Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
J Lipid Res ; 61(1): 33-44, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31722970

RESUMO

Chronic inflammation contributes to cardiovascular disease. Increased levels of the inflammatory cytokine, TNF-α, are often present in conditions associated with cardiovascular disease risk, and TNF-α induces a number of pro-atherogenic effects in macrovascular endothelial cells, including expression of adhesion molecules and chemokines, and lipoprotein uptake and transcytosis to the subendothelial tissue. However, little is known about the roles of acyl-CoA synthetases (ACSLs), enzymes that esterify free fatty acids into their acyl-CoA derivatives, or about the effects of TNF-α on ACSLs in endothelial cells. Therefore, we investigated the effects of TNF-α on ACSLs and downstream lipids in cultured human coronary artery endothelial cells and human umbilical vein endothelial cells. We demonstrated that TNF-α induces ACSL1, ACSL3, and ACSL5, but not ACSL4, in both cell types. TNF-α also increased oleoyl-CoA levels, consistent with the increased ACSL3 expression. RNA-sequencing demonstrated that knockdown of ACSL3 had no marked effects on the TNF-α transcriptome. Instead, ACSL3 was required for TNF-α-induced lipid droplet formation in cells exposed to oleic acid. These results demonstrate that increased acyl-CoA synthesis as a result of ACSL3 induction is part of the TNF-α response in human macrovascular endothelial cells.


Assuntos
Coenzima A Ligases/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Gotículas Lipídicas/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia , Adulto , Células Cultivadas , Coenzima A Ligases/genética , Células Endoteliais/enzimologia , Feminino , Humanos , Gotículas Lipídicas/metabolismo , Masculino
2.
J Lipid Res ; 58(6): 1174-1185, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28416579

RESUMO

Acyl-CoA thioesterase 7 (ACOT7) is an intracellular enzyme that converts acyl-CoAs to FFAs. ACOT7 is induced by lipopolysaccharide (LPS); thus, we investigated downstream effects of LPS-induced induction of ACOT7 and its role in inflammatory settings in myeloid cells. Enzymatic thioesterase activity assays in WT and ACOT7-deficient macrophage lysates indicated that endogenous ACOT7 contributes a significant fraction of total acyl-CoA thioesterase activity toward C20:4-, C20:5-, and C22:6-CoA, but contributes little activity toward shorter acyl-CoA species. Lipidomic analyses revealed that LPS causes a dramatic increase, primarily in bis(monoacylglycero)phosphate species containing long (≥C20) polyunsaturated acyl-chains in macrophages, and that the limited effect observed by ACOT7 deficiency is restricted to glycerophospholipids containing 20-carbon unsaturated acyl-chains. Furthermore, ACOT7 deficiency did not detectably alter the ability of LPS to induce cytokines or prostaglandin E2 production in macrophages. Consistently, although ACOT7 was induced in macrophages from diabetic mice, hematopoietic ACOT7 deficiency did not alter the stimulatory effect of diabetes on systemic inflammation or atherosclerosis in LDL receptor-deficient mice. Thus, inflammatory stimuli induce ACOT7 and remodeling of phospholipids containing unsaturated long (≥C20)-acyl chains in macrophages, and, although ACOT7 has preferential thioesterase activity toward these lipid species, loss of ACOT7 has no major detrimental effect on macrophage inflammatory phenotypes.≥.


Assuntos
Macrófagos/metabolismo , Palmitoil-CoA Hidrolase/biossíntese , Fosfolipídeos/metabolismo , Animais , Citocinas/biossíntese , Dinoprostona/metabolismo , Indução Enzimática/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Glicerofosfolipídeos/metabolismo , Inflamação/enzimologia , Inflamação/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Camundongos , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Palmitoil-CoA Hidrolase/deficiência , Palmitoil-CoA Hidrolase/genética , Palmitoil-CoA Hidrolase/metabolismo
3.
Ann Rheum Dis ; 76(3): 602-611, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27543414

RESUMO

OBJECTIVES: Recent evidence indicates that high-density lipoprotein (HDL) exerts vasculoprotective activities by promoting activating transcription factor 3 (ATF3), leading to downregulation of toll-like receptor (TLR)-induced inflammatory responses. Systemic lupus erythematosus (SLE) is associated with increased cardiovascular disease risk not explained by the Framingham risk score. Recent studies have indicated oxidised HDL as a possible contributor. We investigated the potential mechanisms by which lupus HDL may lose its anti-inflammatory effects and promote immune dysregulation. METHODS: Control macrophages were challenged with control and SLE HDL in vitro and examined for inflammatory markers by real-time qRT-PCR, confocal microscopy, ELISA and flow cytometry. Lupus-prone mice were treated with an HDL mimetic (ETC-642) in vivo and inflammatory cytokine levels measured by real-time qRT-PCR and ELISA. RESULTS: Compared with control HDL, SLE HDL activates NFκB, promotes inflammatory cytokine production and fails to block TLR-induced inflammation in control macrophages. This failure of lupus HDL to block inflammatory responses is due to an impaired ability to promote ATF3 synthesis and nuclear translocation. This inflammation is dependent on lectin-like oxidised low-density lipoprotein receptor 1 (LOX1R) binding and rho-associated, coiled-coil containing protein kinase 1 and 2 (ROCK1/2) kinase activity. HDL mimetic-treated lupus mice showed significant ATF3 induction and proinflammatory cytokine abrogation. CONCLUSIONS: Lupus HDL promotes proinflammatory responses through NFκB activation and decreased ATF3 synthesis and activity in an LOX1R-dependent and ROCK1/2-dependent manner. HDL mimetics should be explored as potential therapies for inflammation and SLE cardiovascular risk.


Assuntos
Fator 3 Ativador da Transcrição/biossíntese , Citocinas/genética , Lipoproteínas HDL/metabolismo , Lipoproteínas HDL/farmacologia , Lúpus Eritematoso Sistêmico/sangue , RNA Mensageiro/metabolismo , 1,2-Dipalmitoilfosfatidilcolina/farmacologia , Fator 3 Ativador da Transcrição/metabolismo , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Amidas/farmacologia , Animais , Células Cultivadas , Feminino , Humanos , Macrófagos , Camundongos , NF-kappa B/metabolismo , Oxirredução , Peptídeos/farmacologia , Biossíntese de Proteínas/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , Receptores Depuradores Classe A/genética , Receptores Depuradores Classe E/genética , Receptores Depuradores Classe E/metabolismo , Esfingomielinas/farmacologia , Baço/citologia , Receptores Toll-Like/metabolismo , Transcrição Gênica/efeitos dos fármacos , Quinases Associadas a rho/metabolismo
4.
Am J Respir Crit Care Med ; 193(8): 861-8, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-26575972

RESUMO

RATIONALE: Interstitial lung diseases (ILDs) are associated with oxidative stress. Plasma biomarkers that are directly linked to oxidative stress responses in this disease have not been identified. Stable oxidation products of tyrosine residues in proteins may reflect the oxidative microenvironment in the lung or a systemic inflammatory state. OBJECTIVES: To determine if levels of protein tyrosine oxidation are elevated in plasma of patients with ILD compared with an age- and sex-matched healthy control cohort. METHODS: Three tyrosine oxidation products (3-chlorotyrosine, 3-nitrotyrosine, and o,o'-dityrosine) were quantified by tandem mass spectrometry in cellular models, a mouse model of injury-induced fibrosis, and in plasma of healthy control subjects and patients with ILD (n = 42 in each group). MEASUREMENTS AND MAIN RESULTS: Plasma levels of 3-chlorotyrosine, 3-nitrotyrosine, and o,o'-dityrosine were markedly elevated in patients with ILD compared with control subjects with receiver operating characteristic curves separating these groups of 0.872, 0.893, and 0.997, respectively. In a murine model of lung fibrosis, levels of all three oxidative tyrosine modifications were increased in plasma and lung tissue. Cellular models support a critical role for a heme peroxidase and enzymatic sources of reactive oxygen species in the generation of these oxidized products. CONCLUSIONS: We demonstrate an increase in oxidized tyrosine moieties within proteins in the circulating plasma of patients with ILD. These data support the potential for development of oxidative stress-related biomarkers in early diagnosis, prognostication, and/or in evaluating responsiveness to emerging therapies for ILD.


Assuntos
Doenças Pulmonares Intersticiais/sangue , Estresse Oxidativo , Tirosina/análogos & derivados , Animais , Biomarcadores/sangue , Células Cultivadas , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Espectrometria de Massas em Tandem , Tirosina/sangue
5.
Proc Natl Acad Sci U S A ; 109(12): E715-24, 2012 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-22308341

RESUMO

The mechanisms that promote an inflammatory environment and accelerated atherosclerosis in diabetes are poorly understood. We show that macrophages isolated from two different mouse models of type 1 diabetes exhibit an inflammatory phenotype. This inflammatory phenotype associates with increased expression of long-chain acyl-CoA synthetase 1 (ACSL1), an enzyme that catalyzes the thioesterification of fatty acids. Monocytes from humans and mice with type 1 diabetes also exhibit increased ACSL1. Furthermore, myeloid-selective deletion of ACSL1 protects monocytes and macrophages from the inflammatory effects of diabetes. Strikingly, myeloid-selective deletion of ACSL1 also prevents accelerated atherosclerosis in diabetic mice without affecting lesions in nondiabetic mice. Our observations indicate that ACSL1 plays a critical role by promoting the inflammatory phenotype of macrophages associated with type 1 diabetes; they also raise the possibilities that diabetic atherosclerosis has an etiology that is, at least in part, distinct from the etiology of nondiabetic vascular disease and that this difference is because of increased monocyte and macrophage ACSL1 expression.


Assuntos
Aterosclerose/metabolismo , Coenzima A Ligases/metabolismo , Diabetes Mellitus/metabolismo , Macrófagos/citologia , Alelos , Animais , Glicemia/metabolismo , Transplante de Medula Óssea , Feminino , Deleção de Genes , Humanos , Inflamação , Lipídeos/química , Masculino , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Monócitos/citologia , Fenótipo , Receptores de LDL/genética
6.
J Am Soc Nephrol ; 25(11): 2511-25, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24722446

RESUMO

Podocytes are the key cells affected in nephrotic glomerular kidney diseases, and they respond uniformly to injury with cytoskeletal rearrangement. In nephrotic diseases, such as membranous nephropathy and FSGS, persistent injury often leads to irreversible structural damage, whereas in minimal change disease, structural alterations are mostly transient. The factors leading to persistent podocyte injury are currently unknown. Proteolysis is an irreversible process and could trigger persistent podocyte injury through degradation of podocyte-specific proteins. We, therefore, analyzed the expression and functional consequence of the two most prominent proteolytic systems, the ubiquitin proteasome system (UPS) and the autophagosomal/lysosomal system, in persistent and transient podocyte injuries. We show that differential upregulation of both proteolytic systems occurs in persistent human and rodent podocyte injury. The expression of specific UPS proteins in podocytes differentiated children with minimal change disease from children with FSGS and correlated with poor clinical outcome. Degradation of the podocyte-specific protein α-actinin-4 by the UPS depended on oxidative modification in membranous nephropathy. Notably, the UPS was overwhelmed in podocytes during experimental glomerular disease, resulting in abnormal protein accumulation and compensatory upregulation of the autophagosomal/lysosomal system. Accordingly, inhibition of both proteolytic systems enhanced proteinuria in persistent nephrotic disease. This study identifies altered proteolysis as a feature of persistent podocyte injury. In the future, specific UPS proteins may serve as new biomarkers or therapeutic targets in persistent nephrotic syndrome.


Assuntos
Podócitos/metabolismo , Podócitos/patologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteinúria/metabolismo , Proteinúria/patologia , Ubiquitina/metabolismo , Actinina/genética , Actinina/metabolismo , Animais , Autofagia/fisiologia , Linhagem Celular Transformada , Modelos Animais de Doenças , Humanos , Glomérulos Renais/metabolismo , Glomérulos Renais/patologia , Lisossomos/metabolismo , Lisossomos/patologia , Complexo de Endopeptidases do Proteassoma/genética , Proteinúria/genética , Ratos Wistar , Transcriptoma , Ubiquitina/genética , Regulação para Cima/fisiologia
7.
J Biol Chem ; 288(14): 9957-9970, 2013 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-23426369

RESUMO

The enzyme acyl-CoA synthetase 1 (ACSL1) is induced by peroxisome proliferator-activated receptor α (PPARα) and PPARγ in insulin target tissues, such as skeletal muscle and adipose tissue, and plays an important role in ß-oxidation in these tissues. In macrophages, however, ACSL1 mediates inflammatory effects without significant effects on ß-oxidation. Thus, the function of ACSL1 varies in different tissues. We therefore investigated the signals and signal transduction pathways resulting in ACSL1 induction in macrophages as well as the consequences of ACSL1 deficiency for phospholipid turnover in LPS-activated macrophages. LPS, Gram-negative bacteria, IFN-γ, and TNFα all induce ACSL1 expression in macrophages, whereas PPAR agonists do not. LPS-induced ACSL1 expression is dependent on Toll-like receptor 4 (TLR4) and its adaptor protein TRIF (Toll-like receptor adaptor molecule 1) but does not require the MyD88 (myeloid differentiation primary response gene 88) arm of TLR4 signaling; nor does it require STAT1 (signal transducer and activator of transcription 1) for maximal induction. Furthermore, ACSL1 deletion attenuates phospholipid turnover in LPS-stimulated macrophages. Thus, the regulation and biological function of ACSL1 in macrophages differ markedly from that in insulin target tissues. These results suggest that ACSL1 may have an important role in the innate immune response. Further, these findings illustrate an interesting paradigm in which the same enzyme, ACSL1, confers distinct biological effects in different cell types, and these disparate functions are paralleled by differences in the pathways that regulate its expression.


Assuntos
Coenzima A Ligases/metabolismo , Bactérias Gram-Negativas/metabolismo , Lipopolissacarídeos/metabolismo , Macrófagos/metabolismo , Fosfolipídeos/metabolismo , Animais , Células da Medula Óssea/citologia , Feminino , Imunidade Inata , Interferon gama/metabolismo , MAP Quinase Quinase 4/metabolismo , Macrófagos/citologia , Macrófagos Peritoneais/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Transdução de Sinais
8.
Kidney Int ; 86(1): 58-66, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24402087

RESUMO

The endothelial dysfunction of Fabry disease results from α-galactosidase A deficiency leading to the accumulation of globotriaosylceramide. Vasculopathy in the α-galactosidase A null mouse is manifested as oxidant-induced thrombosis, accelerated atherogenesis, and impaired arterial reactivity. To better understand the pathogenesis of Fabry disease in humans, we generated a human cell model by using RNA interference. Hybrid endothelial cells were transiently transfected with small interfering RNA (siRNA) specifically directed against α-galactosidase A. Knockdown of α-galactosidase A was confirmed using immunoblotting and globotriaosylceramide accumulation. Endothelial nitric oxide synthase (eNOS) activity was correspondingly decreased by >60%. Levels of 3-nitrotyrosine (3NT), a specific marker for reactive nitrogen species and quantified using mass spectrometry, increased by 40- to 120-fold without corresponding changes in other oxidized amino acids, consistent with eNOS-derived reactive nitrogen species as the source of the reactive oxygen species. eNOS uncoupling was confirmed by the observed increase in free plasma and protein-bound aortic 3NT levels in the α-galactosidase A knockout mice. Finally, 3NT levels, assayed in biobanked plasma samples from patients with classical Fabry disease, were over sixfold elevated compared with age- and gender-matched controls. Thus, 3NT may serve as a biomarker for the vascular involvement in Fabry disease.


Assuntos
Doença de Fabry/complicações , Doença de Fabry/metabolismo , Tirosina/análogos & derivados , Doenças Vasculares/etiologia , Doenças Vasculares/metabolismo , Adolescente , Adulto , Animais , Biomarcadores/metabolismo , Estudos de Casos e Controles , Linhagem Celular , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Doença de Fabry/genética , Células Endoteliais da Veia Umbilical Humana , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Óxido Nítrico Sintase Tipo III/metabolismo , RNA Interferente Pequeno/genética , Tirosina/metabolismo , Adulto Jovem , alfa-Galactosidase/antagonistas & inibidores , alfa-Galactosidase/genética
9.
Arterioscler Thromb Vasc Biol ; 33(2): 232-40, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23241406

RESUMO

OBJECTIVE: Saturated fatty acids, such as palmitic and stearic acid, cause detrimental effects in endothelial cells and have been suggested to contribute to macrophage accumulation in adipose tissue and the vascular wall, in states of obesity and insulin resistance. Long-chain fatty acids are believed to require conversion into acyl-CoA derivatives to exert most of their detrimental effects, a reaction catalyzed by acyl-CoA synthetases (ACSLs). The objective of this study was to investigate the role of ACSL1, an ACSL isoform previously shown to mediate inflammatory effects in myeloid cells, in regulating endothelial cell responses to a saturated fatty acid-rich environment in vitro and in vivo. METHODS AND RESULTS: Saturated fatty acids caused increased inflammatory activation, endoplasmic reticulum stress, and apoptosis in mouse microvascular endothelial cells. Forced ACSL1 overexpression exacerbated the effects of saturated fatty acids on apoptosis and endoplasmic reticulum stress. However, endothelial ACSL1 deficiency did not protect against the effects of saturated fatty acids in vitro, nor did it protect insulin-resistant mice fed a saturated fatty acid-rich diet from macrophage adipose tissue accumulation or increased aortic adhesion molecule expression. CONCLUSIONS: Endothelial ACSL1 is not required for inflammatory and apoptotic effects of a saturated fatty acid-rich environment.


Assuntos
Apoptose , Coenzima A Ligases/metabolismo , Células Endoteliais/enzimologia , Ácidos Graxos/metabolismo , Inflamação/enzimologia , Obesidade/enzimologia , Acil Coenzima A/metabolismo , Tecido Adiposo/imunologia , Tecido Adiposo/metabolismo , Tecido Adiposo/patologia , Animais , Aorta/metabolismo , Bovinos , Células Cultivadas , Coenzima A Ligases/deficiência , Coenzima A Ligases/genética , Modelos Animais de Doenças , Estresse do Retículo Endoplasmático , Células Endoteliais/imunologia , Células Endoteliais/patologia , Ativação Enzimática , Inflamação/imunologia , Inflamação/patologia , Resistência à Insulina , Molécula 1 de Adesão Intercelular/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Macrófagos/imunologia , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/genética , Obesidade/imunologia , Obesidade/patologia , Palmitoil Coenzima A/metabolismo , Interferência de RNA , Fatores de Tempo , Transfecção , Molécula 1 de Adesão de Célula Vascular/metabolismo
10.
Ann Rheum Dis ; 72(10): 1725-31, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23313808

RESUMO

OBJECTIVE: Phagocyte-derived myeloperoxidase (MPO) and pro-inflammatory high density lipoprotein (HDL) associate with rheumatoid arthritis (RA), but the link between MPO and HDL has not been systematically examined. In this study, we investigated whether MPO can oxidise HDL and determined MPO-specific oxidative signature by apoA-1 by peptide mapping in RA subjects with and without known cardiovascular disease (CVD). METHODS: Two MPO oxidation products, 3-chlorotyrosine and 3-nitrotyrosine, were quantified by tandem mass spectrometry (MS/MS) in in vitro model system studies and in plasma and HDL derived from healthy controls and RA subjects. MPO levels and cholesterol efflux were determined. Site-specific nitration and chlorination of apoA-1 peptides were quantified by MS/MS. RESULTS: RA subjects demonstrated higher levels of MPO, MPO-oxidised HDL and diminished cholesterol efflux. There was marked increase in MPO-specific 3-chlorotyrosine and 3-nitrotyrosine content in HDL in RA subjects consistent with specific targeting of HDL, with increased nitration in RA subjects with CVD. Cholesterol efflux capacity was diminished in RA subjects and correlated inversely with HDL 3-chlorotyrosine suggesting a mechanistic role for MPO. Nitrated HDL was elevated in RACVD subjects compared with RA subjects without CVD. Oxidative peptide mapping revealed site-specific unique oxidation signatures on apoA-1 for RA subjects with and without CVD. CONCLUSIONS: We report an increase in MPO-mediated HDL oxidation that is regiospecific in RA and accentuated in those with CVD. Decreased cholesterol efflux capacity due to MPO-mediated chlorination is a potential mechanism for atherosclerosis in RA and raises the possibility that oxidant resistant forms of HDL may attenuate this increased risk.


Assuntos
Artrite Reumatoide/sangue , Lipoproteínas HDL/sangue , Peroxidase/fisiologia , Adulto , Idoso , Apolipoproteína A-I/sangue , Artrite Reumatoide/complicações , Doenças Cardiovasculares/sangue , Doenças Cardiovasculares/etiologia , Estudos de Casos e Controles , Colesterol/sangue , Feminino , Halogenação/fisiologia , Humanos , Masculino , Pessoa de Meia-Idade , Oxirredução , Mapeamento de Peptídeos/métodos , Peroxidase/sangue , Espectrometria de Massas em Tandem/métodos , Tirosina/análogos & derivados , Tirosina/sangue
11.
Arthritis Rheum ; 64(9): 2975-85, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22549550

RESUMO

OBJECTIVE: Patients with systemic lupus erythematosus (SLE) have a notable increase in atherothrombotic cardiovascular disease (CVD) which is not explained by the Framingham risk equation. In vitro studies indicate that type I interferons (IFNs) may play prominent roles in increased CV risk in SLE. However, the in vivo relevance of these findings, with regard to the development of CVD, has not been characterized. This study was undertaken to examine the role of type I IFNs in endothelial dysfunction, aberrant vascular repair, and atherothrombosis in murine models of lupus and atherosclerosis. METHODS: Lupus-prone New Zealand mixed 2328 (NZM) mice and atherosclerosis-prone apolipoprotein E- knockout (apoE(-/-) ) mice were compared to mice lacking type I IFN receptor (INZM and apoE(-/-) IFNAR(-/-) mice, respectively) with regard to endothelial vasodilatory function, endothelial progenitor cell (EPC) function, in vivo neoangiogenesis, plaque development, and occlusive thrombosis. Similar experiments were performed using NZM and apoE(-/-) mice exposed to an IFNα-containing or empty adenovirus. RESULTS: Loss of type I IFN receptor signaling improved endothelium-dependent vasorelaxation, lipoprotein parameters, EPC numbers and function, and neoangiogenesis in lupus-prone mice, independent of disease activity or sex. Further, acute exposure to IFNα impaired endothelial vasorelaxation and EPC function in lupus-prone and non-lupus-prone mice. Decreased atherosclerosis severity and arterial inflammatory infiltrates and increased neoangiogenesis were observed in apoE(-/-) IFNAR(-/-) mice, compared to apoE(-/-) mice, while NZM and apoE(-/-) mice exposed to IFNα developed accelerated thrombosis and platelet activation. CONCLUSION: These results support the hypothesis that type I IFNs play key roles in the development of premature CVD in SLE and, potentially, in the general population, through pleiotropic deleterious effects on the vasculature.


Assuntos
Aterosclerose/metabolismo , Interferon Tipo I/metabolismo , Lúpus Eritematoso Sistêmico/metabolismo , Placa Aterosclerótica/metabolismo , Trombose/metabolismo , Cicatrização/fisiologia , Animais , Aterosclerose/fisiopatologia , Modelos Animais de Doenças , Progressão da Doença , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Feminino , Lúpus Eritematoso Sistêmico/fisiopatologia , Masculino , Camundongos , Vasodilatação/fisiologia
12.
J Lipid Res ; 52(4): 782-93, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21242590

RESUMO

Long-chain acyl-CoA synthetases (ACSLs) catalyze the thioesterification of long-chain FAs into their acyl-CoA derivatives. Purified ACSL4 is an arachidonic acid (20:4)-preferring ACSL isoform, and ACSL4 is therefore a probable regulator of lipid mediator production in intact cells. Eicosanoids play important roles in vascular homeostasis and disease, yet the role of ACSL4 in vascular cells is largely unknown. In the present study, the ACSL4 splice variant expressed in human arterial smooth muscle cells (SMCs) was identified as variant 1. To investigate the function of ACSL4 in SMCs, ACSL4 variant 1 was overexpressed, knocked-down by small interfering RNA, or its enzymatic activity acutely inhibited in these cells. Overexpression of ACSL4 resulted in a markedly increased synthesis of arachidonoyl-CoA, increased 20:4 incorporation into phosphatidylethanolamine, phosphatidylinositol, and triacylglycerol, and reduced cellular levels of unesterified 20:4. Accordingly, secretion of prostaglandin E2 (PGE2) was blunted in ACSL4-overexpressing SMCs compared with controls. Conversely, acute pharmacological inhibition of ACSL4 activity resulted in increased release of PGE2. However, long-term downregulation of ACSL4 resulted in markedly reduced PGE2 secretion. Thus, ACSL4 modulates PGE2 release from human SMCs. ACSL4 may regulate a number of processes dependent on the release of arachidonic acid-derived lipid mediators in the arterial wall.


Assuntos
Artérias/citologia , Coenzima A Ligases/metabolismo , Dinoprostona/metabolismo , Miócitos de Músculo Liso/metabolismo , Western Blotting , Células Cultivadas , Coenzima A Ligases/genética , Vetores Genéticos/genética , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Retroviridae/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
Hypertension ; 77(2): 393-404, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33390039

RESUMO

Activation of AT1 (type 1 Ang) receptors stimulates cardiomyocyte hypertrophy in vitro. Accordingly, it has been suggested that regression of cardiac hypertrophy associated with renin-Ang system blockade is due to inhibition of cellular actions of Ang II in the heart, above and beyond their effects to reduce pressure overload. We generated 2 distinct mouse lines with cell-specific deletion of AT1A receptors, from cardiomyocytes. In the first line (C-SMKO), elimination of AT1A receptors was achieved using a heterologous Cre recombinase transgene under control of the Sm22 promoter, which expresses in cells of smooth muscle lineage including cardiomyocytes and vascular smooth muscle cells of conduit but not resistance vessels. The second line (R-SMKO) utilized a Cre transgene knocked-in to the Sm22 locus, which drives expression in cardiac myocytes and vascular smooth muscle cells in both conduit and resistance arteries. Thus, although both groups lack AT1 receptors in the cardiomyocytes, they are distinguished by presence (C-SMKO) or absence (R-SMKO) of peripheral vascular responses to Ang II. Similar to wild-types, chronic Ang II infusion caused hypertension and cardiac hypertrophy in C-SMKO mice, whereas both hypertension and cardiac hypertrophy were reduced in R-SMKOs. Thus, despite the absence of AT1A receptors in cardiomyocytes, C-SMKOs develop robust cardiac hypertrophy. By contrast, R-SMKOs developed identical levels of hypertrophy in response to pressure overload-induced by transverse aortic banding. Our findings suggest that direct activation of AT1 receptors in cardiac myocytes has minimal influence on cardiac hypertrophy induced by renin-Ang system activation or pressure overload.


Assuntos
Angiotensina II/farmacologia , Cardiomegalia/genética , Hipertensão/genética , Miócitos Cardíacos/metabolismo , Receptor Tipo 1 de Angiotensina/genética , Sistema Renina-Angiotensina/efeitos dos fármacos , Animais , Cardiomegalia/induzido quimicamente , Cardiomegalia/metabolismo , Hipertensão/induzido quimicamente , Hipertensão/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Miocárdio/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Resistência Vascular/efeitos dos fármacos
14.
J Diabetes Complications ; 31(9): 1437-1443, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28709739

RESUMO

AIMS: To assess the role of oxidative stress in mediating adverse outcomes in metabolic syndrome (MetS) and resultant cardiovascular autonomic neuropathy (CAN), and to evaluate the effects of lifestyle interventions on measures of oxidative stress and CAN in subjects with MetS. METHODS: Pilot study in 25 non-diabetic subjects with MetS (age 49±10years, 76% females) participating in a 24-week lifestyle intervention (supervised aerobic exercise/Mediterranean diet), and 25 age-matched healthy controls. CAN was assessed by cardiovascular reflex tests, heart rate variability (HRV) and PET imaging with sympathetic analog [11C] meta-hydroxyephedrine ([11C]HED). Specific oxidative fingerprints were measured by liquid-chromatography/mass-spectrometry (LC/MS). RESULTS: At baseline, MetS subjects had significantly higher oxidative stress markers [3-nitrotyrosine (234±158 vs. 54±47µmol/mol tyrosine), ortho-tyrosine (59±38 vs. 18±10µmol/molphenylalanine, all P<0.0001], and impaired HRV at rest and during deep breathing (P=0.039 and P=0.021 respectively) compared to controls. Twenty-four-week lifestyle intervention significantly reduced all oxidative stress markers (all P<0.01) but did not change any of the CAN measures. CONCLUSIONS: Subjects with MetS present with signs of CAN and increased oxidative stress in the absence of diabetes. The 24-week lifestyle intervention was effective in ameliorating oxidative stress, but did not improve measures of CAN. Larger clinical trials with longer duration are required to confirm these findings.


Assuntos
Doenças do Sistema Nervoso Autônomo/etiologia , Doenças Cardiovasculares/etiologia , Síndrome Metabólica/complicações , Síndrome Metabólica/terapia , Estresse Oxidativo , Comportamento de Redução do Risco , Programas de Redução de Peso/métodos , Adulto , Sistema Nervoso Autônomo/fisiopatologia , Doenças do Sistema Nervoso Autônomo/epidemiologia , Doenças Cardiovasculares/epidemiologia , Sistema Cardiovascular/fisiopatologia , Estudos Transversais , Dieta Mediterrânea , Exercício Físico/fisiologia , Feminino , Humanos , Estilo de Vida , Masculino , Síndrome Metabólica/epidemiologia , Síndrome Metabólica/fisiopatologia , Pessoa de Meia-Idade , Estresse Oxidativo/fisiologia , Projetos Piloto , Fatores de Risco , Resultado do Tratamento
15.
Antioxid Redox Signal ; 21(4): 588-600, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23991914

RESUMO

AIMS: High circulating long chain fatty acids (LCFAs) are implicated in diabetic neuropathy (DN) development. Expression of the long-chain acyl-CoA synthetase 1 (Acsl1) gene, a gene required for LCFA metabolic activation, is altered in human and mouse diabetic peripheral nerve. We assessed the significance of Acsl1 upregulation in primary cultured Schwann cells. RESULTS: Acsl1 overexpression prevented oxidative stress (nitrotyrosine; hydroxyoctadecadienoic acids [HODEs]) and attenuated cellular injury (TUNEL) in Schwann cells following 12 h exposure to LCFAs (palmitate, linoleate, and oleate, 100 µM). Acsl1 overexpression potentiated the observed increase in medium to long-chain acyl-carnitines following 12 h LCFA exposure. Data are consistent with increased mitochondrial LCFA uptake, largely directed to incomplete beta-oxidation. LCFAs uncoupled mitochondrial oxygen consumption from ATP production. Acsl1 overexpression corrected mitochondrial dysfunction, increasing coupling efficiency and decreasing proton leak. INNOVATION: Schwann cell mitochondrial function is critical for peripheral nerve function, but research on Schwann cell mitochondrial dysfunction in response to hyperlipidemia is minimal. We demonstrate that high levels of a physiologically relevant mixture of LCFAs induce Schwann cell injury, but that improved mitochondrial uptake and metabolism attenuate this lipotoxicity. CONCLUSION: Acsl1 overexpression improves Schwann cell function and survival following high LCFA exposure in vitro; however, the observed endogenous Acsl1 upregulation in peripheral nerve in response to diabetes is not sufficient to prevent the development of DN in murine models of DN. Therefore, targeted improvement in Schwann cell metabolic disposal of LCFAs may improve DN phenotypes.


Assuntos
Coenzima A Ligases/genética , Ácidos Graxos/metabolismo , Expressão Gênica , Mitocôndrias/metabolismo , Estresse Oxidativo , Células de Schwann/metabolismo , Animais , Células Cultivadas , Neuropatias Diabéticas/metabolismo , Modelos Animais de Doenças , Ácidos Graxos/farmacologia , Humanos , Hipertrigliceridemia/genética , Hipertrigliceridemia/metabolismo , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/genética , Estresse Oxidativo/efeitos dos fármacos , Consumo de Oxigênio , Cultura Primária de Células , Ratos , Células de Schwann/efeitos dos fármacos
16.
Arthritis Rheumatol ; 66(9): 2532-2544, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24838349

RESUMO

OBJECTIVE: Oxidative stress and oxidized high-density lipoprotein (HDL) are implicated as risk factors for cardiovascular disease (CVD) in systemic lupus erythematosus (SLE). Yet, how HDL is oxidized and rendered dysfunctional in SLE remains unclear. Neutrophil extracellular traps (NETs), the levels of which are elevated in lupus, possess oxidant-generating enzymes, including myeloperoxidase (MPO), NADPH oxidase (NOX), and nitric oxide synthase (NOS). We hypothesized that NETs mediate HDL oxidation, impairing cholesterol efflux capacity (CEC). METHODS: Plasma MPO levels and CEC activity were examined in controls and lupus patients, and 3-chlorotyrosine (MPO specific) and 3-nitrotyrosine (derived from reactive nitrogen species) were quantified in human HDL. Multivariable linear models were used to estimate and test differences between groups. HDL was exposed to NETs from control and lupus neutrophils in the presence or absence of MPO, NOX, NOS inhibitors, and chloroquine (CQ). Murine HDL oxidation was quantified after NET inhibition in vivo. RESULTS: SLE patients displayed higher MPO levels and diminished CEC compared to controls. SLE HDL had higher 3-nitrotyrosine and 3-chlorotyrosine content than control HDL, with site-specific oxidation signatures on apolipoprotein A-I. Experiments with human and murine NETs confirmed that chlorination was mediated by MPO and NOX, and nitration by NOS and NOX. Mice with lupus treated with the NET inhibitor Cl-amidine displayed significantly decreased HDL oxidation. CQ inhibited NET formation in vitro. CONCLUSION: Active NOS, NOX, and MPO within NETs significantly modify HDL, rendering the lipoprotein proatherogenic. Since NET formation is enhanced in SLE, these findings support a novel role for NET-derived lipoprotein oxidation in SLE-associated CVD and identify additional proatherogenic roles of neutrophils and putative protective roles of antimalarials in autoimmunity.


Assuntos
Lipoproteínas HDL/metabolismo , Lúpus Eritematoso Sistêmico/enzimologia , Neutrófilos/enzimologia , Estresse Oxidativo/fisiologia , Adulto , Animais , Doenças Cardiovasculares/enzimologia , Feminino , Humanos , Lúpus Eritematoso Sistêmico/sangue , Masculino , Camundongos , Pessoa de Meia-Idade , NADPH Oxidases/sangue , Óxido Nítrico Sintase/sangue , Oxirredução , Peroxidase/sangue
17.
Cell Rep ; 7(2): 356-365, 2014 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-24726364

RESUMO

Inflammatory activation of myeloid cells is accompanied by increased glycolysis, which is required for the surge in cytokine production. Although in vitro studies suggest that increased macrophage glucose metabolism is sufficient for cytokine induction, the proinflammatory effects of increased myeloid cell glucose flux in vivo and the impact on atherosclerosis, a major complication of diabetes, are unknown. We therefore tested the hypothesis that increased glucose uptake in myeloid cells stimulates cytokine production and atherosclerosis. Overexpression of the glucose transporter GLUT1 in myeloid cells caused increased glycolysis and flux through the pentose phosphate pathway but did not induce cytokines. Moreover, myeloid-cell-specific overexpression of GLUT1 in LDL receptor-deficient mice was ineffective in promoting atherosclerosis. Thus, increased glucose flux is insufficient for inflammatory myeloid cell activation and atherogenesis. If glucose promotes atherosclerosis by increasing cellular glucose flux, myeloid cells do not appear to be the key targets.


Assuntos
Aterosclerose/metabolismo , Transportador de Glucose Tipo 1/metabolismo , Glucose/metabolismo , Células Mieloides/metabolismo , Animais , Transporte Biológico Ativo , Citocinas/genética , Citocinas/metabolismo , Transportador de Glucose Tipo 1/genética , Glicólise , Inflamação/metabolismo , Camundongos , Via de Pentose Fosfato , Receptores de LDL/genética , Receptores de LDL/metabolismo
18.
J Endocrinol ; 216(1): 1-11, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23086140

RESUMO

Diabetic neuropathy (DN) is the most common complication of diabetes and is characterized by distal-to-proximal loss of peripheral nerve axons. The idea of tissue-specific pathological alterations in energy metabolism in diabetic complications-prone tissues is emerging. Altered nerve metabolism in type 1 diabetes models is observed; however, therapeutic strategies based on these models offer limited efficacy to type 2 diabetic patients with DN. Therefore, understanding how peripheral nerves metabolically adapt to the unique type 2 diabetic environment is critical to develop disease-modifying treatments. In the current study, we utilized targeted liquid chromatography-tandem mass spectrometry (LC/MS/MS) to characterize the glycolytic and tricarboxylic acid (TCA) cycle metabolomes in sural nerve, sciatic nerve, and dorsal root ganglia (DRG) from male type 2 diabetic mice (BKS.Cg-m+/+Lepr(db); db/db) and controls (db/+). We report depletion of glycolytic intermediates in diabetic sural nerve and sciatic nerve (glucose-6-phosphate, fructose-6-phosphate, fructose-1,6-bisphosphate (sural nerve only), 3-phosphoglycerate, 2-phosphoglycerate, phosphoenolpyruvate, and lactate), with no significant changes in DRG. Citrate and isocitrate TCA cycle intermediates were decreased in sural nerve, sciatic nerve, and DRG from diabetic mice. Utilizing LC/electrospray ionization/MS/MS and HPLC methods, we also observed increased protein and lipid oxidation (nitrotyrosine; hydroxyoctadecadienoic acids) in db/db tissue, with a proximal-to-distal increase in oxidative stress, with associated decreased aconitase enzyme activity. We propose a preliminary model, whereby the greater change in metabolomic profile, increase in oxidative stress, and decrease in TCA cycle enzyme activity may cause distal peripheral nerves to rely on truncated TCA cycle metabolism in the type 2 diabetes environment.


Assuntos
Ciclo do Ácido Cítrico , Diabetes Mellitus Tipo 2/metabolismo , Neuropatias Diabéticas/metabolismo , Modelos Animais de Doenças , Glicólise , Estresse Oxidativo , Sistema Nervoso Periférico/metabolismo , Aconitato Hidratase/metabolismo , Animais , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/enzimologia , Neuropatias Diabéticas/enzimologia , Regulação para Baixo , Gânglios Espinais/enzimologia , Gânglios Espinais/metabolismo , Peroxidação de Lipídeos , Masculino , Camundongos , Camundongos Mutantes , Neurônios/enzimologia , Neurônios/metabolismo , Sistema Nervoso Periférico/enzimologia , Receptores para Leptina/genética , Receptores para Leptina/metabolismo , Nervo Isquiático/enzimologia , Nervo Isquiático/metabolismo , Nervo Sural/enzimologia , Nervo Sural/metabolismo , Tirosina/análogos & derivados , Tirosina/metabolismo
19.
Sci Transl Med ; 5(178): 178ra40, 2013 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-23536012

RESUMO

The early events leading to the development of rheumatoid arthritis (RA) remain unclear, but formation of autoantibodies to citrullinated protein antigens (ACPAs) is considered a key pathogenic event. Neutrophils isolated from patients with various autoimmune diseases display enhanced neutrophil extracellular trap (NET) formation, a phenomenon that exposes autoantigens in the context of immunostimulatory molecules. We investigated whether aberrant NETosis occurs in RA, determined its triggers, and examined its deleterious inflammatory consequences. Enhanced NETosis was observed in circulating and RA synovial fluid neutrophils compared to neutrophils from healthy controls and from patients with osteoarthritis (OA). Further, netting neutrophils infiltrated RA synovial tissue, rheumatoid nodules, and skin. NETosis correlated with ACPA presence and levels and with systemic inflammatory markers. RA sera and immunoglobulin fractions from RA patients with high levels of ACPA and/or rheumatoid factor significantly enhanced NETosis, and the NETs induced by these autoantibodies displayed distinct protein content. Indeed, during NETosis, neutrophils externalized the citrullinated autoantigens implicated in RA pathogenesis, and anti-citrullinated vimentin antibodies potently induced NET formation. Moreover, the inflammatory cytokines interleukin-17A (IL-17A) and tumor necrosis factor-α (TNF-α) induced NETosis in RA neutrophils. In turn, NETs significantly augmented inflammatory responses in RA and OA synovial fibroblasts, including induction of IL-6, IL-8, chemokines, and adhesion molecules. These observations implicate accelerated NETosis in RA pathogenesis, through externalization of citrullinated autoantigens and immunostimulatory molecules that may promote aberrant adaptive and innate immune responses in the joint and in the periphery, and perpetuate pathogenic mechanisms in this disease.


Assuntos
Artrite Reumatoide/imunologia , Autoanticorpos/imunologia , Autoantígenos/imunologia , Citrulina/metabolismo , Humanos , Interleucina-17/metabolismo , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Microscopia de Fluorescência , Neutrófilos/imunologia , Reação em Cadeia da Polimerase em Tempo Real , Fator de Necrose Tumoral alfa/metabolismo
20.
Methods Enzymol ; 491: 73-89, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21329795

RESUMO

Oxidative stress plays a central role in the pathogenesis of diverse chronic inflammatory disorders including diabetic complications, cardiovascular disease, aging, neurodegenerative disease, autoimmune disorders, and pulmonary fibrosis. Protein misfolding can lead to chronic endoplasmic reticulum (ER) stress which can exacerbate oxidative stress. This can trigger apoptotic cascades resulting in chronic inflammatory disorders. Despite intense interest in origins and magnitude of oxidative stress, ability to quantify oxidants has been limited because they are short lived. We have developed quantitative mass spectrometry (MS)-based analytical strategies to analyze stable end products of protein oxidation. These molecules provide quantitative and mechanistic assessment of degree of oxidative stress in cell cultures, tissues, and biofluids of animal models of disease and human samples. Our studies support the hypothesis that unique reactive intermediates generated in localized microenvironments of vulnerable tissues promote end-organ damage. The ability to quantify these changes and assess response to therapies will be pivotal in understanding disease mechanisms and monitoring efficacy of therapy.


Assuntos
Aminoácidos/metabolismo , Retículo Endoplasmático/metabolismo , Proteínas/metabolismo , Espectrometria de Massas em Tandem/métodos , Aminoácidos/isolamento & purificação , Animais , Cromatografia Líquida/métodos , Cromatografia Gasosa-Espectrometria de Massas/métodos , Humanos , Oxirredução , Estresse Oxidativo , Proteínas/isolamento & purificação , Espectrometria de Massas por Ionização por Electrospray/métodos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa