Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 139
Filter
4.
Antioxidants (Basel) ; 11(10)2022 Sep 30.
Article in English | MEDLINE | ID: mdl-36290688

ABSTRACT

NADPH oxidases (Nox) are one of the main sources of reactive oxygen species (ROS) in the central nervous system (CNS). While these enzymes have been shown to be involved in physiological regulation of cerebral vascular tone, excessive ROS produced by Nox1-5 play a critical role in blood-brain barrier (BBB) dysfunction in numerous neuropathologies. Nox-derived ROS have been implicated in mediating matrix metalloprotease (MMP) activation, downregulation of junctional complexes between adjacent brain endothelial cells and brain endothelial cell apoptosis, leading to brain microvascular endothelial barrier dysfunction and consequently, increases in BBB permeability. In this review, we will highlight recent findings on the role played by these enzymes in BBB disruption induced by ischemic stroke.

5.
J Am Heart Assoc ; 11(10): e025181, 2022 05 17.
Article in English | MEDLINE | ID: mdl-35535614

ABSTRACT

Background Lung injury, a severe adverse outcome of lipopolysaccharide-induced acute respiratory distress syndrome, is attributed to excessive neutrophil recruitment and effector response. Poldip2 (polymerase δ-interacting protein 2) plays a critical role in regulating endothelial permeability and leukocyte recruitment in acute inflammation. Thus, we hypothesized that myeloid Poldip2 is involved in neutrophil recruitment to inflamed lungs. Methods and Results After characterizing myeloid-specific Poldip2 knockout mice, we showed that at 18 hours post-lipopolysaccharide injection, bronchoalveolar lavage from myeloid Poldip2-deficient mice contained fewer inflammatory cells (8 [4-16] versus 29 [12-57]×104/mL in wild-type mice) and a smaller percentage of neutrophils (30% [28%-34%] versus 38% [33%-41%] in wild-type mice), while the main chemoattractants for neutrophils remained unaffected. In vitro, Poldip2-deficient neutrophils responded as well as wild-type neutrophils to inflammatory stimuli with respect to neutrophil extracellular trap formation, reactive oxygen species production, and induction of cytokines. However, neutrophil adherence to a tumor necrosis factor-α stimulated endothelial monolayer was inhibited by Poldip2 depletion (225 [115-272] wild-type [myePoldip2+/+] versus 133 [62-178] myeloid-specific Poldip2 knockout [myePoldip2-/-] neutrophils) as was transmigration (1.7 [1.3-2.1] versus 1.1 [1.0-1.4] relative to baseline transmigration). To determine the underlying mechanism, we examined the surface expression of ß2-integrin, its binding to soluble intercellular adhesion molecule 1, and Pyk2 phosphorylation. Surface expression of ß2-integrins was not affected by Poldip2 deletion, whereas ß2-integrins and Pyk2 were less activated in Poldip2-deficient neutrophils. Conclusions These results suggest that myeloid Poldip2 is involved in ß2-integrin activation during the inflammatory response, which in turn mediates neutrophil-to-endothelium adhesion in lipopolysaccharide-induced acute respiratory distress syndrome.


Subject(s)
Mitochondrial Proteins , Neutrophils , Nuclear Proteins , Pneumonia , Respiratory Distress Syndrome , Animals , Cell Adhesion , Disease Models, Animal , Focal Adhesion Kinase 2/metabolism , Integrins/metabolism , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Neutrophils/metabolism , Neutrophils/pathology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Pneumonia/genetics , Pneumonia/metabolism , Pneumonia/pathology , Respiratory Distress Syndrome/genetics , Respiratory Distress Syndrome/metabolism , Respiratory Distress Syndrome/pathology
6.
Arterioscler Thromb Vasc Biol ; 42(5): e117-e130, 2022 05.
Article in English | MEDLINE | ID: mdl-35354309

ABSTRACT

BACKGROUND: Reactive oxygen species (ROS) and calcium ions (Ca2+) are among the major effectors of Ang II (angiotensin II) in vascular smooth muscle cells. ROS are related to Ca2+ signaling or contraction induced by Ang II, but little is known about their detailed functions. Here, NOX (NADPH oxidase), a major ROS source responsive to Ang II, was investigated regarding its contribution to Ca2+ signaling. METHODS: Vascular smooth muscle cells were primary cultured from rat aorta. Ca2+ and ROS were monitored mainly using fura-2 and HyPer family probes' respectively. Signals activating NOX were examined with relevant pharmacological inhibitors and genetic manipulation techniques. RESULTS: Ang II-induced ROS generation was found to be biphasic: the first phase of ROS production, which was mainly mediated by NOX1, was small and transient, preceding a rise in Ca2+, and the second phase of ROS generation, mediated by NOX1 and NOX4, was slow but sizeable, continuing over tens of minutes. NOX1-derived superoxide in the first phase is required for Ca2+ influx through nonselective cation channels. AT1R (Ang II type 1 receptor)-Gßγ-PI3Kγ (phosphoinositide 3-kinase γ) signaling pathway was responsible for the rapid activation of NOX1 in the first phase, while in the second phase, NOX1 was further activated by a separate AT1R-Gαq/11-PLC (phospholipase C)-PKCß (protein kinase C ß) signaling axis. Consistent with these observations, aortas from NOX1-knockout mice exhibited reduced contractility in response to Ang II, and thus the acute pressor response to Ang II was also attenuated in NOX1-knockout mice. CONCLUSIONS: NOX1 mediates Ca2+ signal generation and thereby contributes to vascular contraction and blood pressure elevation by Ang II.


Subject(s)
Angiotensin II , Calcium , NADPH Oxidase 1/metabolism , Angiotensin II/metabolism , Angiotensin II/pharmacology , Animals , Blood Pressure , Calcium/metabolism , Mice , Muscle, Smooth, Vascular/metabolism , NADH, NADPH Oxidoreductases/genetics , NADH, NADPH Oxidoreductases/metabolism , NADPH Oxidase 4/metabolism , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Rats , Reactive Oxygen Species/metabolism
7.
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
8.
PLoS One ; 16(12): e0247261, 2021.
Article in English | MEDLINE | ID: mdl-34928942

ABSTRACT

POLDIP2 is a multifunctional protein whose roles are only partially understood. Our laboratory previously reported physiological studies performed using a mouse gene trap model, which suffered from three limitations: perinatal lethality in homozygotes, constitutive Poldip2 inactivation and inadvertent downregulation of the adjacent Tmem199 gene. To overcome these limitations, we developed a new conditional floxed Poldip2 model. The first part of the present study shows that our initial floxed mice were affected by an unexpected mutation, which was not readily detected by Southern blotting and traditional PCR. It consisted of a 305 kb duplication around Poldip2 with retention of the wild type allele and could be traced back to the original targeted ES cell clone. We offer simple suggestions to rapidly detect similar accidents, which may affect genome editing using both traditional and CRISPR-based methods. In the second part of the present study, correctly targeted floxed Poldip2 mice were generated and used to produce a new constitutive knockout line by crossing with a Cre deleter. In contrast to the gene trap model, many homozygous knockout mice were viable, in spite of having no POLDIP2 expression. To further characterize the effects of Poldip2 ablation in the vasculature, RNA-seq and RT-qPCR experiments were performed in constitutive knockout arteries. Results show that POLDIP2 inactivation affects multiple cellular processes and provide new opportunities for future in-depth study of its functions.


Subject(s)
CRISPR-Cas Systems , Gene Targeting , Membrane Proteins/genetics , Mitochondrial Proteins/deficiency , Mouse Embryonic Stem Cells/metabolism , Nuclear Proteins/deficiency , RNA-Seq , Animals , Membrane Proteins/metabolism , Mice , Mice, Knockout , Mitochondrial Proteins/metabolism , Nuclear Proteins/metabolism
9.
Circ Res ; 128(7): 993-1020, 2021 04 02.
Article in English | MEDLINE | ID: mdl-33793335

ABSTRACT

A link between oxidative stress and hypertension has been firmly established in multiple animal models of hypertension but remains elusive in humans. While initial studies focused on inactivation of nitric oxide by superoxide, our understanding of relevant reactive oxygen species (superoxide, hydrogen peroxide, and peroxynitrite) and how they modify complex signaling pathways to promote hypertension has expanded significantly. In this review, we summarize recent advances in delineating the primary and secondary sources of reactive oxygen species (nicotinamide adenine dinucleotide phosphate oxidases, uncoupled endothelial nitric oxide synthase, endoplasmic reticulum, and mitochondria), the posttranslational oxidative modifications they induce on protein targets important for redox signaling, their interplay with endogenous antioxidant systems, and the role of inflammasome activation and endoplasmic reticular stress in the development of hypertension. We highlight how oxidative stress in different organ systems contributes to hypertension, describe new animal models that have clarified the importance of specific proteins, and discuss clinical studies that shed light on how these processes and pathways are altered in human hypertension. Finally, we focus on the promise of redox proteomics and systems biology to help us fully understand the relationship between ROS and hypertension and their potential for designing and evaluating novel antihypertensive therapies.


Subject(s)
Hypertension/etiology , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism , Animals , Antioxidants/metabolism , Disease Models, Animal , Endoplasmic Reticulum/metabolism , Humans , Hypertension/metabolism , Inflammasomes/physiology , Kidney/metabolism , Mitochondria/metabolism , NADPH Oxidases/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase/metabolism , Oxidation-Reduction , Signal Transduction/physiology , Superoxides/metabolism , Vascular Diseases/metabolism
10.
Sci Rep ; 11(1): 5533, 2021 03 10.
Article in English | MEDLINE | ID: mdl-33692398

ABSTRACT

Stroke is a multiphasic process involving a direct ischemic brain injury which is then exacerbated by the influx of immune cells into the brain tissue. Activation of brain endothelial cells leads to the expression of adhesion molecules such vascular cell adhesion molecule 1 (VCAM-1) on endothelial cells, further increasing leukocyte recruitment. Polymerase δ-interacting protein 2 (Poldip2) promotes brain vascular inflammation and leukocyte recruitment via unknown mechanisms. This study aimed to define the role of Poldip2 in mediating vascular inflammation and leukocyte recruitment following cerebral ischemia. Cerebral ischemia was induced in Poldip2+/+ and Poldip2+/- mice and brains were isolated and processed for flow cytometry or RT-PCR. Cultured rat brain microvascular endothelial cells were used to investigate the effect of Poldip2 depletion on focal adhesion kinase (FAK)-mediated VCAM-1 induction. Poldip2 depletion in vivo attenuated the infiltration of myeloid cells, inflammatory monocytes/macrophages and decreased the induction of adhesion molecules. Focusing on VCAM-1, we demonstrated mechanistically that FAK activation was a critical intermediary in Poldip2-mediated VCAM-1 induction. In conclusion, Poldip2 is an important mediator of endothelial dysfunction and leukocyte recruitment. Thus, Poldip2 could be a therapeutic target to improve morbidity following ischemic stroke.


Subject(s)
Brain Ischemia/metabolism , Brain/metabolism , Focal Adhesion Kinase 1/metabolism , Ischemic Stroke/metabolism , Leukocytes/metabolism , Mitochondrial Proteins/metabolism , Nuclear Proteins/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Animals , Brain Ischemia/genetics , Focal Adhesion Kinase 1/genetics , Ischemic Stroke/genetics , Mice , Mice, Mutant Strains , Mitochondrial Proteins/genetics , Nuclear Proteins/genetics , Vascular Cell Adhesion Molecule-1/genetics
11.
Am J Physiol Heart Circ Physiol ; 320(2): H734-H739, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33337960

ABSTRACT

The integrin family, an indispensable part of cell-cell and cell-matrix interactions, consists of a group of heterodimeric adhesion receptors formed by α- and ß-integrin subunits. Their wide expression and unique bidirectional signaling pathways allow them to play roles in a variety of biological activities including blood clot formation, cell attachment, and migration. Evidence suggests that integrins are essential regulators of the initiation of acute inflammation, especially two key aspects of this process i.e., vascular permeability and leukocyte recruitment. This mini-review discusses the importance of integrins at the onset of the acute inflammatory response and outlines research advances regarding the function of integrins and their modulators at different stages of this process. Insights into the fine-tuning of integrin signaling during acute inflammation may inspire the design of new drugs for inflammatory diseases.


Subject(s)
CD18 Antigens/metabolism , Capillary Permeability , Chemotaxis, Leukocyte , Endothelium, Vascular/metabolism , Inflammation Mediators/metabolism , Inflammation/metabolism , Integrin beta1/metabolism , Leukocytes/metabolism , Animals , Cell Adhesion , Cell Communication , Endothelium, Vascular/immunology , Endothelium, Vascular/physiopathology , Humans , Inflammation/immunology , Inflammation/physiopathology , Leukocyte Rolling , Leukocytes/immunology , Signal Transduction , Transendothelial and Transepithelial Migration
12.
Cardiovasc Res ; 117(1): 60-73, 2021 01 01.
Article in English | MEDLINE | ID: mdl-32215570

ABSTRACT

ABSTRACT: Sepsis accounts for nearly 700 000 deaths in Europe annually and is caused by an overwhelming host response to infection resulting in organ failure. The endothelium is an active contributor to sepsis and as such represents a major target for therapy. During sepsis, endothelial cells amplify the immune response and activate the coagulation system. They are both a target and source of inflammation and serve as a link between local and systemic immune responses. In response to cytokines produced by immune cells, the endothelium expresses adhesion molecules and produces vasoactive compounds, inflammatory cytokines, and chemoattractants, thus switching from an anticoagulant to procoagulant state. These responses contribute to local control of infection, but systemic activation can lead to microvascular thrombosis, capillary permeability, hypotension, tissue hypoxia, and ultimately tissue damage. This review focuses on the role of the endothelium in leucocyte adhesion and transmigration as well as production of reactive oxygen and nitrogen species, microRNAs and cytokines, formation of signalling microparticles, and disseminated intravascular coagulation. We also discuss alterations in endothelial permeability and apoptosis. Finally, we review the diagnostic potential of endothelial markers and endothelial pathways as therapeutic targets for this devastating disease.


Subject(s)
Blood Coagulation , Cardiovascular Diseases/metabolism , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Inflammation Mediators/metabolism , Inflammation/metabolism , Sepsis/metabolism , Animals , Anti-Inflammatory Agents/therapeutic use , Anticoagulants/therapeutic use , Biomarkers/metabolism , Blood Coagulation/drug effects , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/immunology , Cardiovascular Diseases/pathology , Endothelial Cells/drug effects , Endothelial Cells/immunology , Endothelial Cells/pathology , Endothelium, Vascular/drug effects , Endothelium, Vascular/immunology , Endothelium, Vascular/pathology , Humans , Inflammation/drug therapy , Inflammation/immunology , Inflammation/pathology , Sepsis/drug therapy , Sepsis/immunology , Sepsis/pathology , Signal Transduction
14.
J Cell Biol ; 219(5)2020 05 04.
Article in English | MEDLINE | ID: mdl-32232465

ABSTRACT

Tissue morphogenesis requires dynamic intercellular contacts that are subsequently stabilized as tissues mature. The mechanisms governing these competing adhesive properties are not fully understood. Using gain- and loss-of-function approaches, we tested the role of p120-catenin (p120) and VE-cadherin (VE-cad) endocytosis in vascular development using mouse mutants that exhibit increased (VE-cadGGG/GGG) or decreased (VE-cadDEE/DEE) internalization. VE-cadGGG/GGG mutant mice exhibited reduced VE-cad-p120 binding, reduced VE-cad levels, microvascular hemorrhaging, and decreased survival. By contrast, VE-cadDEE/DEE mutants exhibited normal vascular permeability but displayed microvascular patterning defects. Interestingly, VE-cadDEE/DEE mutant mice did not require endothelial p120, demonstrating that p120 is dispensable in the context of a stabilized cadherin. In vitro, VE-cadDEE mutant cells displayed defects in polarization and cell migration that were rescued by uncoupling VE-cadDEE from actin. These results indicate that cadherin endocytosis coordinates cell polarity and migration cues through actin remodeling. Collectively, our results indicate that regulated cadherin endocytosis is essential for both dynamic cell movements and establishment of stable tissue architecture.


Subject(s)
Antigens, CD/genetics , Blood Vessels/growth & development , Cadherins/genetics , Catenins/genetics , Embryonic Development/genetics , Endothelium, Vascular/growth & development , Actins/genetics , Animals , Aorta/growth & development , Aorta/metabolism , Blood Vessels/metabolism , Body Patterning/genetics , Cell Movement/genetics , Cell Polarity/genetics , Embryo, Mammalian , Endocytosis/genetics , Endothelium, Vascular/metabolism , Mice , Protein Binding/genetics , Delta Catenin
15.
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
16.
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
18.
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
19.
J Clin Invest ; 129(3): 962-964, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30776027

ABSTRACT

Vascular smooth muscle cell (VSMC) phenotype switching from a contractile state to a synthetic phenotype has been implicated in intimal remodeling during vascular injury. While multiple studies have focused on dedifferentiation of VSMCs, prevention of VSMC-mediated excessive repair remains poorly understood. In this issue of the JCI, Zeng et al. identified a mechanism by which platelet-derived microRNA-223 (miRNA-223) reverses VSMC dedifferentiation. The authors show that suppression of proliferation occurs after platelet internalization by VSMCs. Moreover, they demonstrate that miRNA-223 inhibits dedifferentiation and intimal hyperplasia in diabetic mice by decreasing PDGFRß expression in VSMCs. Together, these results identify platelet-derived miRNA-223 as a potential therapeutic target in vascular injury.


Subject(s)
Diabetes Mellitus, Experimental , MicroRNAs , Vascular System Injuries , Animals , Cell Proliferation , Cells, Cultured , Mice , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Phenotype
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...