Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 154
Filtrar
1.
Annu Rev Immunol ; 29: 415-45, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21219177

RESUMO

The modern rise in obesity and its strong association with insulin resistance and type 2 diabetes have elicited interest in the underlying mechanisms of these pathologies. The discovery that obesity itself results in an inflammatory state in metabolic tissues ushered in a research field that examines the inflammatory mechanisms in obesity. Here, we summarize the unique features of this metabolic inflammatory state, termed metaflammation and defined as low-grade, chronic inflammation orchestrated by metabolic cells in response to excess nutrients and energy. We explore the effects of such inflammation in metabolic tissues including adipose, liver, muscle, pancreas, and brain and its contribution to insulin resistance and metabolic dysfunction. Another area in which many unknowns still exist is the origin or mechanism of initiation of inflammatory signaling in obesity. We discuss signals or triggers to the inflammatory response, including the possibility of endoplasmic reticulum stress as an important contributor to metaflammation. Finally, we examine anti-inflammatory therapies for their potential in the treatment of obesity-related insulin resistance and glucose intolerance.


Assuntos
Inflamação/metabolismo , Obesidade/complicações , Obesidade/metabolismo , Transdução de Sinais , Animais , Metabolismo Energético , Humanos , Inflamação/complicações , Resistência à Insulina , Doenças Metabólicas/metabolismo
2.
Cell ; 171(5): 1094-1109.e15, 2017 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-29149604

RESUMO

Cholesterol is a critical nutrient requiring tight constraint in the endoplasmic reticulum (ER) due to its uniquely challenging biophysical properties. While the mechanisms by which the ER defends against cholesterol insufficiency are well described, it remains unclear how the ER senses and effectively defends against cholesterol excess. Here, we identify the ER-bound transcription factor nuclear factor erythroid 2 related factor-1, Nrf1/Nfe2L1, as a critical mediator of this process. We show that Nrf1 directly binds to and specifically senses cholesterol in the ER through a defined domain and that cholesterol regulates Nrf1 turnover, processing, localization, and activity. In Nrf1 deficiency, in vivo cholesterol challenges induce massive hepatic cholesterol accumulation and damage, which is rescued by replacing Nrf1 exogenously. This Nrf1-mediated mechanism involves the suppression of CD36-driven inflammatory signaling and derepression of liver X receptor activity. These findings reveal Nrf1 as a guardian of cholesterol homeostasis and a core component of adaptive responses to excess cellular cholesterol.


Assuntos
Colesterol/metabolismo , Retículo Endoplasmático/metabolismo , Fígado/metabolismo , Fator 1 Nuclear Respiratório/metabolismo , Animais , Antígenos CD36/metabolismo , Fígado Gorduroso/metabolismo , Regulação da Expressão Gênica , Homeostase , Humanos , Fígado/citologia , Camundongos , Transcrição Gênica
3.
Nature ; 603(7902): 736-742, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35264794

RESUMO

Cells display complex intracellular organization by compartmentalization of metabolic processes into organelles, yet the resolution of these structures in the native tissue context and their functional consequences are not well understood. Here we resolved the three-dimensional structural organization of organelles in large (more than 2.8 × 105 µm3) volumes of intact liver tissue (15 partial or full hepatocytes per condition) at high resolution (8 nm isotropic pixel size) using enhanced focused ion beam scanning electron microscopy1,2 imaging followed by deep-learning-based automated image segmentation and 3D reconstruction. We also performed a comparative analysis of subcellular structures in liver tissue of lean and obese mice and found substantial alterations, particularly in hepatic endoplasmic reticulum (ER), which undergoes massive structural reorganization characterized by marked disorganization of stacks of ER sheets3 and predominance of ER tubules. Finally, we demonstrated the functional importance of these structural changes by monitoring the effects of experimental recovery of the subcellular organization on cellular and systemic metabolism. We conclude that the hepatic subcellular organization of the ER architecture are highly dynamic, integrated with the metabolic state and critical for adaptive homeostasis and tissue health.


Assuntos
Retículo Endoplasmático , Homeostase , Fígado , Animais , Retículo Endoplasmático/metabolismo , Fígado/citologia , Camundongos , Microscopia/métodos , Organelas
4.
Nature ; 600(7890): 720-726, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34880500

RESUMO

The liberation of energy stores from adipocytes is critical to support survival in times of energy deficit; however, uncontrolled or chronic lipolysis associated with insulin resistance and/or insulin insufficiency disrupts metabolic homeostasis1,2. Coupled to lipolysis is the release of a recently identified hormone, fatty-acid-binding protein 4 (FABP4)3. Although circulating FABP4 levels have been strongly associated with cardiometabolic diseases in both preclinical models and humans4-7, no mechanism of action has yet been described8-10. Here we show that hormonal FABP4 forms a functional hormone complex with adenosine kinase (ADK) and nucleoside diphosphate kinase (NDPK) to regulate extracellular ATP and ADP levels. We identify a substantial effect of this hormone on beta cells and given the central role of beta-cell function in both the control of lipolysis and development of diabetes, postulate that hormonal FABP4 is a key regulator of an adipose-beta-cell endocrine axis. Antibody-mediated targeting of this hormone complex improves metabolic outcomes, enhances beta-cell function and preserves beta-cell integrity to prevent both type 1 and type 2 diabetes. Thus, the FABP4-ADK-NDPK complex, Fabkin, represents a previously unknown hormone and mechanism of action that integrates energy status with the function of metabolic organs, and represents a promising target against metabolic disease.


Assuntos
Proteínas de Ligação a Ácido Graxo , Ilhotas Pancreáticas , Fosfotransferases , Adipócitos/metabolismo , Diabetes Mellitus/metabolismo , Proteínas de Ligação a Ácido Graxo/metabolismo , Humanos , Insulina/metabolismo , Ilhotas Pancreáticas/enzimologia , Ilhotas Pancreáticas/fisiologia , Lipólise , Nucleosídeos/metabolismo , Fosfotransferases/metabolismo
5.
Immunity ; 47(3): 406-420, 2017 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-28930657

RESUMO

Highly ordered interactions between immune and metabolic responses are evolutionarily conserved and paramount for tissue and organismal health. Disruption of these interactions underlies the emergence of many pathologies, particularly chronic non-communicable diseases such as obesity and diabetes. Here, we examine decades of research identifying the complex immunometabolic signaling networks and the cellular and molecular events that occur in the setting of altered nutrient and energy exposures and offer a historical perspective. Furthermore, we describe recent advances such as the discovery that a broad complement of immune cells play a role in immunometabolism and the emerging evidence that nutrients and metabolites modulate inflammatory pathways. Lastly, we discuss how this work may eventually lead to tangible therapeutic advancements to promote health.


Assuntos
Metabolismo Energético , Imunidade , Doenças Metabólicas/imunologia , Doenças Metabólicas/metabolismo , Imunidade Adaptativa , Animais , Evolução Biológica , Comunicação Celular/imunologia , Citocinas/metabolismo , Glucose/metabolismo , Humanos , Sistema Imunitário/citologia , Sistema Imunitário/imunologia , Sistema Imunitário/metabolismo , Imunidade Inata , Inflamação/imunologia , Inflamação/metabolismo , Mediadores da Inflamação , Doenças Metabólicas/terapia , Especificidade de Órgãos , Transdução de Sinais , Pesquisa Translacional Biomédica
6.
Immunity ; 45(6): 1186-1188, 2016 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-28002726

RESUMO

Chronic metabolic challenges have severe consequences on physiological systems. In this issue of Immunity, Ito et al. (2016) show that defects in cholesterol metabolism in CD11c+ immune cells result in impaired antigen presentation and ultimately in autoimmune disease.


Assuntos
Apresentação de Antígeno , Colesterol , Doenças Autoimunes , Humanos
7.
J Immunol ; 210(8): 1086-1097, 2023 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-36883861

RESUMO

Fatty acid-binding protein 4 (FABP4) is a critical immune-metabolic modulator, mainly expressed in adipocytes and macrophages, secreted from adipocytes in association with lipolysis, and plays essential pathogenic roles in cardiovascular and metabolic diseases. We previously reported Chlamydia pneumoniae infecting murine 3T3-L1 adipocytes and causing lipolysis and FABP4 secretion in vitro. However, it is still unknown whether C. pneumoniae intranasal lung infection targets white adipose tissues (WATs), induces lipolysis, and causes FABP4 secretion in vivo. In this study, we demonstrate that C. pneumoniae lung infection causes robust lipolysis in WAT. Infection-induced WAT lipolysis was diminished in FABP4-/- mice or FABP4 inhibitor-pretreated wild-type mice. Infection by C. pneumoniae in wild-type but not FABP4-/- mice induces the accumulation of TNF-α- and IL-6-producing M1-like adipose tissue macrophages in WAT. Infection-induced WAT pathology is augmented by endoplasmic reticulum (ER) stress/the unfolded protein response (UPR), which is abrogated by treatment with azoramide, a modulator of the UPR. C. pneumoniae lung infection is suggested to target WAT and induce lipolysis and FABP4 secretion in vivo via ER stress/UPR. FABP4 released from infected adipocytes may be taken up by other neighboring intact adipocytes or adipose tissue macrophages. This process can further induce ER stress activation and trigger lipolysis and inflammation, followed by FABP4 secretion, leading to WAT pathology. A better understanding of the role of FABP4 in C. pneumoniae infection-induced WAT pathology will provide the basis for rational intervention measures directed at C. pneumoniae infection and metabolic syndrome, such as atherosclerosis, for which robust epidemiologic evidence exists.


Assuntos
Tecido Adiposo Branco , Infecções por Chlamydophila , Proteínas de Ligação a Ácido Graxo , Pneumonia Bacteriana , Animais , Camundongos , Tecido Adiposo Branco/patologia , Chlamydophila pneumoniae , Proteínas de Ligação a Ácido Graxo/metabolismo , Pulmão/microbiologia , Pulmão/patologia , Infecções por Chlamydophila/patologia , Pneumonia Bacteriana/patologia
8.
Cell ; 140(6): 900-17, 2010 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-20303879

RESUMO

The endoplasmic reticulum (ER) is the major site in the cell for protein folding and trafficking and is central to many cellular functions. Failure of the ER's adaptive capacity results in activation of the unfolded protein response (UPR), which intersects with many different inflammatory and stress signaling pathways. These pathways are also critical in chronic metabolic diseases such as obesity, insulin resistance, and type 2 diabetes. The ER and related signaling networks are emerging as a potential site for the intersection of inflammation and metabolic disease.


Assuntos
Retículo Endoplasmático/fisiologia , Doenças Metabólicas/fisiopatologia , Resposta a Proteínas não Dobradas , Animais , Humanos , Inflamação/imunologia , Inflamação/fisiopatologia , Doenças Metabólicas/imunologia , Dobramento de Proteína , Estresse Fisiológico
9.
Cell ; 140(3): 338-48, 2010 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-20144759

RESUMO

As chronic inflammation is a hallmark of obesity, pathways that integrate nutrient- and pathogen sensing pathways are of great interest in understanding the mechanisms of insulin resistance, type 2 diabetes, and other chronic metabolic pathologies. Here, we provide evidence that double-stranded RNA-dependent protein kinase (PKR) can respond to nutrient signals as well as endoplasmic reticulum (ER) stress and coordinate the activity of other critical inflammatory kinases such as the c-Jun N-terminal kinase (JNK) to regulate insulin action and metabolism. PKR also directly targets and modifies insulin receptor substrate and hence integrates nutrients and insulin action with a defined pathogen response system. Dietary and genetic obesity features marked activation of PKR in adipose and liver tissues and absence of PKR alleviates metabolic deterioration due to nutrient or energy excess in mice. These findings demonstrate PKR as a critical component of an inflammatory complex that responds to nutrients and organelle dysfunction.


Assuntos
Doenças Metabólicas/metabolismo , eIF-2 Quinase/metabolismo , Animais , Feminino , Humanos , Proteínas Substratos do Receptor de Insulina/metabolismo , MAP Quinase Quinase 4/metabolismo , Masculino , Camundongos , eIF-2 Quinase/genética
10.
Mol Cell ; 66(6): 731-733, 2017 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-28622517

RESUMO

A new mechanism linking ER dysfunction to metabolic inflammation is discovered in a recent study by Shan et al. (2017), which demonstrated ER stress-induced rewiring of adipose tissue macrophage polarization by IRE1α activation, leading to impaired systemic glucose homeostasis.


Assuntos
Macrófagos , Obesidade , Tecido Adiposo , Homeostase , Humanos , Inflamação
12.
J Lipid Res ; 64(6): 100386, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37172691

RESUMO

Levels of circulating fatty acid binding protein 4 (FABP4) protein are strongly associated with obesity and metabolic disease in both mice and humans, and secretion is stimulated by ß-adrenergic stimulation both in vivo and in vitro. Previously, lipolysis-induced FABP4 secretion was found to be significantly reduced upon pharmacological inhibition of adipose triglyceride lipase (ATGL) and was absent from adipose tissue explants from mice specifically lacking ATGL in their adipocytes (ATGLAdpKO). Here, we find that upon activation of ß-adrenergic receptors in vivo, ATGLAdpKO mice unexpectedly exhibited significantly higher levels of circulating FABP4 as compared with ATGLfl/fl controls, despite no corresponding induction of lipolysis. We generated an additional model with adipocyte-specific deletion of both FABP4 and ATGL (ATGL/FABP4AdpKO) to evaluate the cellular source of this circulating FABP4. In these animals, there was no evidence of lipolysis-induced FABP4 secretion, indicating that the source of elevated FABP4 levels in ATGLAdpKO mice was indeed from the adipocytes. ATGLAdpKO mice exhibited significantly elevated corticosterone levels, which positively correlated with plasma FABP4 levels. Pharmacological inhibition of sympathetic signaling during lipolysis using hexamethonium or housing mice at thermoneutrality to chronically reduce sympathetic tone significantly reduced FABP4 secretion in ATGLAdpKO mice compared with controls. Therefore, activity of a key enzymatic step of lipolysis mediated by ATGL, per se, is not required for in vivo stimulation of FABP4 secretion from adipocytes, which can be induced through sympathetic signaling.


Assuntos
Lipase , Lipólise , Animais , Camundongos , Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Proteínas de Ligação a Ácido Graxo/genética , Proteínas de Ligação a Ácido Graxo/metabolismo , Lipase/genética , Lipase/metabolismo , Lipólise/fisiologia
13.
Cell ; 135(1): 20-2, 2008 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-18854151

RESUMO

Obesity is characterized by chronic activation of inflammatory pathways in peripheral tissues. In this issue, Zhang et al. (2008) demonstrate that inflammation also occurs in the central nervous system where it disrupts activity of the hypothalamus leading to resistance to leptin that is mediated by activation of IKK and the endoplasmic reticulum stress response.


Assuntos
Hipotálamo/fisiopatologia , Obesidade/fisiopatologia , Hipernutrição/fisiopatologia , Animais , Retículo Endoplasmático/fisiologia , Humanos , Quinase I-kappa B/metabolismo , Leptina/metabolismo , Camundongos
14.
Cell ; 134(6): 933-44, 2008 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-18805087

RESUMO

Dysregulation of lipid metabolism in individual tissues leads to systemic disruption of insulin action and glucose metabolism. Utilizing quantitative lipidomic analyses and mice deficient in adipose tissue lipid chaperones aP2 and mal1, we explored how metabolic alterations in adipose tissue are linked to whole-body metabolism through lipid signals. A robust increase in de novo lipogenesis rendered the adipose tissue of these mice resistant to the deleterious effects of dietary lipid exposure. Systemic lipid profiling also led to identification of C16:1n7-palmitoleate as an adipose tissue-derived lipid hormone that strongly stimulates muscle insulin action and suppresses hepatosteatosis. Our data reveal a lipid-mediated endocrine network and demonstrate that adipose tissue uses lipokines such as C16:1n7-palmitoleate to communicate with distant organs and regulate systemic metabolic homeostasis.


Assuntos
Tecido Adiposo/metabolismo , Ácidos Graxos Monoinsaturados/análise , Hormônios/análise , Metabolismo dos Lipídeos , Animais , Distribuição da Gordura Corporal , Proteínas de Ligação a Ácido Graxo/genética , Proteínas de Ligação a Ácido Graxo/metabolismo , Ácidos Graxos Monoinsaturados/sangue , Ácidos Graxos Monoinsaturados/metabolismo , Insulina/metabolismo , Lipogênese , Camundongos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Estearoil-CoA Dessaturase/metabolismo
15.
Nature ; 542(7640): 177-185, 2017 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-28179656

RESUMO

Proper regulation and management of energy, substrate diversity and quantity, as well as macromolecular synthesis and breakdown processes, are fundamental to cellular and organismal survival and are paramount to health. Cellular and multicellular organization are defended by the immune response, a robust and critical system through which self is distinguished from non-self, pathogenic signals are recognized and eliminated, and tissue homeostasis is safeguarded. Many layers of evolutionarily conserved interactions occur between immune response and metabolism. Proper maintenance of this delicate balance is crucial for health and has important implications for many pathological states such as obesity, diabetes, and other chronic non-communicable diseases.


Assuntos
Inflamação/imunologia , Inflamação/metabolismo , Doenças Metabólicas/imunologia , Doenças Metabólicas/metabolismo , Imunidade Adaptativa/genética , Animais , Ensaios Clínicos como Assunto , Citocinas/metabolismo , Evolução Molecular , Estudo de Associação Genômica Ampla , Hormônios/metabolismo , Humanos , Inflamação/complicações , Inflamação/genética , Invertebrados/imunologia , Invertebrados/metabolismo , Doenças Metabólicas/complicações , Doenças Metabólicas/genética , Obesidade/complicações , Obesidade/genética , Obesidade/imunologia , Obesidade/metabolismo , Organelas/metabolismo , Transdução de Sinais
16.
Nature ; 543(7644): 252-256, 2017 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-28219080

RESUMO

Tissue-resident memory T (TRM) cells persist indefinitely in epithelial barrier tissues and protect the host against pathogens. However, the biological pathways that enable the long-term survival of TRM cells are obscure. Here we show that mouse CD8+ TRM cells generated by viral infection of the skin differentially express high levels of several molecules that mediate lipid uptake and intracellular transport, including fatty-acid-binding proteins 4 and 5 (FABP4 and FABP5). We further show that T-cell-specific deficiency of Fabp4 and Fabp5 (Fabp4/Fabp5) impairs exogenous free fatty acid (FFA) uptake by CD8+ TRM cells and greatly reduces their long-term survival in vivo, while having no effect on the survival of central memory T (TCM) cells in lymph nodes. In vitro, CD8+ TRM cells, but not CD8+ TCM cells, demonstrated increased mitochondrial oxidative metabolism in the presence of exogenous FFAs; this increase was not seen in Fabp4/Fabp5 double-knockout CD8+ TRM cells. The persistence of CD8+ TRM cells in the skin was strongly diminished by inhibition of mitochondrial FFA ß-oxidation in vivo. Moreover, skin CD8+ TRM cells that lacked Fabp4/Fabp5 were less effective at protecting mice from cutaneous viral infection, and lung Fabp4/Fabp5 double-knockout CD8+ TRM cells generated by skin vaccinia virus (VACV) infection were less effective at protecting mice from a lethal pulmonary challenge with VACV. Consistent with the mouse data, increased FABP4 and FABP5 expression and enhanced extracellular FFA uptake were also demonstrated in human CD8+ TRM cells in normal and psoriatic skin. These results suggest that FABP4 and FABP5 have a critical role in the maintenance, longevity and function of CD8+ TRM cells, and suggest that CD8+ TRM cells use exogenous FFAs and their oxidative metabolism to persist in tissue and to mediate protective immunity.


Assuntos
Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/metabolismo , Ácidos Graxos não Esterificados/metabolismo , Memória Imunológica/imunologia , Metabolismo dos Lipídeos , Animais , Transporte Biológico , Linfócitos T CD8-Positivos/imunologia , Sobrevivência Celular , Proteínas de Ligação a Ácido Graxo/deficiência , Proteínas de Ligação a Ácido Graxo/metabolismo , Feminino , Humanos , Camundongos , Proteínas de Neoplasias/deficiência , Proteínas de Neoplasias/metabolismo , Oxirredução , Psoríase , Pele/citologia , Pele/imunologia , Pele/virologia , Vacínia/imunologia , Vacínia/prevenção & controle , Vaccinia virus/imunologia
17.
Eur Heart J ; 41(26): 2456-2468, 2020 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-31821481

RESUMO

AIMS: Obesity is a risk factor of abdominal aortic aneurysm (AAA). Inflammatory cytokine interleukin-18 (IL18) has two receptors: IL18 receptor (IL18r) and Na-Cl co-transporter (NCC). In human and mouse AAA lesions, IL18 colocalizes to its receptors at regions rich in adipocytes, suggesting a role of adipocytes in promoting IL18 actions in AAA development. METHODS AND RESULTS: We localized both IL18r and NCC in human and mouse AAA lesions. Murine AAA development required both receptors. In mouse AAA lesions, IL18 binding to these receptors increased at regions enriched in adipocytes or adjacent to perivascular adipose tissue. 3T3-L1 adipocytes enhanced IL18 binding to macrophages, aortic smooth muscle cells (SMCs), and endothelial cells by inducing the expression of both IL18 receptors on these cells. Adipocytes also enhanced IL18r and IL18 expression from T cells and macrophages, AAA-pertinent protease expression from macrophages, and SMC apoptosis. Perivascular implantation of adipose tissue from either diet-induced obese mice or lean mice but not that from leptin-deficient ob/ob mice exacerbated AAA development in recipient mice. Further experiments established an essential role of adipocyte leptin and fatty acid-binding protein 4 (FABP4) in promoting IL18 binding to macrophages and possibly other inflammatory and vascular cells by inducing their expression of IL18, IL18r, and NCC. CONCLUSION: Interleukin-18 uses both IL18r and NCC to promote AAA formation. Lesion adipocyte and perivascular adipose tissue contribute to AAA pathogenesis by releasing leptin and FABP4 that induce IL18, IL18r, and NCC expression and promote IL18 actions.


Assuntos
Adipócitos , Aneurisma da Aorta Abdominal , Interleucina-18 , Animais , Aneurisma da Aorta Abdominal/etiologia , Modelos Animais de Doenças , Células Endoteliais , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Interleucina-18 , Transdução de Sinais
18.
Nature ; 513(7518): 440-3, 2014 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-25043031

RESUMO

Eukaryotic cells coordinately control anabolic and catabolic processes to maintain cell and tissue homeostasis. Mechanistic target of rapamycin complex 1 (mTORC1) promotes nutrient-consuming anabolic processes, such as protein synthesis. Here we show that as well as increasing protein synthesis, mTORC1 activation in mouse and human cells also promotes an increased capacity for protein degradation. Cells with activated mTORC1 exhibited elevated levels of intact and active proteasomes through a global increase in the expression of genes encoding proteasome subunits. The increase in proteasome gene expression, cellular proteasome content, and rates of protein turnover downstream of mTORC1 were all dependent on induction of the transcription factor nuclear factor erythroid-derived 2-related factor 1 (NRF1; also known as NFE2L1). Genetic activation of mTORC1 through loss of the tuberous sclerosis complex tumour suppressors, TSC1 or TSC2, or physiological activation of mTORC1 in response to growth factors or feeding resulted in increased NRF1 expression in cells and tissues. We find that this NRF1-dependent elevation in proteasome levels serves to increase the intracellular pool of amino acids, which thereby influences rates of new protein synthesis. Therefore, mTORC1 signalling increases the efficiency of proteasome-mediated protein degradation for both quality control and as a mechanism to supply substrate for sustained protein synthesis.


Assuntos
Complexos Multiproteicos/metabolismo , Biossíntese de Proteínas , Proteínas/metabolismo , Proteólise , Serina-Treonina Quinases TOR/metabolismo , Aminoácidos/metabolismo , Animais , Humanos , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Endogâmicos C57BL , Fator 1 Nuclear Respiratório/genética , Fator 1 Nuclear Respiratório/metabolismo , Complexo de Endopeptidases do Proteassoma/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas/química , Transdução de Sinais , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Transcrição Gênica
19.
J Lipid Res ; 60(4): 734-740, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30705117

RESUMO

Although counterregulatory hormones and mediators of the fight-or-flight responses are well defined at many levels, how energy stores per se are integrated into this system remains an enigmatic question. Recent years have seen the adipose tissue become a central focus for mediating intracellular signaling and communication through the release of a variety of bioactive lipids and substrates, as well as various adipokines. A critical integration node among these mediators and responses is controlled by FA binding protein 4 (FABP4), also known as adipocyte protein 2 (aP2), which is highly expressed in adipose tissue and functions as a lipid chaperone protein. Recently, it was demonstrated that FABP4 is a secreted hormone that has roles in maintaining glucose homeostasis, representing a key juncture facilitating communication between energy-storage systems and distant organs to respond to life-threatening situations. However, chronic engagement of FABP4 under conditions of immunometabolic stress, such as obesity, exacerbates a number of immunometabolic diseases, including diabetes, asthma, cancer, and atherosclerosis. In both preclinical mouse models and humans, levels of circulating FABP4 have been correlated with metabolic disease incidence, and reducing FABP4 levels or activity is associated with improved metabolic health. In this review, we will discuss the intriguing emerging biology of this protein, including potential therapeutic options for targeting circulating FABP4.


Assuntos
Proteínas de Ligação a Ácido Graxo/metabolismo , Animais , Metabolismo Energético , Proteínas de Ligação a Ácido Graxo/sangue , Humanos , Camundongos
20.
Nature ; 502(7472): 550-4, 2013 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-24153306

RESUMO

Food intake increases the activity of hepatic de novo lipogenesis, which mediates the conversion of glucose to fats for storage or use. In mice, this program follows a circadian rhythm that peaks with nocturnal feeding and is repressed by Rev-erbα/ß and an HDAC3-containing complex during the day. The transcriptional activators controlling rhythmic lipid synthesis in the dark cycle remain poorly defined. Disturbances in hepatic lipogenesis are also associated with systemic metabolic phenotypes, suggesting that lipogenesis in the liver communicates with peripheral tissues to control energy substrate homeostasis. Here we identify a PPARδ-dependent de novo lipogenic pathway in the liver that modulates fat use by muscle via a circulating lipid. The nuclear receptor PPARδ controls diurnal expression of lipogenic genes in the dark/feeding cycle. Liver-specific PPARδ activation increases, whereas hepatocyte-Ppard deletion reduces, muscle fatty acid uptake. Unbiased metabolite profiling identifies phosphatidylcholine 18:0/18:1 (PC(18:0/18:1) as a serum lipid regulated by diurnal hepatic PPARδ activity. PC(18:0/18:1) reduces postprandial lipid levels and increases fatty acid use through muscle PPARα. High-fat feeding diminishes rhythmic production of PC(18:0/18:1), whereas PC(18:0/18:1) administration in db/db mice (also known as Lepr(-/-)) improves metabolic homeostasis. These findings reveal an integrated regulatory circuit coupling lipid synthesis in the liver to energy use in muscle by coordinating the activity of two closely related nuclear receptors. These data implicate alterations in diurnal hepatic PPARδ-PC(18:0/18:1) signalling in metabolic disorders, including obesity.


Assuntos
Ritmo Circadiano , Ácidos Graxos/metabolismo , Lipídeos/sangue , Lipogênese , Fígado/metabolismo , Acetil-CoA Carboxilase/metabolismo , Animais , Diabetes Mellitus/metabolismo , Regulação da Expressão Gênica , Homeostase , Lipogênese/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculos/metabolismo , Obesidade/metabolismo , PPAR delta/metabolismo , Fosfatidilcolinas/sangue , Análise de Componente Principal
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA