injury outcomes following SAH. However, it is unknown if 12/15-LOX is important in delayed pathophysiological events after SAH. Since 12/15-LOX produces metabolites that induce inflammation and vasoconstriction, we hypothesized that 12/15-LOX leads to microvessel constriction and microthrombi formation after SAH, and thus, 12/15-LOX is an important target to prevent delayed cerebral ischemia. SAH was induced in C57BL/6 and 12/15-LOX-/- mice of both sexes by endovascular perforation. Expression of 12/15-LOX was assessed in brain tissue slices and in vitro. C57BL/6 mice were administered either ML351 (12/15-LOX inhibitor) or vehicle. Mice were evaluated for daily neuroscore and euthanized on day 5 to assess cerebral 12/15-LOX expression, vessel constrictions, platelet activation, microthrombi, neurodegeneration, infarction, cortical perfusion, and development of delayed deficits. Finally, the effect of 12/15-LOX inhibition on platelet activation was assessed in SAH patient samples using a platelet spreading assay. In SAH mice, 12/15-LOX was upregulated in brain vascular cells, and there was an increase in 12-S-HETE. Inhibition of 12/15-LOX improved brain perfusion on days 4-5 and attenuated delayed pathophysiological events, including microvessel constrictions, microthrombi, neuronal degeneration, and infarction. Additionally, 12/15-LOX inhibition reduced platelet activation in human and mouse blood samples. Cerebrovascular 12/15-LOX overexpression plays a major role in brain dysfunction after SAH by triggering microvessel constrictions and microthrombi formation, which reduces brain perfusion. Inhibiting 12/15-LOX may be a therapeutic target to improve outcomes after SAH.
Collapse
Affiliation(s)
- Ari Dienel
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77459, USA.
| | - Sung Ha Hong
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77459, USA
| | - Hussein A Zeineddine
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77459, USA
| | - Sithara Thomas
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77459, USA
| | - Shafeeque C M
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77459, USA
| | - Dania A Jose
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77459, USA
| | - Kiara Torres
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77459, USA
| | - Jose Guzman
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77459, USA
| | - Andrew Dunn
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
- Department of Electrical and Computer Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - P Kumar T
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77459, USA
| | - Gadiparthi N Rao
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Spiros L Blackburn
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77459, USA
| | - Devin W McBride
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77459, USA.
| |
Collapse
|
2
|
Dienel A, Hong SH, Zeineddine HA, Thomas S, Shafeeque CM, Jose DA, Torres K, Guzman J, Dunn A, P Kumar T, Rao GN, Blackburn SL, McBride DW. 12/15-Lipooxygenase Inhibition Reduces Microvessel Constriction and Microthrombi after Subarachnoid Hemorrhage in Mice. RESEARCH SQUARE 2024:rs.3.rs-4468292. [PMID: 38947083 PMCID: PMC11213206 DOI: 10.21203/rs.3.rs-4468292/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Background and Purpose Impaired cerebral circulation, induced by blood vessel constrictions and microthrombi, leads to delayed cerebral ischemia after subarachnoid hemorrhage (SAH). 12/15-Lipooxygenase (12/15-LOX) overexpression has been implicated in worsening early brain injury outcomes following SAH. However, it is unknown if 12/15-LOX is important in delayed pathophysiological events after SAH. Since 12/15-LOX produces metabolites that induce inflammation and vasoconstriction, we hypothesized that 12/15-LOX leads to microvessel constriction and microthrombi formation after SAH, and thus 12/15-LOX is an important target to prevent delayed cerebral ischemia. Methods SAH was induced in C57BL/6 and 12/15-LOX-/- mice of both sexes by endovascular perforation. Expression of 12/15-LOX was assessed in brain tissue slices and in vitro. C57BL/6 mice were administered either ML351 (12/15-LOX inhibitor) or vehicle. Mice were evaluated for daily neuroscore and euthanized on day five to assess cerebral 12/15-LOX expression, vessel constrictions, platelet activation, microthrombi, neurodegeneration, infarction, cortical perfusion, and for development of delayed deficits. Finally, the effect of 12/15-LOX inhibition on platelet activation was assessed in SAH patient samples using a platelet spreading assay. Results In SAH mice, 12/15-LOX was upregulated in brain vascular cells and there was an increase in 12-S-HETE. Inhibition of 12/15-LOX improved brain perfusion on days 4-5 and attenuated delayed pathophysiological events, including microvessel constrictions, microthrombi, neuronal degeneration, and infarction. Additionally, 12/15-LOX inhibition reduced platelet activation in human and mouse blood samples. Conclusions Cerebrovascular 12/15-LOX overexpression plays a major role in brain dysfunction after SAH by triggering microvessel constrictions and microthrombi formation, which reduces brain perfusion. Inhibiting 12/15-LOX may be a therapeutic target to improve outcomes after SAH.
Collapse
Affiliation(s)
- Ari Dienel
- The Vivian L. Smith, The University of Texas Health Science Center at Houston
| | - Sung Ha Hong
- The Vivian L. Smith, The University of Texas Health Science Center at Houston
| | | | - Sithara Thomas
- The Vivian L. Smith, The University of Texas Health Science Center at Houston
| | - C M Shafeeque
- The Vivian L. Smith, The University of Texas Health Science Center at Houston
| | - Dania A Jose
- The Vivian L. Smith, The University of Texas Health Science Center at Houston
| | - Kiara Torres
- The Vivian L. Smith, The University of Texas Health Science Center at Houston
| | - Jose Guzman
- The Vivian L. Smith, The University of Texas Health Science Center at Houston
| | | | - T P Kumar
- The Vivian L. Smith, The University of Texas Health Science Center at Houston
| | | | - Spiros L Blackburn
- The Vivian L. Smith, The University of Texas Health Science Center at Houston
| | - Devin W McBride
- The Vivian L. Smith, The University of Texas Health Science Center at Houston
| |
Collapse
|
3
|
He K, Zhou X, Du H, Zhao J, Deng R, Wang J. A review on the relationship between Arachidonic acid 15-Lipoxygenase (ALOX15) and diabetes mellitus. PeerJ 2023; 11:e16239. [PMID: 37849828 PMCID: PMC10578307 DOI: 10.7717/peerj.16239] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 09/14/2023] [Indexed: 10/19/2023] Open
Abstract
Arachidonic acid 15-lipoxygenase (ALOX15), as one of the lipoxygenase family, is mainly responsible for catalyzing the oxidation of various fatty acids to produce a variety of lipid components, contributing to the pathophysiological processes of various immune and inflammatory diseases. Studies have shown that ALOX15 and its related products are widely distributed in human tissues and related to multiple diseases such as liver, cardiovascular, cerebrovascular diseases, diabetes mellitus and other diseases. Diabetes mellitus (DM), the disease studied in this article, is a metabolic disease characterized by a chronic increase in blood glucose levels, which is significantly related to inflammation, oxidative stress, ferroptosis and other mechanisms, and it has a high incidence in the population, accompanied by a variety of complications. Figuring out how ALOX15 is involved in DM is critical to understanding its role in diseases. Therefore, ALOX15 inhibitors or combination therapy containing inhibitors may deliver a novel research direction for the treatment of DM and its complications. This article aims to review the biological effect and the possible function of ALOX15 in the pathogenesis of DM.
Collapse
Affiliation(s)
- Kaiying He
- Lanzhou University, Lanzhou, Gansu, China
- Lanzhou University Second Hospital, Lanzhou University, LanZhou, Gansu, China
| | - Xiaochun Zhou
- Lanzhou University Second Hospital, Lanzhou University, LanZhou, Gansu, China
| | - Hongxuan Du
- Lanzhou University, Lanzhou, Gansu, China
- Lanzhou University Second Hospital, Lanzhou University, LanZhou, Gansu, China
| | - Jing Zhao
- Lanzhou University, Lanzhou, Gansu, China
- Lanzhou University Second Hospital, Lanzhou University, LanZhou, Gansu, China
| | - Rongrong Deng
- Lanzhou University, Lanzhou, Gansu, China
- Lanzhou University Second Hospital, Lanzhou University, LanZhou, Gansu, China
| | - Jianqin Wang
- Lanzhou University Second Hospital, Lanzhou University, LanZhou, Gansu, China
| |
Collapse
|
4
|
Zhang Y, Liu Y, Sun J, Zhang W, Guo Z, Ma Q. Arachidonic acid metabolism in health and disease. MedComm (Beijing) 2023; 4:e363. [PMID: 37746665 PMCID: PMC10511835 DOI: 10.1002/mco2.363] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 08/13/2023] [Accepted: 08/17/2023] [Indexed: 09/26/2023] Open
Abstract
Arachidonic acid (AA), an n-6 essential fatty acid, is a major component of mammalian cells and can be released by phospholipase A2. Accumulating evidence indicates that AA plays essential biochemical roles, as it is the direct precursor of bioactive lipid metabolites of eicosanoids such as prostaglandins, leukotrienes, and epoxyeicosatrienoic acid obtained from three distinct enzymatic metabolic pathways: the cyclooxygenase pathway, lipoxygenase pathway, and cytochrome P450 pathway. AA metabolism is involved not only in cell differentiation, tissue development, and organ function but also in the progression of diseases, such as hepatic fibrosis, neurodegeneration, obesity, diabetes, and cancers. These eicosanoids are generally considered proinflammatory molecules, as they can trigger oxidative stress and stimulate the immune response. Therefore, interventions in AA metabolic pathways are effective ways to manage inflammatory-related diseases in the clinic. Currently, inhibitors targeting enzymes related to AA metabolic pathways are an important area of drug discovery. Moreover, many advances have also been made in clinical studies of AA metabolic inhibitors in combination with chemotherapy and immunotherapy. Herein, we review the discovery of AA and focus on AA metabolism in relation to health and diseases. Furthermore, inhibitors targeting AA metabolism are summarized, and potential clinical applications are discussed.
Collapse
Affiliation(s)
- Yiran Zhang
- Department of Orthopedic SurgeryOrthopedic Oncology InstituteThe Second Affiliated Hospital of Air Force Medical UniversityXi'anChina
| | - Yingxiang Liu
- Department of Orthopedic SurgeryOrthopedic Oncology InstituteThe Second Affiliated Hospital of Air Force Medical UniversityXi'anChina
| | - Jin Sun
- Department of Orthopedic SurgeryOrthopedic Oncology InstituteThe Second Affiliated Hospital of Air Force Medical UniversityXi'anChina
| | - Wei Zhang
- Department of PathologyThe Second Affiliated Hospital of Air Force Medical UniversityXi'anChina
| | - Zheng Guo
- Department of Orthopedic SurgeryOrthopedic Oncology InstituteThe Second Affiliated Hospital of Air Force Medical UniversityXi'anChina
| | - Qiong Ma
- Department of Orthopedic SurgeryOrthopedic Oncology InstituteThe Second Affiliated Hospital of Air Force Medical UniversityXi'anChina
- Department of PathologyThe Second Affiliated Hospital of Air Force Medical UniversityXi'anChina
| |
Collapse
|
5
|
Abstract
Hypertension is a major healthcare issue that afflicts one in every three adults worldwide and contributes to cardiovascular diseases, morbidity and mortality. Bioactive lipids contribute importantly to blood pressure regulation via actions on the vasculature, kidney, and inflammation. Vascular actions of bioactive lipids include blood pressure lowering vasodilation and blood pressure elevating vasoconstriction. Increased renin release by bioactive lipids in the kidney is pro-hypertensive whereas anti-hypertensive bioactive lipid actions result in increased sodium excretion. Bioactive lipids have pro-inflammatory and anti-inflammatory actions that increase or decrease reactive oxygen species and impact vascular and kidney function in hypertension. Human studies provide evidence that fatty acid metabolism and bioactive lipids contribute to sodium and blood pressure regulation in hypertension. Genetic changes identified in humans that impact arachidonic acid metabolism have been associated with hypertension. Arachidonic acid cyclooxygenase, lipoxygenase and cytochrome P450 metabolites have pro-hypertensive and anti-hypertensive actions. Omega-3 fish oil fatty acids eicosapentaenoic acid and docosahexaenoic acid are known to be anti-hypertensive and cardiovascular protective. Lastly, emerging fatty acid research areas include blood pressure regulation by isolevuglandins, nitrated fatty acids, and short chain fatty acids. Taken together, bioactive lipids are key contributors to blood pressure regulation and hypertension and their manipulation could decrease cardiovascular disease and associated morbidity and mortality.
Collapse
Affiliation(s)
- John D Imig
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States.
| |
Collapse
|
6
|
Kriska T, Herrnreiter A, Pfister SL, Adebesin A, Falck JR, Campbell WB. Macrophage 12(S)-HETE Enhances Angiotensin II-Induced Contraction by a BLT2 (Leukotriene B 4 Type-2 Receptor) and TP (Thromboxane Receptor)-Mediated Mechanism in Murine Arteries. Hypertension 2022; 79:104-114. [PMID: 34784723 PMCID: PMC8849474 DOI: 10.1161/hypertensionaha.121.17824] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
12/15-LO (12/15-lipoxygenase), encoded by Alox15 gene, metabolizes arachidonic acid to 12(S)-HETE (12-hydroxyeicosatetraenoic acid). Macrophages are the major source of 12/15-LO among immune cells, and 12/15-LO plays a crucial role in development of hypertension. Global Alox15- or macrophage-deficient mice are resistant to Ang II (angiotensin II)-induced hypertension. This study tests the hypothesis that macrophage 12(S)-HETE contributes to Ang II-mediated arterial constriction and thus to development of Ang II-induced hypertension. Ang II constricted isolated abdominal aortic and mesenteric arterial rings. 12(S)-HETE (100 nmol/L) alone was without effect; however, it significantly enhanced Ang II-induced constriction. The presence of wild-type macrophages also enhanced the Ang II-induced constriction, while Alox15-/- macrophages did not. Using this model, pretreatment of aortic rings with inhibitors, receptor agonists/antagonists, or removal of the endothelium, systematically uncovered an endothelium-mediated, Ang II receptor-2-mediated and superoxide-mediated enhancing effect of 12(S)-HETE on Ang II constrictions. The role of superoxide was confirmed using aortas from p47phox-/- mice where 12(S)-HETE failed to enhance constriction to Ang II. In cultured arterial endothelial cells, 12(S)-HETE increased the production of superoxide, and 12(S)-HETE or Ang II increased the production of an isothromboxane-like metabolite. A TP (thromboxane receptor) antagonist inhibited 12(S)-HETE enhancement of Ang II constriction. Both Ang II-induced hypertension and the enhancing effect of 12(S)-HETE on Ang II contractions were eliminated by a BLT2 (leukotriene B4 receptor-2) antagonist. These results outline a mechanism where the macrophage 12/15-LO pathway enhances the action of Ang II. 12(S)-HETE, acting on the BLT2, contributes to the hypertensive action of Ang II in part by promoting endothelial synthesis of a superoxide-derived TP agonist.
Collapse
Affiliation(s)
- Tamas Kriska
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee (T.K., A.H., S.L.P., W.B.C.)
| | - Anja Herrnreiter
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee (T.K., A.H., S.L.P., W.B.C.)
| | - Sandra L Pfister
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee (T.K., A.H., S.L.P., W.B.C.)
| | - Adeniyi Adebesin
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas (A.A., J.R.F.)
| | - John R Falck
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas (A.A., J.R.F.)
| | - William B Campbell
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee (T.K., A.H., S.L.P., W.B.C.)
| |
Collapse
|
7
|
Song CY, Singh P, Motiwala M, Shin JS, Lew J, Dutta SR, Gonzalez FJ, Bonventre JV, Malik KU. 2-Methoxyestradiol Ameliorates Angiotensin II-Induced Hypertension by Inhibiting Cytosolic Phospholipase A 2α Activity in Female Mice. Hypertension 2021; 78:1368-1381. [PMID: 34628937 PMCID: PMC8516072 DOI: 10.1161/hypertensionaha.121.18181] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Supplemental Digital Content is available in the text. We tested the hypothesis that CYP1B1 (cytochrome P450 1B1)-17β-estradiol metabolite 2-methoxyestradiol protects against Ang II (angiotensin II)–induced hypertension by inhibiting group IV cPLA2α (cytosolic phospholipase A2α) activity and production of prohypertensive eicosanoids in female mice. Ang II (700 ng/kg per minute, SC) increased mean arterial blood pressure (BP), systolic and diastolic BP measured by radiotelemetry, renal fibrosis, and reactive oxygen species production in wild-type mice (cPLA2α+/+/Cyp1b1+/+) that were enhanced by ovariectomy and abolished in intact and ovariectomized-cPLA2α−/−/Cyp1b1+/+ mice. Ang II–induced increase in SBP measured by tail-cuff, renal fibrosis, reactive oxygen species production, and cPLA2α activity measured by its phosphorylation in the kidney, and urinary excretion of prostaglandin E2 and thromboxane A2 metabolites were enhanced in ovariectomized-cPLA2α+/+/Cyp1b1+/+ and intact cPLA2α+/+/Cyp1b1−/− mice. 2-Methoxyestradiol and arachidonic acid metabolism inhibitor 5,8,11,14-eicosatetraynoic acid attenuated the Ang II–induced increase in SBP, renal fibrosis, reactive oxygen species production, and urinary excretion of prostaglandin E2, and thromboxane A2 metabolites in ovariectomized-cPLA2α+/+/Cyp1b1+/+ and intact cPLA2α+/+/Cyp1b1−/− mice. Antagonists of prostaglandin E2 and thromboxane A2 receptors EP1 and EP3 and TP, respectively, inhibited Ang II–induced increases in SBP and reactive oxygen species production and renal fibrosis in ovariectomized-cPLA2α+/+/Cyp1b1+/+ and intact cPLA2α+/+/Cyp1b1−/− mice. These data suggest that CYP1B1-generated metabolite 2-methoxyestradiol mitigates Ang II–induced hypertension and renal fibrosis by inhibiting cPLA2α activity, reducing prostaglandin E2, and thromboxane A2 production and stimulating EP1 and EP3 and TP receptors, respectively. Thus, 2-methoxyestradiol and the drugs that selectively block EP1 and EP3 and TP receptors could be useful in treating hypertension and its pathogenesis in females.
Collapse
Affiliation(s)
- Chi Young Song
- From the Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee HSC, Memphis (C.Y.S., P.S., M.M., J.S.S., J.L., S.R.D., K.U.M.)
| | - Purnima Singh
- From the Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee HSC, Memphis (C.Y.S., P.S., M.M., J.S.S., J.L., S.R.D., K.U.M.)
| | - Mustafa Motiwala
- From the Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee HSC, Memphis (C.Y.S., P.S., M.M., J.S.S., J.L., S.R.D., K.U.M.)
| | - Ji Soo Shin
- From the Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee HSC, Memphis (C.Y.S., P.S., M.M., J.S.S., J.L., S.R.D., K.U.M.)
| | - Jessica Lew
- From the Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee HSC, Memphis (C.Y.S., P.S., M.M., J.S.S., J.L., S.R.D., K.U.M.)
| | - Shubha R. Dutta
- From the Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee HSC, Memphis (C.Y.S., P.S., M.M., J.S.S., J.L., S.R.D., K.U.M.)
| | - Frank J. Gonzalez
- Laboratory of Metabolism, National Cancer Institute, Bethesda, MD (F.J.G.)
| | - Joseph V. Bonventre
- Renal Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA (J.V.B.)
| | - Kafait U. Malik
- From the Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee HSC, Memphis (C.Y.S., P.S., M.M., J.S.S., J.L., S.R.D., K.U.M.)
| |
Collapse
|
8
|
Singh P, Song CY, Dutta SR, Pingili A, Shin JS, Gonzalez FJ, Bonventre JV, Malik KU. 6β-Hydroxytestosterone Promotes Angiotensin II-Induced Hypertension via Enhanced Cytosolic Phospholipase A 2α Activity. Hypertension 2021; 78:1053-1066. [PMID: 34420370 PMCID: PMC8415516 DOI: 10.1161/hypertensionaha.121.17927] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Supplemental Digital Content is available in the text. This study was conducted to test the hypothesis that the CYP1B1 (cytochrome P450 1B1)-testosterone metabolite 6β-hydroxytestosterone contributes to angiotensin II-induced hypertension by promoting activation of group IV cPLA2α (cytosolic phospholipase A2α) and generation of prohypertensive eicosanoids in male mice. Eight-week-old male intact or orchidectomized cPLA2α+/+/Cyp1b1+/+ and cPLA2α–/–/Cyp1b1+/+ and intact cPLA2α+/+/Cyp1b1–/– mice were infused with angiotensin II (700 ng/kg/min, subcutaneous) for 2 weeks and injected with 6β-hydroxytestosterone (15 μg/g/every third day, intraperitoneal). Systolic blood pressure was measured by tail-cuff and confirmed by radiotelemetry. Angiotensin II-induced increase in systolic blood pressure, cardiac and renal collagen deposition, and reactive oxygen species production were reduced by disruption of the cPLA2α or Cyp1b1 genes or by administration of the arachidonic acid metabolism inhibitor 5,8,11,14-eicosatetraynoic acid to cPLA2α+/+/Cyp1b1+/+ mice. 6β-hydroxytestosterone treatment restored these effects of angiotensin II in cPLA2α+/+/Cyp1b1–/– mice but not in orchidectomized cPLA2α–/–/Cyp1b1+/+ mice, which were lowered by 5,8,11,14-eicosatetraynoic acid in cPLA2α+/+/Cyp1b1–/– mice. Antagonists of prostaglandin E2-EP1/EP3 receptors and thromboxane A2-TP receptors decreased the effect of 6β-hydroxytestosterone in restoring the angiotensin II-induced increase in systolic blood pressure, cardiac and renal collagen deposition, and reactive oxygen species production in cPLA2α+/+/Cyp1b1–/– mice. These data suggest that 6β-hydroxytestosterone promotes angiotensin II-induced increase in systolic blood pressure and associated pathogenesis via cPLA2α activation and generation of eicosanoids, most likely prostaglandin E2 and thromboxane A2 that exerts prohypertensive effects by stimulating EP1/EP3 and TP receptors, respectively. Therefore, agents that selectively block these receptors could be useful in treating testosterone exacerbated angiotensin II-induced hypertension and its pathogenesis.
Collapse
Affiliation(s)
- Purnima Singh
- Department of Pharmacology, Addiction Research, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis (P.S., C.Y.S., S.R.D., A.P., J.S.S., K.U.M.)
| | - Chi Young Song
- Department of Pharmacology, Addiction Research, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis (P.S., C.Y.S., S.R.D., A.P., J.S.S., K.U.M.)
| | - Shubha R Dutta
- Department of Pharmacology, Addiction Research, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis (P.S., C.Y.S., S.R.D., A.P., J.S.S., K.U.M.)
| | - Ajeeth Pingili
- Department of Pharmacology, Addiction Research, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis (P.S., C.Y.S., S.R.D., A.P., J.S.S., K.U.M.)
| | - Ji Soo Shin
- Department of Pharmacology, Addiction Research, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis (P.S., C.Y.S., S.R.D., A.P., J.S.S., K.U.M.)
| | - Frank J Gonzalez
- Laboratory of Metabolism, National Cancer Institute, Bethesda, MD (F.J.G.)
| | - Joseph V Bonventre
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Institute of Medicine, Boston, MA (J.V.B.)
| | - Kafait U Malik
- Department of Pharmacology, Addiction Research, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis (P.S., C.Y.S., S.R.D., A.P., J.S.S., K.U.M.)
| |
Collapse
|
9
|
Turolo S, Edefonti A, Mazzocchi A, Syren ML, Morello W, Agostoni C, Montini G. Role of Arachidonic Acid and Its Metabolites in the Biological and Clinical Manifestations of Idiopathic Nephrotic Syndrome. Int J Mol Sci 2021; 22:5452. [PMID: 34064238 PMCID: PMC8196840 DOI: 10.3390/ijms22115452] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 05/17/2021] [Accepted: 05/18/2021] [Indexed: 11/17/2022] Open
Abstract
Studies concerning the role of arachidonic acid (AA) and its metabolites in kidney disease are scarce, and this applies in particular to idiopathic nephrotic syndrome (INS). INS is one of the most frequent glomerular diseases in childhood; it is characterized by T-lymphocyte dysfunction, alterations of pro- and anti-coagulant factor levels, and increased platelet count and aggregation, leading to thrombophilia. AA and its metabolites are involved in several biological processes. Herein, we describe the main fields where they may play a significant role, particularly as it pertains to their effects on the kidney and the mechanisms underlying INS. AA and its metabolites influence cell membrane fluidity and permeability, modulate platelet activity and coagulation, regulate lymphocyte activity and inflammation, preserve the permeability of the glomerular barrier, influence podocyte physiology, and play a role in renal fibrosis. We also provide suggestions regarding dietary measures that are able to prevent an imbalance between arachidonic acid and its parental compound linoleic acid, in order to counteract the inflammatory state which characterizes numerous kidney diseases. On this basis, studies of AA in kidney disease appear as an important field to explore, with possible relevant results at the biological, dietary, and pharmacological level, in the final perspective for AA to modulate INS clinical manifestations.
Collapse
Affiliation(s)
- Stefano Turolo
- Fondazione IRCCS Ca’ Granda-Ospedale Maggiore Policlinico, Pediatric Nephrology, Dialysis and Transplant Unit, Via della Commenda 9, 20122 Milan, Italy; (A.E.); (W.M.); (G.M.)
| | - Alberto Edefonti
- Fondazione IRCCS Ca’ Granda-Ospedale Maggiore Policlinico, Pediatric Nephrology, Dialysis and Transplant Unit, Via della Commenda 9, 20122 Milan, Italy; (A.E.); (W.M.); (G.M.)
| | - Alessandra Mazzocchi
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy; (A.M.); (M.L.S.); (C.A.)
| | - Marie Louise Syren
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy; (A.M.); (M.L.S.); (C.A.)
| | - William Morello
- Fondazione IRCCS Ca’ Granda-Ospedale Maggiore Policlinico, Pediatric Nephrology, Dialysis and Transplant Unit, Via della Commenda 9, 20122 Milan, Italy; (A.E.); (W.M.); (G.M.)
| | - Carlo Agostoni
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy; (A.M.); (M.L.S.); (C.A.)
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Pediatric Intermediate Care Unit, 20122 Milan, Italy
| | - Giovanni Montini
- Fondazione IRCCS Ca’ Granda-Ospedale Maggiore Policlinico, Pediatric Nephrology, Dialysis and Transplant Unit, Via della Commenda 9, 20122 Milan, Italy; (A.E.); (W.M.); (G.M.)
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy; (A.M.); (M.L.S.); (C.A.)
| |
Collapse
|
10
|
Eicosanoid blood vessel regulation in physiological and pathological states. Clin Sci (Lond) 2021; 134:2707-2727. [PMID: 33095237 DOI: 10.1042/cs20191209] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/26/2020] [Accepted: 10/09/2020] [Indexed: 12/15/2022]
Abstract
Arachidonic acid can be metabolized in blood vessels by three primary enzymatic pathways; cyclooxygenase (COX), lipoxygenase (LO), and cytochrome P450 (CYP). These eicosanoid metabolites can influence endothelial and vascular smooth muscle cell function. COX metabolites can cause endothelium-dependent dilation or constriction. Prostaglandin I2 (PGI2) and thromboxane (TXA2) act on their respective receptors exerting opposing actions with regard to vascular tone and platelet aggregation. LO metabolites also influence vascular tone. The 12-LO metabolite 12S-hydroxyeicosatrienoic acid (12S-HETE) is a vasoconstrictor whereas the 15-LO metabolite 11,12,15-trihydroxyeicosatrienoic acid (11,12,15-THETA) is an endothelial-dependent hyperpolarizing factor (EDHF). CYP enzymes produce two types of eicosanoid products: EDHF vasodilator epoxyeicosatrienoic acids (EETs) and the vasoconstrictor 20-HETE. The less-studied cross-metabolites generated from arachidonic acid metabolism by multiple pathways can also impact vascular function. Likewise, COX, LO, and CYP vascular eicosanoids interact with paracrine and hormonal factors such as the renin-angiotensin system and endothelin-1 (ET-1) to maintain vascular homeostasis. Imbalances in endothelial and vascular smooth muscle cell COX, LO, and CYP metabolites in metabolic and cardiovascular diseases result in vascular dysfunction. Restoring the vascular balance of eicosanoids by genetic or pharmacological means can improve vascular function in metabolic and cardiovascular diseases. Nevertheless, future research is necessary to achieve a more complete understanding of how COX, LO, CYP, and cross-metabolites regulate vascular function in physiological and pathological states.
Collapse
|
11
|
Singh NK, Rao GN. Emerging role of 12/15-Lipoxygenase (ALOX15) in human pathologies. Prog Lipid Res 2019; 73:28-45. [PMID: 30472260 PMCID: PMC6338518 DOI: 10.1016/j.plipres.2018.11.001] [Citation(s) in RCA: 230] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 11/07/2018] [Accepted: 11/09/2018] [Indexed: 02/06/2023]
Abstract
12/15-lipoxygenase (12/15-LOX) is an enzyme, which oxidizes polyunsaturated fatty acids, particularly omega-6 and -3 fatty acids, to generate a number of bioactive lipid metabolites. A large number of studies have revealed the importance of 12/15-LOX role in oxidative and inflammatory responses. The in vitro studies have demonstrated the ability of 12/15-LOX metabolites in the expression of various genes and production of cytokine related to inflammation and resolution of inflammation. The studies with the use of knockout and transgenic animals for 12/15-LOX have further shown its involvement in the pathogenesis of a variety of human diseases, including cardiovascular, renal, neurological and metabolic disorders. This review summarizes our current knowledge on the role of 12/15-LOX in inflammation and various human diseases.
Collapse
Affiliation(s)
- Nikhlesh K Singh
- Department of Physiology, University of Tennessee Health Science Center, 71 S. Manassas Street Memphis, Memphis, TN 38163, USA
| | - Gadiparthi N Rao
- Department of Physiology, University of Tennessee Health Science Center, 71 S. Manassas Street Memphis, Memphis, TN 38163, USA.
| |
Collapse
|
12
|
Gouveia-Figueira S, Karimpour M, Bosson JA, Blomberg A, Unosson J, Sehlstedt M, Pourazar J, Sandström T, Behndig AF, Nording ML. Mass spectrometry profiling reveals altered plasma levels of monohydroxy fatty acids and related lipids in healthy humans after controlled exposure to biodiesel exhaust. Anal Chim Acta 2018; 1018:62-69. [PMID: 29605135 DOI: 10.1016/j.aca.2018.02.032] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 02/08/2018] [Accepted: 02/13/2018] [Indexed: 11/26/2022]
Abstract
Experimental human exposure studies are an effective tool to study adverse health effects from acute inhalation of particulate matter and other constituents of air pollution. In this randomized and double-blinded crossover study, we investigated the systemic effect on bioactive lipid metabolite levels after controlled biodiesel exhaust exposure of healthy humans and compared it to filtered air at a separate exposure occasion. Eicosanoids and other oxylipins, as well as endocannabinoids and related lipids, were quantified in plasma from 14 healthy volunteers at baseline and at three subsequent time points (2, 6, and 24 h) after 1 h exposure sessions. Protocols based on liquid chromatography (LC) coupled to tandem mass spectrometry (MS/MS) methods were developed to detect temporal changes in circulating levels after biodiesel exhaust exposure. The exhaust was generated by a diesel engine fed with an undiluted rapeseed methyl ester fuel. Among the 51 analyzed lipid metabolites, PGF2α, 9,10-DiHOME, 9-HODE, 5-HETE, 11-HETE, 12-HETE, and DEA displayed significant responsiveness to the biodiesel exhaust exposure as opposed to filtered air. Of these, 9-HODE and 5-HETE at 24 h survived the 10% false discovery rate cutoff (p < 0.003). Hence, the majority of the responsive lipid metabolites were monohydroxy fatty acids. We conclude that it is possible to detect alterations in circulating bioactive lipid metabolites in response to biodiesel exhaust exposure using LC-MS/MS, with emphasis on metabolites with inflammation related properties and implications on cardiovascular health and disease. These observations aid future investigations on air pollution effects, especially with regard to cardiovascular outcomes.
Collapse
Affiliation(s)
| | | | - Jenny A Bosson
- Department of Public Health and Clinical Medicine, Division of Medicine, Umeå University, 901 87 Umeå, Sweden
| | - Anders Blomberg
- Department of Public Health and Clinical Medicine, Division of Medicine, Umeå University, 901 87 Umeå, Sweden
| | - Jon Unosson
- Department of Public Health and Clinical Medicine, Division of Medicine, Umeå University, 901 87 Umeå, Sweden
| | - Maria Sehlstedt
- Department of Public Health and Clinical Medicine, Division of Medicine, Umeå University, 901 87 Umeå, Sweden
| | - Jamshid Pourazar
- Department of Public Health and Clinical Medicine, Division of Medicine, Umeå University, 901 87 Umeå, Sweden
| | - Thomas Sandström
- Department of Public Health and Clinical Medicine, Division of Medicine, Umeå University, 901 87 Umeå, Sweden
| | - Annelie F Behndig
- Department of Public Health and Clinical Medicine, Division of Medicine, Umeå University, 901 87 Umeå, Sweden
| | - Malin L Nording
- Department of Chemistry, Umeå University, 901 87 Umeå, Sweden.
| |
Collapse
|
13
|
Kriska T, Thomas MJ, Falck JR, Campbell WB. Deactivation of 12(S)-HETE through (ω-1)-hydroxylation and β-oxidation in alternatively activated macrophages. J Lipid Res 2018; 59:615-624. [PMID: 29472381 PMCID: PMC5880500 DOI: 10.1194/jlr.m081448] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 02/20/2018] [Indexed: 12/15/2022] Open
Abstract
Polarization of macrophages to proinflammatory M1 and to antiinflammatory alternatively activated M2 states has physiological implications in the development of experimental hypertension and other pathological conditions. 12/15-Lipoxygenase (12/15-LO) and its enzymatic products 12(S)- and 15(S)-hydroxyeicosatetraenoic acid (HETE) are essential in the process since disruption of the gene encoding 12/15-LO renders the mice unsusceptible to hypertension. The objective was to test the hypothesis that M2 macrophages catabolize 12(S)-HETE into products that are incapable of promoting vasoconstriction. Cultured M2 macrophages metabolized externally added [14C]12(S)-HETE into more polar metabolites, while M1 macrophages had little effect on the catabolism. The major metabolites were identified by mass spectrometry as (ω-1)-hydroxylation and β-oxidation products. The conversion was inhibited by both peroxisomal β-oxidation inhibitor, thioridazine, and cytochrome P450 inhibitors. Quantitative PCR analysis confirmed that several cytochrome P450 enzymes (CYP2E1 and CYP1B1) and peroxisomal β-oxidation markers were upregulated upon M2 polarization. The identified 12,19-dihydroxy-5,8,10,14-eicosatetraenoic acid and 8-hydroxy-6,10-hexadecadienoic acid metabolites were tested on abdominal aortic rings for biological activity. While 12(S)-HETE enhanced vasoconstrictions to angiotensin II from 15% to 25%, the metabolites did not. These results indicate that M2, but not M1, macrophages degrade 12(S)-HETE into products that no longer enhance the angiotensin II-induced vascular constriction, supporting a possible antihypertensive role of M2 macrophages.
Collapse
Affiliation(s)
- Tamas Kriska
- Department of Pharmacology and Toxicology,* Medical College of Wisconsin, Milwaukee, WI 53226.
| | - Michael J Thomas
- Department of Pharmacology and Toxicology,* Medical College of Wisconsin, Milwaukee, WI 53226
| | - John R Falck
- Department of Biochemistry,† University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - William B Campbell
- Department of Pharmacology and Toxicology,* Medical College of Wisconsin, Milwaukee, WI 53226
| |
Collapse
|
14
|
Liu X, Davis CM, Alkayed NJ. P450 Eicosanoids and Reactive Oxygen Species Interplay in Brain Injury and Neuroprotection. Antioxid Redox Signal 2018; 28:987-1007. [PMID: 28298143 PMCID: PMC5849284 DOI: 10.1089/ars.2017.7056] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Significance: Eicosanoids are endogenous lipid mediators that play important roles in brain function and disease. Acute brain injury such as that which occurs in stroke and traumatic brain injury increases the formation of eicosanoids, which, in turn, exacerbate or diminish injury. In chronic neurodegenerative diseases such as Alzheimer's disease and vascular dementia (VD), eicosanoid synthetic and metabolizing enzymes are altered, disrupting the balance between neuroprotective and neurotoxic eicosanoids. Recent Advances: Human and experimental studies have established the opposing roles of hydroxy- and epoxyeicosanoids and their potential utility as diagnostic biomarkers and therapeutic targets in neural injury. Critical Issues: A gap in knowledge remains in understanding the cellular and molecular mechanisms underlying the neurovascular actions of specific eicosanoids, such as specific isomers of epoxyeicosatrienoic (EETs) and hydroxyeicosatetraenoic acids (HETEs). Future Directions: EETs and HETEs exert their actions on brain cells by targeting multiple mechanisms, which include surface G-protein coupled receptors. The identification of high-affinity receptors for EETs and HETEs and their cellular localization in the brain will be a breakthrough in our understanding of these eicosanoids as mediators of cell-cell communications and contributors to brain development, function, and disease. Antioxid. Redox Signal. 28, 987-1007.
Collapse
Affiliation(s)
- Xuehong Liu
- The Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon
| | - Catherine M Davis
- The Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon.,Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, Oregon
| | - Nabil J Alkayed
- The Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon.,Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
15
|
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
|
16
|
Mote RS, Hill NS, Uppal K, Tran VT, Jones DP, Filipov NM. Metabolomics of fescue toxicosis in grazing beef steers. Food Chem Toxicol 2017; 105:285-299. [PMID: 28428084 DOI: 10.1016/j.fct.2017.04.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 03/25/2017] [Accepted: 04/16/2017] [Indexed: 12/25/2022]
Abstract
Fescue toxicosis (FT) results from consumption of tall fescue (Lolium arundinaceum) infected with an endophyte (Epichloë coenophiala) that produces ergot alkaloids (EA), which are considered key etiological agents of FT. Decreased weight gains, hormonal imbalance, circulating cholesterol disruption, and decreased volatile fatty acid absorption suggest toxic (E+) fescue-induced metabolic perturbations. Employing untargeted high-resolution metabolomics (HRM) to analyze E+ grazing-induced plasma and urine metabolome changes, fescue-naïve Angus steers were placed on E+ or non-toxic (Max-Q) fescue pastures and plasma and urine were sampled before, 1, 2, 14, and 28 days after pasture assignment. Plasma and urine catecholamines and urinary EA concentrations were also measured. In E+ steers, urinary EA appeared early and peaked at 14 days. 13,090 urinary and 20,908 plasma HRM features were detected; the most significant effects were observed earlier (2 days) in the urine and later (≥14 days) in the plasma. Alongside EA metabolite detection, tryptophan and lipid metabolism disruption were among the main consequences of E+ consumption. The E+ grazing-associated metabolic pathways and signatures described herein may accelerate development of novel early FT detection and treatment strategies.
Collapse
Affiliation(s)
- Ryan S Mote
- Interdisciplinary Toxicology Program, University of Georgia, Athens, GA, USA; Department of Physiology and Pharmacology, University of Georgia, Athens, GA, USA
| | - Nicholas S Hill
- Department of Crop and Soil Sciences, University of Georgia, Athens, GA, USA
| | - Karan Uppal
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Emory University, Atlanta, GA, USA
| | - ViLinh T Tran
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Emory University, Atlanta, GA, USA
| | - Dean P Jones
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Emory University, Atlanta, GA, USA
| | - Nikolay M Filipov
- Interdisciplinary Toxicology Program, University of Georgia, Athens, GA, USA; Department of Physiology and Pharmacology, University of Georgia, Athens, GA, USA.
| |
Collapse
|
17
|
Stadler S, Nguyen CH, Schachner H, Milovanovic D, Holzner S, Brenner S, Eichsteininger J, Stadler M, Senfter D, Krenn L, Schmidt WM, Huttary N, Krieger S, Koperek O, Bago-Horvath Z, Brendel KA, Marian B, de Wever O, Mader RM, Giessrigl B, Jäger W, Dolznig H, Krupitza G. Colon cancer cell-derived 12(S)-HETE induces the retraction of cancer-associated fibroblast via MLC2, RHO/ROCK and Ca 2+ signalling. Cell Mol Life Sci 2016; 74:1907-1921. [PMID: 28013338 PMCID: PMC5390003 DOI: 10.1007/s00018-016-2441-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 12/06/2016] [Accepted: 12/09/2016] [Indexed: 12/24/2022]
Abstract
Retraction of mesenchymal stromal cells supports the invasion of colorectal cancer cells (CRC) into the adjacent compartment. CRC-secreted 12(S)-HETE enhances the retraction of cancer-associated fibroblasts (CAFs) and therefore, 12(S)-HETE may enforce invasivity of CRC. Understanding the mechanisms of metastatic CRC is crucial for successful intervention. Therefore, we studied pro-invasive contributions of stromal cells in physiologically relevant three-dimensional in vitro assays consisting of CRC spheroids, CAFs, extracellular matrix and endothelial cells, as well as in reductionist models. In order to elucidate how CAFs support CRC invasion, tumour spheroid-induced CAF retraction and free intracellular Ca2+ levels were measured and pharmacological- or siRNA-based inhibition of selected signalling cascades was performed. CRC spheroids caused the retraction of CAFs, generating entry gates in the adjacent surrogate stroma. The responsible trigger factor 12(S)-HETE provoked a signal, which was transduced by PLC, IP3, free intracellular Ca2+, Ca2+-calmodulin-kinase-II, RHO/ROCK and MYLK which led to the activation of myosin light chain 2, and subsequent CAF mobility. RHO activity was observed downstream as well as upstream of Ca2+ release. Thus, Ca2+ signalling served as central signal amplifier. Treatment with the FDA-approved drugs carbamazepine, cinnarizine, nifedipine and bepridil HCl, which reportedly interfere with cellular calcium availability, inhibited CAF-retraction. The elucidation of signalling pathways and identification of approved inhibitory drugs warrant development of intervention strategies targeting tumour–stroma interaction.
Collapse
Affiliation(s)
- Serena Stadler
- Clinical Institute of Pathology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Institute of Medical Genetics, Medical University of Vienna, Waehringer Strasse 10, 1090, Vienna, Austria
| | - Chi Huu Nguyen
- Clinical Institute of Pathology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Department for Clinical Pharmacy and Diagnostics, Faculty of Life Sciences, University of Vienna, Althanstrasse 14, 1090, Vienna, Austria
| | - Helga Schachner
- Clinical Institute of Pathology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Daniela Milovanovic
- Clinical Institute of Pathology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Silvio Holzner
- Clinical Institute of Pathology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Institute of Medical Genetics, Medical University of Vienna, Waehringer Strasse 10, 1090, Vienna, Austria
- Department of Medicine I, Comprehensive Cancer Centre, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Stefan Brenner
- Department for Clinical Pharmacy and Diagnostics, Faculty of Life Sciences, University of Vienna, Althanstrasse 14, 1090, Vienna, Austria
| | - Julia Eichsteininger
- Clinical Institute of Pathology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Department of Pharmacognosy, Faculty of Life Sciences, University of Vienna, Althanstrasse 14, 1090, Vienna, Austria
| | - Mira Stadler
- Institute of Medical Genetics, Medical University of Vienna, Waehringer Strasse 10, 1090, Vienna, Austria
| | - Daniel Senfter
- Clinical Institute of Pathology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Institute of Medical Genetics, Medical University of Vienna, Waehringer Strasse 10, 1090, Vienna, Austria
- Department of Medicine I, Comprehensive Cancer Centre, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Liselotte Krenn
- Department of Pharmacognosy, Faculty of Life Sciences, University of Vienna, Althanstrasse 14, 1090, Vienna, Austria
| | - Wolfgang M Schmidt
- Neuromuscular Research Department, Centre of Anatomy and Cell Biology, Medical University of Vienna, Waehringer Strasse 13, 1090, Vienna, Austria
| | - Nicole Huttary
- Clinical Institute of Pathology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Sigurd Krieger
- Clinical Institute of Pathology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Oskar Koperek
- Clinical Institute of Pathology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Zsuzsanna Bago-Horvath
- Clinical Institute of Pathology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | | | - Brigitte Marian
- Department of Medicine I, Institute of Cancer Research and Comprehensive Cancer Centre, Medical University of Vienna, Borschkegasse 8a, 1090, Vienna, Austria
| | - Oliver de Wever
- Department of Radiation Oncology and Experimental Cancer Research, Ghent University, De Pintelaan 185, 9000, Ghent, Belgium
| | - Robert M Mader
- Department of Medicine I, Comprehensive Cancer Centre, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Benedikt Giessrigl
- Clinical Institute of Pathology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Department for Clinical Pharmacy and Diagnostics, Faculty of Life Sciences, University of Vienna, Althanstrasse 14, 1090, Vienna, Austria
| | - Walter Jäger
- Department for Clinical Pharmacy and Diagnostics, Faculty of Life Sciences, University of Vienna, Althanstrasse 14, 1090, Vienna, Austria
| | - Helmut Dolznig
- Institute of Medical Genetics, Medical University of Vienna, Waehringer Strasse 10, 1090, Vienna, Austria
| | - Georg Krupitza
- Clinical Institute of Pathology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.
| |
Collapse
|
18
|
Nguyen CH, Brenner S, Huttary N, Li Y, Atanasov AG, Dirsch VM, Holzner S, Stadler S, Riha J, Krieger S, Milovanovic D, Fristiohardy A, Simonitsch-Klupp I, Dolznig H, Saiko P, Szekeres T, Giessrigl B, Jäger W, Krupitza G. 12(S)-HETE increases intracellular Ca2+ in lymph-endothelial cells disrupting their barrier function in vitro; stabilization by clinical drugs impairing calcium supply. Cancer Lett 2016; 380:174-83. [DOI: 10.1016/j.canlet.2016.06.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 06/28/2016] [Accepted: 06/28/2016] [Indexed: 02/06/2023]
|
19
|
Maayah ZH, Althurwi HN, Abdelhamid G, Lesyk G, Jurasz P, El-Kadi AO. CYP1B1 inhibition attenuates doxorubicin-induced cardiotoxicity through a mid-chain HETEs-dependent mechanism. Pharmacol Res 2016; 105:28-43. [DOI: 10.1016/j.phrs.2015.12.016] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 12/01/2015] [Accepted: 12/15/2015] [Indexed: 12/20/2022]
|
20
|
Khan NS, Song CY, Thirunavukkarasu S, Fang XR, Bonventre JV, Malik KU. Cytosolic Phospholipase A2α Is Essential for Renal Dysfunction and End-Organ Damage Associated With Angiotensin II-Induced Hypertension. Am J Hypertens 2016; 29:258-65. [PMID: 26045535 DOI: 10.1093/ajh/hpv083] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 05/13/2015] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND The kidney plays an important role in regulating blood pressure (BP). cPLA2α in the kidney is activated by various agents including angiotensin II (Ang II) and selectively releases arachidonic acid (AA) from tissue lipids, generating pro- and antihypertensive eicosanoids. Since activation of cPLA2α is the rate-limiting step in AA release, this study was conducted to determine its contribution to renal dysfunction and end-organ damage associated with Ang II-induced hypertension. METHODS cPLA2α(+/+) and cPLA2α(-/-) mice were infused with Ang II (700 ng/ kg/min) or its vehicle for 13 days. Mice were placed in metabolic cages to monitor their food and water intake, and urine was collected and its volume was measured. Doppler imaging was performed to assess renal hemodynamics. On the 13th day of Ang II infusion, mice were sacrificed and their tissues and blood collected for further analysis. RESULTS Ang II increased renal vascular resistance, water intake, and urine output and Na(+) excretion, decreased urine osmolality, and produced proteinuria in cPLA2α(+/+) mice. Ang II also caused accumulation of F4/80(+) macrophages and CD3(+) T cells and renal fibrosis, and increased oxidative stress in the kidneys of cPLA2α(+/+) mice. All these effects of Ang II were minimized in cPLA2α(-/-) mice. CONCLUSION cPLA2α contributes to renal dysfunction, inflammation, and end-organ damage, most likely via the action of pro-hypertensive eicosanoids and increased oxidative stress associated with Ang II-induced hypertension. Thus, cPLA2α could serve as a potential therapeutic target for treating renal dysfunction and end-organ damage in hypertension.
Collapse
Affiliation(s)
- Nayaab S Khan
- Department of Pharmacology, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Chi Young Song
- Department of Pharmacology, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Shyamala Thirunavukkarasu
- Department of Pharmacology, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Xiao R Fang
- Department of Pharmacology, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Joseph V Bonventre
- Renal Division, Department of Medicine, Brigham and Women's Hospital Boston, Harvard Medical School, Harvard Institute of Medicine, Boston, Massachusetts, USA
| | - Kafait U Malik
- Department of Pharmacology, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA;
| |
Collapse
|
21
|
Abstract
Arachidonic acid metabolites have a myriad of biological actions including effects on the kidney to alter renal hemodynamics and tubular transport processes. Cyclooxygenase metabolites are products of an arachidonic acid enzymatic pathway that has been extensively studied in regards to renal function. Two lesser-known enzymatic pathways of arachidonic acid metabolism are the lipoxygenase (LO) and cytochrome P450 (CYP) pathways. The importance of LO and CYP metabolites to renal hemodynamics and tubular transport processes is now being recognized. LO and CYP metabolites have actions to alter renal blood flow and glomerular filtration rate. Proximal and distal tubular sodium transport and fluid and electrolyte homeostasis are also significantly influenced by renal CYP and LO levels. Metabolites of the LO and CYP pathways also have renal actions that influence renal inflammation, proliferation, and apoptotic processes at vascular and epithelial cells. These renal LO and CYP pathway actions occur through generation of specific metabolites and cell-signaling mechanisms. Even though the renal physiological importance and actions for LO and CYP metabolites are readily apparent, major gaps remain in our understanding of these lipid mediators to renal function. Future studies will be needed to fill these major gaps regarding LO and CYP metabolites on renal function.
Collapse
Affiliation(s)
- John D Imig
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Md Abdul Hye Khan
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
22
|
The role of mid-chain hydroxyeicosatetraenoic acids in the pathogenesis of hypertension and cardiac hypertrophy. Arch Toxicol 2015; 90:119-36. [PMID: 26525395 DOI: 10.1007/s00204-015-1620-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 10/19/2015] [Indexed: 12/16/2022]
Abstract
The incidence, prevalence, and hospitalization rates associated with cardiovascular diseases (CVDs) are projected to increase substantially in the world. Understanding of the biological and pathophysiological mechanisms of survival can help the researchers to develop new management modalities. Numerous experimental studies have demonstrated that mid-chain HETEs are strongly involved in the pathogenesis of the CVDs. Mid-chain HETEs are biologically active eicosanoids that result from the metabolism of arachidonic acid (AA) by both lipoxygenase and CYP1B1 (lipoxygenase-like reaction). Therefore, identifying the localizations and expressions of the lipoxygenase and CYP1B1 and their associated AA metabolites in the cardiovascular system is of major importance in understanding their pathological roles. Generally, the expression of these enzymes is shown to be induced during several CVDs, including hypertension and cardiac hypertrophy. The induction of these enzymes is associated with the generation of mid-chain HETEs and subsequently causation of cardiovascular events. Of interest, inhibiting the formation of mid-chain HETEs has been reported to confer a protection against different cardiac hypertrophy and hypertension models such as angiotensin II, Goldblatt, spontaneously hypertensive rat and deoxycorticosterone acetate (DOCA)-salt-induced models. Although the exact mechanisms of mid-chain HETEs-mediated cardiovascular dysfunction are not fully understood, the present review proposes several mechanisms which include activating G-protein-coupled receptor, protein kinase C, mitogen-activated protein kinases, and nuclear factor kappa B. This review provides a clear understanding of the role of mid-chain HETEs in the pathogenesis of cardiovascular diseases and their importance as novel targets in the treatment for hypertension and cardiac hypertrophy.
Collapse
|
23
|
Pichler Hefti J, Sonntag D, Hefti U, Risch L, Schoch OD, Turk AJ, Hess T, Bloch KE, Maggiorini M, Merz TM, Weinberger KM, Huber AR. Oxidative stress in hypobaric hypoxia and influence on vessel-tone modifying mediators. High Alt Med Biol 2014; 14:273-9. [PMID: 24067187 DOI: 10.1089/ham.2012.1110] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Increased pulmonary artery pressure is a well-known phenomenon of hypoxia and is seen in patients with chronic pulmonary diseases, and also in mountaineers on high altitude expedition. Different mediators are known to regulate pulmonary artery vessel tone. However, exact mechanisms are not fully understood and a multimodal process consisting of a whole panel of mediators is supposed to cause pulmonary artery vasoconstriction. We hypothesized that increased hypoxemia is associated with an increase in vasoconstrictive mediators and decrease of vasodilatators leading to a vasoconstrictive net effect. Furthermore, we suggested oxidative stress being partly involved in changement of these parameters. Oxygen saturation (Sao2) and clinical parameters were assessed in 34 volunteers before and during a Swiss research expedition to Mount Muztagh Ata (7549 m) in Western China. Blood samples were taken at four different sites up to an altitude of 6865 m. A mass spectrometry-based targeted metabolomic platform was used to detect multiple parameters, and revealed functional impairment of enzymes that require oxidation-sensitive cofactors. Specifically, the tetrahydrobiopterin (BH4)-dependent enzyme nitric oxide synthase (NOS) showed significantly lower activities (citrulline-to-arginine ratio decreased from baseline median 0.21 to 0.14 at 6265 m), indicating lower NO availability resulting in less vasodilatative activity. Correspondingly, an increase in systemic oxidative stress was found with a significant increase of the percentage of methionine sulfoxide from a median 6% under normoxic condition to a median level of 30% (p<0.001) in camp 1 at 5533 m. Furthermore, significant increase in vasoconstrictive mediators (e.g., tryptophan, serotonin, and peroxidation-sensitive lipids) were found. During ascent up to 6865 m, significant altitude-dependent changes in multiple vessel-tone modifying mediators with excess in vasoconstrictive metabolites could be demonstrated. These changes, as well as highly significant increase in systemic oxidative stress, may be predictive for increase in acute mountain sickness score and changes in Sao2.
Collapse
Affiliation(s)
- Jacqueline Pichler Hefti
- 1 Center of Laboratory Medicine, Cantonal Hospital Aarau and University of Bern , Bern, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Lu W, Zhao X, Xu Z, Dong N, Zou S, Shen X, Huang J. Development of a new colorimetric assay for lipoxygenase activity. Anal Biochem 2013; 441:162-8. [DOI: 10.1016/j.ab.2013.06.007] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2013] [Revised: 06/12/2013] [Accepted: 06/13/2013] [Indexed: 01/01/2023]
|
25
|
Dobrian AD, Lieb DC, Cole BK, Taylor-Fishwick DA, Chakrabarti SK, Nadler JL. Functional and pathological roles of the 12- and 15-lipoxygenases. Prog Lipid Res 2011; 50:115-31. [PMID: 20970452 PMCID: PMC3012140 DOI: 10.1016/j.plipres.2010.10.005] [Citation(s) in RCA: 250] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Revised: 10/13/2010] [Accepted: 10/14/2010] [Indexed: 12/25/2022]
Abstract
The 12/15-lipoxygenase enzymes react with fatty acids producing active lipid metabolites that are involved in a number of significant disease states. The latter include type 1 and type 2 diabetes (and associated complications), cardiovascular disease, hypertension, renal disease, and the neurological conditions Alzheimer's disease and Parkinson's disease. A number of elegant studies over the last thirty years have contributed to unraveling the role that lipoxygenases play in chronic inflammation. The development of animal models with targeted gene deletions has led to a better understanding of the role that lipoxygenases play in various conditions. Selective inhibitors of the different lipoxygenase isoforms are an active area of investigation, and will be both an important research tool and a promising therapeutic target for treating a wide spectrum of human diseases.
Collapse
Affiliation(s)
- Anca D Dobrian
- Eastern Virginia Medical School, Department of Physiological Sciences, Lewis Hall, Room 2027, 700 W. Olney Road, Norfolk, VA 23507, United States.
| | | | | | | | | | | |
Collapse
|
26
|
Inscho EW. ATP, P2 receptors and the renal microcirculation. Purinergic Signal 2009; 5:447-60. [PMID: 19294530 PMCID: PMC2776135 DOI: 10.1007/s11302-009-9147-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2007] [Accepted: 05/16/2008] [Indexed: 01/22/2023] Open
Abstract
Purinoceptors are rapidly becoming recognised as important regulators of tissue and organ function. Renal expression of P2 receptors is broad and diverse, as reflected by the fact that P2 receptors have been identified in virtually every major tubular/vascular element. While P2 receptor expression by these renal structures is recognised, the physiological functions that they serve remains to be clarified. Renal vascular P2 receptor expression is complex and poorly understood. Evidence suggests that different complements of P2 receptors are expressed by individual renal vascular segments. This unique distribution has given rise to the postulate that P2 receptors are important for renal vascular function, including regulation of preglomerular resistance and autoregulatory behaviour. More recent studies have also uncovered evidence that hypertension reduces renal vascular reactivity to P2 receptor stimulation in concert with compromised autoregulatory capability. This review will consolidate findings related to the role of P2 receptors in regulating renal microvascular function and will present areas of controversy related to the respective roles of ATP and adenosine in autoregulatory resistance adjustments.
Collapse
Affiliation(s)
- Edward W Inscho
- Department of Physiology, Medical College of Georgia, Augusta, Georgia,
| |
Collapse
|
27
|
Dołegowska B, Błogowski W, Kedzierska K, Safranow K, Jakubowska K, Olszewska M, Rać M, Chlubek D, Ciechanowski K. Platelets arachidonic acid metabolism in patients with essential hypertension. Platelets 2009; 20:242-9. [PMID: 19440924 DOI: 10.1080/09537100902849836] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Arachidonic acid's (AA) metabolites, eicosanoids, exert a tremendous influence on circulatory and vascular homeostasis, and in humans are generated by many organs and cell types. In this study we wanted to verify whether platelets AA metabolism play a significant role in pathogenesis of essential hypertension (EH). Participants were divided into the study (EH) and the control group. Plasma and urine concentrations of isoprostanes (8-iPF(2alpha)-III) and thromboxane B(2) (TxB(2)) were determined using the ELISA method. The levels of 5- and 12-hydroxyeicosatetraenoic (HETE) acids, generated by platelets, were analysed using RP-HPLC. In a suspension of not stimulated and AA-stimulated platelets TxB(2) level was statistically lower in the study than in the control group (p < 0.0001 and 0.001 respectively). The concentration of 12-HETE was significantly elevated in EH patients compared to the control group; however, only in the non-stimulated conditions (p < 0.05). Plasma and urine F2-isoprostanes levels were significantly higher in hypertensive individuals than in the control group (p < 0.00002 and p < 0.01 respectively). Moreover, EH patients excreted more TxB(2) in urine than normotensive individuals (p < 0.05). Our results highlight the mutual connections between the platelets AA metabolism and indicate its possible role in the pathogenesis of arterial hypertension. Moreover, we hypothesize that platelets AA metabolism may exert a pro-atherosclerotic effect. Finally, we suggest the use of (5-HETE+12-HETE)/TxB(2) parameter in further studies.
Collapse
Affiliation(s)
- Barbara Dołegowska
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Szczecin, Poland
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Ponnuchamy B, Khalil RA. Cellular mediators of renal vascular dysfunction in hypertension. Am J Physiol Regul Integr Comp Physiol 2009; 296:R1001-18. [PMID: 19225145 PMCID: PMC2698613 DOI: 10.1152/ajpregu.90960.2008] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2008] [Accepted: 02/17/2009] [Indexed: 02/07/2023]
Abstract
The renal vasculature plays a major role in the regulation of renal blood flow and the ability of the kidney to control the plasma volume and blood pressure. Renal vascular dysfunction is associated with renal vasoconstriction, decreased renal blood flow, and consequent increase in plasma volume and has been demonstrated in several forms of hypertension (HTN), including genetic and salt-sensitive HTN. Several predisposing factors and cellular mediators have been implicated, but the relationship between their actions on the renal vasculature and the consequent effects on renal tubular function in the setting of HTN is not clearly defined. Gene mutations/defects in an ion channel, a membrane ion transporter, and/or a regulatory enzyme in the nephron and renal vasculature may be a primary cause of renal vascular dysfunction. Environmental risk factors, such as high dietary salt intake, vascular inflammation, and oxidative stress further promote renal vascular dysfunction. Renal endothelial cell dysfunction is manifested as a decrease in the release of vasodilatory mediators, such as nitric oxide, prostacyclin, and hyperpolarizing factors, and/or an increase in vasoconstrictive mediators, such as endothelin, angiotensin II, and thromboxane A(2). Also, an increase in the amount/activity of intracellular Ca(2+) concentration, protein kinase C, Rho kinase, and mitogen-activated protein kinase in vascular smooth muscle promotes renal vasoconstriction. Matrix metalloproteinases and their inhibitors could also modify the composition of the extracellular matrix and lead to renal vascular remodeling. Synergistic interactions between the genetic and environmental risk factors on the cellular mediators of renal vascular dysfunction cause persistent renal vasoconstriction, increased renal vascular resistance, and decreased renal blood flow, and, consequently, lead to a disturbance in the renal control mechanisms of water and electrolyte balance, increased plasma volume, and HTN. Targeting the underlying genetic defects, environmental risk factors, and the aberrant renal vascular mediators involved should provide complementary strategies in the management of HTN.
Collapse
|
29
|
Inscho EW, Cook AK, Webb RC, Jin LM. Rho-kinase inhibition reduces pressure-mediated autoregulatory adjustments in afferent arteriolar diameter. Am J Physiol Renal Physiol 2009; 296:F590-7. [PMID: 19129253 DOI: 10.1152/ajprenal.90703.2008] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Preglomerular resistance is regulated by calcium influx- and mobilization-dependent mechanisms; however, the role of Rho-kinase in calcium sensitization in the intact kidney has not been carefully examined. Experiments were performed to test the hypothesis that Rho-kinase inhibition blunts pressure-mediated afferent arteriolar autoregulatory behavior and vasoconstrictor responses evoked by angiotensin II and P2X1 receptor activation. Rat kidneys were studied in vitro using the blood-perfused juxtamedullary nephron technique. Autoregulatory behavior was assessed before and during Rho-kinase inhibition with Y-27632 (1.0 microM; n = 5). Control diameter averaged 14.3 +/- 0.8 microm and increased to 18.1 +/- 0.9 microm (P < 0.05) during Y-27632 treatment. In the continued presence of Y-27632, reducing perfusion pressure to 65 mmHg slightly increased diameter to 18.7 +/- 1.0 microm. Subsequent pressure increases to 130 and 160 mmHg yielded afferent arteriolar diameters of 17.5 +/- 0.8 and 16.6 +/- 0.6 microm (P < 0.05). This 11% decline in diameter is significantly smaller than the 40% decrease obtained in untreated kidneys. The inhibitory effects of Y-27632 on autoregulatory behavior were concentration dependent. Angiotensin II responses were blunted by Y-27632. Angiotensin II (1.0 nM) reduced afferent diameter by 17 +/- 1% in untreated arterioles and by 6 +/- 2% during exposure to Y-27632. The P2X1 receptor agonist, alpha, beta-methylene ATP, reduced afferent arteriolar diameter by 8 +/- 1% but this response was eliminated during exposure to Y-27632. Western blot analysis confirms expression of the Rho-kinase signaling pathway. Thus, Rho-kinase may be important in pressure-mediated autoregulatory adjustments in preglomerular resistance and responsiveness to angiotensin II and autoregulatory P2X1 receptor agonists.
Collapse
Affiliation(s)
- Edward W Inscho
- Department of Physiology, Medical College of Georgia, 1120 15th St., Augusta, Georgia 30912-3000, USA.
| | | | | | | |
Collapse
|
30
|
Navar LG, Arendshorst WJ, Pallone TL, Inscho EW, Imig JD, Bell PD. The Renal Microcirculation. Compr Physiol 2008. [DOI: 10.1002/cphy.cp020413] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
31
|
Legrand M, Mik EG, Johannes T, Payen D, Ince C. Renal hypoxia and dysoxia after reperfusion of the ischemic kidney. MOLECULAR MEDICINE (CAMBRIDGE, MASS.) 2008. [PMID: 18488066 DOI: 10.2119/2008-00006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Ischemia is the most common cause of acute renal failure. Ischemic-induced renal tissue hypoxia is thought to be a major component in the development of acute renal failure in promoting the initial tubular damage. Renal oxygenation originates from a balance between oxygen supply and consumption. Recent investigations have provided new insights into alterations in oxygenation pathways in the ischemic kidney. These findings have identified a central role of microvascular dysfunction related to an imbalance between vasoconstrictors and vasodilators, endothelial damage and endothelium-leukocyte interactions, leading to decreased renal oxygen supply. Reduced microcirculatory oxygen supply may be associated with altered cellular oxygen consumption (dysoxia), because of mitochondrial dysfunction and activity of alternative oxygen-consuming pathways. Alterations in oxygen utilization and/or supply might therefore contribute to the occurrence of organ dysfunction. This view places oxygen pathways' alterations as a potential central player in the pathogenesis of acute kidney injury. Both in regulation of oxygen supply and consumption, nitric oxide seems to play a pivotal role. Furthermore, recent studies suggest that, following acute ischemic renal injury, persistent tissue hypoxia contributes to the development of chronic renal dysfunction. Adaptative mechanisms to renal hypoxia may be ineffective in more severe cases and lead to the development of chronic renal failure following ischemia-reperfusion. This paper is aimed at reviewing the current insights into oxygen transport pathways, from oxygen supply to oxygen consumption in the kidney and from the adaptation mechanisms to renal hypoxia. Their role in the development of ischemia-induced renal damage and ischemic acute renal failure are discussed.
Collapse
Affiliation(s)
- Matthieu Legrand
- Department of Physiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | | | | | | | | |
Collapse
|
32
|
Legrand M, Mik EG, Johannes T, Payen D, Ince C. Renal hypoxia and dysoxia after reperfusion of the ischemic kidney. Mol Med 2008; 14:502-16. [PMID: 18488066 DOI: 10.2119/2008-00006.legrand] [Citation(s) in RCA: 198] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2008] [Accepted: 04/17/2008] [Indexed: 12/18/2022] Open
Abstract
Ischemia is the most common cause of acute renal failure. Ischemic-induced renal tissue hypoxia is thought to be a major component in the development of acute renal failure in promoting the initial tubular damage. Renal oxygenation originates from a balance between oxygen supply and consumption. Recent investigations have provided new insights into alterations in oxygenation pathways in the ischemic kidney. These findings have identified a central role of microvascular dysfunction related to an imbalance between vasoconstrictors and vasodilators, endothelial damage and endothelium-leukocyte interactions, leading to decreased renal oxygen supply. Reduced microcirculatory oxygen supply may be associated with altered cellular oxygen consumption (dysoxia), because of mitochondrial dysfunction and activity of alternative oxygen-consuming pathways. Alterations in oxygen utilization and/or supply might therefore contribute to the occurrence of organ dysfunction. This view places oxygen pathways' alterations as a potential central player in the pathogenesis of acute kidney injury. Both in regulation of oxygen supply and consumption, nitric oxide seems to play a pivotal role. Furthermore, recent studies suggest that, following acute ischemic renal injury, persistent tissue hypoxia contributes to the development of chronic renal dysfunction. Adaptative mechanisms to renal hypoxia may be ineffective in more severe cases and lead to the development of chronic renal failure following ischemia-reperfusion. This paper is aimed at reviewing the current insights into oxygen transport pathways, from oxygen supply to oxygen consumption in the kidney and from the adaptation mechanisms to renal hypoxia. Their role in the development of ischemia-induced renal damage and ischemic acute renal failure are discussed.
Collapse
Affiliation(s)
- Matthieu Legrand
- Department of Physiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | | | | | | | | |
Collapse
|
33
|
Abdel-Rahman EM, Abadir PM, Siragy HM. Regulation of renal 12(S)-hydroxyeicosatetraenoic acid in diabetes by angiotensin AT1 and AT2 receptors. Am J Physiol Regul Integr Comp Physiol 2008; 295:R1473-8. [PMID: 18799632 DOI: 10.1152/ajpregu.90699.2008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Diabetes is associated with increased production of 12(S)-hydroxyeicosatetraenoic acid [12(S)-HETE]. The mechanisms involved in this process remain unclear. We hypothesized that hyperglycemia and angiotensin II (ANG II) regulate renal 12(S)-HETE production via a balance between angiotensin AT(1) and AT(2) receptors activities. Using a microdialysis technique, renal interstitial fluid (RIF) levels of ANG II and 12(S)-HETE were monitored in normal control and streptozotocin-induced diabetic rats at baseline and then weekly thereafter for 12 wk. In a second group of normal and diabetic rats, 3 wk after development of diabetes, we monitored RIF 12(S)-HETE levels in response to acute AT(1) receptor blockade with valsartan or AT(2) receptor blockade with PD123319 individually or combined. Two weeks after induction of diabetes there was a 404% increase in ANG II (P < 0.05), a 149% increase in 12S-HETE (P < 0.05), and a 649% increase in urinary albumin excretion (P < 0.05). These levels remained elevated throughout the study. PD123319 given alone had no effect on 12(S)-HETE. Valsartan decreased 12(S)-HETE by 61.6% (P < 0.0001), a response that was abrogated when PD123319 was given with valsartan. These data demonstrate that hyperglycemia increases renal ANG II and 12(S)-HETE levels. The increase in 12(S)-HETE is mediated via AT(1) receptor. The attenuation of the effects of AT(1) receptor blockade by PD123319 suggests that AT(2) receptor contributes to the downregulation of renal 12(S)-HETE production.
Collapse
Affiliation(s)
- Emaad M Abdel-Rahman
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia 22908-1409, USA
| | | | | |
Collapse
|
34
|
Arachidonic Acid metabolites in the cardiovascular system: the role of lipoxygenase isoforms in atherogenesis with particular emphasis on vascular remodeling. J Cardiovasc Pharmacol 2008; 50:609-20. [PMID: 18091576 DOI: 10.1097/fjc.0b013e318159f177] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Vascular remodeling refers to lasting structural alterations in the vessel wall that are initiated in response to external and internal stimuli. These changes are distinct from acute functional responses of blood vessels when challenged by increased blood pressure, altered hemodynamics, or vasoactive mediators. In early atherogenesis, when lesion formation is starting to impact local hemodynamics, the vessel wall responds with outward vascular remodeling to maintain normal blood flow. However, inward remodeling may also occur during the time course of plaque formation, contributing to vascular stenosis. Lipoxygenases form a heterogeneous family of lipid-peroxidizing enzymes, which have been implicated in atherogenesis. Several lines of in vitro and in vivo evidence indicated their involvement in disease development, but the precise function of different lipoxygenase isoforms is still a matter of discussion. Vascular remodeling is an early response during plaque development; therefore, lipoxygenases may be involved in this process. Unfortunately, little is known about the potential role of lipoxygenase isoforms in vascular remodeling. This review will briefly summarize our knowledge of the role of lipoxygenases in vascular biology and will critically review the activities of the 3 most athero-relevant lipoxygenase isoforms in atherogenesis, with particular emphasis on vascular remodeling.
Collapse
|
35
|
|
36
|
Abstract
Small lipids such as eicosanoids exert diverse and complex functions. In addition to their role in regulating normal kidney function, these lipids also play important roles in the pathogenesis of kidney diseases. Cyclooxygenase (COX)-derived prostanoids play important role in maintaining renal function, body fluid homeostasis, and blood pressure. Renal cortical COX2-derived prostanoids, particularly (PGI2) and PGE2 play critical roles in maintaining blood pressure and renal function in volume contracted states. Renal medullary COX2-derived prostanoids appear to have antihypertensive effect in individuals challenged with a high salt diet. 5-Lipoxygenase (LO)-derived leukotrienes are involved in inflammatory glomerular injury. LO product 12-hydroxyeicosatetraenoic acid (12-HETE) is associated with pathogenesis of hypertension, and may mediate angiotensin II and TGFbeta induced mesengial cell abnormality in diabetic nephropathy. P450 hydroxylase-derived 20-HETE is a potent vasoconstrictor and is involved in the pathogenesis of hypertension. P450 epoxygenase derived epoxyeicosatrienoic acids (EETs) have vasodilator and natriuretic effect. Blockade of EET formation is associated with salt-sensitive hypertension. Ceramide has also been demonstrated to be an important signaling molecule, which is involved in pathogenesis of acute kidney injury caused by ischemia/reperfusion, and toxic insults. Those pathways should provide fruitful targets for intervention in the pharmacologic treatment of renal disease.
Collapse
Affiliation(s)
- C-M Hao
- Division of Nephrology, Department of Medicine, Vanderbilt University, Veterans Administration Medical Center, Nashville, Tennessee 37232, USA.
| | | |
Collapse
|
37
|
Abstract
Small lipids such as eicosanoids exert diverse and complex functions. In addition to their role in regulating normal kidney function, these lipids also play important roles in the pathogenesis of kidney diseases. Increased glomerular cyclooxygenase (COX)1 or COX2 expression has been reported in patients with nephritis and in animal models of nephritis. COX inhibitors have shown beneficial effects on lupus nephritis and passive Heymann nephritis, but not anti-Thy1.1-induced nephritis. 5-Lipoxygenase-derived leukotrienes are involved in inflammatory glomerular injury. Lipoxygenase product 12-hydroxyeicosatetraenoic acid may mediate angiotensin II and transforming growth factor beta-induced mesangial cell abnormality in diabetic nephropathy. P450 arachidonic acid mono-oxygenase-derived 20-hydroxyeicosatetraenoic acid and epoxyeicosatrienoic acids are involved in several forms of kidney injury, including renal injury in metabolic syndrome. Ceramide also has been shown to be an important signaling molecule that is involved in the pathogenesis of acute kidney injury caused by ischemia/reperfusion and toxic insults. Those pathways should provide fruitful targets for intervention in the pharmacologic treatment of renal disease.
Collapse
Affiliation(s)
- Chuan-Ming Hao
- Division of Nephrology, Department of Medicine, Vanderbilt University, and Veterans Affairs Medical Center, Nashville, TN 37232, USA.
| | | |
Collapse
|
38
|
Abstract
Arachidonic acid metabolites are vital for the proper control of renal haemodynamics and, when not properly controlled, can contribute to renal vascular injury and end-stage renal disease. Three major enzymatic pathways, COX (cyclo-oxygenase), CYP450 (cytochrome P450) and LOX (lipoxygenase), are responsible for the metabolism of arachidonic acid metabolites to bioactive eicosanoids. These eicosanoids can dilate or constrict the renal vasculature and maintain vascular resistance in the face of changing vasoactive hormones. Renal vascular generation of eicosanoids is altered in pathophysiological conditions such as hypertension, diabetes, metabolic syndrome and acute renal failure. Experimental evidence supports the concept that altered eicosanoid metabolism contributes to renal haemodynamic alterations and the development and progression of nephropathy. The possible beneficial renal vascular actions of enzymatic inhibitors, eicosanoid analogues and receptor antagonists have been examined in hypertension, diabetes and metabolic syndrome. This review highlights the roles of renal vascular eicosanoids in the pathogenesis of nephropathy and therapeutic targets for renal disease related to hypertension, diabetes, metabolic syndrome and acute renal failure.
Collapse
Affiliation(s)
- John D Imig
- Vascular Biology Center, Department of Physiology, Medical College of Georgia, Augusta, GA 30912, USA.
| |
Collapse
|
39
|
Palmer BF. Disturbances in Renal Autoregulation and the Susceptibility to Hypertension-Induced Chronic Kidney Disease. Am J Med Sci 2004; 328:330-43. [PMID: 15599329 DOI: 10.1016/s0002-9629(15)33943-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The risk of developing chronic kidney disease in the setting of hypertension varies among patient populations. Black hypertensive patients have an increased risk of developing hypertension-induced chronic kidney disease even after taking into account socioeconomic factors. There is evidence to suggest that the kidney is intrinsically more susceptible to the damaging effects of hypertension in black patients. This susceptibility can be traced to disturbances in the way the kidney autoregulates. Impaired renal autoregulation may be the renal manifestation of a more widespread abnormality in endothelial function. Other conditions that can impair renal autoregulation and add to the risk of chronic kidney disease include low birth weight, obesity, insulin resistance, hyperuricemia, and hypercholesterolemia. To minimize the risk of chronic kidney disease in patients with impaired renal autoregulatory capability, strict blood pressure control is required. There is indirect evidence that blocking the renin-angiotensin system may improve renal autoregulation.
Collapse
Affiliation(s)
- Biff F Palmer
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-8856, USA.
| |
Collapse
|