110 results on '"David J. Fulton"'
Search Results
2. Transgenic Overexpression of HDAC9 Promotes Adipocyte Hypertrophy, Insulin Resistance and Hepatic Steatosis in Aging Mice
- Author
-
Praneet Veerapaneni, Brandee Goo, Samah Ahmadieh, Hong Shi, David S. Kim, Mourad Ogbi, Stephen Cave, Ronnie Chouhaita, Nicole Cyriac, David J. Fulton, Alexander D. Verin, Weiqin Chen, Yun Lei, Xin-Yun Lu, Ha Won Kim, and Neal L. Weintraub
- Subjects
HDAC9 ,overexpression ,insulin resistance ,adipose tissue ,liver ,Microbiology ,QR1-502 - Abstract
Histone deacetylase (HDAC) 9 is a negative regulator of adipogenic differentiation, which is required for maintenance of healthy adipose tissues. We reported that HDAC9 expression is upregulated in adipose tissues during obesity, in conjunction with impaired adipogenic differentiation, adipocyte hypertrophy, insulin resistance, and hepatic steatosis, all of which were alleviated by global genetic deletion of Hdac9. Here, we developed a novel transgenic (TG) mouse model to test whether overexpression of Hdac9 is sufficient to induce adipocyte hypertrophy, insulin resistance, and hepatic steatosis in the absence of obesity. HDAC9 TG mice gained less body weight than wild-type (WT) mice when fed a standard laboratory diet for up to 40 weeks, which was attributed to reduced fat mass (primarily inguinal adipose tissue). There was no difference in insulin sensitivity or glucose tolerance in 18-week-old WT and HDAC9 TG mice; however, at 40 weeks of age, HDAC9 TG mice exhibited impaired insulin sensitivity and glucose intolerance. Tissue histology demonstrated adipocyte hypertrophy, along with reduced numbers of mature adipocytes and stromovascular cells, in the HDAC9 TG mouse adipose tissue. Moreover, increased lipids were detected in the livers of aging HDAC9 TG mice, as evaluated by oil red O staining. In conclusion, the experimental aging HDAC9 TG mice developed adipocyte hypertrophy, insulin resistance, and hepatic steatosis, independent of obesity. This novel mouse model may be useful in the investigation of the impact of Hdac9 overexpression associated with metabolic and aging-related diseases.
- Published
- 2024
- Full Text
- View/download PDF
3. Elevated Cytokine Levels in Plasma of Patients with SARS-CoV-2 Do Not Contribute to Pulmonary Microvascular Endothelial Permeability
- Author
-
Anita Kovacs-Kasa, Abdelrahman A. Zaied, Silvia Leanhart, Murat Koseoglu, Supriya Sridhar, Rudolf Lucas, David J. Fulton, Jose A. Vazquez, and Brian H. Annex
- Subjects
SARS-CoV-2 ,barrier dysfunction ,endothelial permeability ,plasma ,cytokine ,complements factors ,Microbiology ,QR1-502 - Abstract
ABSTRACT The vascular endothelial injury occurs in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections, but the mechanisms are poorly understood. We sought to determine the frequency and type of cytokine elevations and their relationship to endothelial injury induced by plasma from patients with SARS-CoV-2 versus controls. Plasma from eight consecutively enrolled patients hospitalized with acute SARS-CoV-2 infection was compared to controls. Endothelial cell (EC) barrier integrity was evaluated using ECIS (electric cell-substrate impedance sensing) on human lung microvascular EC. Plasma from all SARS-CoV-2 but none from controls decreased transendothelial resistance to a greater degree than that produced by tumor necrosis factor-alpha (TNF-α), the positive control for the assay. Thrombin, angiopoietin 2 (Ang2), and vascular endothelial growth factor (VEGF), complement factor C3a and C5a, and spike protein increased endothelial permeability, but to a lesser extent and a shorter duration when compared to SARS-CoV-2 plasma. Analysis of Ang2, VEGF, and 15 cytokines measured in plasma revealed striking patient-to-patient variability within the SARS-CoV-2 patients. Pretreatment with thrombin inhibitors, single, or combinations of neutralizing antibodies against cytokines, Ca3 and C5a receptor antagonists, or with ACE2 antibody failed to lessen the SARS-CoV-2 plasma-induced EC permeability. The EC barrier destructive effects of plasma from patients with SARS-CoV-2 were susceptible to heat inactivation. Plasma from patients hospitalized with acute SARS-CoV-2 infection uniformly disrupts lung microvascular integrity. No predicted single, or set of, cytokine(s) accounted for the enhanced vascular permeability, although the factor(s) were heat-labile. A still unidentified but potent circulating factor(s) appears to cause the EC disruption in SARS-CoV-2 infected patients. IMPORTANCE Lung vascular endothelial injury in SARS-CoV-2 patients is one of the most important causes of morbidity and mortality and has been linked to more severe complications including acute respiratory distress syndrome (ARDS) and subsequent death due to multiorgan failure. We have demonstrated that in eight consecutive patients with SARS-CoV-2, who were not selected for evidence of endothelial injury, the diluted plasma-induced intense lung microvascular damage, in vitro. Known endothelial barrier-disruptive agents and proposed mediators of increased endothelial permeability in SARS-CoV-2, induced changes in permeability that were smaller in magnitude and shorter in duration than plasma from patients with SARS-CoV-2. The effect on endothelial cell permeability of plasma from patients with SARS-CoV-2 was heat-labile. The main plasma factor that causes the increased endothelial permeability remains to be identified. Our study provides a possible approach for future studies to understand the underlying mechanisms leading to vascular injury in SARS-CoV-2 infections.
- Published
- 2022
- Full Text
- View/download PDF
4. Prkaa1 Metabolically Regulates Monocyte/Macrophage Recruitment and Viability in Diet-Induced Murine Metabolic Disorders
- Author
-
Qiuhua Yang, Qian Ma, Jiean Xu, Zhiping Liu, Jianqiu Zou, Jian Shen, Yaqi Zhou, Qingen Da, Xiaoxiao Mao, Sarah Lu, David J. Fulton, Neal L. Weintraub, Zsolt Bagi, Mei Hong, and Yuqing Huo
- Subjects
AMPKα1/PRKAA1 ,glycolysis ,monocyte recruitment ,macrophage viability ,metabolic disorders ,Biology (General) ,QH301-705.5 - Abstract
Myeloid cells, including monocytes/macrophages, primarily rely on glucose and lipid metabolism to provide the energy and metabolites needed for their functions and survival. AMP-activated protein kinase (AMPK, its gene is PRKA for human, Prka for rodent) is a key metabolic sensor that regulates many metabolic pathways. We studied recruitment and viability of Prkaa1-deficient myeloid cells in mice and the phenotype of these mice in the context of cardio-metabolic diseases. We found that the deficiency of Prkaa1 in myeloid cells downregulated genes for glucose and lipid metabolism, compromised glucose and lipid metabolism of macrophages, and suppressed their recruitment to adipose, liver and arterial vessel walls. The viability of macrophages in the above tissues/organs was also decreased. These cellular alterations resulted in decreases in body weight, insulin resistance, and lipid accumulation in liver of mice fed with a high fat diet, and reduced the size of atherosclerotic lesions of mice fed with a Western diet. Our results indicate that AMPKα1/PRKAA1-regulated metabolism supports monocyte recruitment and macrophage viability, contributing to the development of diet-induced metabolic disorders including diabetes and atherosclerosis.
- Published
- 2021
- Full Text
- View/download PDF
5. PRKAA1/AMPKα1-driven glycolysis in endothelial cells exposed to disturbed flow protects against atherosclerosis
- Author
-
Qiuhua Yang, Jiean Xu, Qian Ma, Zhiping Liu, Varadarajan Sudhahar, Yapeng Cao, Lina Wang, Xianqiu Zeng, Yaqi Zhou, Min Zhang, Yiming Xu, Yong Wang, Neal L. Weintraub, Chunxiang Zhang, Tohru Fukai, Chaodong Wu, Lei Huang, Zhen Han, Tao Wang, David J. Fulton, Mei Hong, and Yuqing Huo
- Subjects
Science - Abstract
Increased glycolysis and inflammatory responses have been observed in endothelial cells exposed to disturbed flow. However, the role of endothelial glycolysis in atherosclerosis is unclear. Here the authors unveil a protective role for glycolysis by showing that endothelial deletion of Prkaa1 accelerates atherosclerosis in hyperlipidemic mice through a reduction of glycolytic metabolism.
- Published
- 2018
- Full Text
- View/download PDF
6. Regulation of endothelial intracellular adenosine via adenosine kinase epigenetically modulates vascular inflammation
- Author
-
Yiming Xu, Yong Wang, Siyuan Yan, Qiuhua Yang, Yaqi Zhou, Xianqiu Zeng, Zhiping Liu, Xiaofei An, Haroldo A. Toque, Zheng Dong, Xuejun Jiang, David J. Fulton, Neal L. Weintraub, Qinkai Li, Zsolt Bagi, Mei Hong, Detlev Boison, Chaodong Wu, and Yuqing Huo
- Subjects
Science - Abstract
The molecular mechanisms underlying vascular inflammation are unclear. Here the authors show that pro-inflammatory stimuli lead to endothelial inflammation by increasing adenosine kinase expression, and that its knockdown in endothelial cells inhibits atherosclerosis and cerebral ischemic injury in mice.
- Published
- 2017
- Full Text
- View/download PDF
7. Endothelial adenosine A2a receptor-mediated glycolysis is essential for pathological retinal angiogenesis
- Author
-
Zhiping Liu, Siyuan Yan, Jiaojiao Wang, Yiming Xu, Yong Wang, Shuya Zhang, Xizhen Xu, Qiuhua Yang, Xianqiu Zeng, Yaqi Zhou, Xuejiao Gu, Sarah Lu, Zhongjie Fu, David J. Fulton, Neal L. Weintraub, Ruth B. Caldwell, Wenbo Zhang, Chaodong Wu, Xiao-Ling Liu, Jiang-Fan Chen, Aftab Ahmad, Ismail Kaddour-Djebbar, Mohamed Al-Shabrawey, Qinkai Li, Xuejun Jiang, Ye Sun, Akrit Sodhi, Lois Smith, Mei Hong, and Yuqing Huo
- Subjects
Science - Abstract
Pathological angiogenesis in the retina is a major cause of blindness. Here the authors show that adenosine receptor A2A drives pathological angiogenesis in the oxygen-induced retinopathy mouse model by promoting glycolysis in endothelial cells via the ERK/Akt/HIF-1α pathway, thereby suggesting new therapeutic targets for disease treatment.
- Published
- 2017
- Full Text
- View/download PDF
8. Disruption of Immune Homeostasis in Human Dendritic Cells via Regulation of Autophagy and Apoptosis by Porphyromonas gingivalis
- Author
-
Mohamed M. Meghil, Omnia K. Tawfik, Mahmoud Elashiry, Mythilypriya Rajendran, Roger M. Arce, David J. Fulton, Patricia V. Schoenlein, and Christopher W. Cutler
- Subjects
(MeSH) dendritic cells ,Porphyromonas gingivalis ,autophagy ,apoptosis ,periodontitis ,Immunologic diseases. Allergy ,RC581-607 - Abstract
As fundamental processes of immune homeostasis, autophagy, and apoptosis must be maintained to mitigate risk of chronic inflammation and autoimmune diseases. Periodontitis is a chronic inflammatory disease characterized by oral microbial dysbiosis, and dysregulation of dendritic cell (DC) and T cell responses. The aim of this study was to elucidate the underlying mechanisms by which the oral microbe Porphyromonas gingivalis (P. gingivalis) manipulates dendritic cell signaling to perturb both autophagy and apoptosis. Using a combination of Western blotting, flow cytometry, qRT-PCR and immunofluorescence analysis, we show a pivotal role for the minor (Mfa1) fimbriae of P. gingivalis in nuclear/cytoplasmic shuttling of Akt and FOXO1 in human monocyte-derived DCs. Mfa1-induced Akt nuclear localization and activation ultimately induced mTOR. Activation of the Akt/mTOR axis downregulated intracellular LC3II, also known as Atg8, required for autophagosome formation and maturation. Use of allosteric panAkt inhibitor MK2206 and mTOR inhibitor rapamycin confirmed the role of Akt/mTOR signaling in autophagy inhibition by P. gingivalis in DCs. Interestingly, this pathway was also linked to induction of the anti-apoptotic protein Bcl2, decreased caspase-3 cleavage and decreased expression of pro-apoptotic proteins Bax and Bim, thus promoting longevity of host DCs. Addition of ABT-199 peptide to disrupt the interaction of antiapoptotic Bcl2 and its proapoptotic partners BAK/BAX restored apoptotic death to P. gingivalis-infected DC cells. In summary, we have identified the underlying mechanism by which P. gingivalis promotes its own survival and that of its host DCs.
- Published
- 2019
- Full Text
- View/download PDF
9. Increased Muscle Mass Protects Against Hypertension and Renal Injury in Obesity
- Author
-
Joshua T. Butcher, James D. Mintz, Sebastian Larion, Shuiqing Qiu, Ling Ruan, David J. Fulton, and David W. Stepp
- Subjects
hyperglycemia ,hypertension ,myostatin ,nicotinamide‐adenine dinucleotide phosphate, reduced form, oxidase 4 ,skeletal muscle ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
Background Obesity compromises cardiometabolic function and is associated with hypertension and chronic kidney disease. Exercise ameliorates these conditions, even without weight loss. Although the mechanisms of exercise's benefits remain unclear, augmented lean body mass is a suspected mechanism. Myostatin is a potent negative regulator of skeletal muscle mass that is upregulated in obesity and downregulated with exercise. The current study tested the hypothesis that deletion of myostatin would increase muscle mass and reduce blood pressure and kidney injury in obesity. Methods and Results Myostatin knockout mice were crossed to db/db mice, and metabolic and cardiovascular functions were examined. Deletion of myostatin increased skeletal muscle mass by ≈50% to 60% without concomitant weight loss or reduction in fat mass. Increased blood pressure in obesity was prevented by the deletion of myostatin, but did not confer additional benefit against salt loading. Kidney injury was evident because of increased albuminuria, which was abolished in obese mice lacking myostatin. Glycosuria, total urine volume, and whole kidney NOX‐4 levels were increased in obesity and prevented by myostatin deletion, arguing that increased muscle mass provides a multipronged defense against renal dysfunction in obese mice. Conclusions These experimental observations suggest that loss of muscle mass is a novel risk factor in obesity‐derived cardiovascular dysfunction. Interventions that increase muscle mass, either through exercise or pharmacologically, may help limit cardiovascular disease in obese individuals.
- Published
- 2018
- Full Text
- View/download PDF
10. Effect of myostatin deletion on cardiac and microvascular function
- Author
-
Joshua T. Butcher, M. Irfan Ali, Merry W. Ma, Cameron G. McCarthy, Bianca N. Islam, Lauren G. Fox, James D. Mintz, Sebastian Larion, David J. Fulton, and David W. Stepp
- Subjects
Augmented muscle mass ,cardiac function ,coronary microvasculature ,exercise ,myostatin ,nitric oxide ,Physiology ,QP1-981 - Abstract
Abstract The objective of this study is to test the hypothesis that increased muscle mass has positive effects on cardiovascular function. Specifically, we tested the hypothesis that increases in lean body mass caused by deletion of myostatin improves cardiac performance and vascular function. Echocardiography was used to quantify left ventricular function at baseline and after acute administration of propranolol and isoproterenol to assess β‐adrenergic reactivity. Additionally, resistance vessels in several beds were removed, cannulated, pressurized to 60 mmHg and reactivity to vasoactive stimuli was assessed. Hemodynamics were measured using in vivo radiotelemetry. Myostatin deletion results in increased fractional shortening at baseline. Additionally, arterioles in the coronary and muscular microcirculations are more sensitive to endothelial‐dependent dilation while nonmuscular beds or the aorta were unaffected. β‐adrenergic dilation was increased in both coronary and conduit arteries, suggesting a systemic effect of increased muscle mass on vascular function. Overall hemodynamics and physical characteristics (heart weight and size) remained unchanged. Myostatin deletion mimics in part the effects of exercise on cardiovascular function. It significantly increases lean muscle mass and results in muscle‐specific increases in endothelium‐dependent vasodilation. This suggests that increases in muscle mass may serve as a buffer against pathological states that specifically target cardiac function (heart failure), the β‐adrenergic system (age), and nitric oxide bio‐availability (atherosclerosis). Taken together, pharmacological inhibition of the myostatin pathway could prove an excellent mechanism by which the benefits of exercise can be conferred in patients that are unable to exercise.
- Published
- 2017
- Full Text
- View/download PDF
11. SPARC Ameliorates Ovarian Cancer-Associated Inflammation
- Author
-
Neveen A. Said, Ahmed A. Elmarakby, John D. Imig, David J. Fulton, and Kouros Motamed
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
We have recently identified that the role of secreted protein acidic and rich in cysteine (SPARC) in amelioration of peritoneal ovarian carcinomatosis is mediated, at least in part, through mesothelial cell/lysophosphatidic acid-induced inflammatory response in ovarian cancer cells. The aim of this study was to elucidate the molecular mechanisms of the interactions between tumor cells and the cellular components of the ovarian cancer peritoneal microenvironment, specifically, mesothelial cells and macrophages. We found that SPARC not only significantly reduced macrophage chemoattractant protein-1 production and its macrophage chemotactic effect, but also attenuated the response of ovarian cancer cells to the mitogenic and proinvasive effects of macrophage chemo-attractant protein-1 and decreased macrophage-induced cancer cell invasiveness. Overexpression of SPARC in ovarian cancer cells significantly attenuated macrophage- and mesothelial cell-induced production and activity of interleukin-6, prostanoids (prostaglandins E2 and 8-isoprostanes) as well as matrix metalloproteinases and urokinase plasminogen activator. Moreover, the effects of SPARC overexpression in ovarian cancer cells were mediated, in part, through inhibition of nuclear factor-κB promoter activation. These results indicate, for the first time, that the effects of tumor SPARC as a negative regulator of ovarian cancer are mediated through decreased recruitment of macrophages and downregulation of the associated inflammation.
- Published
- 2008
- Full Text
- View/download PDF
12. SPARC Inhibits LPA-Mediated Mesothelial—Ovarian Cancer Cell Crosstalk
- Author
-
Neveen A. Said, Ida Najwer, Matthew J. Socha, David J. Fulton, Samuel C. Mok, and Kouros Motamed
- Subjects
SPARC ,LPA ,IL-6 ,ovarian cancer ,mesothelial cells ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
The interplay between peritoneal mesothelial cells and ovarian cancer cells is critical for the initiation and peritoneal dissemination of, and ascites formation in, ovarian cancer. The production of lysophosphatidic acid (LPA) by both peritoneal mesothelial cells and ovarian cancer cells has been shown to promote metastatic phenotype in ovarian cancer. Herein, we report that exogenous addition or ectopic overexpression of the matricellular protein SPARC (secreted protein acidic and rich in cysteine) significantly attenuated LPA-induced proliferation, chemotaxis, and invasion in both highly metastatic SKOV3 and less metastatic OVCAR3 ovarian cancer cell lines. SPARC appears to modulate these functions, at least in part, through the regulation of LPA receptor levels and the attenuation of extracellular signal-regulated kinase (ERK) 1/2 and protein kinase B/AKT signaling. Moreover, our results show that SPARC not only significantly inhibited both basal and LPA-induced interleukin (IL) 6 production in both cell lines but also attenuated IL-6-induced mitogenic, chemotactic, and proinvasive effects, in part, through significant suppression of ERK1/2 and, to a lesser extent, of signal transducers and activators of transcription 3 signaling pathways. Our results strongly suggest that SPARC exerts a dual inhibitory effect on LPA-induced mesothelial-ovarian cancer cell crosstalk through the regulation of both LPA-induced IL-6 production and function. Taken together, our findings underscore the use of SPARC as a potential therapeutic candidate in peritoneal ovarian carcinomatosis.
- Published
- 2007
- Full Text
- View/download PDF
13. The biological clock enhancer nobiletin ameliorates steatosis in genetically obese mice by restoring aberrant hepatic circadian rhythm
- Author
-
Sebastian Larion, Caleb A. Padgett, Joshua T. Butcher, James D. Mintz, David J. Fulton, and David W. Stepp
- Subjects
Hepatology ,Tumor Necrosis Factor-alpha ,Physiology ,Insulins ,Gastroenterology ,Mice, Obese ,Period Circadian Proteins ,Lipids ,Circadian Rhythm ,Mice, Inbred C57BL ,Mice ,Non-alcoholic Fatty Liver Disease ,Circadian Clocks ,Physiology (medical) ,Animals ,Obesity ,RNA, Messenger ,Luciferases - Abstract
Nonalcoholic fatty liver disease (NAFLD) is associated with disruption of homeostatic lipid metabolism, but underlying processes are poorly understood. One possible mechanism is impairment in hepatic circadian rhythm, which regulates key lipogenic mediators in the liver and whose circadian oscillation is diminished in obesity. Nobiletin enhances biological rhythms by activating RAR-related orphan receptor nuclear receptor, protecting against metabolic syndrome in a clock-dependent manner. The effect of nobiletin in NAFLD is unclear. In this study, we investigate the clock-enhancing effects of nobiletin in genetically obese (db/db) PER2::LUCIFERASE reporter mice with fatty liver. We report microarray expression data suggesting hepatic circadian signaling is impaired in db/db mice with profound hepatic steatosis. Circadian PER2 activity, as assessed by mRNA and luciferase assay, was significantly diminished in liver of db/db PER2::LUCIFERASE reporter mice. Continuous animal monitoring systems and constant dark studies suggest the primary circadian defect in db/db mice lies within peripheral hepatic oscillators and not behavioral rhythms or the master clock. In vitro, nobiletin restored PER2 amplitude in lipid-laden PER2::LUCIFERASE reporter macrophages. In vivo, nobiletin dramatically upregulated core clock gene expression, hepatic PER2 activity, and ameliorated steatosis in db/db PER2::LUCIFERASE reporter mice. Mechanistically, nobiletin reduced serum insulin levels, decreased hepatic Srebp1c, Acaca1, Tnfα, and Fgf21 expression, but did not improve Plin2, Plin5, or Cpt1, suggesting nobiletin attenuates steatosis in db/db mice via downregulation of hepatic lipid accumulation. These data suggest restoring endogenous rhythm with nobiletin resolves steatosis in obesity, proposing that hypothesis that targeting the biological clock may be an attractive therapeutic strategy for NAFLD.bNEWamp; NOTEWORTHY/bNAFLD is the most common chronic liver disease, but underlying mechanisms are unclear. We show here that genetically obese (idb/db/i) mice with fatty liver have impaired hepatic circadian rhythm. HepaticiPer2/iexpression and PER2 reporter activity are diminished inidb/db/iPER2::LUCIFERASE mice. The biological clock-enhancer nobiletin restores hepatic PER2 inidb/db/iPER2::LUCIFERASE mice, resolving steatosis via downregulation ofiSrebp1c/i. These studies suggest targeting the circadian clock may be beneficial strategy in NAFLD.
- Published
- 2022
- Full Text
- View/download PDF
14. The Long Noncoding RNA Cardiac Mesoderm Enhancer-Associated Noncoding RNA (Carmn) Is a Critical Regulator of Gastrointestinal Smooth Muscle Contractile Function and Motility
- Author
-
Xiangqin He, Kunzhe Dong, Jian Shen, Guoqing Hu, James D. Mintz, Reem T. Atawia, Juanjuan Zhao, Xiuxu Chen, Robert W. Caldwell, Meixiang Xiang, David W. Stepp, David J. Fulton, and Jiliang Zhou
- Subjects
Hepatology ,Gastroenterology - Published
- 2023
- Full Text
- View/download PDF
15. Disruption of endothelial Pfkfb3 ameliorates diet-induced murine insulin resistance
- Author
-
Qiuhua Yang, David W. Stepp, Mei Hong, Yuqing Huo, Zhiping Liu, David J. Fulton, Neal L. Weintraub, Xiaoxiao Mao, Yongfeng Cai, Jiean Xu, Qian Ma, and Yaqi Zhou
- Subjects
Male ,medicine.medical_specialty ,Phosphofructokinase-2 ,Endocrinology, Diabetes and Metabolism ,Adipose tissue ,Mice, Transgenic ,Inflammation ,Diet, High-Fat ,Mice ,chemistry.chemical_compound ,Endocrinology ,Insulin resistance ,Stress, Physiological ,Internal medicine ,medicine ,Animals ,Humans ,Glycolysis ,Cells, Cultured ,Gene knockdown ,Chemistry ,Endothelial Cells ,NF-κB ,medicine.disease ,Mice, Inbred C57BL ,Endothelial stem cell ,Endothelium, Vascular ,Insulin Resistance ,medicine.symptom ,Steatosis - Abstract
Overnutrition-induced endothelial inflammation plays a crucial role in high-fat diet (HFD)-induced insulin resistance in animals. Endothelial glycolysis plays a critical role in endothelial inflammation and proliferation, but its role in diet-induced endothelial inflammation and subsequent insulin resistance has not been elucidated. PFKFB3 is a critical glycolytic regulator, and its increased expression has been observed in adipose vascular endothelium of C57BL/6J mice fed with HFD in vivo, and in palmitate (PA)-treated primary human adipose microvascular endothelial cells (HAMECs) in vitro. We generated mice with Pfkfb3 deficiency selective for endothelial cells to examine the effect of endothelial Pfkfb3 in endothelial inflammation in metabolic organs and in the development of HFD-induced insulin resistance. EC Pfkfb3-deficientmice exhibited mitigated HFD-induced insulin resistance, including decreased body weight and fat mass, improved glucose clearance and insulin sensitivity, and alleviated adiposity and hepatic steatosis. Mechanistically, cultured PFKFB3 knockdown HAMECs showed decreased NF-κB activation induced by PA, and consequent suppressed adhesion molecule expression and monocyte adhesion. Taken together, these results demonstrate that increased endothelial PFKFB3 expression promotes diet-induced inflammatory responses and subsequent insulin resistance, suggesting that endothelial metabolic alteration plays an important role in the development of insulin resistance.
- Published
- 2021
- Full Text
- View/download PDF
16. The LncRNA Carmn is a Critical Regulator for Gastrointestinal Smooth Muscle Contractile Function and Motility
- Author
-
Xiangqin He, Kunzhe Dong, Jian Shen, Guoqing Hu, James D. Mintz, Reem T. Atawia, Juanjuan Zhao, Xiuxu Chen, Robert W. Caldwell, Meixiang Xiang, David W. Stepp, David J. Fulton, and Jiliang Zhou
- Abstract
Background & aimsVisceral smooth muscle cells (SMCs) are an integral component of the gastrointestinal (GI) tract and are critical for regulating motility. SMC contraction is regulated by changes in post-translational signaling and the state of differentiation. Impaired SMC contraction is associated with significant morbidity and mortality but the mechanisms regulating the expression levels of SMC-specific contractile proteins, including the role of long non-coding RNAs (lncRNAs), remains largely unexplored. Herein, we have uncovered an important role of Carmn (Cardiac mesoderm enhancer-associated noncoding RNA), a SMC-specific lncRNA, in regulating the phenotype of visceral SMCs of the GI tract.MethodsAnalysis of GTEx and publicly available single-cell RNA sequencing (scRNA-seq) datasets from embryonic, adult human and mouse GI tissues were used to identify SMC-specific lncRNAs. The functional role of Carmn was investigated using a novel GFP knock-in (KI) reporter/knockout (KO) mouse model. Bulk RNA sequencing (RNA-seq) and single nuclei RNA sequencing (snRNA-seq) of colonic muscularis were used to investigate underlying mechanisms.ResultsUnbiased in silico analyses and GFP expression patterns in Carmn GFP KI mice revealed that Carmn is specifically expressed in SMCs in human and mouse GI tract. Premature lethality was observed in global Carmn KO (gKO) and inducible SMC-specific KO (iKO) mice due to colonic pseudo-obstruction, severe distension of the GI tract with blockages in cecum and colon segments. Histology, whole-gut GI transit time and muscle myography analysis revealed severe dilation, significantly delayed GI transit and impaired GI contractility in Carmn KO mice versus control mice. Bulk RNA-seq of colonic muscularis revealed that Carmn deficiency promotes SMC de-differentiation as evidenced by up-regulation of extracellular matrix genes and down-regulation of SMC contractile genes including Mylk, a key regulator of SMC contraction. SnRNA-seq further revealed SMC Carmn deficiency not only compromised myogenic motility by reducing expression of contractile genes but also impaired neurogenic motility by disrupting cell-cell connectivity in the colonic muscularis. These findings may have translational significance as silencing CARMN in human colonic SMCs significantly attenuated contractile gene expression including MYLK and decreased SMC contractility. Luciferase reporter assays showed that CARMN enhances the transactivation activity of the master regulator of SMC contractile phenotype, myocardin, thereby maintaining the GI SMC myogenic program.ConclusionOur data suggest that Carmn is indispensable for maintaining GI SMC contractile function in mice, and that loss of function of CARMN may contribute to human visceral myopathy. To our knowledge this is the first study showing an essential role of lncRNA in the regulation of visceral SMC phenotype.
- Published
- 2022
- Full Text
- View/download PDF
17. Galectin‐3 Regulates Oxidant Stress in Obesity
- Author
-
Caleb A. Padgett, Andrew C. Speese, Cody L. Rosewater, Zachary L. Corley, James D. Mintz, David J. Fulton, and David W. Stepp
- Subjects
Genetics ,Molecular Biology ,Biochemistry ,Biotechnology - Published
- 2022
- Full Text
- View/download PDF
18. PFKFB3 Mediated Changes in ROS/NO Balance Contribute to Endothelial Dysfunction in Obesity
- Author
-
Robert K. Batori, Zsuzsanna Bordan, Caleb Padgett, Reem Atawia, Eric Belin Chantemele, David Stepp, and David J. Fulton
- Subjects
Genetics ,Molecular Biology ,Biochemistry ,Biotechnology - Published
- 2022
- Full Text
- View/download PDF
19. Adenosine and ATPγS protect against bacterial pneumonia-induced acute lung injury
- Author
-
Anita Kovacs-Kasa, Mary Cherian-Shaw, Christine Gross, Alexander D. Verin, Mary L Meadows, and David J. Fulton
- Subjects
0301 basic medicine ,Male ,Adenosine ,Lipopolysaccharide ,Vasodilator Agents ,Phosphatase ,Acute Lung Injury ,lcsh:Medicine ,Gene delivery ,Lung injury ,Pharmacology ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Adenosine Triphosphate ,Extracellular ,medicine ,Escherichia coli ,Pneumonia, Bacterial ,Animals ,Phosphorylation ,Purine metabolism ,lcsh:Science ,Multidisciplinary ,Chronic obstructive pulmonary disease ,lcsh:R ,Affinity Labels ,respiratory system ,Mice, Inbred C57BL ,030104 developmental biology ,030228 respiratory system ,chemistry ,lcsh:Q ,Bacterial outer membrane ,medicine.drug - Abstract
Lipopolysaccharide (LPS), a component of the outer membrane of gram-negative bacteria, disrupts the alveolar-capillary barrier, triggering pulmonary vascular leak thus inducing acute lung injury (ALI). Extracellular purines, adenosine and ATP, protected against ALI induced by purified LPS. In this study, we investigated whether these purines can impact vascular injury in more clinically-relevant E.coli (non-sterile LPS) murine ALI model. Mice were inoculated with live E. coli intratracheally (i.t.) with or without adenosine or a non-hydrolyzable ATP analog, adenosine 5′-(γ-thio)-triphosphate (ATPγS) added intravenously (i.v.). After 24 h of E. coli treatment, we found that injections of either adenosine or ATPγS 15 min prior or adenosine 3 h after E.coli insult significantly attenuated the E.coli-mediated increase in inflammatory responses. Furthermore, adenosine prevented weight loss, tachycardia, and compromised lung function in E. coli-exposed mice. Accordingly, treatment with adenosine or ATPγS increased oxygen saturation and reduced histopathological signs of lung injury in mice exposed to E. coli. Lastly, lung-targeting gene delivery of adenosine or ATPγS downstream effector, myosin phosphatase, significantly attenuated the E. coli-induced compromise of lung function. Collectively, our study has demonstrated that adenosine or ATPγS mitigates E. coli-induced ALI in mice and may be useful as an adjuvant therapy in future pre-clinical studies.
- Published
- 2020
- Full Text
- View/download PDF
20. Role of prostaglandin D2 receptors in the pathogenesis of abdominal aortic aneurysm formation
- Author
-
Neal L. Weintraub, Andra L. Blomkalns, Mourad Ogbi, Tetsuo Horimatsu, Tyler W. Benson, Yuqing Huo, David J. Fulton, Gautam Agarwal, Richard Lee, Michael A. Winkler, Lufei Young, Ken Fujise, Avirup Guha, Tohru Fukai, Masuko Ushio-Fukai, Xiaochun Long, Brian H. Annex, and Ha Won Kim
- Subjects
Male ,Mice ,Angiotensin II ,Receptors, Prostaglandin ,cardiovascular system ,Animals ,macromolecular substances ,General Medicine ,Receptors, Immunologic ,Aortic Aneurysm, Abdominal - Abstract
Prostaglandin D2 (PGD2) released from immune cells or other cell types activates its receptors, D prostanoid receptor (DP)1 and 2 (DP1 and DP2), to promote inflammatory responses in allergic and lung diseases. Prostaglandin-mediated inflammation may also contribute to vascular diseases such as abdominal aortic aneurysm (AAA). However, the role of DP receptors in the pathogenesis of AAA has not been systematically investigated. In the present study, DP1-deficient mice and pharmacological inhibitors of either DP1 or DP2 were tested in two distinct mouse models of AAA formation: angiotensin II (AngII) infusion and calcium chloride (CaCl2) application. DP1-deficient mice [both heterozygous (DP1+/−) and homozygous (DP1−/−)] were protected against CaCl2-induced AAA formation, in conjunction with decreased matrix metallopeptidase (MMP) activity and adventitial inflammatory cell infiltration. In the AngII infusion model, DP1+/− mice, but not DP1−/− mice, exhibited reduced AAA formation. Interestingly, compensatory up-regulation of the DP2 receptor was detected in DP1−/− mice in response to AngII infusion, suggesting a potential role for DP2 receptors in AAA. Treatment with selective antagonists of DP1 (laropiprant) or DP2 (fevipiprant) protected against AAA formation, in conjunction with reduced elastin degradation and aortic inflammatory responses. In conclusion, PGD2 signaling contributes to AAA formation in mice, suggesting that antagonists of DP receptors, which have been extensively tested in allergic and lung diseases, may be promising candidates to ameliorate AAA.
- Published
- 2022
21. Endothelial AMPKα1/PRKAA1 exacerbates inflammation in HFD-fed mice
- Author
-
Mei Hong, Yaqi Zhou, Yuqing Huo, Qian Ma, Qingen Da, Zhiping Liu, David J. Fulton, Neal L. Weintraub, Jiean Xu, Yongfeng Cai, Qiuhua Yang, Eric J. Belin de Chantemèle, and Xiaoxiao Mao
- Subjects
medicine.medical_specialty ,ATP citrate lyase ,Adipose tissue ,Inflammation ,AMP-Activated Protein Kinases ,Diet, High-Fat ,Mice ,Internal medicine ,medicine ,Animals ,Glycolysis ,Protein kinase A ,Pharmacology ,Metabolic Syndrome ,Mice, Knockout ,Chemistry ,Body Weight ,Endothelial Cells ,medicine.disease ,Endothelial stem cell ,Mice, Inbred C57BL ,Endocrinology ,Glucose ,Steatosis ,medicine.symptom ,Metabolic syndrome ,Insulin Resistance - Abstract
BACKGROUND AND PURPOSE Excess nutrient-induced endothelial cell inflammation is a hallmark in high fat diet (HFD)-induced metabolic syndrome. Pharmacological activation of protein kinase AMP-activated alpha 1(PRKAA1)/5'-Adenosine monophosphate-activated protein kinase alpha1 (AMPKα1) shows its beneficial effects in many studies of cardiometabolic disorders. However, AMPKα1, as a major cellular sensor of energy and nutrients in endothelial cells, has not been studied for its physiological role in excess nutrient-induced endothelial cell (EC) inflammation. EXPERIMENTAL APPROACH Wild-type and EC-specific Prkaa1 knockout mice were fed with an HFD. Body weight, fat mass composition, glucose and lipid levels were monitored regularly. Insulin sensitivity was analyzed systemically and in major metabolic organs/tissues. Inflammation status in metabolic organs/tissues were examined with quantitative RT-PCR and flow cytometry. Additionally, metabolic status, inflammation severity and signaling in cultured ECs were assayed with multiple approaches at the molecular level. KEY RESULTS EC Prkaa1 deficiency unexpectedly alleviated HFD-induced metabolic syndromes including decreased body weight and fat mass, enhanced glucose clearance and insulin sensitivity, and relieved adipose inflammation and hepatic steatosis. Mechanistically, PRKAA1 knockdown in cultured ECs reduced endothelial glycolysis and fatty acid oxidation, decreased the levels of acetyl-coA, and suppressed transcription of inflammatory molecules mediated by ATP citrate lyase (ACLY) and histone acetyltransferase p300. CONCLUSIONS AND IMPLICATIONS This unexpected pro-inflammatory effect of endothelial AMPKα1/PRKAA1 in metabolic context provides additional insight in AMPKα1/PRKAA1 activities, warranting that in-depth study and thoughtful consideration should be applied when AMPKα1/PRKAA1 is used as a therapeutic target in the treatment of metabolic syndrome.
- Published
- 2021
22. Niacin protects against abdominal aortic aneurysm formation via GPR109A independent mechanisms: role of NAD+/nicotinamide
- Author
-
Nicole Gilreath, Yuqing Huo, Gautam Agarwal, Tyler W. Benson, Andra L. Blomkalns, David J. Fulton, Sagar Patel, Adrien Mann, Ashlee Edgell, Mourad Ogbi, Jonathan Pye, Lauren Reid, Samah Ahmadieh, Brian K. Stansfield, Allie Thompson, Mary M. Moses, Nagendra Singh, Neal L. Weintraub, Tetsuo Horimatsu, David Kim, Nathan Robbins, Stefan Offermanns, Ha Won Kim, and Stephanie B. Benjamin
- Subjects
Male ,Niacinamide ,Physiology ,macromolecular substances ,Pharmacology ,Nicotinamide adenine dinucleotide ,Niacin ,Receptors, G-Protein-Coupled ,Calcium Chloride ,chemistry.chemical_compound ,Immune system ,Sirtuin 1 ,NAD+ ,Physiology (medical) ,Humans ,Animals ,AcademicSubjects/MED00200 ,Aorta, Abdominal ,Receptor ,Nicotinamide ,Aorta ,Cells, Cultured ,Mice, Knockout ,Sirt1 ,Vascular Pathophysiology ,Chemistry ,Angiotensin II ,digestive, oral, and skin physiology ,food and beverages ,Original Articles ,NAD ,GPR109A ,Mice, Inbred C57BL ,Disease Models, Animal ,Nicotinic acid ,Receptors, LDL ,cardiovascular system ,Abdominal aortic aneurysm ,NAD+ kinase ,Cardiology and Cardiovascular Medicine ,Aortic Aneurysm, Abdominal ,Dilatation, Pathologic ,Signal Transduction - Abstract
Aims Chronic adventitial and medial infiltration of immune cells play an important role in the pathogenesis of abdominal aortic aneurysms (AAAs). Nicotinic acid (niacin) was shown to inhibit atherosclerosis by activating the anti-inflammatory G protein-coupled receptor GPR109A [also known as hydroxycarboxylic acid receptor 2 (HCA2)] expressed on immune cells, blunting immune activation and adventitial inflammatory cell infiltration. Here, we investigated the role of niacin and GPR109A in regulating AAA formation. Methods and results Mice were supplemented with niacin or nicotinamide, and AAA was induced by angiotensin II (AngII) infusion or calcium chloride (CaCl2) application. Niacin markedly reduced AAA formation in both AngII and CaCl2 models, diminishing adventitial immune cell infiltration, concomitant inflammatory responses, and matrix degradation. Unexpectedly, GPR109A gene deletion did not abrogate the protective effects of niacin against AAA formation, suggesting GPR109A-independent mechanisms. Interestingly, nicotinamide, which does not activate GPR109A, also inhibited AAA formation and phenocopied the effects of niacin. Mechanistically, both niacin and nicotinamide supplementation increased nicotinamide adenine dinucleotide (NAD+) levels and NAD+-dependent Sirt1 activity, which were reduced in AAA tissues. Furthermore, pharmacological inhibition of Sirt1 abrogated the protective effect of nicotinamide against AAA formation. Conclusion Niacin protects against AAA formation independent of GPR109A, most likely by serving as an NAD+ precursor. Supplementation of NAD+ using nicotinamide-related biomolecules may represent an effective and well-tolerated approach to preventing or treating AAA.
- Published
- 2019
- Full Text
- View/download PDF
23. Copper Transporter ATP7A (Copper-Transporting P-Type ATPase/Menkes ATPase) Limits Vascular Inflammation and Aortic Aneurysm Development
- Author
-
Archita Das, Dipankar Ash, Tetsuo Horimatsu, Tohru Fukai, Masuko Ushio-Fukai, Bhupesh Singla, Silvia Leanhart, Ha Won Kim, Olga Antipova, Gabor Csanyi, David J. Fulton, Joseph White, Neal L. Weintraub, Varadarajan Sudhahar, Stefan Vogt, and Jack H. Kaplan
- Subjects
Male ,medicine.medical_specialty ,ATPase ,ATP7A ,Down-Regulation ,Apoptosis ,Mice, Transgenic ,Muscle, Smooth, Vascular ,Article ,Aortic aneurysm ,Copper Transport Proteins ,Internal medicine ,medicine ,Animals ,Humans ,Cells, Cultured ,Chelating Agents ,Inflammation ,Molybdenum ,biology ,Chemistry ,Angiotensin II ,Transporter ,medicine.disease ,Micronutrient ,Up-Regulation ,Mice, Inbred C57BL ,Disease Models, Animal ,MicroRNAs ,Endocrinology ,Copper-Transporting ATPases ,cardiovascular system ,biology.protein ,P-type ATPase ,Female ,Cardiology and Cardiovascular Medicine ,Elastin ,Copper ,Aortic Aneurysm, Abdominal ,Molecular Chaperones - Abstract
Objective: Copper (Cu) is essential micronutrient, and its dysregulation is implicated in aortic aneurysm (AA) development. The Cu exporter ATP7A (copper-transporting P-type ATPase/Menkes ATPase) delivers Cu via the Cu chaperone Atox1 (antioxidant 1) to secretory Cu enzymes, such as lysyl oxidase, and excludes excess Cu. Lysyl oxidase is shown to protect against AA formation. However, the role and mechanism of ATP7A in AA pathogenesis remain unknown. Approach and Results: Here, we show that Cu chelator markedly inhibited Ang II (angiotensin II)–induced abdominal AA (AAA) in which ATP7A expression was markedly downregulated. Transgenic ATP7A overexpression prevented Ang II–induced AAA formation. Conversely, Cu transport dysfunctional ATP7A mut/+ /ApoE −/− mice exhibited robust AAA formation and dissection, excess aortic Cu accumulation as assessed by X-ray fluorescence microscopy, and reduced lysyl oxidase activity. In contrast, AAA formation was not observed in Atox1 −/− /ApoE −/− mice, suggesting that decreased lysyl oxidase activity, which depends on both ATP7A and Atox1, was not sufficient to develop AAA. Bone marrow transplantation suggested importance of ATP7A in vascular cells, not bone marrow cells, in AAA development. MicroRNA (miR) array identified miR-125b as a highly upregulated miR in AAA from ATP7A mut/+ /ApoE −/− mice. Furthermore, miR-125b target genes (histone methyltransferase Suv39h1 and the NF-κB negative regulator TNFAIP3 [tumor necrosis factor alpha induced protein 3]) were downregulated, which resulted in increased proinflammatory cytokine expression, aortic macrophage recruitment, MMP (matrix metalloproteinase)-2/9 activity, elastin fragmentation, and vascular smooth muscle cell loss in ATP7A mut/+ /ApoE −/− mice and reversed by locked nucleic acid-anti-miR-125b infusion. Conclusions: ATP7A downregulation/dysfunction promotes AAA formation via upregulating miR-125b, which augments proinflammatory signaling in a Cu-dependent manner. Thus, ATP7A is a potential therapeutic target for inflammatory vascular disease.
- Published
- 2019
- Full Text
- View/download PDF
24. Galectin-3 is expressed in vascular smooth muscle cells and promotes pulmonary hypertension through changes in proliferation, apoptosis, and fibrosis
- Author
-
Feng Chen, Keyvan Mahboubi, Daniel S. Weintraub, Peter Traber, Jennifer A. Thompson, Yusi Wang, Yunchao Su, Zsuzsanna Bordan, Jennifer C. Sullivan, G. Ryan Crislip, David J. Fulton, Stephen Haigh, Xueyi Li, Joshua T. Butcher, Jiliang Zhou, Danny Jonigk, David W. Stepp, William Snider, Dmitry Kondrikov, and Scott A. Barman
- Subjects
Male ,0301 basic medicine ,Pulmonary and Respiratory Medicine ,Pathology ,medicine.medical_specialty ,Vascular smooth muscle ,Knockout rat ,Physiology ,Galectin 3 ,Galectins ,Hypertension, Pulmonary ,Pulmonary Fibrosis ,Myocytes, Smooth Muscle ,Apoptosis ,030204 cardiovascular system & hematology ,Muscle, Smooth, Vascular ,Rats, Sprague-Dawley ,03 medical and health sciences ,0302 clinical medicine ,Fibrosis ,Physiology (medical) ,medicine ,Animals ,Humans ,Cell Proliferation ,business.industry ,Cell migration ,Blood Proteins ,Cell Biology ,medicine.disease ,Pulmonary hypertension ,Rats ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Gene Expression Regulation ,Galectin-3 ,Vascular resistance ,business ,Research Article - Abstract
A defining characteristic of pulmonary hypertension (PH) is the extensive remodeling of pulmonary arteries (PAs), which results in progressive increases in vascular resistance and stiffness and eventual failure of the right ventricle. There is no cure for PH and identification of novel molecular mechanisms that underlie increased proliferation, reduced apoptosis, and excessive extracellular matrix production in pulmonary artery smooth muscle cells (PASMCs) is a vital objective. Galectin-3 (Gal-3) is a chimeric lectin and potent driver of many aspects of fibrosis, but its role in regulating PASMC behavior in PH remains poorly understood. Herein, we evaluated the importance of increased Gal-3 expression and signaling on PA vascular remodeling and cardiopulmonary function in experimental models of PH. Gal-3 expression was quantified by qRT-PCR, immunoblotting, and immunofluorescence imaging, and its functional role was assessed by specific Gal-3 inhibitors and CRISPR/Cas9-mediated knockout of Gal-3 in the rat. In rat models of PH, we observed increased Gal-3 expression in PASMCs, which stimulated migration and resistance to apoptosis, whereas silencing or genetic deletion reduced cellular migration and PA fibrosis and increased apoptosis. Gal-3 inhibitors attenuated and reversed PA remodeling and fibrosis, as well as hemodynamic indices in monocrotaline (MCT)-treated rats in vivo. These results were supported by genetic deletion of Gal-3 in both MCT and Sugen Hypoxia rat models. In conclusion, our results suggest that elevated Gal-3 levels contribute to inappropriate PA remodeling in PH by enhancing multiple profibrotic mechanisms. Therapeutic strategies targeting Gal-3 may be of benefit in the treatment of PH.
- Published
- 2019
- Full Text
- View/download PDF
25. Inhibition of Class IIa HDACs improves endothelial barrier function in endotoxin-induced acute lung injury
- Author
-
Mary L Meadows, Yunchao Su, Anita Kovacs-Kasa, Alexander D. Verin, Vijay Patel, Laszlo Kovacs, Mary Cherian-Shaw, and David J. Fulton
- Subjects
0301 basic medicine ,Lipopolysaccharides ,rho GTP-Binding Proteins ,Myosin light-chain kinase ,Cell Membrane Permeability ,Lipopolysaccharide ,Physiology ,Clinical Biochemistry ,Acute Lung Injury ,Lung injury ,Models, Biological ,Article ,Histone Deacetylases ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,In vivo ,Heart Rate ,Animals ,Lung ,Barrier function ,Adaptor Proteins, Signal Transducing ,Chemistry ,Endothelial Cells ,Cell Biology ,Pneumonia ,HDAC6 ,Cell biology ,Endotoxins ,Histone Deacetylase Inhibitors ,Mice, Inbred C57BL ,Oxygen ,Disease Models, Animal ,030104 developmental biology ,030220 oncology & carcinogenesis ,Microvessels ,Phosphorylation ,Histone deacetylase ,Signal Transduction - Abstract
Acute lung injury (ALI) is an acute inflammatory process arises from a wide range of lung insults. A major cause of ALI is dysfunction of the pulmonary vascular endothelial barrier but the mechanisms involved are incompletely understood. The therapeutic potential of histone deacetylase (HDAC) inhibitors for the treatment of cardiovascular and inflammatory diseases is increasingly apparent, but the mechanisms by which HDACs regulate pulmonary vascular barrier function remain to be resolved. We found that specific Class IIa HDACs inhibitor, TMP269, significantly attenuated the lipopolysaccharide (LPS)-induced human lung microvascular endothelial cells (HLMVEC) barrier compromise in vitro and improved vascular barrier integrity and lung function in murine model of ALI in vivo. TMP269 decreased LPS-induced myosin light chain phosphorylation suggesting the role for Class IIa HDACs in LPS-induced cytoskeleton reorganization. TMP269 did not affect microtubule structure and tubulin acetylation in contrast to the HDAC6-specific inhibitor, Tubastatin A suggesting that Class IIa HDACs and HDAC6 (Class IIb) regulate endothelial cytoskeleton and permeability via different mechanisms. Furthermore, LPS increased the expression of ArgBP2 which has recently been attributed to HDAC-mediated activation of Rho. Depletion of ArgBP2 abolished the ability of LPS to disrupt barrier function in HLMVEC and both TMP269 and Tubastatin A decreased the level of ArgBP2 expression after LPS stimulation suggesting that both Class IIa and IIb HDACs regulate endothelial permeability via ArgBP2-dependent mechanism. Collectively, our data strongly suggest that Class IIa HDACs are involved in LPS-induced ALI in vitro and in vivo via specific mechanism which involved contractile responses, but not microtubule reorganization.
- Published
- 2020
26. Origins of Hypertension in Obesity: Plain vanilla(oid) or multiple flavors?
- Author
-
David J. Fulton and David W. Stepp
- Subjects
medicine.medical_specialty ,MEDLINE ,A Kinase Anchor Proteins ,TRPV Cation Channels ,Blood Pressure ,Diet, High-Fat ,Article ,Mice ,Transient Receptor Potential Channels ,Physiology (medical) ,Internal medicine ,Peroxynitrous Acid ,medicine ,Animals ,Humans ,Calcium Signaling ,Obesity ,Vanilla ,Mice, Knockout ,business.industry ,Membrane Proteins ,medicine.disease ,Vasodilation ,Hypertension ,Endothelium, Vascular ,Cardiology and Cardiovascular Medicine ,business ,Heme Oxygenase-1 - Abstract
BACKGROUND: Impaired endothelium-dependent vasodilation is a hallmark of obesity-induced hypertension. The recognition that Ca(2+) signaling in endothelial cells promotes vasodilation has led to the hypothesis that endothelial Ca(2+) signaling is compromised during obesity, but the underlying abnormality is unknown. In this regard, TRPV4 ion channels are a major Ca(2+) influx pathway in endothelial cells, and regulatory protein AKAP150 enhances the activity of TRPV4 channels. METHODS: We used endothelium-specific knockout mice and high fat diet-fed mice to assess the role of endothelial AKAP150-TRPV4 signaling in blood pressure regulation under normal and obese conditions. We further determined the role of peroxynitrite, an oxidant molecule generated from the reaction between nitric oxide (NO) and superoxide radicals, in impairing endothelial AKAP150-TRPV4 signaling in obesity, and assessed the effectiveness of peroxynitrite inhibition in rescuing endothelial AKAP150-TRPV4 signaling in obesity. The clinical relevance of our findings was evaluated in arteries from non-obese and obese individuals. RESULTS: We show that Ca(2+) influx through TRPV4 channels at myoendothelial projections (MEPs) to smooth muscle cells decreases resting blood pressure in non-obese mice, a response that is diminished in obese mice. Counterintuitively, release of the vasodilator molecule NO attenuated endothelial TRPV4 channel activity and vasodilation in obese animals. Increased activities of iNOS and NOX1 enzymes at MEPs in obese mice generated higher levels of NO and superoxide radicals, resulting in increased local peroxynitrite formation and subsequent oxidation of the regulatory protein AKAP150 at cysteine 36, to impair AKAP150-TRPV4 channel signaling at MEPs. Strategies that lowered peroxynitrite levels prevented cysteine 36 oxidation of AKAP150, and rescued endothelial AKAP150-TRPV4 signaling, vasodilation, and blood pressure in obesity. Importantly, peroxynitrite-dependent impairment of endothelial TRPV4 channel activity and vasodilation was also observed in the arteries from obese patients. CONCLUSIONS: These data suggest that a spatially restricted impairment of endothelial TRPV4 channels contributes to obesity-induced hypertension, and imply that inhibiting peroxynitrite might represent a strategy for normalizing endothelial TRPV4 channel activity, vasodilation, and blood pressure in obesity.
- Published
- 2020
27. Mice with a specific deficiency of Pfkfb3 in myeloid cells are protected from hypoxia-induced pulmonary hypertension
- Author
-
David J. Fulton, Yunchao Su, Qiuhua Yang, Lina Wang, Yapeng Cao, Mei Hong, Scott A. Barman, Zhiping Liu, Jiean Xu, Yuqing Huo, Qian Ma, Xiaoxiao Mao, Yaqi Zhou, Rudolf Lucas, and Xiaoyu Zhang
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Myeloid ,Phosphofructokinase-2 ,Hypertension, Pulmonary ,Peripheral blood mononuclear cell ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Western blot ,Internal medicine ,medicine ,Animals ,Humans ,Rats, Wistar ,Hypoxia ,Pharmacology ,Lung ,medicine.diagnostic_test ,Chemistry ,Macrophages ,Hypoxia (medical) ,Rats ,030104 developmental biology ,HIF1A ,medicine.anatomical_structure ,Endocrinology ,Leukocytes, Mononuclear ,Bone marrow ,medicine.symptom ,Glycolysis ,030217 neurology & neurosurgery ,Immunostaining - Abstract
BACKGROUND AND PURPOSE Macrophage infiltration into the lungs is a characteristic of pulmonary hypertension (PH). Glycolysis is the main metabolic pathway for macrophage activation. However, the effect of macrophage glycolysis on the development of PH remains unknown. We investigated the effect of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKBF3), a critical enzyme of macrophage glycolysis, on PH development. EXPERIMENTAL APPROACH Lung tissues from PH patients were examined by immunostaining with macrophage markers. PH was induced in Wistar rats with SU5416/hypoxia and in mice with hypoxia. Lungs and macrophages were isolated for analysis by RT-PCR, western blot, flow cytometry, and immunostaining. KEY RESULTS Expression of glycolytic molecules was increased in circulating peripheral blood mononuclear cells (PBMCs) and lung macrophages of PH patients. These results were also found in lung macrophages of SU5416/hypoxia (Su/Hx)-induced PH rats and hypoxia-induced PH mice. PH was ameliorated in myeloid-specific Pfkfb3-deficient mice (Pfkfb3ΔMϕ ) or mice treated with the PFKFB3 inhibitor 3PO, compared with their controls. Alveolar macrophages of PH Pfkfb3ΔMϕ mice produced lower levels of growth factors and pro-inflammatory cytokines than those of control mice. Circulating myeloid cells and lung myeloid cells were much fewer in PH Pfkfb3ΔMϕ mice than controls. Mechanistically, overexpression of Hif1a or Hif2a in bone marrow-derived macrophages (BMDMs) cultured with bone marrow of Pfkfb3ΔMϕ mice restored the decreased expression of pro-inflammatory cytokines and growth factors. CONCLUSIONS AND IMPLICATIONS Myeloid Pfkfb3 deficiency protects mice from PH, thereby suggesting that myeloid PFKFB3 is one of the important targets in the therapeutic effect of PFKFB3 inhibition in PH treatment.
- Published
- 2020
28. Adenosine kinase is critical for neointima formation after vascular injury by inducing aberrant DNA hypermethylation
- Author
-
Yiming Xu, Yue Pan, Xianqiu Zeng, Yunchao Su, Neal L. Weintraub, Kaixiang Cao, Xuejun Jiang, Yong Wang, Yuqing Huo, Xiaoling Wang, Detlev Boison, David J. Fulton, Vijay Patel, Siyuan Yan, Qiuhua Yang, Yaqi Zhou, and Zhiping Liu
- Subjects
Neointima ,Intimal hyperplasia ,Vascular smooth muscle ,Physiology ,Myocytes, Smooth Muscle ,Adenosine kinase ,Vascular Remodeling ,Muscle, Smooth, Vascular ,Epigenesis, Genetic ,Kruppel-Like Factor 4 ,Physiology (medical) ,medicine ,Animals ,Humans ,Vascular Diseases ,Adenosine Kinase ,Cell Proliferation ,Mice, Knockout ,biology ,Chemistry ,Original Articles ,DNA ,DNA Methylation ,Vascular System Injuries ,medicine.disease ,musculoskeletal system ,Adenosine ,Adenosine receptor ,ADK ,Disease Models, Animal ,Carotid Arteries ,KLF4 ,biology.protein ,Cancer research ,cardiovascular system ,Cardiology and Cardiovascular Medicine ,Carotid Artery Injuries ,tissues ,medicine.drug - Abstract
Aim Adenosine receptors and extracellular adenosine have been demonstrated to modulate vascular smooth muscle cell (VSMC) proliferation and neointima formation. Adenosine kinase (ADK) is a major enzyme regulating intracellular adenosine levels, but is function in VSMC remains unclear. Here, we investigated the role of ADK in vascular injury-induced smooth muscle proliferation and delineated the mechanisms underlying its action. Methods and results We found that ADK expression was higher in the neointima of injured vessels and in PDGF-treated VSMCs. Genetic and pharmacological inhibition of ADK was enough to attenuate arterial injury-induced neointima formation due to inhibition of VSMC proliferation. Mechanistically, using infinium methylation assays and bisulfite sequencing, we showed that ADK metabolized the intracellular adenosine and potentiated the transmethylation pathway, then induced the aberrant DNA hypermethylation. Pharmacological inhibition of aberrant DNA hypermethylation increased KLF4 expression and suppressed VSMC proliferation as well as the neointima formation. Importantly, in human femoral arteries, we observed increased ADK expression and DNA hypermethylation as well as decreased KLF4 expression in neointimal VSMCs of stenotic vessels suggesting that our findings in mice are relevant for human disease and may hold translational significance. Conclusions Our study unravels a novel mechanism by which ADK promotes VSMC proliferation via inducing aberrant DNA hypermethylation, thereby downregulating KLF4 expression and promoting neointima formation. These findings advance the possibility of targeting ADK as an epigenetic modulator to combat vascular injury. Translational perspective Abnormal proliferation of vascular smooth muscle cell (VSMC) is key to abundant occlusive vascular diseases in humans, such as atherosclerosis and intimal hyperplasia associated with restenosis. Adenosine has been shown to combat abnormal smooth muscle proliferation. Here, we demonstrate that increased catabolism of adenosine by adenosine kinase (ADK) promotes abnormal VSMC proliferation. The pathological ADK overexpression in both mice and humans with vascular disease promotes VSMC proliferation via inducing aberrant DNA hypermethylation and KLF4 downregulation. Our study suggests that pharmacological augmentation of endogenous adenosine by targeting ADK represents a promising therapeutic strategy for occlusive vascular diseases.
- Published
- 2020
29. Neurofibromin Deficiency Induces Endothelial Cell Proliferation and Retinal Neovascularization
- Author
-
David J. Fulton, Hanfang Zhang, Farlyn Z. Hudson, Brian K. Stansfield, Modesto Rojas, Rebekah Tritz, Chintan Patel, Zsuzsanna Bordan, Stephen Haigh, Zhimin Xu, David A. Ingram, Ruth B. Caldwell, and Neal L. Weintraub
- Subjects
0301 basic medicine ,Vascular Endothelial Growth Factor A ,congenital, hereditary, and neonatal diseases and abnormalities ,genetic structures ,Aorta, Thoracic ,Retinal Neovascularization ,Retina ,Neovascularization ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Cell Movement ,medicine ,retinopathy of prematurity ,Animals ,Humans ,Gene Silencing ,Hypoxia ,Cell Proliferation ,Hyperoxia ,neurofibromatosis ,Neurofibromin 1 ,biology ,Chemistry ,Endothelial Cells ,Retinal Vessels ,Retinal ,medicine.disease ,VEGF ,eye diseases ,Cell biology ,nervous system diseases ,Endothelial stem cell ,Mice, Inbred C57BL ,Oxygen ,Vascular endothelial growth factor A ,030104 developmental biology ,medicine.anatomical_structure ,030221 ophthalmology & optometry ,biology.protein ,endothelial cell ,sense organs ,medicine.symptom ,Retinopathy ,Ras ,Signal Transduction - Abstract
Purpose Neurofibromatosis type 1 (NF1) is the result of inherited mutations in the NF1 tumor suppressor gene, which encodes the protein neurofibromin. Eye manifestations are common in NF1 with recent reports describing a vascular dysplasia in the retina and choroid. Common features of NF1 retinopathy include tortuous and dilated feeder vessels that terminate in capillary tufts, increased endothelial permeability, and neovascularization. Given the retinal vascular phenotype observed in persons with NF1, we hypothesize that preserving neurofibromin may be a novel strategy to control pathologic retinal neovascularization. Methods Nf1 expression in human endothelial cells (EC) was reduced using small hairpin (sh) RNA and EC proliferation, migration, and capacity to form vessel-like networks were assessed in response to VEGF and hypoxia. Wild-type (WT), Nf1 heterozygous (Nf1+/-), and Nf1flox/+;Tie2cre pups were subjected to hyperoxia/hypoxia using the oxygen-induced retinopathy model. Retinas were analyzed quantitatively for extent of retinal vessel dropout, neovascularization, and capillary branching. Results Neurofibromin expression was suppressed in response to VEGF, which corresponded with activation of Mek-Erk and PI3-K-Akt signaling. Neurofibromin-deficient EC exhibited enhanced proliferation and network formation in response to VEGF and hypoxia via an Akt-dependent mechanism. In response to hyperoxia/hypoxia, Nf1+/- retinas exhibited increased vessel dropout and neovascularization when compared with WT retinas. Neovascularization was similar between Nf1+/- and Nf1flox/+;Tie2cre retinas, but capillary drop out in Nf1flox/+;Tie2cre retinas was significantly reduced when compared with Nf1+/- retinas. Conclusions These data suggest that neurofibromin expression is essential for controlling endothelial cell proliferation and retinal neovascularization and therapies targeting neurofibromin-deficient EC may be beneficial.
- Published
- 2018
30. Vasodilator dysfunction and oligodendrocyte dysmaturation in aging white matter
- Author
-
Thuan D. Ngyuen, David W. McNeal, Eric B. Larson, Zsolt Bagi, Huijuan Dou, C. Dirk Keene, Thomas J. Montine, Xi Gong, David J. Fulton, Allison Beller, Dieter D. Brandner, Phuong D. Le, and Stephen A. Back
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Population ,Ischemia ,Vasodilation ,medicine.disease_cause ,White matter ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Prefrontal cortex ,education ,Arteriolar vasodilator ,education.field_of_study ,business.industry ,medicine.disease ,Oligodendrocyte ,030104 developmental biology ,medicine.anatomical_structure ,Neurology ,Neurology (clinical) ,business ,030217 neurology & neurosurgery ,Oxidative stress ,medicine.drug - Abstract
OBJECTIVE Microvascular brain injury (mVBI) is a common pathological correlate of vascular contributions to cognitive impairment and dementia (VCID) that leads to white matter (WM) injury (WMI). VCID appears to arise from chronic recurrent white matter ischemia that triggers oxidative stress and an increase in total oligodendrocyte lineage cells. We hypothesized that mVBI involves vasodilator dysfunction of white matter penetrating arterioles and aberrant oligodendrocyte progenitor cell (OPC) responses to WMI. METHODS We analyzed cases of mVBI with low Alzheimer's disease neuropathological change in prefrontal cortex WM from rapid autopsies in a population-based cohort where VCID frequently occurs. Arteriolar vasodilator function was quantified by videomicroscopy. OPC maturation was quantified using lineage specific markers. RESULTS Acetylcholine-mediated arteriolar dilation in mVBI was significantly reduced in WM penetrators relative to pial arterioles. Astrogliosis-defined WMI was positively associated with increased OPCs and was negatively associated with decreased mature oligodendrocytes. INTERPRETATION Selectively impaired vasodilator function of WM penetrating arterioles in mVBI occurs in association with aberrant differentiation of OPCs in WMI, which supports that myelination disturbances in VCID are related to disrupted maturation of myelinating oligodendrocytes. Ann Neurol 2018;83:142-152.
- Published
- 2018
- Full Text
- View/download PDF
31. Endothelial adenosine A2a receptor-mediated glycolysis is essential for pathological retinal angiogenesis
- Author
-
Sarah Lu, Zhongjie Fu, Ruth B. Caldwell, Neal L. Weintraub, Chaodong Wu, Xuejiao Gu, Yuqing Huo, Zhiping Liu, Mohamed Al-Shabrawey, Shuya Zhang, Akrit Sodhi, Aftab Ahmad, Lois E.H. Smith, Yaqi Zhou, Siyuan Yan, Xizhen Xu, Ismail Kaddour-Djebbar, Mei Hong, Qinkai Li, Qiuhua Yang, Ye Sun, David J. Fulton, Jiang-Fan Chen, Xuejun Jiang, Xiaoling Liu, Yong Wang, Xianqiu Zeng, Jiaojiao Wang, Wenbo Zhang, and Yiming Xu
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,Male ,medicine.medical_specialty ,Receptor, Adenosine A2A ,Angiogenesis ,Science ,General Physics and Astronomy ,Adenosine A2A receptor ,Biology ,Retinal Neovascularization ,General Biochemistry, Genetics and Molecular Biology ,Retina ,Article ,03 medical and health sciences ,Mice ,Retinal Diseases ,Internal medicine ,medicine ,Animals ,Humans ,Receptor ,lcsh:Science ,Protein kinase B ,Mice, Knockout ,Multidisciplinary ,Endothelial Cells ,General Chemistry ,Hypoxia-Inducible Factor 1, alpha Subunit ,Adenosine ,Cell biology ,Endothelial stem cell ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,Adenosine Receptor A2a ,Endocrinology ,Female ,lcsh:Q ,Glycolysis ,medicine.drug - Abstract
Adenosine/adenosine receptor-mediated signaling has been implicated in the development of various ischemic diseases, including ischemic retinopathies. Here, we show that the adenosine A2a receptor (ADORA2A) promotes hypoxia-inducible transcription factor-1 (HIF-1)-dependent endothelial cell glycolysis, which is crucial for pathological angiogenesis in proliferative retinopathies. Adora2a expression is markedly increased in the retina of mice with oxygen-induced retinopathy (OIR). Endothelial cell-specific, but not macrophage-specific Adora2a deletion decreases key glycolytic enzymes and reduces pathological neovascularization in the OIR mice. In human primary retinal microvascular endothelial cells, hypoxia induces the expression of ADORA2A by activating HIF-2α. ADORA2A knockdown decreases hypoxia-induced glycolytic enzyme expression, glycolytic flux, and endothelial cell proliferation, sprouting and tubule formation. Mechanistically, ADORA2A activation promotes the transcriptional induction of glycolytic enzymes via ERK- and Akt-dependent translational activation of HIF-1α protein. Taken together, these findings advance translation of ADORA2A as a therapeutic target in the treatment of proliferative retinopathies and other diseases dependent on pathological angiogenesis., Pathological angiogenesis in the retina is a major cause of blindness. Here the authors show that adenosine receptor A2A drives pathological angiogenesis in the oxygen-induced retinopathy mouse model by promoting glycolysis in endothelial cells via the ERK/Akt/HIF-1α pathway, thereby suggesting new therapeutic targets for disease treatment.
- Published
- 2017
- Full Text
- View/download PDF
32. Genetic Deletion of NADPH Oxidase 1 Rescues Microvascular Function in Mice With Metabolic Disease
- Author
-
David W. Stepp, Eric J. Belin de Chantemèle, Jennifer A. Thompson, Sebastian Larion, David J. Fulton, and James D. Mintz
- Subjects
Blood Glucose ,Male ,0301 basic medicine ,medicine.medical_specialty ,Physiology ,Mice, Obese ,030204 cardiovascular system & hematology ,Article ,Muscle, Smooth, Vascular ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Insulin resistance ,Metabolic Diseases ,Internal medicine ,medicine ,Animals ,NADH, NADPH Oxidoreductases ,Mesenteric arteries ,Mice, Knockout ,chemistry.chemical_classification ,Reactive oxygen species ,NADPH oxidase ,biology ,NADPH Oxidase 1 ,medicine.disease ,Mice, Inbred C57BL ,Oxidative Stress ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,chemistry ,NOX1 ,Microvessels ,Knockout mouse ,cardiovascular system ,biology.protein ,Cardiology and Cardiovascular Medicine ,Gene Deletion ,Dyslipidemia - Abstract
Rationale: Early vascular changes in metabolic disease that precipitate the development of cardiovascular complications are largely driven by reactive oxygen species accumulation, yet the extent to which excess reactive oxygen species derive from specific NADPH oxidase isoforms remains ill defined. Objective: Identify the role of Nox1 in the development of microvascular dysfunction in metabolic disease. Methods and Results: Four genotypes were generated by breeding Nox1 knockout mice with db/db mice: lean (H db W nox1 ), lean Nox1 knockout (H db K nox1 ), obese (K db W nox1 ), and obese KK (K db K nox1 ). The degree of adiposity, insulin resistance, and dyslipidemia in KW mice was not influenced by Nox1 deletion as determined by nuclear magnetic resonance spectroscopy, glucose tolerance tests, and plasma analyses. Endothelium-dependent responses to acetylcholine in pressurized mesenteric arteries were reduced in KW versus HW ( P P Conclusions: Nox1 deletion reduces oxidant load and restores microvascular health in db/db mice without influencing the degree of metabolic dysfunction. Therefore, targeted Nox1 inhibition may be effective in the prevention of vascular complications.
- Published
- 2017
- Full Text
- View/download PDF
33. Leptin Restores Endothelial Function via Endothelial PPARγ-Nox1-Mediated Mechanisms in a Mouse Model of Congenital Generalized Lipodystrophy
- Author
-
Eric J. Belin de Chantemèle, David J. Fulton, Stephen Haigh, Simone Kennard, Vijay Patel, Jessica L. Faulkner, Weiqin Chen, Galina Antonova, and Thiago Bruder-Nascimento
- Subjects
0301 basic medicine ,Leptin ,Male ,medicine.medical_specialty ,Endothelium ,030204 cardiovascular system & hematology ,medicine.disease_cause ,Article ,Congenital generalized lipodystrophy ,03 medical and health sciences ,Mice ,Random Allocation ,0302 clinical medicine ,Lipodystrophy, Congenital Generalized ,Reference Values ,Internal medicine ,Internal Medicine ,medicine ,Animals ,Analysis of Variance ,business.industry ,digestive, oral, and skin physiology ,medicine.disease ,Mice, Inbred C57BL ,PPAR gamma ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,Treatment Outcome ,NOX1 ,cardiovascular system ,NADPH Oxidase 1 ,Endothelium, Vascular ,business ,Reactive Oxygen Species ,Function (biology) ,Oxidative stress ,hormones, hormone substitutes, and hormone antagonists - Abstract
Leptin is the current treatment for metabolic disorders associated with acquired and congenital generalized lipodystrophy (CGL). Although excess leptin levels have been associated with vascular inflammation and cardiovascular disease in the context of obesity, the effects of chronic leptin treatment on vascular function remain unknown in CGL. Here, we hypothesized that leptin treatment will improve endothelial function via direct vascular mechanisms. We investigated the cardiovascular consequences of leptin deficiency and supplementation in male gBscl2 −/− ( Berardinelli-Seip 2 gene–deficient) mice—a mouse model of CGL. CGL mice exhibited reduced adipose mass and leptin levels, as well as impaired endothelium-dependent relaxation. Blood vessels from CGL mice had increased NADPH Oxidase 1 (Nox1) expression and reactive oxygen species production, and selective Nox1 inhibition restored endothelial function. Remarkably, chronic and acute leptin supplementation restored endothelial function via a PPARγ-dependent mechanism that decreased Nox1 expression and reactive oxygen species production. Selective ablation of leptin receptors in endothelial cells promoted endothelial dysfunction, which was restored by Nox1 inhibition. Lastly, we confirmed in aortic tissue from older patients undergoing cardiac bypass surgery that acute leptin can promote signaling in human blood vessels. In conclusion, in gBscl2 −/− mice, leptin restores endothelial function via peroxisome proliferator activated receptor gamma-dependent decreases in Nox1. Furthermore, we provide the first evidence that vessels from aged patients remain leptin sensitive. These data reveal a new direct role of leptin receptors in the control of vascular homeostasis and present leptin as a potential therapy for the treatment of vascular disease associated with low leptin levels.
- Published
- 2019
34. Progesterone predisposes females to obesity-associated leptin-mediated endothelial dysfunction via upregulating endothelial mineralocorticoid receptor expression
- Author
-
Vijay Patel, Galina Antonova, Qing Lu, Jessica L. Faulkner, Anne-Cecile Huby, Iris Z. Jaffe, Eric J. Belin de Chantemèle, David J. Fulton, and Simone Kennard
- Subjects
Leptin ,Male ,medicine.medical_specialty ,Endogeny ,Disease ,030204 cardiovascular system & hematology ,Risk Assessment ,Sensitivity and Specificity ,Article ,03 medical and health sciences ,Mice ,Random Allocation ,0302 clinical medicine ,Mineralocorticoid receptor ,Sex Factors ,Internal medicine ,Progesterone receptor ,Internal Medicine ,medicine ,Animals ,Obesity ,Endothelial dysfunction ,Progesterone ,Mice, Inbred BALB C ,business.industry ,medicine.disease ,Up-Regulation ,Disease Models, Animal ,Endocrinology ,Receptors, Mineralocorticoid ,Gene Expression Regulation ,Female ,Endothelium, Vascular ,business ,030217 neurology & neurosurgery ,Sex characteristics - Abstract
Compelling clinical evidence indicates that obesity and its associated metabolic abnormalities supersede the protective effects of female sex-hormones and predisposes premenopausal women to cardiovascular disease. The underlying mechanisms remain poorly defined; however, recent studies have implicated overactivation of the aldosterone-mineralocorticoid receptor (MR) axis as a cause of sex-specific cardiovascular risk in obese females. Experimental evidence indicates that the MR on endothelial cells contributes to obesity-associated, leptin-induced endothelial dysfunction in female experimental models, however, the vascular-specific mechanisms via which females are predisposed to heightened endothelial MR activation remain unknown. Therefore, we hypothesized that endogenous expression of endothelial MR is higher in females than males which predisposes them to obesity-associated, leptin-mediated endothelial dysfunction. We found that endothelial MR expression is higher in blood vessels from female mice and humans compared to those of males, and further, that progesterone receptor activation in endothelial cells is the driving mechanism for sex-dependent increases in endothelial MR expression in females. In addition, we show that genetic deletion of either the endothelial MR or progesterone receptor in female mice prevents leptin-induced endothelial dysfunction, providing direct evidence that interaction between the progesterone receptor and MR mediates obesity-associated endothelial impairment in females. Collectively, these novel findings suggest that progesterone drives sex-differences in endothelial MR expression and predisposes female mice to leptin-induced endothelial dysfunction, which indicates that MR antagonists may be a promising sex-specific therapy to reduce the risk of cardiovascular diseases in obese premenopausal women.
- Published
- 2019
35. PFKFB3-mediated endothelial glycolysis promotes pulmonary hypertension
- Author
-
Jiean Xu, Jason X.-J. Yuan, Xiaoyu Zhang, Qiuhua Yang, Lina Wang, Junmin Quan, David J. Fulton, Jennifer C. Sullivan, Chaodong Wu, Zheng Dong, Laszlo Kovacs, Mei Hong, Louise Meadows, Zhiping Liu, Qian Ma, Yuqing Huo, Rudolf Lucas, Yiming Xu, Yaqi Zhou, Min Zhang, Scott A. Barman, Jingjing Wang, Yapeng Cao, Neal L. Weintraub, Xianqiu Zeng, Yunchao Su, Ramon J. Ayon, and Yong Wang
- Subjects
Male ,medicine.medical_specialty ,Vascular smooth muscle ,Phosphofructokinase-2 ,Hypertension, Pulmonary ,Proinflammatory cytokine ,Rats, Sprague-Dawley ,Mice ,Western blot ,Internal medicine ,medicine ,Animals ,Humans ,Endothelium ,Hypoxia ,Lung ,Multidisciplinary ,PDGFB ,medicine.diagnostic_test ,Chemistry ,Cell growth ,Hypoxia (medical) ,medicine.disease ,Pulmonary hypertension ,Rats ,Mice, Inbred C57BL ,Disease Models, Animal ,Endocrinology ,medicine.anatomical_structure ,PNAS Plus ,Gene Knockdown Techniques ,medicine.symptom ,Glycolysis - Abstract
Increased glycolysis in the lung vasculature has been connected to the development of pulmonary hypertension (PH). We therefore investigated whether glycolytic regulator 6-phosphofructo-2-kinase/fructose-2, 6-bisphosphatase (PFKFB3)-mediated endothelial glycolysis plays a critical role in the development of PH. Heterozygous global deficiency of Pfkfb3 protected mice from developing hypoxia-induced PH, and administration of the PFKFB3 inhibitor 3PO almost completely prevented PH in rats treated with Sugen 5416/hypoxia, indicating a causative role of PFKFB3 in the development of PH. Immunostaining of lung sections and Western blot with isolated lung endothelial cells showed a dramatic increase in PFKFB3 expression and activity in pulmonary endothelial cells of rodents and humans with PH. We generated mice that were constitutively or inducibly deficient in endothelial Pfkfb3 and found that these mice were incapable of developing PH or showed slowed PH progression. Compared with control mice, endothelial Pfkfb3-knockout mice exhibited less severity of vascular smooth muscle cell proliferation, endothelial inflammation, and leukocyte recruitment in the lungs. In the absence of PFKFB3, lung endothelial cells from rodents and humans with PH produced lower levels of growth factors (such as PDGFB and FGF2) and proinflammatory factors (such as CXCL12 and IL1β). This is mechanistically linked to decreased levels of HIF2A in lung ECs following PFKFB3 knockdown. Taken together, these results suggest that targeting PFKFB3 is a promising strategy for the treatment of PH.
- Published
- 2019
36. Neurofibromin is a novel regulator of Ras-induced reactive oxygen species production in mice and humans
- Author
-
David J. Fulton, Hanfang Zhang, Julie A. Mund, Farlyn Z. Hudson, Brandon D. Downing, Waylan K. Bessler, Valerie Harris, Yusi Wang, Jamie Case, David A. Ingram, and Brian K. Stansfield
- Subjects
0301 basic medicine ,Neointima ,MAPK/ERK pathway ,Heterozygote ,congenital, hereditary, and neonatal diseases and abnormalities ,Neurofibromatosis 1 ,DNA damage ,Myocytes, Smooth Muscle ,medicine.disease_cause ,Biochemistry ,Article ,Proto-Oncogene Proteins p21(ras) ,Mice ,03 medical and health sciences ,Physiology (medical) ,medicine ,Animals ,Humans ,Carotid Stenosis ,neoplasms ,Protein kinase B ,Cell Proliferation ,chemistry.chemical_classification ,Reactive oxygen species ,Neurofibromin 1 ,NADPH oxidase ,biology ,Macrophages ,eye diseases ,nervous system diseases ,Oxidative Stress ,030104 developmental biology ,chemistry ,NADPH Oxidase 2 ,Immunology ,biology.protein ,Cancer research ,Reactive Oxygen Species ,Oxidative stress ,DNA Damage - Abstract
Neurofibromatosis type 1 (NF1) predisposes individuals to early and debilitating cardiovascular disease. Loss of function mutations in the NF1 tumor suppressor gene, which encodes the protein neurofibromin, leads to accelerated p21(Ras) activity and phosphorylation of multiple downstream kinases, including Erk and Akt. Nf1 heterozygous (Nf1(+/-)) mice develop a robust neointima that mimics human disease. Monocytes/macrophages play a central role in NF1 arterial stenosis as Nf1 mutations in myeloid cells alone are sufficient to reproduce the enhanced neointima observed in Nf1(+/-) mice. Though the molecular mechanisms underlying NF1 arterial stenosis remain elusive, macrophages are important producers of reactive oxygen species (ROS) and Ras activity directly regulates ROS production. Here, we use compound mutant and lineage-restricted mice to demonstrate that Nf1(+/-) macrophages produce excessive ROS, which enhance Nf1(+/-) smooth muscle cell proliferation in vitro and in vivo. Further, use of a specific NADPH oxidase-2 inhibitor to limit ROS production prevents neointima formation in Nf1(+/-) mice. Finally, mononuclear cells from asymptomatic NF1 patients have increased oxidative DNA damage, an indicator of chronic exposure to oxidative stress. These data provide genetic and pharmacologic evidence that excessive exposure to oxidant species underlie NF1 arterial stenosis and provide a platform for designing novels therapies and interventions.
- Published
- 2016
- Full Text
- View/download PDF
37. Increased Muscle Mass Restores a Healthy Vascular Endothelial Cell Phenotype in Obesity
- Author
-
Caleb A. Padgett, Joshua T. Butcher, David J. Fulton, James D. Mintz, David W. Stepp, and Zachary L. Corley
- Subjects
medicine.medical_specialty ,business.industry ,Muscle mass ,medicine.disease ,Biochemistry ,Obesity ,Phenotype ,Endothelial stem cell ,Endocrinology ,Internal medicine ,Genetics ,medicine ,business ,Molecular Biology ,Biotechnology - Published
- 2020
- Full Text
- View/download PDF
38. Mechanisms of Myosteatosis in Obesity and the Effects of Muscle Hypertrophy
- Author
-
David J. Fulton, Zachary L. Corley, James D. Mintz, David W. Stepp, and Caleb A. Padgett
- Subjects
medicine.medical_specialty ,Endocrinology ,business.industry ,Internal medicine ,Genetics ,medicine ,medicine.disease ,business ,Molecular Biology ,Biochemistry ,Obesity ,Biotechnology ,Muscle hypertrophy - Published
- 2020
- Full Text
- View/download PDF
39. Ablation of myeloid adenosine kinase epigenetically suppresses atherosclerosis in apolipoprotein E-deficient mice
- Author
-
Chaodong Wu, Neal L. Weintraub, Xiaofei An, David J. Fulton, Qiuhua Yang, Yiming Xu, Tao Wang, Yaqi Zhou, Min Zhang, Mei Hong, Lei Huang, Yapeng Cao, Xiaoyu Zhang, Jiean Xu, Zhen Han, Yuqing Huo, Zhiping Liu, and Xianqiu Zeng
- Subjects
0301 basic medicine ,Apolipoprotein E ,Male ,Myeloid ,Mice, Knockout, ApoE ,Aortic Diseases ,Adenosine kinase ,Article ,Epigenesis, Genetic ,03 medical and health sciences ,medicine ,Animals ,Adenosine Kinase ,Aorta ,Cells, Cultured ,Foam cell ,ATP Binding Cassette Transporter, Subfamily G, Member 1 ,biology ,Chemistry ,DNA Methylation ,Atherosclerosis ,Adenosine receptor ,Molecular biology ,Adenosine ,Plaque, Atherosclerotic ,ADK ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Cholesterol ,biology.protein ,lipids (amino acids, peptides, and proteins) ,Female ,Cardiology and Cardiovascular Medicine ,Intracellular ,medicine.drug ,Foam Cells ,Signal Transduction - Abstract
Objective— Monocyte-derived foam cells are one of the key players in the formation of atherosclerotic plaques. Adenosine receptors and extracellular adenosine have been demonstrated to modulate foam cell formation. ADK (adenosine kinase) is a major enzyme regulating intracellular adenosine levels, but its functional role in myeloid cells remains poorly understood. To enhance intracellular adenosine levels in myeloid cells, ADK was selectively deleted in novel transgenic mice using Cre-LoxP technology, and foam cell formation and the development of atherosclerotic lesions were determined. Approach and Results— ADK was upregulated in macrophages on ox-LDL (oxidized low-density lipoprotein) treatment in vitro and was highly expressed in foam cells in atherosclerotic plaques. Atherosclerotic mice deficient in ADK in myeloid cells were generated by breeding floxed ADK (ADK F/F ) mice with LysM-Cre (myeloid-specific Cre recombinase expressing) mice and ApoE −/− (apolipoprotein E deficient) mice. Mice absent ADK in myeloid cells exhibited much smaller atherosclerotic plaques compared with controls. In vitro assays showed that ADK deletion or inhibition resulted in increased intracellular adenosine and reduced DNA methylation of the ABCG1 (ATP-binding cassette transporter G1) gene. Loss of methylation was associated with ABCG1 upregulation, enhanced cholesterol efflux, and eventually decreased foam cell formation. Conclusions— Augmentation of intracellular adenosine levels through ADK knockout in myeloid cells protects ApoE −/− mice against atherosclerosis by reducing foam cell formation via the epigenetic regulation of cholesterol trafficking. ADK inhibition is a promising approach for the treatment of atherosclerotic diseases.
- Published
- 2018
40. Transcription factor TEAD1 is essential for vascular development by promoting vascular smooth muscle differentiation
- Author
-
Luyi Yu, Qin Yin, Kunzhe Dong, Zeqi Zheng, Tong Wen, Jingtian Peng, Wei Zhang, Xiangqin He, David J. Fulton, Jinhua Liu, Quansheng Du, Jiliang Zhou, Hongbo Xin, Islam Osman, and Guoqing Hu
- Subjects
0301 basic medicine ,Male ,Vascular smooth muscle ,Cell ,Biology ,Muscle, Smooth, Vascular ,Article ,Transcriptome ,03 medical and health sciences ,Mice ,0302 clinical medicine ,medicine ,Animals ,Molecular Biology ,Transcription factor ,TEAD1 ,Mice, Knockout ,TEA Domain Transcription Factors ,Cell Differentiation ,Cell Biology ,musculoskeletal system ,Cell biology ,DNA-Binding Proteins ,030104 developmental biology ,medicine.anatomical_structure ,Myocardin ,Hippo signaling ,030220 oncology & carcinogenesis ,Knockout mouse ,cardiovascular system ,Female ,Gene Deletion ,Transcription Factors - Abstract
TEAD1 (TEA domain transcription factor 1), a transcription factor known for the functional output of Hippo signaling, is important for tumorigenesis. However, the role of TEAD1 in the development of vascular smooth muscle cell (VSMC) is unknown. To investigate cell-specific role of Tead1, we generated cardiomyocyte (CMC) and VSMC-specific Tead1 knockout mice. We found CMC/VSMC-specific deletion of Tead1 led to embryonic lethality by E14.5 in mice due to hypoplastic cardiac and vascular walls, as a result of impaired CMC and VSMC proliferation. Whole transcriptome analysis revealed that deletion of Tead1 in CMCs/VSMCs downregulated expression of muscle contractile genes and key transcription factors including Pitx2c and myocardin. In vitro studies demonstrated that PITX2c and myocardin rescued TEAD1-dependent defects in VSMC differentiation. We further identified Pitx2c as a novel transcriptional target of TEAD1, and PITX2c exhibited functional synergy with myocardin by directly interacting with myocardin, leading to augment the differentiation of VSMC. In summary, our study reveals a critical role of Tead1 in cardiovascular development in mice, but also identifies a novel regulatory mechanism, whereby Tead1 functions upstream of the genetic regulatory hierarchy for establishing smooth muscle contractile phenotype.
- Published
- 2018
41. Increased Muscle Mass Protects Against Hypertension and Renal Injury in Obesity
- Author
-
Shuiqing Qiu, David W. Stepp, James D. Mintz, David J. Fulton, Joshua T. Butcher, Sebastian Larion, and Ling Ruan
- Subjects
Glycosuria ,medicine.medical_specialty ,Mice, Obese ,Blood Pressure ,030209 endocrinology & metabolism ,Myostatin ,Glycosuria, Renal ,030204 cardiovascular system & hematology ,Kidney ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Risk Factors ,Weight loss ,nicotinamide‐adenine dinucleotide phosphate, reduced form, oxidase 4 ,Internal medicine ,medicine ,Animals ,Obesity ,skeletal muscle ,Renal Insufficiency, Chronic ,Sodium Chloride, Dietary ,Muscle, Skeletal ,Exercise ,Original Research ,Mice, Knockout ,biology ,business.industry ,Skeletal muscle ,musculoskeletal system ,medicine.disease ,3. Good health ,medicine.anatomical_structure ,Blood pressure ,Endocrinology ,High Blood Pressure ,NADPH Oxidase 4 ,Hypertension ,Body Composition ,Albuminuria ,biology.protein ,hyperglycemia ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,business ,Kidney disease - Abstract
Background Obesity compromises cardiometabolic function and is associated with hypertension and chronic kidney disease. Exercise ameliorates these conditions, even without weight loss. Although the mechanisms of exercise's benefits remain unclear, augmented lean body mass is a suspected mechanism. Myostatin is a potent negative regulator of skeletal muscle mass that is upregulated in obesity and downregulated with exercise. The current study tested the hypothesis that deletion of myostatin would increase muscle mass and reduce blood pressure and kidney injury in obesity. Methods and Results Myostatin knockout mice were crossed to db/db mice, and metabolic and cardiovascular functions were examined. Deletion of myostatin increased skeletal muscle mass by ≈50% to 60% without concomitant weight loss or reduction in fat mass. Increased blood pressure in obesity was prevented by the deletion of myostatin, but did not confer additional benefit against salt loading. Kidney injury was evident because of increased albuminuria, which was abolished in obese mice lacking myostatin. Glycosuria, total urine volume, and whole kidney NOX‐4 levels were increased in obesity and prevented by myostatin deletion, arguing that increased muscle mass provides a multipronged defense against renal dysfunction in obese mice. Conclusions These experimental observations suggest that loss of muscle mass is a novel risk factor in obesity‐derived cardiovascular dysfunction. Interventions that increase muscle mass, either through exercise or pharmacologically, may help limit cardiovascular disease in obese individuals.
- Published
- 2018
- Full Text
- View/download PDF
42. Abstract 043: Endothelial Adenosine Kinase Deficiency Ameliorates Diet-induced Insulin Resistance
- Author
-
Xianqiu Zeng, David J. Fulton, Mei Hong, Jiean Xu, Yuqing Huo, Xiaoyu Zhang, Yaqi Zhou, Min Zhang, Qiuhua Yang, Lina Wang, Yapeng Cao, Zsolt Bagi, Yong Wang, Yiming Xu, Qian Ma, and Zhiping Liu
- Subjects
biology ,Endothelial nitric oxide ,Adenosine kinase ,Pharmacology ,medicine.disease ,Adenosine ,Nitric oxide ,Bioavailability ,chemistry.chemical_compound ,Insulin resistance ,chemistry ,medicine ,biology.protein ,Endothelial dysfunction ,Cardiology and Cardiovascular Medicine ,medicine.drug - Abstract
Background: Obesity is associated with endothelial dysfunction characterized by reduced bioavailability of nitric oxide (NO). It is well known that adenosine regulates endothelial nitric oxide release and vasodilation. However, it is unclear whether intracellular adenosine is able to modulate endothelial nitric oxide production and diet-induced metabolic disorders. This study is to investigate the effect of elevated intracellular adenosine caused by adenosine kinase (ADK) deletion or knockdown on endothelial nitric oxide production and diet-induced insulin resistance as well as the pathways associated to this effect. Methods and results: Endothelial-specific ADK knockout mice fed a high-fat diet show decreased fasting blood glucose and insulin, suppressed adipose tissue inflammation and hepatic steatosis, increased skeletal muscle arteriole vasodilation in an eNOS dependent manner. Mechanistically, ADK knockdown in Human Umbilical Vein Endothelial Cells (HUVECs) with adenoviral ADK shRNA elevated both intracellular and extracellular adenosine, and consequently increased endothelial nitric oxide synthase (eNOS) expression and activation, resulting in an increase in NO production, insulin uptake and vasodilatory-stimulated phosphoprotein (VASP) phosphorylation. Treatment of HUVECs with adenosine A2b receptor antagonist MRS 1754 abolished ADK-knockdown induced eNOS expression. eNOS phosphorylation in ADK-knockdown HUVECs were not interfered with adenosine receptors antagonists. Conclusion: ADK knockdown-mediated elevation of intracellular adenosine in endothelial cells ameliorates diet-induced insulin resistance and metabolic disorders. This is attributed to an enhancement of NO production caused by increased eNOS activation and expression, the latter is regulated via endothelial adenosine A2b receptor.
- Published
- 2018
- Full Text
- View/download PDF
43. Abstract 056: Endothelial 6-Phosphofructo-2-Kinase/Fructose-2,6-Bisphosphatase, Isoform 3 (PFKFB3) Deficiency Inhibits Hypoxia-Induced Pulmonary Hypertension in Mice
- Author
-
David J. Fulton, Mei Hong, Neal L. Weintraub, Yong Wang, Jiean Xu, Qinkai Li, Yuqing Huo, Yiming Xu, Zhiping Liu, Yapeng Cao, Qiuhua Yang, Lina Wang, Yunchao Su, and Chaodong Wu
- Subjects
Gene isoform ,medicine.medical_specialty ,business.industry ,Proliferative disease ,Small pulmonary arteries ,Inflammation ,Hypoxia (medical) ,medicine.disease ,Pulmonary hypertension ,Endocrinology ,Internal medicine ,medicine.artery ,Pulmonary artery ,medicine ,6 phosphofructo 2 kinase fructose 2 6 bisphosphatase ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,business - Abstract
Background: Pulmonary artery hypertension (PAH) is a severe proliferative disease characterized by the remodeling of small pulmonary arteries leading to a progressive increase in pulmonary vascular resistance and ultimately to right ventricular failure and death. Many studies have indicated that endothelial dysfunction is a key element in the pathogenesis of this disease. In this study, we investigated whether endothelial glycolysis plays a critical role in the development of hypoxia-induced PAH by knocking down endothelial PFKFB3 (encoding 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase, isoform 3), a critical regulator of glycolysis. Methods and Results: Immunostaining results showed that pulmonary artery endothelial PFKFB3 was increased in the lung of PAH patients. Mice deficient in endothelial Pfkfb3 showed decreased right ventricular systolic pressure, attenuated right ventricular hypertrophy and normal morphology of distal pulmonary arteries after four weeks of hypoxia compared with control mice. Furthermore, PFKFB3-deficient pulmonary arterial ECs secreted less growth factors, which ameliorated the proliferation of pulmonary artery smooth muscle cells. Additionally, the levels of endothelial derived inflammatory cytokines were also decreased. Besides, the inhibitory effect of endothelial Pfkfb3 supression on PAH formation were demonstrated in Sugen 5416/hypoxia rat pulmonary hypertension model with 3PO, a specific PFKFB3 inhibitor. Mechanistically, genetic deletion of PFKFB3 or 3PO treatment decreased the TNF-α induced NLRP3 inflammasome activation in pulmonary arterial ECs. Conclusions: Endothelial PFKFB3 inhibition is able to ameliorate the development of hypoxia-induced pulmonary hypertension.
- Published
- 2018
- Full Text
- View/download PDF
44. Myostatin Deletion Prevents Kidney Specific Increases in NOX4 in Obesity and Protects Against Renal Injury and Hypertension
- Author
-
David J. Fulton, Sebastian Larion, David W. Stepp, Joshua T. Butcher, and James D. Mintz
- Subjects
medicine.medical_specialty ,Kidney ,biology ,business.industry ,NOX4 ,Myostatin ,medicine.disease ,Biochemistry ,Obesity ,Endocrinology ,medicine.anatomical_structure ,Renal injury ,Internal medicine ,Genetics ,biology.protein ,medicine ,business ,Molecular Biology ,Biotechnology - Published
- 2018
- Full Text
- View/download PDF
45. Red Blood Cell Dysfunction Induced by High-Fat Diet
- Author
-
Víctor M. Blanco, Wilson Tong, Robert Sturm, Abigail Peairs, Danielle Coyle, Andra L. Blomkalns, W. Sean Davidson, Tyler W. Benson, Bruce J. Aronow, Theodosia A. Kalfa, Stephen Haigh, David Manka, Mary B. Palascak, Dusten Unruh, Ryan D. Keil, Ramprasad Srinivasan, David J. Fulton, David Y. Hui, Vladimir Y. Bogdanov, Julia E. Brittain, Tapan K. Chatterjee, Robert S. Franco, Neal L. Weintraub, and Neil Batra
- Subjects
medicine.medical_specialty ,Chemokine ,biology ,business.industry ,Phagocytosis ,Monocyte ,medicine.disease ,Proinflammatory cytokine ,Red blood cell ,Endocrinology ,medicine.anatomical_structure ,Physiology (medical) ,Internal medicine ,medicine ,biology.protein ,Macrophage ,Endothelial dysfunction ,Cardiology and Cardiovascular Medicine ,business ,Ex vivo - Abstract
Background— High-fat diet (HFD) promotes endothelial dysfunction and proinflammatory monocyte activation, which contribute to atherosclerosis in obesity. We investigated whether HFD also induces the dysfunction of red blood cells (RBCs), which serve as a reservoir for chemokines via binding to Duffy antigen receptor for chemokines (DARC). Methods and Results— A 60% HFD for 12 weeks, which produced only minor changes in lipid profile in C57/BL6 mice, markedly augmented the levels of monocyte chemoattractant protein-1 bound to RBCs, which in turn stimulated macrophage migration through an endothelial monolayer. Levels of RBC-bound KC were also increased by HFD. These effects of HFD were abolished in DARC –/– mice. In RBCs from HFD-fed wild-type and DARC –/– mice, levels of membrane cholesterol and phosphatidylserine externalization were increased, fostering RBC-macrophage inflammatory interactions and promoting macrophage phagocytosis in vitro. When labeled ex vivo and injected into wild-type mice, RBCs from HFD-fed mice exhibited ≈3-fold increase in splenic uptake. Finally, RBCs from HFD-fed mice induced increased macrophage adhesion to the endothelium when they were incubated with isolated aortic segments, indicating endothelial activation. Conclusions— RBC dysfunction, analogous to endothelial dysfunction, occurs early during diet-induced obesity and may serve as a mediator of atherosclerosis. These findings may have implications for the pathogenesis of atherosclerosis in obesity, a worldwide epidemic.
- Published
- 2015
- Full Text
- View/download PDF
46. Obesity Alters the Peripheral Circadian Clock in the Aorta and Microcirculation
- Author
-
Shuiqing Qiu, David W. Stepp, David J Fulton, Suwan Thirawarapan, Nitirut Nernpermpisooth, Daniel Rudic, Wisuda Suvitayavat, and James D. Mintz
- Subjects
Male ,medicine.medical_specialty ,Physiology ,Circadian clock ,Biology ,Article ,Mice ,Enos ,Circadian Clocks ,Physiology (medical) ,Internal medicine ,Gene expression ,medicine ,Animals ,Obesity ,Circadian rhythm ,Molecular Biology ,Aorta ,NPAS2 ,Microcirculation ,biology.organism_classification ,PER2 ,CLOCK ,Endocrinology ,Gene Expression Regulation ,Cardiology and Cardiovascular Medicine ,PER1 - Abstract
Objective Perturbation of daily rhythm increases cardiovascular risk. The aim of this study was to determine whether obesity alters circadian gene expression and microvascular function in lean mice and obese (db/db) mice. Methods Mice were subjected to normal LD or DD to alter circadian rhythm. Metabolic parameters and microvascular vasoreactivity were evaluated. Array studies were conducted in the am and pm cycles to assess the rhythmicity of the entire genomics. Rhythmic expression of specific clock genes (Bmal1, Clock, Npas2, Per1, Per2, and Cry1), clock output genes (dbp), and vascular relaxation-related genes (eNOS, GTPCH1) were assessed. Results Obesity was associated with metabolic dysfunction and impaired endothelial dilation in the microvasculature. Circadian rhythm of gene expression was suppressed 80% in both macro- and microcirculations of obese mice. Circadian disruption with DD increased fasting serum glucose and HbA1c in obese but not lean mice. Endothelium-dependent dilation was attenuated in obese mice and in lean mice subjected to DD. Rhythmic expression of per1 and dbp was depressed in obesity. Expression of eNOS expression was suppressed and GTPCH1 lost rhythmic expression both in obesity and by constant darkness. Conclusion These results suggest that obesity reduces circadian gene expression in concert with impaired endothelial function. The causal relationship remains to be determined.
- Published
- 2015
- Full Text
- View/download PDF
47. Regulation of endothelial intracellular adenosine via adenosine kinase epigenetically modulates vascular inflammation
- Author
-
Qinkai Li, Yuqing Huo, Chaodong Wu, Zheng Dong, Haroldo A. Toque, Mei Hong, Xuejun Jiang, Neal L. Weintraub, Xiaofei An, Qiuhua Yang, Yiming Xu, David J. Fulton, Siyuan Yan, Zhiping Liu, Detlev Boison, Yaqi Zhou, Zsolt Bagi, Yong Wang, and Xianqiu Zeng
- Subjects
0301 basic medicine ,Adenosine ,Mice, Knockout, ApoE ,General Physics and Astronomy ,Cardiovascular ,Epigenesis, Genetic ,Mice ,2.1 Biological and endogenous factors ,lcsh:Science ,Cerebral Cortex ,Multidisciplinary ,biology ,Cell biology ,Vascular endothelial growth factor B ,Reperfusion Injury ,Gene Knockdown Techniques ,medicine.symptom ,Intracellular ,medicine.drug ,ApoE ,Science ,Knockout ,Inflammation ,Adenosine kinase ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Genetic ,medicine ,Animals ,Inflammatory and Immune System ,Adenosine Kinase ,Adenosylhomocysteinase ,Endothelial Cells ,General Chemistry ,medicine.disease ,Adenosine A3 receptor ,Atherosclerosis ,ADK ,030104 developmental biology ,Gene Expression Regulation ,Immunology ,biology.protein ,Blood Vessels ,lcsh:Q ,Reperfusion injury ,Epigenesis - Abstract
The molecular mechanisms underlying vascular inflammation and associated inflammatory vascular diseases are not well defined. Here we show that endothelial intracellular adenosine and its key regulator adenosine kinase (ADK) play important roles in vascular inflammation. Pro-inflammatory stimuli lead to endothelial inflammation by increasing endothelial ADK expression, reducing the level of intracellular adenosine in endothelial cells, and activating the transmethylation pathway through increasing the association of ADK with S-adenosylhomocysteine (SAH) hydrolase (SAHH). Increasing intracellular adenosine by genetic ADK knockdown or exogenous adenosine reduces activation of the transmethylation pathway and attenuates the endothelial inflammatory response. In addition, loss of endothelial ADK in mice leads to reduced atherosclerosis and affords protection against ischemia/reperfusion injury of the cerebral cortex. Taken together, these results demonstrate that intracellular adenosine, which is controlled by the key molecular regulator ADK, influences endothelial inflammation and vascular inflammatory diseases., The molecular mechanisms underlying vascular inflammation are unclear. Here the authors show that pro-inflammatory stimuli lead to endothelial inflammation by increasing adenosine kinase expression, and that its knockdown in endothelial cells inhibits atherosclerosis and cerebral ischemic injury in mice.
- Published
- 2017
- Full Text
- View/download PDF
48. Effect of myostatin deletion on cardiac and microvascular function
- Author
-
David J. Fulton, Bianca N. Islam, Sebastian Larion, James D. Mintz, David W. Stepp, Cameron G. McCarthy, Joshua T. Butcher, M. Irfan Ali, Lauren G. Fox, and Merry W. Ma
- Subjects
0301 basic medicine ,Male ,Physiology ,Adrenergic ,Hemodynamics ,Vasodilation ,Blood Pressure ,Myostatin ,030204 cardiovascular system & hematology ,Cardiovascular Physiology ,lcsh:Physiology ,Mice ,0302 clinical medicine ,Heart Rate ,Ventricular Function ,Augmented muscle mass ,Original Research ,Mice, Inbred ICR ,lcsh:QP1-981 ,biology ,exercise ,Heart ,Adrenergic beta-Agonists ,Coronary Vessels ,Propranolol ,Cardiology ,Cardiac function curve ,medicine.medical_specialty ,Skeletal Muscle ,Adrenergic beta-Antagonists ,β‐adrenergic ,03 medical and health sciences ,nitric oxide ,Physiology (medical) ,Internal medicine ,medicine.artery ,Heart rate ,medicine ,Animals ,coronary microvasculature ,Muscle, Skeletal ,Aorta ,business.industry ,Isoproterenol ,medicine.disease ,Exercise Metabolism ,030104 developmental biology ,Heart failure ,Microvessels ,biology.protein ,cardiac function ,business ,Gene Deletion - Abstract
The objective of this study is to test the hypothesis that increased muscle mass has positive effects on cardiovascular function. Specifically, we tested the hypothesis that increases in lean body mass caused by deletion of myostatin improves cardiac performance and vascular function. Echocardiography was used to quantify left ventricular function at baseline and after acute administration of propranolol and isoproterenol to assess β ‐adrenergic reactivity. Additionally, resistance vessels in several beds were removed, cannulated, pressurized to 60 mmHg and reactivity to vasoactive stimuli was assessed. Hemodynamics were measured using in vivo radiotelemetry. Myostatin deletion results in increased fractional shortening at baseline. Additionally, arterioles in the coronary and muscular microcirculations are more sensitive to endothelial‐dependent dilation while nonmuscular beds or the aorta were unaffected. β ‐adrenergic dilation was increased in both coronary and conduit arteries, suggesting a systemic effect of increased muscle mass on vascular function. Overall hemodynamics and physical characteristics (heart weight and size) remained unchanged. Myostatin deletion mimics in part the effects of exercise on cardiovascular function. It significantly increases lean muscle mass and results in muscle‐specific increases in endothelium‐dependent vasodilation. This suggests that increases in muscle mass may serve as a buffer against pathological states that specifically target cardiac function (heart failure), the β ‐adrenergic system (age), and nitric oxide bio‐availability (atherosclerosis). Taken together, pharmacological inhibition of the myostatin pathway could prove an excellent mechanism by which the benefits of exercise can be conferred in patients that are unable to exercise.
- Published
- 2017
49. Intracellular adenosine regulates epigenetic programming in endothelial cells to promote angiogenesis
- Author
-
Jiean Xu, Detlev Boison, Yaqi Zhou, Yiming Xu, Christian Weber, Mei Hong, Nasrul Hoda, Qiuhua Yang, Lina Wang, Siyuan Yan, Xuejun Jiang, Yue Pan, Yuqing Huo, Neal L. Weintraub, Xiaoling Wang, Xianqiu Zeng, Zsolt Bagi, Zhiping Liu, Yong Wang, Xiaofei An, David J. Fulton, Chaodong Wu, Yapeng Cao, and Qinkai Li
- Subjects
0301 basic medicine ,Vascular Biology & Angiogenesis ,Adenosine ,Angiogenesis ,Bisulfite sequencing ,Neovascularization, Physiologic ,Adenosine kinase ,030204 cardiovascular system & hematology ,Biology ,adenosine kinase ,Cardiovascular ,Chromatin, Epigenetics, Genomics & Functional Genomics ,Medical and Health Sciences ,Epigenesis, Genetic ,Promoter Regions ,03 medical and health sciences ,Mice ,angiogenesis ,0302 clinical medicine ,Genetic ,Genetics ,medicine ,Human Umbilical Vein Endothelial Cells ,2.1 Biological and endogenous factors ,Animals ,Humans ,Aetiology ,Promoter Regions, Genetic ,Physiologic ,Research Articles ,Aorta ,Neovascularization ,Gene knockdown ,DNA methylation ,Biological Sciences ,Vascular Endothelial Growth Factor Receptor-2 ,endothelial cells ,ADK ,030104 developmental biology ,Cancer research ,biology.protein ,Molecular Medicine ,Intracellular ,Research Article ,medicine.drug ,Epigenesis - Abstract
The nucleoside adenosine is a potent regulator of vascular homeostasis, but it remains unclear how expression or function of the adenosine‐metabolizing enzyme adenosine kinase (ADK) and the intracellular adenosine levels influence angiogenesis. We show here that hypoxia lowered the expression of ADK and increased the levels of intracellular adenosine in human endothelial cells. Knockdown (KD) of ADK elevated intracellular adenosine, promoted proliferation, migration, and angiogenic sprouting in human endothelial cells. Additionally, mice deficient in endothelial ADK displayed increased angiogenesis as evidenced by the rapid development of the retinal and hindbrain vasculature, increased healing of skin wounds, and prompt recovery of arterial blood flow in the ischemic hindlimb. Mechanistically, hypomethylation of the promoters of a series of pro‐angiogenic genes, especially for VEGFR2 in ADK KD cells, was demonstrated by the Infinium methylation assay. Methylation‐specific PCR, bisulfite sequencing, and methylated DNA immunoprecipitation further confirmed hypomethylation in the promoter region of VEGFR2 in ADK‐deficient endothelial cells. Accordingly, loss or inactivation of ADK increased VEGFR2 expression and signaling in endothelial cells. Based on these findings, we propose that ADK downregulation‐induced elevation of intracellular adenosine levels in endothelial cells in the setting of hypoxia is one of the crucial intrinsic mechanisms that promote angiogenesis.
- Published
- 2017
- Full Text
- View/download PDF
50. Extracellular adenosine-induced Rac1 activation in pulmonary endothelium: Molecular mechanisms and barrier-protective role
- Author
-
Kyung Mi Kim, Stephen M. Black, Mary Cherian-Shaw, David J. Fulton, Alexander D. Verin, and Anita Kovacs-Kasa
- Subjects
0301 basic medicine ,rac1 GTP-Binding Protein ,VAV2 ,Adenosine ,Physiology ,Clinical Biochemistry ,RAC1 ,030204 cardiovascular system & hematology ,Pulmonary Artery ,Article ,Cell Line ,Dephosphorylation ,03 medical and health sciences ,0302 clinical medicine ,Extracellular ,medicine ,Cyclic AMP ,Electric Impedance ,Guanine Nucleotide Exchange Factors ,Humans ,T-Lymphoma Invasion and Metastasis-inducing Protein 1 ,Proto-Oncogene Proteins c-vav ,Lung ,Chemistry ,Endothelial Cells ,Cell Biology ,Adenosine receptor ,Cell biology ,Endothelial stem cell ,030104 developmental biology ,Guanine nucleotide exchange factor ,Endothelium, Vascular ,medicine.drug ,Signal Transduction - Abstract
We have previously shown that Gs-coupled adenosine receptors (A2a) are primarily involved in adenosine-induced human pulmonary artery endothelial cell (HPAEC) barrier enhancement. However, the downstream events that mediate the strengthening of the endothelial cell (EC) barrier via adenosine signaling are largely unknown. In the current study, we tested the overall hypothesis that adenosine-induced Rac1 activation and EC barrier enhancement is mediated by Gs-dependent stimulation of cAMP-dependent Epac1-mediated signaling cascades. Adenoviral transduction of HPAEC with constitutively-active (C/A) Rac1 (V12Rac1) significantly increases transendothelial electrical resistance (TER) reflecting an enhancement of the EC barrier. Conversely, expression of an inactive Rac1 mutant (N17Rac1) decreases TER reflecting a compromised EC barrier. The adenosine-induced increase in TER was accompanied by activation of Rac1, decrease in contractility (MLC dephosphorylation), but not Rho inhibition. Conversely, inhibition of Rac1 activity attenuates adenosine-induced increase in TER. We next examined the role of cAMP-activated Epac1 and its putative downstream targets Rac1, Vav2, Rap1, and Tiam1. Depletion of Epac1 attenuated the adenosine-induced Rac1 activation and the increase in TER. Furthermore, silencing of Rac1 specific guanine nucleotide exchange factors (GEFs), Vav2 and Rap1a expression significantly attenuated adenosine-induced increases in TER and activation of Rac1. Depletion of Rap1b only modestly impacted adenosine-induced increases in TER and Tiam1 depletion had no effect on adenosine-induced Rac1 activation and TER. Together these data strongly suggest that Rac1 activity is required for adenosine-induced EC barrier enhancement and that the activation of Rac1 and ability to strengthen the EC barrier depends, at least in part, on cAMP-dependent Epac1/Vav2/Rap1-mediated signaling.
- Published
- 2017
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.