Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 995
Filtrar
1.
Epigenetics ; 19(1): 2404198, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-39292753

RESUMO

Maternal hyperglycemia during pregnancy adversely affects maternal and child outcomes. While mechanisms are not fully understood, maternal circulating miRNAs may play a role. We examined whether continuous glucose levels and hyperglycemia subtypes (gestational diabetes, type 2 diabetes, and glucose intolerance) were associated with circulating miRNAs during late pregnancy. Seven miRNAs (hsa-miR-107, hsa-let-7b-5p, hsa-miR-126-3p, hsa-miR-181a-5p, hsa-miR-374a-5p, hsa-miR-382-5p, and hsa-miR-337-5p) were associated (p < 0.05) with either hyperglycemia or continuous glucose levels prior to multiple testing correction. These miRNAs target genes involved in pathways relevant to maternal and child health, including insulin signaling, placental development, energy balance, and appetite regulation.


Assuntos
Diabetes Gestacional , Vesículas Extracelulares , Humanos , Feminino , Gravidez , Adulto , Vesículas Extracelulares/genética , Vesículas Extracelulares/metabolismo , Diabetes Gestacional/genética , Diabetes Gestacional/sangue , Glicemia/metabolismo , MicroRNAs/genética , MicroRNAs/sangue , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/sangue , Hiperglicemia/genética , Hiperglicemia/sangue , MicroRNA Circulante/genética , MicroRNA Circulante/sangue , Intolerância à Glucose/genética , Estudos de Coortes
2.
Diabetes Obes Metab ; 26(10): 4591-4601, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39113250

RESUMO

AIM: To investigate the effect of G protein-coupled receptor 55 (GPR55) deletion on glucose homeostasis and islet function following diet-induced obesity. METHODS: GPR55-/- and wild-type (WT) mice were fed ad libitum either standard chow (SC) or a high-fat diet (HFD) for 20 weeks. Glucose and insulin tolerance tests were performed at 9/10 and 19/20 weeks of dietary intervention. Insulin secretion in vivo and dynamic insulin secretion following perifusion of isolated islets were also determined, as were islet caspase-3/7 activities and ß-cell 5-bromo-20-deoxyuridine (BrdU) incorporation. RESULTS: GPR55-/- mice fed a HFD were more susceptible to diet-induced obesity and were more glucose intolerant and insulin resistant than WT mice maintained on a HFD. Islets isolated from HFD-fed GPR55-/- mice showed impaired glucose- and pcacahorbol 12-myristate 13-acetate-stimulated insulin secretion, and they also displayed increased cytokine-induced apoptosis. While there was a 5.6 ± 1.6-fold increase in ß-cell BrdU incorporation in the pancreases of WT mice fed a HFD, this compensatory increase in ß-cell proliferation in response to the HFD was attenuated in GPR55-/- mice. CONCLUSIONS: Under conditions of diet-induced obesity, GPR55-/- mice show impaired glucose handling, which is associated with reduced insulin secretory capacity, increased islet cell apoptosis and insufficient compensatory increases in ß-cell proliferation. These observations support that GPR55 plays an important role in positively regulating islet function.


Assuntos
Dieta Hiperlipídica , Homeostase , Secreção de Insulina , Células Secretoras de Insulina , Insulina , Camundongos Knockout , Obesidade , Receptores de Canabinoides , Animais , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Obesidade/metabolismo , Obesidade/genética , Camundongos , Dieta Hiperlipídica/efeitos adversos , Receptores de Canabinoides/metabolismo , Receptores de Canabinoides/genética , Resistência à Insulina/genética , Masculino , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Apoptose , Camundongos Endogâmicos C57BL , Teste de Tolerância a Glucose , Glicemia/metabolismo , Intolerância à Glucose/genética , Intolerância à Glucose/metabolismo
3.
Endocrinology ; 165(9)2024 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-39045670

RESUMO

Classic hereditary hemochromatosis (HH) is an autosomal recessive iron-overload disorder resulting from loss-of-function mutations of the HFE gene. Patients with HH exhibit excessive hepatic iron accumulation that predisposes these patients to liver disease, including the risk for developing liver cancer. Chronic iron overload also poses a risk for the development of metabolic disorders such as obesity, type 2 diabetes, and insulin resistance. We hypothesized that liraglutide, GLP1 receptor agonist, alters iron metabolism while also reducing body weight and glucose tolerance in a mouse model of HH (global HFE knockout, HFE KO) and diet-induced obesity and glucose intolerance. The total body HFE KO and wild-type control mice were fed high-fat diet for 8 weeks. Mice were subdivided into liraglutide and vehicle-treated groups and received daily subcutaneous administration of the respective treatment once daily for 18 weeks. Liraglutide improved glucose tolerance and hepatic lipid markers and reduced body weight in a mouse model of HH, the HFE KO mouse, similar to wild-type controls. Importantly, our data show that liraglutide alters iron metabolism in HFE KO mice, leading to decreased circulating and stored iron levels in HFE KO mice. These observations highlight the potential that GLP1 receptor agonist could be used to reduce iron overload in addition to reducing body weight and improving glucose regulation in HH patients.


Assuntos
Modelos Animais de Doenças , Proteína da Hemocromatose , Hemocromatose , Homeostase , Ferro , Liraglutida , Camundongos Knockout , Animais , Hemocromatose/genética , Hemocromatose/metabolismo , Hemocromatose/tratamento farmacológico , Liraglutida/farmacologia , Liraglutida/uso terapêutico , Ferro/metabolismo , Homeostase/efeitos dos fármacos , Camundongos , Proteína da Hemocromatose/genética , Proteína da Hemocromatose/metabolismo , Fígado/metabolismo , Fígado/efeitos dos fármacos , Masculino , Dieta Hiperlipídica/efeitos adversos , Intolerância à Glucose/metabolismo , Intolerância à Glucose/tratamento farmacológico , Intolerância à Glucose/genética , Obesidade/metabolismo , Obesidade/tratamento farmacológico , Obesidade/genética , Camundongos Endogâmicos C57BL , Peso Corporal/efeitos dos fármacos
4.
Elife ; 122024 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-39056292

RESUMO

From a forward mutagenetic screen to discover mutations associated with obesity, we identified mutations in the Spag7 gene linked to metabolic dysfunction in mice. Here, we show that SPAG7 KO mice are born smaller and develop obesity and glucose intolerance in adulthood. This obesity does not stem from hyperphagia, but a decrease in energy expenditure. The KO animals also display reduced exercise tolerance and muscle function due to impaired mitochondrial function. Furthermore, SPAG7-deficiency in developing embryos leads to intrauterine growth restriction, brought on by placental insufficiency, likely due to abnormal development of the placental junctional zone. This insufficiency leads to loss of SPAG7-deficient fetuses in utero and reduced birth weights of those that survive. We hypothesize that a 'thrifty phenotype' is ingrained in SPAG7 KO animals during development that leads to adult obesity. Collectively, these results indicate that SPAG7 is essential for embryonic development and energy homeostasis later in life.


Obesity rates are climbing worldwide, leading to an increase in associated conditions such as type 2 diabetes. While new pharmaceutical approaches are available to help individuals manage their weight, many patients do not respond to them or experience prohibitive side effects. Identifying alternative treatments will likely require pinpointing the genes and molecular actors involved in the biological processes that control weight regulation. Previous research suggests that a protein known as SPAG7 could help shape how mice use and store the energy they extract from food. Flaherty et al. therefore set out to investigate the role this protein plays in the body. To do so, they created a line of mice born without SPAG7, which they monitored closely throughout life. These animals were underweight at birth and did not eat more than other mice, yet they were obese as adults. Their ability to exercise was reduced, their muscles were weaker and contained fibers with functional defects. The mice also exhibited biological changes associated with the onset of diabetes. Yet deleting SPAG7 during adulthood led to no such changes; these mice maintained normal muscle function and body weight. Closely examining how SPAG7-deficient mice developed in the womb revealed placental defects which likely caused these animals to receive fewer nutrients from their mother. Such early-life deprivation is known to be associated with the body shifting towards maximizing its use of resources and privileging fat storage, even into and throughout adulthood. By shedding light on the biological role of SPAG7, the work by Flaherty et al. helps to better understand how developmental events can increase the likelihood of obesity later in life. Further investigations are now needed to explore whether this knowledge could help design interventions relevant to human health.


Assuntos
Retardo do Crescimento Fetal , Camundongos Knockout , Obesidade , Animais , Obesidade/genética , Obesidade/metabolismo , Retardo do Crescimento Fetal/genética , Camundongos , Feminino , Metabolismo Energético/genética , Deleção de Genes , Gravidez , Intolerância à Glucose/genética
5.
J Proteome Res ; 23(8): 3332-3341, 2024 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-38967328

RESUMO

The prevalence of different metabolic syndromes has grown globally, and the farnesoid X receptor (FXR), a metabolic homeostat for glucose, lipid, and bile acid metabolisms, may serve an important role in the progression of metabolic disorders. Glucose intolerance by FXR deficiency was previously reported and observed in our study, but the underlying biology remained unclear. To investigate the ambiguity, we collected the nontargeted profiles of the fecal metaproteome, serum metabolome, and liver proteome in Fxr-null (Fxr-/-) and wild-type (WT) mice with LC-HRMS. FXR deficiency showed a global impact on the different molecular levels we monitored, suggesting its serious disruption in the gut microbiota, hepatic metabolism, and circulating biomolecules. The network and enrichment analyses of the dysregulated metabolites and proteins suggested the perturbation of carbohydrate and lipid metabolism by FXR deficiency. Fxr-/- mice presented lower levels of hepatic proteins involved in glycogenesis. The impairment of glycogenesis by an FXR deficiency may leave glucose to accumulate in the circulation, which may deteriorate glucose tolerance. Lipid metabolism was dysregulated by FXR deficiency in a structural-dependent manner. Fatty acid ß-oxidations were alleviated, but cholesterol metabolism was promoted by an FXR deficiency. Together, we explored the molecular events associated with glucose intolerance by impaired FXR with integrated novel multiomic data.


Assuntos
Intolerância à Glucose , Metabolismo dos Lipídeos , Fígado , Camundongos Knockout , Multiômica , Receptores Citoplasmáticos e Nucleares , Animais , Masculino , Camundongos , Fezes/química , Microbioma Gastrointestinal , Glucose/metabolismo , Intolerância à Glucose/metabolismo , Intolerância à Glucose/sangue , Intolerância à Glucose/genética , Metabolismo dos Lipídeos/genética , Fígado/metabolismo , Metaboloma , Multiômica/métodos , Proteoma/metabolismo , Proteômica/métodos , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/deficiência
6.
Diabetes ; 73(9): 1440-1446, 2024 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-38869455

RESUMO

The T allele at rs7903146 in TCF7L2 increases the rate of conversion from prediabetes to type 2 diabetes. This has been associated with impaired ß-cell function and with defective suppression of α-cell secretion by glucose. However, the temporal relationship of these abnormalities is uncertain. To study the longitudinal changes in islet function, we recruited 128 subjects, with 67 homozygous for the diabetes-associated allele (TT) at rs7903146 and 61 homozygous for the protective allele. Subjects were studied on two occasions, 3 years apart, using an oral 75-g glucose challenge. The oral minimal model was used to quantitate ß-cell function; the glucagon secretion rate was estimated from deconvolution of glucagon concentrations. Glucose tolerance worsened in subjects with the TT genotype. This was accompanied by impaired postchallenge glucagon suppression but appropriate ß-cell responsivity to rising glucose concentrations. These data suggest that α-cell abnormalities associated with the TT genotype (rs7903146) occur early and may precede ß-cell dysfunction in people as they develop glucose intolerance and type 2 diabetes.


Assuntos
Diabetes Mellitus Tipo 2 , Glucagon , Teste de Tolerância a Glucose , Células Secretoras de Insulina , Proteína 2 Semelhante ao Fator 7 de Transcrição , Humanos , Proteína 2 Semelhante ao Fator 7 de Transcrição/genética , Proteína 2 Semelhante ao Fator 7 de Transcrição/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Glucagon/metabolismo , Masculino , Feminino , Adulto , Pessoa de Meia-Idade , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/fisiologia , Estudos Longitudinais , Células Secretoras de Glucagon/metabolismo , Ilhotas Pancreáticas/metabolismo , Genótipo , Glicemia/metabolismo , Intolerância à Glucose/genética , Intolerância à Glucose/metabolismo , Alelos
7.
PLoS One ; 19(6): e0304761, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38843265

RESUMO

Type 2 diabetes predisposes patients to heart disease, which is the primary cause of death across the globe. Type 2 diabetes often accompanies obesity and is defined by insulin resistance and abnormal glucose handling. Insulin resistance impairs glucose uptake and results in hyperglycemia, which damages tissues such as kidneys, liver, and heart. 2-oxoglutarate (2-OG)- and iron-dependent oxygenases (2-OGDOs), a family of enzymes regulating various aspects of cellular physiology, have been studied for their role in obesity and diet-induced insulin resistance. However, nothing is known of the 2-OGDO family member 2-oxoglutarate and iron-dependent prolyl hydroxylase domain containing protein 1 (OGFOD1) in this setting. OGFOD1 deletion leads to protection in cardiac ischemia-reperfusion injury and cardiac hypertrophy, which are two cardiac events that can lead to heart failure. Considering the remarkable correlation between heart disease and diabetes, the cardioprotection observed in OGFOD1-knockout mice led us to challenge these knockouts with high-fat diet. Wildtype mice fed a high-fat diet developed diet-induced obesity, insulin resistance, and glucose intolerance, but OGFOD1 knockout mice fed this same diet were resistant to diet-induced obesity and insulin resistance. These results support OGFOD1 down-regulation as a strategy for preventing obesity and insulin handling defects.


Assuntos
Dieta Hiperlipídica , Resistência à Insulina , Camundongos Knockout , Obesidade , Animais , Obesidade/metabolismo , Obesidade/genética , Camundongos , Dieta Hiperlipídica/efeitos adversos , Masculino , Prolil Hidroxilases/metabolismo , Prolil Hidroxilases/genética , Intolerância à Glucose/metabolismo , Intolerância à Glucose/genética , Camundongos Endogâmicos C57BL , Deleção de Genes , Cardiomegalia/metabolismo , Cardiomegalia/prevenção & controle , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Traumatismo por Reperfusão Miocárdica/genética
8.
Metabolism ; 158: 155939, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38843995

RESUMO

BACKGROUND AND AIM: Diacylglycerol kinase (DGK) isoforms catalyze an enzymatic reaction that removes diacylglycerol (DAG) and thereby terminates protein kinase C signaling by converting DAG to phosphatidic acid. DGKδ (type II isozyme) downregulation causes insulin resistance, metabolic inflexibility, and obesity. Here we determined whether DGKδ overexpression prevents these metabolic impairments. METHODS: We generated a transgenic mouse model overexpressing human DGKδ2 under the myosin light chain promoter (DGKδ TG). We performed deep metabolic phenotyping of DGKδ TG mice and wild-type littermates fed chow or high-fat diet (HFD). Mice were also provided free access to running wheels to examine the effects of DGKδ overexpression on exercise-induced metabolic outcomes. RESULTS: DGKδ TG mice were leaner than wild-type littermates, with improved glucose tolerance and increased skeletal muscle glycogen content. DGKδ TG mice were protected against HFD-induced glucose intolerance and obesity. DGKδ TG mice had reduced epididymal fat and enhanced lipolysis. Strikingly, DGKδ overexpression recapitulated the beneficial effects of exercise on metabolic outcomes. DGKδ overexpression and exercise had a synergistic effect on body weight reduction. Microarray analysis of skeletal muscle revealed common gene ontology signatures of exercise and DGKδ overexpression that were related to lipid storage, extracellular matrix, and glycerophospholipids biosynthesis pathways. CONCLUSION: Overexpression of DGKδ induces adaptive changes in both skeletal muscle and adipose tissue, resulting in protection against HFD-induced obesity. DGKδ overexpression recapitulates exercise-induced adaptations on energy homeostasis and skeletal muscle gene expression profiles.


Assuntos
Diacilglicerol Quinase , Dieta Hiperlipídica , Camundongos Transgênicos , Obesidade , Animais , Diacilglicerol Quinase/metabolismo , Diacilglicerol Quinase/genética , Obesidade/metabolismo , Obesidade/genética , Camundongos , Dieta Hiperlipídica/efeitos adversos , Masculino , Glucose/metabolismo , Condicionamento Físico Animal/fisiologia , Músculo Esquelético/metabolismo , Humanos , Intolerância à Glucose/metabolismo , Intolerância à Glucose/genética , Intolerância à Glucose/prevenção & controle , Camundongos Endogâmicos C57BL , Resistência à Insulina/genética
9.
FASEB J ; 38(10): e23690, 2024 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-38795327

RESUMO

Alterations to gene transcription and DNA methylation are a feature of many liver diseases including fatty liver disease and liver cancer. However, it is unclear whether the DNA methylation changes are a cause or a consequence of the transcriptional changes. It is even possible that the methylation changes are not required for the transcriptional changes. If DNA methylation is just a minor player in, or a consequence of liver transcriptional change, then future studies in this area should focus on other systems such as histone tail modifications. To interrogate the importance of de novo DNA methylation, we generated mice that are homozygous mutants for both Dnmt3a and Dnmt3b in post-natal liver. These mice are viable and fertile with normal sized livers. Males, but not females, showed increased adipose depots, yet paradoxically, improved glucose tolerance on both control diet and high-fat diets (HFD). Comparison of the transcriptome and methylome with RNA sequencing and whole-genome bisulfite sequencing in adult hepatocytes revealed that widespread loss of methylation in CpG-rich regions in the mutant did not induce loss of homeostatic transcriptional regulation. Similarly, extensive transcriptional changes induced by HFD did not require de novo DNA methylation. The improved metabolic phenotype of the Dnmt3a/3b mutant mice may be mediated through the dysregulation of a subset of glucose and fat metabolism genes which increase both glucose uptake and lipid export by the liver. However, further work is needed to confirm this.


Assuntos
DNA (Citosina-5-)-Metiltransferases , Metilação de DNA , DNA Metiltransferase 3A , DNA Metiltransferase 3B , Dieta Hiperlipídica , Intolerância à Glucose , Fígado , Animais , Masculino , Dieta Hiperlipídica/efeitos adversos , Fígado/metabolismo , Camundongos , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , DNA Metiltransferase 3A/metabolismo , Intolerância à Glucose/metabolismo , Intolerância à Glucose/genética , Feminino , Camundongos Endogâmicos C57BL
10.
J Biol Chem ; 300(6): 107328, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38679332

RESUMO

Management of chronic obesity-associated metabolic disorders is a key challenge for biomedical researchers. During chronic obesity, visceral adipose tissue (VAT) undergoes substantial transformation characterized by a unique lipid-rich hypoxic AT microenvironment which plays a crucial role in VAT dysfunction, leading to insulin resistance (IR) and type 2 diabetes. Here, we demonstrate that obese AT microenvironment triggers the release of miR-210-3p microRNA-loaded extracellular vesicles from adipose tissue macrophages, which disseminate miR-210-3p to neighboring adipocytes, skeletal muscle cells, and hepatocytes through paracrine and endocrine actions, thereby influencing insulin sensitivity. Moreover, EVs collected from Dicer-silenced miR-210-3p-overexpressed bone marrow-derived macrophages induce glucose intolerance and IR in lean mice. Mechanistically, miR-210-3p interacts with the 3'-UTR of GLUT4 mRNA and silences its expression, compromising cellular glucose uptake and insulin sensitivity. Therapeutic inhibition of miR-210-3p in VAT notably rescues high-fat diet-fed mice from obesity-induced systemic glucose intolerance. Thus, targeting adipose tissue macrophage-specific miR-210-3p during obesity could be a promising strategy for managing IR and type 2 diabetes.


Assuntos
Transportador de Glucose Tipo 4 , Resistência à Insulina , Macrófagos , MicroRNAs , Obesidade , MicroRNAs/genética , MicroRNAs/metabolismo , Animais , Obesidade/metabolismo , Obesidade/genética , Obesidade/patologia , Macrófagos/metabolismo , Camundongos , Transportador de Glucose Tipo 4/metabolismo , Transportador de Glucose Tipo 4/genética , Masculino , Camundongos Endogâmicos C57BL , Tecido Adiposo/metabolismo , Tecido Adiposo/patologia , Humanos , Dieta Hiperlipídica/efeitos adversos , Intolerância à Glucose/metabolismo , Intolerância à Glucose/genética , Intolerância à Glucose/patologia , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/genética , Gordura Intra-Abdominal/metabolismo , Gordura Intra-Abdominal/patologia
11.
Am J Physiol Endocrinol Metab ; 326(5): E663-E672, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38568150

RESUMO

Despite the fact that genes and the environment are known to play a central role in islet function, our knowledge of how these parameters interact to modulate insulin secretory function remains relatively poor. Presently, we performed ex vivo glucose-stimulated insulin secretion and insulin content assays in islets of 213 mice from 13 inbred mouse strains on chow, Western diet (WD), and a high-fat, carbohydrate-free (KETO) diet. Strikingly, among these 13 strains, islets from the commonly used C57BL/6J mouse strain were the least glucose responsive. Using matched metabolic phenotyping data, we performed correlation analyses of isolated islet parameters and found a positive correlation between basal and glucose-stimulated insulin secretion, but no relationship between insulin secretion and insulin content. Using in vivo metabolic measures, we found that glucose tolerance determines the relationship between ex vivo islet insulin secretion and plasma insulin levels. Finally, we showed that islet glucose-stimulated insulin secretion decreased with KETO in almost all strains, concomitant with broader phenotypic changes, such as increased adiposity and glucose intolerance. This is an important finding as it should caution against the application of KETO diet for beta-cell health. Together these data offer key insights into the intersection of diet and genetic background on islet function and whole body glucose metabolism.NEW & NOTEWORTHY Thirteen strains of mice on chow, Western diet, and high-fat, carbohydrate-free (KETO), correlating whole body phenotypes to ex vivo pancreatic islet functional measurements, were used. The study finds a huge spectrum of functional islet responses and insulin phenotypes across all strains and diets, with the ubiquitous C57Bl/6J mouse exhibiting the lowest secretory response of all strains, highlighting the overall importance of considering genetic background when investigating islet function. Ex vivo basal and stimulated insulin secretion are correlated in the islet, and KETO imparts widescale downregulation of islet insulin secretion.


Assuntos
Dieta Hiperlipídica , Secreção de Insulina , Insulina , Ilhotas Pancreáticas , Camundongos Endogâmicos C57BL , Animais , Camundongos , Ilhotas Pancreáticas/metabolismo , Secreção de Insulina/fisiologia , Insulina/metabolismo , Insulina/sangue , Masculino , Dieta Ocidental , Glucose/metabolismo , Dieta com Restrição de Carboidratos , Camundongos Endogâmicos , Glicemia/metabolismo , Intolerância à Glucose/metabolismo , Intolerância à Glucose/genética
12.
Bone ; 182: 117048, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38378083

RESUMO

Osteocalcin deficient mice (OC-/-), on a mixed 129/BL6J background, were reported to show glucose intolerance, insulin insensitivity and reduced insulin secretion at 1-6 mos of age. This is controversial as two studies in OC-/- mice on different backgrounds (C3H/BL6 (5-6 mos.) and C57BL/6N (5 and 9 mos.)) found no effect on glucose metabolism. To determine the role of OC in glucose metabolism we conducted glucose tolerance tests (GTT), insulin tolerances tests (ITT) and glucose stimulated insulin secretion (GSIS) on 6 and 9.5 month-old male OC-/- and OC+/+ mice on a pure C57BL/6J background and fed a normal chow diet. All results were analyzed with a two-way repeated measures ANOVA. The GTT results showed no effect on males at 6 months of age but glucose intolerance was significantly increased (p < 0.05) in male OC-/- mice at 9.5 months of age. The ITT results indicated significantly increased insulin resistance in male OC-/- mice. Glucose stimulated insulin secretion (GSIS) showed insulin significantly (p < 0.05) reduced in OC-/- at several time points. Mouse Osteocalcin injected into OC-/- mice decreased the glucose level. Our results confirm the role of OC in glucose metabolism and insulin sensitivity and demonstrate a role in insulin secretion in older male mice on a C57BL/6J background. Differences in background, age, or experimental procedures could explain controversial results. A delayed onset of the effect of OC on glucose metabolism at 9.5 months in male C57BL/6J mice highlights the importance of background on phenotype. Consideration of genetic background and age may be beneficial for human studies on osteocalcin and glucose homeostasis and may be relevant to the elderly where osteocalcin is reduced.


Assuntos
Envelhecimento , Intolerância à Glucose , Resistência à Insulina , Osteocalcina , Animais , Camundongos , Glicemia/metabolismo , Glucose , Intolerância à Glucose/genética , Intolerância à Glucose/metabolismo , Insulina , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Osteocalcina/genética , Osteocalcina/metabolismo , Envelhecimento/metabolismo
13.
Diabetes ; 73(3): 474-489, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38064504

RESUMO

Genome-wide association studies have identified several gene polymorphisms, including UBE2E2, associated with type 2 diabetes. Although UBE2E2 is one of the ubiquitin-conjugating enzymes involved in the process of ubiquitin modifications, the pathophysiological roles of UBE2E2 in metabolic dysfunction are not yet understood. Here, we showed upregulated UBE2E2 expression in the islets of a mouse model of diet-induced obesity. The diabetes risk allele of UBE2E2 (rs13094957) in noncoding regions was associated with upregulation of UBE2E2 mRNA in the human pancreas. Although glucose-stimulated insulin secretion was intact in the isolated islets, pancreatic ß-cell-specific UBE2E2-transgenic (TG) mice exhibited reduced insulin secretion and decreased ß-cell mass. In TG mice, suppressed proliferation of ß-cells before the weaning period and while receiving a high-fat diet was accompanied by elevated gene expression levels of p21, resulting in decreased postnatal ß-cell mass expansion and compensatory ß-cell hyperplasia, respectively. In TG islets, proteomic analysis identified enhanced formation of various types of polyubiquitin chains, accompanied by increased expression of Nedd4 E3 ubiquitin protein ligase. Ubiquitination assays showed that UBE2E2 mediated the elongation of ubiquitin chains by Nedd4. The data suggest that UBE2E2-mediated ubiquitin modifications in ß-cells play an important role in regulating glucose homeostasis and ß-cell mass.


Assuntos
Diabetes Mellitus Tipo 2 , Intolerância à Glucose , Células Secretoras de Insulina , Camundongos , Animais , Humanos , Intolerância à Glucose/genética , Intolerância à Glucose/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Estudo de Associação Genômica Ampla , Proteômica , Células Secretoras de Insulina/metabolismo , Glucose/metabolismo , Camundongos Transgênicos , Dieta Hiperlipídica/efeitos adversos , Ubiquitinas/genética , Ubiquitinas/metabolismo , Insulina/metabolismo
14.
Acta Diabetol ; 61(1): 79-90, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37688646

RESUMO

AIM: Gestational diabetes mellitus (GDM) affects a significant number of women worldwide and has been associated with lifelong health consequences for their offspring, including increased susceptibility to obesity, insulin resistance, and type II diabetes. Recent studies have suggested that aberrant expression of the long non-coding RNA Meg3 in the liver may contribute to impaired glucose metabolism in individuals. In this study, we aimed to investigate whether intrauterine exposure to hyperglycemia affects glucose intolerance in puberty by mediating the overexpression of LncMeg3 in the liver. METHODS: To test our hypothesis, we established an animal model of intrauterine hyperglycemia to mimic GDM. The progeny was observed for phenotypic changes, and intraperitoneal glucose tolerance tests, insulin tolerance tests, and pyruvate tolerance tests were conducted to assess glucose and insulin tolerance. We also measured LncMeg3 expression in the liver using real-time quantitative PCR and examined differential methylation areas (DMRs) in the Meg3 gene using pyrophosphoric sequencing. To investigate the role of LncMeg3 in glucose tolerance, we conducted Meg3 intervention by vein tail and analyzed the changes in the phenotype and transcriptome of the progeny using bioinformatics analysis. RESULTS: We found that intrauterine exposure to hyperglycemia led to impaired glucose and insulin tolerance in the progeny, with a tendency toward increased fasting blood glucose in fat offspring at 16 weeks (P = 0.0004). LncMeg3 expression was significantly upregulated (P = 0.0061), DNMT3B expression downregulated (P = 0.0226), and DNMT3A (P = 0.0026), TET2 (P = 0.0180) expression upregulated in the liver. Pyrophosphoric sequencing showed hypomethylation in Meg3-DMRs (P = 0.0005). Meg3 intervention by vein tail led to a decrease in the percentage of obese and emaciated offspring (emaciation: 44% vs. 23%; obesity: 25% vs. 15%) and attenuated glucose intolerance. Bioinformatics analysis revealed significant differences in the transcriptome of the progeny, particularly in circadian rhythm and PPAR signaling pathways. CONCLUSION: In conclusion, our study suggests that hypomethylation of Meg3-DMRs increases the expression of the imprinted gene Meg3 in the liver of males, which is associated with impaired glucose tolerance in GDM-F1. MEG3 interference may attenuate glucose intolerance, which may be related to transcriptional changes. Our findings provide new insights into the mechanisms underlying the long-term effects of intrauterine hyperglycemia on progeny health and highlight the potential of Meg3 as an intervention target for glucose intolerance.


Assuntos
Diabetes Mellitus Tipo 2 , Diabetes Gestacional , Intolerância à Glucose , Hiperglicemia , Insulinas , RNA Longo não Codificante , Animais , Feminino , Humanos , Masculino , Gravidez , Glicemia/metabolismo , Diabetes Gestacional/genética , Glucose , Intolerância à Glucose/genética , Hiperglicemia/genética , Hiperglicemia/metabolismo , Obesidade/complicações , Obesidade/genética , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo
15.
Commun Biol ; 6(1): 1226, 2023 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-38049528

RESUMO

The maternal liver is challenged by metabolic demands throughout pregnancy. However, hepatocyte dynamics and their physiological significance in pregnancy remain unclear. Here, we show in mice that hepatocyte proliferation is spatiotemporally regulated in each liver lobular zone during pregnancy, with transient proliferation of periportal and pericentral hepatocytes during mid and late gestation, respectively. Using adeno-associated virus (AAV)-8-mediated expression of the cell cycle inhibitor p21 in hepatocytes, we show that inhibition of hepatocyte proliferation during mid, but not late, gestation impairs liver growth. Transcriptionally, genes involved in glucose/glycogen metabolism are downregulated in late pregnancy when midgestational hepatocyte proliferation is attenuated. In addition, hepatic glycogen storage is abolished, with concomitant elevated blood glucose concentrations, glucose intolerance, placental glycogen deposition, and fetal overgrowth. Laser capture microdissection and RNA-seq analysis of each liver lobular zone show zone-specific changes in the transcriptome during pregnancy and identify genes that are periportally expressed at midgestation, including the hyaluronan-mediated motility receptor (Hmmr). Knockdown of Hmmr in hepatocytes by AAV8-shHmmr suppresses periportal hepatocyte proliferation at midgestation and induces impaired hepatic glycogen storage, glucose intolerance, placental glycogen deposition and fetal overgrowth. Our results suggest that periportal hepatocyte proliferation during midgestation is critical for maternal glycogen metabolism and fetal size.


Assuntos
Diabetes Gestacional , Intolerância à Glucose , Humanos , Camundongos , Gravidez , Feminino , Animais , Glicogênio Hepático/metabolismo , Placenta/metabolismo , Intolerância à Glucose/genética , Intolerância à Glucose/metabolismo , Macrossomia Fetal/metabolismo , Glucose/metabolismo , Glicogênio/metabolismo , Hepatócitos/metabolismo , Homeostase , Proliferação de Células
16.
Endocrinology ; 164(11)2023 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-37897489

RESUMO

In adipose tissue, growth hormone (GH) stimulates lipolysis, leading to an increase in plasma free fatty acid levels and a reduction in insulin sensitivity. In our previous studies, we have found that GH increases lipolysis by reducing peroxisome proliferator-activated receptor γ (PPARγ) transcription activity, leading to a reduction of tat-specific protein 27 (FSP27, also known as CIDEC) expression. In previous studies, our laboratory uncovered 3 developmentally distinct subpopulations of white adipocytes. In this manuscript, we show that one of the subpopulations, termed type 2 adipocytes, has increased GH-induced signaling and lipolysis compared to other adipocyte subtypes. To assess the physiological role of GH-mediated lipolysis mediated by this adipocyte subpopulation, we specifically expressed human FSP27 (hFSP27) transgene in type 2 adipocytes (type2Ad-hFSP27tg mice). Systemically, male type2Ad-hFSP27tg mice displayed reduced serum glycerol release and nonesterified fatty acids levels after acute GH treatment, and improvement in acute, but not chronic, GH-induced glucose intolerance. Furthermore, we demonstrate that type2Ad-hFSP27tg mice displayed improved hepatic insulin signaling. Taken together, these results indicate that this adipocyte subpopulation is a critical regulator of the GH-mediated lipolytic and metabolic response. Thus, further investigation of adipocyte subpopulations may provide novel treatment strategies to regulate GH-induced glucose intolerance in patients with growth and metabolic disorders.


Assuntos
Intolerância à Glucose , Hormônio do Crescimento Humano , Humanos , Masculino , Camundongos , Animais , Hormônio do Crescimento/metabolismo , Lipólise/genética , Intolerância à Glucose/genética , Hormônio do Crescimento Humano/farmacologia , Hormônio do Crescimento Humano/metabolismo , Adipócitos Brancos/metabolismo , Glucose
17.
mSystems ; 8(5): e0057323, 2023 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-37787527

RESUMO

IMPORTANCE: Alterations in the intestinal environment are associated with various diseases, and FFAR4 is abundantly enriched in the intestine, where it has been shown to have the ability to regulate intestinal hormone secretion and intestinal microbiota; here, we confirmed previous reports. Meanwhile, we found that intestinal FFAR4 regulates glucagon-like peptide 1 secretion by decreasing Akkermansia muciniphila abundance and show that such change is associated with the level of glucose utilization at ZT12 in mice. Intestinal FFAR4 deficiency leads to severely impaired glucose tolerance at the ZT12 moment in mice, and Akkermansia muciniphila supplementation ameliorates the abnormal glucose utilization at the ZT12 moment caused by FFAR4 deficiency, which is very similar to the dawn phenomenon in diabetic patients. Collectively, our data suggest that intestinal Ffar4 deteriorates glucose tolerance at the daily light to dark transition by affecting Akkermansia muciniphila.


Assuntos
Microbioma Gastrointestinal , Intolerância à Glucose , Verrucomicrobia , Animais , Humanos , Camundongos , Suplementos Nutricionais , Glucose/metabolismo , Intestinos , Camundongos Knockout , Verrucomicrobia/química , Verrucomicrobia/metabolismo , Luz , Escuridão , Receptores Acoplados a Proteínas G/metabolismo , Intolerância à Glucose/genética , Intolerância à Glucose/metabolismo
18.
Front Immunol ; 14: 1152003, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37711619

RESUMO

Introduction: Subcutaneous adipose tissue (SAT) is a critical regulator of systemic metabolic homeostasis. Persons with HIV (PWH) have an increased risk of metabolic diseases and significant alterations in the SAT immune environment compared with the general population. Methods: We generated a comprehensive single-cell multi-omic SAT atlas to characterize cellular compositional and transcriptional changes in 59 PWH across a spectrum of metabolic health. Results: Glucose intolerance was associated with increased lipid-associated macrophages, CD4+ and CD8+ T effector memory cells, and decreased perivascular macrophages. We observed a coordinated intercellular regulatory program which enriched for genes related to inflammation and lipid-processing across multiple cell types as glucose intolerance increased. Increased CD4+ effector memory tissue-resident cells most strongly associated with altered expression of adipocyte genes critical for lipid metabolism and cellular regulation. Intercellular communication analysis demonstrated enhanced pro-inflammatory and pro-fibrotic signaling between immune cells and stromal cells in PWH with glucose intolerance compared with non-diabetic PWH. Lastly, while cell type-specific gene expression among PWH with diabetes was globally similar to HIV-negative individuals with diabetes, we observed substantially divergent intercellular communication pathways. Discussion: These findings suggest a central role of tissue-resident immune cells in regulating SAT inflammation among PWH with metabolic disease, and underscore unique mechanisms that may converge to promote metabolic disease.


Assuntos
Intolerância à Glucose , Infecções por HIV , Humanos , Intolerância à Glucose/genética , Gordura Subcutânea , Inflamação , Lipídeos
19.
Am J Physiol Endocrinol Metab ; 325(4): E325-E335, 2023 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-37610411

RESUMO

Complement factor 5 of the innate immune system generates C5a and C5b ligands, which initiate inflammatory and cell lysis events, respectively. C5 activation has been linked with obesity-associated metabolic disorders; however, whether it has a causative role is unclear. We generated a C5 null (C5-/-) mouse using CRISPR-Cas9 gene editing to determine whether loss of C5 improves obesity-linked metabolic dysfunction. Generation of a new mouse model was prompted in part by the observation of off-target gene mutations in commercially available C5-/- lines. Male and female wild-type (WT), heterozygous (Het), and C5-/- mice were fed low-fat diet (LFD) or high-fat diet (HFD) for 22 wk. Body weight gain did not differ between genotypes on LFD or HFD. In lean animals, male C5-/- mice had similar glucose tolerance compared with WT controls; however, in obese conditions, glucose tolerance was worsened in C5-/- compared with controls. In contrast, female mice did not exhibit differences in glucose tolerance between genotypes under either dietary paradigm. Fasting insulin was not different between genotypes, whereas diet-induced obese male C5-/- mice had lower fed insulin concentrations compared with WT controls. No differences in adipose tissue inflammation or adipocyte size were identified between groups. Similarly, susceptibility to fatty liver and hepatic inflammation was similar between WT and C5-/- mice. However, the systemic cytokine response to acute endotoxin exposure was decreased in C5-/- mice. Together, these data suggest that loss of C5 worsens glucose tolerance in obese male but not female mice. Additional work is required to pinpoint the mechanisms by which loss of C5 amplifies glucose intolerance in obesity.NEW & NOTEWORTHY We generated a new mouse model of complement factor 5 deficiency. This work was prompted by a need for improved transgenic mouse lines of C5, due to off-target gene mutations. We find that loss of C5 worsens glucose tolerance in a sex-dependent manner. Though the mechanisms evoking glucose intolerance are not clear, we are confident this model will be useful in interrogating complement activation in obesity-associated diseases.


Assuntos
Intolerância à Glucose , Resistência à Insulina , Masculino , Feminino , Camundongos , Animais , Intolerância à Glucose/genética , Intolerância à Glucose/metabolismo , Fator V , Complemento C5 , Obesidade/genética , Obesidade/metabolismo , Dieta Hiperlipídica , Resistência à Insulina/genética , Inflamação/genética , Inflamação/metabolismo , Camundongos Transgênicos , Proteínas do Sistema Complemento , Glucose/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Obesos
20.
Diabetes ; 72(11): 1534-1546, 2023 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-37552863

RESUMO

It is well established that chronic glucocorticoid exposure causes hyperglycemia. While glucocorticoid receptor (GR) stimulates hepatic gluconeogenic gene transcription, additional mechanisms are activated by chronic glucocorticoid exposure to enhance gluconeogenesis. We found that chronic glucocorticoid treatment activated sphingosine-1-phosphate (S1P)-mediated signaling. Hepatic knockdown of hepatic S1P receptor 1 (S1PR1) had no effect on chronic glucocorticoid-induced glucose intolerance but elevated fasting plasma insulin levels. In contrast, hepatic S1PR3 knockdown exacerbated chronic glucocorticoid-induced glucose intolerance without affecting fasting plasma insulin levels. Finally, hepatic S1PR2 knockdown attenuated chronic glucocorticoid-induced glucose intolerance and reduced fasting plasma insulin levels. Here, we focused on dissecting the role of S1PR2 signaling in chronic glucocorticoid response on glucose homeostasis. We found that chronic glucocorticoid-induced hepatic gluconeogenesis, gluconeogenic gene expression, and GR recruitment to the glucocorticoid response elements (GREs) of gluconeogenic genes were all reduced in hepatic S1PR2 knockdown male mice. Hepatic S1PR2 knockdown also enhanced glucocorticoid suppression of RAR-related orphan receptor γ (RORγ) expression. Hepatic RORγ overexpression in hepatic S1PR2 knockdown mice restored glucocorticoid-induced glucose intolerance, gluconeogenic gene expression, and GR recruitment to their GREs. Conversely, RORγ antagonist and the reduction of hepatic RORγ expression attenuated such glucocorticoid effects. Thus, chronic glucocorticoid exposure induces an S1PR2-RORγ axis to cooperate with GR to enhance hepatic gluconeogenesis. Overall, this work provides novel mechanisms of and pharmaceutical targets against steroid-induced hyperglycemia.


Assuntos
Intolerância à Glucose , Hiperglicemia , Insulinas , Hepatopatias , Camundongos , Masculino , Animais , Glucocorticoides/metabolismo , Gluconeogênese/genética , Intolerância à Glucose/induzido quimicamente , Intolerância à Glucose/genética , Intolerância à Glucose/metabolismo , Fígado/metabolismo , Hiperglicemia/metabolismo , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Insulinas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...