Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Hum Mol Genet ; 32(15): 2473-2484, 2023 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-37162340

RESUMEN

Growth hormone (GH) binding to GH receptor activates janus kinase 2 (JAK2)-signal transducer and activator of transcription 5b (STAT5b) pathway, which stimulates transcription of insulin-like growth factor-1 (IGF1), insulin-like growth factor binding protein 3 (IGFBP3) and insulin-like growth factor acid-labile subunit (IGFALS). Although STAT5B deficiency was established as an autosomal recessive disorder, heterozygous dominant-negative STAT5B variants have been reported in patients with less severe growth deficit and milder immune dysfunction. We developed an in vivo functional assay in zebrafish to characterize the pathogenicity of three human STAT5B variants (p.Ala630Pro, p.Gln474Arg and p.Lys632Asn). Overexpression of human wild-type (WT) STAT5B mRNA and its variants led to a significant reduction of body length together with developmental malformations in zebrafish embryos. Overexpression of p.Ala630Pro, p.Gln474Arg or p.Lys632Asn led to an increased number of embryos with pericardial edema, cyclopia and bent spine compared with WT STAT5B. Although co-injection of WT and p.Gln474Arg and WT and p.Lys632Asn STAT5B mRNA in zebrafish embryos partially or fully rescues the length and the developmental malformations in zebrafish embryos, co-injection of WT and p.Ala630Pro STAT5B mRNA leads to a greater number of embryos with developmental malformations and a reduction in body length of these embryos. These results suggest that these variants could interfere with endogenous stat5.1 signaling through different mechanisms. In situ hybridization of zebrafish embryos overexpressing p.Gln474Arg and p.Lys632Asn STAT5B mRNA shows a reduction in igf1 expression. In conclusion, our study reveals the pathogenicity of the STAT5B variants studied.


Asunto(s)
Factor de Transcripción STAT5 , Pez Cebra , Animales , Humanos , Pez Cebra/genética , Pez Cebra/metabolismo , Factor de Transcripción STAT5/genética , Factor de Transcripción STAT5/metabolismo , Hormona del Crecimiento , Transducción de Señal/genética , ARN Mensajero , Factor I del Crecimiento Similar a la Insulina/genética
2.
Pediatr Endocrinol Rev ; 17(4): 331-342, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32780957

RESUMEN

Pheochromocytomas/paragangliomas (PCCs/PGLs) are rare neuroendocrine tumors, developed from chromaffin cells derived from the neural crest. From a genetic point of view PCCs/PGLs are divided as sporadic cases, and inherited cases as part of hereditary (familial) syndromes. While the majority is benign, up to 26% of PCCs/PGLs will undergo malignant transformation. Validated prognostic pathological parameters for malignant PCCs/PGLs are still lacking. Signaling that follows the interactions between IGFs and their receptor/s in tumor cells received extensive attention when investigating the role of IGF1R in cancer. Increased IGF1R expression has been shown during progression to metastatic phenotypes and associated with worse prognosis in several types of cancer. In this review we provide evidence supporting a role for the IGFs system on PCC/PGL tumor biology and malignant behavior, endocrine actions to sustain tumor phenotype as well as heterotypic interaction´s regulation by IGF1, encouraging further research for targeted therapeutic options for these tumors.


Asunto(s)
Neoplasias de las Glándulas Suprarrenales , Feocromocitoma , Trastornos del Crecimiento , Humanos , Insulinas
3.
Clin Endocrinol (Oxf) ; 79(5): 623-30, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23506534

RESUMEN

CONTEXT: Pheochromocytomas and paragangliomas (pheo/pgl) are neuroendocrine tumours derived from chromaffin cells. Although mostly benign, up to 26% of pheo/pgl will undergo malignant transformation. Reliable histological signs to differentiate benign pheo/pgl from malignant tumours are currently lacking. Increased IGF-1R expression has been shown during progression to metastatic phenotypes of several types of cancer. OBJECTIVE: To analyse the distribution and expression of the IGF-1R in pheo/pgl of different genetic origin and degree of malignancy. MEASUREMENTS: We studied the expression of the IGF-1R protein by immunohistochemistry, in 40 primary tumours from patients with pheo/pgl from different genetic aetiology (11 of 29 metastatic/nonmetastatic diseases). RESULTS: We found a strong association between increased expression of IGF-1R and malignant behaviour regardless of the age at diagnosis and the genetic aetiology. IGF-1R labelling was mostly weak in primary tumours from patients with nonmetastatic pheo/pgl. Conversely, intense IGF-1R labelling was predominant in cases of pheo/pgl with confirmed metastatic disease. The risk of metastases was 11·7 times higher if tumour IGF-1R labelling was intense independently of age at diagnosis. The probability of remaining free of metastases was higher in patients with pheo/pgl scored weak for IGF-1R at 60 months and more than twofold higher at 120 months of follow-up than in patients with intense IGF-1R labelling in their primary tumours. CONCLUSIONS: Our results strongly suggest that IGF-1R is associated with malignancy in familial pheo/pgl and that IGF-1R expression in the primary tumour might be a useful tool to detect those patients harbouring pheo/pgl who have an increased risk of metastasis.


Asunto(s)
Paraganglioma/metabolismo , Paraganglioma/patología , Feocromocitoma/metabolismo , Feocromocitoma/patología , Receptor IGF Tipo 1/metabolismo , Adolescente , Neoplasias de las Glándulas Suprarrenales/metabolismo , Neoplasias de las Glándulas Suprarrenales/patología , Adulto , Anciano , Niño , Preescolar , Femenino , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Adulto Joven
4.
J Clin Endocrinol Metab ; 108(6): 1355-1369, 2023 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-36546343

RESUMEN

CONTEXT: Insulin-like growth factor (IGF)1 gene mutations are extremely rare causes of pre- and postnatal growth retardation. Phenotype can be heterogenous with varying degrees of neurosensory deafness, cognitive defects, glucose metabolism impairment and short stature. OBJECTIVE: This study describes a 12.6-year-old girl presenting with severe short stature and insulin resistance, but with normal hearing and neurological development at the lower limit of normal. METHODS: DNA was obtained from the proband and both parents for whole exome sequencing (WES). In silico analysis was performed to predict the impact of the IGF1 variant on IGF1 and insulin receptors (IGF1R and IR) signaling. Phosphorylation of the IGF1R at activating Tyr residues and cell proliferation analyses were used to assess the ability of each subject's IGF1 to bind and activate IGF1R. RESULTS: The proband had low immunoreactive IGF1 in serum and WES revealed a novel homozygous IGF1 missense variant (c.247A>T), causing a change of serine 83 for cysteine (p.Ser83Cys; p.Ser35Cys in mature peptide). The proband's parents were heterozygous for this mutation. In silico analyses indicated the pathogenic potential of the variant with electrostatic variations with the potential of hampering the interaction with the IGF1R but strengthening the binding to IR. The mutant IGF1 protein had a significantly reduced activity on in vitro bioassays. CONCLUSION: We describe a novel IGF1 mutation leading to severe loss of circulating IGF1 immunoreactivity and bioactivity. In silico modeling predicts that the mutant IGF1 could interfere with IR signaling, providing a possible explanation for the severe insulin resistance observed in the patient. The absence of significant hearing and neurodevelopmental involvement in the present case is unusual and broadens the clinical spectrum of IGF1 mutations.


Asunto(s)
Enanismo , Resistencia a la Insulina , Humanos , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Resistencia a la Insulina/genética , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Mutación , Mutación Missense , Enanismo/genética , Fenotipo
6.
Front Endocrinol (Lausanne) ; 13: 854365, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35388293

RESUMEN

The von Hippel-Lindau (VHL) disease is an autosomal dominant cancer syndrome caused by mutations in the VHL tumor suppressor gene. VHL protein (pVHL) forms a complex (VBC) with Elongins B-C, Cullin2, and Rbx1. Although other functions have been discovered, the most described function of pVHL is to recognize and target hypoxia-inducible factor (HIF) for degradation. This work comprises the functional characterization of two novel variants of the VHL gene (P138R and L163R) that have been described in our center in patients with VHL disease by in vitro, in vivo, and in silico approaches. In vitro, we found that these variants have a significantly shorter half-life compared to wild-type VHL but still form a functional VBC complex. Altered fibronectin deposition was evidenced for both variants using immunofluorescence. In vivo studies revealed that both variants failed to suppress tumor growth. By means of molecular dynamics simulations, we inspected in silico the nature of the changes introduced by each variant in the VBC complex. We have demonstrated the pathogenicity of P138R and L163R novel variants, involving HIF-dependent and HIF-independent mechanisms. These results provide the basis for future studies regarding the impact of structural alterations on posttranslational modifications that drive pVHL's fate and functions.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Enfermedad de von Hippel-Lindau , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Humanos , Factores de Transcripción/metabolismo , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo , Enfermedad de von Hippel-Lindau/genética
7.
Front Endocrinol (Lausanne) ; 13: 849279, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35574033

RESUMEN

Gliomas are the most frequent solid tumors in children. Among these, high-grade gliomas are less common in children than in adults, though they are similar in their aggressive clinical behavior. In adults, glioblastoma is the most lethal tumor of the central nervous system. Insulin-like growth factor 1 receptor (IGF1R) plays an important role in cancer biology, and its nuclear localization has been described as an adverse prognostic factor in different tumors. Previously, we have demonstrated that, in pediatric gliomas, IGF1R nuclear localization is significantly associated with high-grade tumors, worst clinical outcome, and increased risk of death. Herein we explore the role of IGF1R intracellular localization by comparing two glioblastoma cell lines that differ only in their IGF1R capacity to translocate to the nucleus. In vitro, IGF1R nuclear localization enhances glioblastoma cell motility and metabolism without affecting their proliferation. In vivo, IGF1R has the capacity to translocate to the nucleus and allows not only a higher proliferation rate and the earlier development of tumors but also renders the cells sensitive to OSI906 therapy. With this work, we provide evidence supporting the implications of the presence of IGF1R in the nucleus of glioma cells and a potential therapeutic opportunity for patients harboring gliomas with IGF1R nuclear localization.


Asunto(s)
Glioblastoma , Glioma , Adulto , Carcinogénesis/metabolismo , Núcleo Celular/metabolismo , Niño , Glioblastoma/metabolismo , Glioma/metabolismo , Humanos , Receptores de Somatomedina/metabolismo
8.
J Mol Endocrinol ; 69(2): 357-376, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35608964

RESUMEN

Growth hormone (GH) exerts major actions in cardiac growth and metabolism. Considering the important role of insulin in the heart and the well-established anti-insulin effects of GH, cardiac insulin resistance may play a role in the cardiopathology observed in acromegalic patients. As conditions of prolonged exposure to GH are associated with a concomitant increase of circulating GH, IGF1 and insulin levels, to dissect the direct effects of GH, in this study, we evaluated the activation of insulin signaling in the heart using four different models: (i) transgenic mice overexpressing GH, with chronically elevated GH, IGF1 and insulin circulating levels; (ii) liver IGF1-deficient mice, with chronically elevated GH and insulin but decreased IGF1 circulating levels; (iii) mice treated with GH for a short period of time; (iv) primary culture of rat cardiomyocytes incubated with GH. Despite the differences in the development of cardiomegaly and in the metabolic alterations among the three experimental mouse models analyzed, exposure to GH was consistently associated with a decreased response to acute insulin stimulation in the heart at the receptor level and through the PI3K/AKT pathway. Moreover, a blunted response to insulin stimulation of this signaling pathway was also observed in cultured cardiomyocytes of neonatal rats incubated with GH. Therefore, the key novel finding of this work is that impairment of insulin signaling in the heart is a direct and early event observed as a consequence of exposure to GH, which may play a major role in the development of cardiac pathology.


Asunto(s)
Acromegalia , Hormona de Crecimiento Humana , Animales , Hormona del Crecimiento/metabolismo , Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Ratones , Miocitos Cardíacos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Ratas , Transducción de Señal
9.
Eur J Endocrinol ; 186(6): P35-P52, 2022 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-35319491

RESUMEN

Growth hormone (GH) has been used for over 35 years, and its safety and efficacy has been studied extensively. Experimental studies showing the permissive role of GH/insulin-like growth factor 1 (IGF-I) in carcinogenesis have raised concerns regarding the safety of GH replacement in children and adults who have received treatment for cancer and those with intracranial and pituitary tumours. A consensus statement was produced to guide decision-making on GH replacement in children and adult survivors of cancer, in those treated for intracranial and pituitary tumours and in patients with increased cancer risk. With the support of the European Society of Endocrinology, the Growth Hormone Research Society convened a Workshop, where 55 international key opinion leaders representing 10 professional societies were invited to participate. This consensus statement utilized: (1) a critical review paper produced before the Workshop, (2) five plenary talks, (3) evidence-based comments from four breakout groups, and (4) discussions during report-back sessions. Current evidence reviewed from the proceedings from the Workshop does not support an association between GH replacement and primary tumour or cancer recurrence. The effect of GH replacement on secondary neoplasia risk is minor compared to host- and tumour treatment-related factors. There is no evidence for an association between GH replacement and increased mortality from cancer amongst GH-deficient childhood cancer survivors. Patients with pituitary tumour or craniopharyngioma remnants receiving GH replacement do not need to be treated or monitored differently than those not receiving GH. GH replacement might be considered in GH-deficient adult cancer survivors in remission after careful individual risk/benefit analysis. In children with cancer predisposition syndromes, GH treatment is generally contraindicated but may be considered cautiously in select patients.


Asunto(s)
Hormona de Crecimiento Humana , Neoplasias Hipofisarias , Adulto , Niño , Hormona del Crecimiento , Hormona de Crecimiento Humana/efectos adversos , Humanos , Factor I del Crecimiento Similar a la Insulina , Recurrencia Local de Neoplasia/inducido químicamente , Neoplasias Hipofisarias/tratamiento farmacológico , Sobrevivientes
10.
FASEB J ; 23(3): 709-19, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18952711

RESUMEN

Serum insulin-like growth factor (IGF) -1 is secreted mainly by the liver and circulates bound to IGF-binding proteins (IGFBPs), either as binary complexes or ternary complexes with IGFBP-3 or IGFBP-5 and an acid-labile subunit (ALS). The purpose of this study was to genetically dissect the role of IGF-1 circulatory complexes in somatic growth, skeletal integrity, and metabolism. Phenotypic comparisons of controls and four mouse lines with genetic IGF-1 deficits-liver-specific IGF-1 deficiency (LID), ALS knockout (ALSKO), IGFBP-3 (BP3) knockout, and a triply deficient LID/ALSKO/BP3 line-produced several novel findings. 1) All deficient strains had decreased serum IGF-1 levels, but this neither predicted growth potential or skeletal integrity nor defined growth hormone secretion or metabolic abnormalities. 2) IGF-1 deficiency affected development of both cortical and trabecular bone differently, effects apparently dependent on the presence of different circulating IGF-1 complexes. 3) IGFBP-3 deficiency resulted in increased linear growth. In summary, each IGF-1 complex constituent appears to play a distinct role in determining skeletal phenotype, with different effects on cortical and trabecular bone compartments.


Asunto(s)
Densidad Ósea/fisiología , Metabolismo de los Hidratos de Carbono/fisiología , Factor I del Crecimiento Similar a la Insulina/metabolismo , Animales , Composición Corporal , Densidad Ósea/genética , Huesos/anatomía & histología , Huesos/fisiología , Metabolismo de los Hidratos de Carbono/genética , Femenino , Regulación de la Expresión Génica/fisiología , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/genética , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/deficiencia , Factor I del Crecimiento Similar a la Insulina/genética , Masculino , Ratones , Ratones Noqueados , Mutación , Aumento de Peso
11.
Biochimie ; 171-172: 147-157, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32105813

RESUMEN

The importance of cytochrome P450 (CYP)-derived arachidonic acid (AA) metabolites, 20-hydroxyeicosatetraenoic acid (20-HETE) and epoxyeicosatrienoic acids (EETs) as tumor growth promotors has already been described in several cancer types. The aim of this study was to evaluate the role of these compounds in the biology of pheochromocytoma/paraganglioma. These tumors originate from chromaffin cells derived from adrenal medulla (pheochromocytomas) or extra-adrenal autonomic paraganglia (paragangliomas), and they represent the most common hereditary endocrine neoplasia. According to mutations in the driver genes, these tumors are divided in two clusters: pseudo-hypoxic and kinase-signaling EETs, but not 20-HETE, exhibited a potent ability to sustain growth in a murine pheochromocytoma cell line (MPC) in vitro, EETs promoted an increase in cell proliferation and a decrease in cell apoptosis. In a mouse model of pheochromocytoma, the inhibition of CYP-mediated AA metabolism using 1-aminobenzotriazol resulted in slower tumor growth, a decreased vascularization, and a lower final volume. Also, the expression of AA-metabolizing CYP monooxygenases was detected in tumor samples from human origin, being their apparent abundance and the production of both metabolites higher in tumors from the kinase-signaling cluster. This is the first evidence of the importance of CYP- derived AA metabolites in the biology and development of pheochromocytoma/paraganglioma tumors.


Asunto(s)
Ácido 8,11,14-Eicosatrienoico/análogos & derivados , Neoplasias de las Glándulas Suprarrenales/inducido químicamente , Sistema Enzimático del Citocromo P-450/metabolismo , Ácidos Hidroxieicosatetraenoicos/farmacología , Feocromocitoma/inducido químicamente , Ácido 8,11,14-Eicosatrienoico/farmacología , Adolescente , Neoplasias de las Glándulas Suprarrenales/patología , Adulto , Animales , Línea Celular Tumoral , Niño , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Neovascularización Patológica , Feocromocitoma/patología , Adulto Joven
12.
Front Endocrinol (Lausanne) ; 11: 624684, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33613456

RESUMEN

Introduction: Practice guidelines cannot recommend establishing a diagnosis of growth hormone deficiency (GHD) without performing growth hormone stimulation tests (GHST) in children with risk factors, due to the lack of sufficient evidence. Objective: Our goal was to generate an evidence-based prediction rule to diagnose GHD in children with growth failure and clinically identifiable risk factors. Methods: We studied a cohort of children with growth failure to build the prediction model, and a second, independent cohort to validate the prediction rule. To this end, we assessed the existence of: pituitary dysgenesis, midline abnormalities, (supra)sellar tumor/surgery, CNS infection, traumatic brain injury, cranial radiotherapy, chemotherapy, genetic GHD, pituitary hormone deficiencies, and neonatal hypoglycemia, cholestasis, or hypogenitalism. Selection of variables for model building was performed using artificial intelligence protocols. Specificity of the prediction rule was the main outcome measure in the validation set. Results: In the first cohort (n=770), the resulting prediction rule stated that a patient would have GHD if (s)he had: pituitary dysgenesis, or two or more anterior pituitary deficiencies, or one anterior pituitary deficiency plus: neonatal hypoglycemia or hypogenitalism, or diabetes insipidus, or midline abnormalities, or (supra)sellar tumor/surgery, or cranial radiotherapy ≥18 Gy. In the validation cohort (n=161), the specificity of the prediction rule was 99.2% (95% CI: 95.6-100%). Conclusions: This clinical rule predicts the existence of GHD with high specificity in children with growth disorders and clinically identifiable risk factors, thus providing compelling evidence to recommend that GHD can be safely diagnosed without recurring to GHST in neonates and children with growth failure and specific comorbidities.


Asunto(s)
Algoritmos , Estatura/fisiología , Hormona de Crecimiento Humana/deficiencia , Aprendizaje Automático/normas , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Niño , Preescolar , Estudios de Cohortes , Enanismo Hipofisario/sangre , Enanismo Hipofisario/diagnóstico por imagen , Femenino , Humanos , Masculino , Guías de Práctica Clínica como Asunto/normas , Valor Predictivo de las Pruebas , Reproducibilidad de los Resultados , Factores de Riesgo
13.
Growth Horm IGF Res ; 50: 61-70, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31902742

RESUMEN

BACKGROUND: The most frequent monogenic causes of growth hormone insensitivity (GHI) include defects in genes encoding the GH receptor itself (GHR), the signal transducer and activator of transcription (STAT5B), the insulin like-growth factor type I (IGF1) and the acid-labile subunit (IGFALS). GHI is characterized by a continuum of mild to severe post-natal growth failure. OBJECTIVE: To characterize the molecular defect in a patient with short stature and partial GHI. PATIENT AND METHODS: The boy was born at term adequate for gestational age from non-consanguineous normal-stature parents. At 2.2 years, he presented proportionate short stature (height -2.77 SDS), wide forehead and normal mental development. Whole-exome analysis and functional characterization (site-directed mutagenesis, dual luciferase reporter assay, immunofluorescence and western immunoblot) were performed. RESULTS: Biochemical and endocrinological evaluation revealed partial GH insensitivity with normal stimulated GH peak (7.8 ng/mL), undetectable IGF1 and low IGFBP3 levels. Two heterozygous variants in the GH-signaling pathway were found: a novel heterozygous STAT5B variant (c.1896G>T, p.K632N) and a hypomorphic IGFALS variant (c.1642C>T, p.R548W). Functional in vitro characterization demonstrated that p.K632N-STAT5b is an inactivating variant that impairs STAT5b activity through abolished phosphorylation. Remarkably, the patient's immunological evaluation displayed only a mild hypogammaglobulinemia, while a major characteristic of STAT5b deficient patients is severe immunodeficiency. CONCLUSIONS: We reported a novel pathogenic inactivating STAT5b variant, which may be associated with partial GH insensitivity and can present without severe immunological complications in heterozygous state. Our results contribute to expand the spectrum of phenotypes associated to GHI.


Asunto(s)
Agammaglobulinemia/genética , Síndrome de Laron/genética , Factor de Transcripción STAT5/genética , Agammaglobulinemia/inmunología , Preescolar , Heterocigoto , Hormona de Crecimiento Humana/metabolismo , Humanos , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Síndrome de Laron/inmunología , Síndrome de Laron/metabolismo , Síndrome de Laron/fisiopatología , Masculino , Pruebas de Función Hipofisaria , Mutación Puntual , Índice de Severidad de la Enfermedad
14.
Biochimie ; 163: 108-116, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31185266

RESUMEN

Insulin-like growth factor 1 (IGF1) has a critical role in maintaining tumor phenotype and survival of already transformed murine pheochromocytoma (pheo) cells (MPC4/30) and it is required for the initial establishment of these tumors. However, the role of local IGF1/IGF1R system in tumor microenvironment has not been fully understood. In vivo, by subcutaneous injection of pheo cells in heterozygous IGF1R knockout mice (L/n), we found that the time of noticeable tumor appearance was delayed, and incidence was decreased in L/n group compared to control (L/L) mice. Once established, tumor proliferation, vascularization or growth rate did not differ between groups. In vitro, fibroblast from L/n and L/L mice were cultured to generate conditioned media (CM) and differential matrixes on which pheo cells were seeded. Proliferation rate was higher when pheo cells were cultured with CM, or in differential matrix generated by L/L murine fibroblasts. A diminished fibronectin (FN) expression and secretion from L/n fibroblast was associated with decreased expression of integrin subunits in tumor cells. Also, soluble factors as IGF1 and insulin-like growth factor binding protein 2 (IGFBP2) were reduced. Our data suggest that IGF1 signaling through IGF1R may contribute to tumor cells anchorage and survival by interaction with both matrix and soluble factors produced by tumor microenvironment fibroblasts.


Asunto(s)
Neoplasias de las Glándulas Suprarrenales/fisiopatología , Proliferación Celular , Fibroblastos/metabolismo , Haploinsuficiencia , Feocromocitoma/fisiopatología , Receptor IGF Tipo 1/genética , Microambiente Tumoral , Neoplasias de las Glándulas Suprarrenales/genética , Neoplasias de las Glándulas Suprarrenales/metabolismo , Animales , Fibronectinas/genética , Regulación Neoplásica de la Expresión Génica , Masculino , Ratones , Neovascularización Patológica , Feocromocitoma/genética , Feocromocitoma/metabolismo
15.
Eur J Endocrinol ; 181(5): K43-K53, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31539878

RESUMEN

BACKGROUND: IGF1 is a key factor in fetal and postnatal growth. To date, only three homozygous IGF1 gene defects leading to complete or partial loss of IGF1 activity have been reported in three short patients born small for gestational age. We describe the fourth patient with severe short stature presenting a novel homozygous IGF1 gene mutation. RESULTS: We report a boy born from consanguineous parents at 40 weeks of gestational age with intrauterine growth restriction and severe postnatal growth failure. Physical examination revealed proportionate short stature, microcephaly, facial dysmorphism, bilateral sensorineural deafness and mild global developmental delay. Basal growth hormone (GH) fluctuated from 0.2 to 29 ng/mL, while IGF1 levels ranged from -1.15 to 2.95 SDS. IGFBP3 was normal-high. SNP array delimited chromosomal regions of homozygosity, including 12q23.2 where IGF1 is located. IGF1 screening by HRM revealed a homozygous missense variant NM_000618.4(IGF1):c.322T>C, p.(Tyr108His). The change of the highly conserved Tyr60 in the mature IGF1 peptide was consistently predicted as pathogenic by multiple bioinformatic tools. Tyr60 has been described to be critical for IGF1 interaction with type 1 IGF receptor (IGF1R). In vitro, HEK293T cells showed a marked reduction of IGF1R phosphorylation after stimulation with serum from the patient as compared to sera from age-matched controls. Mutant IGF1 was also less efficient in inducing cell growth. CONCLUSION: The present report broadens the spectrum of clinical and biochemical presentation of homozygous IGF1 defects and underscores the variability these patients may present depending on the IGF/IGF1R pathway activity.


Asunto(s)
Trastornos del Crecimiento/genética , Pérdida Auditiva Sensorineural/genética , Factor I del Crecimiento Similar a la Insulina/deficiencia , Mutación Missense/genética , Anomalías Múltiples/genética , Proliferación Celular , Biología Computacional , Simulación por Computador , Retardo del Crecimiento Fetal/genética , Células HEK293 , Homocigoto , Humanos , Lactante , Factor I del Crecimiento Similar a la Insulina/genética , Masculino , Linaje , Polimorfismo de Nucleótido Simple/genética , Receptor IGF Tipo 1 , Receptores de Somatomedina/genética , Tirosina/genética
16.
Horm Cancer ; 9(3): 205-214, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29524179

RESUMEN

Nuclear localization of insulin-like growth factor receptor type 1 (IGF-1R) has been described as adverse prognostic factor in some cancers. We studied the expression and localization of IGF-1R in paediatric patients with gliomas, as well as its association with World Health Organization (WHO) grading and survival. We conducted a single cohort, prospective study of paediatric patients with gliomas. Samples were taken at the time of the initial surgery; IGF-1R expression and localization were characterized by immunohistochemistry (IHC), subcellular fractionation and western blotting. Tumours (47/53) showed positive staining for IGF-1R by IHC. IGF-1R nuclear labelling was observed in 10/47 cases. IGF-1R staining was mostly non-nuclear in low-grade tumours, while IGF-1R nuclear labelling was predominant in high-grade gliomas (p = 0.0001). Survival was significantly longer in patients with gliomas having non-nuclear IGF-1R localization than in patients with nuclear IGF-1R tumours (p = 0.016). In gliomas, IGF-1R nuclear localization was significantly associated with both high-grade tumours and increased risk of death. Based on a prospective design, we provide evidence of a potential usefulness of intracellular localization of IGF-1R as prognostic factor in paediatric patients with gliomas.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Encefálicas/metabolismo , Núcleo Celular/metabolismo , Glioma/metabolismo , Receptor IGF Tipo 1/metabolismo , Transporte Activo de Núcleo Celular , Adolescente , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/mortalidad , Niño , Preescolar , Estudios de Cohortes , Femenino , Glioma/diagnóstico , Glioma/mortalidad , Humanos , Inmunohistoquímica , Lactante , Masculino , Estadificación de Neoplasias , Pronóstico , Estudios Prospectivos , Receptor IGF Tipo 1/genética , Análisis de Supervivencia , Organización Mundial de la Salud
17.
Mol Cell Endocrinol ; 473: 166-177, 2018 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-29378236

RESUMEN

Germinal heterozygous activating STAT3 mutations represent a novel monogenic defect associated with multi-organ autoimmune disease and, in some cases, severe growth retardation. By using whole-exome sequencing, we identified two novel STAT3 mutations, p.E616del and p.C426R, in two unrelated pediatric patients with IGF-I deficiency and immune dysregulation. The functional analyses showed that both variants were gain-of-function (GOF), although they were not constitutively phosphorylated. They presented differences in their dephosphorylation kinetics and transcriptional activities under interleukin-6 stimulation. Both variants increased their transcriptional activities in response to growth hormone (GH) treatment. Nonetheless, STAT5b transcriptional activity was diminished in the presence of STAT3 GOF variants, suggesting a disruptive role of STAT3 GOF variants in the GH signaling pathway. This study highlights the broad clinical spectrum of patients presenting activating STAT3 mutations and explores the underlying molecular pathway responsible for this condition, suggesting that different mutations may drive increased activity by slightly different mechanisms.


Asunto(s)
Células Germinativas/metabolismo , Trastornos del Crecimiento/genética , Pérdida Auditiva Sensorineural/genética , Enfermedades del Sistema Inmune/genética , Factor I del Crecimiento Similar a la Insulina/deficiencia , Mutación/genética , Factor de Transcripción STAT3/genética , Secuencia de Aminoácidos , Preescolar , Femenino , Células HEK293 , Hormona de Crecimiento Humana/farmacología , Humanos , Lactante , Recién Nacido , Factor I del Crecimiento Similar a la Insulina/genética , Interleucina-5/metabolismo , Luciferasas/metabolismo , Masculino , Modelos Moleculares , Fosforilación/efectos de los fármacos , Multimerización de Proteína , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factor de Transcripción STAT3/química , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT5/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas/genética , Proteína 3 Supresora de la Señalización de Citocinas/metabolismo , Transcripción Genética/efectos de los fármacos , Secuenciación del Exoma
18.
Endocrinology ; 147(6): 2619-30, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16513827

RESUMEN

IGF-I and insulin are structurally related polypeptides that mediate a similar pattern of biological effects via receptors that display considerably homology. Administration of recombinant human IGF-I (rhIGF-I) has been proven to improve glucose control and liver and muscle insulin sensitivity in patients with type 2 diabetes mellitus (DM). The effect of rhIGF-I treatment was evaluated in a mouse model of type 2 DM (MKR mouse), which expresses a dominant-negative form of the human IGF-I receptor under the control of the muscle creatine kinase promoter specifically in skeletal muscle. MKR mice have impaired IGF-I and insulin signaling in skeletal muscle, leading to severe insulin resistance in muscle, liver, and fat, developing type 2 DM at 5 wk of age. Six-week-old MKR mice were treated with either saline or rhIGF-I for 3 wk. Blood glucose levels were decreased in response to rhIGF-I treatment in MKR mice. rhIGF-I treatment also increased body weight in MKR with concomitant changes in body composition such as a decrease in fat mass and an increase in lean body mass. Insulin, fatty acid, and triglyceride levels were not affected by rhIGF-I, nor were insulin or glucose tolerance in MKR mice. Hyperinsulinemic-euglycemic clamp analysis demonstrated no improvement in overall insulin sensitivity. Pyruvate and glutamine tolerance tests proved that there was a decrease in the rate of glucose appearance in MKR mice treated with rhIGF-I, suggesting a reduction in the gluconeogenic capacity of liver, kidney, and small intestine. Taken together these results demonstrate that the improvement of the hyperglycemia was achieved by inhibition of gluconeogenesis rather than an improvement in insulin sensitivity. Also, these results suggest that a functional IGF-I receptor in skeletal muscle is required for IGF-I to improve insulin sensitivity in this mouse model of type 2 DM.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Gluconeogénesis/efectos de los fármacos , Factor I del Crecimiento Similar a la Insulina/uso terapéutico , Animales , Glucemia/análisis , Composición Corporal/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Diabetes Mellitus Tipo 2/metabolismo , Ingestión de Líquidos/efectos de los fármacos , Técnica de Clampeo de la Glucosa , Glucosa-6-Fosfatasa/genética , Hormona del Crecimiento/sangre , Resistencia a la Insulina , Factor I del Crecimiento Similar a la Insulina/farmacología , Intestino Delgado/metabolismo , Riñón/metabolismo , Hígado/metabolismo , Masculino , Ratones , Fosfoenolpiruvato Carboxiquinasa (GTP)/genética , Proteínas Recombinantes/uso terapéutico
19.
Endocrinology ; 147(12): 5826-34, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16959846

RESUMEN

Obesity increases the risk of many cancers in both males and females. This study describes a link between obesity, obesity-associated metabolic alterations, and the risk of developing cancer in male and female mice. The goal of this study was to evaluate the relationship between gender and obesity and to determine the role of estrogen status in obese females and its effect on tumor growth. We examined the susceptibility of C57BL/6 mice to diet-induced obesity, insulin resistance/glucose intolerance, and tumors. Mice were injected sc with one of two tumorigenic cell lines, Lewis lung carcinoma, or mouse colon 38-adenocarcinoma. Results show that tumor growth rate was increased in obese mice vs. control mice irrespective of the tumor cell type. To investigate the effect of estrogen status on tumor development in obese females, we compared metabolic parameters and tumor growth in ovariectomized (ovx) and intact obese female mice. Obese ovx female mice developed insulin resistance and glucose intolerance similar to that observed in obese males. Our results demonstrate that body adiposity increased in ovx females irrespective of the diet administered and that tumor growth correlated positively with body adiposity. Overall, these data point to more rapid tumor growth in obese mice and suggest that endogenous sex steroids, together with diet, affect adiposity, insulin sensitivity, and tumor growth in female mice.


Asunto(s)
Dieta/efectos adversos , Hormonas Gonadales/fisiología , Neoplasias Experimentales/complicaciones , Neoplasias Experimentales/patología , Obesidad/complicaciones , Obesidad/etiología , Adenocarcinoma/patología , Animales , Movimiento Celular , Proliferación Celular , Neoplasias del Colon/patología , Estrógenos/sangre , Estrógenos/fisiología , Femenino , Hormonas Gonadales/sangre , Humanos , Neoplasias Pulmonares/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Obesidad/sangre , Ovario/fisiología , Células Tumorales Cultivadas , Ensayo de Tumor de Célula Madre , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Cancer Res ; 62(22): 6529-37, 2002 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-12438247

RESUMEN

Several lines of evidence support an important role for the insulin-like growth factor system in breast cancer. Alterations in insulin-like growth factor I receptor (IGF-IR) have been associated with breast cancer metastasis; however, the specific role played by the IGF-IR in this process remains unclear. To address this issue, we evaluated MCF-7 breast cancer cells stably transfected either with an antisense construct to the IGF-IR, which reduced the expression of the IGF-IRs by approximately 50% (SX13 cells), or with the empty vector as control (NEO cells). Using functional assays for motility, attachment, and aggregation, we found a 3-fold increase in migration using both the wounding assay and the Boyden chamber migration assay. In addition, the SX13 cells attached less, and there was a reduction in cellular aggregation. These functional changes were accompanied by approximately 50% decrease in expression of E-cadherin and approximately 80% increase in p120 protein levels. Moreover, there was a significant reduction in p120 present in the E-cadherin-catenin-p120 complex. There was a 2-fold increase in active Rac1 and Cdc42 and a 35% decrease in active Rho in the SX13 cells. Our findings strongly suggest that the IGF-IR plays a role in the stabilization of the E-cadherin-catenin complex, thereby providing one possible explanation for the association between low levels of IGF-IR and a higher risk of mammary tumor metastasis.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Receptor IGF Tipo 1/fisiología , Neoplasias de la Mama/genética , Adhesión Celular/genética , Adhesión Celular/fisiología , Agregación Celular/fisiología , Movimiento Celular/genética , Movimiento Celular/fisiología , ADN sin Sentido/genética , Humanos , Metástasis de la Neoplasia , Receptor IGF Tipo 1/biosíntesis , Receptor IGF Tipo 1/genética , Transfección , Células Tumorales Cultivadas , Proteína de Unión al GTP cdc42/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA