Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Gen Physiol ; 156(11)2024 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-39302315

RESUMO

Sarcomere activation in striated muscle requires both thin filament-based and thick filament-based activation mechanisms. Recent studies have shown that myosin heads on the thick filaments undergo OFF to ON structural transitions in response to calcium (Ca2+) in permeabilized porcine myocardium in the presence of a small molecule inhibitor that eliminated active force. The changes in X-ray diffraction signatures of OFF to ON transitions were interpreted as Ca2+ acting to activate the thick filaments. Alternatively, Ca2+ binding to troponin could initiate a Ca2+-dependent crosstalk from the thin filament to the thick filament via interfilament connections such as the myosin binding protein-C. Here, we exchanged native troponin in permeabilized porcine myocardium for troponin containing the cTnC D65A mutation, which disallows the activation of troponin through Ca2+ binding to determine if Ca2+-dependent thick filament activation persists in the absence of thin filament activation. After the exchange protocol, over 95% of the Ca2+-activated force was eliminated. Equatorial intensity ratio increased significantly in both WT and D65A exchanged myocardium with increasing Ca2+ concentration. The degree of helical ordering of the myosin heads decreased by the same amount in WT and D65A myocardium when Ca2+ concentration increased. These results are consistent with a direct effect of Ca2+ in activating the thick filament rather than an indirect effect due to Ca2+-mediated crosstalk between the thick and thin filaments.


Assuntos
Cálcio , Miocárdio , Animais , Cálcio/metabolismo , Suínos , Miocárdio/metabolismo , Sarcômeros/metabolismo , Miosinas/metabolismo , Troponina/metabolismo , Contração Miocárdica/efeitos dos fármacos , Contração Miocárdica/fisiologia
2.
Nature ; 632(8023): 192-200, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39020181

RESUMO

Gene replacement using adeno-associated virus (AAV) vectors is a promising therapeutic approach for many diseases1,2. However, this therapeutic modality is challenged by the packaging capacity of AAVs (approximately 4.7 kilobases)3, limiting its application for disorders involving large coding sequences, such as Duchenne muscular dystrophy, with a 14 kilobase messenger RNA. Here we developed a new method for expressing large dystrophins by utilizing the protein trans-splicing mechanism mediated by split inteins. We identified several split intein pairs that efficiently join two or three fragments to generate a large midi-dystrophin or the full-length protein. We show that delivery of two or three AAVs into dystrophic mice results in robust expression of large dystrophins and significant physiological improvements compared with micro-dystrophins. Moreover, using the potent myotropic AAVMYO4, we demonstrate that low total doses (2 × 1013 viral genomes per kg) are sufficient to express large dystrophins in striated muscles body-wide with significant physiological corrections in dystrophic mice. Our data show a clear functional superiority of large dystrophins over micro-dystrophins that are being tested in clinical trials. This method could benefit many patients with Duchenne or Becker muscular dystrophy, regardless of genotype, and could be adapted to numerous other disorders caused by mutations in large genes that exceed the AAV capacity.


Assuntos
Distrofina , Terapia Genética , Inteínas , Distrofia Muscular de Duchenne , Processamento de Proteína , Animais , Humanos , Masculino , Camundongos , Dependovirus/genética , Dependovirus/metabolismo , Modelos Animais de Doenças , Distrofina/biossíntese , Distrofina/deficiência , Distrofina/genética , Distrofina/metabolismo , Terapia Genética/métodos , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Inteínas/genética , Camundongos Endogâmicos mdx , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Distrofia Muscular de Duchenne/metabolismo , Processamento de Proteína/genética
3.
J Mol Cell Cardiol ; 191: 27-39, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38648963

RESUMO

Approximately 40% of hypertrophic cardiomyopathy (HCM) mutations are linked to the sarcomere protein cardiac myosin binding protein-C (cMyBP-C). These mutations are either classified as missense mutations or truncation mutations. One mutation whose nature has been inconsistently reported in the literature is the MYBPC3-c.772G > A mutation. Using patient-derived human induced pluripotent stem cells differentiated to cardiomyocytes (hiPSC-CMs), we have performed a mechanistic study of the structure-function relationship for this MYBPC3-c.772G > A mutation versus a mutation corrected, isogenic cell line. Our results confirm that this mutation leads to exon skipping and mRNA truncation that ultimately suggests ∼20% less cMyBP-C protein (i.e., haploinsufficiency). This, in turn, results in increased myosin recruitment and accelerated myofibril cycling kinetics. Our mechanistic studies suggest that faster ADP release from myosin is a primary cause of accelerated myofibril cross-bridge cycling due to this mutation. Additionally, the reduction in force generating heads expected from faster ADP release during isometric contractions is outweighed by a cMyBP-C phosphorylation mediated increase in myosin recruitment that leads to a net increase of myofibril force, primarily at submaximal calcium activations. These results match well with our previous report on contractile properties from myectomy samples of the patients from whom the hiPSC-CMs were generated, demonstrating that these cell lines are a good model to study this pathological mutation and extends our understanding of the mechanisms of altered contractile properties of this HCM MYBPC3-c.772G > A mutation.


Assuntos
Cardiomiopatia Hipertrófica , Proteínas de Transporte , Haploinsuficiência , Células-Tronco Pluripotentes Induzidas , Mutação , Miócitos Cardíacos , Humanos , Cardiomiopatia Hipertrófica/genética , Cardiomiopatia Hipertrófica/metabolismo , Miócitos Cardíacos/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Miosinas/metabolismo , Miosinas/genética , Diferenciação Celular/genética , Cinética
4.
Circ Res ; 133(5): 430-443, 2023 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-37470183

RESUMO

BACKGROUND: Modulating myosin function is a novel therapeutic approach in patients with cardiomyopathy. Danicamtiv is a novel myosin activator with promising preclinical data that is currently in clinical trials. While it is known that danicamtiv increases force and cardiomyocyte contractility without affecting calcium levels, detailed mechanistic studies regarding its mode of action are lacking. METHODS: Permeabilized porcine cardiac tissue and myofibrils were used for X-ray diffraction and mechanical measurements. A mouse model of genetic dilated cardiomyopathy was used to evaluate the ability of danicamtiv to correct the contractile deficit. RESULTS: Danicamtiv increased force and calcium sensitivity via increasing the number of myosins in the ON state and slowing cross-bridge turnover. Our detailed analysis showed that inhibition of ADP release results in decreased cross-bridge turnover with cross bridges staying attached longer and prolonging myofibril relaxation. Danicamtiv corrected decreased calcium sensitivity in demembranated tissue, abnormal twitch magnitude and kinetics in intact cardiac tissue, and reduced ejection fraction in the whole organ. CONCLUSIONS: As demonstrated by the detailed studies of Danicamtiv, increasing myosin recruitment and altering cross-bridge cycling are 2 mechanisms to increase force and calcium sensitivity in cardiac muscle. Myosin activators such as Danicamtiv can treat the causative hypocontractile phenotype in genetic dilated cardiomyopathy.


Assuntos
Cardiomiopatia Dilatada , Camundongos , Animais , Suínos , Cardiomiopatia Dilatada/tratamento farmacológico , Cálcio/fisiologia , Miocárdio , Miosinas , Miócitos Cardíacos , Cardiotônicos
5.
Proc Natl Acad Sci U S A ; 120(5): e2207615120, 2023 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-36696446

RESUMO

Contraction in striated muscle is initiated by calcium binding to troponin complexes, but it is now understood that dynamic transition of myosin between resting, ordered OFF states on thick filaments and active, disordered ON states that can bind to thin filaments is critical in regulating muscle contractility. These structural OFF to ON transitions of myosin are widely assumed to correspond to transitions from the biochemically defined, energy-sparing, super-relaxed (SRX) state to the higher ATPase disordered-relaxed (DRX) state. Here we examined the effect of 2'-deoxy-ATP (dATP), a naturally occurring energy substrate for myosin, on the structural OFF to ON transitions of myosin motors in porcine cardiac muscle thick filaments. Small-angle X-ray diffraction revealed that titrating dATP in relaxation solutions progressively moves the myosin heads from ordered OFF states on the thick filament backbone to disordered ON states closer to thin filaments. Importantly, we found that the structural OFF to ON transitions are not equivalent to the biochemically defined SRX to DRX transitions and that the dATP-induced structural OFF to ON transitions of myosin motors in relaxed muscle are strongly correlated with submaximal force augmentation by dATP. These results indicate that structural OFF to ON transitions of myosin in relaxed muscle can predict the level of force attained in calcium-activated cardiac muscle. Computational modeling and stiffness measurements suggest a final step in the OFF to ON transition may involve a subset of DRX myosins that form weakly bound cross-bridges prior to becoming active force-producing cross-bridges.


Assuntos
Cálcio , Músculo Estriado , Animais , Suínos , Cálcio/metabolismo , Miocárdio/metabolismo , Miosinas/metabolismo , Músculo Esquelético/metabolismo , Músculo Estriado/metabolismo , Cálcio da Dieta
6.
J Mol Cell Cardiol ; 175: 1-12, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36470336

RESUMO

Hallmark features of systolic heart failure are reduced contractility and impaired metabolic flexibility of the myocardium. Cardiomyocytes (CMs) with elevated deoxy ATP (dATP) via overexpression of ribonucleotide reductase (RNR) enzyme robustly improve contractility. However, the effect of dATP elevation on cardiac metabolism is unknown. Here, we developed proteolysis-resistant versions of RNR and demonstrate that elevation of dATP/ATP to ∼1% in CMs in a transgenic mouse (TgRRB) resulted in robust improvement of cardiac function. Pharmacological approaches showed that CMs with elevated dATP have greater basal respiratory rates by shifting myosin states to more active forms, independent of its isoform, in relaxed CMs. Targeted metabolomic profiling revealed a significant reprogramming towards oxidative phosphorylation in TgRRB-CMs. Higher cristae density and activity in the mitochondria of TgRRB-CMs improved respiratory capacity. Our results revealed a critical property of dATP to modulate myosin states to enhance contractility and induce metabolic flexibility to support improved function in CMs.


Assuntos
Miocárdio , Ribonucleotídeo Redutases , Camundongos , Animais , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Contração Miocárdica , Ribonucleotídeo Redutases/metabolismo , Ribonucleotídeo Redutases/farmacologia , Camundongos Transgênicos , Trifosfato de Adenosina/metabolismo , Miosinas/metabolismo
7.
J Clin Invest ; 132(10)2022 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-35575090

RESUMO

In hypertrophied and failing hearts, fuel metabolism is reprogrammed to increase glucose metabolism, especially glycolysis. This metabolic shift favors biosynthetic function at the expense of ATP production. Mechanisms responsible for the switch are poorly understood. We found that inhibitory factor 1 of the mitochondrial FoF1-ATP synthase (ATPIF1), a protein known to inhibit ATP hydrolysis by the reverse function of ATP synthase during ischemia, was significantly upregulated in pathological cardiac hypertrophy induced by pressure overload, myocardial infarction, or α-adrenergic stimulation. Chemical cross-linking mass spectrometry analysis of hearts hypertrophied by pressure overload suggested that increased expression of ATPIF1 promoted the formation of FoF1-ATP synthase nonproductive tetramer. Using ATPIF1 gain- and loss-of-function cell models, we demonstrated that stalled electron flow due to impaired ATP synthase activity triggered mitochondrial ROS generation, which stabilized HIF1α, leading to transcriptional activation of glycolysis. Cardiac-specific deletion of ATPIF1 in mice prevented the metabolic switch and protected against the pathological remodeling during chronic stress. These results uncover a function of ATPIF1 in nonischemic hearts, which gives FoF1-ATP synthase a critical role in metabolic rewiring during the pathological remodeling of the heart.


Assuntos
Glicólise , ATPases Mitocondriais Próton-Translocadoras , Proteínas/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Camundongos , Miocárdio/metabolismo , Ativação Transcricional , Regulação para Cima , Proteína Inibidora de ATPase
8.
J Mol Cell Cardiol ; 158: 1-10, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33989657

RESUMO

BACKGROUND: Reduced fatty acid oxidation (FAO) is a hallmark of metabolic remodeling in heart failure. Enhancing mitochondrial long-chain fatty acid uptake by Acetyl-CoA carboxylase 2 (ACC2) deletion increases FAO and prevents cardiac dysfunction during chronic stresses, but therapeutic efficacy of this approach has not been determined. METHODS: Male and female ACC2 f/f-MCM (ACC2KO) and their respective littermate controls were subjected to chronic pressure overload by TAC surgery. Tamoxifen injection 3 weeks after TAC induced ACC2 deletion and increased FAO in ACC2KO mice with pathological hypertrophy. RESULTS: ACC2 deletion in mice with pre-existing cardiac pathology promoted FAO in female and male hearts, but improved cardiac function only in female mice. In males, pressure overload caused a downregulation in the mitochondrial oxidative function. Stimulating FAO by ACC2 deletion caused unproductive acyl-carnitine accumulation, which failed to improve cardiac energetics. In contrast, mitochondrial oxidative capacity was sustained in female pressure overloaded hearts and ACC2 deletion improved myocardial energetics. Mechanistically, we revealed a sex-dependent regulation of PPARα signaling pathway in heart failure, which accounted for the differential response to ACC2 deletion. CONCLUSION: Metabolic remodeling in the failing heart is sex-dependent which could determine the response to metabolic intervention. The findings suggest that both mitochondrial oxidative capacity and substrate preference should be considered for metabolic therapy of heart failure.


Assuntos
Acetil-CoA Carboxilase/metabolismo , Ácidos Graxos/metabolismo , Insuficiência Cardíaca/metabolismo , PPAR alfa/metabolismo , Transdução de Sinais/genética , Acetil-CoA Carboxilase/genética , Animais , Carnitina/análogos & derivados , Carnitina/metabolismo , Modelos Animais de Doenças , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/genética , Feminino , Deleção de Genes , Insuficiência Cardíaca/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Miocárdio/metabolismo , Oxirredução , Fatores Sexuais , Transdução de Sinais/efeitos dos fármacos , Tamoxifeno/administração & dosagem
9.
PLoS One ; 15(12): e0242749, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33264332

RESUMO

Cystic fibrosis (CF) is due to mutations in the CF-transmembrane conductance regulator (CFTR) and CF-related diabetes (CFRD) is its most common co-morbidity, affecting ~50% of all CF patients, significantly influencing pulmonary function and longevity. Yet, the complex pathogenesis of CFRD remains unclear. Two non-mutually exclusive underlying mechanisms have been proposed in CFRD: i) damage of the endocrine cells secondary to the severe exocrine pancreatic pathology and ii) intrinsic ß-cell impairment of the secretory response in combination with other factors. The later has proven difficult to determine due to low expression of CFTR in ß-cells, which results in the general perception that this Cl-channel does not participate in the modulation of insulin secretion or the development of CFRD. The objective of the present work is to demonstrate CFTR expression at the molecular and functional levels in insulin-secreting ß-cells in normal human islets, where it seems to play a role. Towards this end, we have used immunofluorescence confocal and immunofluorescence microscopy, immunohistochemistry, RT-qPCR, Western blotting, pharmacology, electrophysiology and insulin secretory studies in normal human, rat and mouse islets. Our results demonstrate heterogeneous CFTR expression in human, mouse and rat ß-cells and provide evidence that pharmacological inhibition of CFTR influences basal and stimulated insulin secretion in normal mouse islets but not in islets lacking this channel, despite being detected by electrophysiological means in ~30% of ß-cells. Therefore, our results demonstrate a potential role for CFTR in the pancreatic ß-cell secretory response suggesting that intrinsic ß-cell dysfunction may also participate in the pathogenesis of CFRD.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Células Secretoras de Insulina/metabolismo , Adulto , Idoso , Animais , Anticorpos/metabolismo , Antígenos/metabolismo , Linhagem Celular , Regulador de Condutância Transmembrana em Fibrose Cística/imunologia , Feminino , Humanos , Lactente , Secreção de Insulina , Masculino , Camundongos , Pessoa de Meia-Idade , Ratos , Reprodutibilidade dos Testes , Adulto Jovem
10.
J Biol Chem ; 294(10): 3707-3719, 2019 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-30587573

RESUMO

Neuroendocrine-type ATP-sensitive K+ (KATP) channels are metabolite sensors coupling membrane potential with metabolism, thereby linking insulin secretion to plasma glucose levels. They are octameric complexes, (SUR1/Kir6.2)4, comprising sulfonylurea receptor 1 (SUR1 or ABCC8) and a K+-selective inward rectifier (Kir6.2 or KCNJ11). Interactions between nucleotide-, agonist-, and antagonist-binding sites affect channel activity allosterically. Although it is hypothesized that opening these channels requires SUR1-mediated MgATP hydrolysis, we show here that ATP binding to SUR1, without hydrolysis, opens channels when nucleotide antagonism on Kir6.2 is minimized and SUR1 mutants with increased ATP affinities are used. We found that ATP binding is sufficient to switch SUR1 alone between inward- or outward-facing conformations with low or high dissociation constant, KD , values for the conformation-sensitive channel antagonist [3H]glibenclamide ([3H]GBM), indicating that ATP can act as a pure agonist. Assembly with Kir6.2 reduced SUR1's KD for [3H]GBM. This reduction required the Kir N terminus (KNtp), consistent with KNtp occupying a "transport cavity," thus positioning it to link ATP-induced SUR1 conformational changes to channel gating. Moreover, ATP/GBM site coupling was constrained in WT SUR1/WT Kir6.2 channels; ATP-bound channels had a lower KD for [3H]GBM than ATP-bound SUR1. This constraint was largely eliminated by the Q1179R neonatal diabetes-associated mutation in helix 15, suggesting that a "swapped" helix pair, 15 and 16, is part of a structural pathway connecting the ATP/GBM sites. Our results suggest that ATP binding to SUR1 biases KATP channels toward open states, consistent with SUR1 variants with lower KD values causing neonatal diabetes, whereas increased KD values cause congenital hyperinsulinism.


Assuntos
Trifosfato de Adenosina/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Receptores de Sulfonilureias/química , Receptores de Sulfonilureias/metabolismo , Difosfato de Adenosina/metabolismo , Regulação Alostérica , Animais , Sítios de Ligação , Cricetinae , Guanosina Trifosfato/metabolismo , Células HEK293 , Humanos , Hidrólise , Ativação do Canal Iônico , Modelos Moleculares , Mutação , Canais de Potássio Corretores do Fluxo de Internalização/química , Ligação Proteica , Conformação Proteica em alfa-Hélice
11.
J Lipid Res ; 56(12): 2337-47, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26489644

RESUMO

In addition to triacylglycerols, adipocytes contain a large reserve of unesterified cholesterol. During adipocyte lipolysis and cell death seen during severe obesity and weight loss, free fatty acids and cholesterol become available for uptake and processing by adipose tissue macrophages (ATMs). We hypothesize that ATMs become cholesterol enriched and participate in cholesterol clearance from adipose tissue. We previously showed that ABCG1 is robustly upregulated in ATMs taken from obese mice and further enhanced by caloric restriction. Here, we found that ATMs taken from obese and calorie-restricted mice derived from transplantation of WT or Abcg1-deficient bone marrow are cholesterol enriched. ABCG1 levels regulate the ratio of classically activated (M1) to alternatively activated (M2) ATMs and their cellular cholesterol content. Using WT and Abcg1(-/-) cultured macrophages, we found that Abcg1 is most highly expressed by M2 macrophages and that ABCG1 deficiency is sufficient to retard macrophage chemotaxis. However, changes in myeloid expression of Abcg1 did not protect mice from obesity or impaired glucose homeostasis. Overall, ABCG1 modulates ATM cholesterol content in obesity and weight loss regimes leading to an alteration in M1 to M2 ratio that we suggest is due to the extent of macrophage egress from adipose tissue.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Tecido Adiposo/citologia , Tecido Adiposo/metabolismo , Restrição Calórica , Colesterol/metabolismo , Lipoproteínas/metabolismo , Macrófagos/metabolismo , Obesidade/metabolismo , Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Lipoproteínas/genética , Macrófagos/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/genética
12.
Physiol Rep ; 2(11)2014 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-25428948

RESUMO

Diabetic nephropathy (DN) is a major complication of diabetes and the leading cause of end-stage renal disease. DN is characterized by changes in kidney structure and function but the underlying genetic and molecular factors are poorly understood. We used a mouse diversity panel to explore the genetic basis of DN traits in mice carrying the Ins2 Akita mutation. Twenty-eight Akita strains were generated by breeding this panel to DBA/2.Akita mice. Male F1 diabetic and nondiabetic littermates were evaluated for DN-related traits. Urine albumin-to-creatinine ratios (ACRs), volume and cystatin C as well as blood urea nitrogen and lipoprotein levels varied significantly among the diabetic strains. For most Akita strains, ACR values increased 2- to 6-fold over euglycemic control values. However, six strains exhibited changes in ACR exceeding 10-fold with two strains (NOD/ShiLt and CBA) showing 50- to 83- fold increases. These increases are larger than previously reported among available DN mouse models establishing these strains as useful for additional studies of renal function. ACRs correlated with cystatin C (P = 0.0286), a measure of hyperfiltration and an interstitial tubular marker associated with DN onset in humans suggesting that tubule damage as well as podocyte-stress contributed to reduced kidney function assessed by ACR. Although large changes were seen for ACRs, severe nephropathology was absent. However, glomerular hypertrophy and collagen IV content were found to vary significantly among strains suggesting a genetic basis for early onset features of DN. Our results define the range of DN phenotypes that occur among common inbred strains of mice.

13.
Am J Physiol Endocrinol Metab ; 302(8): E961-71, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-22318945

RESUMO

Lymphotoxin-α (LTα) is secreted by lymphocytes and acts through tumor necrosis factor-α receptors and the LTß receptor. Our goals were to determine whether LT has a role in obesity and investigate whether LT contributes to the link between obesity and adipose tissue lymphocyte accumulation. LT deficient (LT(-/-)) and wild-type (WT) mice were fed standard pelleted rodent chow or a high-fat/high-sucrose diet (HFHS) for 13 wk. Body weight, body composition, and food intake were measured. Glucose tolerance was assessed. Systemic and adipose tissue inflammatory statuses were evaluated by quantifying plasma adipokine levels and tissue macrophage and T cell-specific gene expression in abdominal fat. LT(-/-) mice were smaller (20%) and leaner (25%) than WT controls after 13 wk of HFHS diet feeding. LT(-/-) mice showed improved glucose tolerance, suggesting that, in WT mice, LT may impair glucose metabolism. Surprisingly, adipose tissue from rodent chow- and HFHS-fed LT(-/-) mice exhibited increased T lymphocyte and macrophage infiltration compared with WT mice. Despite the fact that LT(-/-) mice exhibited an enhanced inflammatory status at the systemic and tissue level even when fed rodent chow, they were protected from enhanced diet-induced obesity and insulin resistance. Thus, LT contributes to body weight and adiposity and is required to modulate the accumulation of immune cells in adipose tissue.


Assuntos
Tecido Adiposo Branco/imunologia , Linfotoxina-alfa/metabolismo , Macrófagos/imunologia , Obesidade/imunologia , Linfócitos T/imunologia , Adipocinas/sangue , Adiposidade , Animais , Linhagem Celular , Citocinas/genética , Citocinas/metabolismo , Dieta Hiperlipídica/efeitos adversos , Sacarose Alimentar/efeitos adversos , Expressão Gênica , Regulação da Expressão Gênica , Intolerância à Glucose/sangue , Intolerância à Glucose/etiologia , Intolerância à Glucose/imunologia , Intolerância à Glucose/metabolismo , Resistência à Insulina , Linfotoxina-alfa/genética , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Knockout , Obesidade/sangue , Obesidade/etiologia , Obesidade/metabolismo , RNA Mensageiro/metabolismo , Distribuição Aleatória , Linfócitos T/metabolismo , Aumento de Peso
14.
Biochim Biophys Acta ; 1821(3): 425-34, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22179025

RESUMO

The prevalence of obesity has reached epidemic proportions and is associated with several co-morbid conditions including diabetes, dyslipidemia, cancer, atherosclerosis and gallstones. Obesity is associated with low systemic inflammation and an accumulation of adipose tissue macrophages (ATMs) that are thought to modulate insulin resistance. ATMs may also modulate adipocyte metabolism and take up lipids released during adipocyte lipolysis and cell death. We suggest that high levels of free cholesterol residing in adipocytes are released during these processes and contribute to ATM activation and accumulation during obesity and caloric restriction. Db/db mice were studied for extent of adipose tissue inflammation under feeding conditions of ad libitum (AL) and caloric restriction (CR). The major finding was a marked elevation in epididymal adipose ABCG1 mRNA levels with obesity and CR (6-fold and 16-fold, respectively) over that seen for lean wild-type mice. ABCG1 protein was also elevated for CR as compared to AL adipose tissue. ABCG1 is likely produced by cholesterol loaded ATMs since this gene is not highly expressed in adipocytes and ABCG1 expression is sterol mediated. Our data supports the concept that metabolic changes in adipocytes due to demand lipolysis and cell death lead to cholesterol loading of ATMs. Based on finding cholesterol-loaded peritoneal leukocytes with elevated levels of ABCG1 in CR as compared to AL mice, we suggest that pathways for cholesterol trafficking out of adipose tissue involve ATM egress as well as ABCG1 mediated cholesterol efflux. This article is part of a Special Issue entitled Advances in High Density Lipoprotein Formation and Metabolism: A Tribute to John F. Oram (1945-2010).


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Gordura Abdominal/metabolismo , Expressão Gênica , Lipoproteínas/metabolismo , Obesidade/metabolismo , Redução de Peso , Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Gordura Abdominal/patologia , Adipócitos Brancos/metabolismo , Adipócitos Brancos/patologia , Animais , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Restrição Calórica , Movimento Celular , Colesterol/metabolismo , Feminino , Lipólise , Lipoproteínas/genética , Macrófagos/enzimologia , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Óxido Nítrico Sintase Tipo II/metabolismo , Obesidade/dietoterapia , Obesidade/fisiopatologia , Triglicerídeos/metabolismo
15.
J Autoimmun ; 34(2): 96-104, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19664906

RESUMO

We developed a panel of non-obese diabetic (NOD) mice deficient in major lysosomal cysteine proteases (cathepsins S, L and B) to identify protease enzymes essential for autoimmune diabetes. Null alleles for cathepsins (Cts) S, L or B were introgressed onto the NOD genetic background with 19 Idd markers at homozygosity. Diabetes onset was determined among females aged up to 6 months. We evaluated insulitis and sialadenitis in tissues using histology and computer assisted morphology. NOD mice deficient in Ctss or Ctsb were partially protected from diabetes with incidence at 33% and 28%, respectively, versus wild-type NOD (69%; p < 0.00001). NODs lacking cathepsin L (Ctsl-/-) are completely protected from IDDM, as originally shown by others. Ctsl, Ctss, or Ctsb heterozygous mice were able to develop IDDM, although incidence levels were significantly lower for Ctsb+/- (50%) and Ctsl+/- (55%) as compared to NODs (69%; p < 0.03). Ctsl-/- mice contain functional, diabetogenic T cells and an enriched Foxp3+ regulatory T cell population, and diabetes resistance was due to the presence of an expanded population of regulatory T cells. These data provide additional information about the potency of the diabetogenic T cell population in Ctsl-/- mice which were comparable in potency to wild-type NOD mice. These data illustrate the critical contribution of each of these proteases in determining IDDM in the NOD mouse and provide a useful set of models for further studies.


Assuntos
Catepsina B/metabolismo , Catepsina L/metabolismo , Catepsinas/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Idade de Início , Animais , Antígenos CD4/biossíntese , Catepsina B/genética , Catepsina B/imunologia , Catepsina L/genética , Catepsina L/imunologia , Catepsinas/genética , Catepsinas/imunologia , Movimento Celular/genética , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/patologia , Diabetes Mellitus Tipo 1/fisiopatologia , Feminino , Fatores de Transcrição Forkhead/biossíntese , Linfopenia , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Knockout , Pancreatite , Sialadenite , Subpopulações de Linfócitos T/patologia , Linfócitos T Reguladores/patologia
16.
Endocrinology ; 150(9): 4124-34, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19477937

RESUMO

TNF-alpha signals through two receptors, TNFR1 and TNFR2. Our goals were: 1) determine the role of TNFRs in obesity and metabolic disease and 2) investigate whether TNFRs contribute to the link between obesity and adipose tissue macrophage infiltration and polarization. R1(-/-)R2(-/-) (RKO) and wild-type (WT) mice were fed standard chow or a high-fat/high-sucrose diet (HFHS) over 14 wk. Body composition, food intake, and energy expenditure were measured. Oral glucose tolerance and insulin sensitivity tests assessed glucose homeostasis. Adipose tissue and systemic inflammatory status were evaluated by quantifying plasma adipokine levels and macrophage-specific gene expression in fat. RKO mice were heavier (10%) and fatter (18%) than WT controls at 4 wk of age and were 26% heavier and 50% fatter than WT after 14 wk of HFHS diet feeding. Age- and diet-adjusted 24-h oxygen consumption, activity, and respiratory exchange ratio were significantly reduced in RKO mice. Obese RKO mice were markedly insulin resistant, suggesting that intact TNFR signaling is not required for the effect of obesity to impair glucose metabolism. Adipose tissue from HFHS-fed RKO mice exhibited increased macrophage infiltration, but compared with WT mice, macrophage phenotypic markers featured a predominance of antiinflammatory M2 over proinflammatory M1 cells. TNFRs play a physiological role to limit body weight and adiposity by modestly increasing metabolic rate and fatty acid oxidation, and they are required for obesity-induced activation of adipose tissue macrophages. Despite these effects, TNFRs are not required for obesity-induced insulin resistance.


Assuntos
Tecido Adiposo/citologia , Macrófagos/fisiologia , Obesidade/prevenção & controle , Receptores do Fator de Necrose Tumoral/fisiologia , Adipocinas/sangue , Tecido Adiposo/patologia , Animais , Resistência à Insulina , Leptina/sangue , Macrófagos/metabolismo , Masculino , Síndrome Metabólica/fisiopatologia , Camundongos , Camundongos Knockout , Obesidade/patologia , Fator de Necrose Tumoral alfa/metabolismo
17.
Arterioscler Thromb Vasc Biol ; 28(4): 685-91, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18239153

RESUMO

OBJECTIVE: Chronic systemic inflammation accompanies obesity and predicts development of cardiovascular disease. Dietary cholesterol has been shown to increase inflammation and atherosclerosis in LDL receptor-deficient (LDLR(-/-)) mice. This study was undertaken to determine whether dietary cholesterol and obesity have additive effects on inflammation and atherosclerosis. METHODS AND RESULTS: LDLR(-/-) mice were fed chow, high-fat, high-carbohydrate (diabetogenic) diets without (DD) or with added cholesterol (DDC) for 24 weeks. Effects on adipose tissue, inflammatory markers, and atherosclerosis were studied. Despite similar weight gain between DD and DDC groups, addition of dietary cholesterol increased insulin resistance relative to DD. Adipocyte hypertrophy, macrophage accumulation, and local inflammation were observed in intraabdominal adipose tissue in DD and DDC, but were significantly higher in the DDC group. Circulating levels of the inflammatory protein serum amyloid A (SAA) were 4.4-fold higher in DD animals and 15-fold higher in DDC animals than controls, suggesting chronic systemic inflammation. Hepatic SAA mRNA levels were similarly elevated. Atherosclerosis was increased in the DD-fed animals and further increased in the DDC group. CONCLUSIONS: Obesity-induced macrophage accumulation in adipose tissue is exacerbated by dietary cholesterol. These local inflammatory changes in adipose tissue are associated with insulin resistance, systemic inflammation, and increased atherosclerosis in this mouse model.


Assuntos
Aterosclerose/etiologia , Colesterol na Dieta/efeitos adversos , Macrófagos/patologia , Obesidade/complicações , Receptores de LDL/deficiência , Gordura Abdominal/metabolismo , Gordura Abdominal/patologia , Adipócitos/patologia , Animais , Aterosclerose/metabolismo , Sequência de Bases , Colesterol na Dieta/administração & dosagem , Primers do DNA/genética , Mediadores da Inflamação/metabolismo , Resistência à Insulina , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/metabolismo , Obesidade/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de LDL/genética , Proteína Amiloide A Sérica/genética , Proteína Amiloide A Sérica/metabolismo
18.
Circulation ; 117(3): 421-8, 2008 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-18158360

RESUMO

BACKGROUND: Vascular inflammation and lipid deposition are prominent features of atherosclerotic lesion formation. We have shown previously that the dithiol compound alpha-lipoic acid (LA) exerts antiinflammatory effects by inhibiting tumor necrosis factor-alpha- and lipopolysaccharide-induced endothelial and monocyte activation in vitro and lipopolysaccharide-induced acute inflammatory responses in vivo. Here, we investigated whether LA inhibits atherosclerosis in apolipoprotein E-deficient (apoE-/-) and apoE/low-density lipoprotein receptor-deficient mice, 2 well-established animal models of human atherosclerosis. METHODS AND RESULTS: Four-week-old female apoE-/- mice (n=20 per group) or apoE/low-density lipoprotein receptor-deficient mice (n=21 per group) were fed for 10 weeks a Western-type chow diet containing 15% fat and 0.125% cholesterol without or with 0.2% (wt/wt) R,S-LA or a normal chow diet containing 4% fat without or with 0.2% (wt/wt) R-LA, respectively. Supplementation with LA significantly reduced atherosclerotic lesion formation in the aortic sinus of both mouse models by approximately 20% and in the aortic arch and thoracic aorta of apoE-/- and apoE/low-density lipoprotein receptor-deficient mice by approximately 55% and 40%, respectively. This strong antiatherogenic effect of LA was associated with almost 40% less body weight gain and lower serum and very low-density lipoprotein levels of triglycerides but not cholesterol. In addition, LA supplementation reduced aortic expression of adhesion molecules and proinflammatory cytokines and aortic macrophage accumulation. These antiinflammatory effects of LA were more pronounced in the aortic arch and the thoracic aorta than in the aortic sinus, reflecting the corresponding reductions in atherosclerosis. CONCLUSIONS: Our study shows that dietary LA supplementation inhibits atherosclerotic lesion formation in 2 mouse models of human atherosclerosis, an inhibition that appears to be due to the "antiobesity," antihypertriglyceridemic, and antiinflammatory effects of LA. LA may be a useful adjunct in the prevention and treatment of atherosclerotic vascular diseases.


Assuntos
Apolipoproteínas E/deficiência , Aterosclerose/tratamento farmacológico , Receptores de LDL/deficiência , Ácido Tióctico/farmacologia , Animais , Aterosclerose/prevenção & controle , Peso Corporal/efeitos dos fármacos , Colesterol/sangue , Suplementos Nutricionais , Modelos Animais de Doenças , Feminino , Inflamação/tratamento farmacológico , Inflamação/prevenção & controle , Camundongos , Ácido Tióctico/administração & dosagem , Triglicerídeos/sangue
19.
Circulation ; 111(21): 2798-804, 2005 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-15911707

RESUMO

BACKGROUND: Myeloperoxidase (MPO) colocalizes with macrophages in the human artery wall, and its characteristic oxidation products have been detected in atherosclerotic lesions. Thus, oxidants produced by the enzyme might promote atherosclerosis. However, macrophages in mouse atherosclerotic tissue do not express MPO. Therefore, mice are an inappropriate model for testing the role of MPO in vascular disease. To overcome this problem, we generated and studied transgenic (Tg) mice that contained the human MPO gene. METHODS AND RESULTS: We produced human MPO-Tg mice with use of a Visna virus promoter. To confine MPO expression to macrophages, we lethally irradiated LDL receptor-deficient mice and repopulated their bone marrow with cells from wild-type mice or MPO-Tg mice. Despite having similarly high levels of cholesterol after maintenance on a high-fat, high-cholesterol diet, the MPO-Tg animals developed a 2-fold greater atherosclerotic area in the aorta than did mice transplanted with wild-type bone marrow (P=0.00003). CONCLUSIONS: Our observations indicate that expression of human MPO in macrophages promotes atherosclerosis in hypercholesterolemic mice, raising the possibility that the enzyme might be a potential therapeutic target for preventing cardiovascular disease in humans.


Assuntos
Aterosclerose/etiologia , Macrófagos/enzimologia , Peroxidase/farmacologia , Animais , Doenças da Aorta/etiologia , Transplante de Medula Óssea , Colesterol/administração & dosagem , Técnicas de Transferência de Genes , Humanos , Hipercolesterolemia/complicações , Macrófagos/transplante , Camundongos , Camundongos Transgênicos , Peroxidase/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA