Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37.697
Filtrar
1.
Gene ; 932: 148896, 2025 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-39209183

RESUMO

Pescadillo ribosomal biogenesis factor 1 (PES1), a nucleolar protein initially identified in zebrafish, plays an important role in embryonic development and ribosomal biogenesis. Notably, PES1 has been found to be overexpressed in a number of cancer types, where it contributes to tumorigenesis and cancer progression by promoting cell proliferation, suppressing cellular senescence, modulating the tumor microenvironment (TME) and promoting drug resistance in cancer cells. Moreover, recent emerging evidence suggests that PES1 expression is significantly elevated in the livers of Type 2 diabetes mellitus (T2DM) and obese patients, indicating its involvement in the pathogenesis of metabolic diseases through lipid metabolism regulation. In this review, we present the structural characteristics and biological functions of PES1, as well as complexes in which PES1 participates. Furthermore, we comprehensively summarize the multifaceted role of PES1 in various diseases and the latest insights into its underlying molecular mechanisms. Finally, we discuss the potential clinical translational perspectives of targeting PES1, highlighting its promising as a therapeutic intervention and treatment target.


Assuntos
Neoplasias , Proteínas de Ligação a RNA , Humanos , Animais , Neoplasias/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/genética , Proteínas de Ligação a RNA/metabolismo , Proteínas de Ligação a RNA/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/tratamento farmacológico , Microambiente Tumoral , Metabolismo dos Lipídeos , Terapia de Alvo Molecular/métodos , Obesidade/metabolismo , Obesidade/genética
2.
Chem Pharm Bull (Tokyo) ; 72(10): 845-855, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39358209

RESUMO

Obesity is a global medical issue that can be effectively treated by relieving adipose inflammation and subsequent insulin resistance. Diosgenin (DIOS) has various effects as a steroidal saponin in inflammatory disorders. This study explored the effects and mechanism of DIOS on adipose inflammation and insulin sensitivity, both in silico and in vivo. The high-fat diet-induced obesity model in C57BL/6 mice was divided into five groups: normal chow (NC), high-fat diet (HFD), HFD with atorvastatin 10 mg/kg (AT), HFD with DIOS 100 mg/kg (DIOS 100), and HFD with DIOS 200 mg/kg (DIOS 200). Each group underwent an oral intervention for seven weeks. DIOS significantly suppressed weight gain in the body, liver, and epididymal fat pads. Additionally, it significantly improved fasting glucose and insulin levels, homeostatic model assessment of insulin resistance (HOMA-IR), and oral glucose tolerance test results, and reduced the proportion of total and M1 adipose tissue macrophages. Significant changes were shown in mRNA expression of janus kinase 2 (JAK2), insulin receptor (INRS), insulin receptor substrate 1 (IRS-1), phosphatidylinositol 3-kinase (PI3K), and protein kinase B (Akt), all of which exhibited high binding affinity in the in silico. Safety indices, including aspartate aminotransferase (AST), alanine transaminase (ALT), and creatinine level indicated the preventive effects of DIOS. In conclusion, DIOS improves insulin resistance and obesity-associated inflammation via the PI3K/Akt signaling pathway.


Assuntos
Dieta Hiperlipídica , Diosgenina , Resistência à Insulina , Camundongos Endogâmicos C57BL , Obesidade , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Transdução de Sinais , Animais , Diosgenina/farmacologia , Diosgenina/química , Diosgenina/uso terapêutico , Dieta Hiperlipídica/efeitos adversos , Obesidade/tratamento farmacológico , Obesidade/metabolismo , Camundongos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Masculino
3.
Sci Rep ; 14(1): 23014, 2024 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-39362932

RESUMO

Metabolic dysfunction-associated steatohepatitis (MASH) is a common but frequently unrecognized complication of obesity and type 2 diabetes. The association between these conditions is multifaceted and involves complex interactions between metabolic, inflammatory, and genetic factors. Here we assess the underlying structural and molecular processes focusing on the immunological phase of MASH in the nonobese inflammation and fibrosis (NIF) mouse model and compare it to the human disease as well as other murine models. Histopathology together with synchrotron-radiation-based x-ray micro-computed tomography (SRµCT) was used to investigate structural changes within the hepatic sinusoids network in the NIF mouse in comparison to patients with different severities of MASH. A time-course, bulk RNA-sequencing analysis of liver tissue from NIF mice was performed to identify the dynamics of key processes associated with the pathogenesis. Transcriptomics profiling of the NIF mouse revealed a gradual transition from an initially reactive inflammatory response to a regenerative, pro-fibrotic inflammatory response suggesting new avenues for treatment strategies that focus on immunological targets. Despite the lack of metabolic stress induced liver phenotype, a large similarity between the NIF mouse and the immunological phase of human MASH was detected. The translational value was further supported by the comparative analyses with MASH patients and additional animal models. Finally, the impact of diets known to induce metabolic stress, was explored in the NIF mouse. An obesogenic diet was found to induce key physiological, metabolic, and histologic changes akin to those observed in human MASH.


Assuntos
Modelos Animais de Doenças , Animais , Humanos , Camundongos , Masculino , Fígado/metabolismo , Fígado/patologia , Fígado Gorduroso/metabolismo , Fígado Gorduroso/patologia , Hepatopatia Gordurosa não Alcoólica/metabolismo , Hepatopatia Gordurosa não Alcoólica/patologia , Microtomografia por Raio-X , Camundongos Endogâmicos C57BL , Inflamação/metabolismo , Inflamação/patologia , Obesidade/metabolismo , Obesidade/patologia , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia
4.
Sci Rep ; 14(1): 22796, 2024 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-39354056

RESUMO

Excessive caloric intake and obesity due to high-fat (HFD) and high-disaccharide (HDD) diets have been recognized as major contributing factors to dyslipidemia and metabolic dysfunction-associated steatotic liver disease (MASLD). However, the effect of HFD and HDD without excessive caloric intake is obscure. The aim of the study was to evaluate the effect of physiological caloric intake delivered through HFD and HDD on liver and lipid profiles. The study was performed on 6-week-old male and female (50/50%) Sprague Dawley rats, receiving either a standard (controls, n = 16), HFD (n = 14) or HDD (n = 14) chow. All groups received the same, standard daily calorie rations, titrated weekly to the age of growing rats, for 12 weeks. A panel of metabolic in vivo measurement were performed, followed by histological, biochemical and molecular biology assays on tissues harvested from sacrificed rats. There was no significant difference between the groups in body weight. In contrast to controls, HFD and HDD groups showed metabolic dysfunction-associated steatohepatitis (MASH) characterized by liver steatosis, inflammation, ballooning of hepatocytes and fibrosis. These changes were more pronounced in the HFD than in the HDD group. The HFD group showed significantly higher serum LDL than controls or HDD rats. Furthermore, the HFD group had higher liver protein levels of low-density lipoprotein receptor (LDLR) but lower plasma levels of proprotein convertase subtilisin/kexin type 9 (PCSK9) than the controls or HDD group. There were no differences between sexes in evaluated parameters. The excessive caloric intake and obesity are not prerequisites for the development of MASH and dyslipidemia in rats. The liver changes induced by the HFD and HDD diets exhibit differences in severity, as well as in the expression patterns of LDLR and PCSK9. Notably, these effects are independent of the sex of the rats.


Assuntos
Dieta Hiperlipídica , Dislipidemias , Ingestão de Energia , Obesidade , Ratos Sprague-Dawley , Animais , Dieta Hiperlipídica/efeitos adversos , Masculino , Dislipidemias/etiologia , Dislipidemias/metabolismo , Feminino , Ratos , Obesidade/metabolismo , Obesidade/etiologia , Fígado/metabolismo , Fígado/patologia , Fígado Gorduroso/etiologia , Fígado Gorduroso/metabolismo , Fígado Gorduroso/patologia , Pró-Proteína Convertase 9/metabolismo
5.
Sci Rep ; 14(1): 22834, 2024 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-39354116

RESUMO

Hyperuricemic nephropathy (HN) is renal injury caused by hyperuricemia (HUA). While sleeve gastrectomy (SG) has shown promise in improving renal injury in patients with obesity-related HN, the mechanisms are not fully understood. This study induced an obesity-combined HN model in male ob/ob mice and measured serum uric acid (SUA), creatinine, and other biochemical indicators 6 weeks post-surgery. Renal histological changes were evaluated through staining techniques, and the study also assessed renal adenosine monophosphate-activated protein kinase (AMPK) and nuclear factor erythroid 2-related factor 2 (Nrf2) phosphorylation levels and urate transporter ABCG2 expression. In vitro experiments involved Nrf2 knockdown in AMPK-activated HK-2 cells and ChIP to confirm Nrf2 binding to the ABCG2 promoter. Results showed that SG reduced SUA levels, serum creatinine, and blood urea nitrogen, increased p-AMPK, p-Nrf2 protein, and ABCG2 expression, and alleviated renal fibrosis and inflammation. In vitro, Nrf2 knockdown down-regulated ABCG2 expression, and ChIP confirmed Nrf2's role in ABCG2 transcription. The study suggests that SG may improve renal injury in HN mice by modulating the AMPK/Nrf2 pathway and upregulating ABCG2 transcription.


Assuntos
Proteínas Quinases Ativadas por AMP , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Gastrectomia , Hiperuricemia , Fator 2 Relacionado a NF-E2 , Obesidade , Animais , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/genética , Fator 2 Relacionado a NF-E2/metabolismo , Fator 2 Relacionado a NF-E2/genética , Hiperuricemia/metabolismo , Camundongos , Proteínas Quinases Ativadas por AMP/metabolismo , Masculino , Obesidade/metabolismo , Obesidade/complicações , Obesidade/cirurgia , Gastrectomia/métodos , Transdução de Sinais , Nefropatias/metabolismo , Nefropatias/etiologia , Nefropatias/patologia , Modelos Animais de Doenças , Rim/metabolismo , Rim/patologia , Humanos , Camundongos Endogâmicos C57BL
6.
Physiol Rep ; 12(19): e70019, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39358834

RESUMO

In the present scenario, obesity is a challenging health problem and its prevalence along with comorbidities are on the rise around the world. Ingestion of fish becomes trendy in daily meals. Recent research has shown that marine fish oil (FO) (found in tuna, sardines, and mackerel) may offer an alternative method for reducing obesity and problems associated with it. Marine FO rich in long-chain omega-3 polyunsaturated fatty acids (LC n-3 PUFA) and long-chain omega-6 polyunsaturated fatty acids (LC n-6 PUFA) plays an important role in reducing abnormalities associated with the metabolic syndrome and has a variety of disease-fighting properties, including cardioprotective activity, anti-atherosclerotic, anti-obesity, anti-cancer, anti-inflammatory activity. Studies in rodents and humans have indicated that LC n-3 PUFA potentially elicit a number of effects which might be useful for reducing obesity, including suppression of appetite, improvements in circulation, enhanced fat oxidation, energy expenditure, and reduced fat deposition. This review discusses the interplay between inflammation and obesity, and their subsequent regulation via the beneficial role of marine FO, suggesting an alternative dietary strategy to ameliorate obesity and obesity-associated chronic diseases.


Assuntos
Óleos de Peixe , Obesidade , Humanos , Animais , Óleos de Peixe/uso terapêutico , Óleos de Peixe/administração & dosagem , Óleos de Peixe/farmacologia , Obesidade/metabolismo , Obesidade/tratamento farmacológico , Ácidos Graxos Ômega-3/uso terapêutico , Doenças Metabólicas/tratamento farmacológico , Doenças Metabólicas/metabolismo , Doenças Metabólicas/prevenção & controle
7.
FASEB J ; 38(20): e70097, 2024 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-39394863

RESUMO

Secondary lymphedema (LE) following breast cancer-related surgery is a life-long complication, which currently has no cure. LE induces significant regional adipose tissue deposition, requiring liposuction as a treatment. Here, we aimed to elucidate the transcriptional, metabolomic, and lipidomic signature of the adipose tissue developed due to the surgery-induced LE in short- and long-term LE patients and compared the transcriptomic landscape of LE adipose tissue to the obesity-induced adipose tissue. Adipose tissue biopsies were obtained from breast cancer-operated females with LE from the affected and non-affected arms (n = 20 patients). To decipher the molecular properties of the LE adipose tissue, we performed RNA sequencing, metabolomics, and lipidomics combined with bioinformatics analyses. Differential gene expression data from a cohort of lean and obese patients without LE was used for comparisons. Integrative analysis of functional genomics revealed that inflammatory response, cell chemotaxis, and angiogenesis were upregulated biological processes in the LE arm, indicating a sustained inflammation in the edematous adipose tissue; whereas, epidermal differentiation, cell-cell junction organization, water homeostasis, and neurogenesis were downregulated in the LE arm. Surprisingly, only a few genes were found to be the same in the LE-induced and the obesity-induced adipose tissue expansion, indicating a different type of adipose tissue development in these two conditions. In metabolomics analysis, we found reduced levels of a branched-chain amino acid valine in the LE arm and downregulation of the mRNA levels of its transporter SLC6A15. Lipidomics analyses did not show any significant differences between the LE and non-LE arms, suggesting that other factors affect the lipid composition of the adipose tissue more than the LE in these patients. Our results provide a detailed molecular characterization of adipose tissue in secondary LE after breast cancer-related surgery. We also show distinct differences in transcriptomic signatures between LE-induced adipose tissue and obesity-induced adipose tissue, but only minor differences in metabolome and lipidome between the LE and the non-LE arm.


Assuntos
Tecido Adiposo , Neoplasias da Mama , Linfedema , Humanos , Feminino , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Neoplasias da Mama/cirurgia , Tecido Adiposo/metabolismo , Pessoa de Meia-Idade , Linfedema/metabolismo , Linfedema/etiologia , Linfedema/genética , Linfedema/patologia , Obesidade/metabolismo , Transcriptoma , Idoso , Adulto , Metabolômica , Lipidômica , Multiômica
8.
Vopr Pitan ; 93(4): 39-48, 2024.
Artigo em Russo | MEDLINE | ID: mdl-39396214

RESUMO

Over the past 50 years, the prevalence of obesity around the world has increased several times and has become a pandemic. The effect of obesity on the lymphatic system, which plays a key role in the regulation of fluid homeostasis, immune cell migration, antigen presentation, and resolution of inflammatory responses, is poorly understood, and there is no data on the contractile activity of the lymph nodes in obesity. The purpose of the research was to investigate the parameters and mechanisms of dysfunction of the contractile function of the mesenteric lymph nodes of rats in obesity caused by the feeding with the high-fat diet (HFD). Material and methods. The study was conducted on 50 male Sprague-Dawley rats. Rats aged 6 weeks were randomly divided into groups: a control group (n=10) fed a standard diet and a group of rats (n=40) kept on HFD (60% fat content by calorie value). Rats received food and water ad libitum for 16 weeks. Before the end of the experiment, four groups of HFD rats were formed: obesity resistant animals (HFD-OR, n=11), without additional interventions (HFD, n=10), rats which were administered dexamethasone three days before the study (HFD+Dexa, n=9), HFD followed by 8-week diet restriction (HFD+DR, n=9). At the end of the experiment, mesenteric lymph nodes (LNs) were taken from rats under anesthesia and their contractile function was studied in a myograph using 1400W, dynastat and Tempol. Results. LNs of control rats had a high level of tone and generated spontaneous high-amplitude phasic contractions. The LNs of HFD rats had a low initial tone, and rare low-amplitude phasic contractions were recorded in them. The parameters of contractile activity of the LNs of rats in HFD-OR and HFD+Dexa groups differed slightly from the corresponding parameters of the LNs of rats in the control group. Calorie restriction for 8 weeks in obese rats (HFD+DR) resulted in an increase in tone, frequency and amplitude of phasic contractions of the LNs compared to those in HFD rats. iNOS inhibition caused a significant increase in the tone, amplitude and frequency of phasic contractions of the LNs in the HFD group. An increase in the frequency of phasic contractions was observed only in the LNs of HFD+Dex and HFD+DR rats. Inhibition of cyclooxygenase 2 did not affect the contractile function of the LNs of rats of all groups, with the exception of animals from the HFD group (increase in the amplitude and frequency of phasic contractions). Tempol significantly increased the tone, frequency and amplitude of phasic contractions of the LNs in rats of the HFD group and increased the frequency of phasic contractions of the LNs of the HFD+DR rats. Conclusion. A high-fat diet leads to impaired contractile function of rat LNs and can create additional obstacles to the movement of lymph, promoting its leakage into surrounding tissues. Obesity is accompanied by the development of inflammation in the LNs and perinodal adipose tissue, which induces the expression of inducible NO synthase, cyclooxygenase-2 and the accumulation of reactive oxygen species (ROS). NO, prostaglandins and ROS have an inhibitory effect on the SMC capsules of the LNs, leading to a decrease in tonic tension and a weakening of spontaneous phasic contractions. The reason for inhibition of LN contractile function is obesity, but not consumption of food high in fat. Transferring obese rats to a calorie-restricted diet results in a decrease in body weight and visceral fat mass and an improvement in LN contractile function.


Assuntos
Linfonodos , Obesidade , Ratos Sprague-Dawley , Animais , Masculino , Ratos , Obesidade/metabolismo , Obesidade/fisiopatologia , Obesidade/patologia , Linfonodos/patologia , Contração Muscular/efeitos dos fármacos , Mesentério , Dieta Hiperlipídica/efeitos adversos , Óxidos N-Cíclicos/farmacologia , Marcadores de Spin
9.
Commun Biol ; 7(1): 1300, 2024 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-39390075

RESUMO

The crosstalk between adipose tissue and the liver is finely controlled to maintain metabolic health. Yet, how adipose tissue controls toxic free fatty acid overflow into the liver remains incompletely understood. Here, we show that adipocyte activating transcription factor 3 (ATF3) was induced in human or mouse obesity. Adipocyte Atf3-/- (Atf3Adi-/-) mice developed obesity, glucose intolerance, and metabolic dysfunction-associated steatohepatitis (MASH) in chow diet, high-fat diet, or Western diet-fed mice. Blocking fatty acid flux by inhibiting hepatocyte CD36, but not the restoration of hepatic AMPK signaling, prevented the aggravation of MASH in Atf3Adi-/- mice. Further studies show that the loss of adipocyte ATF3 increased lipolysis via inducing adipose triglyceride lipase, which in turn induced lipogenesis and inflammation in hepatocytes. Moreover, Atf3Adi-/- mice had reduced energy expenditure and increased adipose lipogenesis and inflammation. Our data demonstrate that adipocyte ATF3 is a gatekeeper in counteracting MASH development under physiological and pathological conditions.


Assuntos
Fator 3 Ativador da Transcrição , Tecido Adiposo , Lipólise , Camundongos Knockout , Animais , Fator 3 Ativador da Transcrição/metabolismo , Fator 3 Ativador da Transcrição/genética , Camundongos , Tecido Adiposo/metabolismo , Humanos , Masculino , Fígado Gorduroso/metabolismo , Fígado Gorduroso/genética , Fígado Gorduroso/patologia , Obesidade/metabolismo , Obesidade/genética , Camundongos Endogâmicos C57BL , Adipócitos/metabolismo , Dieta Hiperlipídica/efeitos adversos , Lipogênese/genética , Metabolismo Energético , Feminino
10.
Mol Med ; 30(1): 169, 2024 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-39390356

RESUMO

External constraints, such as development, disease, and environment, can induce changes in epigenomic patterns that may profoundly impact the health trajectory of fetuses and neonates into adulthood, influencing conditions like obesity. Epigenetic modifications encompass processes including DNA methylation, covalent histone modifications, and RNA-mediated regulation. Beyond forward cellular differentiation (cell programming), terminally differentiated cells are reverted to a pluripotent or even totipotent state, that is, cellular reprogramming. Epigenetic modulators facilitate or erase histone and DNA modifications both in vivo and in vitro during programming and reprogramming. Noticeably, obesity is a complex metabolic disorder driven by both genetic and environmental factors. Increasing evidence suggests that epigenetic modifications play a critical role in the regulation of gene expression involved in adipogenesis, energy homeostasis, and metabolic pathways. Hence, we discuss the mechanisms by which epigenetic interventions influence obesity, focusing on DNA methylation, histone modifications, and non-coding RNAs. We also analyze the methodologies that have been pivotal in uncovering these epigenetic regulations, i.e., Large-scale screening has been instrumental in identifying genes and pathways susceptible to epigenetic control, particularly in the context of adipogenesis and metabolic homeostasis; Single-cell RNA sequencing (scRNA-seq) provides a high-resolution view of gene expression patterns at the individual cell level, revealing the heterogeneity and dynamics of epigenetic regulation during cellular differentiation and reprogramming; Chromatin immunoprecipitation (ChIP) assays, focused on candidate genes, have been crucial for characterizing histone modifications and transcription factor binding at specific genomic loci, thereby elucidating the epigenetic mechanisms that govern cellular programming; Somatic cell nuclear transfer (SCNT) and cell fusion techniques have been employed to study the epigenetic reprogramming accompanying cloning and the generation of hybrid cells with pluripotent characteristics, etc. These approaches have been instrumental in identifying specific epigenetic marks and pathways implicated in obesity, providing a foundation for developing targeted therapeutic interventions. Understanding the dynamic interplay between epigenetic regulation and cellular programming is crucial for advancing mechanism and clinical management of obesity.


Assuntos
Reprogramação Celular , Metilação de DNA , Epigênese Genética , Obesidade , Humanos , Obesidade/genética , Obesidade/metabolismo , Animais , Reprogramação Celular/genética , Diferenciação Celular/genética , Adipogenia/genética , Histonas/metabolismo , Código das Histonas , Epigenômica/métodos
11.
J Biosci ; 492024.
Artigo em Inglês | MEDLINE | ID: mdl-39377444

RESUMO

The regulation of adipose tissue metabolism by irisin involves modulating gene expressions related to energy metabolism and insulin sensitivity via miRNA-mediated signaling pathways within adipose tissue. Understanding the molecular mechanisms behind the role of irisin is vital for addressing obesity and related metabolic complications. In this study, we undertook an extensive miRNA transcriptomic approach to identify differentially expressed miRNAs following irisin exposure in adipocytes and murine white adipose tissue. Our findings spotlighted two miRNAs, miRNA-758 and miRNA-668, as being influenced by irisin. To understand the impact of the modulations of these miRNAs by irisin, we performed a signaling pathway and network analysis. After irisin exposure, both, miRNA-758 and miRNA-668, emerged as key regulators in leptin and CDK5 signaling pathways. Leptin, a hormone originating from adipose tissue, is primarily produced by adipocytes, and its effects are known to be mediated by CDK5. In essence, this study identifies pivotal genes and miRNAs in irisin-driven mechanisms in adipose tissue, offering valuable insights for crafting novel therapeutic strategies for metabolic and associated disorders.


Assuntos
Adipócitos , Quinase 5 Dependente de Ciclina , Fibronectinas , MicroRNAs , Transdução de Sinais , MicroRNAs/genética , MicroRNAs/metabolismo , Fibronectinas/metabolismo , Fibronectinas/genética , Animais , Camundongos , Adipócitos/metabolismo , Quinase 5 Dependente de Ciclina/metabolismo , Quinase 5 Dependente de Ciclina/genética , Leptina/metabolismo , Leptina/genética , Regulação da Expressão Gênica , Tecido Adiposo/metabolismo , Tecido Adiposo Branco/metabolismo , Células 3T3-L1 , Humanos , Perfilação da Expressão Gênica , Metabolismo Energético/genética , Obesidade/metabolismo , Obesidade/genética
12.
Compr Physiol ; 14(4): 5581-5640, 2024 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-39382163

RESUMO

For a long time, white adipocytes were thought to function as lipid storages due to the sizeable unilocular lipid droplet that occupies most of their space. However, recent discoveries have highlighted the critical role of white adipocytes in maintaining energy homeostasis and contributing to obesity and related metabolic diseases. These physiological and pathological functions depend heavily on the mitochondria that reside in white adipocytes. This article aims to provide an up-to-date overview of the recent research on the function and dysfunction of white adipocyte mitochondria. After briefly summarizing the fundamental aspects of mitochondrial biology, the article describes the protective role of functional mitochondria in white adipocyte and white adipose tissue health and various roles of dysfunctional mitochondria in unhealthy white adipocytes and obesity. Finally, the article emphasizes the importance of enhancing mitochondrial quantity and quality as a therapeutic avenue to correct mitochondrial dysfunction, promote white adipocyte browning, and ultimately improve obesity and its associated metabolic diseases. © 2024 American Physiological Society. Compr Physiol 14:5581-5640, 2024.


Assuntos
Adipócitos Brancos , Mitocôndrias , Humanos , Mitocôndrias/metabolismo , Mitocôndrias/fisiologia , Adipócitos Brancos/metabolismo , Adipócitos Brancos/fisiologia , Animais , Obesidade/metabolismo , Obesidade/fisiopatologia , Tecido Adiposo Branco/metabolismo , Metabolismo Energético/fisiologia
13.
Front Endocrinol (Lausanne) ; 15: 1325286, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39381436

RESUMO

Introduction: Reducing Optic Atrophy 1 (OPA1) expression in skeletal muscle in male mice induces Activation Transcription Factor 4 (ATF4) and the integrated stress response (ISR). Additionally, skeletal muscle secretion of Fibroblast Growth Factor 21 (FGF21) is increased, which mediates metabolic adaptations including resistance to diet-induced obesity (DIO) and glucose intolerance in these mice. Although FGF21 induction in this model can be reversed with pharmacological attenuation of ER stress, it remains to be determined if ATF4 is responsible for FGF21 induction and its metabolic benefits in this model. Methods: We generated mice with homozygous floxed Opa1 and Atf4 alleles and a tamoxifen-inducible Cre transgene controlled by the human skeletal actin promoter to enable simultaneous depletion of OPA1 and ATF4 in skeletal muscle (mAO DKO). Mice were fed high fat (HFD) or control diet and evaluated for ISR activation, body mass, fat mass, glucose tolerance, insulin tolerance and circulating concentrations of FGF21 and growth differentiation factor 15 (GDF15). Results: In mAO DKO mice, ATF4 induction is absent. Other indices of ISR activation, including XBP1s, ATF6, and CHOP were induced in mAO DKO males, but not in mOPA1 or mAO DKO females. Resistance to diet-induced obesity was not reversed in mAO DKO mice of both sexes. Circulating FGF21 and GDF15 illustrated sexually dimorphic patterns. Loss of OPA1 in skeletal muscle increases circulating FGF21 in mOPA1 males, but not in mOPA1 females. Additional loss of ATF4 decreased circulating FGF21 in mAO DKO male mice, but increased circulating FGF21 in female mAO DKO mice. Conversely, circulating GDF15 was increased in mAO DKO males and mOPA1 females, but not in mAO DKO females. Conclusion: Sex differences exist in the transcriptional outputs of the ISR following OPA deletion in skeletal muscle. Deletion of ATF4 in male and female OPA1 KO mice does not reverse the resistance to DIO. Induction of circulating FGF21 is ATF4 dependent in males, whereas induction of circulating GDF15 is ATF4 dependent in females. Elevated GDF15 in males and FGF21 in females could reflect activation by other transcriptional outputs of the ISR, that maintain mitokine-dependent metabolic protection in an ATF4-independent manner.


Assuntos
Fator 4 Ativador da Transcrição , Fatores de Crescimento de Fibroblastos , GTP Fosfo-Hidrolases , Camundongos Knockout , Músculo Esquelético , Caracteres Sexuais , Animais , Fator 4 Ativador da Transcrição/metabolismo , Fator 4 Ativador da Transcrição/genética , Camundongos , Masculino , Músculo Esquelético/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Fatores de Crescimento de Fibroblastos/genética , Feminino , GTP Fosfo-Hidrolases/metabolismo , GTP Fosfo-Hidrolases/genética , Fator 15 de Diferenciação de Crescimento/metabolismo , Fator 15 de Diferenciação de Crescimento/genética , Obesidade/metabolismo , Obesidade/genética , Dieta Hiperlipídica , Camundongos Endogâmicos C57BL
14.
Sci Signal ; 17(856): eadt4125, 2024 10.
Artigo em Inglês | MEDLINE | ID: mdl-39353039

RESUMO

Obesity exacerbates inflammation to a greater extent in female patients and mice with multiple sclerosis.


Assuntos
Autoimunidade , Esclerose Múltipla , Obesidade , Animais , Feminino , Humanos , Obesidade/imunologia , Obesidade/metabolismo , Camundongos , Esclerose Múltipla/imunologia , Masculino , Inflamação/imunologia , Inflamação/metabolismo , Fatores Sexuais , Caracteres Sexuais
15.
Med Sci Monit ; 30: e946675, 2024 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-39350608

RESUMO

Rising levels of obesity in all age groups are associated with profound effects on health and economies in developed and developing countries. This year, the scientific research behind the development of glucagon-like peptide 1 (GLP-1) receptor agonists (GLP-1 analogs or incretin mimetics) has been recognized. On 19 September 2024, three scientists were presented with a Lasker Award for their basic clinical research on identifying and studying the roles of GLP-1. The research by Joel Habener, Svetlana Mojsov, and Lotte Bjerre Knudsen began three decades ago and has led to new anti-obesity drugs, which mimic GLP-1 to lower blood glucose levels and control appetite. The efficacy of GLP-1 receptor agonists in the management of obesity in adults, as well as in children and adolescents, has now been supported by several clinical trials. This editorial aims to describe the research behind developing GLP-1 receptor agonists and their potential and pitfalls in managing obesity in all age groups.


Assuntos
Peptídeo 1 Semelhante ao Glucagon , Receptor do Peptídeo Semelhante ao Glucagon 1 , Obesidade , Humanos , Obesidade/tratamento farmacológico , Obesidade/metabolismo , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Receptor do Peptídeo Semelhante ao Glucagon 1/agonistas , Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Fármacos Antiobesidade/uso terapêutico , Fármacos Antiobesidade/farmacologia , Incretinas/uso terapêutico , Incretinas/metabolismo , Glicemia/metabolismo , Glicemia/efeitos dos fármacos
16.
FASEB J ; 38(19): e70093, 2024 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-39373976

RESUMO

The risk of developing type 2 diabetes (T2D) is heterogeneous among individuals with obesity. Functional decline of adipocyte precursor cells (APCs) and accumulation of senescent cells in the subcutaneous adipose tissue contributes to the progression toward T2D. LncRNAs regulate cell senescence and may be implicated in determining this abnormality in APCs. Here, we report that APCs from individuals with obesity show a gradual increase in multiple senescence markers, which worsens in parallel with the progression from normal glucose tolerance (NGT) to impaired glucose tolerance (IGT) or T2D. Transcriptomic analysis identified PANDAR as the top-ranked lncRNA differentially expressed in APCs from individuals with obesity and T2D and non-obese subjects. Q-PCR confirmed PANDAR up-regulation in APCs from individuals with obesity, at progressively increased levels in those who developed, respectively, IGT and T2D. Bisulfite sequencing and luciferase assays revealed that, in parallel with glucose tolerance deterioration, the -1317 CpG at the PANDAR promoter became hypo-methylated in obesity, resulting in enhanced PANDAR induction by p53. PANDAR silencing in senescent APCs from individuals with obesity and T2D caused repression of senescence programs and cell cycle re-entry. PANDAR transcription in white blood cells (WBCs) mirrored that in APCs. Also, individuals with obesity exhibited rescue of PANDAR transcription in WBCs following bariatric surgery, accompanied by enhanced methylation at the regulatory PANDAR -1317 CpG. In conclusion, PANDAR dysregulation is a newly identified mechanism determining the early senescence of APCs from individuals with obesity, which worsens along the progression toward T2D. In the future, PANDAR targeting may represent a valuable strategy to delay this progression.


Assuntos
Adipócitos , Senescência Celular , Metilação de DNA , Diabetes Mellitus Tipo 2 , Obesidade , Regiões Promotoras Genéticas , RNA Longo não Codificante , Adulto , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Adipócitos/metabolismo , Senescência Celular/genética , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Obesidade/genética , Obesidade/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo
17.
PLoS One ; 19(10): e0311502, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39374222

RESUMO

BACKGROUND: Penthorum chinense Pursh (PCP), a medicinal and edible plant, has been reported to protect against liver damage by suppressing oxidative stress. Type 2 diabetes mellitus (T2DM) is associated with liver dysfunction and oxidative stress. In the present study, we aim to investigate the hypoglycemic effect of PCP on db/db mice and further explore the underlying mechanisms. METHODS: Thirty-two db/db mice were randomized into four groups, including a diabetic model control group (MC) and three diabetic groups treated with low (LPCP, 300 mg/kg/d), medium (MPLP, 600 mg/kg/d), and high doses of PCP (HPCP, 1200 mg/kg/d), and the normal control group (NC) of eight db/m mice were included. Mice in the NC and MC groups received the ultrapure water. After four weeks of intervention, parameters of fasting blood glucose (FBG), insulin resistance (IR), blood lipid levels, hepatic oxidative stress, and enzymes related to hepatic glucose metabolism were compared in the groups. RESULTS: PCP administration significantly reduced FBG and IR in diabetic db/db mice, and improved hepatic glucose metabolism by increasing glucose transporter 2 (GLUT2) and glucokinase (GCK) protein expression. Meanwhile, PCP supplementation ameliorated hepatic oxidative stress by decreasing malonaldehyde content and increasing the activities of superoxide dismutase and glutathione peroxidase in db/db mice. Furthermore, PCP treatment reduced obesity and food intake in db/db mice, and improved dyslipidemia demonstrated by increasing high-density lipoprotein cholesterol (HDL-C) while decreasing total cholesterol (TC), triglyceride (TG), and low-density lipoprotein cholesterol (HDL-C). All doses of PCP treatment decreased the values of LDL-C/HDL-C in a dose-response relationship. CONCLUSION: PCP significantly alleviated hyperglycemia, hyperinsulinemia, hyperlipidemia, and obesity, inhibited hepatic oxidative stress, and enhanced hepatic glucose transport in T2DM mice. Based on the above findings, the hypoglycemic effect of PCP may be attributed to the activation of the GLUT2/GCK expression in the liver and the reduction of hepatic oxidative stress.


Assuntos
Glicemia , Diabetes Mellitus Tipo 2 , Resistência à Insulina , Obesidade , Estresse Oxidativo , Extratos Vegetais , Animais , Estresse Oxidativo/efeitos dos fármacos , Camundongos , Extratos Vegetais/farmacologia , Extratos Vegetais/uso terapêutico , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Glicemia/metabolismo , Obesidade/tratamento farmacológico , Obesidade/metabolismo , Masculino , Fígado/metabolismo , Fígado/efeitos dos fármacos , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/metabolismo , Hipoglicemiantes/farmacologia , Hipoglicemiantes/uso terapêutico , Transportador de Glucose Tipo 2/metabolismo
18.
Front Endocrinol (Lausanne) ; 15: 1458848, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39351529

RESUMO

Obesity is increasing globally and is closely associated with a range of metabolic disorders, including metabolic associated fatty liver disease, diabetes, and cardiovascular diseases. An effective strategy to combat obesity involves stimulating brown and beige adipocyte thermogenesis, which significantly enhances energy expenditure. Recent research has underscored the vital role of PRDM16 in the development and functionality of thermogenic adipocytes. Consequently, PRDM16 has been identified as a potential therapeutic target for obesity and its related metabolic disorders. This review comprehensively examines various studies that focus on combating obesity by directly targeting PRDM16 in adipose tissue.


Assuntos
Tecido Adiposo , Proteínas de Ligação a DNA , Doenças Metabólicas , Obesidade , Termogênese , Fatores de Transcrição , Animais , Humanos , Tecido Adiposo/metabolismo , Tecido Adiposo Marrom/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/genética , Metabolismo Energético , Doenças Metabólicas/metabolismo , Obesidade/metabolismo , Fatores de Transcrição/metabolismo
19.
Nat Commun ; 15(1): 8588, 2024 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-39362888

RESUMO

Excessive glucocorticoid (GC) action is linked to various metabolic disorders. Recent findings suggest that disrupting skeletal GC signaling prevents bone loss and alleviates metabolic disorders in high-fat diet (HFD)-fed obese mice, underpinning the neglected contribution of skeletal GC action to obesity and related bone loss. Here, we show that the elevated expression of 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1), the enzyme driving local GC activation, and GC signaling in osteoblasts, are associated with bone loss and obesity in HFD-fed male mice. Osteoblast-specific 11ß-HSD1 knockout male mice exhibit resistance to HFD-induced bone loss and metabolic disorders. Mechanistically, elevated 11ß-HSD1 restrains glucose uptake and osteogenic activity in osteoblast. Pharmacologically inhibiting osteoblastic 11ß-HSD1 by using bone-targeted 11ß-HSD1 inhibitor markedly promotes bone formation, ameliorates glucose handling and mitigated obesity in HFD-fed male mice. Taken together, our study demonstrates that osteoblastic 11ß-HSD1 directly contributes to HFD-induced bone loss, glucose handling impairment and obesity.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 1 , Dieta Hiperlipídica , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade , Osteoblastos , Animais , Humanos , Masculino , Camundongos , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/metabolismo , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/genética , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/antagonistas & inibidores , Reabsorção Óssea/metabolismo , Reabsorção Óssea/prevenção & controle , Dieta Hiperlipídica/efeitos adversos , Glucocorticoides/metabolismo , Glucose/metabolismo , Obesidade/metabolismo , Obesidade/etiologia , Obesidade/genética , Osteoblastos/metabolismo , Osteoblastos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Transdução de Sinais
20.
Clin Transl Med ; 14(10): e70029, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39350476

RESUMO

INTRODUCTION: Temporomandibular joint osteoarthritis (TMJ OA) is a major disease that affects maxillofacial health and is characterised by cartilage degeneration and subchondral bone remodelling. Obesity is associated with the exacerbation of pathological manifestations of TMJ OA. However, the underlying mechanism between adipose tissue and the TMJ axis remains limited. OBJECTIVES: To evaluate the effects of obesity and the adipose tissue on the development of TMJ OA. METHODS: The obesity-related metabolic changes in TMJ OA patients were detected by physical signs and plasma metabolites. The effects of adipose tissue-derived EVs (Ad-EVs) on TMJ OA was investigated through histological and cytological experiments as well as gene editing technology. Alterations of Ad-EVs in obese state were identified by microRNA-seq analysis and the mechanism by which EVs affect TMJ OA was explored in vitro and in vivo. RESULTS: Obesity and the related metabolic changes were important influencing factors for TMJ OA. Ad-EVs from obese mice induced marked chondrocyte apoptosis, cartilage matrix degradation and subchondral bone remodelling, which exacerbated the development of TMJ OA. Depletion of Ad-EVs secretion by knocking out the geranylgeranyl diphosphate synthase (Ggpps) gene in adipose tissue significantly inhibited the obesity-induced aggravation of TMJ OA. MiR-3074-5p played an important role in this process . CONCLUSIONS: Our work unveils an unknown link between obese adipose tissue and TMJ OA. Targeting the Ad-EVs and the miR-3074-5p may represent a promising therapeutic strategy for obesity-related TMJ OA. KEY POINTS: High-fat-diet-induced obesity aggravate the progression of TMJ OA in mice. Obese adipose tissue participates in cartilage damage through the altered miRNA in extracellular vesicles. Inhibition of miR-3074-5p/SMAD4 pathway in chondrocyte alleviated the effect of HFD-EVs on TMJ OA.


Assuntos
Tecido Adiposo , Vesículas Extracelulares , Obesidade , Osteoartrite , Vesículas Extracelulares/metabolismo , Animais , Osteoartrite/metabolismo , Osteoartrite/etiologia , Obesidade/metabolismo , Obesidade/complicações , Camundongos , Tecido Adiposo/metabolismo , Humanos , Masculino , Feminino , Articulação Temporomandibular/metabolismo , Articulação Temporomandibular/patologia , Transtornos da Articulação Temporomandibular/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA