Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Cell ; 167(3): 843-857.e14, 2016 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-27720451

RESUMEN

Glucagon and thyroid hormone (T3) exhibit therapeutic potential for metabolic disease but also exhibit undesired effects. We achieved synergistic effects of these two hormones and mitigation of their adverse effects by engineering chemical conjugates enabling delivery of both activities within one precisely targeted molecule. Coordinated glucagon and T3 actions synergize to correct hyperlipidemia, steatohepatitis, atherosclerosis, glucose intolerance, and obesity in metabolically compromised mice. We demonstrate that each hormonal constituent mutually enriches cellular processes in hepatocytes and adipocytes via enhanced hepatic cholesterol metabolism and white fat browning. Synchronized signaling driven by glucagon and T3 reciprocally minimizes the inherent harmful effects of each hormone. Liver-directed T3 action offsets the diabetogenic liability of glucagon, and glucagon-mediated delivery spares the cardiovascular system from adverse T3 action. Our findings support the therapeutic utility of integrating these hormones into a single molecular entity that offers unique potential for treatment of obesity, type 2 diabetes, and cardiovascular disease.


Asunto(s)
Glucagón/uso terapéutico , Enfermedades Metabólicas/tratamiento farmacológico , Triyodotironina/efectos de los fármacos , Animales , Aterosclerosis/tratamiento farmacológico , Peso Corporal/efectos de los fármacos , Huesos/efectos de los fármacos , Ingeniería Química/métodos , Colesterol/metabolismo , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Modelos Animales de Enfermedad , Combinación de Medicamentos , Sistemas de Liberación de Medicamentos , Sinergismo Farmacológico , Glucagón/efectos adversos , Glucagón/química , Glucagón/farmacología , Hiperglucemia/tratamiento farmacológico , Hígado/efectos de los fármacos , Hígado/metabolismo , Ratones , Terapia Molecular Dirigida , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Obesidad/tratamiento farmacológico , Triyodotironina/efectos adversos , Triyodotironina/química , Triyodotironina/farmacología
2.
Diabetes Obes Metab ; 23(1): 195-207, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33001570

RESUMEN

AIMS: Unimolecular peptides targeting the receptors for glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) (GLP-1/GIP co-agonist) have been shown to outperform each single peptide in the treatment of obesity and cardiometabolic disease in preclinical and clinical trials. By combining physiological treatment endpoints with plasma proteomic profiling (PPP), we aimed to identify biomarkers to advance non-invasive metabolic monitoring of compound treatment success and exploration of ulterior treatment effects on an individual basis. MATERIALS AND METHODS: We performed metabolic phenotyping along with PPP in body weight-matched male and female diet-induced obese (DIO) mice treated for 21 days with phosphate-buffered saline, single GIP and GLP-1 mono-agonists, or a GLP-1/GIP co-agonist. RESULTS: GLP-1R/GIPR co-agonism improved obesity, glucose intolerance, non-alcoholic fatty liver disease (NAFLD) and dyslipidaemia with superior efficacy in both male and female mice compared with mono-agonist treatments. PPP revealed broader changes of plasma proteins after GLP-1/GIP co-agonist compared with mono-agonist treatments in both sexes, including established and potential novel biomarkers for systemic inflammation, NAFLD and atherosclerosis. Subtle sex-specific differences have been observed in metabolic phenotyping and PPP. CONCLUSIONS: We herein show that a recently developed unimolecular GLP-1/GIP co-agonist is more efficient in improving metabolic disease than either mono-agonist in both sexes. PPP led to the identification of a sex-independent protein panel with the potential to monitor non-invasively the treatment efficacies on metabolic function of this clinically advancing GLP-1/GIP co-agonist.


Asunto(s)
Incretinas , Proteoma , Animales , Dieta , Femenino , Polipéptido Inhibidor Gástrico , Receptor del Péptido 1 Similar al Glucagón , Masculino , Ratones , Ratones Obesos , Obesidad/tratamiento farmacológico , Proteómica , Resultado del Tratamiento
3.
Diabetologia ; 63(6): 1236-1247, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32140744

RESUMEN

AIMS/HYPOTHESIS: Treatment with the α3ß4 nicotinic acetylcholine receptor (nAChR) agonist, 1,1-dimethyl-4-phenylpiperazinium iodide (DMPP), improves glucose tolerance in diet-induced obese (DIO) mice, but the physiological and molecular mechanisms are unknown. METHODS: DMPP (10 mg/kg body weight, s.c.) was administered either in a single injection (acute) or daily for up to 14 days (chronic) in DIO wild-type (WT) and Chrnb4 knockout (KO) mice and glucose tolerance, tissue-specific tracer-based glucose metabolism, and insulin signalling were assessed. RESULTS: In WT mice, but not in Chrnb4 KO mice, single acute treatment with DMPP induced transient hyperglycaemia, which was accompanied by high plasma adrenaline (epinephrine) levels, upregulated hepatic gluconeogenic genes, and decreased hepatic glycogen content. In contrast to these acute effects, chronic DMPP treatment in WT mice elicited improvements in glucose tolerance already evident after three consecutive days of DMPP treatment. After seven days of DMPP treatment, glucose tolerance was markedly improved, also in comparison with mice that were pair-fed to DMPP-treated mice. The glycaemic benefit of chronic DMPP was absent in Chrnb4 KO mice. Chronic DMPP increased insulin-stimulated glucose clearance into brown adipose tissue (+69%), heart (+93%), gastrocnemius muscle (+74%) and quadriceps muscle (+59%), with no effect in white adipose tissues. After chronic DMPP treatment, plasma adrenaline levels did not increase following an injection with DMPP. In glucose-stimulated skeletal muscle, we detected a decreased phosphorylation of the inhibitory Ser640 phosphorylation site on glycogen synthase and a congruent increase in glycogen accumulation following chronic DMPP treatment. CONCLUSIONS/INTERPRETATION: Our data suggest that DMPP acutely induces adrenaline release and hepatic glycogenolysis, while chronic DMPP-mediated activation of ß4-containing nAChRs improves peripheral insulin sensitivity independently of changes in body weight via mechanisms that could involve increased non-oxidative glucose disposal into skeletal muscle.


Asunto(s)
Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Receptores Nicotínicos/metabolismo , Animales , Glucemia/efectos de los fármacos , Catecolaminas/metabolismo , Yoduro de Dimetilfenilpiperazina/uso terapéutico , Hiperglucemia/tratamiento farmacológico , Hiperglucemia/metabolismo , Resistencia a la Insulina/fisiología , Masculino , Ratones , Ratones Noqueados , Agonistas Nicotínicos/uso terapéutico
4.
Nat Metab ; 2(2): 192-209, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32694693

RESUMEN

Dedifferentiation of insulin-secreting ß cells in the islets of Langerhans has been proposed to be a major mechanism of ß-cell dysfunction. Whether dedifferentiated ß cells can be targeted by pharmacological intervention for diabetes remission, and ways in which this could be accomplished, are unknown as yet. Here we report the use of streptozotocin-induced diabetes to study ß-cell dedifferentiation in mice. Single-cell RNA sequencing (scRNA-seq) of islets identified markers and pathways associated with ß-cell dedifferentiation and dysfunction. Single and combinatorial pharmacology further show that insulin treatment triggers insulin receptor pathway activation in ß cells and restores maturation and function for diabetes remission. Additional ß-cell selective delivery of oestrogen by Glucagon-like peptide-1 (GLP-1-oestrogen conjugate) decreases daily insulin requirements by 60%, triggers oestrogen-specific activation of the endoplasmic-reticulum-associated protein degradation system, and further increases ß-cell survival and regeneration. GLP-1-oestrogen also protects human ß cells against cytokine-induced dysfunction. This study not only describes mechanisms of ß-cell dedifferentiation and regeneration, but also reveals pharmacological entry points to target dedifferentiated ß cells for diabetes remission.


Asunto(s)
Diabetes Mellitus Experimental/tratamiento farmacológico , Hipoglucemiantes/uso terapéutico , Células Secretoras de Insulina/patología , Insulina/uso terapéutico , Animales , Diabetes Mellitus Experimental/patología , Estrógenos/uso terapéutico , Péptido 1 Similar al Glucagón/uso terapéutico , Homeostasis , Humanos , Ratones , Polifarmacología , Inducción de Remisión , Estreptozocina
5.
Mol Metab ; 23: 75-81, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30902502

RESUMEN

OBJECTIVE: The contribution of brown adipose tissue (BAT) to adult human metabolic control is a topic of ongoing investigation. In context, understanding the cellular events leading to BAT uncoupling, heat production, and energy expenditure is anticipated to produce significant insight into this endeavor. The phosphoinositide interacting regulator of transient receptor potentials (Pirt) was recently put forward as a key protein regulating cold sensing downstream of the transient receptor potential melastatin 8 (TRPM8). Notably, TRPM8 has been identified as a non-canonical regulator of BAT thermogenesis. The aim of this investigation was to delineate the role of Pirt in energy homeostasis and glucose metabolism - and the possible involvement of Pirt in TRPM8-elicited energy expenditure. METHODS: To this end, we metabolically phenotyped male and female Pirt deficient (Pirt-/-) mice exposed to a low-fat chow diet or to a high-fat, high-sugar (HFHS) diet. RESULTS: We identified that chow-fed female Pirt-/- mice have an increased susceptibility to develop obesity and glucose intolerance. This effect is abrogated when the mice are exposed to a HFHS diet. Conversely, Pirt-/- male mice display no metabolic phenotype on either diet relative to wild-type (WT) control mice. Finally, we observed that Pirt is dispensable for TRPM8-evoked energy expenditure. CONCLUSION: We here report subtle metabolic abnormalities in female, but not male, Pirt-/- mice. Future studies are required to tease out if metabolic stressors beyond dietary interventions, e.g. temperature fluctuations, are interacting with Pirt-signaling and metabolic control in a sex-specific fashion.


Asunto(s)
Proteínas Portadoras/genética , Metabolismo Energético/fisiología , Glucosa/metabolismo , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Tejido Adiposo Pardo/metabolismo , Animales , Agonistas de los Canales de Calcio/farmacología , Dieta con Restricción de Grasas , Dieta Alta en Grasa , Metabolismo Energético/efectos de los fármacos , Femenino , Técnicas de Inactivación de Genes , Intolerancia a la Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/metabolismo , Pirimidinonas/farmacología , Factores Sexuales , Canales Catiónicos TRPM/agonistas , Canales Catiónicos TRPM/metabolismo , Termogénesis/efectos de los fármacos , Transcriptoma
6.
Nat Commun ; 9(1): 4975, 2018 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-30459311

RESUMEN

In the original PDF version of this article, affiliation 1, 'Institute for Diabetes and Obesity, Helmholtz Diabetes Center (HDC), Helmholtz Zentrum Muenchen & German Center for Diabetes Research (DZD), Neuherberg, Germany', was incorrectly given as 'Institute of Diabetes and Regeneration Research, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health (GmbH), Neuherberg, Germany '. This has now been corrected in the PDF version of the article; the HTML version was correct at the time of publication.

7.
Nat Commun ; 9(1): 4304, 2018 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-30353008

RESUMEN

Pharmacological stimulation of brown adipose tissue (BAT) thermogenesis to increase energy expenditure is progressively being pursued as a viable anti-obesity strategy. Here, we report that pharmacological activation of the cold receptor transient receptor potential cation channel subfamily M member 8 (TRPM8) with agonist icilin mimics the metabolic benefits of cold exposure. In diet-induced obese (DIO) mice, treatment with icilin enhances energy expenditure, and decreases body weight, without affecting food intake. To further potentiate the thermogenic action profile of icilin and add complementary anorexigenic mechanisms, we set out to identify pharmacological partners next to icilin. To that end, we specifically targeted nicotinic acetylcholine receptor (nAChR) subtype alpha3beta4 (α3ß4), which we had recognized as a potential regulator of energy homeostasis and glucose metabolism. Combinatorial targeting of TRPM8 and nAChR α3ß4 by icilin and dimethylphenylpiperazinium (DMPP) orchestrates synergistic anorexic and thermogenic pathways to reverse diet-induced obesity, dyslipidemia, and glucose intolerance in DIO mice.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Obesidad/metabolismo , Receptores Nicotínicos/metabolismo , Canales Catiónicos TRPM/antagonistas & inhibidores , Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Pardo/metabolismo , Animales , Peso Corporal/efectos de los fármacos , Frío , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Dieta , Yoduro de Dimetilfenilpiperazina/farmacología , Yoduro de Dimetilfenilpiperazina/uso terapéutico , Metabolismo Energético/efectos de los fármacos , Hígado Graso/patología , Intolerancia a la Glucosa/patología , Resistencia a la Insulina , Masculino , Melanocortinas/metabolismo , Ratones Endogámicos C57BL , Ratones Obesos , Obesidad/tratamiento farmacológico , Pirimidinonas/farmacología , Pirimidinonas/uso terapéutico , Receptor de Melanocortina Tipo 4/metabolismo , Canales Catiónicos TRPM/metabolismo , Termogénesis/efectos de los fármacos
8.
Mol Metab ; 6(5): 440-446, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28462078

RESUMEN

OBJECTIVE: Obesity is a major health threat that affects men and women equally. Despite this fact, weight-loss potential of pharmacotherapies is typically first evaluated in male mouse models of diet-induced obesity (DIO). To address this disparity we herein determined whether a monomeric peptide with agonism at the receptors for glucagon-like peptide 1 (GLP-1), glucose-dependent insulinotropic polypeptide (GIP), and glucagon is equally efficient in correcting DIO, dyslipidemia, and glucose metabolism in DIO female mice as it has been previously established for DIO male mice. METHODS: Female C57BL/6J mice and a cohort of fatmass-matched C57BL/6J male mice were treated for 27 days via subcutaneous injections with either the GLP-1/GIP/glucagon triagonist or PBS. A second cohort of C57BL/6J male mice was included to match the females in the duration of the high-fat, high-sugar diet (HFD) exposure. RESULTS: Our results show that GLP-1/GIP/glucagon triple agonism inhibits food intake and decreases body weight and body fat mass with comparable potency in male and female mice that have been matched for body fat mass. Treatment improved dyslipidemia in both sexes and reversed diet-induced steatohepatitis to a larger extent in female mice compared to male mice. CONCLUSIONS: We herein show that a recently developed unimolecular peptide triagonist is equally efficient in both sexes, suggesting that this polypharmaceutical strategy might be a relevant alternative to bariatric surgery for the treatment of obesity and related metabolic disorders.


Asunto(s)
Dislipidemias/metabolismo , Hígado Graso/metabolismo , Polipéptido Inhibidor Gástrico/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Glucagón/metabolismo , Obesidad/metabolismo , Adiposidad , Animales , Dieta Alta en Grasa/efectos adversos , Dislipidemias/etiología , Ingestión de Alimentos , Hígado Graso/etiología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/etiología , Factores Sexuales
9.
Diabetes ; 66(2): 272-286, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27650854

RESUMEN

Glucocorticoids (GCs) are important regulators of systemic energy metabolism, and aberrant GC action is linked to metabolic dysfunctions. Yet, the extent to which normal and pathophysiological energy metabolism depend on the GC receptor (GR) in adipocytes remains unclear. Here, we demonstrate that adipocyte GR deficiency in mice significantly impacts systemic metabolism in different energetic states. Plasma metabolomics and biochemical analyses revealed a marked global effect of GR deficiency on systemic metabolite abundance and, thus, substrate partitioning in fed and fasted states. This correlated with a decreased lipolytic capacity of GR-deficient adipocytes under postabsorptive and fasting conditions, resulting from impaired signal transduction from ß-adrenergic receptors to adenylate cyclase. Upon prolonged fasting, the impaired lipolytic response resulted in abnormal substrate utilization and lean mass wasting. Conversely, GR deficiency attenuated aging-/diet-associated obesity, adipocyte hypertrophy, and liver steatosis. Systemic glucose tolerance was improved in obese GR-deficient mice, which was associated with increased insulin signaling in muscle and adipose tissue. We conclude that the GR in adipocytes exerts central but diverging roles in the regulation of metabolic homeostasis depending on the energetic state. The adipocyte GR is indispensable for the feeding-fasting transition but also promotes adiposity and associated metabolic disorders in fat-fed and aged mice.


Asunto(s)
Adipocitos/metabolismo , Envejecimiento/genética , Ayuno , Conducta Alimentaria , Metabolismo de los Lípidos/genética , Hígado/metabolismo , Obesidad/genética , Receptores de Glucocorticoides/genética , Adenilil Ciclasas/metabolismo , Tejido Adiposo/metabolismo , Adiposidad/genética , Envejecimiento/metabolismo , Animales , Western Blotting , Cromatografía Liquida , Dieta Alta en Grasa , Metabolismo Energético , Hígado Graso/genética , Hipertrofia , Insulina/metabolismo , Lipólisis , Espectrometría de Masas , Metabolómica , Ratones , Obesidad/metabolismo , Receptores Adrenérgicos beta/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA