57 results on '"Kolluru GK"'
Search Results
2. A Potential Role for MAGI-1 in the Bi-Directional Relationship Between Major Depressive Disorder and Cardiovascular Disease.
- Author
-
Banerjee P, Chau K, Kotla S, Davis EL, Turcios EB, Li S, Pengzhi Z, Wang G, Kolluru GK, Jain A, Cooke JP, Abe J, and Le NT
- Subjects
- Humans, Animals, Cell Adhesion Molecules metabolism, Cell Adhesion Molecules genetics, Polymorphism, Single Nucleotide, Depressive Disorder, Major metabolism, Depressive Disorder, Major genetics, Cardiovascular Diseases genetics, Cardiovascular Diseases metabolism, Adaptor Proteins, Signal Transducing genetics, Adaptor Proteins, Signal Transducing metabolism, Guanylate Kinases genetics, Guanylate Kinases metabolism
- Abstract
Purpose of Review: Major Depressive Disorder (MDD) is characterized by persistent symptoms such as fatigue, loss of interest in activities, feelings of sadness and worthlessness. MDD often coexist with cardiovascular disease (CVD), yet the precise link between these conditions remains unclear. This review explores factors underlying the development of MDD and CVD, including genetic, epigenetic, platelet activation, inflammation, hypothalamic-pituitary-adrenal (HPA) axis activation, endothelial cell (EC) dysfunction, and blood-brain barrier (BBB) disruption., Recent Findings: Single nucleotide polymorphisms (SNPs) in the membrane-associated guanylate kinase WW and PDZ domain-containing protein 1 (MAGI-1) are associated with neuroticism and psychiatric disorders including MDD. SNPs in MAGI-1 are also linked to chronic inflammatory disorders such as spontaneous glomerulosclerosis, celiac disease, ulcerative colitis, and Crohn's disease. Increased MAGI-1 expression has been observed in colonic epithelial samples from Crohn's disease and ulcerative colitis patients. MAGI-1 also plays a role in regulating EC activation and atherogenesis in mice and is essential for Influenza A virus (IAV) infection, endoplasmic reticulum stress-induced EC apoptosis, and thrombin-induced EC permeability. Despite being understudied in human disease; evidence suggests that MAGI-1 may play a role in linking CVD and MDD. Therefore, further investigation of MAG-1 could be warranted to elucidate its potential involvement in these conditions., (© 2024. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2024
- Full Text
- View/download PDF
3. Deletion of Sigmar1 leads to increased arterial stiffness and altered mitochondrial respiration resulting in vascular dysfunction.
- Author
-
Remex NS, Abdullah CS, Aishwarya R, Kolluru GK, Traylor J, Bhuiyan MAN, Kevil CG, Orr AW, Rom O, Pattillo CB, and Bhuiyan MS
- Abstract
Sigmar1 is a ubiquitously expressed, multifunctional protein known for its cardioprotective roles in cardiovascular diseases. While accumulating evidence indicate a critical role of Sigmar1 in cardiac biology, its physiological function in the vasculature remains unknown. In this study, we characterized the expression of Sigmar1 in the vascular wall and assessed its physiological function in the vascular system using global Sigmar1 knockout (Sigmar1
-/- ) mice. We determined the expression of Sigmar1 in the vascular tissue using immunostaining and biochemical experiments in both human and mouse blood vessels. Deletion of Sigmar1 globally in mice (Sigmar1-/- ) led to blood vessel wall reorganizations characterized by nuclei disarray of vascular smooth muscle cells, altered organizations of elastic lamina, and higher collagen fibers deposition in and around the arteries compared to wildtype littermate controls (Wt). Vascular function was assessed in mice using non-invasive time-transit method of aortic stiffness measurement and flow-mediated dilation (FMD) of the left femoral artery. Sigmar1-/- mice showed a notable increase in arterial stiffness in the abdominal aorta and failed to increase the vessel diameter in response to reactive-hyperemia compared to Wt. This was consistent with reduced plasma and tissue nitric-oxide bioavailability (NOx) and decreased phosphorylation of endothelial nitric oxide synthase (eNOS) in the aorta of Sigmar1-/- mice. Ultrastructural analysis by transmission electron microscopy (TEM) of aorta sections showed accumulation of elongated shaped mitochondria in both vascular smooth muscle and endothelial cells of Sigmar1-/- mice. In accordance, decreased mitochondrial respirometry parameters were found in ex-vivo aortic rings from Sigmar1 deficient mice compared to Wt controls. These data indicate a potential role of Sigmar1 in maintaining vascular homeostasis., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. The author(s) declared that they were an editorial board member of Frontiers, at the time of submission. This had no impact on the peer review process and the final decision., (Copyright © 2024 Remex, Abdullah, Aishwarya, Kolluru, Traylor, Bhuiyan, Kevil, Orr, Rom, Pattillo and Bhuiyan.)- Published
- 2024
- Full Text
- View/download PDF
4. Neurogranin expression regulates mitochondrial function and redox balance in endothelial cells.
- Author
-
Jorgensen AN, Rashdan NA, Rao KNS, Delgadillo LF, Kolluru GK, Krzywanski DM, Pattillo CB, Kevil CG, and Nam HW
- Subjects
- Animals, Humans, Mice, Hydrogen Peroxide metabolism, Mitochondrial Diseases metabolism, Nitric Oxide metabolism, Oxidation-Reduction, Reactive Oxygen Species metabolism, Endothelial Cells metabolism, Mitochondria genetics, Mitochondria metabolism, Neurogranin metabolism
- Abstract
Endothelial dysfunction and endothelial activation are common early events in vascular diseases and can arise from mitochondrial dysfunction. Neurogranin (Ng) is a 17kD protein well known to regulate intracellular Ca
2+ -calmodulin (CaM) complex signaling, and its dysfunction is significantly implicated in brain aging and neurodegenerative diseases. We found that Ng is also expressed in human aortic endothelial cells (HAECs), and depleting Ng promotes Ca2+ -CaM complex-dependent endothelial activation and redox imbalances. Endothelial-specific Ng knockout (Cre-CDH5-Ngf/f ) mice demonstrate a significant delay in the flow-mediated dilation (FMD) response. Therefore, it is critical to characterize how endothelial Ng expression regulates reactive oxygen species (ROS) generation and affects cardiovascular disease. Label-free quantification proteomics identified that mitochondrial dysfunction and the oxidative phosphorylation pathway are significantly changed in the aorta of Cre-CDH5-Ngf/f mice. We found that a significant amount of Ng is expressed in the mitochondrial fraction of HAECs using western blotting and colocalized with the mitochondrial marker, COX IV, using immunofluorescence staining. Seahorse assay demonstrated that a lack of Ng decreases mitochondrial respiration. Treatment with MitoEbselen significantly restores the oxygen consumption rate in Ng knockdown cells. With the RoGFP-Orp1 approach, we identified that Ng knockdown increases mitochondrial-specific hydrogen peroxide (H2 O2 ) production, and MitoEbselen treatment significantly reduced mitochondrial ROS (mtROS) levels in Ng knockdown cells. These results suggest that Ng plays a significant role in mtROS production. We discovered that MitoEbselen treatment also rescues decreased eNOS expression and nitric oxide (NO) levels in Ng knockdown cells, which implicates the critical role of Ng in mtROS-NO balance in the endothelial cells., Competing Interests: Declaration of competing interest Declarations of interest: none., (Copyright © 2024 The Authors. Published by Elsevier B.V. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
5. Hypoxia increases persulfide and polysulfide formation by AMP kinase dependent cystathionine gamma lyase phosphorylation.
- Author
-
Alam S, Pardue S, Shen X, Glawe JD, Yagi T, Bhuiyan MAN, Patel RP, Dominic PS, Virk CS, Bhuiyan MS, Orr AW, Petit C, Kolluru GK, and Kevil CG
- Subjects
- Humans, Phosphorylation, Adenylate Kinase metabolism, Cystathionine gamma-Lyase genetics, Hypoxia, Hydrogen Sulfide metabolism
- Abstract
Hydropersulfide and hydropolysulfide metabolites are increasingly important reactive sulfur species (RSS) regulating numerous cellular redox dependent functions. Intracellular production of these species is known to occur through RSS interactions or through translational mechanisms involving cysteinyl t-RNA synthetases. However, regulation of these species under cell stress conditions, such as hypoxia, that are known to modulate RSS remain poorly understood. Here we define an important mechanism of increased persulfide and polysulfide production involving cystathionine gamma lyase (CSE) phosphorylation at serine 346 and threonine 355 in a substrate specific manner, under acute hypoxic conditions. Hypoxic phosphorylation of CSE occurs in an AMP kinase dependent manner increasing enzyme activity involving unique inter- and intramolecular interactions within the tetramer. Importantly, both cellular hypoxia and tissue ischemia result in AMP Kinase dependent CSE phosphorylation that regulates blood flow in ischemic tissues. Our findings reveal hypoxia molecular signaling pathways regulating CSE dependent persulfide and polysulfide production impacting tissue and cellular response to stress., Competing Interests: Declaration of competing interest C.G.K., G.K·K., S.A., and X.S. have a provisional patent on CSE phosphorylation mutants and uses., (Copyright © 2023 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
6. Sulfide regulation of cardiovascular function in health and disease.
- Author
-
Kolluru GK, Shackelford RE, Shen X, Dominic P, and Kevil CG
- Subjects
- Humans, Sulfides, Heart, Hydrogen Sulfide metabolism, Myocardial Infarction, Heart Failure
- Abstract
Hydrogen sulfide (H
2 S) has emerged as a gaseous signalling molecule with crucial implications for cardiovascular health. H2 S is involved in many biological functions, including interactions with nitric oxide, activation of molecular signalling cascades, post-translational modifications and redox regulation. Various preclinical and clinical studies have shown that H2 S and its synthesizing enzymes - cystathionine γ-lyase, cystathionine β-synthase and 3-mercaptosulfotransferase - can protect against cardiovascular pathologies, including arrhythmias, atherosclerosis, heart failure, myocardial infarction and ischaemia-reperfusion injury. The bioavailability of H2 S and its metabolites, such as hydropersulfides and polysulfides, is substantially reduced in cardiovascular disease and has been associated with single-nucleotide polymorphisms in H2 S synthesis enzymes. In this Review, we highlight the role of H2 S, its synthesizing enzymes and metabolites, their roles in the cardiovascular system, and their involvement in cardiovascular disease and associated pathologies. We also discuss the latest clinical findings from the field and outline areas for future study., (© 2022. Springer Nature Limited.)- Published
- 2023
- Full Text
- View/download PDF
7. Mitochondrial dysfunction and autophagy activation are associated with cardiomyopathy developed by extended methamphetamine self-administration in rats.
- Author
-
Abdullah CS, Remex NS, Aishwarya R, Nitu S, Kolluru GK, Traylor J, Hartman B, King J, Bhuiyan MAN, Hall N, Murnane KS, Goeders NE, Kevil CG, Orr AW, and Bhuiyan MS
- Subjects
- Humans, Rats, Animals, Autophagy, Mitochondria, Methamphetamine toxicity, Central Nervous System Stimulants pharmacology, Cardiomyopathies
- Abstract
The recent rise in illicit use of methamphetamine (METH), a highly addictive psychostimulant, is a huge health care burden due to its central and peripheral toxic effects. Mounting clinical studies have noted that METH use in humans is associated with the development of cardiomyopathy; however, preclinical studies and animal models to dissect detailed molecular mechanisms of METH-associated cardiomyopathy development are scarce. The present study utilized a unique very long-access binge and crash procedure of METH self-administration to characterize the sequelae of pathological alterations that occur with METH-associated cardiomyopathy. Rats were allowed to intravenously self-administer METH for 96 h continuous weekly sessions over 8 weeks. Cardiac function, histochemistry, ultrastructure, and biochemical experiments were performed 24 h after the cessation of drug administration. Voluntary METH self-administration induced pathological cardiac remodeling as indicated by cardiomyocyte hypertrophy, myocyte disarray, interstitial and perivascular fibrosis accompanied by compromised cardiac systolic function. Ultrastructural examination and native gel electrophoresis revealed altered mitochondrial morphology and reduced mitochondrial oxidative phosphorylation (OXPHOS) supercomplexes (SCs) stability and assembly in METH exposed hearts. Redox-sensitive assays revealed significantly attenuated mitochondrial respiratory complex activities with a compensatory increase in pyruvate dehydrogenase (PDH) activity reminiscent of metabolic remodeling. Increased autophagy flux and increased mitochondrial antioxidant protein level was observed in METH exposed heart. Treatment with mitoTEMPO reduced the autophagy level indicating the involvement of mitochondrial dysfunction in the adaptive activation of autophagy in METH exposed hearts. Altogether, we have reported a novel METH-associated cardiomyopathy model using voluntary drug seeking behavior. Our studies indicated that METH self-administration profoundly affects mitochondrial ultrastructure, OXPHOS SCs assembly and redox activity accompanied by increased PDH activity that may underlie observed cardiac dysfunction., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2022 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
8. Methamphetamine causes cardiovascular dysfunction via cystathionine gamma lyase and hydrogen sulfide depletion.
- Author
-
Kolluru GK, Glawe JD, Pardue S, Kasabali A, Alam S, Rajendran S, Cannon AL, Abdullah CS, Traylor JG, Shackelford RE, Woolard MD, Orr AW, Goeders NE, Dominic P, Bhuiyan MSS, and Kevil CG
- Abstract
Methamphetamine (METH) is an addictive illicit drug used worldwide that causes significant damage to blood vessels resulting in cardiovascular dysfunction. Recent studies highlight increased prevalence of cardiovascular disease (CVD) and associated complications including hypertension, vasospasm, left ventricular hypertrophy, and coronary artery disease in younger populations due to METH use. Here we report that METH administration in a mouse model of 'binge and crash' decreases cardiovascular function via cystathionine gamma lyase (CSE), hydrogen sulfide (H
2 S), nitric oxide (NO) (CSE/H2 S/NO) dependent pathway. METH significantly reduced H2 S and NO bioavailability in plasma and skeletal muscle tissues co-incident with a significant reduction in flow-mediated vasodilation (FMD) and blood flow velocity revealing endothelial dysfunction. METH administration also reduced cardiac ejection fraction (EF) and fractional shortening (FS) associated with increased tissue and perivascular fibrosis. Importantly, METH treatment selectively decreased CSE expression and sulfide bioavailability along with reduced eNOS phosphorylation and NO levels. Exogenous sulfide therapy or endothelial CSE transgenic overexpression corrected cardiovascular and associated pathological responses due to METH implicating a central molecular regulatory pathway for tissue pathology. These findings reveal that therapeutic intervention targeting CSE/H2 S bioavailability may be useful in attenuating METH mediated cardiovascular disease., (Copyright © 2022 The Authors. Published by Elsevier B.V. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
9. SGLT2 Inhibitors-A Medical Revelation: Molecular Signaling of Canagliflozin Underlying Hypertension and Vascular Remodeling.
- Author
-
Yagi T and Kolluru GK
- Subjects
- Canagliflozin therapeutic use, Humans, Hypoglycemic Agents, Sodium-Glucose Transporter 2, Vascular Remodeling, Diabetes Mellitus, Type 2 complications, Diabetes Mellitus, Type 2 drug therapy, Hypertension drug therapy, Sodium-Glucose Transporter 2 Inhibitors therapeutic use
- Published
- 2022
- Full Text
- View/download PDF
10. Neurogranin regulates calcium-dependent cardiac hypertrophy.
- Author
-
Jorgensen AN, Abdullah CS, Bhuiyan MS, Watt M, Dominic P, Kolluru GK, Kevil CG, and Nam HW
- Subjects
- Animals, Calcium-Calmodulin-Dependent Protein Kinase Type 2 genetics, Calcium-Calmodulin-Dependent Protein Kinase Type 2 metabolism, Calmodulin metabolism, Cardiomegaly metabolism, Fibrosis, Mice, Mice, Knockout, Myocytes, Cardiac metabolism, Calcium metabolism, Neurogranin genetics, Neurogranin metabolism
- Abstract
Intracellular Ca
2+ -calmodulin (CaM) signaling plays an important role in Ca2+ -CaM-dependent kinase (CaMKII) and calcineurin (CaN)-mediated cardiac biology. While neurogranin (Ng) is known as a major Ca2+ -CaM modulator in the brain, its pathophysiological role in cardiac hypertrophy has never been studied before. In the present study, we report that Ng is expressed in the heart and depletion of Ng dysregulates Ca2+ homeostasis and promotes cardiac failure in mice. 10-month-old Ng null mice demonstrate significantly increased heart-to-body weight ratios compared to wild-type. Using histological approaches, we identified that depletion of Ng increases cardiac hypertrophy, fibrosis, and collagen deposition near perivascular areas in the heart tissue of Ng null mice. Ca2+ spark experiments revealed that cardiac myocytes isolated from Ng null mice have decreased spark frequency and width, while the duration of sparks is significantly increased. We also identified that a lack of Ng increases CaMKIIδ signaling and periostin protein expression in these mouse hearts. Overall, we are the first study to explore how Ng expression in the heart plays an important role in Ca2+ homeostasis in cardiac myocytes as well as the pathophysiology of cardiac hypertrophy and fibrosis., (Copyright © 2022 Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
11. Decreased availability of nitric oxide and hydrogen sulfide is a hallmark of COVID-19.
- Author
-
Dominic P, Ahmad J, Bhandari R, Pardue S, Solorzano J, Jaisingh K, Watts M, Bailey SR, Orr AW, Kevil CG, and Kolluru GK
- Subjects
- Humans, Nitric Oxide, SARS-CoV-2, COVID-19, Gasotransmitters, Hydrogen Sulfide
- Abstract
Background: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is involved in a global outbreak affecting millions of people who manifest a variety of symptoms. Coronavirus disease 2019 (COVID-19) caused by SARS-CoV-2 is increasingly associated with cardiovascular complications requiring hospitalizations; however, the mechanisms underlying these complications remain unknown. Nitric oxide (NO) and hydrogen sulfide (H
2 S) are gasotransmitters that regulate key cardiovascular functions., Methods: Blood samples were obtained from 68 COVID-19 patients and 33 controls and NO and H2 S metabolites were assessed. H2 S and NO levels were compared between cases and controls in the entire study population and subgroups based on race. The availability of gasotransmitters was examined based on severity and outcome of COVID-19 infection. The performance of H2 S and NO levels in predicting COVID-19 infection was also analyzed. Multivariable regression analysis was performed to identify the effects of traditional determinants of gasotransmitters on NO and H2 S levels in the patients with COVID-19 infection., Results: Significantly reduced NO and H2 S levels were observed in both Caucasian and African American COVID-19 patients compared to healthy controls. COVID-19 patients who died had significantly higher NO and H2 S levels compared to COVID-19 patients who survived. Receiver-operating characteristic analysis of NO and H2 S metabolites in the study population showed free sulfide levels to be highly predictive of COVID-19 infection based on reduced availability. Traditional determinants of gasotransmitters, namely age, race, sex, diabetes, and hypertension had no effect on NO and H2 S levels in COVID-19 patients., Conclusion: These observations provide the first insight into the role of NO and H2 S in COVID-19 infection, where their low availability may be a result of reduced synthesis secondary to endotheliitis, or increased consumption from scavenging of reactive oxygen species., (Copyright © 2021 The Author(s). Published by Elsevier B.V. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
12. Decreased bioavailability of hydrogen sulfide links vascular endothelium and atrial remodeling in atrial fibrillation.
- Author
-
Watts M, Kolluru GK, Dherange P, Pardue S, Si M, Shen X, Trosclair K, Glawe J, Al-Yafeai Z, Iqbal M, Pearson BH, Hamilton KA, Orr AW, Glasscock E, Kevil CG, and Dominic P
- Subjects
- Animals, Biological Availability, Case-Control Studies, Endothelium, Vascular, Humans, Mice, Mice, Knockout, Atrial Fibrillation, Atrial Remodeling, Hydrogen Sulfide
- Abstract
Oxidative stress drives the pathogenesis of atrial fibrillation (AF), the most common arrhythmia. In the cardiovascular system, cystathionine γ-lyase (CSE) serves as the primary enzyme producing hydrogen sulfide (H
2 S), a mammalian gasotransmitter that reduces oxidative stress. Using a case control study design in patients with and without AF and a mouse model of CSE knockout (CSE-KO), we evaluated the role of H2 S in the etiology of AF. Patients with AF (n = 51) had significantly reduced plasma acid labile sulfide levels compared to patients without AF (n = 65). In addition, patients with persistent AF (n = 25) showed lower plasma free sulfide levels compared to patients with paroxysmal AF (n = 26). Consistent with an important role for H2 S in AF, CSE-KO mice had decreased atrial sulfide levels, increased atrial superoxide levels, and enhanced propensity for induced persistent AF compared to wild type (WT) mice. Rescuing H2 S signaling in CSE-KO mice by Diallyl trisulfide (DATS) supplementation or reconstitution with endothelial cell specific CSE over-expression significantly reduced atrial superoxide, increased sulfide levels, and lowered AF inducibility. Lastly, low H2 S levels in CSE KO mice was associated with atrial electrical remodeling including longer effective refractory periods, slower conduction velocity, increased myocyte calcium sparks, and increased myocyte action potential duration that were reversed by DATS supplementation or endothelial CSE overexpression. Our findings demonstrate an important role of CSE and H2 S bioavailability in regulating electrical remodeling and susceptibility to AF., (Copyright © 2020 The Authors. Published by Elsevier B.V. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
13. Methamphetamine induces cardiomyopathy by Sigmar1 inhibition-dependent impairment of mitochondrial dynamics and function.
- Author
-
Abdullah CS, Aishwarya R, Alam S, Morshed M, Remex NS, Nitu S, Kolluru GK, Traylor J, Miriyala S, Panchatcharam M, Hartman B, King J, Bhuiyan MAN, Chandran S, Woolard MD, Yu X, Goeders NE, Dominic P, Arnold CL, Stokes K, Kevil CG, Orr AW, and Bhuiyan MS
- Subjects
- Animals, Cardiomyopathies prevention & control, Cyclic AMP Response Element-Binding Protein genetics, Cyclic AMP Response Element-Binding Protein metabolism, Drug Administration Schedule, Gene Expression Regulation drug effects, Heart drug effects, Humans, Methamphetamine administration & dosage, Mice, Mitochondria metabolism, Mitochondrial Proteins genetics, Mitochondrial Proteins metabolism, Myocardium pathology, Myocytes, Cardiac drug effects, Receptors, sigma genetics, Sigma-1 Receptor, Cardiomyopathies chemically induced, Methamphetamine toxicity, Mitochondria drug effects, Receptors, sigma metabolism
- Abstract
Methamphetamine-associated cardiomyopathy is the leading cause of death linked with illicit drug use. Here we show that Sigmar1 is a therapeutic target for methamphetamine-associated cardiomyopathy and defined the molecular mechanisms using autopsy samples of human hearts, and a mouse model of "binge and crash" methamphetamine administration. Sigmar1 expression is significantly decreased in the hearts of human methamphetamine users and those of "binge and crash" methamphetamine-treated mice. The hearts of methamphetamine users also show signs of cardiomyopathy, including cellular injury, fibrosis, and enlargement of the heart. In addition, mice expose to "binge and crash" methamphetamine develop cardiac hypertrophy, fibrotic remodeling, and mitochondrial dysfunction leading to contractile dysfunction. Methamphetamine treatment inhibits Sigmar1, resulting in inactivation of the cAMP response element-binding protein (CREB), decreased expression of mitochondrial fission 1 protein (FIS1), and ultimately alteration of mitochondrial dynamics and function. Therefore, Sigmar1 is a viable therapeutic agent for protection against methamphetamine-associated cardiomyopathy.
- Published
- 2020
- Full Text
- View/download PDF
14. Neurogranin regulates eNOS function and endothelial activation.
- Author
-
Cheriyan VT, Alfaidi M, Jorgensen AN, Alam MA, Abdullah CS, Kolluru GK, Bhuiyan MS, Kevil CG, Orr AW, and Nam HW
- Subjects
- Animals, Cells, Cultured, Endothelial Cells metabolism, Mice, Nitric Oxide metabolism, Phosphorylation, Neurogranin, Nitric Oxide Synthase Type III genetics, Nitric Oxide Synthase Type III metabolism
- Abstract
Endothelial nitric oxide (NO) is a critical mediator of vascular function and vascular remodeling. NO is produced by endothelial nitric oxide synthase (eNOS), which is activated by calcium (Ca
2+ )-dependent and Ca2+ -independent pathways. Here, we report that neurogranin (Ng), which regulates Ca2+ -calmodulin (CaM) signaling in the brain, is uniquely expressed in endothelial cells (EC) of human and mouse vasculature, and is also required for eNOS regulation. To test the role of Ng in eNOS activation, Ng knockdown in human aortic endothelial cells (HAEC) was performed using Ng SiRNA along with Ng knockout (Ng-/- ) in mice. Depletion of Ng expression decreased eNOS activity in HAEC and NO production in mice. We show that Ng expression was decreased by short-term laminar flow and long-them oscillating flow shear stress, and that Ng siRNA with shear stress decreased eNOS expression as well as eNOS phosphorylation at S1177. We further reveled that lack of Ng expression decreases both AKT-dependent eNOS phosphorylation, NF-κB-mediated eNOS expression, and promotes endothelial activation. Our findings also indicate that Ng modulates Ca2+ -dependent calcineurin (CaN) activity, which suppresses Ca2+ -independent AKT-dependent eNOS signaling. Moreover, deletion of Ng in mice also reduced eNOS activity and caused endothelial dysfunction in flow-mediated dilation experiments. Our results demonstrate that Ng plays a crucial role in Ca2+ -CaM-dependent eNOS regulation and contributes to vascular remodeling, which is important for the pathophysiology of cardiovascular disease., Competing Interests: Declaration of competing interest All authors declare no conflicts of interest., (Copyright © 2020 The Authors. Published by Elsevier B.V. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
15. Hydrogen sulfide stimulates xanthine oxidoreductase conversion to nitrite reductase and formation of NO.
- Author
-
Pardue S, Kolluru GK, Shen X, Lewis SE, Saffle CB, Kelley EE, and Kevil CG
- Subjects
- Animals, Endothelial Cells, Mice, Nitric Oxide, Nitrite Reductases, Hydrogen Sulfide, Xanthine Dehydrogenase genetics
- Abstract
Cardiovascular disease is the leading cause of death and disability worldwide with increased oxidative stress and reduced NO bioavailability serving as key risk factors. For decades, elevation in protein abundance and enzymatic activity of xanthine oxidoreductase (XOR) under hypoxic/inflammatory conditions has been associated with organ damage and vascular dysfunction. Recent reports have challenged this dogma by identifying a beneficial function for XOR, under similar hypoxic/acidic conditions, whereby XOR catalyzes the reduction of nitrite (NO2
- ) to nitric oxide (NO) through poorly defined mechanisms. We previously reported that hydrogen sulfide (H2 S/sulfide) confers significant vascular benefit under these same conditions via NO2- mediated mechanisms independent of nitric oxide synthase (NOS). Here we report for the first time the convergence of H2 S, XOR, and nitrite to form a concerted triad for NO generation. Specifically, hypoxic endothelial cells show a dose-dependent, sulfide and polysulfide (diallyl trisulfide (DATS)-induced, NOS-independent NO2 - reduction to NO that is dependent upon the enzymatic activity of XOR. Interestingly, nitrite reduction to NO was found to be slower and more sustained with DATS compared to H2 S. Capacity for sulfide/polysulfide to produce an XOR-dependent impact on NO generation translates to salutary actions in vivo as DATS administration in cystathionine-γ-lyase (CSE) knockout mice significantly improved hindlimb ischemia blood flow post ligation, while the XOR-specific inhibitor, febuxostat (Febx), abrogated this benefit. Moreover, flow-mediated vasodilation (FMD) in CSE knockout mice following administration of DATS resulted in greater than 4-fold enhancement in femoral artery dilation while co-treatment with Febx completely completely abrogated this effect. Together, these results identify XOR as a focal point of convergence between sulfide- and nitrite-mediated signaling, as well as affirm the critical need to reexamine current dogma regarding inhibition of XOR in the context of vascular dysfunction., Competing Interests: Declaration of competing interest C.G.K. has provisional patents regarding nitric oxide/nitrite and hydrogen sulfide chemistry and is a co-founder of Innolyzer LLC. X.S. has intellectual property regarding hydrogen sulfide chemistry., (Copyright © 2020. Published by Elsevier B.V.)- Published
- 2020
- Full Text
- View/download PDF
16. It's a "Gut Feeling": Association of Microbiota, Trimethylamine N-Oxide and Cardiovascular Outcomes.
- Author
-
Kolluru GK and Kevil CG
- Subjects
- Case-Control Studies, Humans, Methylamines, Patients, Gastrointestinal Microbiome, Microbiota, Myocardial Infarction
- Published
- 2020
- Full Text
- View/download PDF
17. Reactive Sulfur Species: A New Redox Player in Cardiovascular Pathophysiology.
- Author
-
Kolluru GK, Shen X, and Kevil CG
- Subjects
- Biological Availability, Cardiovascular Diseases physiopathology, Cystathionine gamma-Lyase genetics, Humans, Nitrogen Oxides metabolism, Oxidation-Reduction, Polymorphism, Genetic, Sulfides metabolism, Vascular Remodeling, Cardiovascular Diseases metabolism, Hydrogen Sulfide metabolism, Oxidative Stress, Reactive Oxygen Species metabolism
- Abstract
Hydrogen sulfide has emerged as an important gaseous signaling molecule and a regulator of critical biological processes. However, the physiological significance of hydrogen sulfide metabolites such as persulfides, polysulfides, and other reactive sulfur species (RSS) has only recently been appreciated. Emerging evidence suggests that these RSS molecules may have similar or divergent regulatory roles compared with hydrogen sulfide in various biological activities. However, the chemical nature of persulfides and polysulfides is complex and remains poorly understood within cardiovascular and other pathophysiological conditions. Recent reports suggest that RSS can be produced endogenously, with different forms having unique chemical properties and biological implications involving diverse cellular responses such as protein biosynthesis, cell-cell barrier functions, and mitochondrial bioenergetics. Enzymes of the transsulfuration pathway, CBS (cystathionine beta-synthase) and CSE (cystathionine gamma-lyase), may also produce RSS metabolites besides hydrogen sulfide. Moreover, CARSs (cysteinyl-tRNA synthetase) are also able to generate protein persulfides via cysteine persulfide (CysSSH) incorporation into nascently formed polypeptides suggesting a new biologically relevant amino acid. This brief review discusses the biochemical nature and potential roles of RSS, associated oxidative stress redox signaling, and future research opportunities in cardiovascular disease.
- Published
- 2020
- Full Text
- View/download PDF
18. Macrophage Metabolism of Apoptotic Cell-Derived Arginine Promotes Continual Efferocytosis and Resolution of Injury.
- Author
-
Yurdagul A Jr, Subramanian M, Wang X, Crown SB, Ilkayeva OR, Darville L, Kolluru GK, Rymond CC, Gerlach BD, Zheng Z, Kuriakose G, Kevil CG, Koomen JM, Cleveland JL, Muoio DM, and Tabas I
- Subjects
- Animals, Apoptosis genetics, Arginase metabolism, ELAV-Like Protein 1 metabolism, Gene Deletion, Guanine Nucleotide Exchange Factors genetics, Guanine Nucleotide Exchange Factors metabolism, Humans, Jurkat Cells, Macrophages drug effects, Male, Mice, Inbred C57BL, Myeloid Cells drug effects, Myeloid Cells metabolism, Ornithine Decarboxylase metabolism, Phagocytosis genetics, Putrescine biosynthesis, RNA Stability drug effects, RNA Stability genetics, RNA, Messenger genetics, RNA, Messenger metabolism, rac1 GTP-Binding Protein metabolism, Apoptosis drug effects, Arginine pharmacology, Macrophages metabolism, Macrophages pathology, Phagocytosis drug effects
- Abstract
Continual efferocytic clearance of apoptotic cells (ACs) by macrophages prevents necrosis and promotes injury resolution. How continual efferocytosis is promoted is not clear. Here, we show that the process is optimized by linking the metabolism of engulfed cargo from initial efferocytic events to subsequent rounds. We found that continual efferocytosis is enhanced by the metabolism of AC-derived arginine and ornithine to putrescine by macrophage arginase 1 (Arg1) and ornithine decarboxylase (ODC). Putrescine augments HuR-mediated stabilization of the mRNA encoding the GTP-exchange factor Dbl, which activates actin-regulating Rac1 to facilitate subsequent rounds of AC internalization. Inhibition of any step along this pathway after first-AC uptake suppresses second-AC internalization, whereas putrescine addition rescues this defect. Mice lacking myeloid Arg1 or ODC have defects in efferocytosis in vivo and in atherosclerosis regression, while treatment with putrescine promotes atherosclerosis resolution. Thus, macrophage metabolism of AC-derived metabolites allows for optimal continual efferocytosis and resolution of injury., Competing Interests: Declaration of Interests The authors declare no competing interests., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
19. Methamphetamine Use and Cardiovascular Disease.
- Author
-
Kevil CG, Goeders NE, Woolard MD, Bhuiyan MS, Dominic P, Kolluru GK, Arnold CL, Traylor JG, and Orr AW
- Subjects
- Arrhythmias, Cardiac chemically induced, Atherosclerosis chemically induced, Cardiomyopathies chemically induced, Humans, Hypertension, Pulmonary chemically induced, Vasoconstriction drug effects, Cardiovascular Diseases chemically induced, Methamphetamine toxicity
- Abstract
While the opioid epidemic has garnered significant attention, the use of methamphetamines is growing worldwide independent of wealth or region. Following overdose and accidents, the leading cause of death in methamphetamine users is cardiovascular disease, because of significant effects of methamphetamine on vasoconstriction, pulmonary hypertension, atherosclerotic plaque formation, cardiac arrhythmias, and cardiomyopathy. In this review, we examine the current literature on methamphetamine-induced changes in cardiovascular health, discuss the potential mechanisms regulating these varied effects, and highlight our deficiencies in understanding how to treat methamphetamine-associated cardiovascular dysfunction.
- Published
- 2019
- Full Text
- View/download PDF
20. Nitric Oxide and Hydrogen Sulfide Regulation of Ischemic Vascular Growth and Remodeling.
- Author
-
Rajendran S, Shen X, Glawe J, Kolluru GK, and Kevil CG
- Subjects
- Cardiovascular Diseases genetics, Cardiovascular Diseases physiopathology, Humans, Intercellular Signaling Peptides and Proteins physiology, Ischemia therapy, Mutation, Neovascularization, Pathologic physiopathology, Nitric Oxide chemistry, Signal Transduction physiology, Hydrogen Sulfide metabolism, Ischemia physiopathology, Neovascularization, Physiologic physiology, Nitric Oxide physiology, Vascular Remodeling physiology
- Abstract
Ischemic vascular remodeling occurs in response to stenosis or arterial occlusion leading to a change in blood flow and tissue perfusion. Altered blood flow elicits a cascade of molecular and cellular physiological responses leading to vascular remodeling of the macro- and micro-circulation. Although cellular mechanisms of vascular remodeling such as arteriogenesis and angiogenesis have been studied, therapeutic approaches in these areas have had limited success due to the complexity and heterogeneous constellation of molecular signaling events regulating these processes. Understanding central molecular players of vascular remodeling should lead to a deeper understanding of this response and aid in the development of novel therapeutic strategies. Hydrogen sulfide (H
2 S) and nitric oxide (NO) are gaseous signaling molecules that are critically involved in regulating fundamental biochemical and molecular responses necessary for vascular growth and remodeling. This review examines how NO and H2 S regulate pathophysiological mechanisms of angiogenesis and arteriogenesis, along with important chemical and experimental considerations revealed thus far. The importance of NO and H2 S bioavailability, their synthesis enzymes and cofactors, and genetic variations associated with cardiovascular risk factors suggest that they serve as pivotal regulators of vascular remodeling responses. © 2019 American Physiological Society. Compr Physiol 9:1213-1247, 2019., (Copyright © 2019 American Physiological Society. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
21. Total sulfane sulfur bioavailability reflects ethnic and gender disparities in cardiovascular disease.
- Author
-
Rajpal S, Katikaneni P, Deshotels M, Pardue S, Glawe J, Shen X, Akkus N, Modi K, Bhandari R, Dominic P, Reddy P, Kolluru GK, and Kevil CG
- Subjects
- Adult, Black or African American genetics, Aged, Biological Availability, Bridged Bicyclo Compounds chemistry, Cardiovascular Diseases genetics, Cardiovascular Diseases pathology, Chromatography, Liquid, Female, Gene Frequency, High-Throughput Nucleotide Sequencing, Humans, Hydrogen Sulfide isolation & purification, Male, Middle Aged, Polymorphism, Single Nucleotide, White People genetics, Cardiovascular Diseases blood, Cystathionine gamma-Lyase genetics, Hydrogen Sulfide blood, Sulfides blood
- Abstract
Hydrogen sulfide (H
2 S) has emerged as an important physiological and pathophysiological signaling molecule in the cardiovascular system influencing vascular tone, cytoprotective responses, redox reactions, vascular adaptation, and mitochondrial respiration. However, bioavailable levels of H2 S in its various biochemical metabolite forms during clinical cardiovascular disease remain poorly understood. We performed a case-controlled study to quantify and compare the bioavailability of various biochemical forms of H2 S in patients with and without cardiovascular disease (CVD). In our study, we used the reverse-phase high performance liquid chromatography monobromobimane assay to analytically measure bioavailable pools of H2 S. Single nucleotide polymorphisms (SNPs) were also identified using DNA Pyrosequencing. We found that plasma acid labile sulfide levels were significantly reduced in Caucasian females with CVD compared with those without the disease. Conversely, plasma bound sulfane sulfur levels were significantly reduced in Caucasian males with CVD compared with those without the disease. Surprisingly, gender differences of H2 S bioavailability were not observed in African Americans, although H2 S bioavailability was significantly lower overall in this ethnic group compared to Caucasians. We also performed SNP analysis of H2 S synthesizing enzymes and found a significant increase in cystathionine gamma-lyase (CTH) 1364 G-T allele frequency in patients with CVD compared to controls. Lastly, plasma H2 S bioavailability was found to be predictive for cardiovascular disease in Caucasian subjects as determined by receiver operator characteristic analysis. These findings reveal that plasma H2 S bioavailability could be considered a biomarker for CVD in an ethnic and gender manner. Cystathionine gamma-lyase 1346 G-T SNP might also contribute to the risk of cardiovascular disease development., (Copyright © 2018 The Authors. Published by Elsevier B.V. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
22. Hypoxia perturbs endothelium by re-organizing cellular actin architecture: Nitric oxide offers limited protection.
- Author
-
Swaminathan A, Kasiviswanathan D, Balaguru UM, Kolluru GK, SuryaKumar G, and Chatterjee S
- Subjects
- Actin Cytoskeleton genetics, Actin Cytoskeleton metabolism, Actins metabolism, Animals, Cell Hypoxia genetics, Cell Line, Cell Membrane Permeability, Chick Embryo, Endothelial Cells physiology, Endothelium, Vascular metabolism, Humans, Cell Hypoxia physiology, Endothelial Cells metabolism, Endothelium, Vascular growth & development, Nitric Oxide metabolism
- Abstract
Exposure to hypoxia causes structural changes in the endothelial cell (EC) monolayer that alter its permeability. There was a report earlier of impairment of nitric oxide (NO) production in endothelium. The intervention of NO in the altered cellular arrangements of actin cytoskeleton in endothelium for rectification of paracellular gaps in endothelium under hypoxia was observed. The present study demonstrates hypoxia inducing paracellular gaps in hypoxia-exposed blood capillaries in chick embryo extravascular model. Phalloidin staining confirmed significant polymerization of actin and unique cellular localization of the F-actin bands under hypoxia treatments. Addition of spermine NONOate (SPNO), a NO donor, or reoxygenation to endothelial monolayer attenuated hypoxia-mediated effects on endothelial permeability with partial recovery of endothelial integrity through actin remodeling. The present study indicates link of hypoxia-induced actin-associated cytoskeletal rearrangements and paracellular gaps in the endothelium with a low NO availability in the hypoxia milieu. The author concludes that NO confers protection against hypoxia-mediated cytoskeletal remodeling and endothelial leakiness., (Copyright © 2017 Elsevier Ltd. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
23. Gasotransmitter Heterocellular Signaling.
- Author
-
Kolluru GK, Shen X, Yuan S, and Kevil CG
- Subjects
- Animals, Carbon Monoxide, Humans, Hydrogen Sulfide metabolism, Models, Biological, Nitric Oxide metabolism, Signal Transduction, Gasotransmitters metabolism, Oxidation-Reduction
- Abstract
Significance: The family of gasotransmitter molecules, nitric oxide (NO), carbon monoxide (CO), and hydrogen sulfide (H
2 S), has emerged as an important mediator of numerous cellular signal transduction and pathophysiological responses. As such, these molecules have been reported to influence a diverse array of biochemical, molecular, and cell biology events often impacting one another. Recent Advances: Discrete regulation of gasotransmitter molecule formation, movement, and reaction is critical to their biological function. Due to the chemical nature of these molecules, they can move rapidly throughout cells and tissues acting on targets through reactions with metal groups, reactive chemical species, and protein amino acids., Critical Issues: Given the breadth and complexity of gasotransmitter reactions, this field of research is expanding into exciting, yet sometimes confusing, areas of study with significant promise for understanding health and disease. The precise amounts of tissue and cellular gasotransmitter levels and where they are formed, as well as how they react with molecular targets or themselves, all remain poorly understood., Future Directions: Elucidation of specific molecular targets, characteristics of gasotransmitter molecule heterotypic interactions, and spatiotemporal formation and metabolism are all important to better understand their true pathophysiological importance in various organ systems. Antioxid. Redox Signal. 26, 936-960.- Published
- 2017
- Full Text
- View/download PDF
24. Small Molecule, Big Prospects: MicroRNA in Pregnancy and Its Complications.
- Author
-
Cai M, Kolluru GK, and Ahmed A
- Subjects
- Female, Fetal Growth Retardation metabolism, Gene Expression Regulation, Developmental, Humans, MicroRNAs genetics, Obstetric Labor, Premature metabolism, Pre-Eclampsia metabolism, Pregnancy, Uterus metabolism, Fetal Growth Retardation genetics, MicroRNAs metabolism, Obstetric Labor, Premature genetics, Placenta metabolism, Pre-Eclampsia genetics
- Abstract
MicroRNAs are small, noncoding RNA molecules that regulate target gene expression in the posttranscriptional level. Unlike siRNA, microRNAs are "fine-tuners" rather than "switches" in the regulation of gene expression; thus they play key roles in maintaining tissue homeostasis. The aberrant microRNA expression is implicated in the disease process. To date, numerous studies have demonstrated the regulatory roles of microRNAs in various pathophysiological conditions. In contrast, the study of microRNA in pregnancy and its associated complications, such as preeclampsia (PE), fetal growth restriction (FGR), and preterm labor, is a young field. Over the last decade, the knowledge of pregnancy-related microRNAs has increased and the molecular mechanisms by which microRNAs regulate pregnancy or its associated complications are emerging. In this review, we focus on the recent advances in the research of pregnancy-related microRNAs, especially their function in pregnancy-associated complications and the potential clinical applications. Here microRNAs that associate with pregnancy are classified as placenta-specific, placenta-associated, placenta-derived circulating, and uterine microRNA according to their localization and origin. MicroRNAs offer a great potential for developing diagnostic and therapeutic targets in pregnancy-related disorders.
- Published
- 2017
- Full Text
- View/download PDF
25. AltitudeOmics: Red Blood Cell Metabolic Adaptation to High Altitude Hypoxia.
- Author
-
D'Alessandro A, Nemkov T, Sun K, Liu H, Song A, Monte AA, Subudhi AW, Lovering AT, Dvorkin D, Julian CG, Kevil CG, Kolluru GK, Shiva S, Gladwin MT, Xia Y, Hansen KC, and Roach RC
- Subjects
- Acclimatization physiology, Adult, Altitude, Altitude Sickness physiopathology, Arginine metabolism, Glutathione metabolism, Glycolysis, Healthy Volunteers, Humans, Pentose Phosphate Pathway, Purines metabolism, Sulfur metabolism, Time Factors, Young Adult, Adaptation, Physiological, Altitude Sickness metabolism, Erythrocytes metabolism
- Abstract
Red blood cells (RBCs) are key players in systemic oxygen transport. RBCs respond to in vitro hypoxia through the so-called oxygen-dependent metabolic regulation, which involves the competitive binding of deoxyhemoglobin and glycolytic enzymes to the N-terminal cytosolic domain of band 3. This mechanism promotes the accumulation of 2,3-DPG, stabilizing the deoxygenated state of hemoglobin, and cytosol acidification, triggering oxygen off-loading through the Bohr effect. Despite in vitro studies, in vivo adaptations to hypoxia have not yet been completely elucidated. Within the framework of the AltitudeOmics study, erythrocytes were collected from 21 healthy volunteers at sea level, after exposure to high altitude (5260 m) for 1, 7, and 16 days, and following reascent after 7 days at 1525 m. UHPLC-MS metabolomics results were correlated to physiological and athletic performance parameters. Immediate metabolic adaptations were noted as early as a few hours from ascending to >5000 m, and maintained for 16 days at high altitude. Consistent with the mechanisms elucidated in vitro, hypoxia promoted glycolysis and deregulated the pentose phosphate pathway, as well purine catabolism, glutathione homeostasis, arginine/nitric oxide, and sulfur/H
2 S metabolism. Metabolic adaptations were preserved 1 week after descent, consistently with improved physical performances in comparison to the first ascendance, suggesting a mechanism of metabolic memory.- Published
- 2016
- Full Text
- View/download PDF
26. Oxygen tension, H2S, and NO bioavailability: is there an interaction?
- Author
-
Kolluru GK, Prasai PK, Kaskas AM, Letchuman V, and Pattillo CB
- Subjects
- Animals, Biological Availability, Homeostasis physiology, Humans, Hydrogen Sulfide metabolism, Hypoxia metabolism, Nitric Oxide metabolism, Oxygen metabolism
- Abstract
Molecular oxygen (O2) is an essential component for survival and development. Variation in O2 levels leads to changes in molecular signaling and ultimately affects the physiological functions of many organisms. Nitric oxide (NO) and hydrogen sulfide (H2S) are two gaseous cellular signaling molecules that play key roles in several physiological functions involved in maintaining vascular homeostasis including vasodilation, anti-inflammation, and vascular growth. Apart from the aforementioned functions, NO and H2S are believed to mediate hypoxic responses and serve as O2 chemosensors in biological systems. In this literature review, we briefly discuss NO and H2S and their roles during hypoxia., (Copyright © 2016 the American Physiological Society.)
- Published
- 2016
- Full Text
- View/download PDF
27. Cystathionine γ-lyase regulates arteriogenesis through NO-dependent monocyte recruitment.
- Author
-
Kolluru GK, Bir SC, Yuan S, Shen X, Pardue S, Wang R, and Kevil CG
- Subjects
- Animals, Cytokines metabolism, Disease Models, Animal, Ischemia metabolism, Male, Mice, Inbred C57BL, Mice, Knockout, Neovascularization, Physiologic, Sulfides metabolism, Cystathionine gamma-Lyase genetics, Cystathionine gamma-Lyase metabolism, Hydrogen Sulfide metabolism, Monocytes metabolism, Nitric Oxide metabolism
- Abstract
Aims: Hydrogen sulfide (H2S) is a vasoactive gasotransmitter that is endogenously produced in the vasculature by the enzyme cystathionine γ-lyase (CSE). However, the importance of CSE activity and local H2S generation for ischaemic vascular remodelling remains completely unknown. In this study, we examine the hypothesis that CSE critically regulates ischaemic vascular remodelling involving H2S-dependent mononuclear cell regulation of arteriogenesis., Methods and Results: Arteriogenesis including mature vessel density, collateral formation, blood flow, and SPY angiographic blush rate were determined in wild-type (WT) and CSE knockout (KO) mice at different time points following femoral artery ligation (FAL). The role of endogenous H2S in regulation of IL-16 expression and subsequent recruitment of monocytes, and expression of VEGF and bFGF in ischaemic tissues, were determined along with endothelial progenitor cell (CD34/Flk1) formation and function. FAL of WT mice significantly increased CSE activity, expression and endogenous H2S generation in ischaemic tissues, and monocyte infiltration, which was absent in CSE-deficient mice. Treatment of CSE KO mice with the polysulfide donor diallyl trisulfide restored ischaemic vascular remodelling, monocyte infiltration, and cytokine expression. Importantly, exogenous H2S therapy restored nitric oxide (NO) bioavailability in CSE KO mice that was responsible for monocyte recruitment and arteriogenesis., Conclusion: Endogenous CSE/H2S regulates ischaemic vascular remodelling mediated during hind limb ischaemia through NO-dependent monocyte recruitment and cytokine induction revealing a previously unknown mechanism of arteriogenesis., (Published on behalf of the European Society of Cardiology. All rights reserved. © The Author 2015. For permissions please email: journals.permissions@oup.com.)
- Published
- 2015
- Full Text
- View/download PDF
28. Measurement of H2S in vivo and in vitro by the monobromobimane method.
- Author
-
Shen X, Kolluru GK, Yuan S, and Kevil CG
- Subjects
- Animals, Chromatography, High Pressure Liquid, Humans, Hydrogen Sulfide isolation & purification, Hydrogen Sulfide metabolism, Reference Standards, Spectrometry, Fluorescence, Spectrometry, Mass, Electrospray Ionization, Bridged Bicyclo Compounds chemistry, Fluorescent Dyes chemistry, Hydrogen Sulfide chemistry
- Abstract
The gasotransmitter hydrogen sulfide (H2S) is known as an important regulator in several physiological and pathological responses. Among the challenges facing the field is the accurate and reliable measurement of hydrogen sulfide bioavailability. We have reported an approach to discretely measure sulfide and sulfide pools using the monobromobimane (MBB) method coupled with reversed phase high-performance liquid chromatography (RP-HPLC). The method involves the derivatization of sulfide with excess MBB under precise reaction conditions at room temperature to form sulfide dibimane (SDB). The resultant fluorescent SDB is analyzed by RP-HPLC using fluorescence detection with the limit of detection for SDB (2 nM). Care must be taken to avoid conditions that may confound H2S measurement with this method. Overall, RP-HPLC with fluorescence detection of SDB is a useful and powerful tool to measure biological sulfide levels., (© 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
29. H2S regulation of nitric oxide metabolism.
- Author
-
Kolluru GK, Yuan S, Shen X, and Kevil CG
- Subjects
- Animals, Blotting, Western, Cells, Cultured, Fluorescent Dyes chemistry, Gene Expression, Human Umbilical Vein Endothelial Cells, Humans, Hydrogen Sulfide chemistry, Luminescent Measurements standards, Nitric Oxide chemistry, Nitric Oxide Synthase genetics, Nitric Oxide Synthase metabolism, Real-Time Polymerase Chain Reaction, Reference Standards, Signal Transduction, Hydrogen Sulfide metabolism, Nitric Oxide metabolism
- Abstract
Nitric oxide (NO) and hydrogen sulfide (H2S) are two major gaseous signaling molecules that regulate diverse physiological functions. Recent publications indicate the regulatory role of H2S on NO metabolism. In this chapter, we discuss the latest findings on H2S-NO interactions through formation of novel chemical derivatives and experimental approaches to study these adducts. This chapter also addresses potential H2S interference on various NO detection techniques, along with precautions for analyzing biological samples from various sources. This information will facilitate critical evaluation and clearer insight into H2S regulation of NO signaling and its influence on various physiological functions., (© 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
30. Rho-kinase as a therapeutic target in vascular diseases: striking nitric oxide signaling.
- Author
-
Kolluru GK, Majumder S, and Chatterjee S
- Subjects
- Amides pharmacology, Amides therapeutic use, Drug Delivery Systems, Humans, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Pyridines pharmacology, Pyridines therapeutic use, Signal Transduction drug effects, Vascular Diseases drug therapy, rho-Associated Kinases antagonists & inhibitors, Nitric Oxide metabolism, Vascular Diseases enzymology, rho-Associated Kinases metabolism
- Abstract
Rho GTPases are a globular, monomeric group of small signaling G-protein molecules. Rho-associated protein kinase/Rho-kinase (ROCK) is a downstream effector protein of the Rho GTPase. Rho-kinases are the potential therapeutic targets in the treatment of cardiovascular diseases. Here, we have primarily discussed the intriguing roles of ROCK in cardiovascular health in relation to nitric oxide signaling. Further, we highlighted the biphasic effects of Y-27632, a ROCK inhibitor under shear stress, which acts as an agonist of nitric oxide production in endothelial cells. The biphasic effects of this inhibitor raised the question of safety of the drug usage in treating cardiovascular diseases., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
31. Biological activities of fusarochromanone: a potent anti-cancer agent.
- Author
-
Mahdavian E, Palyok P, Adelmund S, Williams-Hart T, Furmanski BD, Kim YJ, Gu Y, Barzegar M, Wu Y, Bhinge KN, Kolluru GK, Quick Q, Liu YY, Kevil CG, Salvatore BA, Huang S, and Clifford JL
- Subjects
- Apoptosis drug effects, Cell Line, Tumor, Cell Proliferation, Drug Screening Assays, Antitumor, Humans, Vascular Endothelial Growth Factor A antagonists & inhibitors, Vascular Endothelial Growth Factor A physiology, Antineoplastic Agents pharmacology, Chromones pharmacology
- Abstract
Background: Fusarochromanone (FC101) is a small molecule fungal metabolite with a host of interesting biological functions, including very potent anti-angiogenic and direct anti-cancer activity., Results: Herein, we report that FC101 exhibits very potent in-vitro growth inhibitory effects (IC50 ranging from 10nM-2.5 μM) against HaCat (pre-malignant skin), P9-WT (malignant skin), MCF-7 (low malignant breast), MDA-231 (malignant breast), SV-HUC (premalignant bladder), UM-UC14 (malignant bladder), and PC3 (malignant prostate) in a time-course and dose-dependent manner, with the UM-UC14 cells being the most sensitive. FC101 induces apoptosis and an increase in proportion of cells in the sub-G1 phase in both HaCat and P9-WT cell lines as evidenced by cell cycle profile analysis. In a mouse xenograft SCC tumor model, FC101 was well tolerated, non-toxic, and achieved a 30% reduction in tumor size at a dose of 8 mg/kg/day. FC101 is also a potent anti-angiogenenic agent. At nanomolar doses, FC101 inhibits the vascular endothelial growth factor-A (VEGF-A)-mediated proliferation of endothelial cells., Conclusions: Our data presented here indicates that FC101 is an excellent lead candidate for a small molecule anti-cancer agent that simultaneously affects angiogenesis signaling, cancer signal transduction, and apoptosis. Further understanding of the underlying FC101's molecular mechanism may lead to the design of novel targeted and selective therapeutics, both of which are pursued targets in cancer drug discovery.
- Published
- 2014
- Full Text
- View/download PDF
32. A comparative study of NONOate based NO donors: spermine NONOate is the best suited NO donor for angiogenesis.
- Author
-
Majumder S, Sinha S, Siamwala JH, Muley A, Reddy Seerapu H, Kolluru GK, Veeriah V, Nagarajan S, Sridhara SR, Priya MK, Kuppusamy M, Srinivasan S, Konikkat S, Soundararajan G, Venkataraman S, Saran U, and Chatterjee S
- Subjects
- Animals, Aorta metabolism, Cattle, Cells, Cultured, Chick Embryo, Egg Yolk, Endothelium, Vascular metabolism, Gene Expression Profiling, Ischemia metabolism, Male, Nitric Oxide chemistry, Rats, Rats, Wistar, Signal Transduction, Spermine chemistry, Wound Healing, Angiogenesis Inducing Agents chemistry, Neovascularization, Physiologic, Nitric Oxide Donors chemistry, Spermine analogs & derivatives
- Abstract
Nitric oxide (NO) is a known modulator of angiogenesis. The NONOate subfamily of NO donors has long been used in experimental and clinical studies to promote angiogenesis. However, no studies have been conducted yet to compare the angiogenesis potential of these NO donors in respect to their pattern of NO release. We hypothesize that having different pattern of NO release, each of the NO donors in NONOate subfamily can promote key stages of angiogenesis in differential manner. To verify our hypothesis, NO donors with half life ranging from seconds to several hours and having very different pattern of NO release were selected to evaluate their efficacy in modulating angiogenesis. Endothelial tube formation using EAhy926 cells was maximally increased by Spermine NONOate (SP) treatment. SP treatment maximally induced both ex vivo and in vivo angiogenesis using egg yolk and cotton plug angiogenesis models respectively. Experiment using chick embryo partial ischemia model revealed SP as the best suited NO donor to recover ischemia driven hampered angiogenesis. The present study elaborated that differential release pattern of NO by different NO donors can modulate angiogenesis differentially and also suggested that SP have a unique pattern of NO release that best fits for angiogenesis., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
33. Nitrite anion therapy protects against chronic ischemic tissue injury in db/db diabetic mice in a NO/VEGF-dependent manner.
- Author
-
Bir SC, Pattillo CB, Pardue S, Kolluru GK, Shen X, Giordano T, and Kevil CG
- Subjects
- Animals, Diabetes Mellitus, Type 2 metabolism, Diabetes Mellitus, Type 2 pathology, Femoral Artery drug effects, Femoral Artery metabolism, Hindlimb blood supply, Hindlimb drug effects, Ischemia complications, Ischemia metabolism, Ischemia pathology, Mice, Neovascularization, Pathologic complications, Neovascularization, Pathologic metabolism, Neovascularization, Pathologic pathology, Neovascularization, Pathologic prevention & control, Nitrates pharmacology, Nitrates therapeutic use, Sodium Nitrite pharmacology, Time Factors, Diabetes Mellitus, Type 2 complications, Ischemia drug therapy, Neovascularization, Pathologic drug therapy, Nitric Oxide metabolism, Sodium Nitrite therapeutic use, Vascular Endothelial Growth Factor A metabolism
- Abstract
Nitrite anion has been demonstrated to be a prodrug of nitric oxide (NO) with positive effects on tissue ischemia/reperfusion injury, cytoprotection, and vasodilation. However, effects of nitrite anion therapy for ischemic tissue vascular remodeling during diabetes remain unknown. We examined whether sodium nitrite therapy altered ischemic revascularization in BKS-Lepr(db/db) mice subjected to permanent unilateral femoral artery ligation. Sodium nitrite therapy completely restored ischemic hind limb blood flow compared with nitrate or PBS therapy. Importantly, delayed nitrite therapy 5 days after ischemia restored ischemic limb blood flow in aged diabetic mice. Restoration of blood flow was associated with increases in ischemic tissue angiogenesis activity and cell proliferation. Moreover, nitrite but not nitrate therapy significantly prevented ischemia-mediated tissue necrosis in aged mice. Nitrite therapy significantly increased ischemic tissue vascular endothelial growth factor (VEGF) protein expression that was essential for nitrite-mediated reperfusion of ischemic hind limbs. Nitrite significantly increased ischemic tissue NO bioavailability along with concomitant reduction of superoxide formation. Lastly, nitrite treatment also significantly stimulated hypoxic endothelial cell proliferation and migration in the presence of high glucose in an NO/VEGF-dependent manner. These results demonstrate that nitrite therapy effectively stimulates ischemic tissue vascular remodeling in the setting of metabolic dysfunction that may be clinically useful.
- Published
- 2014
- Full Text
- View/download PDF
34. Hydrogen sulfide chemical biology: pathophysiological roles and detection.
- Author
-
Kolluru GK, Shen X, Bir SC, and Kevil CG
- Subjects
- Animals, Cysteine metabolism, Humans, Mice, Nitric Oxide metabolism, Signal Transduction, Biochemistry, Hydrogen Sulfide analysis, Hydrogen Sulfide metabolism
- Abstract
Hydrogen sulfide (H2S) is the most recent endogenous gasotransmitter that has been reported to serve many physiological and pathological functions in different tissues. Studies over the past decade have revealed that H2S can be synthesized through numerous pathways and its bioavailability regulated through its conversion into different biochemical forms. H2S exerts its biological effects in various manners including redox regulation of protein and small molecular weight thiols, polysulfides, thiosulfate/sulfite, iron-sulfur cluster proteins, and anti-oxidant properties that affect multiple cellular and molecular responses. However, precise measurement of H2S bioavailability and its associated biochemical and pathophysiological roles remains less well understood. In this review, we discuss recent understanding of H2S chemical biology, its relationship to tissue pathophysiological responses and possible therapeutic uses., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
35. Intravascular radiocontrast iodixanol increases permeability of proximal tubule epithelium: a possible mechanism of contrast-induced nephropathy.
- Author
-
Yao L, Kolluru GK, Kevil CG, and Zhang WW
- Subjects
- Apoptosis drug effects, Cell Line, Cell Proliferation drug effects, Cell Survival drug effects, Claudins genetics, Claudins metabolism, Dose-Response Relationship, Drug, Epithelial Cells metabolism, Epithelial Cells pathology, Humans, Intercellular Junctions metabolism, Intercellular Junctions pathology, Kidney Tubules, Proximal metabolism, Kidney Tubules, Proximal pathology, Permeability, Up-Regulation, Contrast Media toxicity, Epithelial Cells drug effects, Intercellular Junctions drug effects, Kidney Tubules, Proximal drug effects, Triiodobenzoic Acids toxicity
- Abstract
Purpose: To investigate the effect of Iodixanol on kidney proximal tubular cell line human kidney 2 (HK-2)., Methods: The HK-2 cells were treated with Iodixanol. A Terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) assay was used to evaluate apoptosis. Cell viability was measured by proliferation assay kit. Cell permeability changes were assessed by transwell assay and intercellular gaps measurement. Expression of claudin-2 was assessed by quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR) and Western blot., Results: Iodixanol reduced tubule cell viability (P < .01) but did not cause apoptosis. The intercellular gap formation (P < .01) and transwell (P < .05) assays revealed that cell permeability significantly increased after Iodixanol treatment of monolayer cells. Western blot and qRT-PCR showed significant upregulation of claudin-2 protein (P < .05) and messenger RNA expression (P < .01)., Conclusions: Our in vitro data do not support the hypothesis that direct kidney cell death from Iodixanol is a major mechanism of contrast-induced nephropathy (CIN). However, increased permeability of proximal tubule epithelium caused by Iodixanol may play an important role in CIN.
- Published
- 2013
- Full Text
- View/download PDF
36. A tale of two gases: NO and H2S, foes or friends for life?
- Author
-
Kolluru GK, Shen X, and Kevil CG
- Subjects
- Signal Transduction, Hydrogen Sulfide metabolism, Nitric Oxide metabolism
- Abstract
Nitric oxide (NO) and hydrogen sulfide (H2S) have emerged as dominant redox regulators of numerous aspects of cellular and physiological functions within several organ systems included cardiovascular, immune and neurological tissues. Recent studies have begun to reveal that these two gaseous molecules may have redundant or overlapping pathophysiological functions often involving similar molecular targets. However, it remains less clear when and how NO and H2S may interact under biological and disease processes. In this graphical review, we discuss the current understanding of NO and H2S interactions and how they may functionally influence each other and what this may mean for biology and mechanisms of disease.
- Published
- 2013
- Full Text
- View/download PDF
37. Hydrogen sulfide stimulates ischemic vascular remodeling through nitric oxide synthase and nitrite reduction activity regulating hypoxia-inducible factor-1α and vascular endothelial growth factor-dependent angiogenesis.
- Author
-
Bir SC, Kolluru GK, McCarthy P, Shen X, Pardue S, Pattillo CB, and Kevil CG
- Subjects
- Animals, Enzyme-Linked Immunosorbent Assay, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Ischemia metabolism, Ischemia physiopathology, Male, Mice, Mice, Inbred C57BL, Regional Blood Flow, Vascular Endothelial Growth Factor A metabolism, Xanthine Oxidase, Hydrogen Sulfide administration & dosage, Hypoxia-Inducible Factor 1, alpha Subunit drug effects, Ischemia drug therapy, Neovascularization, Physiologic drug effects, Nitric Oxide Synthase drug effects, Nitrites metabolism, Vascular Endothelial Growth Factor A drug effects
- Abstract
Background: Hydrogen sulfide (H(2)S) therapy is recognized as a modulator of vascular function during tissue ischemia with the notion of potential interactions of nitric oxide (NO) metabolism. However, little is known about specific biochemical mechanisms or the importance of H(2)S activation of NO metabolism during ischemic tissue vascular remodeling. The goal of this study was to determine the effect of H(2)S on NO metabolism during chronic tissue ischemia and subsequent effects on ischemic vascular remodeling responses., Methods and Results: The unilateral, permanent femoral artery ligation model of hind-limb ischemia was performed in C57BL/6J wild-type and endothelial NO synthase-knockout mice to evaluate exogenous H(2)S effects on NO bioavailability and ischemic revascularization. We found that H(2)S selectively restored chronic ischemic tissue function and viability by enhancing NO production involving both endothelial NO synthase and nitrite reduction mechanisms. Importantly, H(2)S increased ischemic tissue xanthine oxidase activity, hind-limb blood flow, and angiogenesis, which were blunted by the xanthine oxidase inhibitor febuxostat. H(2)S treatment increased ischemic tissue and endothelial cell hypoxia-inducible factor-1α expression and activity and vascular endothelial growth factor protein expression and function in a NO-dependent manner that was required for ischemic vascular remodeling., Conclusions: These data demonstrate that H(2)S differentially regulates NO metabolism during chronic tissue ischemia, highlighting novel biochemical pathways to increase NO bioavailability for ischemic vascular remodeling.
- Published
- 2012
- Full Text
- View/download PDF
38. Redox balance dynamically regulates vascular growth and remodeling.
- Author
-
Bir SC, Kolluru GK, Fang K, and Kevil CG
- Subjects
- Humans, Oxidation-Reduction, Reactive Nitrogen Species metabolism, Reactive Oxygen Species metabolism, Signal Transduction, Neovascularization, Physiologic
- Abstract
Vascular growth and remodeling responses entail several complex biochemical, molecular, and cellular responses centered primarily on endothelial cell activation and function. Recent studies reveal that changes in endothelial cell redox status critically influence numerous cellular events that are important for vascular growth under different conditions. It has been known for some time that oxidative stress actively participates in many aspects of angiogenesis and vascular remodeling. Initial studies in this field were largely exploratory with minimal insight into specific molecular mechanisms and how these responses could be regulated. However, it is now clear that intracellular redox mechanisms involving hypoxia, NADPH oxidases (NOX), xanthine oxidase (XO), nitric oxide and its synthases, and intracellular antioxidant defense pathways collectively orchestrate a redox balance system whereby reactive oxygen and nitrogen species integrate cues controlling vascular growth and remodeling. In this review, we discuss key redox regulation pathways that are centrally important for vascular growth in tissue health and disease. Important unresolved questions and issues are also addressed that requires future investigation., (Copyright © 2012. Published by Elsevier Ltd.)
- Published
- 2012
- Full Text
- View/download PDF
39. Nitrite anion stimulates ischemic arteriogenesis involving NO metabolism.
- Author
-
Bir SC, Pattillo CB, Pardue S, Kolluru GK, Docherty J, Goyette D, Dvorsky P, and Kevil CG
- Subjects
- Angiography, Animals, Chronic Disease, Collateral Circulation drug effects, Collateral Circulation physiology, Femoral Artery drug effects, Femoral Artery physiopathology, Femoral Artery surgery, Femoral Vein drug effects, Femoral Vein physiopathology, Femoral Vein surgery, Hindlimb blood supply, Hindlimb drug effects, Hindlimb physiopathology, Ischemia drug therapy, Ischemia physiopathology, Mice, Mice, Inbred C57BL, Regional Blood Flow drug effects, Regional Blood Flow physiology, Severity of Illness Index, Neovascularization, Physiologic drug effects, Nitric Oxide metabolism, Nitric Oxide Donors administration & dosage, Prodrugs administration & dosage, Sodium Nitrite administration & dosage
- Abstract
Nitric oxide (NO) is a potential regulator of ischemic vascular remodeling, and as such therapies augmenting its bioavailability may be useful for the treatment of ischemic tissue diseases. Here we examine the effect of administering the NO prodrug sodium nitrite on arteriogenesis activity during established tissue ischemia. Chronic hindlimb ischemia was induced by permanent unilateral femoral artery and vein ligation. Five days postligation; animals were randomized to control PBS or sodium nitrite (165 μg/kg) therapy twice daily. In situ vascular remodeling was measured longitudinally using SPY angiography and Microfil vascular casting. Delayed sodium nitrite therapy rapidly increased ischemic limb arterial vessel diameter and branching in a NO-dependent manner. SPY imaging angiography over time showed that nitrite therapy enhanced ischemic gracillis collateral vessel formation from the profunda femoris to the saphenous artery. Immunofluorescent staining of smooth muscle cell actin also confirmed that sodium nitrite therapy increased arteriogenesis in a NO-dependent manner. The NO prodrug sodium nitrite significantly increases arteriogenesis and reperfusion of established severe chronic tissue ischemia.
- Published
- 2012
- Full Text
- View/download PDF
40. Endothelial dysfunction and diabetes: effects on angiogenesis, vascular remodeling, and wound healing.
- Author
-
Kolluru GK, Bir SC, and Kevil CG
- Abstract
Diabetes mellitus (DM) is a chronic metabolic disorder characterized by inappropriate hyperglycemia due to lack of or resistance to insulin. Patients with DM are frequently afflicted with ischemic vascular disease or wound healing defect. It is well known that type 2 DM causes amplification of the atherosclerotic process, endothelial cell dysfunction, glycosylation of extracellular matrix proteins, and vascular denervation. These complications ultimately lead to impairment of neovascularization and diabetic wound healing. Therapeutic angiogenesis remains an attractive treatment modality for chronic ischemic disorders including PAD and/or diabetic wound healing. Many experimental studies have identified better approaches for diabetic cardiovascular complications, however, successful clinical translation has been limited possibly due to the narrow therapeutic targets of these agents or the lack of rigorous evaluation of pathology and therapeutic mechanisms in experimental models of disease. This paper discusses the current body of evidence identifying endothelial dysfunction and impaired angiogenesis during diabetes.
- Published
- 2012
- Full Text
- View/download PDF
41. Inorganic nitrite therapy: historical perspective and future directions.
- Author
-
Kevil CG, Kolluru GK, Pattillo CB, and Giordano T
- Subjects
- Animals, Autoimmune Diseases metabolism, Humans, Inflammation, Ischemia metabolism, Mouth metabolism, Mouth microbiology, Neoplasms metabolism, Nitric Oxide metabolism, Oxidation-Reduction, Autoimmune Diseases drug therapy, Bacteria metabolism, Inorganic Chemicals therapeutic use, Ischemia drug therapy, Neoplasms drug therapy, Sodium Nitrite therapeutic use
- Abstract
Over the past several years, investigators studying nitric oxide (NO) biology and metabolism have come to learn that the one-electron oxidation product of NO, nitrite anion, serves as a unique player in modulating tissue NO bioavailability. Numerous studies have examined how this oxidized metabolite of NO can act as a salvage pathway for maintaining NO equivalents through multiple reduction mechanisms in permissive tissue environments. Moreover, it is now clear that nitrite anion production and distribution throughout the body can act in an endocrine manner to augment NO bioavailability, which is important for physiological and pathological processes. These discoveries have led to renewed hope and efforts for an effective NO-based therapeutic agent through the unique action of sodium nitrite as an NO prodrug. More recent studies also indicate that sodium nitrate may also increase plasma nitrite levels via the enterosalivary circulatory system resulting in nitrate reduction to nitrite by microorganisms found within the oral cavity. In this review, we discuss the importance of nitrite anion in several disease models along with an appraisal of sodium nitrite therapy in the clinic, potential caveats of such clinical uses, and future possibilities for nitrite-based therapies., (Copyright © 2011 Elsevier Inc. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
42. Detection of hydrogen sulfide in biological samples: current and future.
- Author
-
Kolluru GK, Shen X, and Kevil CG
- Abstract
Evaluation of: Levitt MD, Abdel-Rehim MS, Furne J. Free and acid-labile hydrogen sulfide concentrations in mouse tissues: anomalously high free hydrogen sulfide in aortic tissue. Antioxid. Redox Signal. DOI: 10.1089/ars.2010.3525 (2010) (Epub ahead of print). Hydrogen sulfide (H(2)S), known as a pungent toxic gas, has recently emerged as a novel critical mediator in the cardiovascular system, the nervous system and various biological signaling functions, as well as a therapeutic agent. However, much less certainty exists regarding biological levels of H(2)S in these systems and during disease. Many papers have reported various methods of measuring the levels of sulfide through different techniques both in vitro and in vivo. Complicating this matter is the fact that sulfide exists in multiple forms - free sulfides such as S(2) (-), HS(-), H(2)S, acid-labile and bound sulfides. These different forms of sulfide make quantitative measurement of bioavailable H(2)S difficult and have led to variable reported levels in the literature. The sensitive detection of sulfide in its multiple forms is needed to establish reliable bioavailable concentrations of sulfide species in order to understand their role in various aspects of physiology and pathology, and to address possible therapeutic approaches. A recent method by Levitt et al. describes a unique gas chromatography chemiluminescence-based technique to measure free and acid-labile H(2)S in multiple tissues from mouse.
- Published
- 2011
- Full Text
- View/download PDF
43. eNOS phosphorylation in health and disease.
- Author
-
Kolluru GK, Siamwala JH, and Chatterjee S
- Subjects
- Animals, Humans, Models, Biological, Nitric Oxide metabolism, Phosphorylation, Serine metabolism, Nitric Oxide Synthase Type III metabolism
- Abstract
Endothelium plays a fundamental role in maintaining the vascular tone by releasing various biochemical factors that modulate the contractile and relaxatory behavior of the underlying vascular smooth muscle, regulation of inflammation, immunomodulation, platelet aggregation, and thrombosis. Endothelium regulates these cellular processes by activating endothelial nitric oxide synthase (eNOS) responsible for nitric oxide (NO) production. eNOS is constitutively expressed in ECs in response to humoral, mechanical or pharmacological stimulus. eNOS activity is regulated mainly by protein-protein interactions and multisite phosphorylations. The phosphorylation state of specific serine, threonine and tyrosine residues of the enzyme plays a pivotal role in regulation of eNOS activity. Perturbations of eNOS phosphorylation have been reported in a number of diseases thereby emphasizing the importance of regulation of eNOS activity. This review summarizes the mechanism of eNOS regulation through multi-site phosphorylation in different pathologies. Attempts have been made to highlight phosphorylation of eNOS at various residues, regulation of the enzyme activity via posttranslational modifications and its implications on health and disease., (Copyright (c) 2010 Elsevier Masson SAS. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
44. Simulated microgravity promotes nitric oxide-supported angiogenesis via the iNOS-cGMP-PKG pathway in macrovascular endothelial cells.
- Author
-
Siamwala JH, Majumder S, Tamilarasan KP, Muley A, Reddy SH, Kolluru GK, Sinha S, and Chatterjee S
- Subjects
- Animals, Cattle, Cell Movement, Cell Proliferation, Chickens, Endothelial Cells cytology, Humans, Nitric Oxide Synthase Type III metabolism, Signal Transduction, Sus scrofa, Weightlessness, Wound Healing, Cyclic GMP metabolism, Cyclic GMP-Dependent Protein Kinases metabolism, Endothelial Cells enzymology, Neovascularization, Physiologic, Nitric Oxide metabolism, Nitric Oxide Synthase Type II metabolism, Weightlessness Simulation
- Abstract
Angiogenesis is a physiological process involving the growth of blood vessel in response to specific stimuli. The present study shows that limited microgravity treatments induce angiogenesis by activating macrovascular endothelial cells. Inhibition of nitric oxide production using pharmacological inhibitors and inducible nitric oxide synthase (iNOS) small interfering ribo nucleic acid (siRNA) abrogated microgravity induced nitric oxide production in macrovascular cells. The study further delineates that iNOS acts as a molecular switch for the heterogeneous effects of microgravity on macrovascular, endocardial and microvascular endothelial cells. Further dissection of nitric oxide downstream signaling confirms that simulated microgravity induces angiogenesis via the cyclic guanosine monophosphate (cGMP)-PKG dependent pathway., (Copyright (c) 2010 Federation of European Biochemical Societies. Published by Elsevier B.V. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
45. Inhibition of dynamin-2 confers endothelial barrier dysfunctions by attenuating nitric oxide production.
- Author
-
Seerapu Hb, Subramaniam GP, Majumder S, Sinha S, Bisana S, Mahajan S, Kolluru GK, Muley A, Siamwala JH, Illavazagan G, and Chatterjee S
- Subjects
- Antioxidants metabolism, Cell Line, Dynamin II metabolism, Endothelial Cells cytology, Endothelial Cells metabolism, Endothelium, Vascular cytology, Humans, Hypoxia metabolism, Nitric Oxide Donors metabolism, Nitric Oxide Synthase Type III metabolism, Nitroprusside metabolism, Peroxynitrous Acid metabolism, Uric Acid metabolism, Capillary Permeability physiology, Dynamin II antagonists & inhibitors, Endothelium, Vascular metabolism, Nitric Oxide biosynthesis
- Abstract
Hypoxia induces barrier dysfunctions in endothelial cells. Nitric oxide is an autacoid signalling molecule that confers protection against hypoxia-mediated barrier dysfunctions. Dyn-2 (dynamin-2), a large GTPase and a positive modulator of eNOS (endothelial nitric oxide synthase), plays an important role in maintaining vascular homeostasis. The present study aims to elucidate the role of dyn-2 in hypoxia-mediated leakiness of the endothelial monolayer in relation to redox milieu. Inhibition of dyn-2 by transfecting the cells with K44A, a dominant negative construct of dyn-2, elevated leakiness of the endothelial monolayer under hypoxia. Sodium nitroprusside (nitric oxide donor) and uric acid (peroxynitrite quencher) were used to evaluate the role of nitric oxide and peroxynitrite in maintaining endothelial barrier functions under hypoxia. Administration of nitric oxide and uric acid recovered hypoxia-mediated leakiness of K44A-overexpressed endothelial monolayer. Our study confirms that inhibition of dyn-2 induces leakiness in the endothelial monolayer by increasing the load of peroxynitrite under hypoxia.
- Published
- 2010
- Full Text
- View/download PDF
46. Simulated microgravity perturbs actin polymerization to promote nitric oxide-associated migration in human immortalized Eahy926 cells.
- Author
-
Siamwala JH, Reddy SH, Majumder S, Kolluru GK, Muley A, Sinha S, and Chatterjee S
- Subjects
- Androstadienes pharmacology, Bradykinin pharmacology, Cell Line, Cell Line, Transformed, Cell Surface Extensions drug effects, Cell Surface Extensions metabolism, Endothelial Cells drug effects, Endothelial Cells enzymology, Enzyme Inhibitors pharmacology, Humans, Mitogen-Activated Protein Kinases metabolism, Models, Biological, Neovascularization, Physiologic drug effects, Nitric Oxide biosynthesis, Nitric Oxide Synthase Type III antagonists & inhibitors, Phosphoinositide-3 Kinase Inhibitors, Wortmannin, Wound Healing drug effects, Actins metabolism, Cell Movement drug effects, Endothelial Cells cytology, Endothelial Cells metabolism, Nitric Oxide metabolism, Weightlessness Simulation instrumentation
- Abstract
Microgravity causes endothelium dysfunctions and vascular endothelium remodeling in astronauts returning from space flight. Cardiovascular deconditioning occurs as a consequence of an adaptive response to microgravity partially due to the effects exerted at cellular level. Directional migration of endothelial cell which are central in maintaining the structural integrity of vascular walls is regulated by chemotactic, haptotactic, and mechanotactic stimuli which are essential for vasculogenesis. We explored the migration property of transformed endothelial cells (EC) exposed to 2-h microgravity, simulated using a three-dimensional clinostat constructed based on blueprint published by the Fokker Space, Netherlands. Migration of EC was measured using the scrap wound healing in the presence or absence of actin polymerization inhibitor-cytochalasin D (CD) in Eahy926 cell lines. Simulated microgravity increased cellular migration by 25% while CD-blocked microgravity induced cellular migration. The key migratory structures of cells, filopodia and lamellipodia, formed by EC were more in simulated microgravity compared to gravity. Parallel experiments with phalloidin and diaminorhodamine-4M (DAR-4M) showed that simulated microgravity caused actin rearrangements that lead to 25% increase in nitric oxide production. Further nitric oxide measurements showed a higher nitric oxide production which was not abrogated by phosphoinositol 3 kinase inhibitor (Wortmanin). Bradykinin, an inducer of nitric oxide, prompted two folds higher nitric oxide production along with simulated microgravity in a synergistic manner. We suggest that limited exposure to simulated microgravity increases Eahy926 cell migration by modulating actin and releasing nitric oxide.
- Published
- 2010
- Full Text
- View/download PDF
47. Shear stress promotes nitric oxide production in endothelial cells by sub-cellular delocalization of eNOS: A basis for shear stress mediated angiogenesis.
- Author
-
Kolluru GK, Sinha S, Majumder S, Muley A, Siamwala JH, Gupta R, and Chatterjee S
- Subjects
- Animals, Chick Embryo, Endothelial Cells metabolism, Endothelial Cells ultrastructure, Endothelium-Dependent Relaxing Factors metabolism, Humans, Phosphorylation, Endothelial Cells enzymology, Neovascularization, Physiologic, Nitric Oxide biosynthesis, Nitric Oxide Synthase Type III analysis, Nitric Oxide Synthase Type III metabolism, Stress, Mechanical
- Abstract
This study aims to investigate the role of shear stress in cellular remodeling and angiogenesis with relation to nitric oxide (NO). We observed a 2-fold increase in endothelial cell (EC) migration in relation to actin re-arrangements under 15 dyne/cm(2) shear stress. Blocking NO production inhibited the migration and ring formation of ECs by 6-fold and 5-fold, respectively under shear stress. eNOS-siRNA knockdown technique also ascertained a 3-fold reduction in shear stress mediated ring formation. In ovo artery ligation model with a half and complete flow block for 30 min showed a reduction of angiogenesis by 50% and 70%, respectively. External stimulation with NO donor showed a 2-fold recovery in angiogenesis under both half and complete flow block conditions. NO intensity clustering studies by using Diaminofluorescein diacetate (DAF-2DA) probed endothelial monolayer depicted pattern-changes in NO distribution and cluster formation of ECs under shear stress. Immunofluorescence and live cell studies revealed an altered sub-cellular localization pattern of eNOS and phospho-eNOS under shear stress. In conclusion, shear-induced angiogenesis is mediated by nitric oxide dependent EC migration., (Copyright 2010 Elsevier Inc. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
48. Secreted frizzled-related protein 4: an angiogenesis inhibitor.
- Author
-
Muley A, Majumder S, Kolluru GK, Parkinson S, Viola H, Hool L, Arfuso F, Ganss R, Dharmarajan A, and Chatterjee S
- Subjects
- Animals, Biological Assay, Calcium metabolism, Cell Line, Tumor, Cell Polarity, Chickens, Female, Humans, Mice, Mice, Inbred BALB C, Models, Biological, Oxidation-Reduction, Rats, Rats, Wistar, Reactive Oxygen Species metabolism, Signal Transduction, Sus scrofa, Wnt Proteins metabolism, Xenograft Model Antitumor Assays, beta Catenin metabolism, Angiogenesis Inhibitors metabolism, Frizzled Receptors metabolism, Neovascularization, Pathologic metabolism
- Abstract
Wnt signaling is involved in developmental processes, cell proliferation, and cell migration. Secreted frizzled-related protein 4 (sFRP4) has been demonstrated to be a Wnt antagonist; however, its effects on endothelial cell migration and angiogenesis have not yet been reported. Using various in vitro assays, we show that sFRP4 inhibits endothelial cell migration and the development of sprouts and pseudopodia as well as disrupts the stability of endothelial rings in addition to inhibiting proliferation. sFRP4 interfered with endothelial cell functions by antagonizing the canonical Wnt/beta-catenin signaling pathway and the Wnt/planar cell polarity pathway. Furthermore, sFRP4 blocked the effect of vascular endothelial growth factor on endothelial cells. sFRP4 also selectively induced apoptotic events in endothelial cells by increasing cellular levels of reactive oxygen species. In vivo assays demonstrated a reduction in vascularity after sFRP4 treatment. Most importantly, sFRP4 restricted tumor growth in mice by interfering with endothelial cell function. The data demonstrate sFRP4 to be a potent angiogenesis inhibitor that warrants further investigation as a therapeutic agent in the control of angiogenesis-associated pathology.
- Published
- 2010
- Full Text
- View/download PDF
49. Thalidomide attenuates nitric oxide-driven angiogenesis by interacting with soluble guanylyl cyclase.
- Author
-
Majumder S, Rajaram M, Muley A, Reddy HS, Tamilarasan KP, Kolluru GK, Sinha S, Siamwala JH, Gupta R, Ilavarasan R, Venkataraman S, Sivakumar KC, Anishetty S, Kumar PG, and Chatterjee S
- Subjects
- Angiogenesis Inhibitors administration & dosage, Animals, Catalytic Domain drug effects, Cattle, Cells, Cultured, Cyclic GMP metabolism, Dose-Response Relationship, Drug, Drug Delivery Systems, Endothelial Cells drug effects, Endothelial Cells metabolism, Endothelium, Vascular drug effects, Endothelium, Vascular metabolism, Guanylate Cyclase metabolism, Humans, Male, Models, Molecular, Rats, Rats, Wistar, Receptors, Cytoplasmic and Nuclear metabolism, Soluble Guanylyl Cyclase, Thalidomide administration & dosage, Umbilical Veins, Wound Healing drug effects, Angiogenesis Inhibitors pharmacology, Guanylate Cyclase drug effects, Neovascularization, Physiologic drug effects, Nitric Oxide metabolism, Receptors, Cytoplasmic and Nuclear drug effects, Thalidomide pharmacology
- Abstract
Background and Purpose: Nitric oxide (NO) promotes angiogenesis by activating endothelial cells. Thalidomide arrests angiogenesis by interacting with the NO pathway, but its putative targets are not known. Here, we have attempted to identify these targets., Experimental Approach: Cell-based angiogenesis assays (wound healing of monolayers and tube formation in ECV304, EAhy926 and bovine arterial endothelial cells), along with ex vivo and in vivo angiogenesis assays, were used to explore interactions between thalidomide and NO. We also carried out in silico homology modelling and docking studies to elucidate possible molecular interactions of thalidomide and soluble guanylyl cyclase (sGC)., Key Results: Thalidomide inhibited pro-angiogenic functions in endothelial cell cultures, whereas 8-bromo-cGMP, sildenafil (a phosphodiesterase inhibitor) or a NO donor [sodium nitroprusside (SNP)] increased these functions. The inhibitory effects of thalidomide were reversed by adding 8-bromo-cGMP or sildenafil, but not by SNP. Immunoassays showed a concentration-dependent decrease of cGMP in endothelial cells with thalidomide, without affecting the expression level of sGC protein. These results suggested that thalidomide inhibited the activity of sGC. Molecular modelling and docking experiments revealed that thalidomide could interact with the catalytic domain of sGC, which would explain the inhibitory effects of thalidomide on NO-dependent angiogenesis., Conclusion and Implications: Our results showed that thalidomide interacted with sGC, suppressing cGMP levels in endothelial cells, thus exerting its anti-angiogenic effects. These results could lead to the formulation of thalidomide-based drugs to curb angiogenesis by targeting sGC.
- Published
- 2009
- Full Text
- View/download PDF
50. Cadmium attenuates bradykinin-driven nitric oxide production by interplaying with the localization pattern of endothelial nitric oxide synthase.
- Author
-
Majumder S, Gupta R, Reddy H, Sinha S, Muley A, Kolluru GK, and Chatterjee S
- Subjects
- Animals, Cattle, Cells, Cultured, Endothelium, Vascular cytology, Endothelium, Vascular enzymology, Endothelium, Vascular metabolism, Bradykinin physiology, Cadmium toxicity, Endothelium, Vascular drug effects, Nitric Oxide biosynthesis, Nitric Oxide Synthase Type III metabolism
- Abstract
Cadmium, a ubiquitous heavy metal, interferes with endothelial functions and angiogenesis. Bradykinin is a Ca-mobilizing soluble peptide that acts via nitric oxide to promote vasodilation and capillary permeability. The objective of the present study was to explore the Cd implications in bradykinin-dependent endothelial functions. An egg yolk angiogenesis model was employed to evaluate the effect of Cd on bradykinin-induced angiogenesis. The results demonstrate that 100 nmol/L Cd attenuated bradykinin-dependent angiogenesis. The results of the in vitro wound healing and tube formation assays by using EAhy 926, a transformed endothelial cell line, suggest that Cd blocked bradykinin-mediated endothelial migration and tube formation by 38% and 67%, respectively, while nitric oxide supplementation could reverse the effect of Cd on bradykinin-induced endothelial migration by 94%. The detection of nitric oxide by using a DAF-2DA fluorescent probe, Griess assay, and ultrasensitive electrode suggests that Cd blocked bradykinin-induced nitric oxide production. Fluorescence imaging of eNOS-GFP transfected endothelial cells, immunofluorescence, and Western blot studies of Cd and bradykinin-treated cells show that Cd interfered with the localization pattern of eNOS, which possibly attenuates nitric oxide production in part. Additionally, Ca imaging of Cd- and bradykinin-treated cells suggests that Cd blocked bradykinin-dependent Ca influx into the cells, thus partially blocking Ca-dependent nitric oxide production in endothelial cells. The results of this study conclude that Cd blunted the effect of bradykinin by interfering with the Ca-associated NOS activity specifically by impeding subcellular trafficking of eNOS.
- Published
- 2009
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.