Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
J Biol Chem ; 298(9): 102347, 2022 09.
Article in English | MEDLINE | ID: mdl-35963433

ABSTRACT

Cell death-inducing DNA fragmentation factor-like effector C (CIDEC) expression in adipose tissue positively correlates with insulin sensitivity in obese humans. Further, E186X, a single-nucleotide CIDEC variant is associated with lipodystrophy, hypertriglyceridemia, and insulin resistance. To establish the unknown mechanistic link between CIDEC and maintenance of systemic glucose homeostasis, we generated transgenic mouse models expressing CIDEC (Ad-CIDECtg) and CIDEC E186X variant (Ad-CIDECmut) transgene specifically in the adipose tissue. We found that Ad-CIDECtg but not Ad-CIDECmut mice were protected against high-fat diet-induced glucose intolerance. Furthermore, we revealed the role of CIDEC in lipid metabolism using transcriptomics and lipidomics. Serum triglycerides, cholesterol, and low-density lipoproteins were lower in high-fat diet-fed Ad-CIDECtg mice compared to their littermate controls. Mechanistically, we demonstrated that CIDEC regulates the enzymatic activity of adipose triglyceride lipase via interacting with its activator, CGI-58, to reduce free fatty acid release and lipotoxicity. In addition, we confirmed that CIDEC is indeed a vital regulator of lipolysis in adipose tissue of obese humans, and treatment with recombinant CIDEC decreased triglyceride breakdown in visceral human adipose tissue. Our study unravels a central pathway whereby adipocyte-specific CIDEC plays a pivotal role in regulating adipose lipid metabolism and whole-body glucose homeostasis. In summary, our findings identify human CIDEC as a potential 'drug' or a 'druggable' target to reverse obesity-induced lipotoxicity and glucose intolerance.


Subject(s)
Glucose Intolerance , Insulin Resistance , Animals , Cholesterol , Diet, High-Fat/adverse effects , Fatty Acids, Nonesterified , Glucose , Glucose Intolerance/genetics , Glucose Intolerance/prevention & control , Humans , Insulin Resistance/genetics , Lipase/genetics , Lipid Metabolism , Lipoproteins, LDL/metabolism , Mice , Nucleotides/metabolism , Obesity/genetics , Proteins/metabolism , Transgenes , Triglycerides
2.
Arterioscler Thromb Vasc Biol ; 41(3): 1239-1250, 2021 03.
Article in English | MEDLINE | ID: mdl-33504180

ABSTRACT

OBJECTIVE: There is growing recognition that adipose tissue-derived proatherogenic mediators contribute to obesity-related cardiovascular disease. We sought to characterize regional differences in perivascular adipose tissue (PVAT) phenotype in relation to atherosclerosis susceptibility. Approach and Results: We examined thoracic PVAT samples in 34 subjects (body mass index 32±6 kg/m2, age 59±11 years) undergoing valvular, aortic, or coronary artery bypass graft surgeries and performed transcriptomic characterization using whole-genome expression profiling and quantitative polymerase chain reaction analyses. We identified a highly inflamed region of PVAT surrounding the human aortic root in close proximity to coronary takeoff and adjoining epicardial fat. In subjects undergoing coronary artery bypass graft, we found 300 genes significantly upregulated (false discovery rate Q<0.1) in paired samples of PVAT surrounding the aortic root compared with nonatherosclerotic left internal mammary artery. Genes encoding proteins mechanistically implicated in atherogenesis were enriched in aortic PVAT consisting of signaling pathways linked to inflammation, WNT (wingless-related integration site) signaling, matrix remodeling, coagulation, and angiogenesis. Overexpression of several proatherogenic transcripts, including IL1ß, CCL2 (MCP-1), and IL6, were confirmed by quantitative polymerase chain reaction and significantly bolstered in coronary artery disease subjects. Angiographic coronary artery disease burden quantified by the Gensini score positively correlated with the expression of inflammatory genes in PVAT. Moreover, periaortic adipose inflammation was markedly higher in obese subjects with striking upregulation (≈8-fold) of IL1ß expression compared to nonobese individuals. CONCLUSIONS: Proatherogenic mediators that originate from dysfunctional PVAT may contribute to vascular disease mechanisms in human vessels. Moreover, PVAT may adopt detrimental properties under obese conditions that play a key role in the pathophysiology of ischemic heart disease. Graphic Abstract: A graphic abstract is available for this article.


Subject(s)
Adipose Tissue/pathology , Myocardial Ischemia/pathology , Adipose Tissue/metabolism , Aged , Aorta/metabolism , Aorta/pathology , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Chemokine CCL2/genetics , Female , Gene Expression Profiling , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Inflammation Mediators/metabolism , Interleukin-1beta/genetics , Interleukin-6/genetics , Male , Middle Aged , Myocardial Ischemia/genetics , Myocardial Ischemia/metabolism , Obesity/genetics , Obesity/metabolism , Obesity/pathology , Up-Regulation , Wnt Signaling Pathway
3.
Am J Physiol Endocrinol Metab ; 316(2): E168-E177, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30576243

ABSTRACT

Obesity, particularly visceral adiposity, has been linked to mitochondrial dysfunction and increased oxidative stress, which have been suggested as mechanisms of insulin resistance. The mechanism(s) behind this remains incompletely understood. In this study, we hypothesized that mitochondrial complex II dysfunction plays a role in impaired insulin sensitivity in visceral adipose tissue of subjects with obesity. We obtained subcutaneous and visceral adipose tissue biopsies from 43 subjects with obesity (body mass index ≥ 30 kg/m2) during planned bariatric surgery. Compared with subcutaneous adipose tissue, visceral adipose tissue exhibited decreased complex II activity, which was restored with the reducing agent dithiothreitol (5 mM) ( P < 0.01). A biotin switch assay identified that cysteine oxidative posttranslational modifications (OPTM) in complex II subunit A (succinate dehydrogenase A) were increased in visceral vs. subcutaneous fat ( P < 0.05). Insulin treatment (100 nM) stimulated complex II activity in subcutaneous fat ( P < 0.05). In contrast, insulin treatment of visceral fat led to a decrease in complex II activity ( P < 0.01), which was restored with addition of the mitochondria-specific oxidant scavenger mito-TEMPO (10 µM). In a cohort of 10 subjects with severe obesity, surgical weight loss decreased OPTM and restored complex II activity, exclusively in the visceral depot. Mitochondrial complex II may be an unrecognized and novel mediator of insulin resistance associated with visceral adiposity. The activity of complex II is improved by weight loss, which may contribute to metabolic improvements associated with bariatric surgery.


Subject(s)
Electron Transport Complex II/metabolism , Insulin Resistance , Intra-Abdominal Fat/metabolism , Obesity/metabolism , Protein Processing, Post-Translational , Adult , Bariatric Surgery , Cysteine , Female , Humans , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Intra-Abdominal Fat/drug effects , Male , Middle Aged , Obesity/surgery , Organophosphorus Compounds/pharmacology , Oxidation-Reduction , Piperidines/pharmacology , Subcutaneous Fat/drug effects , Subcutaneous Fat/metabolism
4.
Am J Physiol Heart Circ Physiol ; 313(1): H200-H206, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28411232

ABSTRACT

Experimental studies have suggested that Wingless-related integration site 5A (WNT5A) is a proinflammatory secreted protein that is associated with metabolic dysfunction in obesity. Impaired angiogenesis in fat depots has been implicated in the development of adipose tissue capillary rarefaction, hypoxia, inflammation, and metabolic dysfunction. We have recently demonstrated that impaired adipose tissue angiogenesis is associated with overexpression of antiangiogenic factor VEGF-A165b in human fat and the systemic circulation. In the present study, we postulated that upregulation of WNT5A is associated with angiogenic dysfunction and examined its role in regulating VEGF-A165b expression in human obesity. We biopsied subcutaneous and visceral adipose tissue from 38 obese individuals (body mass index: 44 ± 7 kg/m2, age: 37 ± 11 yr) during planned bariatric surgery and characterized depot-specific protein expression of VEGF-A165b and WNT5A using Western blot analysis. In both subcutaneous and visceral fat, VEGF-A165b expression correlated strongly with WNT5A protein (r = 0.9, P < 0.001). In subcutaneous adipose tissue where angiogenic capacity is greater than in the visceral depot, exogenous human recombinant WNT5A increased VEGF-A165b expression in both whole adipose tissue and isolated vascular endothelial cell fractions (P < 0.01 and P < 0.05, respectively). This was associated with markedly blunted angiogenic capillary sprout formation in human fat pad explants. Moreover, recombinant WNT5A increased secretion of soluble fms-like tyrosine kinase-1, a negative regulator of angiogenesis, in the sprout media (P < 0.01). Both VEGF-A165b-neutralizing antibody and secreted frizzled-related protein 5, which acts as a decoy receptor for WNT5A, significantly improved capillary sprout formation and reduced soluble fms-like tyrosine kinase-1 production (P < 0.05). We demonstrated a significant regulatory nexus between WNT5A and antiangiogenic VEGF-A165b in the adipose tissue of obese subjects that was linked to angiogenic dysfunction. Elevated WNT5A expression in obesity may function as a negative regulator of angiogenesis.NEW & NOTEWORTHY Wingless-related integration site 5a (WNT5A) negatively regulates adipose tissue angiogenesis via VEGF-A165b in human obesity.


Subject(s)
Adipose Tissue/blood supply , Adipose Tissue/physiopathology , Angiogenesis Inhibitors/metabolism , Neovascularization, Pathologic/physiopathology , Obesity/physiopathology , Wnt-5a Protein/metabolism , Adult , Female , Humans , Male , Transcriptome , Vascular Endothelial Growth Factor A/metabolism
5.
Arterioscler Thromb Vasc Biol ; 35(6): 1498-506, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25908760

ABSTRACT

OBJECTIVE: Increased visceral adiposity has been closely linked to insulin resistance, endothelial dysfunction, and cardiometabolic disease in obesity, but pathophysiological mechanisms are poorly understood. We sought to investigate mechanisms of vascular insulin resistance by characterizing depot-specific insulin responses and gain evidence that altered functionality of transcription factor forkhead box O-1 (FOXO-1) may play an important role in obesity-related endothelial dysfunction. APPROACH AND RESULTS: We intraoperatively collected paired subcutaneous and visceral adipose tissue samples from 56 severely obese (body mass index, 43 ± 7 kg/m(2)) and 14 nonobese subjects during planned surgical operations, and characterized depot-specific insulin-mediated responses using Western blot and quantitative immunofluorescence techniques. Insulin signaling via phosphorylation of FOXO-1 and consequent endothelial nitric oxide synthase stimulation was selectively impaired in the visceral compared with subcutaneous adipose tissue and endothelial cells of obese subjects. In contrast, tissue actions of insulin were preserved in nonobese individuals. Pharmacological antagonism with AS1842856 and biological silencing using small interfering RNA-mediated FOXO-1 knockdown reversed insulin resistance and restored endothelial nitric oxide synthase activation in the obese. CONCLUSIONS: We observed profound endothelial insulin resistance in the visceral adipose tissue of obese humans which improved with FOXO-1 inhibition. FOXO-1 modulation may represent a novel therapeutic target to diminish vascular insulin resistance. In addition, characterization of endothelial insulin resistance in the adipose microenvironment may provide clues to mechanisms of systemic disease in human obesity.


Subject(s)
Endothelium, Vascular/physiopathology , Forkhead Transcription Factors/metabolism , Insulin Resistance/physiology , Obesity/physiopathology , Adult , Delayed-Action Preparations , Endothelial Cells/metabolism , Enzyme Activation/drug effects , Female , Forkhead Box Protein O1 , Forkhead Transcription Factors/antagonists & inhibitors , Humans , Insulin/pharmacology , Male , Middle Aged , Nitric Oxide Synthase Type III/metabolism , Phosphorylation , Quinolones/pharmacology
6.
Vasc Med ; 21(6): 489-496, 2016 12.
Article in English | MEDLINE | ID: mdl-27688298

ABSTRACT

Obesity is associated with the development of vascular insulin resistance; however, pathophysiological mechanisms are poorly understood. We sought to investigate the role of WNT5A-JNK in the regulation of insulin-mediated vasodilator responses in human adipose tissue arterioles prone to endothelial dysfunction. In 43 severely obese (BMI 44±11 kg/m2) and five metabolically normal non-obese (BMI 26±2 kg/m2) subjects, we isolated arterioles from subcutaneous and visceral fat during planned surgeries. Using videomicroscopy, we examined insulin-mediated, endothelium-dependent vasodilator responses and characterized adipose tissue gene and protein expression using real-time polymerase chain reaction and Western blot analyses. Immunofluorescence was used to quantify endothelial nitric oxide synthase (eNOS) phosphorylation. Insulin-mediated vasodilation was markedly impaired in visceral compared to subcutaneous vessels from obese subjects (p<0.001), but preserved in non-obese individuals. Visceral adiposity was associated with increased JNK activation and elevated expression of WNT5A and its non-canonical receptors, which correlated negatively with insulin signaling. Pharmacological JNK antagonism with SP600125 markedly improved insulin-mediated vasodilation by sixfold (p<0.001), while endothelial cells exposed to recombinant WNT5A developed insulin resistance and impaired eNOS phosphorylation (p<0.05). We observed profound vascular insulin resistance in the visceral adipose tissue arterioles of obese subjects that was associated with up-regulated WNT5A-JNK signaling and impaired endothelial eNOS activation. Pharmacological JNK antagonism markedly improved vascular endothelial function, and may represent a potential therapeutic target in obesity-related vascular disease.


Subject(s)
Adiposity , Arterioles/drug effects , Endothelium, Vascular/drug effects , Insulin Resistance , Insulin/pharmacology , Intra-Abdominal Fat/blood supply , JNK Mitogen-Activated Protein Kinases/metabolism , Obesity/enzymology , Vasodilation/drug effects , Vasodilator Agents/pharmacology , Wnt Signaling Pathway/drug effects , Wnt-5a Protein/metabolism , Adolescent , Adult , Arterioles/enzymology , Arterioles/physiopathology , Case-Control Studies , Cells, Cultured , Endothelium, Vascular/enzymology , Endothelium, Vascular/physiopathology , Female , Humans , In Vitro Techniques , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Male , Middle Aged , Nitric Oxide Synthase Type III/metabolism , Obesity/physiopathology , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Young Adult
7.
Circulation ; 130(13): 1072-80, 2014 Sep 23.
Article in English | MEDLINE | ID: mdl-25116954

ABSTRACT

BACKGROUND: Experimental studies suggest that visceral adiposity and adipose tissue dysfunction play a central role in obesity-related cardiometabolic complications. Impaired angiogenesis in fat has been implicated in the development of adipose tissue hypoxia, capillary rarefaction, inflammation, and metabolic dysregulation, but pathophysiological mechanisms remain unknown. In this study, we examined the role of a novel antiangiogenic isoform of vascular endothelial growth factor-A (VEGF-A), VEGF-A165b, in human obesity. METHODS AND RESULTS: We biopsied paired subcutaneous and visceral adipose tissue in 40 obese subjects (body mass index, 45±8 kg/m(2); age, 45±11 years) during bariatric surgery and characterized depot-specific adipose tissue angiogenic capacity using an established ex vivo assay. Visceral adipose tissue exhibited significantly blunted angiogenic growth compared with subcutaneous fat (P<0.001) that was associated with marked tissue upregulation of VEGF-A165b (P=0.004). The extent of VEGF-A165b expression correlated negatively with angiogenic growth (r=-0.6, P=0.006). Although recombinant VEGF-A165b significantly impaired angiogenesis, targeted inhibition of VEGF-A165b with neutralizing antibody stimulated fat pad neovascularization and restored VEGF receptor activation. Blood levels of VEGF-A165b were significantly higher in obese subjects compared with lean control subjects (P=0.02), and surgical weight loss induced a marked decline in serumVEGF-A165b (P=0.003). CONCLUSIONS: We demonstrate that impaired adipose tissue angiogenesis is associated with overexpression of a novel antiangiogenic factor, VEGF-A165b, that may play a pathogenic role in human adiposopathy. Moreover, systemic upregulation of VEGF-A165b in circulating blood may have wider-ranging implications beyond the adipose milieu. VEGF-A165b may represent a novel area of investigation to gain further understanding of mechanisms that modulate the cardiometabolic consequences of obesity.


Subject(s)
Angiogenesis Inhibitors/physiology , Obesity/physiopathology , Vascular Endothelial Growth Factor A/physiology , Adult , Biopsy , Female , Humans , Intra-Abdominal Fat/pathology , Intra-Abdominal Fat/physiopathology , Male , Middle Aged , Obesity/pathology , Protein Isoforms/physiology , Retrospective Studies , Signal Transduction/physiology , Subcutaneous Fat/pathology , Subcutaneous Fat/physiopathology , Vascular Endothelial Growth Factor Receptor-2/physiology
8.
Mediators Inflamm ; 2015: 106237, 2015.
Article in English | MEDLINE | ID: mdl-26663986

ABSTRACT

BACKGROUND: Dysregulated lipolysis has been implicated in mechanisms of cardiometabolic disease and inflammation in obesity. PURPOSE: We sought to examine the effect of bariatric weight loss on adipose tissue lipolytic gene expression and their relationship to systemic metabolic parameters in obese subjects. METHODS/RESULTS: We biopsied subcutaneous adipose tissue in 19 obese individuals (BMI 42 ± 5 kg/m(2), 79% female) at baseline and after a mean period of 8 ± 5 months (range 3-15 months) following bariatric surgery. We performed adipose tissue mRNA expression of proteins involved in triglyceride hydrolysis and correlated their weight loss induced alterations with systemic parameters associated with cardiovascular disease risk. mRNA transcripts of adipose triglyceride lipase (ATGL), hormone-sensitive lipase (HSL), and lipid droplet proteins comparative gene identification 58 (CGI-58) and perilipin increased significantly after weight loss (p < 0.05 for all). ATGL expression correlated inversely with plasma triglyceride (TG), hemoglobin A1C (HbA1C), and glucose, and HSL expression correlated negatively with glucose, while CGI-58 was inversely associated with HbA1C. CONCLUSION: We observed increased expression of adipose tissue lipolytic genes following bariatric weight loss which correlated inversely with systemic markers of lipid and glucose metabolism. Functional alterations in lipolysis in human adipose tissue may play a role in shaping cardiometabolic phenotypes in human obesity.


Subject(s)
Adipose Tissue/metabolism , Bariatric Surgery , Lipolysis , Obesity/surgery , Transcriptome , Female , Humans , Lipase/genetics , Male , Obesity/metabolism , Sterol Esterase/genetics , Weight Loss
9.
Am J Cardiol ; 210: 201-207, 2024 01 01.
Article in English | MEDLINE | ID: mdl-37863116

ABSTRACT

Accumulation of ectopic pericardial adipose tissue has been associated with cardiovascular complications which, in part, may relate to adipose-derived factors that regulate vascular responses and angiogenesis. We sought to characterize adipose tissue microvascular angiogenic capacity in subjects who underwent elective cardiac surgeries including aortic, valvular, and coronary artery bypass grafting. Pericardial adipose tissue was collected intraoperatively and examined for angiogenic capacity. Capillary sprouting was significantly blunted (twofold, p <0.001) in subjects with coronary artery disease (CAD) (age 60 ± 9 years, body mass index [BMI] 32 ± 4 kg/m2, low-density lipoprotein cholesterol [LDL-C] 95 ± 46 mg/100 ml, n = 29) compared with age-, BMI-, and LDL-C matched subjects without angiographic obstructive CAD (age 59 ± 10 y, BMI 35 ± 9 kg/m2, LDL-C 101 ± 40 mg/100 ml, n = 12). For potential mechanistic insight, we performed mRNA expression analyses using quantitative real-time polymerase chain reaction and observed no significant differences in pericardial fat gene expression of proangiogenic mediators vascular endothelial growth factor-A (VEGF-A), fibroblast growth factor-2 (FGF-2), and angiopoietin-1 (angpt1), or anti-angiogenic factors soluble fms-like tyrosine kinase-1 (sFlt-1) and endostatin. In contrast, mRNA expression of anti-angiogenic thrombospondin-1 (TSP-1) was significantly upregulated (twofold, p = 0.008) in CAD compared with non-CAD subjects, which was confirmed by protein western-immunoblot analysis. TSP-1 gene knockdown using short hairpin RNA lentiviral delivery significantly improved angiogenic deficiency in CAD (p <0.05). In conclusion, pericardial fat in subjects with CAD may be associated with an antiangiogenic profile linked to functional defects in vascularization capacity. Local paracrine actions of TSP-1 in adipose depots surrounding the heart may play a role in mechanisms of ischemic heart disease.


Subject(s)
Coronary Artery Disease , Myocardial Ischemia , Humans , Middle Aged , Aged , Vascular Endothelial Growth Factor A/metabolism , Thrombospondin 1/genetics , Thrombospondin 1/metabolism , Cholesterol, LDL/metabolism , Myocardial Ischemia/complications , Adipose Tissue , Coronary Artery Disease/etiology , RNA, Messenger/metabolism
10.
Diabetes ; 72(1): 19-32, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36256836

ABSTRACT

Cell death-inducing DNA fragmentation factor-α-like effector C (CIDEC), originally identified to be a lipid droplet-associated protein in adipocytes, positively associates with insulin sensitivity. Recently, we discovered that it is expressed abundantly in human endothelial cells and regulates vascular function. The current study was designed to characterize the physiological effects and molecular actions of endothelial CIDEC in the control of vascular phenotype and whole-body glucose homeostasis. To achieve this, we generated a humanized mouse model expressing endothelial-specific human CIDEC (E-CIDECtg). E-CIDECtg mice exhibited protection against high-fat diet-induced glucose intolerance, insulin resistance, and dyslipidemia. Moreover, these mice displayed improved insulin signaling and endothelial nitric oxide synthase activation, enhanced endothelium-dependent vascular relaxation, and improved vascularization of adipose tissue, skeletal muscle, and heart. Mechanistically, we identified a novel interplay of CIDEC-vascular endothelial growth factor A (VEGFA)-vascular endothelial growth factor receptor 2 (VEGFR2) that reduced VEGFA and VEGFR2 degradation, thereby increasing VEGFR2 activation. Overall, our results demonstrate a protective role of endothelial CIDEC against obesity-induced metabolic and vascular dysfunction, in part, by modulation of VEGF signaling. These data suggest that CIDEC may be investigated as a potential future therapeutic target for mitigating obesity-related cardiometabolic disease.


Subject(s)
Insulin Resistance , Vascular Endothelial Growth Factor A , Humans , Mice , Animals , Diet, High-Fat/adverse effects , Endothelial Cells/metabolism , Obesity/metabolism , Endothelium/metabolism
11.
J Lipid Res ; 52(9): 1693-701, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21743036

ABSTRACT

Recent studies have established SIRT1 as an important regulator of lipid metabolism, although the mechanism of its action at the molecular level has not been revealed. Here, we show that knockdown of SIRT1 with the help of small hairpin RNA decreases basal and isoproterenol-stimulated lipolysis in cultured adipocytes. This effect is attributed, at least in part, to the suppression of the rate-limiting lipolytic enzyme, adipose triglyceride lipase (ATGL), at the level of transcription. Mechanistically, SIRT1 controls acetylation status and functional activity of FoxO1 that directly binds to the ATGL promoter and regulates ATGL gene transcription. We have also found that depletion of SIRT1 decreases AMP-dependent protein kinase (AMPK) activity in adipocytes. To determine the input of AMPK in regulation of lipolysis, we have established a stable adipose cell line that expresses a dominant-negative α1 catalytic subunit of AMPK under the control of the inducible TET-OFF lentiviral expression vector. Reduction of AMPK activity does not have a significant effect on the rates of lipolysis in this cell model. We conclude, therefore, that SIRT1 controls ATGL transcription primarily by deacetylating FoxO1.


Subject(s)
Adipocytes/enzymology , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Enzymologic , Lipase/metabolism , Lipid Metabolism , Lipolysis/physiology , Sirtuin 1/metabolism , 3T3-L1 Cells , Adenylate Kinase/metabolism , Adipocytes/cytology , Adipocytes/physiology , Animals , Down-Regulation , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Gene Knockdown Techniques , Lipase/genetics , Mice , PPAR gamma/metabolism , Promoter Regions, Genetic , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Sirtuin 1/genetics
12.
JAMA Netw Open ; 4(7): e2115267, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34251443

ABSTRACT

Importance: Bariatric surgical weight loss is associated with reduced cardiovascular mortality; however, the mechanisms underlying this association are incompletely understood. Objectives: To identify variables associated with vascular remodeling after bariatric surgery and to examine how sex, race, and metabolic status are associated with microvascular and macrovascular outcomes. Design, Setting, and Participants: This population-based longitudinal cohort included 307 individuals who underwent bariatric surgery. Participants were enrolled in the bariatric weight loss program at Boston Medical Center, a large, multi-ethnic urban hospital, with presurgical and postsurgical assessments. Data were collected from December 11, 2001 to August 27, 2019. Data were analyzed in September 2019. Exposure: Bariatric surgery. Main Outcomes and Measures: Flow-mediated dilation (FMD) and reactive hyperemia (RH) (as measures of macrovascular and microvascular function, respectively) and clinical variables were measured preoperatively at baseline and at least once postoperatively within 12 months of the bariatric intervention. Results: A total of 307 participants with obesity (mean [SD] age, 42 [12] years; 246 [80%] women; 199 [65%] White; mean [SD] body mass index, 46 [8]) were enrolled in this study. Bariatric surgery was associated with significant weight loss and improved macrovascular and microvascular function across subgroups of sex, race, and traditional metabolic syndrome (mean [SD] pre- vs postsurgery weight: 126 [25] kg vs 104 [25] kg; P < .001; mean [SD] pre- vs postsurgery FMD: 9.1% [5.3] vs 10.2% [5.1]; P < .001; mean [SD] pre- vs postsurgery RH: 764% [400] vs 923% [412]; P < .001). Factors associated with change in vascular phenotype correlated most strongly with adiposity markers and several metabolic variables depending on vascular territory (eg, association of weight change with change in RH: estimate, -3.2; 95% CI, -4.7 to -1.8; association of hemoglobin A1c with change in FMD: estimate, -0.5; 95% CI, -0.95 to -0.05). While changes in macrovascular function among individuals with metabolically healthy obesity were not observed, the addition of biomarker assessment using high-sensitivity C-reactive protein plasma levels greater than 2 mg/dL identified participants with seemingly metabolically healthy obesity who had low-grade inflammation and achieved microvascular benefit from weight loss surgery. Conclusions and Relevance: The findings of this study suggest that bariatric intervention is associated with weight loss and favorable remodeling of the vasculature among a wide range of individuals with cardiovascular risk. Moreover, differences in arterial responses to weight loss surgery by metabolic status were identified, underscoring heterogeneity in physiological responses to adiposity change and potential activation of distinct pathological pathways in clinical subgroups. As such, individuals with metabolically healthy obesity represent a mixed population that may benefit from more refined phenotypic classification.


Subject(s)
Bariatric Surgery/adverse effects , Cardiovascular Diseases/etiology , Obesity/surgery , Treatment Outcome , Adult , Bariatric Surgery/methods , Bariatric Surgery/standards , Body Mass Index , Boston/epidemiology , Cardiovascular Diseases/epidemiology , Female , Humans , Longitudinal Studies , Male , Middle Aged , Obesity/complications
13.
Curr Obes Rep ; 8(3): 255-261, 2019 Sep.
Article in English | MEDLINE | ID: mdl-30919313

ABSTRACT

PURPOSE OF REVIEW: Obesity is a pandemic, yet preventable healthcare problem. Insulin resistance, diabetes mellitus, dyslipidemia, and cardiovascular complications are core manifestation of obesity. While adipose tissue is a primary site of energy storage, it is also an endocrine organ, secreting a large number of adipokines and cytokines. Nonetheless in obesity, the secretion of cytokines and free fatty acids increases significantly and is associated with the degree of adiposity and insulin resistance. Fat-specific protein 27 (FSP27) has emerged as one of the major proteins that promote physiological storage of fat in adipose tissue. RECENT FINDINGS: Review of number of recent findings suggests that FSP27 plays a crucial role in physiological storage of fat within the adipose tissue especially in humans. However, in disease conditions such as obesity, FSP27 may contribute to ectopic fat accumulation in non-adipose tissue. More studies are required to highlight the tissue-specific role of FSP27, especially in humans.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Diabetes Mellitus/metabolism , Obesity/metabolism , Adipokines/metabolism , Adipose Tissue/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Diabetes Complications/metabolism , Humans , Insulin Resistance , Liver/metabolism , Obesity/complications
14.
J Am Heart Assoc ; 8(11): e011431, 2019 06 04.
Article in English | MEDLINE | ID: mdl-31433737

ABSTRACT

Background Pathophysiological mechanisms that connect obesity to cardiovascular disease are incompletely understood. FSP27 (Fat-specific protein 27) is a lipid droplet-associated protein that regulates lipolysis and insulin sensitivity in adipocytes. We unexpectedly discovered extensive FSP27 expression in human endothelial cells that is downregulated in association with visceral obesity. We sought to examine the functional role of FSP27 in the control of vascular phenotype. Methods and Results We biopsied paired subcutaneous and visceral fat depots from 61 obese individuals (body mass index 44±8 kg/m2, age 48±4 years) during planned bariatric surgery. We characterized depot-specific FSP27 expression in relation to adipose tissue microvascular insulin resistance, endothelial function and angiogenesis, and examined differential effects of FSP27 modification on vascular function. We observed markedly reduced vasodilator and angiogenic capacity of microvessels isolated from the visceral compared with subcutaneous adipose depots. Recombinant FSP27 and/or adenoviral FSP27 overexpression in human tissue increased endothelial nitric oxide synthase phosphorylation and nitric oxide production, and rescued vasomotor and angiogenic dysfunction (P<0.05), while siRNA-mediated FSP27 knockdown had opposite effects. Mechanistically, we observed that FSP27 interacts with vascular endothelial growth factor-A and exerts robust regulatory control over its expression. Lastly, in a subset of subjects followed longitudinally for 12±3 months after their bariatric surgery, 30% weight loss improved metabolic parameters and increased angiogenic capacity that correlated positively with increased FSP27 expression (r=0.79, P<0.05). Conclusions Our data strongly support a key role and functional significance of FSP27 as a critical endogenous modulator of human microvascular function that has not been previously described. FSP27 may serve as a previously unrecognized regulator of arteriolar vasomotor capacity and angiogenesis which are pivotal in the pathogenesis of cardiometabolic diseases linked to obesity.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Cardiovascular Diseases/metabolism , Endothelial Cells/metabolism , Intra-Abdominal Fat/blood supply , Microvessels/metabolism , Neovascularization, Physiologic , Obesity/metabolism , Subcutaneous Fat/blood supply , Vasodilation , Adiposity , Adult , Apoptosis Regulatory Proteins/genetics , Cardiovascular Diseases/etiology , Cardiovascular Diseases/physiopathology , Cells, Cultured , Female , Humans , Male , Microvessels/physiopathology , Middle Aged , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Obesity/complications , Obesity/physiopathology , Phosphorylation , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism
15.
J Nutr Biochem ; 19(2): 129-37, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18061429

ABSTRACT

A blood-brain barrier (BBB) model composed of porcine brain capillary endothelial cells (BCEC) was exposed to a moderately excessive zinc environment (50 micromol/L Zn) in cell culture, and longitudinal measurements were made of zinc transport kinetics, ZnT-1 (SLC30A1) expression and changes in the protein concentration of metallothionein (MT), ZnT-1, ZnT-2 (SLC30A2) and Zip1 (SLC39A1). Zinc release by cells of the BBB model significantly increased after 12-24 h of exposure, but decreased back to control levels after 48-96 h, as indicated by transport across the BBB from both the ablumenal (brain) and the lumenal (blood) directions. Expression of ZnT-1, the zinc export protein, increased by 169% within 12 h, but was no longer different from controls after 24 h. Likewise, ZnT-1 protein content increased transiently after 12 h of exposure, but returned to control levels by 24 h. Capacity for zinc uptake and retention increased from both the lumenal and the ablumenal directions within 12-24 h of exposure and remained elevated. MT and ZnT-2 were elevated within 12 h and remained elevated throughout the study. Zip1 was unchanged by the treatment. The BBB's response to a moderately high zinc environment was dynamic and involved multiple mechanisms. The initial response was to increase the cells' capacity to sequester zinc with additional MT and to increase zinc export with the ZnT-1 protein. But the longer-term strategy involved increasing ZnT-2 transporters, presumably to sequester zinc into intracellular vesicles as a mechanism to protect the brain and to maintain brain zinc homeostasis.


Subject(s)
Blood-Brain Barrier , Carrier Proteins/metabolism , Metallothionein/metabolism , Zinc/metabolism , Amino Acid Sequence , Animals , Base Sequence , Biological Transport , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cells, Cultured , DNA Primers , Homeostasis , Kinetics , Molecular Sequence Data , RNA, Messenger/genetics , Swine , Swine, Miniature
16.
J Vis Exp ; (127)2017 09 29.
Article in English | MEDLINE | ID: mdl-28994775

ABSTRACT

While obesity is closely linked to the development of metabolic and cardiovascular disease, little is known about mechanisms that govern these processes. It is hypothesized that pro-atherogenic mediators released from fat tissues particularly in association with central/visceral adiposity may promote pathogenic vascular changes locally and systemically, and the notion that cardiovascular disease may be the consequence of adipose tissue dysfunction continues to evolve. Here, we describe a unique method of videomicroscopy that involves analysis of vasodilator and vasoconstrictor responses of intact small human arterioles removed from the adipose depot of living human subjects. Videomicroscopy is used to examine functional properties of isolated microvessels in response to pharmacological or physiological stimuli using a pressured system that mimics in vivo conditions. The technique is a useful approach to gain understanding of the pathophysiology and molecular mechanisms that contribute to vascular dysfunction locally within the adipose tissue milieu. Moreover, abnormalities in the adipose tissue microvasculature have also been linked with systemic diseases. We applied this technique to examine depot-specific vascular responses in obese subjects. We assessed endothelium-dependent vasodilation to both increased flow and acetylcholine in adipose arterioles (50 - 350 µm internal diameter, 2 - 3 mm in length) isolated from two different adipose depots during bariatric surgery from the same individual. We demonstrated that arterioles from visceral fat exhibit impaired endothelium-dependent vasodilation compared to vessels isolated from the subcutaneous depot. The findings suggest that the visceral microenvironment is associated with vascular endothelial dysfunction which may be relevant to clinical observation linking increased visceral adiposity to systemic disease mechanisms. The videomicroscopy technique can be used to examine vascular phenotypes from different fat depots as well as compare findings across individuals with different degrees of obesity and metabolic dysfunction. The method can also be used to examine vascular responses longitudinally in response to clinical interventions.


Subject(s)
Adipose Tissue/pathology , Microscopy, Video/methods , Female , Humans , Male
17.
Sci Rep ; 7(1): 17326, 2017 12 11.
Article in English | MEDLINE | ID: mdl-29229927

ABSTRACT

The accumulation of visceral adiposity is strongly associated with systemic inflammation and increased cardiometabolic risk. WNT5A, a non-canonical WNT ligand, has been shown to promote adipose tissue inflammation and insulin resistance in animal studies. Among other non-canonical pathways, WNT5A activates planar cell polarity (PCP) signaling. The current study investigated the potential contribution of non-canonical WNT5A/PCP signaling to visceral adipose tissue (VAT) inflammation and associated metabolic dysfunction in individuals with obesity. VAT and subcutaneous adipose tissue (SAT) samples obtained from subjects undergoing bariatric surgery were analyzed by qRT-PCR for expression of WNT/PCP genes. In vitro experiments were conducted with preadipocytes isolated from VAT and SAT biopsies. The expression of 23 out of 33 PCP genes was enriched in VAT compared to SAT. Strong positive expression correlations of individual PCP genes were observed in VAT. WNT5A expression in VAT, but not in SAT, correlated with indexes of JNK signaling activity, IL6, waist-to-hip ratio and hsCRP. In vitro, WNT5A promoted the expression of IL6 in human preadipocytes. In conclusion, elevated non-canonical WNT5A signaling in VAT contributes to the exacerbated IL-6 production in this depot and the low-grade systemic inflammation typically associated with visceral adiposity.


Subject(s)
Gene Expression Regulation , Panniculitis/metabolism , Subcutaneous Fat/metabolism , Wnt Signaling Pathway , Adult , Female , Humans , Inflammation/metabolism , Inflammation/pathology , Male , Panniculitis/pathology , Subcutaneous Fat/pathology
18.
Expert Rev Endocrinol Metab ; 9(2): 93-95, 2014 Mar.
Article in English | MEDLINE | ID: mdl-30743752

ABSTRACT

The global obesity epidemic has emerged as one of the most important health care problems worldwide. Insulin resistance represents a prevalent pathophysiological abnormality that underlies mechanisms of cardiometabolic disease associated with obesity. Increasing basic, animal, and clinical data support a mechanistic link between insulin resistance and vascular dysfunction, and suggest that improving insulin sensitivity may represent a therapeutic target for combating atherosclerosis and cardiovascular disease. As clinical studies suggest that insulin resistance may play a key role in the cardiovascular benefit achieved with weight loss intervention, we will discuss our clinical perspective and provide evidence that obese individuals with hyperinsulinemia may derive the greatest improvement in vascular function with weight reduction. Lastly, we will address several important unanswered questions in the field that are likely to drive future clinical investigation.

19.
Obesity (Silver Spring) ; 22(2): 349-55, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23640904

ABSTRACT

OBJECTIVE: The purpose of this study was to determine whether cyclooxygenase inhibition improves vascular dysfunction of adipose microvessels from obese humans. DESIGN AND METHODS: In 20 obese subjects (age 37 ± 12 years, BMI 47 ± 8 kg/m²), subcutaneous and visceral fat were collected during bariatric surgery and characterized for adipose depot-specific gene expression, endothelial cell phenotype, and microvascular function. Vasomotor function was assessed in response to endothelium-dependent agonists using videomicroscopy of small arterioles from fat. RESULTS: Arterioles from visceral fat exhibited impaired endothelium-dependent, acetylcholine-mediated vasodilation, compared to the subcutaneous depot (P < 0.001). Expression of mRNA transcripts relevant to the cyclooxygenase pathway was upregulated in visceral compared to subcutaneous fat. Pharmacological inhibition of cyclooxygenase with indomethacin improved endothelium-dependent vasodilator function of arterioles from visceral fat by twofold (P = 0.01), whereas indomethacin had no effect in the subcutaneous depot. Indomethacin increased activation via serine-1177 phosphorylation of endothelial nitric oxide synthase in response to acetylcholine in endothelial cells from visceral fat. Inhibition of endothelial nitric oxide synthase with N(ω)-nitro-L-arginine methyl ester abrogated the effects of cyclooxygenase-inhibition suggesting that vascular actions of indomethacin were related to increased nitric oxide bioavailability. CONCLUSIONS: Our findings suggest that cyclooxygenase-mediated vasoconstrictor prostanoids partly contribute to endothelial dysfunction of visceral adipose arterioles in human obesity.


Subject(s)
Arterioles/drug effects , Cyclooxygenase Inhibitors/pharmacology , Endothelium, Vascular/drug effects , Intra-Abdominal Fat/drug effects , Obesity/drug therapy , Vasoconstriction/drug effects , Vasomotor System/drug effects , Adult , Arterioles/metabolism , Arterioles/pathology , Arterioles/physiopathology , Body Mass Index , Cells, Cultured , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation/drug effects , Humans , Intra-Abdominal Fat/blood supply , Intra-Abdominal Fat/metabolism , Intra-Abdominal Fat/pathology , Male , Microscopy, Video , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type III/chemistry , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Obesity/metabolism , Obesity/pathology , Obesity/physiopathology , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Subcutaneous Fat, Abdominal/blood supply , Subcutaneous Fat, Abdominal/drug effects , Subcutaneous Fat, Abdominal/metabolism , Subcutaneous Fat, Abdominal/pathology , Tissue Culture Techniques , Vasomotor System/metabolism , Vasomotor System/pathology , Vasomotor System/physiopathology
20.
J Am Coll Cardiol ; 62(24): 2297-305, 2013 Dec 17.
Article in English | MEDLINE | ID: mdl-23978693

ABSTRACT

OBJECTIVES: The aim of this study was to determine whether the effects of weight loss on arterial function are differentially modified by insulin status. BACKGROUND: Clinical studies suggest that plasma insulin levels may predict the extent of cardiovascular benefit achieved with weight loss in obese individuals, but mechanisms are currently unknown. METHODS: We prospectively followed 208 overweight or obese patients (body mass index [BMI] ≥25 kg/m(2)) receiving medical/dietary (48%) or bariatric surgical (52%) weight-loss treatment during a median period of 11.7 months (interquartile range: 4.6 to 13 months). We measured plasma metabolic parameters and vascular endothelial function using ultrasound at baseline and following weight-loss intervention and stratified analyses by median plasma insulin levels. RESULTS: Patients age 45 ± 1 years, with BMI 45 ± 9 kg/m(2), experienced 14 ± 14% weight loss during the study period. In individuals with higher baseline plasma insulin levels (above median >12 µIU/ml; n = 99), ≥10% weight loss (compared with <10%) significantly improved brachial artery macrovascular flow-mediated vasodilation and microvascular reactive hyperemia (p < 0.05 for all). By contrast, vascular function did not change significantly in the lower insulin group (≤12 µIU/ml; n = 109) despite a similar degree of weight loss. In analyses using a 5% weight loss cut point, only microvascular responses improved in the higher insulin group (p = 0.02). CONCLUSIONS: Insulin status is an important determinant of the positive effect of weight reduction on vascular function with hyperinsulinemic patients deriving the greatest benefit. Integrated improvement in both microvascular and macrovascular function was associated with ≥10% weight loss. Reversal of insulin resistance and endothelial dysfunction may represent key therapeutic targets for cardiovascular risk reduction in obesity.


Subject(s)
Brachial Artery/diagnostic imaging , Insulin/blood , Obesity/therapy , Overweight/therapy , Weight Loss , Blood Flow Velocity/physiology , Body Mass Index , Cholesterol/blood , Female , Follow-Up Studies , Glycated Hemoglobin/analysis , Homeostasis/physiology , Humans , Hyperemia/physiopathology , Male , Microcirculation/physiology , Middle Aged , Prospective Studies , Regional Blood Flow/physiology , Triglycerides/blood , Ultrasonography, Doppler, Pulsed
SELECTION OF CITATIONS
SEARCH DETAIL