Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Horm Res Paediatr ; 95(6): 619-630, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36446332

RESUMO

The growth hormone (GH)-insulin-like growth factor (IGF) cascade is central to the regulation of growth and metabolism. This article focuses on the history of the components of the IGF system, with an emphasis on the peptide hormones, IGF-I and -II, their cell surface receptors, and the IGF binding proteins (IGFBPs) and IGFBP proteases that regulate the availability of the peptide hormones for interaction with their receptors in relevant target tissues. We describe landmark events in the evolution of the somatomedin hypothesis, including evidence that has become available from experiments at the molecular and cellular levels, whole animal and tissue-specific gene knockouts, studies of cancer epidemiology, identification of prismatic human cases, and short- and long-term clinical trials of IGF-I therapy in humans. In addition, this new evidence has expanded our clinical definition of GH insensitivity (GHI) beyond growth hormone receptor mutations (classic Laron syndrome) to include conditions that cause primary IGF deficiency by impacting post-receptor signal transduction, IGF production, IGF availability to interact with the IGF-I receptor (IGF-1R), and defects in the IGF-1R, itself. We also discuss the clinical aspects of IGFs, from their description as insulin-like activity, to the use of IGF-I in the diagnosis and treatment of GH deficiency, and to the use of recombinant human IGF-I for therapy of children with GHI.


Assuntos
Fator de Crescimento Insulin-Like II , Fator de Crescimento Insulin-Like I , Síndrome de Laron , Animais , Humanos , Fator de Crescimento Insulin-Like I/deficiência , Fator de Crescimento Insulin-Like I/história , Fator de Crescimento Insulin-Like I/fisiologia , Fator de Crescimento Insulin-Like I/uso terapêutico , Síndrome de Laron/tratamento farmacológico , Síndrome de Laron/genética , Síndrome de Laron/história , Síndrome de Laron/fisiopatologia , Hormônios Peptídicos , Processamento de Proteína Pós-Traducional , Transdução de Sinais , Somatomedinas/deficiência , Somatomedinas/história , Somatomedinas/fisiologia , Fator de Crescimento Insulin-Like II/deficiência , Fator de Crescimento Insulin-Like II/história , Fator de Crescimento Insulin-Like II/fisiologia , Fator de Crescimento Insulin-Like II/uso terapêutico
2.
J Endocrinol ; 246(1): 93-108, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32380473

RESUMO

The placenta regulates materno-fetal nutrient transfer and secretes hormones that enable maternal physiological support of the pregnancy. In mice, these functions are performed by the labyrinth (Lz) and junctional (Jz) zones, respectively. Insulin-like growth factor 2 (Igf2) is an imprinted gene expressed by the conceptus that is important for promoting fetal growth and placenta formation. However, the specific role of Igf2 in the Jz in regulating placental endocrine function and fetal development is unknown. This study used a novel model to investigate the effect of conditional loss of Igf2 in the Jz (Jz-Igf2UE) on placental endocrine cell formation and the expression of hormones and IGF signaling components in placentas from female and male fetuses. Jz-Igf2UE altered gross placental structure and expression of key endocrine and signaling genes in a sexually dimorphic manner. The volumes of spongiotrophoblast and glycogen trophoblast in the Jz were decreased in placentas from female but not male fetuses. Expression of insulin receptor was increased and expression the MAPK pathway genes (Mek1, P38α) decreased in the placental Jz of female but not male fetuses. In contrast, expression of the type-1 and -2 IGF receptors and the MAPK pathway genes (H-ras, N-ras, K-ras) was decreased in the placental Jz from male but not female fetuses. Expression of the steroidogenic gene, Cyp17a1, was increased and placental lactogen-2 was decreased in the placenta of both sexes. In summary, we report that Jz-Igf2UE alters the cellular composition, IGF signaling components and hormone expression of the placental Jz in a manner largely dependent on fetal sex.


Assuntos
Fator de Crescimento Insulin-Like II/metabolismo , Placenta/metabolismo , Animais , Células Endócrinas/metabolismo , Feminino , Fator de Crescimento Insulin-Like II/deficiência , Fator de Crescimento Insulin-Like II/genética , Masculino , Camundongos , Lactogênio Placentário/genética , Lactogênio Placentário/metabolismo , Gravidez , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Somatomedinas/genética , Somatomedinas/metabolismo , Trofoblastos/metabolismo
3.
Biol Reprod ; 99(4): 864-876, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-29741588

RESUMO

Preterm birth (PTB), parturition prior to 37 weeks' gestation, is the leading cause of neonatal mortality. The causes of spontaneous PTB are poorly understood; however, recent studies suggest that this condition may arise as a consequence of the parental fetal environment. Specifically, we previously demonstrated that developmental exposure of male mice (F1 animals) to the environmental endocrine disruptor 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) was associated with reduced sperm quantity/quality in adulthood and control female partners frequently delivered preterm. Reproductive defects persisted in the F2 and F3 descendants, and spontaneous PTB was common. Reproductive changes in the F3 males, the first generation without direct TCDD exposure, suggest the occurrence of epigenetic alterations in the sperm, which have the potential to impact placental development. Herein, we conducted an epigenetic microarray analysis of control and F1 male-derived placentae, which identified 2171 differentially methylated regions, including the progesterone receptor (Pgr) and insulin-like growth factor (Igf2). To assess if Pgr and Igf2 DNA methylation changes were present in sperm and persist in future generations, we assessed methylation and expression of these genes in F1/F3 sperm and F3-derived placentae. Although alterations in methylation and gene expression were observed, in most tissues, only Pgr reached statistical significance. Despite the modest gene expression changes in Igf2, offspring of F1 and F3 males consistently exhibited IUGR. Taken together, our data indicate that paternal developmental TCDD exposure is associated with transgenerational placental dysfunction, suggesting epigenetic modifications within the sperm have occurred. An evaluation of additional genes and alternative epigenetic mechanisms is warranted.


Assuntos
Epigênese Genética , Fator de Crescimento Insulin-Like II/genética , Exposição Paterna/efeitos adversos , Placenta/metabolismo , Receptores de Progesterona/genética , Espermatozoides/metabolismo , Animais , DNA (Citosina-5-)-Metiltransferases/genética , Metilação de DNA , Modelos Animais de Doenças , Disruptores Endócrinos/toxicidade , Epigênese Genética/efeitos dos fármacos , Feminino , Retardo do Crescimento Fetal/etiologia , Fator de Crescimento Insulin-Like II/deficiência , Fator de Crescimento Insulin-Like II/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Placentação/genética , Dibenzodioxinas Policloradas/toxicidade , Gravidez , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Progesterona/deficiência , Receptores de Progesterona/metabolismo
4.
Diab Vasc Dis Res ; 15(4): 302-313, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29499628

RESUMO

AIMS: Despite vast clinical experience linking diabetes and atherosclerosis, the molecular mechanisms leading to accelerated vascular damage are still unclear. Here, we investigated the effects of nuclear factor of activated T-cells inhibition on plaque burden in a novel mouse model of type 2 diabetes that better replicates human disease. METHODS & RESULTS: IGF-II/LDLR-/-ApoB100/100 mice were generated by crossbreeding low-density lipoprotein receptor-deficient mice that synthesize only apolipoprotein B100 (LDLR-/-ApoB100/100) with transgenic mice overexpressing insulin-like growth factor-II in pancreatic ß cells. Mice have mild hyperglycaemia and hyperinsulinaemia and develop complex atherosclerotic lesions. In vivo treatment with the nuclear factor of activated T-cells blocker A-285222 for 4 weeks reduced atherosclerotic plaque area and degree of stenosis in the brachiocephalic artery of IGF-II/LDLR-/-ApoB100/100 mice, as assessed non-invasively using ultrasound biomicroscopy prior and after treatment, and histologically after termination. Treatment had no impact on plaque composition (i.e. muscle, collagen, macrophages). The reduced plaque area could not be explained by effects of A-285222 on plasma glucose, insulin or lipids. Inhibition of nuclear factor of activated T-cells was associated with increased expression of atheroprotective NOX4 and of the anti-oxidant enzyme catalase in aortic vascular smooth muscle cells. CONCLUSION: Targeting the nuclear factor of activated T-cells signalling pathway may be an attractive approach for the treatment of diabetic macrovascular complications.


Assuntos
Apolipoproteínas B/deficiência , Aterosclerose/prevenção & controle , Tronco Braquiocefálico/efeitos dos fármacos , Fator de Crescimento Insulin-Like II/deficiência , Fatores de Transcrição NFATC/antagonistas & inibidores , Placa Aterosclerótica , Pirazóis/farmacologia , Receptores de LDL/deficiência , Animais , Apolipoproteína B-100 , Apolipoproteínas B/genética , Aterosclerose/genética , Aterosclerose/metabolismo , Aterosclerose/patologia , Tronco Braquiocefálico/metabolismo , Tronco Braquiocefálico/patologia , Catalase/metabolismo , Células Cultivadas , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Modelos Animais de Doenças , Feminino , Predisposição Genética para Doença , Fator de Crescimento Insulin-Like II/genética , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , NADPH Oxidase 4/metabolismo , Fatores de Transcrição NFATC/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Fenótipo , Receptores de LDL/genética , Transdução de Sinais
5.
Science ; 356(6339): 757-759, 2017 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-28522536

RESUMO

Insulin-like growth factor 2 (IGF2) is the major fetal growth hormone in mammals. We identify zinc finger protein 568 (ZFP568), a member of the rapidly evolving Kruppel-associated box-zinc finger protein (KRAB-ZFP) family linked primarily to silencing of endogenous retroelements, as a direct repressor of a placental-specific Igf2 transcript (designated Igf2-P0) in mice. Loss of Zfp568, which causes gastrulation failure, or mutation of the ZFP568-binding site at the Igf2-P0 promoter causes inappropriate Igf2-P0 activation. Deletion of Igf2 can completely rescue Zfp568 gastrulation phenotypes through late gestation. Our data highlight the exquisite selectivity with which members of the KRAB-ZFP family repress their targets and identify an additional layer of transcriptional control of a key growth factor regulating fetal and placental development.


Assuntos
Embrião de Mamíferos/metabolismo , Fator de Crescimento Insulin-Like II/deficiência , Fator de Crescimento Insulin-Like II/genética , Proteínas Nucleares/metabolismo , Animais , Feminino , Gastrulação/genética , Regulação da Expressão Gênica , Camundongos , Camundongos Knockout , Mutação , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Placenta/metabolismo , Gravidez , Regiões Promotoras Genéticas/genética , Proteínas Repressoras/metabolismo
6.
Neurobiol Aging ; 44: 9-21, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27318130

RESUMO

Aging is accompanied by declines in memory performance, and particularly affects memories that rely on hippocampal-cortical systems, such as episodic and explicit. With aged populations significantly increasing, the need for preventing or rescuing memory deficits is pressing. However, effective treatments are lacking. Here, we show that the level of the mature form of insulin-like growth factor 2 (IGF-2), a peptide regulated in the hippocampus by learning, required for memory consolidation and a promoter of memory enhancement in young adult rodents, is significantly reduced in hippocampal synapses of aged rats. By contrast, the hippocampal level of the immature form proIGF-2 is increased, suggesting an aging-related deficit in IGF-2 processing. In agreement, aged compared to young adult rats are deficient in the activity of proprotein convertase 2, an enzyme that likely mediates IGF-2 posttranslational processing. Hippocampal administration of the recombinant, mature form of IGF-2 rescues hippocampal-dependent memory deficits and working memory impairment in aged rats. Thus, IGF-2 may represent a novel therapeutic avenue for preventing or reversing aging-related cognitive impairments.


Assuntos
Envelhecimento/psicologia , Fator de Crescimento Insulin-Like II/administração & dosagem , Fator de Crescimento Insulin-Like II/fisiologia , Transtornos da Memória/etiologia , Transtornos da Memória/prevenção & controle , Memória , Animais , Hipocampo/metabolismo , Fator de Crescimento Insulin-Like II/deficiência , Fator de Crescimento Insulin-Like II/metabolismo , Masculino , Memória de Curto Prazo , Terapia de Alvo Molecular , Pró-Proteína Convertase 2/metabolismo , Processamento de Proteína Pós-Traducional , Ratos , Ratos Endogâmicos F344 , Proteínas Recombinantes/administração & dosagem
7.
Eur J Endocrinol ; 174(4): R145-73, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26578640

RESUMO

The fast technological development, particularly single nucleotide polymorphism array, array-comparative genomic hybridization, and whole exome sequencing, has led to the discovery of many novel genetic causes of growth failure. In this review we discuss a selection of these, according to a diagnostic classification centred on the epiphyseal growth plate. We successively discuss disorders in hormone signalling, paracrine factors, matrix molecules, intracellular pathways, and fundamental cellular processes, followed by chromosomal aberrations including copy number variants (CNVs) and imprinting disorders associated with short stature. Many novel causes of GH deficiency (GHD) as part of combined pituitary hormone deficiency have been uncovered. The most frequent genetic causes of isolated GHD are GH1 and GHRHR defects, but several novel causes have recently been found, such as GHSR, RNPC3, and IFT172 mutations. Besides well-defined causes of GH insensitivity (GHR, STAT5B, IGFALS, IGF1 defects), disorders of NFκB signalling, STAT3 and IGF2 have recently been discovered. Heterozygous IGF1R defects are a relatively frequent cause of prenatal and postnatal growth retardation. TRHA mutations cause a syndromic form of short stature with elevated T3/T4 ratio. Disorders of signalling of various paracrine factors (FGFs, BMPs, WNTs, PTHrP/IHH, and CNP/NPR2) or genetic defects affecting cartilage extracellular matrix usually cause disproportionate short stature. Heterozygous NPR2 or SHOX defects may be found in ∼3% of short children, and also rasopathies (e.g., Noonan syndrome) can be found in children without clear syndromic appearance. Numerous other syndromes associated with short stature are caused by genetic defects in fundamental cellular processes, chromosomal abnormalities, CNVs, and imprinting disorders.


Assuntos
Transtornos do Crescimento/genética , Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/metabolismo , Nanismo Hipofisário/genética , Fatores de Crescimento de Fibroblastos/deficiência , Fatores de Crescimento de Fibroblastos/genética , Hormônio do Crescimento Humano/deficiência , Hormônio do Crescimento Humano/genética , Humanos , Fator de Crescimento Insulin-Like I/deficiência , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like I/metabolismo , Fator de Crescimento Insulin-Like II/deficiência , Fator de Crescimento Insulin-Like II/genética , Proteína Relacionada ao Hormônio Paratireóideo/deficiência , Proteína Relacionada ao Hormônio Paratireóideo/genética , Receptor IGF Tipo 1/genética , Transdução de Sinais/genética , Síndrome , Via de Sinalização Wnt/genética
8.
Horm Res Paediatr ; 85(1): 2-10, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26649429

RESUMO

Human growth, from fetal life to adolescence, is dynamic and a good marker of health. Growth is a complex process influenced by genetic, hormonal, nutritional and environmental factors, both pre- and postnatally. To date, no international agreement regarding normal height has been established. Auxological parameters are fundamental to investigate potential short stature (SS), either with a known diagnosis, e.g. disproportionate or proportionate, prenatal and/or postnatal onset, or an unknown diagnosis, i.e. idiopathic SS. The incidence/prevalence of SS is difficult to establish. The measurement of choice in children aged <2 years is length, while in those >2 years of age it is height. A number of monogenic diseases that lead to proportionate SS due to either isolated growth hormone deficiency, multiple pituitary hormone deficiency, growth hormone insensitivity, primary acid-labile subunit deficiency, primary IGF-1 deficiency, IGF-1 resistance, primary IGF-2 deficiency or primary protease deficiency have been discovered in the last 30 years. In addition, the Nosology and Classification of Genetic Skeletal Disorders revised in 2015 includes 436 conditions, with a number of genes of 364. A practical algorithm for the evaluation of SS as well as therapeutic options are discussed.


Assuntos
Transtornos do Crescimento , Perda Auditiva Neurossensorial , Hormônio do Crescimento Humano/deficiência , Fator de Crescimento Insulin-Like II/deficiência , Fator de Crescimento Insulin-Like I/deficiência , Adolescente , Criança , Pré-Escolar , Feminino , Transtornos do Crescimento/genética , Transtornos do Crescimento/metabolismo , Transtornos do Crescimento/patologia , Transtornos do Crescimento/terapia , Perda Auditiva Neurossensorial/genética , Perda Auditiva Neurossensorial/metabolismo , Perda Auditiva Neurossensorial/patologia , Perda Auditiva Neurossensorial/terapia , Humanos , Lactente , Recém-Nascido , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like I/metabolismo , Masculino
9.
N Engl J Med ; 373(4): 349-56, 2015 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-26154720

RESUMO

In humans, mutations in IGF1 or IGF1R cause intrauterine and postnatal growth restriction; however, data on mutations in IGF2, encoding insulin-like growth factor (IGF) II, are lacking. We report an IGF2 variant (c.191C→A, p.Ser64Ter) with evidence of pathogenicity in a multigenerational family with four members who have growth restriction. The phenotype affects only family members who have inherited the variant through paternal transmission, a finding that is consistent with the maternal imprinting status of IGF2. The severe growth restriction in affected family members suggests that IGF-II affects postnatal growth in addition to prenatal growth. Furthermore, the dysmorphic features of affected family members are consistent with a role of deficient IGF-II levels in the cause of the Silver-Russell syndrome. (Funded by Bundesministerium für Bildung und Forschung and the European Union.).


Assuntos
Códon sem Sentido , Transtornos do Crescimento/genética , Fator de Crescimento Insulin-Like II/genética , Síndrome de Silver-Russell/genética , Pai , Feminino , Retardo do Crescimento Fetal/genética , Humanos , Recém-Nascido , Fator de Crescimento Insulin-Like II/deficiência , Masculino , Linhagem , Fenótipo
10.
Am J Physiol Regul Integr Comp Physiol ; 307(6): R746-54, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-25056105

RESUMO

Fetal growth restriction (FGR) affects 3-8% of human pregnancies. Mouse models have provided important etiological data on FGR; they permit the assessment of treatment strategies on the physiological function of both mother and her developing offspring. Our study aimed to 1) develop a method to assess vascular function in fetal mice and 2) as a proof of principle ascertain whether a high dose of sildenafil citrate (SC; Viagra) administered to the pregnant dam affected fetal vascular reactivity. We developed a wire myography methodology for evaluation of fetal vascular function in vitro using the placenta-specific insulin-like growth factor II (Igf2) knockout mouse (P0; a model of FGR). Vascular function was determined in abdominal aortas isolated from P0 and wild-type (WT) fetuses at embryonic day (E) 18.5 of gestation. A subset of dams received SC 0.8 mg/ml via drinking water from E12.5; data were compared with water-only controls. Using wire myography, we found that fetal aortic rings exhibited significant agonist-induced contraction, and endothelium-dependent and endothelium-independent relaxation. Sex-specific alterations in reactivity were noted in both strains. Maternal treatment with SC significantly attenuated endothelium-dependent and endothelium-independent relaxation of fetal aortic rings. Mouse fetal abdominal aortas reproducibly respond to vasoactive agents. Study of these vessels in mouse genetic models of pregnancy complications may 1) help to delineate early signs of abnormal vascular reactivity and 2) inform whether treatments given to the mother during pregnancy may impact upon fetal vascular function.


Assuntos
Aorta Abdominal/fisiopatologia , Retardo do Crescimento Fetal/fisiopatologia , Animais , Aorta Abdominal/efeitos dos fármacos , Aorta Abdominal/embriologia , Aorta Abdominal/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Retardo do Crescimento Fetal/genética , Retardo do Crescimento Fetal/metabolismo , Idade Gestacional , Fator de Crescimento Insulin-Like II/deficiência , Fator de Crescimento Insulin-Like II/genética , Camundongos , Camundongos Knockout , Fenótipo , Inibidores da Fosfodiesterase 5/farmacologia , Piperazinas/farmacologia , Gravidez , Purinas/farmacologia , Citrato de Sildenafila , Sulfonas/farmacologia , Vasoconstrição , Vasoconstritores/farmacologia , Vasodilatação , Vasodilatadores/farmacologia
11.
J Mol Endocrinol ; 52(3): 333-44, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24711643

RESUMO

Mutations in CUL7, OBSL1 and CCDC8, leading to disordered ubiquitination, cause one of the commonest primordial growth disorders, 3-M syndrome. This condition is associated with i) abnormal p53 function, ii) GH and/or IGF1 resistance, which may relate to failure to recycle signalling molecules, and iii) cellular IGF2 deficiency. However the exact molecular mechanisms that may link these abnormalities generating growth restriction remain undefined. In this study, we have used immunoprecipitation/mass spectrometry and transcriptomic studies to generate a 3-M 'interactome', to define key cellular pathways and biological functions associated with growth failure seen in 3-M. We identified 189 proteins which interacted with CUL7, OBSL1 and CCDC8, from which a network including 176 of these proteins was generated. To strengthen the association to 3-M syndrome, these proteins were compared with an inferred network generated from the genes that were differentially expressed in 3-M fibroblasts compared with controls. This resulted in a final 3-M network of 131 proteins, with the most significant biological pathway within the network being mRNA splicing/processing. We have shown using an exogenous insulin receptor (INSR) minigene system that alternative splicing of exon 11 is significantly changed in HEK293 cells with altered expression of CUL7, OBSL1 and CCDC8 and in 3-M fibroblasts. The net result is a reduction in the expression of the mitogenic INSR isoform in 3-M syndrome. From these preliminary data, we hypothesise that disordered ubiquitination could result in aberrant mRNA splicing in 3-M; however, further investigation is required to determine whether this contributes to growth failure.


Assuntos
Antígenos CD/genética , Proteínas de Transporte/genética , Proteínas Culina/genética , Proteínas do Citoesqueleto/genética , Nanismo/genética , Hipotonia Muscular/genética , Receptor de Insulina/genética , Coluna Vertebral/anormalidades , Processamento Alternativo/genética , Proteínas de Transporte/biossíntese , Linhagem Celular Tumoral , Proteínas Culina/biossíntese , Proteínas do Citoesqueleto/biossíntese , Fibroblastos , Perfilação da Expressão Gênica , Transtornos do Crescimento/genética , Células HEK293 , Hormônio do Crescimento Humano/genética , Humanos , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like II/deficiência , Fator de Crescimento Insulin-Like II/genética , Proteína Supressora de Tumor p53/genética , Ubiquitinação/genética
12.
Nat Commun ; 4: 2311, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23921428

RESUMO

Imprinted, maternally silenced insulin-like growth factor-2 is expressed in both the foetus and placenta and has been shown to have roles in foetal and placental development in animal models. Here we compared mice engineered to be null for the placenta-specific P0 transcript (insulin-like growth factor-2-P0 KO) to mice with disruptions of all four insulin-like growth factor-2 transcripts, and therefore null for insulin-like growth factor-2 in both placenta and foetus (insulin-like growth factor-2-total KO). Both models lead to intrauterine growth restriction but dissociate between a situation where there is an imbalance between foetal demand and placental supply of nutrients (the insulin-like growth factor-2-P0 KO) and one where demand and supply is more balanced (the insulin-like growth factor-2-total KO). Increased reactivity to anxiety-provoking stimuli is manifested later in life only in those animals where there is a mismatch between placental supply and foetal demand for nutrients during gestation. Our findings further distinguish placental dysfunction from intrauterine growth restriction and reveal a role for the placenta in long-term programming of emotional behaviour.


Assuntos
Envelhecimento/patologia , Ansiedade/patologia , Fator de Crescimento Insulin-Like II/deficiência , Placenta/metabolismo , Estimulação Acústica , Animais , Animais Recém-Nascidos , Ansiedade/genética , Comportamento Animal , Peso ao Nascer , Peso Corporal , Modelos Animais de Doenças , Comportamento Exploratório , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Impressão Genômica/genética , Hipocampo/metabolismo , Hipocampo/patologia , Fator de Crescimento Insulin-Like II/genética , Fator de Crescimento Insulin-Like II/metabolismo , Aprendizagem em Labirinto , Camundongos , Camundongos Knockout , Gravidez
13.
Endocrinology ; 153(1): 241-53, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22087027

RESUMO

Myostatin inhibition by follistatin (FS) offers a new approach for muscle mass enhancement. The aim of the present study was to characterize the mediators responsible for the FS hypertrophic action on skeletal muscle in male mice. Because IGF-I and IGF-II, two crucial skeletal muscle growth factors, are induced by myostatin inhibition, we assessed their role in FS action. First, we tested whether type 1 IGF receptor (IGF-IR) is required for FS-induced hypertrophy. By using mice expressing a dominant-negative IGF-IR in skeletal muscle, we showed that IGF-IR inhibition blunted by 63% fiber hypertrophy caused by FS. Second, we showed that FS caused the same degree of fiber hypertrophy in wild-type and IGF-II knockout mice. We then tested the role of the signaling molecules stimulated by IGF-IR, in particular the Akt/mammalian target of rapamycin (mTOR)/70-kDa ribosomal protein S6 kinase (S6K) pathway. We investigated whether Akt phosphorylation is required for the FS action. By cotransfecting a dominant-negative form of Akt together with FS, we showed that Akt inhibition reduced by 65% fiber hypertrophy caused by FS. Second, we evaluated the role of mTOR in FS action. Fiber hypertrophy induced by FS was reduced by 36% in rapamycin-treated mice. Finally, because the activity of S6K is increased by FS, we tested its role in FS action. FS caused the same degree of fiber hypertrophy in wild-type and S6K1/2 knockout mice. In conclusion, the IGF-IR/Akt/mTOR pathway plays a critical role in FS-induced muscle hypertrophy. In contrast, induction of IGF-II expression and S6K activity by FS are not required for the hypertrophic action of FS.


Assuntos
Folistatina/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Receptor IGF Tipo 1/metabolismo , Animais , Sequência de Bases , Primers do DNA/genética , Folistatina/genética , Humanos , Hipertrofia , Fator de Crescimento Insulin-Like II/deficiência , Fator de Crescimento Insulin-Like II/genética , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , Camundongos Transgênicos , Músculo Esquelético/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor IGF Tipo 1/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Quinases S6 Ribossômicas/metabolismo , Transdução de Sinais , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Transfecção
14.
Endocrinology ; 152(8): 3202-12, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21673101

RESUMO

The pattern of fetal growth is a major determinant of the subsequent health of the infant. We recently showed in undernourished (UN) mice that fetal growth is maintained until late pregnancy, despite reduced placental weight, through adaptive up-regulation of placental nutrient transfer. Here, we determine the role of the placental-specific transcript of IGF-II (Igf2P0), a major regulator of placental transport capacity in mice, in adapting placental phenotype to UN. We compared the morphological and functional responses of the wild-type (WT) and Igf2P0-deficient placenta in WT mice fed ad libitium or 80% of the ad libitium intake. We observed that deletion of Igf2P0 prevented up-regulation of amino acid transfer normally seen in UN WT placenta. This was associated with a reduction in the proportion of the placenta dedicated to nutrient transport, the labyrinthine zone, and its constituent volume of trophoblast in Igf2P0-deficient placentas exposed to UN on d 16 of pregnancy. Additionally, Igf2P0-deficient placentas failed to up-regulate their expression of the amino acid transporter gene, Slc38a2, and down-regulate phosphoinositide 3-kinase-protein kinase B signaling in response to nutrient restriction on d 19. Furthermore, deleting Igf2P0 altered maternal concentrations of hormones (insulin and corticosterone) and metabolites (glucose) in both nutritional states. Therefore, Igf2P0 plays important roles in adapting placental nutrient transfer capacity during UN, via actions directly on the placenta and/or indirectly through the mother.


Assuntos
Adaptação Fisiológica , Fator de Crescimento Insulin-Like II/fisiologia , Desnutrição/metabolismo , Placenta/metabolismo , Aminoácidos/metabolismo , Animais , Transporte Biológico , Feminino , Fator de Crescimento Insulin-Like II/deficiência , Masculino , Desnutrição/patologia , Camundongos , Camundongos Endogâmicos C57BL , Fosfatidilinositol 3-Quinases/fisiologia , Placenta/patologia , Gravidez , Proteínas Proto-Oncogênicas c-akt/fisiologia
15.
Arterioscler Thromb Vasc Biol ; 31(5): 1011-5, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21372297

RESUMO

OBJECTIVE: Atherosclerotic plaques with large lipid cores and inflammation contain regions of hypoxia. We examined the uptake of 2-(2-nitro-1H-imidazol-1-yl)-N-(2,2,3,3,3-pentafluoropropyl) acetamide ([18F]EF5), a specific marker of hypoxia labeled for positron emission tomography, in mouse atherosclerotic plaques. METHODS AND RESULTS: Atherosclerotic mice of 2 different genetic backgrounds (low-density lipoprotein receptor-/- apolipoprotein B100/100 and insulin-like growth factor II/low-density lipoprotein receptor-/- apolipoprotein B100/100) were first fed a Western diet to induce development of plaques with variable phenotypes and then injected with [18F]EF5. C57BL/6N mice served as controls. Aortas were dissected for biodistribution studies, autoradiography, histology, and immunohistochemistry. Uptake of [18F]EF5 was significantly higher in the aortas of mice with large atherosclerotic plaques than in the C57BL/6N controls. Furthermore, autoradiography demonstrated, on average, 2.0-fold higher [18F]EF5 uptake in atherosclerotic plaques than in the adjacent normal vessel wall. Hypoxia in plaques was verified by using an EF5 adduct-specific antibody and pimonidazole. The blood clearance of [18F]EF5 was slow, with blood radioactivity remaining relatively high up to 180 minutes after injection. CONCLUSIONS: Large atherosclerotic plaques in mice contained hypoxic areas and showed uptake of [18F]EF5. Despite its slow blood clearance, the high uptake of [18F]EF5 in plaques suggested that plaque hypoxia is a potential target for identifying high-risk plaques noninvasively.


Assuntos
Aorta/diagnóstico por imagem , Aterosclerose/diagnóstico por imagem , Etanidazol/análogos & derivados , Radioisótopos de Flúor , Hidrocarbonetos Fluorados , Hipóxia/diagnóstico por imagem , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos , Análise de Variância , Animais , Aorta/metabolismo , Aorta/patologia , Apolipoproteína B-100/deficiência , Apolipoproteína B-100/genética , Aterosclerose/genética , Aterosclerose/metabolismo , Aterosclerose/patologia , Autorradiografia , Modelos Animais de Doenças , Etanidazol/farmacocinética , Feminino , Radioisótopos de Flúor/farmacocinética , Genótipo , Hidrocarbonetos Fluorados/farmacocinética , Hipóxia/genética , Hipóxia/metabolismo , Hipóxia/patologia , Imuno-Histoquímica , Fator de Crescimento Insulin-Like II/deficiência , Fator de Crescimento Insulin-Like II/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nitroimidazóis , Fenótipo , Compostos Radiofarmacêuticos/farmacocinética , Receptores de LDL/deficiência , Receptores de LDL/genética , Distribuição Tecidual
16.
Exp Diabetes Res ; 2011: 171376, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21331382

RESUMO

At the beginning of the third week of pregnancy, mouse fetuses with targeted disruption of their paternally-transmitted insulin-like growth factor 2 gene placental-specific transcripts have growth-restricted placentas but normal body weights due to upregulated placental nutrient transport. We assessed whether increased placental glucose transport rates were associated with raised maternal glucose concentrations by performing intraperitoneal glucose tolerance tests (ipGTT) in pregnant mice carrying knockout pups and comparing them with mice carrying genotype-matched phenotypically wild type pups. Mean ± SD body weights of affected pups were 95 ± 8% of control values at e16 and 73 ± 7% at e18. There were no differences in areas under the maternal ipGTT curves at either e16 (mean ± SD being 99.0 ± 9.1% of control values; P = .9) or e18 (91.4 ± 13.4%; P = .3), suggesting that effects on transplacental glucose transport in these mice are not mediated through changes in maternal glucose concentrations.


Assuntos
Glicemia/análise , Sangue Fetal/química , Glucose/metabolismo , Fator de Crescimento Insulin-Like II/genética , Fator de Crescimento Insulin-Like II/fisiologia , Placenta/metabolismo , Animais , Feminino , Retardo do Crescimento Fetal/genética , Expressão Gênica , Idade Gestacional , Teste de Tolerância a Glucose , Insulina/sangue , Fator de Crescimento Insulin-Like II/deficiência , Masculino , Troca Materno-Fetal , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Gravidez
17.
Development ; 138(2): 203-13, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21148188

RESUMO

During embryonic development, Igf2 gene transcription is highly regulated through the use of several promoters whose specific roles are not defined. Here, we show that loss-of-function of one of these promoters, Igf2-P2, results in growth defects that are temporally and quantitatively different from those seen in Igf2-null mutants. In particular, Igf2-P2 mutants exhibit skeletal abnormalities characterized by thin and short bones with reduced mineralization and medullar cavity and with altered bone remodeling. These abnormalities are associated with decreased numbers of embryonic mesenchymal chondroprogenitors, adult mesenchymal stem cells and osteoprogenitors. Differentiation of osteoprogenitors into osteoblasts is impaired in the Igf2-P2 mutant mice in a cell-autonomous manner, and osteopontin is a target of the IGF2 signaling pathway during this differentiation. Igf2-P2 mutant mice also display impaired formation of giant osteoclasts owing to a defective micro-environment. These results support a model wherein transcriptional activity of the Igf2-P2 promoter regulates the fate of mesenchymal progenitors during bone development and remodeling in the adult, and regulates osteogenesis in a cell-autonomous and non-autonomous manner.


Assuntos
Fator de Crescimento Insulin-Like II/deficiência , Fator de Crescimento Insulin-Like II/genética , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Osteoclastos/citologia , Osteoclastos/metabolismo , Células-Tronco Adultas/citologia , Células-Tronco Adultas/metabolismo , Animais , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Ensaio de Unidades Formadoras de Colônias , Nanismo/embriologia , Nanismo/genética , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Feminino , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Mutantes , Mutação , Osteogênese/genética , Osteogênese/fisiologia , Gravidez , Regiões Promotoras Genéticas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais , Células Estromais/citologia , Células Estromais/metabolismo
18.
Endocrinology ; 151(2): 741-7, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20032056

RESUMO

Whether insulin or IGFs regulate glycogen synthesis in the fetal liver remains to be determined. In this study, we used several knockout mouse strains, including those lacking Pdx-1 (pancreatic duodenal homeobox-1), Insr (insulin receptor), and Igf2 (IGF-II) to determine the role of these genes in the regulation of fetal hepatic glycogen synthesis. Our data show that insulin deficiency does not alter hepatic glycogen stores, whereas Insr and Igf2 deficiency do. We found that both insulin receptor isoforms (IR-A and IR-B) are present in the fetal liver, and their expression is gestationally regulated. IR-B is highly expressed in the fetal liver; nonetheless, the percentage of hepatic IR-A isoform, which binds Igf2, was significantly higher in the fetus than the adult. In vitro experiments demonstrate that Igf2 increases phosphorylation of hepatic Insr, insulin receptor substrate-2, and Akt proteins and also the activity of glycogen synthase. Igf2 ultimately increased glycogen synthesis in fetal hepatocytes. This increase could be blocked by the phosphoinositide 3-kinase inhibitor LY294008. Taken together, we propose Igf2 as a major regulator of fetal hepatic glycogen metabolism, the insulin receptor as its target receptor, and phosphoinositide 3-kinase as the signaling pathway leading to glycogen formation in the fetal liver.


Assuntos
Hepatócitos/fisiologia , Fator de Crescimento Insulin-Like II/fisiologia , Insulina/fisiologia , Glicogênio Hepático/sangue , Receptor de Insulina/fisiologia , Animais , Células Cultivadas , Cruzamentos Genéticos , Primers do DNA , Feminino , Feto/efeitos dos fármacos , Feto/fisiologia , Glicogênio Sintase/metabolismo , Hepatócitos/citologia , Hepatócitos/enzimologia , Heterozigoto , Homeostase , Insulina/deficiência , Insulina/genética , Fator de Crescimento Insulin-Like II/deficiência , Fator de Crescimento Insulin-Like II/genética , Masculino , Camundongos , Camundongos Knockout , RNA Mensageiro/genética , Receptor de Insulina/deficiência , Receptor de Insulina/genética
19.
Placenta ; 30(10): 919-22, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19733906

RESUMO

Bisphosphoglycerate mutase (BPGM) catalyses the formation of 2,3 bisphosphoglycerate (BPG) a ligand of haemoglobin. BPG facilitates liberation of oxygen from haemoglobin at low oxygen tension enabling efficient delivery of oxygen to tissues. We describe expression of BPGM in mouse labyrinthine trophoblasts, located at the maternal-placental interface. Expression is lower in placentae of igf2(+/-) knockout mice, a widely used model of growth restriction, compared to wild type placentae. Circulating maternal BPG increased throughout gestation but this increase was less in wt mothers carrying igf2(+/-) pups than in those carrying exclusively wt pups. This reduction was observed well before term and may contribute to the low birth weight of igf2(+/-) pups. Strikingly, we also measured reductions of fetal and placental weight in wt littermates of igf2(+/-) pups compared to pups developing in an exclusively wt environment. These data suggest that placental expression of BPGM can influence maternal BPG concentrations and supports a hypothesis under which BPG synthesized in the placenta may act on maternal haemoglobin to enhance delivery of oxygen to the developing fetus.


Assuntos
2,3-Difosfoglicerato/sangue , Bisfosfoglicerato Mutase/metabolismo , Desenvolvimento Fetal/genética , Fator de Crescimento Insulin-Like II/deficiência , Placenta/metabolismo , Animais , Bisfosfoglicerato Mutase/genética , Feminino , Peso Fetal/genética , Deleção de Genes , Expressão Gênica/genética , Idade Gestacional , Fator de Crescimento Insulin-Like II/genética , Masculino , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Tamanho do Órgão , Placenta/citologia , Placenta/enzimologia , Placenta/patologia , Gravidez , Trofoblastos/metabolismo
20.
Neuroreport ; 20(16): 1414-8, 2009 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-19738502

RESUMO

The search for therapeutic targets to prevent neurons from dying is ongoing and involves the exploration of a long list of neurotrophic factors. Insulin-like growth factor 2 (IGF2) is a member of the insulin family with known neurotrophic properties. In this study, we used Igf2 knockout (Igf2) neonate mice to determine whether Igf2 deficiency is detrimental to motor neuron survival after axonal injury. Results show that Igf2 neonatal mice are more susceptible to motor neuron damage than Igf2 mice, as they have a significantly lower percentage of motor neuron survival after a sciatic nerve transection. Neuronal survival was significantly improved in Igf2 mice when IGF2 was administered. These results support the role of IGF2 in neonatal motor neuron survival.


Assuntos
Fator de Crescimento Insulin-Like II/deficiência , Neurônios Motores/fisiologia , Neuropatia Ciática/patologia , Neuropatia Ciática/fisiopatologia , Animais , Animais Recém-Nascidos , Contagem de Células/métodos , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Fator de Crescimento Insulin-Like II/uso terapêutico , Camundongos , Camundongos Knockout , Neurônios Motores/efeitos dos fármacos , Neuropatia Ciática/tratamento farmacológico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...