1
|
Ibrahim DM, Fomina A, Bouten CVC, Smits AIPM. Functional regeneration at the blood-biomaterial interface. Adv Drug Deliv Rev 2023; 201:115085. [PMID: 37690484 DOI: 10.1016/j.addr.2023.115085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 06/01/2023] [Accepted: 09/07/2023] [Indexed: 09/12/2023]
Abstract
The use of cardiovascular implants is commonplace in clinical practice. However, reproducing the key bioactive and adaptive properties of native cardiovascular tissues with an artificial replacement is highly challenging. Exciting new treatment strategies are under development to regenerate (parts of) cardiovascular tissues directly in situ using immunomodulatory biomaterials. Direct exposure to the bloodstream and hemodynamic loads is a particular challenge, given the risk of thrombosis and adverse remodeling that it brings. However, the blood is also a source of (immune) cells and proteins that dominantly contribute to functional tissue regeneration. This review explores the potential of the blood as a source for the complete or partial in situ regeneration of cardiovascular tissues, with a particular focus on the endothelium, being the natural blood-tissue barrier. We pinpoint the current scientific challenges to enable rational engineering and testing of blood-contacting implants to leverage the regenerative potential of the blood.
Collapse
Affiliation(s)
- Dina M Ibrahim
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands.
| | - Aleksandra Fomina
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Graduate School of Life Sciences, Utrecht University, Utrecht, the Netherlands.
| | - Carlijn V C Bouten
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands.
| | - Anthal I P M Smits
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands.
| |
Collapse
|
2
|
Wang S, Tang C, Liu Y, Border JJ, Roman RJ, Fan F. Impact of impaired cerebral blood flow autoregulation on cognitive impairment. FRONTIERS IN AGING 2022; 3:1077302. [PMID: 36531742 PMCID: PMC9755178 DOI: 10.3389/fragi.2022.1077302] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 11/23/2022] [Indexed: 11/16/2023]
Abstract
Although the causes of cognitive impairment are multifactorial, emerging evidence indicates that cerebrovascular dysfunction plays an essential role in dementia. One of the most critical aspects of cerebrovascular dysfunction is autoregulation of cerebral blood flow (CBF), mainly mediated by the myogenic response, which is often impaired in dementia individuals with comorbidities, such as diabetes and hypertension. However, many unsolved questions remain. How do cerebrovascular networks coordinately modulate CBF autoregulation in health and disease? Does poor CBF autoregulation have an impact on cognitive impairment, and what are the underlying mechanisms? This review summarizes the cerebral vascular structure and myogenic (a three-phase model), metabolic (O2, CO2, adenosine, and H+), and endothelial (shear stress) factors in the regulation of CBF; and the consequences of CBF dysautoregulation. Other factors contributing to cerebrovascular dysfunction, such as impaired functional hyperemia and capillary abnormalities, are included as well. Moreover, this review highlights recent studies from our lab in terms of novel mechanisms involved in CBF autoregulation and addresses a hypothesis that there is a three-line of defense for CBF autoregulation in the cerebral vasculature.
Collapse
Affiliation(s)
- Shaoxun Wang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Chengyun Tang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Yedan Liu
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Jane J Border
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Richard J Roman
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Fan Fan
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| |
Collapse
|
3
|
Ishii T, Warabi E, Mann GE. Mechanisms underlying unidirectional laminar shear stress-mediated Nrf2 activation in endothelial cells: Amplification of low shear stress signaling by primary cilia. Redox Biol 2021; 46:102103. [PMID: 34425388 PMCID: PMC8379703 DOI: 10.1016/j.redox.2021.102103] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/07/2021] [Accepted: 08/12/2021] [Indexed: 12/14/2022] Open
Abstract
Endothelial cells are sensitive to mechanical stress and respond differently to oscillatory flow versus unidirectional flow. This review highlights the mechanisms by which a wide range of unidirectional laminar shear stress induces activation of the redox sensitive antioxidant transcription factor nuclear factor-E2-related factor 2 (Nrf2) in cultured endothelial cells. We propose that fibroblast growth factor-2 (FGF-2), brain-derived neurotrophic factor (BDNF) and 15-Deoxy-Δ12,14-prostaglandin J2 (15d-PGJ2) are potential Nrf2 activators induced by laminar shear stress. Shear stress-dependent secretion of FGF-2 and its receptor-mediated signaling is tightly controlled, requiring neutrophil elastase released by shear stress, αvβ3 integrin and the cell surface glycocalyx. We speculate that primary cilia respond to low laminar shear stress (<10 dyn/cm2), resulting in secretion of insulin-like growth factor 1 (IGF-1), which facilitates αvβ3 integrin-dependent FGF-2 secretion. Shear stress induces generation of heparan-binding epidermal growth factor-like growth factor (HB-EGF), which contributes to FGF-2 secretion and gene expression. Furthermore, HB-EGF signaling modulates FGF-2-mediated NADPH oxidase 1 activation that favors casein kinase 2 (CK2)-mediated phosphorylation/activation of Nrf2 associated with caveolin 1 in caveolae. Higher shear stress (>15 dyn/cm2) induces vesicular exocytosis of BDNF from endothelial cells, and we propose that BDNF via the p75NTR receptor could induce CK2-mediated Nrf2 activation. Unidirectional laminar shear stress upregulates gene expression of FGF-2 and BDNF and generation of 15d-PGJ2, which cooperate in sustaining Nrf2 activation to protect endothelial cells against oxidative damage.
Collapse
Affiliation(s)
- Tetsuro Ishii
- School of Medicine, University of Tsukuba, Tsukuba, Ibaraki, 305-8577, Japan.
| | - Eiji Warabi
- School of Medicine, University of Tsukuba, Tsukuba, Ibaraki, 305-8577, Japan.
| | - Giovanni E Mann
- King's British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, King's College London, 150 Stamford Street, London, SE1 9NH, UK.
| |
Collapse
|
4
|
Estronca L, Francisco V, Pitrez P, Honório I, Carvalho L, Vazão H, Blersch J, Rai A, Nissan X, Simon U, Grãos M, Saúde L, Ferreira L. Induced pluripotent stem cell-derived vascular networks to screen nano-bio interactions. NANOSCALE HORIZONS 2021; 6:245-259. [PMID: 33576750 DOI: 10.1039/d0nh00550a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The vascular bioactivity/safety of nanomaterials is typically evaluated by animal testing, which is of low throughput and does not account for biological differences between animals and humans such as ageing, metabolism and disease profiles. The development of personalized human in vitro platforms to evaluate the interaction of nanomaterials with the vascular system would be important for both therapeutic and regenerative medicine. A library of 30 nanoparticle (NP) formulations, in use in imaging, antimicrobial and pharmaceutical applications, was evaluated in a reporter zebrafish model of vasculogenesis and then tested in personalized humanized models composed of human-induced pluripotent stem cell (hiPSC)-derived endothelial cells (ECs) with "young" and "aged" phenotypes in 3 vascular network formats: 2D (in polystyrene dish), 3D (in Matrigel) and in a blood vessel on a chip. As a proof of concept, vascular toxicity was used as the main readout. The results show that the toxicity profile of NPs to hiPSC-ECs was dependent on the "age" of the endothelial cells and vascular network format. hiPSC-ECs were less susceptible to the cytotoxicity effect of NPs when cultured in flow than in static conditions, the protective effect being mediated, at least in part, by glycocalyx. Overall, the results presented here highlight the relevance of in vitro hiPSC-derived vascular systems to screen vascular nanomaterial interactions.
Collapse
Affiliation(s)
- Luís Estronca
- Faculty of Medicine, University of Coimbra, 3000-548, Coimbra, Portugal.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Triantafyllidi H, Benas D. Endothelium: The Trivial Fence Became a Powerful Gatekeeper of Vascular Health. Angiology 2021; 72:601-603. [PMID: 33535810 DOI: 10.1177/0003319721992238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Helen Triantafyllidi
- 2nd Department of Cardiology, Medical School, 68989National and Kapodistrian University, Attikon University Hospital, Athens, Greece
| | - Dimitris Benas
- 2nd Department of Cardiology, Medical School, 68989National and Kapodistrian University, Attikon University Hospital, Athens, Greece
| |
Collapse
|
6
|
Hirata T, Yamamoto K, Ikeda K, Arita M. Functional lipidomics of vascular endothelial cells in response to laminar shear stress. FASEB J 2021; 35:e21301. [PMID: 33421194 DOI: 10.1096/fj.202002144r] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 12/03/2020] [Accepted: 12/08/2020] [Indexed: 11/11/2022]
Abstract
Laminar shear stress generated by blood flow stimulates endothelial cells and activates signal transduction, which plays an important role in vascular homeostasis. Several lines of evidence indicate that membrane and intracellular lipids are involved in the signal transduction of biomechanical stresses. In this study, we performed global profiling of cellular lipids from human pulmonary artery endothelial cells (HPAEC) exposed to laminar shear stress. A total of 761 species of lipids were successfully annotated, with 198 of these species significantly changed in response to shear stress for 24 hours. Ether-linked lipids containing an alkyl moiety with a medium chain length (C11-C14) were uniquely upregulated, and the administration of their biosynthetic precursor 1-O-dodecyl-rac-glycerol attenuated phorbol 12-myristate 13-acetate (PMA) induced vascular cell adhesion molecule-1 (VCAM-1) expression. Given the pro-inflammatory and atherogenic roles of VCAM-1, our findings suggest that the induction of a specific group of lipids (ie, ether-linked lipids with medium length alkyl side chain) may confer atheroprotective and anti-inflammatory roles to vascular endothelial cells under flow conditions.
Collapse
Affiliation(s)
- Tsuyoshi Hirata
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.,Cellular and Molecular Epigenetics Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Kimiko Yamamoto
- Laboratory of System Physiology, Department of Biomedical Engineering, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Kazutaka Ikeda
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.,Laboratory of Biomolecule Analysis, Kazusa DNA Research Institute, Japan
| | - Makoto Arita
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.,Cellular and Molecular Epigenetics Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan.,Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, Tokyo, Japan
| |
Collapse
|
7
|
Mitchell JA, Kirkby NS, Ahmetaj-Shala B, Armstrong PC, Crescente M, Ferreira P, Lopes Pires ME, Vaja R, Warner TD. Cyclooxygenases and the cardiovascular system. Pharmacol Ther 2021; 217:107624. [DOI: 10.1016/j.pharmthera.2020.107624] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 06/30/2020] [Indexed: 02/07/2023]
|
8
|
Ovalı F. Molecular and Mechanical Mechanisms Regulating Ductus Arteriosus Closure in Preterm Infants. Front Pediatr 2020; 8:516. [PMID: 32984222 PMCID: PMC7477801 DOI: 10.3389/fped.2020.00516] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 07/21/2020] [Indexed: 12/14/2022] Open
Abstract
Failure of ductus arteriosus closure after preterm birth is associated with significant morbidities. Ductal closure requires and is regulated by a complex interplay of molecular and mechanical mechanisms with underlying genetic factors. In utero patency of the ductus is maintained by low oxygen tension, high levels of prostaglandins, nitric oxide and carbon monoxide. After birth, ductal closure occurs first by functional closure, followed by anatomical remodeling. High oxygen tension and decreased prostaglandin levels mediated by numerous factors including potassium channels, endothelin-1, isoprostanes lead to the contraction of the ductus. Bradykinin and corticosteroids also induce ductal constriction by attenuating the sensitivity of the ductus to PGE2. Smooth muscle cells of the ductus can sense oxygen through a mitochondrial network by the role of Rho-kinase pathway which ends up with increased intracellular calcium levels and contraction of myosin light chains. Anatomical closure of the ductus is also complex with various mechanisms such as migration and proliferation of smooth muscle cells, extracellular matrix production, endothelial cell proliferation which mediate cushion formation with the interaction of blood cells. Regulation of vessel walls is affected by retinoic acid, TGF-β1, notch signaling, hyaluronan, fibronectin, chondroitin sulfate, elastin, and vascular endothelial cell growth factor (VEGF). Formation of the platelet plug facilitates luminal remodeling by the obstruction of the constricted ductal lumen. Vasa vasorum are more pronounced in the term ductus but are less active in the preterm ductus. More than 100 genes are effective in the prostaglandin pathway or in vascular smooth muscle development and structure may affect the patency of ductus. Hemodynamic changes after birth including fluid load and flow characteristics as well as shear forces within the ductus also stimulate closure. Current pharmacological treatment for the closure of a patent ductus is based on the blockage of the prostaglandin pathway mainly through COX or POX inhibition, albeit with some limitations and side effects. Further research for new agents aiming ductal closure should focus on a clear understanding of vascular biology of the ductus.
Collapse
Affiliation(s)
- Fahri Ovalı
- Division of Neonatology, Department of Pediatrics, Istanbul Medeniyet University, Istanbul, Turkey
| |
Collapse
|
9
|
Matsumoto T, Takayanagi K, Kojima M, Katome T, Taguchi K, Kobayashi T. Direct Impairment of the Endothelial Function by Acute Indoxyl Sulfate through Declined Nitric Oxide and Not Endothelium-Derived Hyperpolarizing Factor or Vasodilator Prostaglandins in the Rat Superior Mesenteric Artery. Biol Pharm Bull 2019; 42:1236-1242. [PMID: 31257300 DOI: 10.1248/bpb.b19-00177] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Upon stimulation, endothelial cells release various factors to regulate the vascular tone. In particular, vasorelaxing factors, called endothelium-derived relaxing factors (EDRFs), are altered in the production and/or release, as well as their signaling every vessel and under pathophysiological states, including cardiovascular, kidney, and metabolic diseases. Although indoxyl sulfate is known as a protein-bound uremic toxin and circulating levels are elevated in the impaired kidney functions, direct impact on the vascular function, especially EDRF's signaling, remains unclear. In this study, we hypothesize that acute exposure to indoxyl sulfate could alter vascular relaxation in the rat superior mesenteric artery. Accordingly, we measured acetylcholine (ACh)-induced endothelium-dependent relaxation in the absence and presence of several inhibitors to divide into each EDRF, including nitric oxide (NO), vasodilator prostaglandins (PGs), and endothelium-derived hyperpolarizing factor (EDHF). Indoxyl sulfate reduced the sensitivity to ACh but not sodium nitroprusside. Under cyclooxygenase (COX) inhibition or inhibitions of COX plus source of EDHF, such as small (SKCa)- and intermediate (IKCa)-conductance calcium-activated K+ channels, the decreased sensitivity to ACh in indoxyl sulfate exposed vessel was still preserved. However, under inhibition of NO synthase (NOS) or inhibitions of NOS and COX, the difference of sensitivity to ACh between vehicle and indoxyl sulfate was eliminated. These findings indicated that acute exposure of indoxyl sulfate in the rat superior mesenteric artery specifically explicitly impaired NO signaling but not EDHF or vasodilator PGs.
Collapse
Affiliation(s)
- Takayuki Matsumoto
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University
| | - Keisuke Takayanagi
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University
| | - Mihoka Kojima
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University
| | - Tomoki Katome
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University
| | - Kumiko Taguchi
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University
| | - Tsuneo Kobayashi
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University
| |
Collapse
|
10
|
Raees A, Bakhamis A, Mohamed-Ali V, Bashah M, Al-Jaber M, Abraham D, Clapp LH, Orie NN. Altered cyclooxygenase-1 and enhanced thromboxane receptor activities underlie attenuated endothelial dilatory capacity of omental arteries in obesity. Life Sci 2019; 239:117039. [PMID: 31704447 DOI: 10.1016/j.lfs.2019.117039] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 10/29/2019] [Accepted: 10/31/2019] [Indexed: 01/05/2023]
Abstract
AIMS Obesity is a risk factor for endothelial dysfunction, the severity of which is likely to vary depending on extent and impact of adiposity on the vasculature. This study investigates the roles of cyclooxygenase isoforms and thromboxane receptor activities in the differential endothelial dilatory capacities of arteries derived from omental and subcutaneous adipose tissues in obesity. MAIN METHODS Small arteries were isolated from omental and subcutaneous adipose tissues obtained from consented morbidly obese patients (n = 65, BMI 45 ± 6 kg m-2 [Mean ± SD]) undergoing bariatric surgery. Relaxation to acetylcholine was studied by wire myography in the absence or presence of indomethacin (10 μM, cyclooxygenase inhibitor), FR122047 (1 μM, cyclooxygenase-1 inhibitor), Celecoxib (4 μM, cyclooxygenase-2 inhibitor), Nω-Nitro-L-arginine methyl ester (L-NAME, 100 μM, nitric oxide synthase inhibitor) or combination of apamin (0.5 μM) and charybdotoxin (0.1 μM) that together inhibit endothelium-derived hyperpolarizing factor (EDHF). Contractions to U46619 (thromboxane A2 mimetic) were also studied. KEY FINDINGS Acetylcholine relaxation was significantly attenuated in omental compared with subcutaneous arteries from same patients (p < 0.01). Indomethacin (p < 0.01) and FR122047 (p < 0.001) but not Celecoxib significantly improved the omental arteriolar relaxation. Cyclooxygenase-1 mRNA and U46619 contractions were both increased in omental compared with subcutaneous arteries (p < 0.05). L-NAME comparably inhibited acetylcholine relaxation in both arteries, while apamin+charybdotoxin were less effective in omental compared with subcutaneous arteries. SIGNIFICANCE The results show that the depot-specific reduction in endothelial dilatory capacity of omental compared with subcutaneous arteries in obesity is in large part due to altered cyclooxygenase-1 and enhanced thromboxane receptor activities, which cause EDHF deficiency.
Collapse
Affiliation(s)
- Asmaa Raees
- Qatar Analytics and BioResearch Lab, Anti-Doping Lab Qatar, Qatar
| | - Aysha Bakhamis
- Qatar Analytics and BioResearch Lab, Anti-Doping Lab Qatar, Qatar
| | | | - Moataz Bashah
- Metabolic and Bariatric Surgery Department, Hamad Medical Corporation, Doha, Qatar
| | - Mashael Al-Jaber
- Qatar Analytics and BioResearch Lab, Anti-Doping Lab Qatar, Qatar
| | - David Abraham
- Centre for Rheumatology and Connective Tissue Diseases, Division of Medicine, University College London, UK
| | - Lucie H Clapp
- Institute of Cardiovascular Sciences, University College London, UK
| | - Nelson N Orie
- Qatar Analytics and BioResearch Lab, Anti-Doping Lab Qatar, Qatar.
| |
Collapse
|
11
|
Nyunt T, Britton M, Wanichthanarak K, Budamagunta M, Voss JC, Wilson DW, Rutledge JC, Aung HH. Mitochondrial oxidative stress-induced transcript variants of ATF3 mediate lipotoxic brain microvascular injury. Free Radic Biol Med 2019; 143:25-46. [PMID: 31356870 PMCID: PMC6848793 DOI: 10.1016/j.freeradbiomed.2019.07.024] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 06/30/2019] [Accepted: 07/23/2019] [Indexed: 12/06/2022]
Abstract
Elevation of blood triglycerides, primarily triglyceride-rich lipoproteins (TGRL), is an independent risk factor for cardiovascular disease and vascular dementia (VaD). Accumulating evidence indicates that both atherosclerosis and VaD are linked to vascular inflammation. However, the role of TGRL in vascular inflammation, which increases risk for VaD, remains largely unknown and its underlying mechanisms are still unclear. We strived to determine the effects of postprandial TGRL exposure on brain microvascular endothelial cells, the potential risk factor of vascular inflammation, resulting in VaD. We showed in Aung et al., J Lipid Res., 2016 that postprandial TGRL lipolysis products (TL) activate mitochondrial reactive oxygen species (ROS) and increase the expression of the stress-responsive protein, activating transcription factor 3 (ATF3), which injures human brain microvascular endothelial cells (HBMECs) in vitro. In this study, we deployed high-throughput sequencing (HTS)-based RNA sequencing methods and mito stress and glycolytic rate assays with an Agilent Seahorse XF analyzer and profiled the differential expression of transcripts, constructed signaling pathways, and measured mitochondrial respiration, ATP production, proton leak, and glycolysis of HBMECs treated with TL. Conclusions: TL potentiate ROS by mitochondria which activate mitochondrial oxidative stress, decrease ATP production, increase mitochondrial proton leak and glycolysis rate, and mitochondria DNA damage. Additionally, CPT1A1 siRNA knockdown suppresses oxidative stress and prevents mitochondrial dysfunction and vascular inflammation in TL treated HBMECs. TL activates ATF3-MAPKinase, TNF, and NRF2 signaling pathways. Furthermore, the NRF2 signaling pathway which is upstream of the ATF3-MAPKinase signaling pathway, is also regulated by the mitochondrial oxidative stress. We are the first to report differential inflammatory characteristics of transcript variants 4 (ATF3-T4) and 5 (ATF3-T5) of the stress responsive gene ATF3 in HBMECs induced by postprandial TL. Specifically, our data indicates that ATF3-T4 predominantly regulates the TL-induced brain microvascular inflammation and TNF signaling. Both siRNAs of ATF3-T4 and ATF3-T5 suppress cells apoptosis and lipotoxic brain microvascular endothelial cells. These novel signaling pathways triggered by oxidative stress-responsive transcript variants, ATF3-T4 and ATF3-T5, in the brain microvascular inflammation induced by TGRL lipolysis products may contribute to pathophysiological processes of vascular dementia.
Collapse
Affiliation(s)
- Tun Nyunt
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, Davis, CA, 95616, USA
| | - Monica Britton
- Genome Center and Bioinformatics Core Facility, University of California, Davis, CA, 95616, USA
| | - Kwanjeera Wanichthanarak
- West Coast Metabolomics Center, Genome Center, University of California Davis, Davis, CA, USA; Department of Biochemistry and Siriraj Metabolomics and Phenomics Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Madhu Budamagunta
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Davis, CA, 95616, USA
| | - John C Voss
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Davis, CA, 95616, USA
| | - Dennis W Wilson
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA, 95616, USA
| | - John C Rutledge
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, Davis, CA, 95616, USA
| | - Hnin H Aung
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, Davis, CA, 95616, USA.
| |
Collapse
|
12
|
Riccardello GJ, Shastri DN, Changa AR, Thomas KG, Roman M, Prestigiacomo CJ, Gandhi CD. Influence of Relative Residence Time on Side-Wall Aneurysm Inception. Neurosurgery 2019; 83:574-581. [PMID: 28945849 DOI: 10.1093/neuros/nyx433] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 07/21/2017] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Relative residence time (RRT) is a marker of disturbed blood flow, marked by low magnitude and high oscillatory wall shear stress (WSS). The relation between solute residence time in proximity to the vascular endothelium and the atherosclerotic process is well appreciated in the literature. OBJECTIVE To assess the influence of RRT on side-wall aneurysm inception to better understand the role of atherosclerosis in aneurysm formation. METHODS Fourteen side-wall internal carotid artery aneurysms from the Aneurisk repository which met criteria for parent vessel reconstruction were reconstructed with Vascular Modeling Toolkit. Computational fluid dynamics analysis was carried out in Fluent. RRT was calculated in MATLAB (The MathWorks Inc, Natick, Massachusetts). We analyzed the results for correlations, defined as presence or absence of local elevations in RRT in specific regions of vasculature. RESULTS RRT was concluded to be negatively correlated with aneurysm inception in this study of side-wall internal carotid artery aneurysms, with 12/14 cases yielding the absence of local RRT elevations within or in close proximity of the removed ostium. Subsequent analysis of WSS showed that 11 of 14 aneurysms were formed in an atheroprotective environment, with only 1 of 14 formed in an atherogenic environment. Two models were found to be of indeterminate environment. CONCLUSION Atherogenesis and atherosclerosis have long been thought to be a major inciting factor responsible for the formation of aneurysms in the cerebral vasculature. We propose that inception of side-wall aneurysms occurs in hemodynamic environments that promote an atheroprotective endothelial phenotype and that the atheroprotective phenotype is therefore aneurysmogenic.
Collapse
Affiliation(s)
- Gerald J Riccardello
- Department of Neurological Surgery, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Darshan N Shastri
- Department of Neurological Surgery, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Abhinav R Changa
- Department of Neurological Surgery, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Kiran G Thomas
- Department of Neurological Surgery, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Max Roman
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey
| | | | - Chirag D Gandhi
- Department of Neurological Surgery, Rutgers New Jersey Medical School, Newark, New Jersey.,Department of Radiology, Rutgers New Jersey Medical School, Newark, New Jersey
| |
Collapse
|
13
|
Abstract
Regulation of coronary blood flow is maintained through a delicate balance of ventriculoarterial and neurohumoral mechanisms. The aortic valve is integral to the functions of these systems, and disease states that compromise aortic valve integrity have the potential to seriously disrupt coronary blood flow. Aortic stenosis (AS) is the most common cause of valvular heart disease requiring medical intervention, and the prevalence and associated socio-economic burden of AS are set to increase with population ageing. Valvular stenosis precipitates a cascade of structural, microcirculatory, and neurohumoral changes, which all lead to impairment of coronary flow reserve and myocardial ischaemia even in the absence of notable coronary stenosis. Coronary physiology can potentially be normalized through interventions that relieve severe AS, but normality is often not immediately achievable and probably requires continued adaptation. Finally, the physiological assessment of coronary artery disease in patients with AS represents an ongoing challenge, as the invasive physiological measures used in current cardiology practice are yet to be validated in this population. This Review discusses the key concepts of coronary pathophysiology in patients with AS through presentation of contemporary basic science and data from animal and human studies.
Collapse
|
14
|
Abstract
Passive exercise/movement has a long history in both medicine and physiology. Early clinical applications of passive exercise/movement utilized pneumatic and direct limb compression to stimulate the vasculature and evoke changes in blood flow to avoid complications brought about by stasis and vascular disease. Over the last 50 years, passive exercise/movement has continued to progress and has provided physiologists with a reductionist approach to mechanistically examine the cardiorespiratory, hyperemic, and afferent responses to movement without the confounding influence of metabolism that accompanies active exercise. This review, in addition to providing an historical perspective, focuses on the recent advancements utilizing passive leg movement, and how the hyperemic response at the onset of this passive movement has evolved from a method to evaluate the central and peripheral regulation of blood flow during exercise to an innovative and promising tool to assess vascular function. As an assessment of vascular function, passive leg movement is relatively simple to perform and provides a nitric oxide-dependent evaluation of endothelial function across the lifespan that is sensitive to changes in activity/fitness and disease state (heart failure, peripheral artery disease, sepsis). The continual refinement and characterization of passive leg movement are aimed at improving our understanding of blood flow regulation and the development of a clinically ready approach to predict and monitor the progression of cardiovascular disease.
Collapse
Affiliation(s)
- Joel D Trinity
- George E Wahlen Veterans Affairs Medical Center, Geriatric Research, Education and Clinical Center, Bldg 2, RM 1D29A, 500 Foothill Dr., Salt Lake City, UT, 84148, USA.
- Division of Geriatrics, Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA.
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA.
| | - Russell S Richardson
- George E Wahlen Veterans Affairs Medical Center, Geriatric Research, Education and Clinical Center, Bldg 2, RM 1D29A, 500 Foothill Dr., Salt Lake City, UT, 84148, USA
- Division of Geriatrics, Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
15
|
Rendic SP, Peter Guengerich F. Human cytochrome P450 enzymes 5-51 as targets of drugs and natural and environmental compounds: mechanisms, induction, and inhibition - toxic effects and benefits. Drug Metab Rev 2019; 50:256-342. [PMID: 30717606 DOI: 10.1080/03602532.2018.1483401] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cytochrome P450 (P450, CYP) enzymes have long been of interest due to their roles in the metabolism of drugs, pesticides, pro-carcinogens, and other xenobiotic chemicals. They have also been of interest due to their very critical roles in the biosynthesis and metabolism of steroids, vitamins, and certain eicosanoids. This review covers the 22 (of the total of 57) human P450s in Families 5-51 and their substrate selectivity. Furthermore, included is information and references regarding inducibility, inhibition, and (in some cases) stimulation by chemicals. We update and discuss important aspects of each of these 22 P450s and questions that remain open.
Collapse
Affiliation(s)
| | - F Peter Guengerich
- b Department of Biochemistry , Vanderbilt University School of Medicine , Nashville , TN , USA
| |
Collapse
|
16
|
Crockett SL, Berger CD, Shelton EL, Reese J. Molecular and mechanical factors contributing to ductus arteriosus patency and closure. CONGENIT HEART DIS 2018; 14:15-20. [PMID: 30468303 DOI: 10.1111/chd.12714] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 10/16/2018] [Indexed: 12/30/2022]
Abstract
Regulation of the ductus arteriosus, an essential fetal vessel connecting the pulmonary artery and aorta, is complex. Failure of this vessel to close after birth may result in a persistent left-to-right shunt through the patent ductus arteriosus, a condition associated with significant morbidities. Numerous factors contribute to the shift from fetal ductus patency to postnatal closure, requiring precise coordination of molecular cues with biomechanical forces and underlying genetic influences. Despite significant advances, questions remain regarding signaling dynamics and the natural time course of ductus closure, particularly in preterm neonates. This review highlights the contributions of early investigators and more recent clinician scientists to our understanding of the molecular and mechanical factors that mediate ductus patency and closure.
Collapse
Affiliation(s)
- Stacey L Crockett
- Mildred T. Stahlman Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN
| | - Courtney D Berger
- Mildred T. Stahlman Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN
| | - Elaine L Shelton
- Mildred T. Stahlman Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN
| | - Jeff Reese
- Mildred T. Stahlman Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
17
|
Activation of TP receptors induces high release of PGI2 in coronary arteries of renal hypertensive rats. J Mol Cell Cardiol 2018; 122:125-133. [DOI: 10.1016/j.yjmcc.2018.08.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 08/02/2018] [Accepted: 08/08/2018] [Indexed: 12/19/2022]
|
18
|
Rieg AD, Suleiman S, Anker C, Verjans E, Rossaint R, Uhlig S, Martin C. PDGF-BB regulates the pulmonary vascular tone: impact of prostaglandins, calcium, MAPK- and PI3K/AKT/mTOR signalling and actin polymerisation in pulmonary veins of guinea pigs. Respir Res 2018; 19:120. [PMID: 29921306 PMCID: PMC6009037 DOI: 10.1186/s12931-018-0829-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 06/13/2018] [Indexed: 12/15/2022] Open
Abstract
Background Platelet-derived growth factor (PDGF)-BB and its receptor PDGFR are highly expressed in pulmonary hypertension (PH) and mediate proliferation. Recently, we showed that PDGF-BB contracts pulmonary veins (PVs) and that this contraction is prevented by inhibition of PDGFR-β (imatinib/SU6668). Here, we studied PDGF-BB-induced contraction and downstream-signalling in isolated perfused lungs (IPL) and precision-cut lung slices (PCLS) of guinea pigs (GPs). Methods In IPLs, PDGF-BB was perfused after or without pre-treatment with imatinib (perfused/nebulised), the effects on the pulmonary arterial pressure (PPA), the left atrial pressure (PLA) and the capillary pressure (Pcap) were studied and the precapillary (Rpre) and postcapillary resistance (Rpost) were calculated. Perfusate samples were analysed (ELISA) to detect the PDGF-BB-induced release of prostaglandin metabolites (TXA2/PGI2). In PCLS, the contractile effect of PDGF-BB was evaluated in pulmonary arteries (PAs) and PVs. In PVs, PDGF-BB-induced contraction was studied after inhibition of PDGFR-α/β, L-Type Ca2+-channels, ROCK/PKC, prostaglandin receptors, MAP2K, p38-MAPK, PI3K-α/γ, AKT/PKB, actin polymerisation, adenyl cyclase and NO. Changes of the vascular tone were measured by videomicroscopy. In PVs, intracellular cAMP was measured by ELISA. Results In IPLs, PDGF-BB increased PPA, Pcap and Rpost. In contrast, PDGF-BB had no effect if lungs were pre-treated with imatinib (perfused/nebulised). In PCLS, PDGF-BB significantly contracted PVs/PAs which was blocked by the PDGFR-β antagonist SU6668. In PVs, inhibition of actin polymerisation and inhibition of L-Type Ca2+-channels reduced PDGF-BB-induced contraction, whereas inhibition of ROCK/PKC had no effect. Blocking of EP1/3- and TP-receptors or inhibition of MAP2K-, p38-MAPK-, PI3K-α/γ- and AKT/PKB-signalling prevented PDGF-BB-induced contraction, whereas inhibition of EP4 only slightly reduced it. Accordingly, PDGF-BB increased TXA2 in the perfusate, whereas PGI2 was increased in all groups after 120 min and inhibition of IP-receptors did not enhance PDGF-BB-induced contraction. Moreover, PDGF-BB increased cAMP in PVs and inhibition of adenyl cyclase enhanced PDGF-BB-induced contraction, whereas inhibition of NO-formation only slightly increased it. Conclusions PDGF-BB/PDGFR regulates the pulmonary vascular tone by the generation of prostaglandins, the increase of calcium, the activation of MAPK- or PI3K/AKT/mTOR signalling and actin remodelling. More insights in PDGF-BB downstream-signalling may contribute to develop new therapeutics for PH.
Collapse
Affiliation(s)
- Annette D Rieg
- Department of Anaesthesiology, Medical Faculty RWTH-Aachen, Aachen, Germany.
| | - Said Suleiman
- Institute of Pharmacology and Toxicology, Medical Faculty RWTH-Aachen, Aachen, Germany
| | - Carolin Anker
- Institute of Pharmacology and Toxicology, Medical Faculty RWTH-Aachen, Aachen, Germany
| | - Eva Verjans
- Institute of Pharmacology and Toxicology, Medical Faculty RWTH-Aachen, Aachen, Germany
| | - Rolf Rossaint
- Department of Anaesthesiology, Medical Faculty RWTH-Aachen, Aachen, Germany
| | - Stefan Uhlig
- Institute of Pharmacology and Toxicology, Medical Faculty RWTH-Aachen, Aachen, Germany
| | - Christian Martin
- Institute of Pharmacology and Toxicology, Medical Faculty RWTH-Aachen, Aachen, Germany
| |
Collapse
|
19
|
Lu T, Wang XL, Chai Q, Sun X, Sieck GC, Katusic ZS, Lee HC. Role of the endothelial caveolae microdomain in shear stress-mediated coronary vasorelaxation. J Biol Chem 2017; 292:19013-19023. [PMID: 28924052 DOI: 10.1074/jbc.m117.786152] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 09/08/2017] [Indexed: 12/31/2022] Open
Abstract
In this study, we determined the role of caveolae and the ionic mechanisms that mediate shear stress-mediated vasodilation (SSD). We found that both TRPV4 and SK channels are targeted to caveolae in freshly isolated bovine coronary endothelial cells (BCECs) and that TRPV4 and KCa2.3 (SK3) channels are co-immunoprecipitated by anti-caveolin-1 antibodies. Acute exposure of BCECs seeded in a capillary tube to 10 dynes/cm2 of shear stress (SS) resulted in activation of TRPV4 and SK currents. However, after incubation with HC067047 (TRPV4 inhibitor), SK currents could no longer be activated by SS, suggesting SK channel activation by SS was mediated through TRPV4. SK currents in BCECs were also activated by isoproterenol or by GSK1016790A (TRPV4 activator). In addition, preincubation of isolated coronary arterioles with apamin (SK inhibitor) resulted in a significant diminution of SSD whereas preincubation with HC067047 produced vasoconstriction by SS. Exposure of BCECs to SS (15 dynes/cm2 16 h) enhanced the production of nitric oxide and prostacyclin (PGI2) and facilitated the translocation of TRPV4 to the caveolae. Inhibition of TRPV4 abolished the SS-mediated intracellular Ca2+ ([Ca2+] i ) increase in BCECs. These results indicate a dynamic interaction in the vascular endothelium among caveolae TRPV4 and SK3 channels. This caveolae-TRPV4-SK3 channel complex underlies the molecular and ionic mechanisms that modulate SSD in the coronary circulation.
Collapse
Affiliation(s)
- Tong Lu
- From the Department of Cardiovascular Medicine
| | | | - Qiang Chai
- From the Department of Cardiovascular Medicine.,the Department of Physiology, Institute of Basic Medicine, Shandong Academy of Medical Science, Jinan 250062, China
| | | | - Garry C Sieck
- Department of Physiology and Biomedical Engineering, and
| | - Zvonimir S Katusic
- Department of Anesthesiology, Mayo Clinic, Rochester, Minnesota 55905 and
| | - Hon-Chi Lee
- From the Department of Cardiovascular Medicine,
| |
Collapse
|
20
|
Gori T, von Henning U, Muxel S, Schaefer S, Fasola F, Vosseler M, Schnorbus B, Binder H, Parker JD, Münzel T. Both flow-mediated dilation and constriction are associated with changes in blood flow and shear stress: Two complementary perspectives on endothelial function. Clin Hemorheol Microcirc 2017; 64:255-266. [DOI: 10.3233/ch-168102] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Tommaso Gori
- Kardiologie I, Zentrum für Kardiologie und DZHK Standort Rhein-Main, Mainz, Germany
| | - Urs von Henning
- Kardiologie I, Zentrum für Kardiologie und DZHK Standort Rhein-Main, Mainz, Germany
| | - Selina Muxel
- Kardiologie I, Zentrum für Kardiologie und DZHK Standort Rhein-Main, Mainz, Germany
| | - Sarina Schaefer
- Kardiologie I, Zentrum für Kardiologie und DZHK Standort Rhein-Main, Mainz, Germany
| | - Federica Fasola
- Kardiologie I, Zentrum für Kardiologie und DZHK Standort Rhein-Main, Mainz, Germany
| | - Marcus Vosseler
- Kardiologie I, Zentrum für Kardiologie und DZHK Standort Rhein-Main, Mainz, Germany
| | - Boris Schnorbus
- Kardiologie I, Zentrum für Kardiologie und DZHK Standort Rhein-Main, Mainz, Germany
| | - Harald Binder
- Institut für Medizinische Biometrie, Epidemiologie und Informatik, University Medical Center Mainz, Germany
| | - John D. Parker
- Department of Medicine, Division of Cardiology, Mount Sinai and University Health Network Hospitals, University of Toronto, Canada
| | - Thomas Münzel
- Kardiologie I, Zentrum für Kardiologie und DZHK Standort Rhein-Main, Mainz, Germany
| |
Collapse
|
21
|
Zhou R, Xu Q, Xu Y, Xiong A, Wang Y, Ma P. Oxymatrine attenuated isoproterenol-induced heart failure in rats via regulation of COX-2/PGI 2 pathway. Biomed Pharmacother 2016; 84:1359-1366. [PMID: 27802898 DOI: 10.1016/j.biopha.2016.10.070] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 10/14/2016] [Accepted: 10/24/2016] [Indexed: 12/22/2022] Open
Abstract
Oxymatrine (OMT) is an active constituent of traditional Chinese herb Sophora japonica Ait which has been shown to exert potent anti-inflammatory,anti-oxidant and anti-fibrosis properties. Our previous studies have demonstrated that OMT has protective effects on isoproterenol-induced heart failure in rats through regulation of DDAH/ADMA metabolism pathway.In this study,we further investigated whether OMT could attenuate isoproterenol-induced heart failure through the regulation of COX-2/PGI2 pathway. Heart failure was induced in Sprague-Dawley rats by 5mg/kg isoproterenol subcutaneous injection for 7days. The rats were maintained on normal diet and randomly divided into five groups: control, isoproterenol, isoproterenol with OMT (50, 100mg/kg), and OMT alone groups (n=12 in each group). Serum brain natruretic peptide (BNP, a heart failure biomarker), histopathological variables, expression of Cytosolic phospholipase A2 (cPLA2), cyclooxygenase-1 (COX-1), cyclooxygenase-2 (COX-2) and Prostacyclin synthase (PGIS) were analysed. Administration of OMT significantly reduced the increased BNP in plasm of isoproterenol-induced rats, attenuated cardiac fibrosis,suppressed overexpression of myocardial COX-1 expression, up-regulated COX-2 and PGIS expression, but had no effects on isoproterenol-induced elevated protein cPLA2. And compared with control group, any indexes in sham rats treated with OMT (100mg/kg) alone were unaltered. These results demonstrated that OMT has cardioprotective effects on isoproterenol-induced heart failure in rats by regulating COX-2/PGI2 pathway.
Collapse
Affiliation(s)
- Ru Zhou
- Department of Pharmacology, School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China; Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan 750004, China; Ningxia Hui Medicine Modern Engineering Research Center and Collaborative Innovation Center, Ningxia Medical University, Yinchuan 750004, China
| | - Qingbin Xu
- Department of Cardiology, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Yehua Xu
- Department of Cardiology, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Aiqin Xiong
- Department of Cardiology, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Yang Wang
- Department of Cardiology, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Ping Ma
- Department of Cardiology, General Hospital of Ningxia Medical University, Yinchuan 750004, China.
| |
Collapse
|
22
|
Di T, Sullivan JA, Rupnow HL, Magness RR, Bird IM. Pregnancy Induces Expression of cPLA2 in Ovine Uterine Artery but Not Systemic Artery Endothelium. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/107155769900600604] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
| | | | | | - Ronald R. Magness
- Department of Obstetrics and Gynecology, Perinatal Research Laboratories and Department of Animal Sciences, University of Wisconsin-Madison, Madison, Wisconsin
| | - Ian M. Bird
- Department of Obstetrics and Gynecology, Perinatal Research Laboratories and Department of Animal Sciences, University of Wisconsin-Madison, Madison, Wisconsin
| |
Collapse
|
23
|
Cahill PA, Redmond EM. Vascular endothelium - Gatekeeper of vessel health. Atherosclerosis 2016; 248:97-109. [PMID: 26994427 PMCID: PMC6478391 DOI: 10.1016/j.atherosclerosis.2016.03.007] [Citation(s) in RCA: 354] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 03/03/2016] [Accepted: 03/04/2016] [Indexed: 02/08/2023]
Abstract
The vascular endothelium is an interface between the blood stream and the vessel wall. Changes in this single cell layer of the artery wall are believed of primary importance in the pathogenesis of vascular disease/atherosclerosis. The endothelium responds to humoral, neural and especially hemodynamic stimuli and regulates platelet function, inflammatory responses, vascular smooth muscle cell growth and migration, in addition to modulating vascular tone by synthesizing and releasing vasoactive substances. Compromised endothelial function contributes to the pathogenesis of cardiovascular disease; endothelial 'dysfunction' is associated with risk factors, correlates with disease progression, and predicts cardiovascular events. Therapies for atherosclerosis have been developed, therefore, that are directed towards improving endothelial function.
Collapse
Affiliation(s)
- Paul A Cahill
- Vascular Biology and Therapeutics Laboratory, Dublin City University, Dublin, Ireland
| | - Eileen M Redmond
- Department of Surgery, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
24
|
Randhawa PK, Jaggi AS. TRPV4 channels: physiological and pathological role in cardiovascular system. Basic Res Cardiol 2015; 110:54. [PMID: 26415881 DOI: 10.1007/s00395-015-0512-7] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 09/11/2015] [Accepted: 09/22/2015] [Indexed: 12/12/2022]
Abstract
TRPV4 channels are non-selective cation channels permeable to Ca(2+), Na(+), and Mg(2+) ions. Recently, TRPV4 channels have received considerable attention as these channels are widely expressed in the cardiovascular system including endothelial cells, cardiac fibroblasts, vascular smooth muscles, and peri-vascular nerves. Therefore, these channels possibly play a pivotal role in the maintenance of cardiovascular homeostasis. TRPV4 channels critically regulate flow-induced arteriogenesis, TGF-β1-induced differentiation of cardiac fibroblasts into myofibroblasts, and heart failure-induced pulmonary edema. These channels also mediate hypoxia-induced increase in proliferation and migration of pulmonary artery smooth muscle cells and progression of pulmonary hypertension. These channels also maintain flow-induced vasodilation and preserve vascular function by directly activating Ca(2+)-dependent KCa channels. Furthermore, these may also induce vasodilation and maintain blood pressure indirectly by evoking the release of NO, CGRP, and substance P. The present review discusses the evidences and the potential mechanisms implicated in diverse responses including arteriogenesis, cardiac remodeling, congestive heart failure-induced pulmonary edema, pulmonary hypertension, flow-induced dilation, regulation of blood pressure, and hypoxic preconditioning.
Collapse
Affiliation(s)
- Puneet Kaur Randhawa
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University Patiala, Patiala, 147002, India
| | - Amteshwar Singh Jaggi
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University Patiala, Patiala, 147002, India.
| |
Collapse
|
25
|
Sacerdoti D, Pesce P, Di Pascoli M, Brocco S, Cecchetto L, Bolognesi M. Arachidonic acid metabolites and endothelial dysfunction of portal hypertension. Prostaglandins Other Lipid Mediat 2015; 120:80-90. [PMID: 26072731 DOI: 10.1016/j.prostaglandins.2015.05.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 05/20/2015] [Accepted: 05/25/2015] [Indexed: 12/12/2022]
Abstract
Increased resistance to portal flow and increased portal inflow due to mesenteric vasodilatation represent the main factors causing portal hypertension in cirrhosis. Endothelial cell dysfunction, defined as an imbalance between the synthesis, release, and effect of endothelial mediators of vascular tone, inflammation, thrombosis, and angiogenesis, plays a major role in the increase of resistance in portal circulation, in the decrease in the mesenteric one, in the development of collateral circulation. Reduced response to vasodilators in liver sinusoids and increased response in the mesenteric arterioles, and, viceversa, increased response to vasoconstrictors in the portal-sinusoidal circulation and decreased response in the mesenteric arterioles are also relevant to the pathophysiology of portal hypertension. Arachidonic acid (AA) metabolites through the three pathways, cyclooxygenase (COX), lipoxygenase, and cytochrome P450 monooxygenase and epoxygenase, are involved in endothelial dysfunction of portal hypertension. Increased thromboxane-A2 production by liver sinusoidal endothelial cells (LSECs) via increased COX-1 activity/expression, increased leukotriens, increased epoxyeicosatrienoic acids (EETs) (dilators of the peripheral arterial circulation, but vasoconstrictors of the portal-sinusoidal circulation), represent a major component in the increased portal resistance, in the decreased portal response to vasodilators and in the hyper-response to vasoconstrictors. Increased prostacyclin (PGI2) via COX-1 and COX-2 overexpression, and increased EETs/heme-oxygenase-1/K channels/gap junctions (endothelial derived hyperpolarizing factor system) play a major role in mesenteric vasodilatation, hyporeactivity to vasoconstrictors, and hyper-response to vasodilators. EETs, mediators of liver regeneration after hepatectomy and of angiogenesis, may play a role in the development of regenerative nodules and collateral circulation, through stimulation of vascular endothelial growth factor (VEGF) inside the liver and in the portal circulation. Pharmacological manipulation of AA metabolites may be beneficial for cirrhotic portal hypertension.
Collapse
Affiliation(s)
- David Sacerdoti
- Department of Medicine, University of Padova Via Giustiniani, 2, 35100 Padova, Italy.
| | - Paola Pesce
- Department of Medicine, University of Padova Via Giustiniani, 2, 35100 Padova, Italy
| | - Marco Di Pascoli
- Department of Medicine, University of Padova Via Giustiniani, 2, 35100 Padova, Italy
| | - Silvia Brocco
- Department of Medicine, University of Padova Via Giustiniani, 2, 35100 Padova, Italy
| | - Lara Cecchetto
- Department of Medicine, University of Padova Via Giustiniani, 2, 35100 Padova, Italy
| | - Massimo Bolognesi
- Department of Medicine, University of Padova Via Giustiniani, 2, 35100 Padova, Italy
| |
Collapse
|
26
|
Korbecki J, Baranowska-Bosiacka I, Gutowska I, Chlubek D. Vanadium Compounds as Pro-Inflammatory Agents: Effects on Cyclooxygenases. Int J Mol Sci 2015; 16:12648-68. [PMID: 26053397 PMCID: PMC4490466 DOI: 10.3390/ijms160612648] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 05/12/2015] [Accepted: 05/19/2015] [Indexed: 01/30/2023] Open
Abstract
This paper discusses how the activity and expression of cyclooxygenases are influenced by vanadium compounds at anticancer concentrations and recorded in inorganic vanadium poisonings. We refer mainly to the effects of vanadate (orthovanadate), vanadyl and pervanadate ions; the main focus is placed on their impact on intracellular signaling. We describe the exact mechanism of the effect of vanadium compounds on protein tyrosine phosphatases (PTP), epidermal growth factor receptor (EGFR), PLCγ, Src, mitogen-activated protein kinase (MAPK) cascades, transcription factor NF-κB, the effect on the proteolysis of COX-2 and the activity of cPLA2. For a better understanding of these processes, a lot of space is devoted to the transformation of vanadium compounds within the cell and the molecular influence on the direct targets of the discussed vanadium compounds.
Collapse
Affiliation(s)
- Jan Korbecki
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Powstańców Wlkp. 72 Av., 70-111 Szczecin, Poland.
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Powstańców Wlkp. 72 Av., 70-111 Szczecin, Poland.
| | - Izabela Gutowska
- Department of Biochemistry and Human Nutrition, Pomeranian Medical University, Broniewskiego 24 Str., 71-460 Szczecin, Poland.
| | - Dariusz Chlubek
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Powstańców Wlkp. 72 Av., 70-111 Szczecin, Poland.
| |
Collapse
|
27
|
Chu LY, Liou JY, Wu KK. Prostacyclin protects vascular integrity via PPAR/14-3-3 pathway. Prostaglandins Other Lipid Mediat 2015; 118-119:19-27. [PMID: 25910681 DOI: 10.1016/j.prostaglandins.2015.04.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 03/25/2015] [Accepted: 04/13/2015] [Indexed: 12/20/2022]
Abstract
Vascular integrity is protected by the lining endothelial cells (ECs) through structural and molecular protective mechanisms. In response to external stresses, ECs are dynamic in producing protective molecules such as prostacyclin (PGI2). PGI2 is known to inhibit platelet aggregation and controls smooth muscle cell contraction via IP receptors. Recent studies indicate that PGI2 defends endothelial survival and protects vascular smooth muscle cell from apoptosis via peroxisome-proliferator activated receptors (PPAR). PPAR activation results in 14-3-3 upregulation. Increase in cytosolic 14-3-3ɛ or 14-3-3β enhances binding and sequestration of Akt-mediated phosphorylated Bad and reduces Bad-mediated apoptosis via the mitochondrial pathway. Experimental data indicate that administration of PGI2 analogs or augmentation of PGI2 production by gene transfer attenuates endothelial damage and organ infarction caused by ischemia-reperfusion injury. The protective effect of PGI2 is attributed in part to preserving endothelial integrity.
Collapse
Affiliation(s)
- Ling-yun Chu
- Metabolomic Medicine Research Center, China Medical University, Taichung, Taiwan
| | - Jun-Yang Liou
- Metabolomic Medicine Research Center, China Medical University, Taichung, Taiwan; Institute of Cell and System Medicine, National Health Research Institute, Chunan, Taiwan
| | - Kenneth K Wu
- Metabolomic Medicine Research Center, China Medical University, Taichung, Taiwan; Institute of Cell and System Medicine, National Health Research Institute, Chunan, Taiwan; Department of Medical Sciences, National Tsing-Hua University, Hsin-chu, Taiwan.
| |
Collapse
|
28
|
Horn P, Stern D, Veulemans V, Heiss C, Zeus T, Merx MW, Kelm M, Westenfeld R. Improved endothelial function and decreased levels of endothelium-derived microparticles after transcatheter aortic valve implantation. EUROINTERVENTION 2015; 10:1456-63. [DOI: 10.4244/eijy14m10_02] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
29
|
Abstract
In the mammalian kidney, prostaglandins (PGs) are important mediators of physiologic processes, including modulation of vascular tone and salt and water. PGs arise from enzymatic metabolism of free arachidonic acid (AA), which is cleaved from membrane phospholipids by phospholipase A2 activity. The cyclooxygenase (COX) enzyme system is a major pathway for metabolism of AA in the kidney. COX are the enzymes responsible for the initial conversion of AA to PGG2 and subsequently to PGH2, which serves as the precursor for subsequent metabolism by PG and thromboxane synthases. In addition to high levels of expression of the "constitutive" rate-limiting enzyme responsible for prostanoid production, COX-1, the "inducible" isoform of cyclooxygenase, COX-2, is also constitutively expressed in the kidney and is highly regulated in response to alterations in intravascular volume. PGs and thromboxane A2 exert their biological functions predominantly through activation of specific 7-transmembrane G-protein-coupled receptors. COX metabolites have been shown to exert important physiologic functions in maintenance of renal blood flow, mediation of renin release and regulation of sodium excretion. In addition to physiologic regulation of prostanoid production in the kidney, increases in prostanoid production are also seen in a variety of inflammatory renal injuries, and COX metabolites may serve as mediators of inflammatory injury in renal disease.
Collapse
Affiliation(s)
- Raymond C Harris
- George M. O'Brien Kidney and Urologic Diseases Center and Division of Nephrology, Vanderbilt University School of Medicine and Nashville Veterans Affairs Hospital, Nashville, Tennessee, USA.
| | | |
Collapse
|
30
|
Mechanosensitive properties in the endothelium and their roles in the regulation of endothelial function. J Cardiovasc Pharmacol 2013; 61:461-70. [PMID: 23429585 DOI: 10.1097/fjc.0b013e31828c0933] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
: Vascular endothelial cells (ECs) line the luminal surface of blood vessels, which are exposed constantly to mechanical stimuli, such as fluid shear stress, cyclic strain, and blood pressure. In recent years, more and more evidence indicates that ECs sense these mechanical stimuli and subsequently convert mechanical stimuli into intracellular signals. The properties of ECs that sense the mechanical stimuli are defined as mechanosensors. There are a variety of mechanosensors that have been identified in ECs. These mechanosensors play an important role in regulating the function of the endothelium and vascular function, including blood pressure. This review focuses on the mechanosensors that have been identified in ECs and on the roles that mechanosensors play in the regulation of endothelium function, and in the regulation of vascular function.
Collapse
|
31
|
Abstract
At least 468 individual genes have been manipulated by molecular methods to study their effects on the initiation, promotion, and progression of atherosclerosis. Most clinicians and many investigators, even in related disciplines, find many of these genes and the related pathways entirely foreign. Medical schools generally do not attempt to incorporate the relevant molecular biology into their curriculum. A number of key signaling pathways are highly relevant to atherogenesis and are presented to provide a context for the gene manipulations summarized herein. The pathways include the following: the insulin receptor (and other receptor tyrosine kinases); Ras and MAPK activation; TNF-α and related family members leading to activation of NF-κB; effects of reactive oxygen species (ROS) on signaling; endothelial adaptations to flow including G protein-coupled receptor (GPCR) and integrin-related signaling; activation of endothelial and other cells by modified lipoproteins; purinergic signaling; control of leukocyte adhesion to endothelium, migration, and further activation; foam cell formation; and macrophage and vascular smooth muscle cell signaling related to proliferation, efferocytosis, and apoptosis. This review is intended primarily as an introduction to these key signaling pathways. They have become the focus of modern atherosclerosis research and will undoubtedly provide a rich resource for future innovation toward intervention and prevention of the number one cause of death in the modern world.
Collapse
Affiliation(s)
- Paul N Hopkins
- Cardiovascular Genetics, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA.
| |
Collapse
|
32
|
Toniolo A, Buccellati C, Pinna C, Gaion RM, Sala A, Bolego C. Cyclooxygenase-1 and prostacyclin production by endothelial cells in the presence of mild oxidative stress. PLoS One 2013; 8:e56683. [PMID: 23441213 PMCID: PMC3575487 DOI: 10.1371/journal.pone.0056683] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Accepted: 01/14/2013] [Indexed: 11/19/2022] Open
Abstract
This study aimed at evaluating the relative contribution of endothelial cyclooxygenase-1 and -2 (COX-1 and COX-2) to prostacyclin (PGI2) production in the presence of mild oxidative stress resulting from autooxidation of polyphenols such as (-)-epigallocatechin 3-gallate (EGCG), using both endothelial cells in culture and isolated blood vessels. EGCG treatment resulted in an increase in hydrogen peroxide formation in human umbilical vein endothelial cells. In the presence of exogenous arachidonic acid and EGCG, PGI2 production was preferentially inhibited by a selective COX-1 inhibitor. This effect of selective inhibition was also substantially reversed by catalase. In addition, EGCG caused vasorelaxation of rat aortic ring only partially abolished by a nitric oxide synthase inhibitor. Concomitant treatment with a selective COX-1 inhibitor completely prevented the vasorelaxation as well as the increase in PGI2 accumulation in the perfusate observed in EGCG-treated aortic rings, while a selective COX-2 inhibitor was completely uneffective. Our data strongly support the notions that H2O2 generation affects endothelial PGI2 production, making COX-1, and not COX-2, the main source of endothelial PGI2 under altered oxidative tone conditions. These results might be relevant to the reappraisal of the impact of COX inhibitors on vascular PGI2 production in patients undergoing significant oxidative stress.
Collapse
Affiliation(s)
- Alice Toniolo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Carola Buccellati
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Milano, Italy
| | - Christian Pinna
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Milano, Italy
| | - Rosa Maria Gaion
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Angelo Sala
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Milano, Italy
- IBIM, Consiglio Nazionale delle Ricerche, Palermo, Italy
- * E-mail:
| | - Chiara Bolego
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| |
Collapse
|
33
|
|
34
|
Cyclooxygenase-1, not cyclooxygenase-2, is responsible for physiological production of prostacyclin in the cardiovascular system. Proc Natl Acad Sci U S A 2012; 109:17597-602. [PMID: 23045674 DOI: 10.1073/pnas.1209192109] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Prostacyclin is an antithrombotic hormone produced by the endothelium, whose production is dependent on cyclooxygenase (COX) enzymes of which two isoforms exist. It is widely believed that COX-2 drives prostacyclin production and that this explains the cardiovascular toxicity associated with COX-2 inhibition, yet the evidence for this relies on indirect evidence from urinary metabolites. Here we have used a range of experimental approaches to explore which isoform drives the production of prostacyclin in vitro and in vivo. Our data show unequivocally that under physiological conditions it is COX-1 and not COX-2 that drives prostacyclin production in the cardiovascular system, and that urinary metabolites do not reflect prostacyclin production in the systemic circulation. With the idea that COX-2 in endothelium drives prostacyclin production in healthy individuals removed, we must seek new answers to why COX-2 inhibitors increase the risk of cardiovascular events to move forward with drug discovery and to enable more informed prescribing advice.
Collapse
|
35
|
Nakabayashi S, Nagaoka T, Tani T, Sogawa K, Hein TW, Kuo L, Yoshida A. Retinal arteriolar responses to acute severe elevation in systemic blood pressure in cats: role of endothelium-derived factors. Exp Eye Res 2012; 103:63-70. [PMID: 22940370 DOI: 10.1016/j.exer.2012.08.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2012] [Revised: 08/15/2012] [Accepted: 08/17/2012] [Indexed: 11/24/2022]
Abstract
The purpose of this study was to investigate the roles of endothelium-derived factors in the retinal arteriolar responses to acute severe elevation in systemic blood pressure (BP) in cats. Acute elevation of mean arterial BP by 60% for 5 min was achieved by inflating a balloon-tipped catheter in the descending aorta. The retinal arteriolar diameter, flow velocity, wall shear rate (WSR) and blood flow (RBF) changes during BP elevation were assessed with laser Doppler velocimetry 2 h after intravitreal injections of nitric oxide (NO) synthase inhibitor l-NAME, cyclooxygenase inhibitor indomethacin, endothelin-1 receptor antagonists (BQ-123 for type A and BQ-788 for type B), or Rho kinase inhibitor fasudil. BP elevation caused a marked increase in retinal arteriolar flow velocity and WSR with slight vasoconstriction, resulting in an increase in RBF. The increases in velocity, WSR and RBF, but not diameter, were correlated with the increase in ocular perfusion pressure. With l-NAME or indomethacin, the increase in RBF upon BP elevation was significantly attenuated due to enhanced retinal arteriolar vasoconstriction. In contrast, BQ-123 and fasudil potentiated the increased RBF. BQ-788 had no effect on arteriolar diameter and hemodynamics. Our data suggest that acute elevation of BP by 60% leads to an increase in RBF due to the release of NO and prostanoids probably through a shear stress-induced vasodilation mechanism. The release of endothelin-1 and Rho kinase activation help to limit RBF augmentation by counteracting the vasodilation. It appears that the retinal endothelium, by releasing vasoactive substances, contributes to RBF regulation during acute severe elevation of systemic blood pressure.
Collapse
Affiliation(s)
- Seigo Nakabayashi
- Department of Ophthalmology, Asahikawa Medical University, Midorigaoka Higashi 2-1-1-1, Asahikawa 078-8510, Japan
| | | | | | | | | | | | | |
Collapse
|
36
|
Association between peripheral vascular endothelial dysfunction and livedoid vasculopathy. J Am Acad Dermatol 2011; 67:107-12. [PMID: 21982058 DOI: 10.1016/j.jaad.2011.07.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2011] [Revised: 06/27/2011] [Accepted: 07/21/2011] [Indexed: 11/23/2022]
Abstract
BACKGROUND Livedoid vasculopathy (LV) is a disease characterized by multiple painful and recurrent ulcerations on the feet, accompanied by atrophic scars. Many researchers suggest that a hypercoagulable status is the pathogenetic factor for LV. However, the cause of LV remains elusive. OBJECTIVE We sought to determine if endothelial dysfunction is present in patients with LV. METHODS This prospective study included 16 patients with LV and active ulcers and 16 matched control subjects. We reviewed detailed clinical parameters, including antinuclear antibody, high-sensitivity C-reactive protein, protein C, protein S, homocysteine, anti-SSA, anti-SSB, anticardiolipin antibody, and serum lipid profiles. Flow-mediated vasodilation of the brachial artery was used as an indicator of vascular endothelial function using high-resolution 2-dimensional ultrasonic imaging. RESULTS Blood pressure, blood biochemistry, high-sensitivity C-reactive protein, and homocysteine were not significantly different in patients with LV and control subjects. Nitroglycerin-mediated vasodilation was not significantly different in patients with LV and control subjects. However, flow-mediated vasodilation was much less in patients with LV than in the control group (3.58 ± 2.32% vs 7.51 ± 2.40%, P < .001). LIMITATIONS The study was performed at a single site with a limited sample size. CONCLUSION Peripheral vascular endothelial dysfunction was demonstrated in patients with LV by reduction of brachial flow-mediated vasodilation.
Collapse
|
37
|
Jazuli F, Pyke KE. The impact of baseline artery diameter on flow-mediated vasodilation: a comparison of brachial and radial artery responses to matched levels of shear stress. Am J Physiol Heart Circ Physiol 2011; 301:H1667-77. [DOI: 10.1152/ajpheart.00487.2011] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
An inverse relationship between baseline artery diameter (BAD) and flow-mediated vasodilation (FMD) has been identified using reactive hyperemia (RH) to create a shear stress (SS) stimulus in human conduit arteries. However, RH creates a SS stimulus that is inversely related to BAD. The purpose of this study was to compare FMD in response to matched levels of SS in two differently sized upper limb arteries [brachial (BA) and radial (RA) artery]. With the use of exercise, three distinct, shear rate (SR) stimuli were created (SR = blood velocity/vessel diameter; estimate of SS) in the RA and BA. Artery diameter and mean blood velocity were assessed with echo and Doppler ultrasound in 15 healthy male subjects (19–25 yr). Data are means ± SE. Subjects performed 6 min of adductor pollicis and handgrip exercise to increase SR in the RA and BA, respectively. Exercise intensity was modulated to achieve uniformity in SR between arteries. The three distinct SR levels were as follows: steady-state exercise 39.8 ± 0.6, 57.3 ± 0.7, and 72.4 ± 1.2 s−1 ( P < 0.001). %FMD and AbsFMD (mm) at the end of exercise were greater in the RA vs. the BA at each shear level [at the highest level: RA = 15.7 ± 1.5%, BA = 5.4 ± 0.8% ( P < 0.001)]. The mean slope of the within-subject SR-%FMD regression line was greater in the RA (RA = 0.33 ± 0.04, BA = 0.13 ± 0.02, P < 0.001), and a strong within-subjects relationship between %FMD and SR was observed in both arteries (RA: r2 = 0.92 ± 0.02; BA: r2 = 0.90 ± 0.03). Within the RA, there was a significant relationship between baseline diameter and %FMD; however, this relationship was not present in the BA (RA: r2 = 0.76, P < 0.001; BA: r2 = 0.03, P = 0.541). These findings suggest that the response to SS is not uniform across differently sized vessels, which is in agreement with previous studies.
Collapse
Affiliation(s)
- F. Jazuli
- Queen's University, Kingston, Ontario, Canada
| | - K. E. Pyke
- Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
38
|
COX-derived prostanoid pathways in gastrointestinal cancer development and progression: novel targets for prevention and intervention. Biochim Biophys Acta Rev Cancer 2011; 1825:49-63. [PMID: 22015819 DOI: 10.1016/j.bbcan.2011.09.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Revised: 09/22/2011] [Accepted: 09/24/2011] [Indexed: 12/27/2022]
Abstract
Arachidonic acid metabolism through cyclooxygenase (COX) pathways leads to the generation of biologically active eicosanoids. Eicosanoid expression levels vary during development and progression of gastrointestinal (GI) malignancies. COX-2 is the major COX-isoform responsible for G.I. cancer development/progression. COX-2 expression increases during progression from a normal to cancerous state. Evidence from observational studies has demonstrated that chronic NSAID use reduces the risk of cancer development, while both incidence and risk of death due to G.I. cancers were significantly reduced by daily aspirin intake. A number of randomized controlled trials (APC trial, Prevention of Sporadic Adenomatous Polyps trial, APPROVe trial) have also shown a significant protective effect in patients receiving selective COX-2 inhibitors. However, chronic use of selective COX-2 inhibitors at high doses was associated with increased cardiovascular risk, while NSAIDs have also been associated with increased risk. More recently, downstream effectors of COX-signaling have been investigated in cancer development/progression. PGE(2), which binds to both EP and PPAR receptors, is the major prostanoid implicated in the carcinogenesis of G.I. cancers. The role of TXA(2) in G.I. cancers has also been examined, although further studies are required to uncover its role in carcinogenesis. Other prostanoids investigated include PGD(2) and its metabolite 15d-PGJ2, PGF(1α) and PGI(2). Targeting these prostanoids in G.I. cancers has the promise of avoiding cardiovascular toxicity associated with chronic selective COX-2 inhibition, while maintaining anti-tumor reactivity. A progressive sequence from normal to pre-malignant to a malignant state has been identified in G.I. cancers. In this review, we will discuss the role of the COX-derived prostanoids in G.I. cancer development and progression. Targeting these downstream prostanoids for chemoprevention and/or treatment of G.I. cancers will also be discussed. Finally, we will highlight the latest pre-clinical technologies as well as avenues for future investigation in this highly topical research field.
Collapse
|
39
|
Lu D, Kassab GS. Role of shear stress and stretch in vascular mechanobiology. J R Soc Interface 2011; 8:1379-85. [PMID: 21733876 DOI: 10.1098/rsif.2011.0177] [Citation(s) in RCA: 179] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Blood vessels are under constant mechanical loading from blood pressure and flow which cause internal stresses (endothelial shear stress and circumferential wall stress, respectively). The mechanical forces not only cause morphological changes of endothelium and blood vessel wall, but also trigger biochemical and biological events. There is considerable evidence that physiologic stresses and strains (stretch) exert vasoprotective roles via nitric oxide and provide a homeostatic oxidative balance. A perturbation of tissue stresses and strains can disturb biochemical homeostasis and lead to vascular remodelling and possible dysfunction (e.g. altered vasorelaxation, tone, stiffness, etc.). These distinct biological endpoints are caused by some common biochemical pathways. The focus of this brief review is to point out some possible commonalities in the molecular pathways in response to endothelial shear stress and circumferential wall stretch.
Collapse
Affiliation(s)
- Deshun Lu
- Department of Biomedical Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | | |
Collapse
|
40
|
Williamson S, Varma D, Brown M, Jansen S. Eicosanoid Production following One Bout of Exercise in Middle-Aged African American Pre- and Stage 1 Hypertensives. J Aging Res 2011; 2011:302802. [PMID: 21629748 PMCID: PMC3100569 DOI: 10.4061/2011/302802] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2010] [Revised: 01/31/2011] [Accepted: 03/10/2011] [Indexed: 12/31/2022] Open
Abstract
Endothelial dysfunction and a sedentary lifestyle may be involved in the development of hypertension which is proliferative among middle-aged African Americans (AA). Signaling molecules derived from the oxidation of 20-carbon fatty acid molecules known as eicosanoids influence vascular tone. The relationship between aerobic fitness and eicosanoid formation following exercise in middle-aged African American hypertensives is unknown. Purpose. To determine the relationship between aerobic capacity and eicosanoid formation after a bout of moderate-intensity exercise in middle-aged AA hypertensives. Methods. Ten sedentary hypertensive AA underwent 50 min of aerobic exercise at 65% VO2max. Urine was collected for 24 hr on two occasions, prior to testing and immediately following the bout of exercise. Urinary metabolites of prostacyclin (6-keto PGF1α) and thromboxane (11-dTXB2) were measured during the day and night periods by high-performance liquid chromatography (HPLC). Results. 6-keto PGF1α levels significantly increased (P = .04) following the bout of exercise compared to the control day. There was a significant relationship (r = .49, P < .05) between 6-keto PGF1α levels and VO2max during the exercise day. Conclusion. Based on this preliminary study, there appears to be a relationship between aerobic capacity and exercise-induced 6-keto PGF1α production in middle-aged hypertensive AAs. AAs with lower VO2max had lower 6-keto PGF1α formation.
Collapse
Affiliation(s)
- Sheara Williamson
- Hypertension, Molecular, and Applied Physiology Laboratory, Department of Kinesiology, Temple University, 1800 North Broad Street, Philadelphia, PA 19122, USA
| | | | | | | |
Collapse
|
41
|
Kenagy RD, Min SK, Mulvihill E, Clowes AW. A link between smooth muscle cell death and extracellular matrix degradation during vascular atrophy. J Vasc Surg 2011; 54:182-191.e24. [PMID: 21493032 DOI: 10.1016/j.jvs.2010.12.070] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2010] [Revised: 12/07/2010] [Accepted: 12/11/2010] [Indexed: 12/12/2022]
Abstract
OBJECTIVE High blood flow induces neointimal atrophy in polytetrafluoroethylene (PTFE) aortoiliac grafts and a tight external PTFE wrap of the iliac artery induces medial atrophy. In both nonhuman primate models, atrophy with loss of smooth muscle cells and extracellular matrix (ECM) begins at ≤4 days. We hypothesized that matrix loss would be linked to cell death, but the factors and mechanisms involved are not known. The purpose of this study was to determine commonly regulated genes in these two models, which we hypothesized would be a small set of genes that might be key regulators of vascular atrophy. METHODS DNA microarray analysis (Sentrix Human Ref 8; Illumina, San Diego, Calif; ∼23,000 genes) was performed on arterial tissue from the wrap model (n = 9) and graft neointima from the graft model (n = 5) 1 day after wrapping or the switch to high flow, respectively. Quantitative reverse-transcription polymerase chain reaction (qRT-PCR) was also performed. Expression of this vascular atrophy gene set was also studied after Fas ligand-induced cell death in cultured smooth muscle cells and organ cultured arteries. RESULTS Microarray analysis showed 15 genes were regulated in the same direction in both atrophy models: 9 upregulated and 6 downregulated. Seven of nine upregulated genes were confirmed by qRT-PCR in both models. Upregulated genes included the ECM-degrading enzymes ADAMTS4, tissue plasminogen activator (PLAT), and hyaluronidase 2; possible growth regulatory factors, including chromosome 8 open reading frame 4 and leucine-rich repeat family containing 8; a differentiation regulatory factor (musculoskeletal embryonic nuclear protein 1); a dead cell removal factor (ficolin 3); and a prostaglandin transporter (solute carrier organic anion transporter family member 2A1). Five downregulated genes were confirmed but only in one or the other model. Of the seven upregulated genes, ADAMTS4, PLAT, hyaluronidase 2, solute carrier organic anion transporter family member 2A1, leucine-rich repeat family containing 8, and chromosome 8 open reading frame 4 were also upregulated in vitro in cultured smooth muscle cells or cultured iliac artery by treatment with FasL, which causes cell death. However, blockade of caspase activity with Z-VAD inhibited FasL-mediated cell death, but not gene induction. CONCLUSION Seven gene products were upregulated in two distinctly different in vivo nonhuman primate vascular atrophy models. Induction of cell death by FasL in vitro induced six of these genes, including the ECM-degrading factors ADAMTS4, hyaluronidase 2, and PLAT, suggesting a mechanism by which the program of tissue atrophy coordinately removes extracellular matrix as cells die. These genes may be key regulators of vascular atrophy.
Collapse
Affiliation(s)
- Richard D Kenagy
- Department of Surgery, University of Washington, Seattle, WA 98195-6410, USA
| | | | | | | |
Collapse
|
42
|
Abstract
This article reviews existing methods for the isolation, fractionation, or capture of rare cells in microfluidic devices. Rare cell capture devices face the challenge of maintaining the efficiency standard of traditional bulk separation methods such as flow cytometers and immunomagnetic separators while requiring very high purity of the target cell population, which is typically already at very low starting concentrations. Two major classifications of rare cell capture approaches are covered: (1) non-electrokinetic methods (e.g., immobilization via antibody or aptamer chemistry, size-based sorting, and sheath flow and streamline sorting) are discussed for applications using blood cells, cancer cells, and other mammalian cells, and (2) electrokinetic (primarily dielectrophoretic) methods using both electrode-based and insulative geometries are presented with a view towards pathogen detection, blood fractionation, and cancer cell isolation. The included methods were evaluated based on performance criteria including cell type modeled and used, number of steps/stages, cell viability, and enrichment, efficiency, and/or purity. Major areas for improvement are increasing viability and capture efficiency/purity of directly processed biological samples, as a majority of current studies only process spiked cell lines or pre-diluted/lysed samples. Despite these current challenges, multiple advances have been made in the development of devices for rare cell capture and the subsequent elucidation of new biological phenomena; this article serves to highlight this progress as well as the electrokinetic and non-electrokinetic methods that can potentially be combined to improve performance in future studies.
Collapse
|
43
|
Enhanced cyclooxygenase 2-mediated vasorelaxation in coronary arteries from insulin-resistant obese Zucker rats. Atherosclerosis 2010; 213:392-9. [DOI: 10.1016/j.atherosclerosis.2010.09.022] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2010] [Revised: 09/13/2010] [Accepted: 09/17/2010] [Indexed: 01/02/2023]
|
44
|
Walshe TE, Connell P, Cryan L, Ferguson G, O'Brien C, Cahill PA. The role of pulsatile flow in controlling microvascular retinal endothelial and pericyte cell apoptosis and proliferation. Cardiovasc Res 2010; 89:661-70. [PMID: 21030535 DOI: 10.1093/cvr/cvq341] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
AIMS Aberrant retinal blood flow is a hallmark of various retinopathies and may be a causative factor in the pathology associated with these conditions. We examined the effects of pulsatile flow on bovine retinal endothelial cell (BREC) and bovine retinal pericyte (BRP) apoptosis and proliferation. METHODS AND RESULTS Co-cultured BRECs and BRPs were exposed to low (0.3 mL/min) or high (25 mL/min) pulsatile flow for 72 h using a perfused transcapillary culture system. Pulsatile flow increased BREC nitric oxide synthase (eNOS) and cyclooxygenase-2 (COX-2) expression and activity concomitant with a significant decrease in pre-pro-endothelin-1 (ET-1) mRNA and peptide. BREC apoptosis was significantly attenuated following exposure to high flow. The inhibition of NOS, COX, and ET receptors significantly reduced the pro-survival effects of flow on BREC. In contrast, BRP apoptosis was significantly enhanced following exposure to high flow. The inhibition of COX and ET receptors significantly attenuated the high flow-induced increase in BRP apoptosis when compared with untreated controls. Treatment of static BREC with NO donor (S-nitroso-N-acetylpenicillamine, SNAP), ET-1, or iloprost inhibited serum deprivation-induced apoptosis, whereas treatment of BRP with ET-1 and iloprost, but not SNAP, was ineffective. High pulsatile flow decreased BRP proliferation, in the absence of any changes in BREC proliferation. CONCLUSION Increased pulsatile flow promotes BREC survival and enhances BRP apoptosis through the activation of endothelial-derived vasoactive substances. Altered pulsatile flow does not alter BREC proliferation in co-culture with BRP, whereas BRP proliferation was significantly decreased at high flow rates. These interactions have important implications for vessel growth and regression during retinal vascular pathogenesis.
Collapse
Affiliation(s)
- Tony E Walshe
- Vascular Health Research Centre, Faculty of Science and Health, Dublin City University, Dublin 9, Ireland.
| | | | | | | | | | | |
Collapse
|
45
|
Warboys CM, Eric Berson R, Mann GE, Pearson JD, Weinberg PD. Acute and chronic exposure to shear stress have opposite effects on endothelial permeability to macromolecules. Am J Physiol Heart Circ Physiol 2010; 298:H1850-6. [PMID: 20363882 PMCID: PMC2886649 DOI: 10.1152/ajpheart.00114.2010] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2010] [Accepted: 04/01/2010] [Indexed: 11/22/2022]
Abstract
Endothelial properties are affected by mechanical stresses. Several studies have shown that an acute application of shear stress increases the permeability of endothelial monolayers in culture. We investigated whether more prolonged application of shear has the opposite effect. Porcine aortic endothelial cells were cultured on Transwell filters to assess monolayer permeability to albumin. The medium above the cells was swirled using an orbital shaker; resultant shears were computed to lie within the physiological range. Acute application of shear increased permeability, but chronic application reduced it. The effect of chronic but not acute shear was reversed by inhibiting nitric oxide (NO) synthesis. The effect of chronic shear was also reversed by inhibiting phosphatidylinositol 3-OH kinase (PI3K) and soluble guanylyl cyclase. None of these interventions affected permeability under static conditions, and inhibition of cyclooxygenase was without effect. Chronic shear decreased mitosis rates by a fraction comparable to the reduction in permeability, but this effect was not reversed by inhibiting NO synthesis. We conclude that chronic application of shear stress reduces endothelial permeability to macromolecules by a PI3K-NO-cGMP-dependent mechanism. Since atherosclerosis can be triggered by excessive entry of plasma macromolecules into the arterial wall, the phenomenon may help explain the atheroprotective effects of shear and NO.
Collapse
|
46
|
Watanabe J, Lin JA, Narasimha AJ, Shahbazian A, Ishikawa TO, Martin MG, Herschman HR, Reddy ST. Novel anti-inflammatory functions for endothelial and myeloid cyclooxygenase-2 in a new mouse model of Crohn's disease. Am J Physiol Gastrointest Liver Physiol 2010; 298:G842-50. [PMID: 20299600 PMCID: PMC8875131 DOI: 10.1152/ajpgi.00468.2009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Cyclooxygenase-2 (COX-2) is an important regulator of inflammation implicated in the development of a variety of diseases, including inflammatory bowel disease (IBD). However, the regulation of intestinal inflammation by COX-2 is poorly understood. We previously reported that COX-2(-/-) mice fed a cholate-containing high-fat (CCHF) diet had high mortality of unknown mechanisms attributable to severe intestinal inflammation in the ileo-ceco-colic junction that presented characteristics similar to Crohn's disease (CD). To further characterize the role of COX-2 in intestinal inflammation, we established cell-specific conditional COX-2(-/-) mice. Endothelial cell-specific (COX-2(-E/-E)) and myeloid cell-specific (COX-2(-M/-M)) COX-2(-/-) mice, but not wild-type mice, on the CCHF diet developed localized CD-like pathology at the ileo-ceco-colic junction that was associated with cellular infiltration, increased expression of myeloperoxidase and IL-5, and decreased IL-10 expression. The CD-like pathology in COX-2(-E/-E) mice was also accompanied by increased expression of cytokines (IL-6, TNF-alpha, and INF-gamma), compared with wild-type mice and COX-2(-M/-M) mice. In contrast, the ileo-ceco-colic inflammation in COX-2(-M/-M) mice was associated with more pronounced infiltration of granulocytes and macrophages than COX-2(-E/-E) mice. COX-2(-ME/-ME) (COX-2(-M/-M) x COX-2(-E/-E)) mice on the CCHF diet developed CD-like pathology in the ileo-ceco-colic junction reminiscent of total COX-2(-/-) mice on CCHF diet and wild-type mice on CCHF diet treated with COX-2 inhibitor, celecoxib. The pathology of diet-mediated ileo-ceco-colic inflammation in COX-2(-/-) mice offers an excellent model system to elucidate the protective roles of endothelial and myeloid COX-2 and the molecular pathogenesis of CD.
Collapse
Affiliation(s)
- Junji Watanabe
- 1Atherosclerosis Research Unit, Department of Medicine/Cardiology,
David Geffen School of Medicine, University of California, Los Angeles, California
| | - James A. Lin
- 2Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Ajay J. Narasimha
- 3Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Ani Shahbazian
- 1Atherosclerosis Research Unit, Department of Medicine/Cardiology,
David Geffen School of Medicine, University of California, Los Angeles, California
| | - Tomo-o Ishikawa
- 3Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Martin G. Martin
- 2Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Harvey R. Herschman
- 3Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, California,4Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, California,5Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Srinivasa T. Reddy
- 1Atherosclerosis Research Unit, Department of Medicine/Cardiology,
David Geffen School of Medicine, University of California, Los Angeles, California,3Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, California,5Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, California
| |
Collapse
|
47
|
Human endothelial dysfunction: EDRFs. Pflugers Arch 2010; 459:1005-13. [DOI: 10.1007/s00424-010-0822-4] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2010] [Revised: 03/03/2010] [Accepted: 03/04/2010] [Indexed: 02/02/2023]
|
48
|
Sánchez A, Contreras C, Villalba N, Martínez P, Martínez AC, Bríones A, Salaíces M, García-Sacristán A, Hernández M, Prieto D. Altered arachidonic acid metabolism via COX-1 and COX-2 contributes to the endothelial dysfunction of penile arteries from obese Zucker rats. Br J Pharmacol 2010; 159:604-16. [PMID: 20082610 DOI: 10.1111/j.1476-5381.2009.00553.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND AND PURPOSE The aim of the current study was to investigate the role of arachidonic acid (AA) metabolism via cyclooxygenase (COX) in the endothelial dysfunction of penile arteries from pre-diabetic, obese Zucker rats (OZR). EXPERIMENTAL APPROACH Penile arteries from OZR and from lean Zucker rats (LZR) were mounted in microvascular myographs to assess vascular function and COX expression was determined by immunohistochemistry. KEY RESULTS Acetylcholine (ACh) and AA elicited relaxations that were impaired in arteries from OZR. Inhibition of both COX-1 and COX-2 reduced the relaxant effects of ACh and AA in LZR but not in OZR. Inhibitors of COX-1 and of the TXA(2)/PGH(2) (TP) receptor enhanced the relaxations induced by AA in both LZR and OZR, whereas COX-2 inhibition enhanced these responses only in OZR. TP receptor blockade did not restore ACh relaxant responses in arteries from OZR. Inhibition of COX-1 increased basal tension in OZR and this contraction was blunted by TP receptor blockade. The vasoconstrictor responses to noradrenaline were augmented by indomethacin and by COX-2 inhibition in LZR but not in OZR. Immunohistochemical staining showed that both COX-1 and COX-2 are expressed in the endothelium of penile arteries from both LZR and OZR. CONCLUSIONS AND IMPLICATIONS Vasoactive prostanoids were formed via constitutively active COX-1 and COX-2 pathways in normal rat penile arteries. Under conditions of insulin resistance, the release and/or effects of vasodilator prostanoids were impaired, contributing to the blunted endothelium-dependent vasodilatation and to the enhanced vasoconstriction.
Collapse
Affiliation(s)
- A Sánchez
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense, Madrid, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Mbonye UR, Song I. Posttranscriptional and posttranslational determinants of cyclooxygenase expression. BMB Rep 2009; 42:552-60. [PMID: 19788855 DOI: 10.5483/bmbrep.2009.42.9.552] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Cyclooxygenases (COX-1 and COX-2) are ER-resident proteins that catalyze the committed step in prostanoid synthesis. COX-1 is constitutively expressed in many mammalian cells, whereas COX-2 is usually expressed inducibly and transiently. Abnormal expression of COX-2 has been implicated in the pathogenesis of chronic inflammation and various cancers; therefore, it is subject to tight and complex regulation. Differences in regulation of the COX enzymes at the posttranscriptional and posttranslational levels also contribute significantly to their distinct patterns of expression. Rapid degradation of COX-2 mRNA has been attributed to AU-rich elements (AREs) at its 3' UTR. Recently, microRNAs that can selectively repress COX-2 protein synthesis have been identified. The mature forms of these COX proteins are very similar in structure except that COX-2 has a unique 19-amino acid (19-aa) segment located near the C-terminus. This C-terminal 19-aa cassette plays an important role in mediation of the entry of COX-2 into the ER-associated degradation (ERAD) system, which transports ER proteins to the cytoplasm for degradation by the 26S proteasome. A second pathway for COX-2 protein degradation is initiated after the enzyme undergoes suicide inactivation following cyclooxygenase catalysis. Here, we discuss these molecular determinants of COX-2 expression in detail. [BMB reports 2009; 42(9): 552-560].
Collapse
Affiliation(s)
- Uri R Mbonye
- Department of Life Science, University of Seoul, Seoul 130-743, Korea
| | | |
Collapse
|
50
|
Cyclic stretch induces cyclooxygenase-2 gene expression in vascular endothelial cells via activation of nuclear factor kappa-beta. Biochem Biophys Res Commun 2009; 389:599-601. [PMID: 19748489 DOI: 10.1016/j.bbrc.2009.09.028] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2009] [Accepted: 09/04/2009] [Indexed: 01/11/2023]
Abstract
Vascular endothelial cells respond to biomechanical forces, such as cyclic stretch and shear stress, by altering gene expression. Since endothelial-derived prostanoids, such as prostacyclin and thromboxane A(2), are key mediators of endothelial function, we investigated the effects of cyclic stretch on the expression of genes in human umbilical vein endothelial cells controlling prostanoid synthesis: cyclooxygenase-1 (COX-1), cyclooxygenase-2 (COX-2), prostacyclin synthase (PGIS) and thromboxane A(2) synthase (TXAS). COX-2 and TXAS mRNAs were upregulated by cyclic stretch for 24h. In contrast, PGIS mRNA was decreased and stretch had no effect on COX-1 mRNA expression. We further show that stretch-induced upregulation of COX-2 is mediated by activation of the NF-kappabeta signaling pathway.
Collapse
|