Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell Physiol Biochem ; 45(3): 1165-1171, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29448249

RESUMEN

BACKGROUND/AIMS: Insulin-secreting islet ß-cells adapt to the insulin resistance associated with pregnancy by increasing functional ß-cell mass, but the placental signals involved in this process are not well defined. In the current study, we analysed expression of G-protein coupled receptor (GPCR) mRNAs in mouse islets and islet GPCR ligand mRNAs in placenta during pregnancy to generate an atlas of potential interactions between the placenta and ß-cells to inform future functional studies of islet adaptive responses to pregnancy. METHODS: Quantative RT-PCR arrays were used to measure mRNA expression levels of: (i) 342 GPCRs in islets from non-pregnant mice, and in islets isolated from mice on gestational days 12 and 18; (ii) 126 islet GPCR ligands in mouse placenta at gestational days 12 and 18. RESULTS: At gestational day 12, a time of rapid expansion of the ß-cell mass, 189 islet GPCR mRNAs were quantifiable, while 79 of the 126 known islet GPCR ligand mRNAs were detectable in placental extracts. Approximately half of the quantifiable placental GPCR ligand genes were of unknown function in ß-cells. The expression of some islet GPCR and placental ligand mRNAs varied during pregnancy, with altered expression of both GPCR and ligand mRNAs by gestational day 18. CONCLUSION: The current study has revealed numerous potential routes for interaction between the placenta and islets, and offers an atlas to inform further functional studies of their roles in adaptive responses to pregnancy, and in the regulation of the ß-cell mass.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Placenta/metabolismo , Animales , Femenino , Edad Gestacional , Ratones , Embarazo , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo
2.
J Endocrinol ; 260(2)2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-37997938

RESUMEN

During pregnancy the maternal pancreatic islets of Langerhans undergo adaptive changes to compensate for gestational insulin resistance. The lactogenic hormones are well established to play a key role in regulating the islet adaptation to pregnancy, and one of the mechanisms through which they act is through upregulating ß-cell serotonin production. During pregnancy islet serotonin levels are significantly elevated, where it is released from the ß-cells to drive the adaptive response through paracrine and autocrine effects. We have previously shown that placental kisspeptin (KP) also plays a role in promoting the elevated insulin secretion and ß-cell proliferation observed during pregnancy, although the precise mechanisms involved are unclear. In the present study we investigated the effects of KP on expression of pro-proliferative genes and serotonin biosynthesis within rodent islets. Whilst KP had limited effect on pro-proliferative gene expression at the time points tested, KP did significantly stimulate expression of the serotonin biosynthesis enzyme Tph-1. Furthermore, the islets of pregnant ß-cell-specific GPR54 knockdown mice were found to contain significantly fewer serotonin-positive ß-cells when compared to pregnant controls. Our previous studies suggested that reduced placental kisspeptin production, with consequent impaired kisspeptin-dependent ß-cell compensation, may be a factor in the development of GDM in humans. These current data suggest that, similar to the lactogenic hormones, KP may also contribute to serotonin biosynthesis and subsequent islet signalling during pregnancy. Furthermore, upregulation of serotonin biosynthesis may represent a common mechanism through which multiple signals might influence the islet adaptation to pregnancy.


Asunto(s)
Células Secretoras de Insulina , Islotes Pancreáticos , Humanos , Embarazo , Ratones , Femenino , Animales , Kisspeptinas/metabolismo , Insulina/metabolismo , Serotonina/metabolismo , Placenta/metabolismo , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Prolactina/metabolismo
3.
Diabetologia ; 56(11): 2467-76, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23979485

RESUMEN

AIMS/HYPOTHESIS: Chemokine (C-C motif) ligand 5 (CCL5) acts at C-C chemokine receptors (CCRs) to promote immune cell recruitment to sites of inflammation, but is also an agonist at G-protein-coupled receptor 75 (GPR75), which has very limited homology with CCRs. GPR75 is coupled to Gq to elevate intracellular calcium, so we investigated whether islets express this receptor and whether its activation by CCL5 increases beta cell calcium levels and insulin secretion. METHODS: Islet CCL5 receptor mRNA expression was measured by quantitative RT-PCR and GPR75 was detected in islets by western blotting and immunohistochemistry. In some experiments GPR75 was downregulated by transient transfection with small interfering RNA. Real-time changes in intracellular calcium were determined by single-cell microfluorimetry. Dynamic insulin secretion from perifused islets was quantified by radioimmunoassay. Glucose homeostasis in lean and obese mice was determined by measuring glucose and insulin tolerance, and insulin secretion in vivo. RESULTS: Mouse and human islets express GPR75 and its ligand CCL5. Exogenous CCL5 reversibly increased intracellular calcium in beta cells via GPR75, this phenomenon being dependent on phospholipase C activation and calcium influx. CCL5 also stimulated insulin secretion from mouse and human islets in vitro, and improved glucose tolerance in lean mice and in a mouse model of hyperglycaemia and insulin resistance (ob/ob). The improvement in glucose tolerance was associated with enhanced insulin secretion in vivo, without changes in insulin sensitivity. CONCLUSIONS/INTERPRETATION: Although CCL5 is implicated in the pathogenesis of diabetes through activation of CCRs, it has beneficial effects on beta cells through GPR75 activation.


Asunto(s)
Glucosa/metabolismo , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Quimiocina CCL5/metabolismo , Homeostasis/genética , Homeostasis/fisiología , Humanos , Inmunohistoquímica , Secreción de Insulina , Masculino , Ratones , Ratones Endogámicos ICR , Receptores Acoplados a Proteínas G/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
4.
Diabetologia ; 56(11): 2477-86, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23900510

RESUMEN

AIMS/HYPOTHESIS: The stress-activated nuclear protein transcription regulator 1 (NUPR1) is induced in response to glucose and TNF-α, both of which are elevated in type 2 diabetes, and Nupr1 has been implicated in cell proliferation and apoptosis cascades. We used Nupr1(-/-) mice to study the role of Nupr1 in glucose homeostasis under normal conditions and following maintenance on a high-fat diet (HFD). METHODS: Glucose homeostasis in vivo was determined by measuring glucose tolerance, insulin sensitivity and insulin secretion. Islet number, morphology and beta cell area were assessed by immunofluorescence and morphometric analysis, and islet cell proliferation was quantified by analysis of BrdU incorporation. Islet gene expression was measured by gene arrays and quantitative RT-PCR, and gene promoter activities were monitored by measuring luciferase activity. RESULTS: Nupr1(-/-) mice had increased beta cell mass as a consequence of enhanced islet cell proliferation. Nupr1-dependent suppression of beta cell Ccna2 and Tcf19 promoter activities was identified as a mechanism through which Nupr1 may regulate beta cell cycle progression. Nupr1(-/-) mice maintained on a normal diet were mildly insulin resistant, but were normoglycaemic with normal glucose tolerance because of compensatory increases in basal and glucose-induced insulin secretion. Nupr1 deletion was protective against HFD-induced obesity, insulin resistance and glucose intolerance. CONCLUSIONS/INTERPRETATION: Inhibition of NUPR1 expression or activity has the potential to protect against the metabolic defects associated with obesity and type 2 diabetes.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Intolerancia a la Glucosa/metabolismo , Células Secretoras de Insulina/metabolismo , Proteínas de Neoplasias/metabolismo , Animales , Western Blotting , Proteínas de Unión al ADN/genética , Femenino , Intolerancia a la Glucosa/genética , Humanos , Inmunohistoquímica , Células Secretoras de Insulina/citología , Masculino , Ratones , Ratones Noqueados , Proteínas de Neoplasias/genética
5.
Cytotherapy ; 15(4): 449-59, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23321626

RESUMEN

BACKGROUND AIMS: We recently showed that co-transplantation of mesenchymal stromal cells (MSCs) improves islet function and revascularization in vivo. Pre-transplant islet culture is associated with the loss of islet cells. MSCs may enhance islet cell survival or function by direct cell contact mechanisms and soluble mediators. We investigated the capacity of MSCs to improve islet cell survival or ß-cell function in vitro using direct and indirect contact islet-MSC configurations. We also investigated whether pre-culturing islets with MSCs improves islet transplantation outcome. METHODS: The effect of pre-culturing islets with MSCs on islet function in vitro was investigated by measuring glucose-stimulated insulin secretion. The endothelial cell density of fresh islets and islets cultured with or without MSCs was determined by immunohistochemistry. The efficacy of transplanted islets was tested in vivo using a syngeneic streptozotocin-diabetic minimal islet mass model. Graft function was investigated by monitoring blood glucose concentrations. RESULTS: Indirect islet-MSC co-culture configurations did not improve islet function in vitro. Pre-culturing islets using a direct contact MSC monolayer configuration improved glucose-stimulated insulin secretion in vitro, which correlated with superior islet graft function in vivo. MSC pre-culture had no effect on islet endothelial cell number in vitro or in vivo. CONCLUSIONS: Pre-culturing islets with MSCs using a direct contact configuration maintains functional ß-cell mass in vitro and the capacity of cultured islets to reverse hyperglycemia in diabetic mice.


Asunto(s)
Diabetes Mellitus Experimental/terapia , Trasplante de Islotes Pancreáticos , Islotes Pancreáticos/citología , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Animales , Supervivencia Celular , Células Cultivadas , Técnicas de Cocultivo , Diabetes Mellitus Experimental/metabolismo , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL
6.
Redox Biol ; 67: 102883, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37774548

RESUMEN

In adverse pregnancy a perturbed redox environment is associated with abnormal early-life cardiovascular development and function. Previous studies have noted alterations in the expression and/or activity of Nuclear Factor E2 Related Factor 2 (NRF2) and its antioxidant targets during human gestational diabetic (GDM) pregnancy, however to our knowledge the functional role of NRF2 in fetal 'priming' of cardiovascular dysfunction in obese and GDM pregnancy has not been investigated. Using a murine model of obesity-induced glucose dysregulated pregnancy, we demonstrate that NRF2 activation by maternal sulforaphane (SFN) supplementation normalizes NRF2-linked NQO1, GCL and CuZnSOD expression in maternal and fetal liver placental and fetal heart tissue by gestational day 17.5. Activation of NRF2 in utero in wild type but not NRF2 deficient mice improved markers of placental efficiency and partially restored fetal growth. SFN supplementation was associated with reduced markers of fetal cardiac oxidative stress, including Nox2 and 3-nitrotyrosine, as well as attenuation of cardiac mass and cardiomyocyte area in male offspring by postnatal day 52 and improved vascular function in male and female offspring by postnatal day 98. Our findings are the first to highlight the functional consequences of NRF2 modulation in utero on early-life cardiovascular function in offspring, demonstrating that activation of NRF2 affords cardiovascular protection in offspring of pregnancies affected by redox dysregulation.


Asunto(s)
Factor 2 Relacionado con NF-E2 , Placenta , Humanos , Ratones , Masculino , Femenino , Embarazo , Animales , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Placenta/metabolismo , Oxidación-Reducción , Isotiocianatos/farmacología , Obesidad/metabolismo , Estrés Oxidativo , Miocitos Cardíacos/metabolismo
7.
Diabetes ; 71(4): 837-852, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35073578

RESUMEN

Serum progesterone sulfates were evaluated in the etiology of gestational diabetes mellitus (GDM). Serum progesterone sulfates were measured using ultra-performance liquid chromatography-tandem mass spectrometry in four patient cohorts: 1) the Hyperglycemia and Adverse Pregnancy Outcomes study; 2) London-based women of mixed ancestry and 3) U.K.-based women of European ancestry with or without GDM; and 4) 11-13 weeks pregnant women with BMI ≤25 or BMI ≥35 kg/m2 with subsequent uncomplicated pregnancies or GDM. Glucose-stimulated insulin secretion (GSIS) was evaluated in response to progesterone sulfates in mouse islets and human islets. Calcium fluorescence was measured in HEK293 cells expressing transient receptor potential cation channel subfamily M member 3 (TRPM3). Computer modeling using Molecular Operating Environment generated three-dimensional structures of TRPM3. Epiallopregnanolone sulfate (PM5S) concentrations were reduced in GDM (P < 0.05), in women with higher fasting plasma glucose (P < 0.010), and in early pregnancy samples from women who subsequently developed GDM with BMI ≥35 kg/m2 (P < 0.05). In islets, 50 µmol/L PM5S increased GSIS by at least twofold (P < 0.001); isosakuranetin (TRPM3 inhibitor) abolished this effect. PM5S increased calcium influx in TRPM3-expressing HEK293 cells. Computer modeling and docking showed identical positioning of PM5S to the natural ligand in TRPM3. PM5S increases GSIS and is reduced in GDM serum. The activation of GSIS by PM5S is mediated by TRPM3 in both mouse and human islets.


Asunto(s)
Diabetes Gestacional , Canales Catiónicos TRPM , Animales , Glucemia/metabolismo , Calcio/metabolismo , Femenino , Células HEK293 , Humanos , Insulina/metabolismo , Secreción de Insulina , Ratones , Embarazo , Progesterona , Sulfatos/metabolismo
8.
Methods Mol Biol ; 2128: 181-205, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32180194

RESUMEN

Beta-cell-specific transgenic mice provide an invaluable model for dissecting the direct signaling mechanisms involved in regulating beta-cell structure and function. Furthermore, generating novel transgenic models is now easier and more cost-effective than ever, thanks to exciting novel approaches such as CRISPR.Here, we describe the commonly used approaches for generating and maintaining beta-cell-specific transgenic models and some of the considerations involved in their use. This includes the use of different beta-cell-specific promoters (e.g., pancreatic and duodenal homeobox factor 1 (Pdx1), rat insulin 2 promoter (RIP), and mouse insulin 1 promoter (MIP)) to drive site-specific recombinase technology. Important considerations during selection include level and uniformity of expression in the beta-cell population, ectopic transgene expression, and the use of inducible models.This chapter provides a guide to the procurement, generation, and maintenance of a beta-cell-specific transgene colony from preexisting Cre and loxP mouse strains, providing methods for crossbreeding and genotyping, as well as subsequent maintenance and, in the case of inducible models, transgenic induction.


Asunto(s)
Técnicas de Inactivación de Genes/métodos , Ingeniería Genética/métodos , Técnicas de Genotipaje/métodos , Células Secretoras de Insulina , Integrasas/genética , Animales , Cruzamientos Genéticos , Expresión Génica , Genes Reporteros , Ratones , Ratones Transgénicos , Especificidad de Órganos/genética , Regiones Promotoras Genéticas
9.
J Endocrinol ; 245(2): 247-257, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32106091

RESUMEN

The corticotropin-releasing hormone (CRH) family of peptides, including urocortin (UCN) 1, 2 and 3, are established hypothalamic neuroendocrine peptides, regulating the physiological and behaviour responses to stress indirectly, via the hypothalamic-pituitary-adrenal (HPA) axis. More recently, these peptides have been implicated in diverse roles in peripheral organs through direct signalling, including in placental and pancreatic islet physiology. CRH has been shown to stimulate insulin release through activation of its cognate receptors, CRH receptor 1 (CRHR1) and 2. However, the physiological significance of this is unknown. We have previously reported that during mouse pregnancy, expression of CRH peptides increase in mouse placenta suggesting that these peptides may play a role in various biological functions associated with pregnancy, particularly the pancreatic islet adaptations that occur in the pregnant state to compensate for the physiological increase in maternal insulin resistance. In the current study, we show that mouse pregnancy is associated with increased circulating levels of UCN2 and that when we pharmacologically block endogenous CRHR signalling in pregnant mice, impairment of glucose tolerance is observed. This effect on glucose tolerance was comparable to that displayed with specific CRHR2 blockade and not with specific CRHR1 blockade. No effects on insulin sensitivity or the proliferative capacity of ß-cells were detected. Thus, CRHR2 signalling appears to be involved in ß-cell adaptive responses to pregnancy in the mouse, with endogenous placental UCN2 being the likely signal mediating this.


Asunto(s)
Hormona Liberadora de Corticotropina/metabolismo , Resistencia a la Insulina/fisiología , Células Secretoras de Insulina/fisiología , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Transducción de Señal/fisiología , Urocortinas/metabolismo , Animales , Femenino , Sistema Hipotálamo-Hipofisario/metabolismo , Ratones , Sistema Hipófiso-Suprarrenal/metabolismo , Placenta/metabolismo , Embarazo
10.
Semin Reprod Med ; 37(3): 141-146, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31869842

RESUMEN

Kisspeptin has well-established critical roles in the control of reproduction and fertility. Recently, evidence has emerged that suggests kisspeptin may have additional roles in the regulation of glucose homeostasis. Conflicting reports on the effects of kisspeptin on insulin secretion in animal models have been published, which cannot be fully accounted for by the different kisspeptin isoforms and range of kisspeptin doses used in these studies. Human studies have demonstrated associations between circulating kisspeptin levels and measures of insulin secretion and insulin resistance; and the only published interventional study has confirmed kisspeptin enhances glucose-stimulated insulin secretion in humans. Further studies are required to elucidate the mechanisms underlying the effects of kisspeptin on the pancreatic ß-cell and to determine the therapeutic potential of kisspeptin receptor agonist in the treatment of disorders of glucose homeostasis.


Asunto(s)
Trastornos del Metabolismo de la Glucosa/etiología , Glucosa/metabolismo , Kisspeptinas/fisiología , Animales , Fertilidad/efectos de los fármacos , Fertilidad/genética , Trastornos del Metabolismo de la Glucosa/tratamiento farmacológico , Trastornos del Metabolismo de la Glucosa/genética , Homeostasis/efectos de los fármacos , Homeostasis/genética , Humanos , Insulina/metabolismo , Secreción de Insulina/efectos de los fármacos , Secreción de Insulina/genética , Kisspeptinas/farmacología , Kisspeptinas/uso terapéutico , Receptores de Kisspeptina-1/agonistas , Reproducción/efectos de los fármacos , Reproducción/genética
11.
JCI Insight ; 4(20)2019 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-31619585

RESUMEN

During pregnancy the maternal pancreatic islets of Langerhans undergo adaptive changes to compensate for gestational insulin resistance. Kisspeptin has been shown to stimulate insulin release, through its receptor, GPR54. The placenta releases high levels of kisspeptin into the maternal circulation, suggesting a role in modulating the islet adaptation to pregnancy. In the present study we show that pharmacological blockade of endogenous kisspeptin in pregnant mice resulted in impaired glucose homeostasis. This glucose intolerance was due to a reduced insulin response to glucose as opposed to any effect on insulin sensitivity. A ß cell-specific GPR54-knockdown mouse line was found to exhibit glucose intolerance during pregnancy, with no phenotype observed outside of pregnancy. Furthermore, in pregnant women circulating kisspeptin levels significantly correlated with insulin responses to oral glucose challenge and were significantly lower in women with gestational diabetes (GDM) compared with those without GDM. Thus, kisspeptin represents a placental signal that plays a physiological role in the islet adaptation to pregnancy, maintaining maternal glucose homeostasis by acting through the ß cell GPR54 receptor. Our data suggest reduced placental kisspeptin production, with consequent impaired kisspeptin-dependent ß cell compensation, may be a factor in the development of GDM in humans.


Asunto(s)
Diabetes Gestacional/fisiopatología , Intolerancia a la Glucosa/fisiopatología , Células Secretoras de Insulina/fisiología , Kisspeptinas/metabolismo , Placenta/metabolismo , Adaptación Fisiológica , Adulto , Animales , Diabetes Gestacional/sangre , Diabetes Gestacional/diagnóstico , Diabetes Gestacional/metabolismo , Femenino , Glucosa/análisis , Glucosa/metabolismo , Intolerancia a la Glucosa/sangre , Intolerancia a la Glucosa/diagnóstico , Intolerancia a la Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Humanos , Insulina/administración & dosificación , Insulina/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Kisspeptinas/antagonistas & inhibidores , Kisspeptinas/sangre , Ratones , Ratones Noqueados , Ratones Transgénicos , Placenta/efectos de los fármacos , Circulación Placentaria/fisiología , Embarazo , Receptores de Kisspeptina-1/genética , Receptores de Kisspeptina-1/metabolismo
12.
Curr Opin Pharmacol ; 43: 59-65, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30199758

RESUMEN

Pregnancy involves a progressive increase in insulin resistance and the ß-cells must adapt to compensate and prevent gestational diabetes (GDM). In this review we discuss the evidence for placental peptides, including placental lactogen, hepatocyte growth factor, adiponectin and leptin, playing a role in the islet adaptation to pregnancy. The difficulties of translating data from rodent models into human pregnancy are covered and we summarise studies investigating associations between serum placental peptides and GDM risk. In conclusion, current data support important roles for placental peptides interacting to support ß-cells during pregnancy, however mechanisms involved in humans are unclear. Further work in humans is required, but placental peptides have clinical potential from both a diagnostic and therapeutic perspective.


Asunto(s)
Glucemia/metabolismo , Diabetes Gestacional/metabolismo , Islotes Pancreáticos/metabolismo , Péptidos/metabolismo , Placenta/metabolismo , Adaptación Fisiológica , Adipoquinas/metabolismo , Animales , Biomarcadores/sangre , Glucemia/efectos de los fármacos , Diabetes Gestacional/sangre , Diabetes Gestacional/tratamiento farmacológico , Diabetes Gestacional/fisiopatología , Femenino , Factor de Crecimiento de Hepatocito/metabolismo , Humanos , Hipoglucemiantes/uso terapéutico , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/fisiopatología , Péptidos/uso terapéutico , Placenta/fisiopatología , Lactógeno Placentario/metabolismo , Embarazo , Transducción de Señal
14.
J Endocrinol ; 191(2): 399-405, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17088409

RESUMEN

The mechanisms underlying menopausal hot flushes are poorly understood, although it is generally assumed they result from disturbances of thermoregulatory centres in the hypothalamus. 8-Prenylnaringenin (8-PN) has been identified as a potent phytoestrogen in hops (Humulus lupulus) and there are claims that hop-containing preparations can reduce hot flushes. We have investigated the site of action of 8-PN in a rat model of menopausal hot flushes, in which the tail skin temperature (TST) is increased after oestrogen withdrawal induced by ovariectomy. Daily s.c. administration of either 17beta-oestradiol (E2; 4 microg/kg) or 8-PN (400 microg/kg) significantly reduced the elevated TST after 2 days of treatment. Subcutaneous co-administration of either E2 or 8-PN with the oestrogen receptor (ER) antagonist, ICI 182,780 (200 microg/kg), which is thought not to cross the blood-brain barrier, completely blocked the effect of E2 and 8-PN on TST. The ERalpha- and ERbeta-specific agonists, 4,4',4''-(4-propyl-[1H]-pyrazole-1,3,5-triyl)trisphenol (100 microg/kg) and 2,3-bis(4-hydroxyphenyl)-propionitrile (60 microg/kg) respectively, both significantly reversed the raised TST in ovariectomised rats. These observations suggest that the regulation of the vasomotor response by oestrogens and phytoestrogens is mediated, at least in part, by peripheral mechanisms involving both ERalpha and ERbeta.


Asunto(s)
Flavanonas/uso terapéutico , Sofocos/tratamiento farmacológico , Humulus , Fitoterapia , Temperatura Cutánea/efectos de los fármacos , Animales , Cerveza , Estradiol/análogos & derivados , Estradiol/farmacología , Estradiol/uso terapéutico , Antagonistas de Estrógenos/farmacología , Receptor alfa de Estrógeno/agonistas , Receptor beta de Estrógeno/agonistas , Femenino , Fulvestrant , Modelos Animales , Nitrilos/farmacología , Ovariectomía , Fenoles/farmacología , Pirazoles/farmacología , Ratas , Ratas Wistar , Cola (estructura animal)
15.
Biochem Pharmacol ; 99: 1-10, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26432954

RESUMEN

Diabetes mellitus is a lifelong, metabolic disease that is characterised by an inability to maintain normal glucose homeostasis. There are several different forms of diabetes, however the two most common are Type 1 and Type 2 diabetes. Type 1 diabetes is caused by the autoimmune destruction of pancreatic beta cells and a subsequent lack of insulin production, whilst Type 2 diabetes is due to a combination of both insulin resistance and an inability of the beta cells to compensate adequately with increased insulin release. Animal models are increasingly being used to elucidate the mechanisms underlying both Type 1 and Type 2 diabetes as well as to identify and refine novel treatments. However, a wide range of different animal models are currently in use. The majority of these models are suited to addressing certain specific aspects of diabetes research, but may be of little use in other studies. All have pros and cons, and selecting an appropriate model for addressing a specific question is not always a trivial task and will influence the study results and their interpretation. Thus, as the number of available animal models increases it is important to consider the potential roles of these models in the many different aspects of diabetes research. This review gathers information on the currently used experimental animal models of both Type 1 and Type 2 diabetes and evaluates their advantages and disadvantages for research purposes and details the factors that should be taken into account in their use.


Asunto(s)
Diabetes Mellitus Tipo 1/sangre , Diabetes Mellitus Tipo 1/terapia , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/terapia , Modelos Animales de Enfermedad , Animales , Diabetes Mellitus Tipo 1/etiología , Diabetes Mellitus Tipo 2/etiología , Humanos , Hipoglucemiantes/uso terapéutico , Resistencia a la Insulina/fisiología , Obesidad/sangre , Obesidad/complicaciones , Obesidad/terapia , Conducta de Reducción del Riesgo , Resultado del Tratamiento
16.
Endocrinology ; 146(1): 318-22, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15388651

RESUMEN

Corticotropin-releasing factor (CRF) has been implicated as an important mediator of stress-induced inhibition of reproduction. The role of specific CRF receptor subtypes in this effect is unknown, and in the current study, we investigated the role of the CRF-R2 receptor in stress-mediated suppression of pulsatile LH section. Ovariectomized rats with sc 17beta-estradiol capsules were implanted with intracerebroventricular (i.c.v.) and i.v. cannulae. Blood samples (25 microl) were collected every 5 min for 5 h for LH measurement. Central administration of urocortin II (0.24, 2.4, 24, or 240 nmol, i.c.v.), which selectively binds to CRF-R2, resulted in a dose-dependent suppression of LH pulses. Restraint stress (1 h) induced a profound suppression of pulsatile LH secretion and astressin2-B, a selective CRF-R2 antagonist (28 nmol i.c.v., 10-min prerestraint), was effective in blocking this inhibitory response. These findings suggest that CRF-R2 mediates, at least in part, restraint stress-induced inhibition of LH pulses and may play a pivotal role in the normal physiological response of stress-induced suppression of the hypothalamic GnRH pulse generator and hence the reproductive system.


Asunto(s)
Hormona Luteinizante/antagonistas & inhibidores , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Estrés Fisiológico/metabolismo , Animales , Hormona Liberadora de Corticotropina/administración & dosificación , Hormona Liberadora de Corticotropina/farmacología , Relación Dosis-Respuesta a Droga , Femenino , Inyecciones Intraventriculares , Hormona Luteinizante/metabolismo , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/farmacología , Ratas , Ratas Wistar , Receptores de Hormona Liberadora de Corticotropina/agonistas , Restricción Física , Estrés Fisiológico/etiología , Urocortinas
17.
J Endocrinol ; 225(1): 19-26, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25630331

RESUMEN

The inhibitory effect of somatostatin (SST) on insulin secretion in vivo is attributed to a direct effect on pancreatic beta cells, but this is inconsistent with some in vitro results in which exogenous SST is ineffective in inhibiting secretion from isolated islets. We therefore investigated whether insulin secretion from the pancreatic islets may partly be regulated by an indirect effect of SST mediated via the CNS. Islet hormone secretion was assessed in vitro by perifusion and static incubations of isolated islets and in vivo by i.v. or i.c.v. administration of the SST analogue BIM23014C with an i.v. glucose challenge to conscious, chronically catheterised rats. Hormone content of samples was assessed by ELISA or RIA and blood glucose levels using a glucose meter. Exogenous SST14/SST28 or BIM23014C did not inhibit the release of insulin from isolated rodent islets in vitro, whereas peripheral i.v. administration of BIM23014C (7.5 µg) with glucose (1 g/kg) led to decreased plasma insulin content (2.3±0.5 ng insulin/ml versus 4.5±0.5 ng/ml at t=5 min, P<0.001) and elevated blood glucose levels compared with those of the controls (29.19±1.3 mmol/l versus 23.5±1.7 mmol/l, P<0.05). In contrast, central i.c.v. injection of BIM23014C (0.75 µg) had no significant effect on either plasma insulin (3.3±0.4 ng/ml, P>0.05) or blood glucose levels (23.5±1.7 mmol/l, P>0.05) although i.v. administration of this dose increased blood glucose concentrations (32.3±0.7 mmol/l, P<0.01). BIM23014C did not measurably alter plasma glucagon, SST, GLP1 or catecholamine levels whether injected i.v. or i.c.v. These results indicate that SST does not suppress insulin secretion by a centrally mediated effect but acts peripherally on islet cells.


Asunto(s)
Sistema Nervioso Central/fisiología , Insulina/metabolismo , Somatostatina/farmacología , Animales , Glucemia , Vías de Administración de Medicamentos , Masculino , Ratones , Ratones Endogámicos ICR , Péptidos Cíclicos/farmacología , Ratas , Ratas Wistar , Somatostatina/administración & dosificación , Somatostatina/análogos & derivados
18.
Endocrinology ; 145(4): 1556-63, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-14736738

RESUMEN

In addition to its role as a potent vasodilator, calcitonin gene-related peptide (CGRP) is centrally involved in a variety of stress responses, including activation of the hypothalamo-pituitary-adrenocortical axis. It is well known that stress suppresses the activity of the hypothalamic GnRH pulse generator, the central regulator of LH and FSH pulses, resulting in reproductive dysfunction. The aim of this study was to test the hypothesis that CGRP has a critical role in mediating stress-induced suppression of pulsatile LH secretion in the rat. Ovariectomized rats were implanted with intracerebroventricular and iv cannulae. Central administration of CGRP (75 pmol-1.2 nmol) into the lateral cerebral ventricle resulted in a profound, dose-dependent suppression of LH pulses, which was reversed by a CGRP receptor antagonist (CGRP(8-37),1 nmol). Although the site of action of CGRP remains to be established, the induction of c-Fos expression in the preoptic area and hypothalamic paraventricular nucleus might suggest an involvement of these brain regions. Intravenous administration of CGRP did not affect LH pulses. Coadministration (intracerebroventricular) of CGRP (400 pmol) with a CRH antagonist (alpha-helical CRF(9-41), 26 nmol) partly blocked the CGRP-induced suppression of LH pulses. Furthermore, CGRP(8-37) (1 nmol) completely blocked hypoglycemic stress-induced suppression of LH pulses. These results suggest that the suppression of pulsatile LH secretion by central administration of CGRP may be mediated in part by CRH, and that CGRP may play a pivotal role in the normal physiological response of stress-induced suppression of the hypothalamic GnRH pulse generator, and hence the reproductive system.


Asunto(s)
Péptido Relacionado con Gen de Calcitonina/fisiología , Hormona Liberadora de Gonadotropina/antagonistas & inhibidores , Hipotálamo/metabolismo , Hormona Luteinizante/metabolismo , Estrés Fisiológico/metabolismo , Animales , Péptido Relacionado con Gen de Calcitonina/farmacología , Antagonistas del Receptor Peptídico Relacionado con el Gen de la Calcitonina , Femenino , Hormona Liberadora de Gonadotropina/metabolismo , Hormona Liberadora de Gonadotropina/farmacología , Hormona Liberadora de Gonadotropina/fisiología , Hipoglucemiantes/farmacología , Insulina/farmacología , Fragmentos de Péptidos/farmacología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Wistar
19.
J Endocrinol ; 222(3): G13-25, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25056117

RESUMEN

The pathophysiology of diabetes as a disease is characterised by an inability to maintain normal glucose homeostasis. In type 1 diabetes, this is due to autoimmune destruction of the pancreatic ß-cells and subsequent lack of insulin production, and in type 2 diabetes it is due to a combination of both insulin resistance and an inability of the ß-cells to compensate adequately with increased insulin release. Animal models, in particular genetically modified mice, are increasingly being used to elucidate the mechanisms underlying both type 1 and type 2 diabetes, and as such the ability to study glucose homeostasis in vivo has become an essential tool. Several techniques exist for measuring different aspects of glucose tolerance and each of these methods has distinct advantages and disadvantages. Thus the appropriate methodology may vary from study to study depending on the desired end-points, the animal model, and other practical considerations. This review outlines the most commonly used techniques for assessing glucose tolerance in rodents and details the factors that should be taken into account in their use. Representative scenarios illustrating some of the practical considerations of designing in vivo experiments for the measurement of glucose homeostasis are also discussed.


Asunto(s)
Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Glucosa/metabolismo , Animales , Glucemia/metabolismo , Modelos Animales de Enfermedad , Ayuno/metabolismo , Femenino , Glucagón/sangre , Prueba de Tolerancia a la Glucosa/métodos , Guías como Asunto , Homeostasis , Humanos , Insulina/sangre , Resistencia a la Insulina , Masculino , Ratones , Fenotipo , Embarazo , Embarazo en Diabéticas/metabolismo , Ratas , Somatostatina/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA