Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Am J Obstet Gynecol ; 230(1): 89.e1-89.e12, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37481152

RESUMO

BACKGROUND: Asymptomatic isthmic contractions are a frequent physiological phenomenon in pregnancy, sometimes triggered by bladder voiding. They can interfere with proper cervical length assessment and may lead to false images of placenta previa. However, there is limited research on the prevalence and characteristics of these contractions. OBJECTIVE: This study aimed to determine the prevalence and characteristics of isthmic contractions after bladder voiding in the second trimester of pregnancy, to evaluate their effect on cervical length assessment, and to propose a new method for the objective assessment of the presence and intensity of isthmic contractions. STUDY DESIGN: In this prospective observational study, long videos of the uterine cervix were recorded in 30 singleton pregnancies during the second trimester of pregnancy after bladder voiding. Isthmic length and cervicoisthmic length changes were assessed over time. The isthmic length was measured using a new approach, which involved calculating the distance from the base of the cervix to the internal os, including the isthmus. RESULTS: Isthmic contractions were observed in 43% of pregnant women (95% confidence interval, 26%-62%) after bladder voiding. The median time for complete isthmus relaxation was 19.7 minutes (95% confidence interval, 15.0 to not available). No substantial differences in maternal characteristics were found between individuals with and without contractions. The proposed method for measuring isthmic length provided an objective assessment of the presence and intensity of isthmic contractions. A cutoff of 18 mm in isthmic length allowed for the distinction of pregnant women presenting a contraction. In addition, the study identified a characteristic undulatory pattern in the relaxation of the isthmus in half of the cases with contractions. CONCLUSION: Isthmic contractions are a common occurrence after bladder voiding in the second trimester of pregnancy and may interfere with proper cervical length assessment. We recommend performing cervical assessment at least 20 minutes after bladder voiding to reduce the risk of bias in cervical length measurement and to avoid false images of placenta previa. The new method for measuring isthmic length provides an objective way to assess the presence and intensity of isthmic contractions. Further research is needed to understand the role of isthmic contractions in the physiology of pregnancy and birth.


Assuntos
Placenta Prévia , Nascimento Prematuro , Gravidez , Feminino , Humanos , Útero/diagnóstico por imagem , Colo do Útero/diagnóstico por imagem , Contração Uterina , Ultrassonografia , Medida do Comprimento Cervical/métodos , Segundo Trimestre da Gravidez , Nascimento Prematuro/epidemiologia
2.
Exp Eye Res ; 212: 108801, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34688624

RESUMO

Endostatin, a naturally cleaved fragment of type XVIII collagen with antiangiogenic activity, has been involved in the regulation of neovascularization during diabetic retinopathy. Here, the intracellular distribution of endostatin in healthy mouse and human neuroretinas has been analyzed. In addition, to study the effect of experimental hyperglycemia on retinal endostatin, the db/db mouse model has been used. Endostatin protein expression in mouse and human retinas was studied by immunofluorescence and Western blot, and compared with db/db mice. Eye fundus angiography, histology, and immunofluorescence were used to visualize mouse retinal and intravitreal vessels. For the first time, our results revealed the presence of endostatin in neurons of mouse and human retinas. Endostatin was mainly expressed in bipolar cells and photoreceptors, in contrast to the optic disc, where endostatin expression was undetectable. Diabetic mice showed a reduction of endostatin in their retinas associated with the appearance of intravitreal vessels at the optic disc in 50% of db/db mice. Intravitreal vessels showed GFAP positive neuroglia sheath, basement membrane thickening by collagen IV deposition, and presence of MMP-2 and MMP-9 in the vascular wall. All together, these results point that decreased retinal endostatin during experimental diabetes is associated with optic disc intravitreal vascularization. Based on their phenotype, these intravitreal vessels could be neovessels. However, it cannot be ruled out the possibility that they may also represent persistent hyaloid vessels.


Assuntos
Diabetes Mellitus Experimental , Retinopatia Diabética/metabolismo , Endostatinas/metabolismo , Disco Óptico/metabolismo , Neovascularização Retiniana/metabolismo , Vasos Retinianos/metabolismo , Corpo Vítreo/irrigação sanguínea , Animais , Retinopatia Diabética/diagnóstico , Humanos , Masculino , Camundongos , Disco Óptico/patologia , Neovascularização Retiniana/patologia , Neovascularização Retiniana/prevenção & controle , Vasos Retinianos/diagnóstico por imagem , Corpo Vítreo/diagnóstico por imagem
3.
J Biol Chem ; 290(27): 16772-85, 2015 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-25971976

RESUMO

The human insulin-like growth factor 2 (IGF2) and insulin genes are located within the same genomic region. Although human genomic studies have demonstrated associations between diabetes and the insulin/IGF2 locus or the IGF2 mRNA-binding protein 2 (IGF2BP2), the role of IGF2 in diabetes pathogenesis is not fully understood. We previously described that transgenic mice overexpressing IGF2 specifically in ß-cells (Tg-IGF2) develop a pre-diabetic state. Here, we characterized the effects of IGF2 on ß-cell functionality. Overexpression of IGF2 led to ß-cell dedifferentiation and endoplasmic reticulum stress causing islet dysfunction in vivo. Both adenovirus-mediated overexpression of IGF2 and treatment of adult wild-type islets with recombinant IGF2 in vitro further confirmed the direct implication of IGF2 on ß-cell dysfunction. Treatment of Tg-IGF2 mice with subdiabetogenic doses of streptozotocin or crossing these mice with a transgenic model of islet lymphocytic infiltration promoted the development of overt diabetes, suggesting that IGF2 makes islets more susceptible to ß-cell damage and immune attack. These results indicate that increased local levels of IGF2 in pancreatic islets may predispose to the onset of diabetes. This study unravels an unprecedented role of IGF2 on ß-cells function.


Assuntos
Diabetes Mellitus/genética , Fator de Crescimento Insulin-Like II/genética , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Animais , Desdiferenciação Celular , Linhagem Celular Tumoral , Diabetes Mellitus/metabolismo , Diabetes Mellitus/fisiopatologia , Humanos , Insulina/metabolismo , Fator de Crescimento Insulin-Like II/metabolismo , Ilhotas Pancreáticas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Ratos
4.
Mol Ther Methods Clin Dev ; 25: 190-204, 2022 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-35434177

RESUMO

Type 2 diabetes, insulin resistance, and obesity are strongly associated and are a major health problem worldwide. Obesity largely results from a sustained imbalance between energy intake and expenditure. Therapeutic approaches targeting metabolic rate may counteract body weight gain and insulin resistance. Bone morphogenic protein 7 (BMP7) has proven to enhance energy expenditure by inducing non-shivering thermogenesis in short-term studies in mice treated with the recombinant protein or adenoviral vectors encoding BMP7. To achieve long-term BMP7 effects, the use of adeno-associated viral (AAV) vectors would provide sustained production of the protein after a single administration. Here, we demonstrated that treatment of high-fat-diet-fed mice and ob/ob mice with liver-directed AAV-BMP7 vectors enabled a long-lasting increase in circulating levels of this factor. This rise in BMP7 concentration induced browning of white adipose tissue (WAT) and activation of brown adipose tissue, which enhanced energy expenditure, and reversed WAT hypertrophy, hepatic steatosis, and WAT and liver inflammation, ultimately resulting in normalization of body weight and insulin resistance. This study highlights the potential of AAV-BMP7-mediated gene therapy for the treatment of insulin resistance, type 2 diabetes, and obesity.

5.
Diabetes ; 69(5): 927-939, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32086292

RESUMO

Vitamin D deficiency has been associated with increased incidence of diabetes, both in humans and in animal models. In addition, an association between vitamin D receptor (VDR) gene polymorphisms and diabetes has also been described. However, the involvement of VDR in the development of diabetes, specifically in pancreatic ß-cells, has not been elucidated yet. Here, we aimed to study the role of VDR in ß-cells in the pathophysiology of diabetes. Our results indicate that Vdr expression was modulated by glucose in healthy islets and decreased in islets from both type 1 diabetes and type 2 diabetes mouse models. In addition, transgenic mice overexpressing VDR in ß-cells were protected against streptozotocin-induced diabetes and presented a preserved ß-cell mass and a reduction in islet inflammation. Altogether, these results suggest that sustained VDR levels in ß-cells may preserve ß-cell mass and ß-cell function and protect against diabetes.


Assuntos
Fator de Crescimento Insulin-Like II/metabolismo , Células Secretoras de Insulina/metabolismo , Receptores de Calcitriol/metabolismo , Animais , Glicemia , Diabetes Mellitus , Diabetes Mellitus Experimental , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Glucose/administração & dosagem , Glucose/farmacologia , Fator de Crescimento Insulin-Like II/genética , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos Transgênicos , Receptores de Calcitriol/genética
6.
Invest Ophthalmol Vis Sci ; 60(2): 473-487, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30707220

RESUMO

Purpose: It has been suggested that arteriolar annuli localized in retinal arterioles regulate retinal blood flow acting as sphincters. Here, the morphology and protein expression profile of arteriolar annuli have been analyzed under physiologic conditions in the retina of wild-type, ß-actin-Egfp, and Nestin-gfp transgenic mice. Additionally, to study the effect of hypertension, the KAP transgenic mouse has been used. Methods: Cellular architecture has been studied using digested whole mount retinas and transmission electron microscopy. The profile of protein expression has been analyzed on paraffin sections and whole mount retinas by immunofluorescence and histochemistry. Results: The ultrastructural analysis of arteriolar annuli showed a different cell population found between endothelial and muscle cells that matched most of the morphologic criteria established to define interstitial Cajal cells. The profile of protein expression of these vascular interstitial cells (VICs) was similar to that of interstitial Cajal cells and different from the endothelial and smooth muscle cells, because they expressed ß-actin, nestin, and CD44, but they did not express CD31 and α-SMA or scarcely express F-actin. Furthermore, VICs share with pericytes the expression of NG2 and platelet-derived growth factor receptor beta (PDGFR-ß). The high expression of Ano1 and high activity of nicotinamide adenine dinucleotide phosphate (NADPH)-diaphorase observed in VICs was diminished during hypertensive retinopathy suggesting that these cells might play a role on the motility of arteriolar annuli and that this function is altered during hypertension. Conclusions: A novel type of VICs has been described in the arteriolar annuli of mouse retina. Remarkably, these cells undergo important molecular modifications during hypertensive retinopathy and might thus be a therapeutic target against this disease.


Assuntos
Células Endoteliais/patologia , Hipertensão/patologia , Retinopatia Hipertensiva/patologia , Células Intersticiais de Cajal/patologia , Artéria Retiniana/patologia , Actinas/metabolismo , Animais , Anoctamina-1/metabolismo , Pressão Arterial , Arteríolas/patologia , Células Endoteliais/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Histocitoquímica , Receptores de Hialuronatos/metabolismo , Retinopatia Hipertensiva/metabolismo , Células Intersticiais de Cajal/metabolismo , Camundongos , Camundongos Endogâmicos ICR , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão , Microscopia de Fluorescência , NADPH Desidrogenase/metabolismo , Nestina/metabolismo
7.
EMBO Mol Med ; 10(8)2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29987000

RESUMO

Prevalence of type 2 diabetes (T2D) and obesity is increasing worldwide. Currently available therapies are not suited for all patients in the heterogeneous obese/T2D population, hence the need for novel treatments. Fibroblast growth factor 21 (FGF21) is considered a promising therapeutic agent for T2D/obesity. Native FGF21 has, however, poor pharmacokinetic properties, making gene therapy an attractive strategy to achieve sustained circulating levels of this protein. Here, adeno-associated viral vectors (AAV) were used to genetically engineer liver, adipose tissue, or skeletal muscle to secrete FGF21. Treatment of animals under long-term high-fat diet feeding or of ob/ob mice resulted in marked reductions in body weight, adipose tissue hypertrophy and inflammation, hepatic steatosis, inflammation and fibrosis, and insulin resistance for > 1 year. This therapeutic effect was achieved in the absence of side effects despite continuously elevated serum FGF21. Furthermore, FGF21 overproduction in healthy animals fed a standard diet prevented the increase in weight and insulin resistance associated with aging. Our study underscores the potential of FGF21 gene therapy to treat obesity, insulin resistance, and T2D.


Assuntos
Diabetes Mellitus Tipo 2/terapia , Fatores de Crescimento de Fibroblastos/genética , Terapia Genética , Resistência à Insulina , Obesidade/terapia , Adipócitos/metabolismo , Tecido Adiposo Branco/efeitos dos fármacos , Tecido Adiposo Branco/metabolismo , Animais , Peso Corporal , Diabetes Mellitus Tipo 2/genética , Dieta Hiperlipídica , Metabolismo Energético , Fígado Gorduroso/terapia , Fatores de Crescimento de Fibroblastos/metabolismo , Fibrose/terapia , Técnicas de Transferência de Genes , Hiperplasia/terapia , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos , Músculo Esquelético/metabolismo , Obesidade/genética , Pancreatite/terapia
8.
Diabetes ; 55(12): 3246-55, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17130467

RESUMO

Type 1 diabetic patients are diagnosed when beta-cell destruction is almost complete. Reversal of type 1 diabetes will require beta-cell regeneration from islet cell precursors and prevention of recurring autoimmunity. IGF-I expression in beta-cells of streptozotocin (STZ)-treated transgenic mice regenerates the endocrine pancreas by increasing beta-cell replication and neogenesis. Here, we examined whether IGF-I also protects islets from autoimmune destruction. Expression of interferon (IFN)-beta in beta-cells of transgenic mice led to islet beta(2)-microglobulin and Fas hyperexpression and increased lymphocytic infiltration. Pancreatic islets showed high insulitis, and these mice developed overt diabetes when treated with very-low doses of STZ, which did not affect control mice. IGF-I expression in IFN-beta-expressing beta-cells of double-transgenic mice reduced beta(2)-microglobulin, blocked Fas expression, and counteracted islet infiltration. This was parallel to a decrease in beta-cell death by apoptosis in islets of STZ-treated IGF-I+IFN-beta-expressing mice. These mice were normoglycemic, normoinsulinemic, and showed normal glucose tolerance. They also presented similar pancreatic insulin content and beta-cell mass to healthy mice. Thus, local expression of IGF-I prevented islet infiltration and beta-cell death in mice with increased susceptibility to diabetes. These results indicate that pancreatic expression of IGF-I may regenerate and protect beta-cell mass in type 1 diabetes.


Assuntos
Diabetes Mellitus Experimental/imunologia , Fator de Crescimento Insulin-Like I/genética , Animais , Glicemia/metabolismo , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Tipo 1/imunologia , Regulação da Expressão Gênica , Células Secretoras de Insulina/patologia , Interferon beta/genética , Linfócitos/imunologia , Camundongos , Camundongos Endogâmicos , Camundongos Transgênicos , Infiltração de Neutrófilos
9.
J Clin Invest ; 109(9): 1153-63, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-11994404

RESUMO

Patients with type 1 diabetes are identified after the onset of the disease, when beta cell destruction is almost complete. beta cell regeneration from islet cell precursors might reverse this disease, but factors that can induce beta cell neogenesis and replication and prevent a new round of autoimmune destruction remain to be identified. Here we show that expression of IGF-I in beta cells of transgenic mice (in both C57BL/6-SJL and CD-1 genetic backgrounds) counteracts cytotoxicity and insulitis after treatment with multiple low doses of streptozotocin (STZ). STZ-treated nontransgenic mice developed high hyperglycemia and hypoinsulinemia, lost body weight, and died. In contrast, STZ-treated C57BL/6-SJL transgenic mice showed mild hyperglycemia for about 1 month, after which they normalized glycemia and survived. After STZ treatment, all CD-1 mice developed high hyperglycemia, hypoinsulinemia, polydipsia, and polyphagia. However, STZ-treated CD-1 transgenic mice gradually normalized all metabolic parameters and survived. beta cell mass increased in parallel as a result of neogenesis and beta cell replication. Thus, our results indicate that local expression of IGF-I in beta cells regenerates pancreatic islets and counteracts type 1 diabetes, suggesting that IGF-I gene transfer to the pancreas might be a suitable therapy for this disease.


Assuntos
Diabetes Mellitus Tipo 1/fisiopatologia , Fator de Crescimento Insulin-Like I/metabolismo , Ilhotas Pancreáticas/metabolismo , Animais , Apoptose/fisiologia , Glicemia/metabolismo , Peso Corporal , Divisão Celular , Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/terapia , Ingestão de Líquidos , Ingestão de Alimentos , Glucagon/metabolismo , Humanos , Insulina/genética , Insulina/metabolismo , Fator de Crescimento Insulin-Like I/genética , Ilhotas Pancreáticas/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
10.
J Clin Invest ; 113(8): 1149-57, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15085194

RESUMO

IGF-1 has been associated with the pathogenesis of diabetic retinopathy, although its role is not fully understood. Here we show that normoglycemic/normoinsulinemic transgenic mice overexpressing IGF-1 in the retina developed most alterations seen in human diabetic eye disease. A paracrine effect of IGF-1 in the retina initiated vascular alterations that progressed from nonproliferative to proliferative retinopathy and retinal detachment. Eyes from 2-month-old transgenic mice showed loss of pericytes and thickening of basement membrane of retinal capillaries. In mice 6 months and older, venule dilatation, intraretinal microvascular abnormalities, and neovascularization of the retina and vitreous cavity were observed. Neovascularization was consistent with increased IGF-1 induction of VEGF expression in retinal glial cells. In addition, IGF-1 accumulated in aqueous humor, which may have caused rubeosis iridis and subsequently adhesions between the cornea and iris that hampered aqueous humor drainage and led to neovascular glaucoma. Furthermore, all transgenic mice developed cataracts. These findings suggest a role of IGF-1 in the development of ocular complications in long-term diabetes. Thus, these transgenic mice may be used to study the mechanisms that lead to diabetes eye disease and constitute an appropriate model in which to assay new therapies.


Assuntos
Retinopatia Diabética/etiologia , Fator de Crescimento Insulin-Like I/fisiologia , Retina/metabolismo , Animais , Catarata/etiologia , Glaucoma Neovascular/etiologia , Proteína Glial Fibrilar Ácida/análise , Imuno-Histoquímica , Fator de Crescimento Insulin-Like I/análise , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fator A de Crescimento do Endotélio Vascular/análise
11.
Mol Metab ; 6(7): 664-680, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28702323

RESUMO

OBJECTIVE: Type 1 diabetes is characterized by autoimmune destruction of ß-cells leading to severe insulin deficiency. Although many improvements have been made in recent years, exogenous insulin therapy is still imperfect; new therapeutic approaches, focusing on preserving/expanding ß-cell mass and/or blocking the autoimmune process that destroys islets, should be developed. The main objective of this work was to test in non-obese diabetic (NOD) mice, which spontaneously develop autoimmune diabetes, the effects of local expression of Insulin-like growth factor 1 (IGF1), a potent mitogenic and pro-survival factor for ß-cells with immunomodulatory properties. METHODS: Transgenic NOD mice overexpressing IGF1 specifically in ß-cells (NOD-IGF1) were generated and phenotyped. In addition, miRT-containing, IGF1-encoding adeno-associated viruses (AAV) of serotype 8 (AAV8-IGF1-dmiRT) were produced and administered to 4- or 11-week-old non-transgenic NOD females through intraductal delivery. Several histological, immunological, and metabolic parameters were measured to monitor disease over a period of 28-30 weeks. RESULTS: In transgenic mice, local IGF1 expression led to long-term suppression of diabetes onset and robust protection of ß-cell mass from the autoimmune insult. AAV-mediated pancreatic-specific overexpression of IGF1 in adult animals also dramatically reduced diabetes incidence, both when vectors were delivered before pathology onset or once insulitis was established. Transgenic NOD-IGF1 and AAV8-IGF1-dmiRT-treated NOD animals had much less islet infiltration than controls, preserved ß-cell mass, and normal insulinemia. Transgenic and AAV-treated islets showed less expression of antigen-presenting molecules, inflammatory cytokines, and chemokines important for tissue-specific homing of effector T cells, suggesting IGF1 modulated islet autoimmunity in NOD mice. CONCLUSIONS: Local expression of Igf1 by AAV-mediated gene transfer counteracts progression to diabetes in NOD mice. This study suggests a therapeutic strategy for autoimmune diabetes in humans.


Assuntos
Diabetes Mellitus Tipo 1/genética , Fator de Crescimento Insulin-Like I/genética , Células Secretoras de Insulina/metabolismo , Animais , Células Cultivadas , Dependovirus/genética , Diabetes Mellitus Tipo 1/terapia , Feminino , Terapia Genética , Fator de Crescimento Insulin-Like I/metabolismo , Camundongos , Camundongos Endogâmicos ICR , Camundongos Endogâmicos NOD
12.
Mol Ther Methods Clin Dev ; 5: 16072, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27909699

RESUMO

Type 2 diabetes is characterized by triglyceride accumulation and reduced lipid oxidation capacity in skeletal muscle. SIRT1 is a key protein in the regulation of lipid oxidation and its expression is reduced in the skeletal muscle of insulin resistant mice. In this tissue, Sirt1 up-regulates the expression of genes involved in oxidative metabolism and improves mitochondrial function mainly through PPARGC1 deacetylation. Here we examined whether Sirt1 overexpression mediated by adeno-associated viral vectors of serotype 1 (AAV1) specifically in skeletal muscle can counteract the development of insulin resistance induced by a high fat diet in mice. AAV1-Sirt1-treated mice showed up-regulated expression of key genes related to ß-oxidation together with increased levels of phosphorylated AMP protein kinase. Moreover, SIRT1 overexpression in skeletal muscle also increased basal phosphorylated levels of AKT. However, AAV1-Sirt1 treatment was not enough to prevent high fat diet-induced obesity and insulin resistance. Although Sirt1 gene transfer to skeletal muscle induced changes at the muscular level related with lipid and glucose homeostasis, our data indicate that overexpression of SIRT1 in skeletal muscle is not enough to improve whole-body insulin resistance and that suggests that SIRT1 has to be increased in other metabolic tissues to prevent insulin resistance.

13.
Diabetes ; 65(8): 2139-50, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27207555

RESUMO

Eicosanoids, such as leukotriene B4 (LTB4) and lipoxin A4 (LXA4), may play a key role during obesity. While LTB4 is involved in adipose tissue inflammation and insulin resistance, LXA4 may exert anti-inflammatory effects and alleviate hepatic steatosis. Both lipid mediators derive from the same pathway, in which arachidonate 5-lipoxygenase (ALOX5) and its partner, arachidonate 5-lipoxygenase-activating protein (ALOX5AP), are involved. ALOX5 and ALOX5AP expression is increased in humans and rodents with obesity and insulin resistance. We found that transgenic mice overexpressing ALOX5AP in adipose tissue had higher LXA4 rather than higher LTB4 levels, were leaner, and showed increased energy expenditure, partly due to browning of white adipose tissue (WAT). Upregulation of hepatic LXR and Cyp7a1 led to higher bile acid synthesis, which may have contributed to increased thermogenesis. In addition, transgenic mice were protected against diet-induced obesity, insulin resistance, and inflammation. Finally, treatment of C57BL/6J mice with LXA4, which showed browning of WAT, strongly suggests that LXA4 is responsible for the transgenic mice phenotype. Thus, our data support that LXA4 may hold great potential for the future development of therapeutic strategies for obesity and related diseases.


Assuntos
Proteínas Ativadoras de 5-Lipoxigenase/genética , Proteínas Ativadoras de 5-Lipoxigenase/metabolismo , Tecido Adiposo/metabolismo , Expressão Gênica , Resistência à Insulina/genética , Lipoxinas/metabolismo , Obesidade/genética , Tecido Adiposo Branco/metabolismo , Animais , Dieta Hiperlipídica/efeitos adversos , Células Hep G2 , Humanos , Resistência à Insulina/fisiologia , Leucotrieno B4/metabolismo , Camundongos , Camundongos Transgênicos , Obesidade/etiologia , Obesidade/metabolismo , Obesidade/prevenção & controle , Termogênese/genética , Termogênese/fisiologia
14.
Diabetes ; 51(3): 624-30, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11872659

RESUMO

Adipose tissue glyceroneogenesis generates glycerol 3-phosphate, which could be used for fatty acid esterification during starvation. To determine whether increased glyceroneogenesis leads to increased fat mass and to explore the role of obesity in the development of insulin resistance, we overexpressed PEPCK, a regulatory enzyme of glyceroneogenesis in adipose tissue. Transgenic mice showed a chronic increase in PEPCK activity, which led to increased glyceroneogenesis, re-esterification of free fatty acids (FFAs), increased adipocyte size and fat mass, and higher body weight. In spite of increased fat mass, transgenic mice showed decreased circulating FFAs and normal leptin levels. Moreover, glucose tolerance and whole-body insulin sensitivity were preserved. Skeletal muscle basal and insulin-stimulated glucose uptake and glycogen content were not affected, suggesting that skeletal muscle insulin sensitivity is normal in transgenic obese mice. Our results indicate the key role of PEPCK in the control of FFA re-esterification in adipose tissue and, thus, the contribution of glyceroneogenesis to fat accumulation. Moreover, they suggest that higher fat mass without increased circulating FFAs does not lead to insulin resistance or type 2 diabetes in these mice.


Assuntos
Tecido Adiposo/enzimologia , Ácidos Graxos/metabolismo , Expressão Gênica , Resistência à Insulina , Obesidade/enzimologia , Fosfoenolpiruvato Carboxiquinase (GTP)/genética , Adipócitos/patologia , Animais , Radioisótopos de Carbono , Desoxiglucose/metabolismo , Esterificação , Ácidos Graxos não Esterificados/sangue , Ácidos Graxos não Esterificados/metabolismo , Glicerol/metabolismo , Glicerofosfatos/metabolismo , Heterozigoto , Homozigoto , Hipertrofia , Leptina/sangue , Masculino , Camundongos , Camundongos Transgênicos , RNA Mensageiro/análise , Fator de Necrose Tumoral alfa/genética
15.
PLoS One ; 10(12): e0145116, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26690959

RESUMO

Reprogramming acinar cells into insulin producing cells using adenoviral (Ad)-mediated delivery of Pdx1, Ngn3 and MafA (PNM) is an innovative approach for the treatment of diabetes. Here, we aimed to investigate the molecular mechanisms involved in this process and in particular, the role of microRNAs. To this end, we performed a comparative study of acinar-to-ß cell reprogramming efficiency in the rat acinar cell line AR42J and its subclone B13 after transduction with Ad-PNM. B13 cells were more efficiently reprogrammed than AR42J cells, which was demonstrated by a strong activation of ß cell markers (Ins1, Ins2, IAPP, NeuroD1 and Pax4). miRNome panels were used to analyze differentially expressed miRNAs in acinar cells under four experimental conditions (i) non-transduced AR42J cells, (ii) non-transduced B13 cells, (iii) B13 cells transduced with Ad-GFP vectors and (iv) B13 cells transduced with Ad-PNM vectors. A total of 59 miRNAs were found to be differentially expressed between non-transduced AR42J and B13 cells. Specifically, the miR-200 family was completely repressed in B13 cells, suggesting that these cells exist in a less differentiated state than AR42J cells and as a consequence they present a greater plasticity. Adenoviral transduction per se induced dedifferentiation of acinar cells and 11 miRNAs were putatively involved in this process, whereas 8 miRNAs were found to be associated with PNM expression. Of note, Ad-PNM reprogrammed B13 cells presented the same levels of miR-137-3p, miR-135a-5p, miR-204-5p and miR-210-3p of those detected in islets, highlighting their role in the process. In conclusion, this study led to the identification of miRNAs that might be of compelling importance to improve acinar-to-ß cell conversion for the future treatment of diabetes.


Assuntos
Células Acinares/metabolismo , Adenoviridae , Técnicas de Reprogramação Celular , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Fatores de Transcrição , Transdução Genética , Células Acinares/citologia , Animais , Antígenos de Diferenciação/biossíntese , Antígenos de Diferenciação/genética , Linhagem Celular , Insulina/genética , Secreção de Insulina , Células Secretoras de Insulina/citologia , Ratos , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética
16.
Mol Ther Methods Clin Dev ; 1: 14039, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-26015978

RESUMO

Nonalcoholic fatty liver disease (NAFLD) is the most common hepatic disease worldwide, and evidence suggests that it promotes insulin resistance and type 2 diabetes. Caloric restriction (CR) is the only available strategy for NAFLD treatment. The protein deacetylase Sirtuin1 (SIRT1), which is activated by CR, increases catabolic metabolism and decreases lipogenesis and inflammation, both involved in the development of NAFLD. Here we show that adeno-associated viral vectors of serotype 8 (AAV8)-mediated liver-specific Sirt1 gene transfer prevents the development of NAFLD induced by a high carbohydrate (HC) diet. Long-term hepatic SIRT1 overexpression led to upregulation of key hepatic genes involved in ß-oxidation, prevented HC diet-induced lipid accumulation and reduced liver inflammation. AAV8-Sirt1-treated mice showed improved insulin sensitivity, increased oxidative capacity in skeletal muscle and reduced white adipose tissue inflammation. Moreover, HC feeding induced leptin resistance, which was also attenuated in AAV8-Sirt1-treated mice. Therefore, AAV-mediated gene transfer to overexpress SIRT1 specifically in the liver may represent a new gene therapy strategy to counteract NAFLD and related diseases such as type 2 diabetes.

17.
Diabetes ; 62(2): 551-60, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23099863

RESUMO

In type 1 diabetes, loss of tolerance to ß-cell antigens results in T-cell-dependent autoimmune destruction of ß cells. The abrogation of autoreactive T-cell responses is a prerequisite to achieve long-lasting correction of the disease. The liver has unique immunomodulatory properties and hepatic gene transfer results in tolerance induction and suppression of autoimmune diseases, in part by regulatory T-cell (Treg) activation. Hence, the liver could be manipulated to treat or prevent diabetes onset through expression of key genes. IGF-I may be an immunomodulatory candidate because it prevents autoimmune diabetes when expressed in ß cells or subcutaneously injected. Here, we demonstrate that transient, plasmid-derived IGF-I expression in mouse liver suppressed autoimmune diabetes progression. Suppression was associated with decreased islet inflammation and ß-cell apoptosis, increased ß-cell replication, and normalized ß-cell mass. Permanent protection depended on exogenous IGF-I expression in liver nonparenchymal cells and was associated with increased percentage of intrapancreatic Tregs. Importantly, Treg depletion completely abolished IGF-I-mediated protection confirming the therapeutic potential of these cells in autoimmune diabetes. This study demonstrates that a nonviral gene therapy combining the immunological properties of the liver and IGF-I could be beneficial in the treatment of the disease.


Assuntos
Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Tipo 1/terapia , Terapia Genética , Fator de Crescimento Insulin-Like I/genética , Fígado/metabolismo , Linfócitos T Reguladores/imunologia , Animais , Apoptose/genética , Apoptose/imunologia , Divisão Celular/genética , Divisão Celular/imunologia , Humanos , Células Secretoras de Insulina/imunologia , Células Secretoras de Insulina/patologia , Fígado/imunologia , Camundongos , Camundongos Transgênicos , Pancreatite/genética , Pancreatite/imunologia , Plasmídeos/genética
18.
Diabetes ; 61(11): 2851-61, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22961079

RESUMO

Type 2 diabetes (T2D) results from insulin resistance and inadequate insulin secretion. Insulin resistance initially causes compensatory islet hyperplasia that progresses to islet disorganization and altered vascularization, inflammation, and, finally, decreased functional ß-cell mass and hyperglycemia. The precise mechanism(s) underlying ß-cell failure remain to be elucidated. In this study, we show that in insulin-resistant high-fat diet-fed mice, the enhanced islet vascularization and inflammation was parallel to an increased expression of vascular endothelial growth factor A (VEGF). To elucidate the role of VEGF in these processes, we have genetically engineered ß-cells to overexpress VEGF (in transgenic mice or after adeno-associated viral vector-mediated gene transfer). We found that sustained increases in ß-cell VEGF levels led to disorganized, hypervascularized, and fibrotic islets, progressive macrophage infiltration, and proinflammatory cytokine production, including tumor necrosis factor-α and interleukin-1ß. This resulted in impaired insulin secretion, decreased ß-cell mass, and hyperglycemia with age. These results indicate that sustained VEGF upregulation may participate in the initiation of a process leading to ß-cell failure and further suggest that compensatory islet hyperplasia and hypervascularization may contribute to progressive inflammation and ß-cell mass loss during T2D.


Assuntos
Ilhotas Pancreáticas/irrigação sanguínea , Ilhotas Pancreáticas/metabolismo , Neovascularização Patológica/metabolismo , Estado Pré-Diabético/metabolismo , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/biossíntese , Animais , Citocinas/metabolismo , Diabetes Mellitus Tipo 2/etiologia , Diabetes Mellitus Tipo 2/imunologia , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Dieta Hiperlipídica/efeitos adversos , Progressão da Doença , Fibrose , Técnicas de Transferência de Genes , Hiperplasia , Resistência à Insulina , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/patologia , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neovascularização Patológica/imunologia , Neovascularização Patológica/patologia , Estado Pré-Diabético/etiologia , Estado Pré-Diabético/imunologia , Estado Pré-Diabético/patologia , Isoformas de Proteínas/biossíntese , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA