Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Circ Res ; 135(5): 614-628, 2024 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-39011638

RESUMEN

BACKGROUND: Heart failure with preserved ejection fraction (HFpEF) is an emerging major unmet need and one of the most significant clinic challenges in cardiology. The pathogenesis of HFpEF is associated with multiple risk factors. Hypertension and metabolic disorders associated with obesity are the 2 most prominent comorbidities observed in patients with HFpEF. Although hypertension-induced mechanical overload has long been recognized as a potent contributor to heart failure with reduced ejection fraction, the synergistic interaction between mechanical overload and metabolic disorders in the pathogenesis of HFpEF remains poorly characterized. METHOD: We investigated the functional outcome and the underlying mechanisms from concurrent mechanic and metabolic stresses in the heart by applying transverse aortic constriction in lean C57Bl/6J or obese/diabetic B6.Cg-Lepob/J (ob/ob) mice, followed by single-nuclei RNA-seq and targeted manipulation of a top-ranked signaling pathway differentially affected in the 2 experimental cohorts. RESULTS: In contrast to the post-transverse aortic constriction C57Bl/6J lean mice, which developed pathological features of heart failure with reduced ejection fraction over time, the post-transverse aortic constriction ob/ob mice showed no significant changes in ejection fraction but developed characteristic pathological features of HFpEF, including diastolic dysfunction, worsened cardiac hypertrophy, and pathological remodeling, along with further deterioration of exercise intolerance. Single-nuclei RNA-seq analysis revealed significant transcriptome reprogramming in the cardiomyocytes stressed by both pressure overload and obesity/diabetes, markedly distinct from the cardiomyocytes singularly stressed by pressure overload or obesity/diabetes. Furthermore, glucagon signaling was identified as the top-ranked signaling pathway affected in the cardiomyocytes associated with HFpEF. Treatment with a glucagon receptor antagonist significantly ameliorated the progression of HFpEF-related pathological features in 2 independent preclinical models. Importantly, cardiomyocyte-specific genetic deletion of the glucagon receptor also significantly improved cardiac function in response to pressure overload and metabolic stress. CONCLUSIONS: These findings identify glucagon receptor signaling in cardiomyocytes as a critical determinant of HFpEF progression and provide proof-of-concept support for glucagon receptor antagonism as a potential therapy for the disease.


Asunto(s)
Insuficiencia Cardíaca , Ratones Endogámicos C57BL , Volumen Sistólico , Animales , Insuficiencia Cardíaca/fisiopatología , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/etiología , Volumen Sistólico/efectos de los fármacos , Ratones , Masculino , Receptores de Glucagón/antagonistas & inhibidores , Receptores de Glucagón/metabolismo , Receptores de Glucagón/genética , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Ratones Obesos , Función Ventricular Izquierda/efectos de los fármacos , Obesidad/metabolismo , Obesidad/fisiopatología , Obesidad/complicaciones , Modelos Animales de Enfermedad , Transducción de Señal
2.
Diabetologia ; 67(8): 1602-1615, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38705923

RESUMEN

AIMS/HYPOTHESES: Glucagon and glucagon-like peptide-1 (GLP-1) are derived from the same precursor; proglucagon, and dual agonists of their receptors are currently being explored for the treatment of obesity and metabolic dysfunction-associated steatotic liver disease (MASLD). Elevated levels of endogenous glucagon (hyperglucagonaemia) have been linked with hyperglycaemia in individuals with type 2 diabetes but are also observed in individuals with obesity and MASLD. GLP-1 levels have been reported to be largely unaffected or even reduced in similar conditions. We investigated potential determinants of plasma proglucagon and associations of glucagon receptor signalling with metabolic diseases based on data from the UK Biobank. METHODS: We used exome sequencing data from the UK Biobank for ~410,000 white participants to identify glucagon receptor variants and grouped them based on their known or predicted signalling. Data on plasma levels of proglucagon estimated using Olink technology were available for a subset of the cohort (~40,000). We determined associations of glucagon receptor variants and proglucagon with BMI, type 2 diabetes and liver fat (quantified by liver MRI) and performed survival analyses to investigate if elevated proglucagon predicts type 2 diabetes development. RESULTS: Obesity, MASLD and type 2 diabetes were associated with elevated plasma levels of proglucagon independently of each other. Baseline proglucagon levels were associated with the risk of type 2 diabetes development over a 14 year follow-up period (HR 1.13; 95% CI 1.09, 1.17; n=1562; p=1.3×10-12). This association was of the same magnitude across strata of BMI. Carriers of glucagon receptor variants with reduced cAMP signalling had elevated levels of proglucagon (ß 0.847; 95% CI 0.04, 1.66; n=17; p=0.04), and carriers of variants with a predicted frameshift mutation had higher levels of liver fat compared with the wild-type reference group (ß 0.504; 95% CI 0.03, 0.98; n=11; p=0.04). CONCLUSIONS/INTERPRETATION: Our findings support the suggestion that glucagon receptor signalling is involved in MASLD, that plasma levels of proglucagon are linked to the risk of type 2 diabetes development, and that proglucagon levels are influenced by genetic variation in the glucagon receptor, obesity, type 2 diabetes and MASLD. Determining the molecular signalling pathways downstream of glucagon receptor activation may guide the development of biased GLP-1/glucagon co-agonist with improved metabolic benefits. DATA AVAILABILITY: All coding is available through https://github.com/nicwin98/UK-Biobank-GCG.


Asunto(s)
Bancos de Muestras Biológicas , Diabetes Mellitus Tipo 2 , Obesidad , Proglucagón , Receptores de Glucagón , Transducción de Señal , Humanos , Receptores de Glucagón/genética , Receptores de Glucagón/metabolismo , Reino Unido , Femenino , Proglucagón/metabolismo , Proglucagón/genética , Masculino , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/metabolismo , Persona de Mediana Edad , Obesidad/sangre , Anciano , Adulto , Índice de Masa Corporal , Glucagón/sangre , Péptido 1 Similar al Glucagón/sangre , Biobanco del Reino Unido
3.
Diabetes Obes Metab ; 26(6): 2368-2378, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38560764

RESUMEN

AIM: To describe the biomarker strategy that was applied to select survodutide (BI 456906), BI 456908 and BI 456897 from 19 dual glucagon receptor (GCGR)/ glucagon-like peptide-1 receptor (GLP-1R) agonists for in-depth pharmacological profiling, which led to the qualification of survodutide as the clinical development candidate. MATERIALS AND METHODS: Potencies to increase cyclic adenosine monophosphate (cAMP) were determined in Chinese hamster ovary (CHO)-K1 cells stably expressing human GCGR and GLP-1R. Agonism for endogenously expressed receptors was investigated in insulinoma cells (MIN6) for mouse GLP-1R, and in rat primary hepatocytes for the GCGR. In vivo potencies to engage the GLP-1R or GCGR were determined, measuring improvement in oral glucose tolerance (30 nmol/kg) and increase in plasma fibroblast growth factor-21 (FGF21) and liver nicotinamide N-methyltransferase (NNMT) mRNA expression (100 nmol/kg), respectively. Body weight- and glucose-lowering efficacies were investigated in diet-induced obese (DIO) mice and diabetic db/db mice, respectively. RESULTS: Upon acute dosing in lean mice, target engagement biomarkers for the GCGR and GLP-1R demonstrated a significant correlation (Spearman correlation coefficient with p < 0.05) to the in vitro GCGR and GLP-1R potencies for the 19 dual agonists investigated. Survodutide, BI 456908 and BI 456897 were selected for in-depth pharmacological profiling based on the significant improvement in acute oral glucose tolerance achieved (area under the curve [AUC] of 54%, 57% and 60% vs. vehicle) that was comparable to semaglutide (AUC of 45% vs. vehicle), while showing different degrees of in vivo GCGR engagement, as determined by hepatic NNMT mRNA expression (increased by 15- to 17-fold vs. vehicle) and plasma FGF21 concentrations (increased by up to sevenfold vs. vehicle). In DIO mice, survodutide (30 nmol/kg/once daily), BI 456908 (30 nmol/kg/once daily) and BI 456897 (10 nmol/kg/once daily) achieved a body weight-lowering efficacy from baseline of 25%, 27% and 26%, respectively. In db/db mice, survodutide and BI 456908 (10 and 20 nmol/kg/once daily) significantly lowered glycated haemoglobin (0.4%-0.6%); no significant effect was observed for BI 456897 (3 and 7 nmol/kg/once daily). CONCLUSIONS: Survodutide was selected as the clinical candidate based on its balanced dual GCGR/GLP-1R pharmacology, engaging the GCGR for robust body weight-lowering efficacy exceeding that of selective GLP-1R agonists, while achieving antidiabetic efficacy that was comparable to selective GLP-1R agonism. Survodutide is currently being investigated in Phase 3 clinical trials in people living with obesity.


Asunto(s)
Cricetulus , Receptor del Péptido 1 Similar al Glucagón , Hipoglucemiantes , Receptores de Glucagón , Animales , Receptores de Glucagón/agonistas , Receptores de Glucagón/genética , Ratones , Receptor del Péptido 1 Similar al Glucagón/agonistas , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Receptor del Péptido 1 Similar al Glucagón/genética , Células CHO , Hipoglucemiantes/farmacología , Hipoglucemiantes/uso terapéutico , Humanos , Biomarcadores/sangre , Masculino , Ratas , Ratones Obesos , Ratones Endogámicos C57BL , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Diabetes Mellitus Tipo 2/tratamiento farmacológico
4.
Mol Metab ; 85: 101947, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38677509

RESUMEN

OBJECTIVE: Type 2 diabetes (T2D) is characterised by the loss of first-phase insulin secretion. We studied mice with ß-cell selective loss of the glucagon receptor (Gcgrfl/fl X Ins-1Cre), to investigate the role of intra-islet glucagon receptor (GCGR) signalling on pan-islet [Ca2+]I activity and insulin secretion. METHODS: Metabolic profiling was conducted on Gcgrß-cell-/- and littermate controls. Crossing with GCaMP6f (STOP flox) animals further allowed for ß-cell specific expression of a fluorescent calcium indicator. These islets were functionally imaged in vitro and in vivo. Wild-type mice were transplanted with islets expressing GCaMP6f in ß-cells into the anterior eye chamber and placed on a high fat diet. Part of the cohort received a glucagon analogue (GCG-analogue) for 40 days and the control group were fed to achieve weight matching. Calcium imaging was performed regularly during the development of hyperglycaemia and in response to GCG-analogue treatment. RESULTS: Gcgrß-cell-/- mice exhibited higher glucose levels following intraperitoneal glucose challenge (control 12.7 mmol/L ± 0.6 vs. Gcgrß-cell-/- 15.4 mmol/L ± 0.0 at 15 min, p = 0.002); fasting glycaemia was not different to controls. In vitro, Gcgrß-cell-/- islets showed profound loss of pan-islet [Ca2+]I waves in response to glucose which was only partially rescued in vivo. Diet induced obesity and hyperglycaemia also resulted in a loss of co-ordinated [Ca2+]I waves in transplanted islets. This was reversed with GCG-analogue treatment, independently of weight-loss (n = 8). CONCLUSION: These data provide novel evidence for the role of intra-islet GCGR signalling in sustaining synchronised [Ca2+]I waves and support a possible therapeutic role for glucagonergic agents to restore the insulin secretory capacity lost in T2D.


Asunto(s)
Diabetes Mellitus Tipo 2 , Glucagón , Glucosa , Homeostasis , Secreción de Insulina , Células Secretoras de Insulina , Receptores de Glucagón , Transducción de Señal , Animales , Glucagón/metabolismo , Ratones , Células Secretoras de Insulina/metabolismo , Glucosa/metabolismo , Receptores de Glucagón/metabolismo , Receptores de Glucagón/genética , Diabetes Mellitus Tipo 2/metabolismo , Insulina/metabolismo , Masculino , Islotes Pancreáticos/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Dieta Alta en Grasa , Glucemia/metabolismo , Femenino
5.
Adv Sci (Weinh) ; 11(29): e2400819, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38837628

RESUMEN

Glucagon receptor (GCGR) agonism offers potentially greater effects on the mitigation of hepatic steatosis. However, its underlying mechanism is not fully understood. Here, it screened tetraspanin CD9 might medicate hepatic effects of GCGR agonist. CD9 is decreased in the fatty livers of patients and upregulated upon GCGR activation. Deficiency of CD9 in the liver exacerbated diet-induced hepatic steatosis via complement factor D (CFD) regulated fatty acid metabolism. Specifically, CD9 modulated hepatic fatty acid synthesis and oxidation genes through regulating CFD expression via the ubiquitination-proteasomal degradation of FLI1. In addition, CD9 influenced body weight by modulating lipogenesis and thermogenesis of adipose tissue through CFD. Moreover, CD9 reinforcement in the liver alleviated hepatic steatosis, and blockage of CD9 abolished the remission of hepatic steatosis induced by cotadutide treatment. Thus, CD9 medicates the hepatic beneficial effects of GCGR signaling, and may server as a promising therapeutic target for hepatic steatosis.


Asunto(s)
Hígado Graso , Tetraspanina 29 , Tetraspanina 29/metabolismo , Tetraspanina 29/genética , Animales , Ratones , Humanos , Hígado Graso/metabolismo , Hígado Graso/tratamiento farmacológico , Modelos Animales de Enfermedad , Masculino , Receptores de Glucagón/agonistas , Receptores de Glucagón/metabolismo , Receptores de Glucagón/genética , Ratones Endogámicos C57BL , Hígado/metabolismo , Hígado/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
6.
Endocrinol. nutr. (Ed. impr.) ; 58(6): 258-266, jun.-jul. 2011. ilus
Artículo en Inglés | IBECS (España) | ID: ibc-97119

RESUMEN

Background and aim: To elucidate the pathogenetic mechanisms of a mutant P86S glucagon receptor (GCGR) in causing a novel human disease (Mahvash disease).Material and method: Enhanced green fluorescent protein (EGFP)-tagged WT and P86S GCGR were expressed in HEK 293 or H1299 cells either transiently or stably. Receptor localization and internalization, and cell apoptosis were studied by fluorescence microscopy, and calcium signaling by Rhod-3 labeling. Gene expression was assayed by RT-PCR or Western blot. Cell fate was determined by live cell imaging. Results: Unlike WT GCGR, P86S was partially localized to the plasma membrane and partially in the cytoplasm as previously reported and did not undergo internalization upon glucagon treatment. P86S did not elicit calcium response after treatment with 1 M glucagon. Cells transiently expressing P86S exhibited more apoptosis than those expressing WT GCGR (18.3% vs 2.1%,P < 0.05) but the X-box binding protein 1 mRNA cleavage, a marker of endoplasmic reticulum(ER) stress, was not evident, suggesting that the apoptosis did not result from ER stress. Cells stably expressing P86S did not exhibit apoptosis and a quarter of them harbored a novel inclusion body-like circular structure that was marked by P86S and ER residential proteins. These circular ER bodies were not seen in cells expressing WT GCGR or transiently expressing P86Sand were not affected by treatment with protea some inhibitor or microtubule depolymerizer, suggesting that they do not represent aggresome structures. The circular ER bodies could fuse and split to form new bodies. Conclusion: The naturally-occurring P86S mutant GCGR exhibits abnormal receptor internalization and calcium mobilization, and causes apoptosis. The novel dynamic circular ER bodies may be adaptive in nature to nullify the toxic effects on P86S. These findings provide further in sights into the pathogenetic mechanisms of Mahvash disease (AU)


Antecedentes y objetivo: Conocer los mecanismos patogenéticos de un receptor del glucagón (GCGR) con la mutación P86S en una nueva enfermedad humana (enfermedad de Mahvash). Material y método: Se expresaron tipos silvestre y P86S del GCGR marcados con proteína verde fluorescente mejorada (EGFP) en células de HEK 293 o H1299 de forma estable o transitoria. Se estudió la localización e internalización del receptor, y la apoptosis celular mediante microscopio de fluorescencia, y la señalización del calcio mediante marcadores Rhod-3. La expresión génica se analizó mediante RT-PCR o Western blot. El destino de la célula se determinó mediante imágenes de las células vivas. Resultados: A diferencia del tipo silvestre, la mutación P86S se localizó parte en la membrana plasmática y parte en el citoplasma como se ha notificado con anterioridad, y no se produjo internalización tras tratamiento con glucagón. La P86S no indujo respuesta de calcio después del tratamiento con 1 M de glucagón. Las células que expresaban transitoriamente P86S exhibieron más la apoptosis que aquella con GCGR silvestre (18,3% frente a 2,1%, p < 0,05), pero no resultó patente la existencia de ARNm escindido en proteína de unión X-box 1, un marcador de estrés en el retículo endoplasmático (RE), lo que indica que la apoptosis no fue el resultado de estrés en el RE. Las células que expresan establemente la P86S no mostraron apoptosis y una cuarta parte de ellas albergaba la inclusión de una estructura circular corpusculoide nueva marcada por P86S y proteínas residentes del RE. Estos cuerpos circulares no se observaron en las células que expresaban GCGR silvestre o transitoriamente P86S y no se vieron afectados por el tratamiento con el inhibidor del (..) (AU)


Asunto(s)
Humanos , Señalización del Calcio/genética , Glucagón/sangre , Mutación Missense , Síndromes Neoplásicos Hereditarios/genética , Tumores Neuroendocrinos/genética , Neoplasias Pancreáticas/genética , Mutación Puntual , Receptores de Glucagón/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA