Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 183(3): 684-701.e14, 2020 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-33058756

RESUMO

Positive selection in Europeans at the 2q21.3 locus harboring the lactase gene has been attributed to selection for the ability of adults to digest milk to survive famine in ancient times. However, the 2q21.3 locus is also associated with obesity and type 2 diabetes in humans, raising the possibility that additional genetic elements in the locus may have contributed to evolutionary adaptation to famine by promoting energy storage, but which now confer susceptibility to metabolic diseases. We show here that the miR-128-1 microRNA, located at the center of the positively selected locus, represents a crucial metabolic regulator in mammals. Antisense targeting and genetic ablation of miR-128-1 in mouse metabolic disease models result in increased energy expenditure and amelioration of high-fat-diet-induced obesity and markedly improved glucose tolerance. A thrifty phenotype connected to miR-128-1-dependent energy storage may link ancient adaptation to famine and modern metabolic maladaptation associated with nutritional overabundance.


Assuntos
Doenças Metabólicas/genética , MicroRNAs/genética , Adipócitos Marrons/patologia , Adiposidade , Alelos , Animais , Diferenciação Celular , Linhagem Celular , Células Cultivadas , Dieta Hiperlipídica , Metabolismo Energético , Epigênese Genética , Loci Gênicos , Glucose/metabolismo , Homeostase , Humanos , Hipertrofia , Resistência à Insulina , Leptina/deficiência , Leptina/metabolismo , Masculino , Mamíferos/genética , Camundongos Endogâmicos C57BL , Camundongos Obesos , MicroRNAs/metabolismo , Obesidade/genética , Oligonucleotídeos/metabolismo , Especificidade da Espécie
2.
Pharm Res ; 40(2): 467-480, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36050546

RESUMO

Enhancing thermogenesis by increasing the amount and activity of brown and brite adipocytes is a potential therapeutic target for obesity and its associated diseases. Diet plays important roles in energy metabolism and a myriad of dietary components including lipids are known to regulate thermogenesis through recruitment and activation of brown and brite adipocytes. Depending on types of fatty acids (FAs), the major constituent in lipids, their health benefits differ. Long-chain polyunsaturated FAs (PUFAs), especially n-3 PUFAs remodel adipose tissues in a healthier manner with reduced inflammation and enhanced thermogenesis, while saturated FAs exhibit contrasting effects. Lipid mediators derived from FAs act as autocrine/paracrine as well as endocrine factors to regulate thermogenesis. We discuss lipid mediators that may contribute to the differential effects of FAs on adipose tissue remodeling and hence, cardiometabolic diseases. We also discuss current understanding of molecular and cellular mechanisms through which n-3 PUFAs enhance thermogenesis. Elucidating molecular details of beneficial effects of n-3 PUFAs on thermogenesis is expected to provide information that can be used for development of novel therapeutics for obesity and its associated diseases.


Assuntos
Tecido Adiposo Marrom , Ácidos Graxos Ômega-3 , Humanos , Tecido Adiposo Marrom/metabolismo , Ácidos Graxos/metabolismo , Ácidos Graxos Ômega-3/metabolismo , Ácidos Graxos Ômega-3/farmacologia , Obesidade/metabolismo , Tecido Adiposo Branco , Termogênese , Metabolismo Energético
3.
Fungal Genet Biol ; 160: 103683, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35278684

RESUMO

Fusarium graminearum is the causal agent of Fusarium head blight in cereal crops. As in other filamentous ascomycetes, F. graminearum contains genes encoding putative hydrophobins, which are small secreted amphiphilic proteins with eight conserved cysteine residues. Here, we investigated the roles of all five hydrophobin genes (designated FgHyd1, FgHyd2, FgHyd3, FgHyd4, and FgHyd5) in various mycological traits of F. graminearum. Gene expression analyses revealed that the five FgHyd genes, all of which were under the control of G protein signaling or velvet complex proteins, were differentially expressed under various developmental conditions. Three genes (FgHyd1, FgHyd2, and FgHyd3) were constitutively expressed in all aerial structures examined (hyphae, conidia, and perithecia), and two genes (FgHyd1 and FgHyd2) were also expressed in submerged hyphae. FgHyd3 was exclusively expressed in aerial hyphae on solid surfaces, including rice grains. These genes showed markedly reduced expression in F. asiaticum, which was a closely related to F. graminearum but exhibited different mycological traits from F. graminearum. Phenotypic analyses of various gene deletion strains, including the quintuple deletion (ΔFgHyd12345) strain, confirmed that in addition to their typical functions, all five FgHyd genes were involved in other traits, such as conidiation, pathogenicity, and secondary metabolism in F. graminearum. Both RNA-seq and chemical analyses confirmed that ΔFgHyd led to overproduction of specific terpenoid compounds (e.g., trichothecenes), which has not been reported previously. Nevertheless, the lack of complete phenotypic loss of any of the traits examined, even in the ΔFgHyd12345 strain, and little cumulative action of all five FgHyd genes strongly suggest that all five hydrophobins are redundant in function and are not absolutely essential for these fungal traits in F. graminearum.


Assuntos
Fusarium , Proteínas Fúngicas/metabolismo , Doenças das Plantas/microbiologia , Metabolismo Secundário/genética , Esporos Fúngicos
4.
Int J Mol Sci ; 23(16)2022 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-36012691

RESUMO

Adaptive natural killer (NK) cells expressing self-specific inhibitory killer-cell immunoglobulin-like receptors (KIRs) can be expanded in vivo in response to human cytomegalovirus (HCMV) infection. Developing a method to preferentially expand this subset is essential for effective targeting of allogeneic cancer cells. A previous study developed an in vitro method to generate single KIR+ NK cells for enhanced targeting of the primary acute lymphoblastic leukemia cells; however, the expansion rate was quite low. Here, we present an effective expansion method using genetically modified K562-HLA-E feeder cells for long-term proliferation of adaptive NK cells displaying highly differentiated phenotype and comparable cytotoxicity, CD107a, and interferon-γ (IFN-γ) production. More importantly, our expansion method achieved more than a 10,000-fold expansion of adaptive NK cells after 6 weeks of culture, providing a high yield of alloreactive NK cells for cell therapy against cancer.


Assuntos
Infecções por Citomegalovirus , Subfamília C de Receptores Semelhantes a Lectina de Células NK , Citomegalovirus , Antígenos de Histocompatibilidade Classe I , Humanos , Células K562 , Células Matadoras Naturais , Subfamília C de Receptores Semelhantes a Lectina de Células NK/genética , Receptores KIR , Antígenos HLA-E
5.
Int J Mol Sci ; 22(7)2021 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-33916086

RESUMO

Fibrates, including fenofibrate, are a class of hypolipidemic drugs that activate peroxisome proliferator-activated receptor α (PPARα), which in-turn regulates the expression of lipid and lipoprotein metabolism genes. We investigated whether fenofibrate can reduce visceral obesity and nonalcoholic fatty liver disease via adipose tissue PPARα activation in female ovariectomized (OVX) C57BL/6J mice fed a high-fat diet (HFD), a mouse model of obese postmenopausal women. Fenofibrate reduced body weight gain (-38%, p < 0.05), visceral adipose tissue mass (-46%, p < 0.05), and visceral adipocyte size (-20%, p < 0.05) in HFD-fed obese OVX mice. In addition, plasma levels of alanine aminotransferase and aspartate aminotransferase, as well as free fatty acids, triglycerides, and total cholesterol, were decreased. Fenofibrate also inhibited hepatic lipid accumulation (-69%, p < 0.05) and infiltration of macrophages (-72%, p < 0.05), while concomitantly upregulating the expression of fatty acid ß-oxidation genes targeted by PPARα and decreasing macrophage infiltration and mRNA expression of inflammatory factors in visceral adipose tissue. These results suggest that fenofibrate inhibits visceral obesity, as well as hepatic steatosis and inflammation, in part through visceral adipose tissue PPARα activation in obese female OVX mice.


Assuntos
Dislipidemias/prevenção & controle , Fígado Gorduroso/prevenção & controle , Fenofibrato/uso terapêutico , Hipolipemiantes/uso terapêutico , Obesidade Abdominal/prevenção & controle , Adipócitos/efeitos dos fármacos , Animais , Dieta Hiperlipídica , Avaliação Pré-Clínica de Medicamentos , Feminino , Fenofibrato/farmacologia , Hipolipemiantes/farmacologia , Gordura Intra-Abdominal/metabolismo , Camundongos Endogâmicos C57BL , Ovariectomia , PPAR gama/metabolismo
6.
Surg Radiol Anat ; 43(7): 1041-1044, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33048246

RESUMO

Extensor hallucis capsularis (EHC) is an accessory tendon located medially to extensor halluces longus (EHL) tendon. Most EHC is known to originate as a tendinous slip of the EHL tendon, although it may be splitted from the tibialis anterior (TA) tendon or the extensor halluces brevis (EHB) tendon. During routine dissection of a 49-year-old male cadaver, independent muscle bellies of EHC were discovered bilaterally. The EHL muscle arose from the middle anteromedial aspect of fibula, lateral to the origin of TA muscle and medial to extensor digitorum longus (EDL) muscle. An additional muscle bellies were separated from EHL muscle at the point of 6 cm away from EHL origin in the right leg, and 3 cm away in the left. They coursed downward as EHC to reach the first metatarsophalangeal joint capsule. This muscle, unlike the variations identified to date, is considered to extend to EHC, and the name "extensor hallucis capsularis muscle" is offered. This kind of variation may be important for investigating the development of deformity at the first metatarsophalangeal joint, such as hallux valgus.


Assuntos
Variação Anatômica , Hallux/anormalidades , Músculo Esquelético/anormalidades , Tendões/anormalidades , Cadáver , Hallux Valgus/etiologia , Humanos , Cápsula Articular/anormalidades , Masculino , Articulação Metatarsofalângica/anormalidades , Pessoa de Meia-Idade
7.
Adv Exp Med Biol ; 1250: 35-48, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32601936

RESUMO

Injectable in situ-forming hydrogels have been used clinically in diverse biomedical applications. These hydrogels have distinct advantages such as easy management and minimal invasiveness. The hydrogels are aqueous formulations, and a simple injection at the target site replaces a traditional surgical procedure. Here, we review injectable in situ-forming hydrogels that are formulated by physical and chemical methods to deliver proteins and peptides. Prospects for using in situ-forming hydrogels for several specific applications are also discussed.


Assuntos
Hidrogéis , Peptídeos , Proteínas , Sistemas de Liberação de Medicamentos , Humanos , Hidrogéis/administração & dosagem , Hidrogéis/química , Injeções , Peptídeos/administração & dosagem , Proteínas/administração & dosagem
8.
J Lipid Res ; 60(4): 856-868, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30782959

RESUMO

Treatment with PPARγ agonists in vivo improves human adipocyte metabolism, but the cellular mechanisms and possible depot differences in responsiveness to their effects are poorly understood. To examine the ex vivo metabolic effects of rosiglitazone (Rosi), we cultured explants of human visceral (omental) and abdominal subcutaneous adipose tissues for 7 days. Rosi increased mRNA levels of transcriptional regulators of brite/beige adipocytes (PGC1α, PRDM16), triglyceride synthesis (GPAT3, DGAT1), and lipolysis (ATGL) similarly in adipose tissues from both depots. In parallel, Rosi increased key modulators of FA oxidation (UCP1, FABP3, PLIN5 protein), rates of FA oxidation, and protein levels of electron transport complexes, suggesting an enhanced respiratory capacity as confirmed in newly differentiated adipocytes. Rosi led to the formation of small lipid droplets (SLDs) around the adipocyte central lipid droplet; each SLD was decorated with redistributed mitochondria that colocalized with PLIN5. SLD maintenance required lipolysis and FA reesterification. Rosi thus coordinated a structural and metabolic remodeling in adipocytes from both visceral and subcutaneous depots that enhanced oxidative capacity. Selective targeting of these cellular mechanisms to improve adipocyte FA handling may provide a new approach to treat metabolic complications of obesity and diabetes.


Assuntos
Adipócitos/efeitos dos fármacos , Tecido Adiposo/efeitos dos fármacos , Hipoglicemiantes/farmacologia , Gotículas Lipídicas/efeitos dos fármacos , Rosiglitazona/farmacologia , Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Adulto , Idoso , Feminino , Humanos , Gotículas Lipídicas/metabolismo , Masculino , Pessoa de Meia-Idade , Oxirredução , Fenótipo
9.
Biochim Biophys Acta Mol Basis Dis ; 1864(4 Pt A): 1160-1171, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29409985

RESUMO

Accumulation of dysfunctional white adipose tissues increases risks for cardiometabolic diseases in obesity. In addition to white, brown or brite adipose tissues are also present in adult humans and increasing their amount may be protective. Therefore, understanding factors regulating the amount and function of each adipose depot is crucial for developing therapeutic targets for obesity and its associated metabolic diseases. The transforming growth factor beta (TGFß) superfamily, which consists of TGFß, BMPs, GDFs, and activins, controls multiple aspects of adipose biology. This review focuses on the recent development in understanding the role of TGFß superfamily in the regulation of white, brite and brown adipocyte differentiation, adipose tissue fibrosis, and adipocyte metabolic and endocrine functions. TGFß family and their antagonists are produced locally within adipose tissues and their expression levels are altered in obesity. We also discuss their potential contribution to adipose tissue dysfunction in obesity.


Assuntos
Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Diferenciação Celular , Obesidade/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Adipócitos/patologia , Tecido Adiposo/patologia , Animais , Fibrose , Humanos , Obesidade/patologia
10.
Am J Physiol Endocrinol Metab ; 312(1): E58-E71, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-27879248

RESUMO

Increased adipocyte size is hypothesized to signal the recruitment of adipose progenitor cells (APCs) to expand tissue storage capacity. To investigate depot and sex differences in adipose growth, male and female C57BL/6J mice (10 wk-old) were challenged with high-fat (HF) or low-fat (LF) diets (D) for 14 wk. The HFD increased gonadal (GON) depot weight by adipocyte hypertrophy and hyperplasia in females but hypertrophy alone in males. In both sexes, inguinal (ING) adipocytes were smaller than GON, and depot expansion was due to hypertrophy. Matrix metalloproteinase 3 (Mmp3), an antiadipogenic factor, and its inhibitor Timps modulate the extracellular matrix remodeling needed for depot expansion. Mmp3 mRNA was depot different (ING > GON), higher in females than males and mainly expressed in APCs. In males, HFD-induced obesity increased tissue and APC Mmp3 mRNA levels and MMP3 protein and enzymatic activity. In females however, HFD significantly decreased MMP3 protein without affecting its mRNA levels. MMP3 activity also decreased (significant in ING). Timp4 mRNA was expressed mainly in adipocytes, and HFD-induced obesity tended to increase the ratio of TIMP4 to MMP3 protein in females, whereas it decreased it in males. Overexpression of Mmp3 in 3T3-L1 preadipocytes or rhMMP3 protein added to primary human preadipocytes inhibited differentiation, whereas rhTIMP4 improved adipogenesis and attenuated the inhibitory effect of rhMMP3. These data suggest that HFD-induced obesity downregulates APC MMP3 expression to trigger adipogenesis, and adipocyte TIMP4 may modulate this process to regulate hyperplastic vs. hypertrophic adipose tissue expansion, fat distribution, and metabolic health in a sex- and depot-dependent manner.


Assuntos
Adipócitos/metabolismo , Tecido Adiposo/crescimento & desenvolvimento , Dieta Hiperlipídica , Metaloproteinase 3 da Matriz/genética , Obesidade/genética , RNA Mensageiro/metabolismo , Inibidores Teciduais de Metaloproteinases/genética , Células 3T3-L1 , Adipócitos/efeitos dos fármacos , Adipócitos/patologia , Adipogenia/efeitos dos fármacos , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Dieta com Restrição de Gorduras , Feminino , Humanos , Hiperplasia , Hipertrofia , Masculino , Metaloproteinase 3 da Matriz/metabolismo , Metaloproteinase 3 da Matriz/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/metabolismo , Proteínas Recombinantes/farmacologia , Inibidores Teciduais de Metaloproteinases/metabolismo , Inibidores Teciduais de Metaloproteinases/farmacologia , Inibidor Tecidual 4 de Metaloproteinase
11.
J Lipid Res ; 57(7): 1256-63, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27178044

RESUMO

The glucocorticoid-induced leucine zipper (GILZ), a primary target of glucocorticoids, is expressed in human adipocytes, but its importance in adipocyte function is unknown. Because TNFα is increased in obese adipose tissue and antagonizes a number of glucocorticoid actions, we investigated the interplay of these pathways. GILZ knockdown increased and GILZ overexpression decreased interleukin-6 (IL-6) and leptin mRNA and protein secretion. GILZ knockdown increased the magnitude of the glucocorticoid effect on leptin secretion, but did not affect the glucocorticoid suppression of IL-6. Although GILZ silencing decreased adiponectin mRNA levels, it did not affect the amount of adiponectin secreted. GILZ negatively modulated pro-inflammatory signaling pathways, blocking basal and TNFα-stimulated (1 h) p65 nuclear factor κB nuclear translocation and transcriptional activity by binding to p65 in the cytoplasm. GILZ silencing increased basal ERK1/2 and JNK phosphorylation, and decreased MAPK phosphatase-1 protein levels. Longer term TNFα (4 h or 24 h) treatment decreased GILZ expression in human adipocytes. Furthermore, adipose tissue GILZ mRNA levels were reduced in proportion to the degree of obesity and expression of inflammatory markers. Overall, these results suggest that GILZ antagonizes the pro-inflammatory effects of TNFα in human adipocytes, and its downregulation in obesity may contribute to adipose inflammation and dysregulated adipokine production, and thereby systemic metabolism.


Assuntos
Inflamação/genética , Obesidade/genética , Fatores de Transcrição/genética , Fator de Necrose Tumoral alfa/metabolismo , Adipocinas/metabolismo , Tecido Adiposo/metabolismo , Tecido Adiposo/patologia , Adulto , Biópsia , Fosfatase 1 de Especificidade Dupla/metabolismo , Feminino , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Inflamação/metabolismo , Inflamação/patologia , Interleucina-6/metabolismo , Leptina/metabolismo , Sistema de Sinalização das MAP Quinases/genética , Masculino , NF-kappa B/genética , NF-kappa B/metabolismo , Obesidade/metabolismo , Obesidade/patologia , RNA Mensageiro/biossíntese , Fator de Transcrição RelA/genética , Fator de Transcrição RelA/metabolismo , Fatores de Transcrição/biossíntese , Fator de Necrose Tumoral alfa/genética
12.
J Biol Chem ; 289(12): 8473-83, 2014 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-24509860

RESUMO

Mice and humans lacking caveolae due to gene knock-out or inactivating mutations of cavin-1/PTRF have numerous pathologies including markedly aberrant fuel metabolism, lipodystrophy, and muscular dystrophy. We characterized the physiologic/metabolic profile of cavin-1 knock-out mice and determined that they were lean because of reduced white adipose depots. The knock-out mice were resistant to diet-induced obesity and had abnormal lipid metabolism in the major metabolic organs of white and brown fat and liver. Epididymal white fat cells from cavin-1-null mice were small and insensitive to insulin and ß-adrenergic agonists resulting in reduced adipocyte lipid storage and impaired lipid tolerance. At the molecular level, the lipolytic defects in white fat were caused by impaired perilipin phosphorylation, and the reduced triglyceride accumulation was caused by decreased fatty acid uptake and incorporation as well as the virtual absence of insulin-stimulated glucose transport. The livers of cavin-1-null mice were mildly steatotic and did not accumulate more lipid after high-fat feeding. The brown adipose tissues of cavin-1-null mice exhibited decreased mitochondria protein expression, which was restored upon high fat feeding. Taken together, these data suggest that dysfunction in fat, muscle, and liver metabolism in cavin-1-null mice causes a pleiotropic phenotype, one apparently identical to that of humans lacking caveolae in all tissues.


Assuntos
Adipócitos/metabolismo , Adipócitos/patologia , Deleção de Genes , Metabolismo dos Lipídeos , Proteínas de Membrana/genética , Animais , Cavéolas/metabolismo , Cavéolas/patologia , Glucose/metabolismo , Humanos , Insulina/metabolismo , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas de Ligação a RNA , Triglicerídeos/metabolismo
13.
J Biol Chem ; 289(21): 14481-7, 2014 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-24742676

RESUMO

Lipolysis in fat tissue represents a major source of circulating fatty acids. Previously, we have found that lipolysis in adipocytes is controlled by early growth response transcription factor Egr1 that directly inhibits transcription of adipose triglyceride lipase, ATGL (Chakrabarti, P., Kim, J. Y., Singh, M., Shin, Y. K., Kim, J., Kumbrink, J., Wu, Y., Lee, M. J., Kirsch, K. H., Fried, S. K., and Kandror, K. V. (2013) Mol. Cell. Biol. 33, 3659-3666). Here we demonstrate that knockdown of the lipid droplet protein FSP27 (a.k.a. CIDEC) in human adipocytes increases expression of ATGL at the level of transcription, whereas overexpression of FSP27 has the opposite effect. FSP27 suppresses the activity of the ATGL promoter in vitro, and the proximal Egr1 binding site is responsible for this effect. FSP27 co-immunoprecipitates with Egr1 and increases its association with and inhibition of the ATGL promoter. Knockdown of Egr1 attenuates the inhibitory effect of FSP27. These results provide a new model of transcriptional regulation of ATGL.


Assuntos
Adipócitos/metabolismo , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Lipase/metabolismo , Proteínas/metabolismo , Células 3T3-L1 , Adipócitos/citologia , Animais , Proteínas Reguladoras de Apoptose , Sítios de Ligação/genética , Células Cultivadas , Proteína 1 de Resposta de Crescimento Precoce/genética , Expressão Gênica , Células HEK293 , Humanos , Immunoblotting , Lipase/genética , Lipólise/genética , Camundongos , Microscopia Confocal , Mutagênese Sítio-Dirigida , Regiões Promotoras Genéticas/genética , Ligação Proteica , Proteínas/genética , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa
14.
J Biol Chem ; 289(17): 12029-12039, 2014 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-24627478

RESUMO

In adipocytes, lipolysis is a highly regulated process involving hormonal signals, lipid droplet-associated proteins, and lipases. The discovery of new lipid droplet-associated proteins added complexity to the current model of lipolysis. In this study, we used cultured human adipocytes to demonstrate that fat-specific protein 27 (FSP27), an abundantly expressed protein in adipocytes, regulates both basal and stimulated lipolysis by interacting with adipose triglyceride lipase (ATGL, also called desnutrin or PNPLA2). We identified a core domain of FSP27, amino acids 120-220, that interacts with ATGL to inhibit its lipolytic function and promote triglyceride storage. We also defined the role of FSP27 in free fatty acid-induced insulin resistance in adipocytes. FSP27 depletion in human adipocytes increased lipolysis and inhibited insulin signaling by decreasing AKT phosphorylation. However, reducing lipolysis by either depletion of ATGL or expression of exogenous full-length FSP27 or amino acids 120-220 protected human adipocytes against the adverse effects of free fatty acids on insulin signaling. In embryonic fibroblasts derived from ATGL KO mice, exogenous free fatty acids did not affect insulin sensitivity. Our results demonstrate a crucial role for FSP27-ATGL interactions in regulating lipolysis, triglyceride accumulation, and insulin signaling in human adipocytes.


Assuntos
Adipócitos/enzimologia , Resistência à Insulina , Lipase/metabolismo , Lipólise/fisiologia , Proteínas/fisiologia , Células 3T3-L1 , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Animais , Proteínas Reguladoras de Apoptose , Humanos , Insulina/metabolismo , Insulina/farmacologia , Camundongos , Camundongos Knockout , Fosforilação , Ligação Proteica , Proteínas/genética , Transdução de Sinais , Triglicerídeos/metabolismo
15.
Biochim Biophys Acta ; 1842(3): 473-81, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23735216

RESUMO

Central obesity is associated with insulin resistance and dyslipidemia. Thus, the mechanisms that control fat distribution and its impact on systemic metabolism have importance for understanding the risk for diabetes and cardiovascular disease. Hypercortisolemia at the systemic (Cushing's syndrome) or local levels (due to adipose-specific overproduction via 11ß-hydroxysteroid dehydrogenase 1) results in the preferential expansion of central, especially visceral fat depots. At the same time, peripheral subcutaneous depots can become depleted. The biochemical and molecular mechanisms underlying the depot-specific actions of glucocorticoids (GCs) on adipose tissue function remain poorly understood. GCs exert pleiotropic effects on adipocyte metabolic, endocrine and immune functions, and dampen adipose tissue inflammation. GCs also regulate multiple steps in the process of adipogenesis. Acting synergistically with insulin, GCs increase the expression of numerous genes involved in fat deposition. Variable effects of GC on lipolysis are reported, and GC can improve or impair insulin action depending on the experimental conditions. Thus, the net effect of GC on fat storage appears to depend on the physiologic context. The preferential effects of GC on visceral adipose tissue have been linked to higher cortisol production and glucocorticoid receptor expression, but the molecular details of the depot-dependent actions of GCs are only beginning to be understood. In addition, increasing evidence underlines the importance of circadian variations in GCs in relationship to the timing of meals for determining their anabolic actions on the adipocyte. In summary, although the molecular mechanisms remain to be fully elucidated, there is increasing evidence that GCs have multiple, depot-dependent effects on adipocyte gene expression and metabolism that promote central fat deposition. This article is part of a Special Issue entitled: Modulation of Adipose Tissue in Health and Disease.


Assuntos
Adipócitos/metabolismo , Tecido Adiposo/patologia , Síndrome de Cushing/patologia , Obesidade Abdominal/metabolismo , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/genética , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/metabolismo , Adipócitos/patologia , Tecido Adiposo/metabolismo , Síndrome de Cushing/metabolismo , Glucocorticoides/metabolismo , Humanos , Resistência à Insulina/genética , Lipólise/genética , Obesidade Abdominal/genética , Obesidade Abdominal/patologia , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo
16.
Biochem Pharmacol ; 222: 116116, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38460909

RESUMO

Central pattern of fat distribution, especially fat accumulation within the intraabdominal cavity increases risks for cardiometabolic diseases. Portal hypothesis combined with a pathological remodeling in visceral fat is considered the major etiological factor explaining the independent contribution of visceral obesity to cardiometabolic diseases. Excessive remodeling in visceral fat during development of obesity leads to dysfunctions in the depot, characterized by hypertrophy and death of adipocytes, hypoxia, inflammation, and fibrosis. Dysfunctional visceral fat secretes elevated levels of fatty acids, glycerol, and proinflammatory and profibrotic cytokines into the portal vein directly impacting the liver, the central regulator of systemic metabolism. These metabolic and endocrine products induce ectopic fat accumulation, insulin resistance, inflammation, and fibrosis in the liver, which in turn causes or exacerbates systemic metabolic derangements. Elucidation of underlying mechanisms that lead to the pathological remodeling and higher degree of dysfunctions in visceral adipose tissue is therefore, critical for the development of therapeutics to prevent deleterious sequelae in obesity. We review depot differences in metabolic and endocrine properties and expendabilities as well as underlying mechanisms that contribute to the pathophysiological aspects of visceral adiposity in cardiometabolic diseases. We also discuss impacts of different weight loss interventions on visceral adiposity and cardiometabolic diseases.


Assuntos
Doenças Cardiovasculares , Resistência à Insulina , Humanos , Gordura Intra-Abdominal/metabolismo , Obesidade/metabolismo , Inflamação/metabolismo , Doenças Cardiovasculares/metabolismo , Fibrose , Tecido Adiposo/metabolismo
17.
Obesity (Silver Spring) ; 32(1): 70-79, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37929774

RESUMO

OBJECTIVE: This study investigated remodeling of cellular metabolism and structures during browning of primary human adipocytes derived from both visceral and subcutaneous adipose tissues. Effects of glucocorticoids on the browning were also assessed. METHODS: Differentiated omental and subcutaneous human adipocytes were treated with rosiglitazone, with or without dexamethasone, and expression levels of brite adipocyte markers, lipolysis, and lipid droplet and mitochondrial structures were examined. RESULTS: Both omental and subcutaneous adipocytes acquired brite phenotypes upon peroxisome proliferator-activated receptor-γ agonist treatment, and dexamethasone tended to enhance the remodeling. Although rosiglitazone increased lipolysis during treatment, brite adipocytes exhibited lower basal lipolytic rates and enhanced responses to ß-adrenergic agonists or atrial natriuretic peptide. Transcriptome analysis identified induction of both breakdown and biosynthesis of lipids in brite adipocytes. After 60+ days in culture, lipid droplet size increased to ~50 microns, becoming almost unilocular in control adipocytes, and after browning, they acquired paucilocular morphology, clusters of small lipid droplets (1-2 micron) surrounded by mitochondria appearing on the periphery of the central large one. CONCLUSIONS: Metabolic and structural remodeling during browning of primary human adipocytes is similar to previous findings in human adipocytes in vivo, supporting their uses for mechanical studies investigating browning with translational relevance.


Assuntos
Adipócitos , Gordura Subcutânea , Humanos , Rosiglitazona/farmacologia , Rosiglitazona/metabolismo , Adipócitos/metabolismo , Gordura Subcutânea/metabolismo , Lipólise , Dexametasona
18.
Sci Rep ; 14(1): 7390, 2024 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-38548803

RESUMO

Intravesical treatment using either reovirus or natural killer (NK) cells serves as an efficient strategy for the treatment of bladder cancer cells (BCCs); however, corresponding monotherapies have often shown modest cytotoxicity. The potential of a locoregional combination using high-dose reovirus and NK cell therapy in an intravesical approach has not yet been studied. In this study, we evaluated the effectiveness of reoviruses and expanded NK cells (eNK) as potential strategies for the treatment of bladder cancer. The anti-tumor effects of mono-treatment with reovirus type 3 Dearing strain (RC402 and RP116) and in combination with interleukin (IL)-18/-21-pretreated eNK cells were investigated on BCC lines (5637, HT-1376, and 253J-BV) using intravesical therapy to simulate in vitro model. RP116 and IL-18/-21-pretreated eNK cells exhibited effective cytotoxicity against grade 1 carcinoma (5637 cells) when used alone, but not against HT-1376 (grade 2 carcinoma) and 253J-BV cells (derived from a metastatic site). Notably, combining RP116 with IL-18/-21-pretreated eNK cells displayed effective cytotoxicity against both HT-1376 and 253J-BV cells. Our findings underscore the potential of a combination therapy using reoviruses and NK cells as a promising strategy for treating bladder cancer.


Assuntos
Carcinoma , Orthoreovirus , Reoviridae , Neoplasias da Bexiga Urinária , Humanos , Interleucina-18/farmacologia , Interleucina-18/uso terapêutico , Neoplasias da Bexiga Urinária/patologia , Células Matadoras Naturais/patologia , Terapia Combinada
19.
Biochem Biophys Res Commun ; 432(2): 296-301, 2013 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-23399566

RESUMO

Human adipocytes express high levels of two distinct lipid droplet proteins, fat specific protein 27 (FSP27; also called CIDEC), a member of the CIDE family, and perilipin1 (PLIN1), a member of the PAT family. Both proteins play a role in fat metabolism in adipocytes, but how they interact is not known. Our present study demonstrates that FSP27 and PLIN1 co-localize and interact in cultured human primary adipocytes. We also found that the C-terminal domain of FSP27, aa 120-220, interacts with PLIN1. Individual expression of exogenous FSP27 or PLIN1 increased triglyceride content and decreased glycerol release (a measure of lipolysis), but co-expression of both proteins did not further increase triglyceride content or decrease lipolysis in human adipocytes. However, the combination of PLIN1 and FSP27 increased the average size of lipid droplets or caused the formation of unilocular adipocytes. Our data suggest that FSP27 interacts with PLIN1 to regulate lipid droplet size in human adipocytes in a concerted manner.


Assuntos
Adipócitos/metabolismo , Proteínas de Transporte/metabolismo , Fosfoproteínas/metabolismo , Proteínas/metabolismo , Triglicerídeos/metabolismo , Proteínas Reguladoras de Apoptose , Células Cultivadas , Humanos , Lipólise , Perilipina-1 , Proteínas/genética
20.
Nutr Diabetes ; 13(1): 19, 2023 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-37935669

RESUMO

BACKGROUND: Fenofibrate is a hypolipidemic peroxisome proliferator-activated receptor α (PPARα) agonist used clinically to reduce hypercholesterolemia and hypertriglyceridemia. OBJECTIVE: We investigated the effects of fenofibrate on insulin resistance and tissue inflammation in a high-fat diet (HFD)-fed ovariectomized (OVX) C57BL/6J mice, a mouse model of obese postmenopausal women. METHODS: Female OVX mice were randomly divided into 3 groups and received a low-fat diet, an HFD, or an HFD supplemented with 0.05% (w/w) fenofibrate for 9 weeks. Parameters of insulin resistance and tissue inflammation were measured using blood analysis, histological analysis, immunohistochemistry, and quantitative real-time polymerase chain reaction. RESULTS: When fenofibrate was administered to HFD-fed OVX mice for 9 weeks, we observed reductions in body weight gain, adipose tissue mass, and the size of visceral adipocytes without the change of food intake. Fenofibrate improved mild hyperglycemia, severe hyperinsulinemia, and glucose tolerance in these mice. It also reduced pancreatic islet size and insulin-positive ß-cell area to levels similar to those in OVX mice fed a low-fat diet. Concomitantly, administration of fenofibrate not only suppressed pancreatic lipid accumulation but also decreased CD68-positive macrophages in both the pancreas and visceral adipose tissue. Treatment with fenofibrate reduced tumor necrosis factor α (TNFα) mRNA levels in adipose tissue and lowered serum TNFα levels. CONCLUSION: These results suggest that fenofibrate treatment attenuates insulin resistance in part by reducing tissue inflammation and TNFα expression in HFD-fed OVX mice.


Assuntos
Fenofibrato , Hiperlipidemias , Resistência à Insulina , Humanos , Feminino , Camundongos , Animais , Fenofibrato/farmacologia , Fenofibrato/uso terapêutico , Fenofibrato/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Camundongos Obesos , Fígado/metabolismo , Camundongos Endogâmicos C57BL , Obesidade/tratamento farmacológico , Obesidade/metabolismo , Inflamação/tratamento farmacológico , Inflamação/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA