Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Respir Res ; 25(1): 153, 2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38566174

RESUMEN

BACKGROUND: Wnt/ß-catenin signaling is critical for lung development and AT2 stem cell maintenance in adults, but excessive pathway activation has been associated with pulmonary fibrosis, both in animal models and human diseases such as idiopathic pulmonary fibrosis (IPF). IPF is a detrimental interstitial lung disease, and although two approved drugs limit functional decline, transplantation is the only treatment that extends survival, highlighting the need for regenerative therapies. METHODS: Using our antibody-based platform of Wnt/ß-catenin modulators, we investigated the ability of a pathway antagonist and pathway activators to reduce pulmonary fibrosis in the acute bleomycin model, and we tested the ability of a WNT mimetic to affect alveolar organoid cultures. RESULTS: A WNT mimetic agonist with broad FZD-binding specificity (FZD1,2,5,7,8) potently expanded alveolar organoids. Upon therapeutic dosing, a broad FZD-binding specific Wnt mimetic decreased pulmonary inflammation and fibrosis and increased lung function in the bleomycin model, and it impacted multiple lung cell types in vivo. CONCLUSIONS: Our results highlight the unexpected capacity of a WNT mimetic to effect tissue repair after lung damage and support the continued development of Wnt/ß-catenin pathway modulation for the treatment of pulmonary fibrosis.


Asunto(s)
Fibrosis Pulmonar Idiopática , beta Catenina , Adulto , Animales , Humanos , beta Catenina/metabolismo , Pulmón/metabolismo , Fibrosis Pulmonar Idiopática/inducido químicamente , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Fibrosis Pulmonar Idiopática/metabolismo , Vía de Señalización Wnt , Bleomicina/toxicidad
2.
J Biol Chem ; 289(44): 30470-30480, 2014 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-25204652

RESUMEN

The role of fibroblast growth factor receptor 4 (FGFR4) in regulating bile acid synthesis has been well defined; however, its reported role on glucose and energy metabolism remains unresolved. Here, we show that FGFR4 deficiency in mice leads to improvement in glucose metabolism, insulin sensitivity, and reduction in body weight under high fat conditions. Mechanism of action studies in FGFR4-deficient mice suggest that the effects are mediated in part by increased plasma levels of adiponectin and the endocrine FGF factors FGF21 and FGF15, the latter of which increase in response to an elevated bile acid pool. Direct actions of increased bile acids on bile acid receptors, and other potential indirect mechanisms, may also contribute to the observed metabolic changes. The results described herein suggest that FGFR4 antagonists alone, or in combination with other agents, could serve as a novel treatment for diabetes.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Resistencia a la Insulina , Obesidad/metabolismo , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/genética , Adiponectina/sangre , Tejido Adiposo/metabolismo , Animales , Femenino , Factores de Crecimiento de Fibroblastos/administración & dosificación , Factores de Crecimiento de Fibroblastos/sangre , Glucosa/metabolismo , Células HEK293 , Humanos , Íleon/metabolismo , Hígado/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/etiología , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/deficiencia , Transcriptoma
3.
Am J Physiol Endocrinol Metab ; 307(12): E1144-52, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25370851

RESUMEN

Elucidating the role of secreted frizzled-related protein 5 (SFRP5) in metabolism and obesity has been complicated by contradictory findings when knockout mice were used to determine metabolic phenotypes. By overexpressing SFRP5 in obese, prediabetic mice we consistently observed elevated hyperglycemia and glucose intolerance, supporting SFRP5 as a negative regulator of glucose metabolism. Accordingly, Sfrp5 mRNA expression analysis of both epididymal and subcutaneous adipose depots of mice indicated a correlation with obesity. Thus, we generated a monoclonal antibody (mAb) against SFRP5 to ascertain the effect of SFRP5 inhibition in vivo. Congruent with SFRP5 overexpression worsening blood glucose levels and glucose intolerance, anti-SFRP5 mAb therapy improved these phenotypes in vivo. The results from both the overexpression and mAb inhibition studies suggest a role for SFRP5 in glucose metabolism and pancreatic ß-cell function and thus establish the use of an anti-SFRP5 mAb as a potential approach to treat type 2 diabetes.


Asunto(s)
Glucosa/metabolismo , Células Secretoras de Insulina/metabolismo , Péptidos y Proteínas de Señalización Intercelular/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Anticuerpos Monoclonales/inmunología , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Inmunoglobulina G/inmunología , Células Secretoras de Insulina/efectos de los fármacos , Péptidos y Proteínas de Señalización Intercelular/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Obesidad/complicaciones , Obesidad/genética , Obesidad/metabolismo
4.
J Lipid Res ; 54(2): 325-32, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23204296

RESUMEN

Elevated triglyceride (TG) and cholesterol levels are risk factors for cardiovascular disease and are often associated with diabetes and metabolic syndrome. Recent reports suggest that fibroblast growth factor (FGF)19 and FGF21 can dramatically improve metabolic dysfunction, including hyperglycemia, hypertriglyceridemia, and hypercholesterolemia. Due to their similar receptor specificities and co-receptor requirements, FGF19 and FGF21 share many common properties and have been thought to be interchangeable in metabolic regulation. Here we directly compared how pharmacological administration of recombinant FGF19 or FGF21 proteins affect metabolism in B6.V-Lep(ob)/J leptin-deficient mice. FGF19 and FGF21 equally improved glucose parameters; however, we observed increased serum TG and cholesterol levels after treatment with FGF19 but not with FGF21. Increases in serum TGs were also observed after a 4-day treatment with FGF19 in C57BL6/J mice on a high-fat diet. This is in contrast to many literature reports that showed significant improvements in hyperlipidemia after chronic treatment with FGF19 or FGF21 in high-fat diet models. We propose that FGF19 has lipid-raising and lipid-lowering actions mediated through different FGF receptors and target tissues, and the results described here provide a potential mechanism that may explain the inconsistency in the reported effects of FGF19 on lipid metabolism.


Asunto(s)
Factores de Crecimiento de Fibroblastos/farmacología , Metabolismo de los Lípidos/efectos de los fármacos , Animales , Línea Celular , Colesterol/sangre , Dieta/efectos adversos , Factores de Crecimiento de Fibroblastos/química , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Obesidad/sangre , Obesidad/etiología , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/metabolismo , Triglicéridos/sangre
5.
Nature ; 446(7132): 203-7, 2007 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-17322907

RESUMEN

Complement-derived anaphylatoxins regulate immune and inflammatory responses through G-protein-coupled receptor (GPCR)-mediated signalling. C5L2 (also known as GPR77) is a relatively new GPCR thought to be a non-signalling receptor binding to C5a, on the basis of sequence information and experimental evidence. Here we show, using gene targeting, that C5L2 is required to facilitate C5a signalling in neutrophils, macrophages and fibroblasts in vitro. Deficiency of C5L2 results in reduced inflammatory cell infiltration, suggesting that C5L2 is critical for optimal C5a-mediated cell infiltration in certain in vivo settings. C5L2 is also involved in optimizing C3a-induced signals. Furthermore, like mice incapable of C3a/complement 3a receptor (C3aR) signalling, C5L2-deficient mice are hypersensitive to lipopolysaccharide (LPS)-induced septic shock, show reduced ovalbumin (OVA)-induced airway hyper-responsiveness and inflammation, and are mildly delayed in haematopoietic cell regeneration after gamma-irradiation. Our data indicate that C5L2 can function as a positive modulator for both C5a- and C3a-anaphylatoxin-induced responses.


Asunto(s)
Complemento C3a/metabolismo , Complemento C5a/metabolismo , Actinas/química , Actinas/metabolismo , Animales , Bovinos , Células Cultivadas , Activación de Complemento , Complemento C3a/inmunología , Complemento C5a/inmunología , Fibroblastos , Regulación de la Expresión Génica , Células Madre Hematopoyéticas/citología , Humanos , Inflamación , Pulmón/citología , Neutrófilos/citología , Neutrófilos/metabolismo , Receptor de Anafilatoxina C5a , Receptores de Quimiocina/deficiencia , Receptores de Quimiocina/genética , Receptores de Quimiocina/metabolismo , Transducción de Señal
6.
Proc Natl Acad Sci U S A ; 107(32): 14158-63, 2010 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-20660733

RESUMEN

FGF19 and FGF21 are distinctive members of the FGF family that function as endocrine hormones. Their potent effects on normalizing glucose, lipid, and energy homeostasis in disease models have made them an interesting focus of research for combating the growing epidemics of diabetes and obesity. Despite overlapping functions, FGF19 and FGF21 have many discrete effects, the most important being that FGF19 has both metabolic and proliferative effects, whereas FGF21 has only metabolic effects. Here we identify the structural determinants dictating differential receptor interactions that explain and distinguish these two physiological functions. We also have generated FGF19 variants that have lost the ability to induce hepatocyte proliferation but that still are effective in lowering plasma glucose levels and improving insulin sensitivity in mice. Our results add valuable insight into the structure-function relationship of FGF19/FGF21 and identify the structural basis underpinning the distinct proliferative feature of FGF19 compared with FGF21. In addition, these studies provide a road map for engineering FGF19 as a potential therapeutic candidate for treating diabetes and obesity.


Asunto(s)
Glucemia , Proliferación Celular , Factores de Crecimiento de Fibroblastos/fisiología , Hepatocitos/citología , Secuencia de Aminoácidos , Animales , Diabetes Mellitus/tratamiento farmacológico , Variación Genética , Resistencia a la Insulina , Ratones , Obesidad/tratamiento farmacológico , Fragmentos de Péptidos/farmacología , Proteínas Recombinantes de Fusión/farmacología , Proteínas Recombinantes/farmacología
7.
Biochem Biophys Res Commun ; 418(1): 1-5, 2012 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-22155242

RESUMEN

GPR21 is an orphan G-protein-coupled receptor. We found that mice deficient for the GPR21 gene were resistant to diet-induced obesity. Knockout mice were leaner than their wildtype counterpart, despite that no difference was observed in food intake. No differences were observed in the respiratory exchange rate and thermogenesis. However, knockout mice were more active than wildtype littermates, and this level of activity may be an underlying reason for the difference in energy balance. Mutant mice were more sensitive to insulin than their wildtype control and showed an improved glucose tolerance. Several inflammatory markers MCP-1, CRP and IP-10 were decreased in mutant animals, suggesting that GPR21 may also mediate its effect through anti-inflammatory mechanisms. We found that GPR21 is widely expressed in all tissues, with the highest levels found in the brain and in the spleen. Overall, these findings suggest that GPR21 may play an important role in regulating body weight and glucose metabolism.


Asunto(s)
Resistencia a la Insulina/genética , Insulina/farmacología , Obesidad/genética , Receptores Acoplados a Proteínas G/genética , Animales , Biomarcadores/metabolismo , Peso Corporal/genética , Proteína C-Reactiva/metabolismo , Quimiocina CCL2/metabolismo , Quimiocina CXCL10/metabolismo , Dieta/efectos adversos , Expresión Génica , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Ratones , Ratones Noqueados , Obesidad/etiología , Distribución Tisular
8.
Cell Mol Gastroenterol Hepatol ; 14(2): 435-464, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35569814

RESUMEN

BACKGROUND AND AIMS: Current management of inflammatory bowel disease leaves a clear unmet need to treat the severe epithelial damage. Modulation of Wnt signaling might present an opportunity to achieve histological remission and mucosal healing when treating IBD. Exogenous R-spondin, which amplifies Wnt signals by maintaining cell surface expression of Frizzled (Fzd) and low-density lipoprotein receptor-related protein receptors, not only helps repair intestine epithelial damage, but also induces hyperplasia of normal epithelium. Wnt signaling may also be modulated with the recently developed Wnt mimetics, recombinant antibody-based molecules mimicking endogenous Wnts. METHODS: We first compared the epithelial healing effects of RSPO2 and a Wnt mimetic with broad Fzd specificity in an acute dextran sulfate sodium mouse colitis model. Guided by Fzd expression patterns in the colon epithelium, we also examined the effects of Wnt mimetics with subfamily Fzd specificities. RESULTS: In the DSS model, Wnt mimetics repaired damaged colon epithelium and reduced disease activity and inflammation and had no apparent effect on uninjured tissue. We further identified that the FZD5/8 and LRP6 receptor-specific Wnt mimetic, SZN-1326-p, was associated with the robust repair effect. Through a range of approaches including single-cell transcriptome analyses, we demonstrated that SZN-1326-p directly impacted epithelial cells, driving transient expansion of stem and progenitor cells, promoting differentiation of epithelial cells, histologically restoring the damaged epithelium, and secondarily to epithelial repair, reducing inflammation. CONCLUSIONS: It is feasible to design Wnt mimetics such as SZN-1326-p that impact damaged intestine epithelium specifically and restore its physiological functions, an approach that holds promise for treating epithelial damage in inflammatory bowel disease.


Asunto(s)
Colitis , Enfermedades Inflamatorias del Intestino , Animales , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Modelos Animales de Enfermedad , Inflamación , Enfermedades Inflamatorias del Intestino/patología , Ratones , Regeneración , Vía de Señalización Wnt
9.
Nat Commun ; 12(1): 3247, 2021 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-34059688

RESUMEN

The Wnt signaling pathway is intricately connected with bone mass regulation in humans and rodent models. We designed an antibody-based platform that generates potent and selective Wnt mimetics. Using this platform, we engineer bi-specific Wnt mimetics that target Frizzled and low-density lipoprotein receptor-related proteins and evaluate their effects on bone accrual in murine models. These synthetic Wnt agonists induce rapid and robust bone building effects, and correct bone mass deficiency and bone defects in various disease models, including osteoporosis, aging, and long bone fracture. Furthermore, when these Wnt agonists are combined with antiresorptive bisphosphonates or anti-sclerostin antibody therapies, additional bone accrual/maintenance effects are observed compared to monotherapy, which could benefit individuals with severe and/or acute bone-building deficiencies. Our data support the continued development of Wnt mimetics for the treatment of diseases of low bone mineral density, including osteoporosis.


Asunto(s)
Conservadores de la Densidad Ósea/farmacología , Resorción Ósea/tratamiento farmacológico , Fracturas del Fémur/tratamiento farmacológico , Osteoporosis Posmenopáusica/tratamiento farmacológico , Proteínas Wnt/agonistas , Anciano , Envejecimiento/fisiología , Animales , Densidad Ósea/efectos de los fármacos , Densidad Ósea/fisiología , Conservadores de la Densidad Ósea/uso terapéutico , Resorción Ósea/fisiopatología , Difosfonatos/farmacología , Difosfonatos/uso terapéutico , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Sinergismo Farmacológico , Quimioterapia Combinada/métodos , Femenino , Fracturas del Fémur/patología , Fémur/efectos de los fármacos , Fémur/lesiones , Fémur/patología , Humanos , Ratones , Osteoporosis Posmenopáusica/fisiopatología , Vía de Señalización Wnt/efectos de los fármacos , Adulto Joven
10.
J Cell Biol ; 171(6): 931-7, 2005 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-16365160

RESUMEN

Keratins 8 and 18 (K8/18) are major constituents of Mallory bodies (MBs), which are hepatocyte cytoplasmic inclusions seen in several liver diseases. K18-null but not K8-null or heterozygous mice form MBs, which indicates that K8 is important for MB formation. Early stages in MB genesis include K8/18 hyperphosphorylation and overexpression. We used transgenic mice that overexpress K8, K18, or K8/18 to test the importance of K8 and/or K18 in MB formation. MBs were induced by feeding 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC). Livers of young K8 or K8/K18 overexpressors had no histological abnormalities despite increased keratin protein and phosphorylation. In aging mice, only K8-overexpressing livers spontaneously developed small "pre-MB" aggregates. Only K8-overexpressing young mice are highly susceptible to MB formation after short-term DDC feeding. Thus, the K8 to K18 ratio, rather than K8/18 overexpression by itself, plays an essential role in MB formation. K8 overexpression is sufficient to form pre-MB and primes animals to accumulate MBs upon DDC challenge, which may help explain MB formation in human liver diseases.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Hepatocitos/ultraestructura , Cuerpos de Inclusión/ultraestructura , Queratinas/metabolismo , Proteínas/metabolismo , Animales , Humanos , Queratina-18 , Queratina-8 , Hígado/enzimología , Hígado/metabolismo , Ratones , Ratones Transgénicos , Microscopía Fluorescente , Modelos Biológicos , ARN Mensajero/metabolismo
11.
Sci Rep ; 10(1): 13951, 2020 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-32811902

RESUMEN

R-spondin (RSPO) proteins amplify Wnt signaling and stimulate regeneration in a variety of tissues. To repair tissue in a tissue-specific manner, tissue-targeted RSPO mimetic molecules are desired. Here, we mutated RSPO (RSPO2 F105R/F109A) to eliminate LGR binding while preserving ZNRF3/RNF43 binding and targeted the mutated RSPO to a liver specific receptor, ASGR1. The resulting bi-specific molecule (αASGR1-RSPO2-RA) enhanced Wnt signaling effectively in vitro, and its activity was limited to ASGR1 expressing cells. Systemic administration of αASGR1-RSPO2-RA in mice specifically upregulated Wnt target genes and stimulated cell proliferation in liver but not intestine (which is more responsive to non-targeted RSPO2) in healthy mice, and improved liver function in diseased mice. These results not only suggest that a tissue-specific RSPO mimetic protein can stimulate regeneration in a cell-specific manner, but also provide a blueprint of how a tissue-specific molecule might be constructed for applications in a broader context.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/farmacología , Regeneración Hepática/efectos de los fármacos , Regeneración Hepática/fisiología , Animales , Receptor de Asialoglicoproteína/efectos de los fármacos , Receptor de Asialoglicoproteína/metabolismo , Línea Celular , Proliferación Celular , Descubrimiento de Drogas/métodos , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/efectos de los fármacos , Trombospondinas/metabolismo , Trombospondinas/uso terapéutico , Ubiquitina-Proteína Ligasas/metabolismo , Vía de Señalización Wnt/efectos de los fármacos , beta Catenina/metabolismo
12.
J Cell Biol ; 161(4): 749-56, 2003 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-12771125

RESUMEN

The intermediate filament protein keratin 8 (K8) is critical for the development of most mouse embryos beyond midgestation. We find that 68% of K8-/- embryos, in a sensitive genetic background, are rescued from placental bleeding and subsequent death by cellular complementation with wild-type tetraploid extraembryonic cells. This indicates that the primary defect responsible for K8-/- lethality is trophoblast giant cell layer failure. Furthermore, the genetic absence of maternal but not paternal TNF doubles the number of viable K8-/- embryos. Finally, we show that K8-/- concepti are more sensitive to a TNF-dependent epithelial apoptosis induced by the administration of concanavalin A (ConA) to pregnant mothers. The ConA-induced failure of the trophoblast giant cell barrier results in hematoma formation between the trophoblast giant cell layer and the embryonic yolk sac in a phenocopy of dying K8-deficient concepti in a sensitive genetic background. We conclude the lethality of K8-/- embryos is due to a TNF-sensitive failure of trophoblast giant cell barrier function. The keratin-dependent protection of trophoblast giant cells from a maternal TNF-dependent apoptotic challenge may be a key function of simple epithelial keratins.


Asunto(s)
Queratinas/metabolismo , Placenta/fisiología , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Apoptosis/efectos de los fármacos , Concanavalina A/farmacología , Desarrollo Embrionario y Fetal/efectos de los fármacos , Femenino , Eliminación de Gen , Células Gigantes/efectos de los fármacos , Células Gigantes/metabolismo , Células Gigantes/patología , Hematoma/metabolismo , Hematoma/patología , Queratina-8 , Queratinas/genética , Masculino , Ratones , Ratones Noqueados , Placenta/efectos de los fármacos , Embarazo , Receptores del Factor de Necrosis Tumoral/deficiencia , Receptores del Factor de Necrosis Tumoral/genética , Receptores del Factor de Necrosis Tumoral/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral , Trofoblastos/efectos de los fármacos , Trofoblastos/metabolismo , Trofoblastos/patología , Factor de Necrosis Tumoral alfa/metabolismo
13.
Mol Cell Biol ; 26(2): 709-17, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16382160

RESUMEN

GPR103 is a G-protein-coupled receptor with reported expression in brain, heart, kidney, adrenal gland, retina, and testis. It encodes a 455-amino-acid protein homologous to neuropeptide FF2, neuropeptide Y2, and galanin GalR1 receptors. Its natural ligand was recently identified as 26RFa, a novel human RF-amide-related peptide with orexigenic activity. To identify the function of GPR103, we generated GPR103-deficient mice. Homozygous mutant mice were viable and fertile. Their body weight was undistinguishable from that of their wild-type littermates. Histological analysis revealed that GPR103-/- mice exhibited a thinned osteochondral growth plate, a thickening of trabecular branches, and a reduction in osteoclast number, suggestive of an early arrest of osteochondral bone formation. Microcomputed tomography confirmed the reduction in trabecular bone and connective tissue densities in GPR103 knockout animals. Whole-body radiography followed by morphometric analysis revealed a kyphosis in mutant animals. Reverse transcription-PCR analysis showed that GPR103 was expressed in human skull, mouse spine, and several osteoblast cell lines. Dexamethasone, a known inhibitor of osteoblast growth and inducer of osteoblast differentiation, inhibited GPR103 expression in human osteoblast primary cultures. Altogether, these results suggest that osteopenia in GPR103-/- mice may be mediated directly by the loss of GPR103 expression in bone.


Asunto(s)
Osteogénesis/fisiología , Receptores Acoplados a Proteínas G/fisiología , Animales , Densidad Ósea , Huesos/metabolismo , Encéfalo/metabolismo , Diferenciación Celular , Línea Celular , Femenino , Placa de Crecimiento/fisiología , Humanos , Riñón/metabolismo , Cifosis/genética , Cifosis/patología , Masculino , Ratones , Ratones Noqueados , Especificidad de Órganos , Osteoblastos/citología , Osteoblastos/metabolismo , Osteoclastos/citología , Osteoclastos/metabolismo , Ovariectomía , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Cráneo/metabolismo , Columna Vertebral/diagnóstico por imagen , Columna Vertebral/metabolismo , Tomografía Computarizada por Rayos X
14.
Endocrinology ; 149(9): 4519-26, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18499755

RESUMEN

G protein-coupled receptor 43 (GPR43) has been identified as a receptor for short-chain fatty acids that include acetate and propionate. A potential involvement of GPR43 in immune and inflammatory response has been previously suggested because its expression is highly enriched in immune cells. GPR43 is also expressed in a number of other tissues including adipocytes; however, the functional consequences of GPR43 activation in these other tissues are not clear. In this report, we focus on the potential functions of GPR43 in adipocytes. We show that adipocytes treated with GPR43 natural ligands, acetate and propionate, exhibit a reduction in lipolytic activity. This inhibition of lipolysis is the result of GPR43 activation, because this effect is abolished in adipocytes isolated from GPR43 knockout animals. In a mouse in vivo model, we show that the activation of GPR43 by acetate results in the reduction in plasma free fatty acid levels without inducing the flushing side effect that has been observed by the activation of nicotinic acid receptor, GPR109A. These results suggest a potential role for GPR43 in regulating plasma lipid profiles and perhaps aspects of metabolic syndrome.


Asunto(s)
Adipocitos/metabolismo , Ácidos Grasos no Esterificados/sangre , Lipólisis/genética , Receptores Acoplados a Proteínas G/fisiología , Células 3T3-L1 , Ácido Acético/farmacología , Adipocitos/efectos de los fármacos , Animales , Regulación hacia Abajo , Rubor/inducido químicamente , Lipólisis/efectos de los fármacos , Masculino , Síndrome Metabólico/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Obesos , Niacina/farmacología , Propionatos/farmacología , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo
15.
Mol Cell Biol ; 24(22): 9736-43, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15509778

RESUMEN

The physiological role of an orphan G protein-coupled receptor, LGR5, was investigated by targeted deletion of this seven-transmembrane protein containing a large N-terminal extracellular domain with leucine-rich repeats. LGR5 null mice exhibited 100% neonatal lethality characterized by gastrointestinal tract dilation with air and an absence of milk in the stomach. Gross and histological examination revealed fusion of the tongue to the floor of oral cavity in the mutant newborns and immunostaining of LGR5 expression in the epithelium of the tongue and in the mandible of the wild-type embryos. The observed ankyloglossia phenotype provides a model for understanding the genetic basis of this craniofacial defect in humans and an opportunity to elucidate the physiological role of the LGR5 signaling system during embryonic development.


Asunto(s)
Receptores Acoplados a Proteínas G/deficiencia , Lengua/anomalías , Animales , Animales Recién Nacidos , Secuencia de Bases , Anomalías Craneofaciales/genética , ADN/genética , Femenino , Tracto Gastrointestinal/anomalías , Marcación de Gen , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/fisiología
16.
Neuropsychopharmacology ; 31(1): 112-20, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15988472

RESUMEN

The biological effects of the melanin-concentrating hormone (MCH) are mediated by the melanin concentrating hormone receptor 1 (MCHR1) in mice. This receptor is enriched in brain areas that are involved in the modulation of mood and affect, suggesting that MCH-dependent signaling may influence neurobiological mechanisms underlying fear and anxiety processes. To test this, we have generated mice lacking functional MCHR1 and characterized phenotypic traits using a number of behavioral tests. Mice carrying a null mutation of the MCHR1 gene display anxiolytic-like behavior across a battery different behavioral paradigms commonly used to assess fear and anxiety responses in rodents: open field, elevated plus maze, social interaction, and stress-induced hyperthermia. The brain serotonin (5-HT) system is central to the control of mood- and anxiety-related processes. To examine the impact of MCHR1 receptor deletion on 5-HT neurotransmission, we used in vivo microdialysis in freely moving knockout and wild-type mice. Baseline dialysate 5-HT levels were significantly lower in MCHR1 knockout mice as compared with wild-type controls (9.53+/-0.24 fmol for wild types vs 6.91+/-0.36 fmol for knockouts) in the prefrontal cortex (PFC), one of the main target structures of the serotonergic system and one that is highly associated with the control of emotional processes. Moreover, forced swim increased 5-HT efflux in the PFC of wild-type but not MCHR1 knockout mice. In summary, we show that MCHR1 can modulate stress- and anxiety-like behaviors and suggest that this may be due to changes in serotonergic transmission in forebrain regions.


Asunto(s)
Ansiedad/genética , Receptores de la Hormona Hipofisaria/genética , Animales , Conducta Animal/fisiología , Química Encefálica/genética , Química Encefálica/fisiología , Cromatografía Líquida de Alta Presión , Ambiente , Espacio Extracelular/metabolismo , Fiebre/genética , Fiebre/fisiopatología , Genotipo , Relaciones Interpersonales , Ratones , Ratones Noqueados , Microdiálisis , Actividad Motora/fisiología , Fenotipo , Corteza Prefrontal/metabolismo , Serotonina/metabolismo , Estrés Psicológico/genética , Estrés Psicológico/fisiopatología
17.
Methods Mol Biol ; 1438: 153-75, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27150090

RESUMEN

Type 2 diabetes is a fast-growing epidemic in industrialized countries, associated with obesity, lack of physical exercise, aging, family history, and ethnic background. Diagnostic criteria are elevated fasting or postprandial blood glucose levels, a consequence of insulin resistance. Early intervention can help patients to revert the progression of the disease together with lifestyle changes or monotherapy. Systemic glucose toxicity can have devastating effects leading to pancreatic beta cell failure, blindness, nephropathy, and neuropathy, progressing to limb ulceration or even amputation. Existing treatments have numerous side effects and demonstrate variability in individual patient responsiveness. However, several emerging areas of discovery research are showing promises with the development of novel classes of antidiabetic drugs.The mouse has proven to be a reliable model for discovering and validating new treatments for type 2 diabetes mellitus. We review here commonly used methods to measure endpoints relevant to glucose metabolism which show good translatability to the diagnostic of type 2 diabetes in humans: baseline fasting glucose and insulin, glucose tolerance test, insulin sensitivity index, and body type composition. Improvements on these clinical values are essential for the progression of a novel potential therapeutic molecule through a preclinical and clinical pipeline.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Descubrimiento de Drogas , Hipoglucemiantes/uso terapéutico , Animales , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Prueba de Tolerancia a la Glucosa , Humanos , Insulina/metabolismo , Resistencia a la Insulina , Ratones , Obesidad/complicaciones
18.
F1000Res ; 5: 136, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27081476

RESUMEN

Gpr21 KO mice generated with Gpr21 KO ES cells obtained from Deltagen showed improved glucose tolerance and insulin sensitivity when fed a high fat diet. Further mRNA expression analysis revealed changes in Rabgap1 levels and raised the possibility that Rabgap1 gene may have been modified. To assess this hypothesis a new Gpr21 KO mouse line using TALENS technology was generated. Gpr21 gene deletion was confirmed by PCR and Gpr21 and Rabgap1 mRNA expression levels were determined by RT-PCR. The newly generated Gpr21 KO mice when fed a normal or high fat diet chow did not maintain their improved metabolic phenotype. In conclusion, Rabgap1 disturbance mRNA expression levels may have contributed to the phenotype of the originally designed Gpr21 KO mice.

19.
PLoS One ; 11(2): e0147254, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26840340

RESUMEN

Pancreatic amyloid formation by islet amyloid polypeptide (IAPP) is a hallmark pathological feature of type 2 diabetes. IAPP is stored in the secretory granules of pancreatic beta-cells and co-secreted with insulin to maintain glucose homeostasis. IAPP is innocuous under homeostatic conditions but imbalances in production or processing of IAPP may result in homodimer formation leading to the rapid production of cytotoxic oligomers and amyloid fibrils. The consequence is beta-cell dysfunction and the accumulation of proteinaceous plaques in and around pancreatic islets. Beta-site APP-cleaving enzyme 2, BACE2, is an aspartyl protease commonly associated with BACE1, a related homolog responsible for amyloid processing in the brain and strongly implicated in Alzheimer's disease. Herein, we identify two distinct sites of the mature human IAPP sequence that are susceptible to BACE2-mediated proteolytic activity. The result of proteolysis is modulation of human IAPP fibrillation and human IAPP protein degradation. These results suggest a potential therapeutic role for BACE2 in type 2 diabetes-associated hyperamylinaemia.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Ácido Aspártico Endopeptidasas/metabolismo , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Secuencia de Aminoácidos , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Animales , Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Línea Celular , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Humanos , Insulina/metabolismo , Insulina/farmacología , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Polipéptido Amiloide de los Islotes Pancreáticos/química , Polipéptido Amiloide de los Islotes Pancreáticos/genética , Espectrometría de Masas , Ratones , Datos de Secuencia Molecular , Mutación , Placa Amiloide/metabolismo , Proteolisis/efectos de los fármacos , Proteínas Recombinantes , Especificidad por Sustrato
20.
EBioMedicine ; 2(3): 214-24, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26151067

RESUMEN

The polarization of tissue resident macrophages toward the alternatively activated, anti-inflammatory M2 phenotype is believed to positively impact obesity and insulin resistance. Here we show that the soluble form of the extracellular domain (ECD) of C-type lectin-like receptor 2, CLEC2, regulates Kupffer cell polarization in the liver and improves glucose and lipid parameters in diabetic animal models. Over-expression of Fc-CLEC2(ECD) in mice via in vivo gene delivery, or injection of recombinant Fc-CLEC2(ECD) protein, results in a reduction of blood glucose and liver triglyceride levels and improves glucose tolerance. Furthermore, Fc-CLEC2(ECD) treatment improves cytokine profiles and increases both the M2 macrophage population and the genes involved in the oxidation of lipid metabolism in the liver. These data reveal a previously unidentified role for CLEC2 as a regulator of macrophage polarity, and establish CLEC2 as a promising therapeutic target for treatment of diabetes and liver disease.


Asunto(s)
Glucosa/metabolismo , Macrófagos del Hígado/metabolismo , Lectinas Tipo C/metabolismo , Metabolismo de los Lípidos/fisiología , Animales , Polaridad Celular , Hígado Graso/genética , Hígado Graso/metabolismo , Homeostasis/efectos de los fármacos , Humanos , Macrófagos del Hígado/citología , Macrófagos del Hígado/efectos de los fármacos , Lectinas Tipo C/genética , Metabolismo de los Lípidos/efectos de los fármacos , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos , Estructura Terciaria de Proteína , Receptores Fc/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Solubilidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA