Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Cardiovasc Res ; 118(11): 2506-2518, 2022 08 24.
Article in English | MEDLINE | ID: mdl-34528082

ABSTRACT

AIMS: Sepsis-induced lung injury is associated with significant morbidity and mortality. Previously, we showed that heterozygous deletion of polymerase δ-interacting protein 2 (Poldip2) was protective against sepsis-induced lung injury. Since endothelial barrier disruption is thought to be the main mechanism of sepsis-induced lung injury, we sought to determine if the observed protection was specifically due to the effect of reduced endothelial Poldip2. METHODS AND RESULTS: Endothelial-specific Poldip2 knock-out mice (EC-/-) and their wild-type littermates (EC+/+) were injected with saline or lipopolysaccharide (18 mg/kg) to model sepsis-induced lung injury. At 18 h post-injection mice, were euthanized and bronchoalveolar lavage (BAL) fluid and lung tissue were collected to assess leucocyte infiltration. Poldip2 EC-/- mice showed reduced lung leucocyte infiltration in BAL (0.21 ± 0.9×106 vs. 1.29 ± 1.8×106 cells/mL) and lung tissue (12.7 ± 1.8 vs. 23 ± 3.7% neutrophils of total number of cells) compared to Poldip2 EC+/+ mice. qPCR analysis of the lung tissue revealed a significantly dampened induction of inflammatory gene expression (TNFα 2.23 ± 0.39 vs. 4.15 ± 0.5-fold, IκBα 4.32 ± 1.53 vs. 8.97 ± 1.59-fold), neutrophil chemoattractant gene expression (CXCL1 68.8 ± 29.6 vs. 147 ± 25.7-fold, CXCL2 65 ± 25.6 vs. 215 ± 27.3-fold) and a marker of endothelial activation (VCAM1 1.25 ± 0.25 vs. 3.8 ± 0.38-fold) in Poldip2 EC-/- compared to Poldip2 EC+/+ lungs. An in vitro model using human pulmonary microvascular endothelial cells was used to assess the effect of Poldip2 knock-down on endothelial activation and permeability. TNFα-induced endothelial permeability and VE-cadherin disruption were significantly reduced with siRNA-mediated knock-down of Poldip2 (5 ± 0.5 vs. 17.5 ± 3-fold for permeability, 1.5 ± 0.4 vs. 10.9 ± 1.3-fold for proportion of disrupted VE-cadherin). Poldip2 knock-down altered expression of Rho-GTPase-related genes, which correlated with reduced RhoA activation by TNFα (0.94 ± 0.05 vs. 1.29 ± 0.01 of relative RhoA activity) accompanied by redistribution of active-RhoA staining to the centre of the cell. CONCLUSION: Poldip2 is a potent regulator of endothelial dysfunction during sepsis-induced lung injury, and its endothelium-specific inhibition may provide clinical benefit.


Subject(s)
Lung Injury , Mitochondrial Proteins/metabolism , Nuclear Proteins/metabolism , Sepsis , Animals , Endothelium/metabolism , Humans , Lung/metabolism , Lung Injury/genetics , Mice , Mitochondrial Proteins/genetics , Nuclear Proteins/genetics , Sepsis/complications , Sepsis/genetics , Sepsis/metabolism , Tumor Necrosis Factor-alpha/metabolism
2.
Hypertension ; 76(1): 267-276, 2020 07.
Article in English | MEDLINE | ID: mdl-32389075

ABSTRACT

Endothelial inflammation and mitochondrial dysfunction have been implicated in cardiovascular diseases, yet, a unifying mechanism tying them together remains limited. Mitochondrial dysfunction is frequently associated with mitochondrial fission/fragmentation mediated by the GTPase Drp1 (dynamin-related protein 1). Nuclear factor (NF)-κB, a master regulator of inflammation, is implicated in endothelial dysfunction and resultant complications. Here, we explore a causal relationship between mitochondrial fission and NF-κB activation in endothelial inflammatory responses. In cultured endothelial cells, TNF-α (tumor necrosis factor-α) or lipopolysaccharide induces mitochondrial fragmentation. Inhibition of Drp1 activity or expression suppresses mitochondrial fission, NF-κB activation, vascular cell adhesion molecule-1 induction, and leukocyte adhesion induced by these proinflammatory factors. Moreover, attenuations of inflammatory leukocyte adhesion were observed in Drp1 heterodeficient mice as well as endothelial Drp1 silenced mice. Intriguingly, inhibition of the canonical NF-κB signaling suppresses endothelial mitochondrial fission. Mechanistically, NF-κB p65/RelA seems to mediate inflammatory mitochondrial fission in endothelial cells. In addition, the classical anti-inflammatory drug, salicylate, seems to maintain mitochondrial fission/fusion balance against TNF-α via inhibition of NF-κB. In conclusion, our results suggest a previously unknown mechanism whereby the canonical NF-κB cascade and a mitochondrial fission pathway interdependently regulate endothelial inflammation.


Subject(s)
Dynamins/physiology , Endothelial Cells/physiology , Endothelium, Vascular/pathology , Mitochondrial Dynamics/physiology , NF-kappa B/metabolism , Vasculitis/physiopathology , 3T3 Cells , Animals , Aorta/cytology , Cell Adhesion , Cells, Cultured , Dynamins/antagonists & inhibitors , Dynamins/genetics , Endothelial Cells/drug effects , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , Membrane Proteins/physiology , Mice , Mitochondrial Proteins/physiology , Mutation, Missense , Phosphorylation , Phosphoserine/metabolism , Protein Processing, Post-Translational , Proteome , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Rats , Sodium Salicylate/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Vascular Cell Adhesion Molecule-1/biosynthesis , Vascular Cell Adhesion Molecule-1/genetics
3.
Int J Mol Sci ; 21(9)2020 Apr 28.
Article in English | MEDLINE | ID: mdl-32354103

ABSTRACT

Angiotensin II (AngII) has a crucial role in cardiovascular pathologies, including endothelial inflammation and premature vascular aging. However, the precise molecular mechanism underlying aging-related endothelial inflammation induced by AngII remains elusive. Here, we have tested a hypothesis in cultured rat aortic endothelial cells (ECs) that the removal of AngII-induced senescent cells, preservation of proteostasis, or inhibition of mitochondrial fission attenuates the pro-inflammatory EC phenotype. AngII stimulation in ECs resulted in cellular senescence assessed by senescence-associated ß galactosidase activity. The number of ß galactosidase-positive ECs induced by AngII was attenuated by treatment with a senolytic drug ABT737 or the chemical chaperone 4-phenylbutyrate. Monocyte adhesion assay revealed that the pro-inflammatory phenotype in ECs induced by AngII was alleviated by these treatments. AngII stimulation also increased mitochondrial fission in ECs, which was mitigated by mitochondrial division inhibitor-1. Pretreatment with mitochondrial division inhibitor-1 attenuated AngII-induced senescence and monocyte adhesion in ECs. These findings suggest that mitochondrial fission and endoplasmic reticulum stress have causative roles in endothelial senescence-associated inflammatory phenotype induced by AngII exposure, thus providing potential therapeutic targets in age-related cardiovascular diseases.


Subject(s)
Angiotensin II/pharmacology , Endothelial Cells/cytology , Mitochondria/metabolism , Monocytes/cytology , Animals , Biphenyl Compounds/pharmacology , Cell Adhesion/drug effects , Cells, Cultured , Cellular Senescence/drug effects , Endoplasmic Reticulum Stress/drug effects , Endothelial Cells/metabolism , Humans , Mitochondria/drug effects , Mitochondrial Dynamics/drug effects , Monocytes/drug effects , Nitrophenols/pharmacology , Phenotype , Phenylbutyrates/pharmacology , Piperazines/pharmacology , Proteostasis , Rats , Sulfonamides/pharmacology , THP-1 Cells
4.
Am J Physiol Heart Circ Physiol ; 318(5): H1162-H1175, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32216616

ABSTRACT

Nitric oxide (NO) and S-nitrosothiol (SNO) are considered cardio- and vasoprotective substances. We now understand that one mechanism in which NO/SNOs provide cardiovascular protection is through their direct inhibition of cardiac G protein-coupled receptor (GPCR) kinase 2 (GRK2) activity via S-nitrosylation of GRK2 at cysteine 340 (C340). This maintains GPCR homeostasis, including ß-adrenergic receptors, through curbing receptor GRK2-mediated desensitization. Previously, we have developed a knockin mouse (GRK2-C340S) where endogenous GRK2 is resistant to dynamic S-nitrosylation, which led to increased GRK2 desensitizing activity. This unchecked regulation of cardiac GRK2 activity resulted in significantly more myocardial damage after ischemic injury that was resistant to NO-mediated cardioprotection. Although young adult GRK2-C340S mice show no overt phenotype, we now report that as these mice age, they develop significant cardiovascular dysfunction due to the loss of SNO-mediated GRK2 regulation. This pathological phenotype is apparent as early as 12 mo of age and includes reduced cardiac function, increased cardiac perivascular fibrosis, and maladaptive cardiac hypertrophy, which are common maladies found in patients with cardiovascular disease (CVD). There are also vascular reactivity and aortic abnormalities present in these mice. Therefore, our data demonstrate that a chronic and global increase in GRK2 activity is sufficient to cause cardiovascular remodeling and dysfunction, likely due to GRK2's desensitizing effects in several tissues. Because GRK2 levels have been reported to be elevated in elderly CVD patients, GRK2-C340 mice can give insight into the aged-molecular landscape leading to CVD.NEW & NOTEWORTHY Research on G protein-coupled receptor kinase 2 (GRK2) in the setting of cardiovascular aging is largely unknown despite its strong established functions in cardiovascular physiology and pathophysiology. This study uses a mouse model of chronic GRK2 overactivity to further investigate the consequences of long-term GRK2 on cardiac function and structure. We report for the first time that chronic GRK2 overactivity was able to cause cardiac dysfunction and remodeling independent of surgical intervention, highlighting the importance of GRK activity in aged-related heart disease.


Subject(s)
Aging/physiology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Heart Diseases/etiology , Heart/physiology , Myocardium/metabolism , Nitric Oxide/metabolism , Aging/metabolism , Animals , Female , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , Heart/growth & development , Heart/physiopathology , Heart Diseases/metabolism , Homeostasis , Male , Mice , Mutation
5.
PLoS One ; 15(1): e0225207, 2020.
Article in English | MEDLINE | ID: mdl-31940349

ABSTRACT

BACKGROUND: Hypertension-related mortality has been increasing in recent years; however, limited information exists concerning rate, temporal, secular, and geographic trends in the United States. METHODS AND RESULTS: Using CDC death certificate data spanning 1999-2016, we sought to delineate trends in deaths attributable to an underlying cause of hypertension using joinpoint regression and proportion testing. From 1999-2016, the hypertension-related mortality rate increased by 36.4% with an average annual percent change (AAPC) of 1.8% for individuals ≥ 35 years of age. Interestingly, there was a notable acceleration in the AAPC of hypertension mortality between 2011 and 2016 (2.7% per year). This increase was due to a significant uptick in mortality for individuals ≥ 55 years of age with the greatest AAPC occurring in individuals 55-64 (4.5%) and 65-74 (5.1%) years of age. Increased mortality and AAPC were pervasive throughout sex, ethnicity, and White and American Indian or Alaska Native race, but not Black or African American race. From 2011-2016, there were significant increases in AAPC for hypertension-related mortality with contributing causes of atrial fibrillation, heart failure, diabetes, obesity, and vascular dementia. Elevated mortality was observed for conditions with a contributing cause of hypertension that included chronic obstructive pulmonary disease, diabetes, Alzheimer's, Parkinson's, and all types of falls. Geographically, increases in AAPCs and mortality rates were observed for 25/51 States between 2011 and 2016. CONCLUSIONS: Our results indicate hypertension-related mortality may have accelerated since 2011 for middle-aged and older Americans, which may create new challenges in care and healthcare planning.


Subject(s)
Diabetes Mellitus/mortality , Heart Failure/mortality , Hypertension/mortality , Adult , Black or African American , Age Distribution , Aged , Cause of Death , Death Certificates , Diabetes Mellitus/physiopathology , Female , Heart Failure/complications , Heart Failure/physiopathology , Humans , Hypertension/complications , Hypertension/physiopathology , Male , Middle Aged , United States/epidemiology , White People
6.
J Neuroinflammation ; 16(1): 241, 2019 Nov 28.
Article in English | MEDLINE | ID: mdl-31779628

ABSTRACT

BACKGROUND: Sepsis-associated encephalopathy (SAE), a diffuse cerebral dysfunction in the absence of direct CNS infection, is associated with increased rates of mortality and morbidity in patients with sepsis. Increased cytokine production and disruption of the blood-brain barrier (BBB) are implicated in the pathogenesis of SAE. The induction of pro-inflammatory mediators is driven, in part, by activation of NF-κΒ. Lipopolysaccharide (LPS), an endotoxin produced by gram-negative bacteria, potently activates NF-κΒ and its downstream targets, including cyclooxygenase-2 (Cox-2). Cox-2 catalyzes prostaglandin synthesis and in the brain prostaglandin, E2 is capable of inducing endothelial permeability. Depletion of polymerase δ-interacting protein 2 (Poldip2) has previously been reported to attenuate BBB disruption, possibly via regulation of NF-κΒ, in response to ischemic stroke. Here we investigated Poldip2 as a novel regulator of NF-κΒ/cyclooxygenase-2 signaling in an LPS model of SAE. METHODS: Intraperitoneal injections of LPS (18 mg/kg) were used to induce BBB disruption in Poldip2+/+ and Poldip2+/- mice. Changes in cerebral vascular permeability and the effect of meloxicam, a selective Cox-2 inhibitor, were assessed by Evans blue dye extravasation. Cerebral cortices of Poldip2+/+ and Poldip2+/- mice were further evaluated by immunoblotting and ELISA. To investigate the role of endothelial Poldip2, immunofluorescence microscopy and immunoblotting were performed to study the effect of siPoldip2 on LPS-mediated NF-κΒ subunit p65 translocation and Cox-2 induction in rat brain microvascular endothelial cells. Finally, FITC-dextran transwell assay was used to assess the effect of siPoldip2 on LPS-induced endothelial permeability. RESULTS: Heterozygous deletion of Poldip2 conferred protection against LPS-induced BBB permeability. Alterations in Poldip2+/+ BBB integrity were preceded by induction of Poldip2, p65, and Cox-2, which was not observed in Poldip2+/- mice. Consistent with these findings, prostaglandin E2 levels were significantly elevated in Poldip2+/+ cerebral cortices compared to Poldip2+/- cortices. Treatment with meloxicam attenuated LPS-induced BBB permeability in Poldip2+/+ mice, while having no significant effect in Poldip2+/- mice. Moreover, silencing of Poldip2 in vitro blocked LPS-induced p65 nuclear translocation, Cox-2 expression, and endothelial permeability. CONCLUSIONS: These data suggest Poldip2 mediates LPS-induced BBB disruption by regulating NF-κΒ subunit p65 activation and Cox-2 and prostaglandin E2 induction. Consequently, targeted inhibition of Poldip2 may provide clinical benefit in the prevention of sepsis-induced BBB disruption.


Subject(s)
Blood-Brain Barrier/metabolism , Mitochondrial Proteins/metabolism , Nuclear Proteins/metabolism , Sepsis-Associated Encephalopathy/metabolism , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/pathology , Capillary Permeability/drug effects , Capillary Permeability/physiology , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Knockout , Mitochondrial Proteins/genetics , NF-kappa B/metabolism , Nuclear Proteins/genetics , Permeability , Sepsis-Associated Encephalopathy/genetics , Sepsis-Associated Encephalopathy/pathology
8.
Am J Physiol Cell Physiol ; 316(5): C621-C631, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30726115

ABSTRACT

Polymerase-δ-interacting protein 2 (Poldip2) controls a wide variety of cellular functions and vascular pathologies. To mediate these effects, Poldip2 interacts with numerous proteins and generates reactive oxygen species via the enzyme NADPH oxidase 4 (Nox4). We have previously shown that Poldip2 can activate the Rho family GTPase RhoA, another signaling node within the cell. In this study, we aimed to better understand how Poldip2 activates Rho family GTPases and the functions of the involved proteins in vascular smooth muscle cells (VSMCs). RhoA is activated by guanine nucleotide exchange factors. Using nucleotide-free RhoA (isolated from bacteria) to pulldown active RhoGEFs, we found that the RhoGEF epithelial cell transforming sequence 2 (Ect2) is activated by Poldip2. Ect2 is a critical RhoGEF for Poldip2-mediated RhoA activation, because siRNA against Ect2 prevented Poldip2-mediated RhoA activity (measured by rhotekin pulldowns). Surprisingly, we were unable to detect a direct interaction between Poldip2 and Ect2, as they did not coimmunoprecipitate. Nox4 is not required for Poldip2-driven Ect2 activation, as Poldip2 overexpression induced Ect2 activation in Nox4 knockout VSMCs similar to wild-type cells. However, antioxidant treatment blocked Poldip2-induced Ect2 activation. This indicates a novel reactive oxygen species-driven mechanism by which Poldip2 regulates Rho family GTPases. Finally, we examined the function of these proteins in VSMCs, using siRNA against Poldip2 or Ect2 and determined that Poldip2 and Ect2 are both essential for vascular smooth muscle cell cytokinesis and proliferation.


Subject(s)
Muscle, Smooth, Vascular/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism , Animals , Cell Proliferation/physiology , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/cytology , Nuclear Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , RNA, Small Interfering/pharmacology , Rats , Rats, Sprague-Dawley
9.
Clin Sci (Lond) ; 133(2): 321-334, 2019 01 31.
Article in English | MEDLINE | ID: mdl-30622219

ABSTRACT

Acute respiratory distress syndrome (ARDS) in a deadly disease that can be brought on by endotoxins such as lipopolysaccharide (LPS). ARDS is characterized by vascular permeability, a severe inflammatory response, lung leukocyte infiltration, and resultant lung edema. Polymerase δ-interacting protein 2 (Poldip2) is a novel regulator of blood-brain barrier permeability; however, its role in regulating lung permeability and vascular inflammation is unknown. Here, the role of Poldip2 in regulating vascular permeability and inflammation in a mouse model of ARDS was assessed. Heterozygous deletion of Poldip2 was found to reduce LPS-induced mortality within 20 h, lung inflammatory signaling, and leukocyte infiltration. Moreover, reduced Poldip2-suppressed LP-induced vascular cell adhesion molecule (VCAM)-1 induction, leukocyte recruitment, and mitochondrial reactive oxygen species (ROS) production in vitro These data indicate that Poldip2 is an important regulator of the debilitating consequences of ARDS, potentially through the regulation of mitochondrial ROS-induced inflammatory signaling. Consequently, inhibition of Poldip2 may be a viable option for therapeutic discovery moving forward.


Subject(s)
Capillary Permeability , Endothelial Cells/metabolism , Lung/blood supply , Mitochondrial Proteins/deficiency , Nuclear Proteins/deficiency , Pulmonary Edema/prevention & control , Respiratory Distress Syndrome/metabolism , Vasculitis/prevention & control , Animals , Cell Adhesion , Coculture Techniques , Cytokines/metabolism , Disease Models, Animal , Endothelial Cells/pathology , Female , Humans , Leukocytes/metabolism , Leukocytes/pathology , Male , Mice, Inbred C57BL , Mitochondrial Proteins/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Pulmonary Edema/genetics , Pulmonary Edema/metabolism , Pulmonary Edema/pathology , Reactive Oxygen Species/metabolism , Respiratory Distress Syndrome/genetics , Respiratory Distress Syndrome/pathology , Signal Transduction , THP-1 Cells , Vascular Cell Adhesion Molecule-1/metabolism , Vasculitis/genetics , Vasculitis/metabolism , Vasculitis/pathology
10.
Physiol Rev ; 98(3): 1627-1738, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29873596

ABSTRACT

The renin-angiotensin-aldosterone system plays crucial roles in cardiovascular physiology and pathophysiology. However, many of the signaling mechanisms have been unclear. The angiotensin II (ANG II) type 1 receptor (AT1R) is believed to mediate most functions of ANG II in the system. AT1R utilizes various signal transduction cascades causing hypertension, cardiovascular remodeling, and end organ damage. Moreover, functional cross-talk between AT1R signaling pathways and other signaling pathways have been recognized. Accumulating evidence reveals the complexity of ANG II signal transduction in pathophysiology of the vasculature, heart, kidney, and brain, as well as several pathophysiological features, including inflammation, metabolic dysfunction, and aging. In this review, we provide a comprehensive update of the ANG II receptor signaling events and their functional significances for potential translation into therapeutic strategies. AT1R remains central to the system in mediating physiological and pathophysiological functions of ANG II, and participation of specific signaling pathways becomes much clearer. There are still certain limitations and many controversies, and several noteworthy new concepts require further support. However, it is expected that rigorous translational research of the ANG II signaling pathways including those in large animals and humans will contribute to establishing effective new therapies against various diseases.


Subject(s)
Angiotensin II/metabolism , Receptors, Angiotensin/metabolism , Signal Transduction , Adipocytes/metabolism , Animals , Blood Vessels/metabolism , Brain/metabolism , Heart Diseases/metabolism , Humans , Inflammation/metabolism , Kidney/metabolism , Kidney Diseases/metabolism
11.
Circ Res ; 122(6): 877-902, 2018 03 16.
Article in English | MEDLINE | ID: mdl-29700084

ABSTRACT

Reactive oxygen species (ROS) are well known for their role in mediating both physiological and pathophysiological signal transduction. Enzymes and subcellular compartments that typically produce ROS are associated with metabolic regulation, and diseases associated with metabolic dysfunction may be influenced by changes in redox balance. In this review, we summarize the current literature surrounding ROS and their role in metabolic and inflammatory regulation, focusing on ROS signal transduction and its relationship to disease progression. In particular, we examine ROS production in compartments such as the cytoplasm, mitochondria, peroxisome, and endoplasmic reticulum and discuss how ROS influence metabolic processes such as proteasome function, autophagy, and general inflammatory signaling. We also summarize and highlight the role of ROS in the regulation metabolic/inflammatory diseases including atherosclerosis, diabetes mellitus, and stroke. In order to develop therapies that target oxidative signaling, it is vital to understand the balance ROS signaling plays in both physiology and pathophysiology, and how manipulation of this balance and the identity of the ROS may influence cellular and tissue homeostasis. An increased understanding of specific sources of ROS production and an appreciation for how ROS influence cellular metabolism may help guide us in the effort to treat cardiovascular diseases.


Subject(s)
Cardiovascular Diseases/metabolism , Metabolic Diseases/metabolism , Reactive Oxygen Species/metabolism , Animals , Humans , Signal Transduction
13.
Hypertension ; 70(5): 959-963, 2017 11.
Article in English | MEDLINE | ID: mdl-28947615

ABSTRACT

Angiotensin II (AngII)-activated epidermal growth factor receptor has been implicated in abdominal aortic aneurysm (AAA) development. In vascular smooth muscle cells (VSMCs), AngII activates epidermal growth factor receptor via a metalloproteinase, ADAM17 (a disintegrin and metalloproteinase domain 17). We hypothesized that AngII-dependent AAA development would be prevented in mice lacking ADAM17 in VSMCs. To test this concept, control and VSMC ADAM17-deficient mice were cotreated with AngII and a lysyl oxidase inhibitor, ß-aminopropionitrile, to induce AAA. We found that 52.4% of control mice did not survive because of aortic rupture. All other surviving control mice developed AAA and demonstrated enhanced expression of ADAM17 in the AAA lesions. In contrast, all AngII and ß-aminopropionitrile-treated VSMC ADAM17-deficient mice survived and showed reduction in external/internal diameters (51%/28%, respectively). VSMC ADAM17 deficiency was associated with lack of epidermal growth factor receptor activation, interleukin-6 induction, endoplasmic reticulum/oxidative stress, and matrix deposition in the abdominal aorta of treated mice. However, both VSMC ADAM17-deficient and control mice treated with AngII and ß-aminopropionitrile developed comparable levels of hypertension. Treatment of C57Bl/6 mice with an ADAM17 inhibitory antibody but not with control IgG also prevented AAA development. In conclusion, VSMC ADAM17 silencing or systemic ADAM17 inhibition seems to protect mice from AAA formation. The mechanism seems to involve suppression of epidermal growth factor receptor activation.


Subject(s)
ADAM17 Protein , Aminopropionitrile/metabolism , Angiotensin II/metabolism , Aortic Aneurysm, Abdominal , Hypertension , Muscle, Smooth, Vascular , ADAM17 Protein/antagonists & inhibitors , ADAM17 Protein/metabolism , Animals , Aorta, Abdominal/metabolism , Aorta, Abdominal/pathology , Aortic Aneurysm, Abdominal/etiology , Aortic Aneurysm, Abdominal/metabolism , Aortic Aneurysm, Abdominal/pathology , Aortic Aneurysm, Abdominal/prevention & control , ErbB Receptors/metabolism , Hypertension/etiology , Hypertension/metabolism , Hypertension/prevention & control , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Protein-Lysine 6-Oxidase/metabolism , Receptor Activity-Modifying Proteins/metabolism , Signal Transduction/physiology
14.
Pharmacol Res ; 125(Pt A): 4-13, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28527699

ABSTRACT

The importance of the renin angiotensin aldosterone system in cardiovascular physiology and pathophysiology has been well described whereas the detailed molecular mechanisms remain elusive. The angiotensin II type 1 receptor (AT1 receptor) is one of the key players in the renin angiotensin aldosterone system. The AT1 receptor promotes various intracellular signaling pathways resulting in hypertension, endothelial dysfunction, vascular remodeling and end organ damage. Accumulating evidence shows the complex picture of AT1 receptor-mediated signaling; AT1 receptor-mediated heterotrimeric G protein-dependent signaling, transactivation of growth factor receptors, NADPH oxidase and ROS signaling, G protein-independent signaling, including the ß-arrestin signals and interaction with several AT1 receptor interacting proteins. In addition, there is functional cross-talk between the AT1 receptor signaling pathway and other signaling pathways. In this review, we will summarize an up to date overview of essential AT1 receptor signaling events and their functional significances in the cardiovascular system.


Subject(s)
Cardiovascular System/metabolism , Receptor, Angiotensin, Type 1/metabolism , Signal Transduction/physiology , Animals , GTP-Binding Proteins/metabolism , Humans , Renin-Angiotensin System/physiology , beta-Arrestins/metabolism
15.
Hypertension ; 69(1): 79-86, 2017 01.
Article in English | MEDLINE | ID: mdl-27895190

ABSTRACT

It has been proposed that membrane microdomains, caveolae, in vascular cells are critical for signal transduction and downstream functions induced by angiotensin II (AngII). We have tested our hypothesis that caveolin-1 (Cav1), a major structural protein of vascular caveolae, plays a critical role in the development of vascular remodeling by AngII via regulation of epidermal growth factor receptor and vascular endothelial adhesion molecule-1. Cav1-/- and control Cav+/+ mice were infused with AngII for 2 weeks to induce vascular remodeling and hypertension. On AngII infusion, histological assessments demonstrated medial hypertrophy and perivascular fibrosis of aorta and coronary and renal arteries in Cav1+/+ mice compared with sham-operated Cav1+/+ mice. AngII-infused Cav1+/+ mice also showed a phenotype of cardiac hypertrophy with increased heart weight to body weight ratio compared with control Cav1+/+ mice. In contrast, Cav1-/- mice infused with AngII showed attenuation of vascular remodeling but not cardiac hypertrophy. Similar levels of AngII-induced hypertension were found in both Cav1+/+ and Cav1-/- mice as assessed by telemetry. In Cav1+/+ mice, AngII enhanced tyrosine-phosphorylated epidermal growth factor receptor staining in the aorta, which was attenuated in Cav1-/- mice infused with AngII. Enhanced Cav1 and vascular endothelial adhesion molecule-1 expression was also observed in aorta from AngII-infused Cav1+/+ mice but not in Cav1-/- aorta. Experiments with vascular cells further provided a potential mechanism for our in vivo findings. These data suggest that Cav1, and presumably caveolae, in vascular smooth muscle and the endothelium plays a critical role in vascular remodeling and inflammation independent of blood pressure or cardiac hypertrophy regulation.


Subject(s)
Angiotensin II/pharmacology , Caveolin 1/genetics , Gene Deletion , Hypertension/genetics , Vascular Remodeling/genetics , Animals , Blood Pressure/physiology , Caveolin 1/metabolism , Disease Models, Animal , Hypertension/metabolism , Hypertension/pathology , Male , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Rats , Rats, Sprague-Dawley , Signal Transduction , Vascular Remodeling/drug effects
16.
Hypertension ; 68(4): 949-955, 2016 10.
Article in English | MEDLINE | ID: mdl-27480833

ABSTRACT

Angiotensin II (AngII) has been strongly implicated in hypertension and its complications. Evidence suggests the mechanisms by which AngII elevates blood pressure and enhances cardiovascular remodeling and damage may be distinct. However, the signal transduction cascade by which AngII specifically initiates cardiovascular remodeling, such as hypertrophy and fibrosis, remains insufficiently understood. In vascular smooth muscle cells, a metalloproteinase ADAM17 mediates epidermal growth factor receptor transactivation, which may be responsible for cardiovascular remodeling but not hypertension induced by AngII. Thus, the objective of this study was to test the hypothesis that activation of vascular ADAM17 is indispensable for vascular remodeling but not for hypertension induced by AngII. Vascular ADAM17-deficient mice and control mice were infused with AngII for 2 weeks. Control mice infused with AngII showed cardiac hypertrophy, vascular medial hypertrophy, and perivascular fibrosis. These phenotypes were prevented in vascular ADAM17-deficient mice independent of blood pressure alteration. AngII infusion enhanced ADAM17 expression, epidermal growth factor receptor activation, and endoplasmic reticulum stress in the vasculature, which were diminished in ADAM17-deficient mice. Treatment with a human cross-reactive ADAM17 inhibitory antibody also prevented cardiovascular remodeling and endoplasmic reticulum stress but not hypertension in C57Bl/6 mice infused with AngII. In vitro data further supported these findings. In conclusion, vascular ADAM17 mediates AngII-induced cardiovascular remodeling via epidermal growth factor receptor activation independent of blood pressure regulation. ADAM17 seems to be a unique therapeutic target for the prevention of hypertensive complications.


Subject(s)
ADAM17 Protein/drug effects , ADAM17 Protein/metabolism , Angiotensin II/pharmacology , Cardiomegaly/metabolism , ErbB Receptors/metabolism , Hypertension/complications , Animals , Cardiomegaly/prevention & control , Cells, Cultured , Disease Models, Animal , Fibrosis/metabolism , Fibrosis/pathology , Humans , Hypertension/chemically induced , Hypertension/physiopathology , Male , Mice , Mice, Inbred C57BL , Molecular Targeted Therapy , Myocytes, Cardiac/metabolism , Random Allocation , Renin-Angiotensin System/physiology , Sensitivity and Specificity , Signal Transduction/drug effects , Vascular Remodeling/drug effects , Vascular Remodeling/physiology , Ventricular Remodeling/drug effects , Ventricular Remodeling/physiology
17.
Annu Rev Pharmacol Toxicol ; 56: 627-53, 2016.
Article in English | MEDLINE | ID: mdl-26566153

ABSTRACT

Epidermal growth factor receptor (EGFR) activation impacts the physiology and pathophysiology of the cardiovascular system, and inhibition of EGFR activity is emerging as a potential therapeutic strategy to treat diseases including hypertension, cardiac hypertrophy, renal fibrosis, and abdominal aortic aneurysm. The capacity of G protein-coupled receptor (GPCR) agonists, such as angiotensin II (AngII), to promote EGFR signaling is called transactivation and is well described, yet delineating the molecular processes and functional relevance of this crosstalk has been challenging. Moreover, these critical findings are dispersed among many different fields. The aim of our review is to highlight recent advancements in defining the signaling cascades and downstream consequences of EGFR transactivation in the cardiovascular renal system. We also focus on studies that link EGFR transactivation to animal models of the disease, and we discuss potential therapeutic applications.


Subject(s)
Cardiovascular System/metabolism , ErbB Receptors/metabolism , Signal Transduction/physiology , Transcriptional Activation/physiology , Animals , Humans
19.
Physiol Rev ; 96(3): 1025-1069, 2016 Jul.
Article in English | MEDLINE | ID: mdl-33003261

ABSTRACT

The epidermal growth factor receptor (EGFR) is the prototypical member of a family of membrane-associated intrinsic tyrosine kinase receptors, the ErbB family. EGFR is activated by multiple ligands, including EGF, transforming growth factor (TGF)-α, HB-EGF, betacellulin, amphiregulin, epiregulin, and epigen. EGFR is expressed in multiple organs and plays important roles in proliferation, survival, and differentiation in both development and normal physiology, as well as in pathophysiological conditions. In addition, EGFR transactivation underlies some important biologic consequences in response to many G protein-coupled receptor (GPCR) agonists. Aberrant EGFR activation is a significant factor in development and progression of multiple cancers, which has led to development of mechanism-based therapies with specific receptor antibodies and tyrosine kinase inhibitors. This review highlights the current knowledge about mechanisms and roles of EGFR in physiology and disease.

20.
Ethn Dis ; 25(4): 413-8, 2015 Nov 05.
Article in English | MEDLINE | ID: mdl-26674844

ABSTRACT

BACKGROUND: African Americans have a predisposition to heightened systemic inflammation and a high prevalence of hypertension. OBJECTIVE: The purpose of this study was to evaluate the influence of interleukin-10 (IL-10) and laminar shear stress (LSS) on African American endothelial cells by measuring total endothelial nitric oxide synthase (eNOS) protein expression and its phosphorylated form (p-eNOS) at Serine 1177, and nitric oxide (NO) levels, in response to IL-10 incubation and high physiological levels of LSS, used as an in vitro mimetic for aerobic exercise training (AEXT). DESIGN: Human umbilical vein endothelial cells (HUVEC) from an African American donor were cultured. The experimental conditions included Static, Static with IL-10 Incubation, LSS at 20 dynes/cm², and LSS at 20 dynes/cm² with IL-10 Incubation. Western blotting was used to measure eNOS and p-eNOS protein expression in the cells. A modified Griess assay was used to measure NO metabolites in the cell culture media. RESULTS: There were significant increases in p-eNOS, eNOS, and NO in the LSS at 20 dynes/cm² and LSS at 20 dynes/cm² with IL-10 Incubation experimental conditions when compared to the Static experimental condition. There were no other statistically significant differences demonstrating that IL-10 did not have an additive effect on eNOS activity in our study. CONCLUSION: The significant increases in p-eNOS, eNOS, and NO as a result of LSS in African American HUVECs suggest that AEXT may be a viable, nonpharmacologic method to improve vascular inflammation status and vasodilation, and thereby contribute to hypertension reduction in the African American population.


Subject(s)
Black or African American , Human Umbilical Vein Endothelial Cells/drug effects , Interleukin-10/pharmacology , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide/metabolism , Stress, Mechanical , Cell Culture Techniques , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Shear Strength
SELECTION OF CITATIONS
SEARCH DETAIL
...