Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.440
Filtrar
Más filtros











Intervalo de año de publicación
1.
Horm Res Paediatr ; 95(6): 619-630, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36446332

RESUMEN

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.


Asunto(s)
Factor II del Crecimiento Similar a la Insulina , Factor I del Crecimiento Similar a la Insulina , Síndrome de Laron , Animales , Humanos , Factor I del Crecimiento Similar a la Insulina/deficiencia , Factor I del Crecimiento Similar a la Insulina/historia , Factor I del Crecimiento Similar a la Insulina/fisiología , Factor I del Crecimiento Similar a la Insulina/uso terapéutico , Síndrome de Laron/tratamiento farmacológico , Síndrome de Laron/genética , Síndrome de Laron/historia , Síndrome de Laron/fisiopatología , Hormonas Peptídicas , Procesamiento Proteico-Postraduccional , Transducción de Señal , Somatomedinas/deficiencia , Somatomedinas/historia , Somatomedinas/fisiología , Factor II del Crecimiento Similar a la Insulina/deficiencia , Factor II del Crecimiento Similar a la Insulina/historia , Factor II del Crecimiento Similar a la Insulina/fisiología , Factor II del Crecimiento Similar a la Insulina/uso terapéutico
2.
Cells ; 11(20)2022 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-36291127

RESUMEN

The growth hormone (GH)-insulin-like growth factor-1 (IGF1) signaling pathway plays a major role in orchestrating cellular interactions, metabolism, growth and aging. Studies from worms to mice showed that downregulated activity of the GH/IGF1 pathway could be beneficial for the extension of lifespan. Laron syndrome (LS) is an inherited autosomal recessive disorder caused by molecular defects of the GH receptor (GHR) gene, leading to congenital IGF1 deficiency. Life-long exposure to minute endogenous IGF1 levels in LS is associated with low stature as well as other endocrine and metabolic deficits. Epidemiological surveys reported that patients with LS have a reduced risk of developing cancer. Studies conducted on LS-derived lymphoblastoid cells led to the identification of a novel link between IGF1 and thioredoxin-interacting protein (TXNIP), a multifunctional mitochondrial protein. TXNIP is highly expressed in LS patients and plays a critical role in cellular redox regulation by thioredoxin. Given that IGF1 affects the levels of TXNIP under various stress conditions, including high glucose and oxidative stress, we hypothesized that the IGF1-TXNIP axis plays an essential role in helping maintain a physiological balance in cellular homeostasis. In this study, we show that TXNIP is vital for the cell fate choice when cells are challenged by various stress signals. Furthermore, prolonged IGF1 treatment leads to the establishment of a premature senescence phenotype characterized by a unique senescence network signature. Combined IGF1/TXNIP-induced premature senescence can be associated with a typical secretory inflammatory phenotype that is mediated by STAT3/IL-1A signaling. Finally, these mechanistic insights might help with the understanding of basic aspects of IGF1-related pathologies in the clinical setting.


Asunto(s)
Proteínas Portadoras , Senescencia Celular , Factor I del Crecimiento Similar a la Insulina , Síndrome de Laron , Tiorredoxinas , Animales , Ratones , Proteínas Portadoras/metabolismo , Senescencia Celular/efectos de los fármacos , Senescencia Celular/fisiología , Glucosa/metabolismo , Hormona del Crecimiento/metabolismo , Factor I del Crecimiento Similar a la Insulina/farmacología , Factor I del Crecimiento Similar a la Insulina/fisiología , Síndrome de Laron/metabolismo , Proteínas Mitocondriales/metabolismo , Tiorredoxinas/metabolismo , Humanos , Fibroblastos/efectos de los fármacos , Células 3T3-L1
3.
Front Endocrinol (Lausanne) ; 13: 852382, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35370981

RESUMEN

Prostate cancer (PCa) is the most common malignancy in men worldwide, thus developing effective prevention strategies remain a critical challenge. Insulin-like growth factor 1 (IGF-1) is produced mainly in the liver by growth hormone signaling and is necessary for normal physical growth. However, several studies have shown an association between increased levels of circulating IGF-1 and the risk of developing solid malignancies, including PCa. Because the IGF-1 receptor is overexpressed in PCa, IGF-1 can accelerate PCa growth by activating phosphoinositide 3-kinase and mitogen-activated protein kinase, or increasing sex hormone sensitivity. Short-chain fatty acids (SCFAs) are beneficial gut microbial metabolites, mainly because of their anti-inflammatory effects. However, we have demonstrated that gut microbiota-derived SCFAs increase the production of IGF-1 in the liver and prostate. This promotes the progression of PCa by the activation of IGF-1 receptor downstream signaling. In addition, the relative abundance of SCFA-producing bacteria, such as Alistipes, are increased in gut microbiomes of patients with high-grade PCa. IGF-1 production is therefore affected by the gut microbiome, dietary habits, and genetic background, and may play a central role in prostate carcinogenesis. The pro-tumor effects of bacteria and diet-derived metabolites might be potentially countered through dietary regimens and supplements. The specific diets or supplements that are effective are unclear. Further research into the "Gut-IGF-1-Prostate Axis" may help discover optimal diets and nutritional supplements that could be implemented for prevention of PCa.


Asunto(s)
Microbioma Gastrointestinal , Factor I del Crecimiento Similar a la Insulina , Carcinogénesis , Microbioma Gastrointestinal/fisiología , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/fisiología , Masculino , Fosfatidilinositol 3-Quinasas , Próstata
4.
Int J Mol Sci ; 22(20)2021 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-34681810

RESUMEN

There is considerable evidence of a positive association between the incidence of type 2 diabetes mellitus (T2DM) and obesity with bladder cancer (BCa), with the link between T2DM and obesity having already been established. There also appear to be potential associations between Pleckstrin homology domain containing S1 (PLEKHS1) and the Insulin-like Growth Factor (IGF) axis. Seven literature searches were carried out to investigate the backgrounds of these potential links. PLEKHS1 is a candidate biomarker in BCa, with mutations that are easily detectable in urine and increased expression seemingly associated with worse disease states. PLEKHS1 has also been implicated as a potential mediator for the onset of T2DM in people with obesity. The substantial evidence of the involvement of IGF in BCa, the role of the IGF axis in obesity and T2DM, and the global prevalence of T2DM and obesity suggest there is scope for investigating the links between these components. Preliminary findings on the relationship between PLEKHS1 and the IGF axis signal possible associations with BCa progression. This indicates that PLEKHS1 plays a role in the pathogenesis of BCa that may be mediated by members of the IGF axis. Further detailed research is needed to establish the relationship between PLEKHS1 and the IGF axis in BCa and determine how these phenomena overlap with T2DM and obesity.


Asunto(s)
Diabetes Mellitus Tipo 2/complicaciones , Obesidad/complicaciones , Neoplasias de la Vejiga Urinaria/etiología , Animales , Biomarcadores de Tumor/genética , Diabetes Mellitus Tipo 2/epidemiología , Diabetes Mellitus Tipo 2/genética , Humanos , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/fisiología , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/fisiología , Obesidad/epidemiología , Obesidad/genética , Factores de Riesgo , Transducción de Señal/genética , Neoplasias de la Vejiga Urinaria/epidemiología , Neoplasias de la Vejiga Urinaria/genética
5.
Int J Mol Sci ; 22(17)2021 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-34502376

RESUMEN

Inflammation induces a wide response of the neuroendocrine system, which leads to modifications in all the endocrine axes. The hypothalamic-growth hormone (GH)-insulin-like growth factor-1 (IGF-1) axis is deeply affected by inflammation, its response being characterized by GH resistance and a decrease in circulating levels of IGF-1. The endocrine and metabolic responses to inflammation allow the organism to survive. However, in chronic inflammatory conditions, the inhibition of the hypothalamic-GH-IGF-1 axis contributes to the catabolic process, with skeletal muscle atrophy and cachexia. Here, we review the changes in pituitary GH secretion, IGF-1, and IGF-1 binding protein-3 (IGFBP-3), as well as the mechanism that mediated those responses. The contribution of GH and IGF-1 to muscle wasting during inflammation has also been analyzed.


Asunto(s)
Caquexia/metabolismo , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Caquexia/fisiopatología , Hormona del Crecimiento/metabolismo , Hormona de Crecimiento Humana/metabolismo , Humanos , Hipotálamo/metabolismo , Inflamación/fisiopatología , Insulina/metabolismo , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/fisiología , Factor I del Crecimiento Similar a la Insulina/fisiología , Atrofia Muscular/metabolismo , Atrofia Muscular/fisiopatología
6.
Commun Biol ; 4(1): 775, 2021 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-34163008

RESUMEN

Patients with long-standing diabetes have a high risk for cardiac complications that is exacerbated by increased reactive oxygen species (ROS) production. We found that feeding cyanocobalamin (B12), a scavenger of superoxide, not only prevented but reversed signs of cardiomyopathy in type 1 diabetic Elmo1H/H Ins2Akita/+ mice. ROS reductions in plasma and hearts were comparable to those in mice treated with other antioxidants, N-acetyl-L-cysteine or tempol, but B12 produced better cardioprotective effects. Diabetes markedly decreased plasma insulin-like growth factor (IGF)-1 levels, while B12, but not N-acetyl-L-cysteine nor tempol, restored them. B12 activated hepatic IGF-1 production via normalization of S-adenosylmethionine levels, DNA methyltransferase (DNMT)-1/3a/3b mRNA, and DNA methylation of promoters for suppressor of cytokine signaling (SOCS)-1/3. Reductions of cardiac IGF-1 mRNA and phosphorylated IGF-1 receptors were also restored. Thus, B12 is a promising option for preventing diabetic cardiomyopathy via ROS reduction and IGF-1 retrieval through DNMT-SOCS1/3 signaling.


Asunto(s)
Cardiomiopatías/prevención & control , ADN-Citosina Metilasas/fisiología , Diabetes Mellitus Tipo 1/complicaciones , Factor I del Crecimiento Similar a la Insulina/fisiología , Estrés Oxidativo/efectos de los fármacos , Proteína 1 Supresora de la Señalización de Citocinas/fisiología , Proteína 3 Supresora de la Señalización de Citocinas/fisiología , Vitamina B 12/farmacología , Proteínas Adaptadoras Transductoras de Señales/fisiología , Animales , Diabetes Mellitus Tipo 1/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/efectos de los fármacos
7.
FASEB J ; 35(5): e21563, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33818810

RESUMEN

One of the endogenous estrogens, 17ß-estradiol (E2 ) is a female steroid hormone secreted from the ovary. It is well established that E2 causes biochemical and histological changes in the uterus. However, it is not completely understood how E2 regulates the oviductal environment in vivo. In this study, we assessed the effect of E2 on each oviductal cell type, using an ovariectomized-hormone-replacement mouse model, single-cell RNA-sequencing (scRNA-seq), in situ hybridization, and cell-type-specific deletion in mice. We found that each cell type in the oviduct responded to E2 distinctively, especially ciliated and secretory epithelial cells. The treatment of exogenous E2 did not drastically alter the transcriptomic profile from that of endogenous E2 produced during estrus. Moreover, we have identified and validated genes of interest in our datasets that may be used as cell- and region-specific markers in the oviduct. Insulin-like growth factor 1 (Igf1) was characterized as an E2 -target gene in the mouse oviduct and was also expressed in human fallopian tubes. Deletion of Igf1 in progesterone receptor (Pgr)-expressing cells resulted in female subfertility, partially due to an embryo developmental defect and embryo retention within the oviduct. In summary, we have shown that oviductal cell types, including epithelial, stromal, and muscle cells, are differentially regulated by E2 and support gene expression changes, such as growth factors that are required for normal embryo development and transport in mouse models. Furthermore, we have identified cell-specific and region-specific gene markers for targeted studies and functional analysis in vivo.


Asunto(s)
Biomarcadores/metabolismo , Estradiol/farmacología , Trompas Uterinas/fisiología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Factor I del Crecimiento Similar a la Insulina/fisiología , Oviductos/fisiología , Análisis de la Célula Individual/métodos , Animales , Estrógenos/farmacología , Trompas Uterinas/citología , Trompas Uterinas/efectos de los fármacos , Femenino , Perfilación de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oviductos/citología , Oviductos/efectos de los fármacos , Receptores de Progesterona/fisiología
8.
Otolaryngol Head Neck Surg ; 165(6): 757-758, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-33781112

RESUMEN

Teprotumumab is a human monoclonal antibody and IGF-1R (insulin-like growth factor 1 receptor) inhibitor approved for treatment of thyroid eye disease in adults. Recent clinical trials have demonstrated side effects, notably hearing loss, in the treatment cohort as compared with the placebo cohort. These unexpected otologic side effects may be understood through a mechanistic understanding of IGF-1 (insulin-like growth factor 1). As otolaryngologists who historically play a significant role in the multidisciplinary treatment of thyroid disease and its associated complications, we should be aware of and monitor the otologic side effects of teprotumumab. Clinicians who prescribe teprotumumab should strongly consider monitoring patients' hearing with an audiologist and otolaryngologist.


Asunto(s)
Anticuerpos Monoclonales Humanizados/efectos adversos , Oftalmopatía de Graves/tratamiento farmacológico , Pérdida Auditiva/inducido químicamente , Factor I del Crecimiento Similar a la Insulina/fisiología , Receptor IGF Tipo 1/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales Humanizados/uso terapéutico , Humanos , Ratones , Otorrinolaringólogos
10.
Ann Endocrinol (Paris) ; 82(2): 107-111, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33727117

RESUMEN

Acromegaly is an endocrine disease that leads to elevated production and secretion of growth hormone (GH). It can occur in adult and aged cats and is usually associated with neoplasms, such as functional pituitary macroadenoma of somatotropic cells. In dogs it is usually related to an increase in serum progesterone that induces production of GH by the mammary glands. The main clinical signs are related to insulin resistance and the anabolic effect induced by GH: polyuria, polydipsia, polyphagia, increased tissue growth, weight gain, prognathism, and other changes. The condition can be diagnosed from clinical signals and imaging associated to measurement of serum concentrations of GH and insulin-like growth factor 1 (IGF-1, also known as somatomedin C). The main therapeutic modalities are radiotherapy, hypophysectomy, and several drugs such as somatostatin analogs, dopaminergic agonists and GH receptor antagonists. The present review aims to provide a relevant animal model of acromegaly with an update on the therapeutic approach that may help clinicians to consider the GH axis-IGF-1 system, its pathogenesis and the clinical signs induced by this hormonal disorder.


Asunto(s)
Acromegalia/veterinaria , Enfermedades de los Gatos , Enfermedades de los Perros , Acromegalia/diagnóstico , Acromegalia/terapia , Animales , Enfermedades de los Gatos/diagnóstico , Enfermedades de los Gatos/terapia , Gatos , Enfermedades de los Perros/diagnóstico , Enfermedades de los Perros/terapia , Perros , Agonistas de Dopamina/uso terapéutico , Hormona del Crecimiento/metabolismo , Hipofisectomía , Resistencia a la Insulina , Factor I del Crecimiento Similar a la Insulina/fisiología , Radioterapia , Somatostatina/análogos & derivados
11.
J Endocrinol ; 248(1): 17-30, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33112794

RESUMEN

Both estrogen and hydrogen sulfide (H2S) inhibit the proliferation of vascular smooth muscle cells (SMCs) and development of atherosclerosis. In the absence of endogenous H2S as occurred in CSE-knockout (KO) mouse, however, estrogen stimulates the proliferation of vascular SMCs. The underlying mechanisms for this seemingly controversial vascular effect of estrogen are unclear. In the present study, we demonstrated that the stimulatory effect of estrogen on the proliferation of CSE-KO SMCs was suppressed by the inhibitor of insulin-like growth factor-1 receptor (IGF-1R) or knockdown of IGF-1R protein expression. Estrogen downregulated the expression of insulin-like growth factor-1 (IGF-1) and IGF-1R in aortic tissues or aortic SMCs isolated from WT and CSE-KO mice. Furthermore, endogenous H2S downregulated IGF-1R, but upregulated estrogen receptor (ER)-α, in aortic tissues or SMCs. ER-α and IGF-1R were co-located in SMCs and co-immunoprecipitated, which was decreased by H2S. Finally, both endogenous and exogenous H2S induced the S-sulfhydration of IGF-1R, but not ER-α, in WT-SMCs and CSE-KO SMCs, which underlies the decreased formation of IGF-1R/ER-α hybrid in the presence of H2S. Thus, the absence of H2S favors the interaction of estrogen with IGF-1R/ER-α hybrid to stimulate SMCs proliferation. The appreciation of a critical role of H2S in preventing estrogen-induced SMCs proliferation will help better understand the regulation of complex vascular effects of estrogen and sex-related cardiovascular diseases.


Asunto(s)
Proliferación Celular , Estradiol/fisiología , Sulfuro de Hidrógeno/metabolismo , Factor I del Crecimiento Similar a la Insulina/fisiología , Miocitos del Músculo Liso/fisiología , Animales , Aorta/metabolismo , Receptor alfa de Estrógeno/metabolismo , Femenino , Masculino , Ratones Noqueados , Proteína Fosfatasa 2/metabolismo , Receptor IGF Tipo 1/metabolismo
12.
J Endocrinol ; 248(1): 31-44, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33112796

RESUMEN

A reduction in hepatocyte growth hormone (GH)-signaling promotes non-alcoholic fatty liver disease (NAFLD). However, debate remains as to the relative contribution of the direct effects of GH on hepatocyte function vs indirect effects, via alterations in insulin-like growth factor 1 (IGF1). To isolate the role of hepatocyte GH receptor (GHR) signaling, independent of changes in IGF1, mice with adult-onset, hepatocyte-specific GHR knockdown (aHepGHRkd) were treated with a vector expressing rat IGF1 targeted specifically to hepatocytes. Compared to GHR-intact mice, aHepGHRkd reduced circulating IGF1 and elevated GH. In male aHepGHRkd, the shift in IGF1/GH did not alter plasma glucose or non-esterified fatty acids (NEFA), but was associated with increased insulin, enhanced systemic lipid oxidation and reduced white adipose tissue (WAT) mass. Livers of male aHepGHRkd exhibited steatosis associated with increased de novo lipogenesis, hepatocyte ballooning and inflammation. In female aHepGHRkd, hepatic GHR protein levels were not detectable, but moderate levels of IGF1 were maintained, with minimal alterations in systemic metabolism and no evidence of steatosis. Reconstitution of hepatocyte IGF1 in male aHepGHRkd lowered GH and normalized insulin, whole body lipid utilization and WAT mass. However, IGF1 reconstitution did not reduce steatosis or eliminate liver injury. RNAseq analysis showed IGF1 reconstitution did not impact aHepGHRkd-induced changes in liver gene expression, despite changes in systemic metabolism. These results demonstrate the impact of aHepGHRkd is sexually dimorphic and the steatosis and liver injury observed in male aHepGHRkd mice is autonomous of IGF1, suggesting GH acts directly on the adult hepatocyte to control NAFLD progression.


Asunto(s)
Hígado Graso/etiología , Hormona del Crecimiento/fisiología , Hepatocitos/fisiología , Factor I del Crecimiento Similar a la Insulina/fisiología , Hígado/metabolismo , Animales , Femenino , Metabolismo de los Lípidos , Masculino , Ratones , Receptores de Somatotropina/fisiología , Caracteres Sexuales , Somatotrofos/metabolismo
13.
Mol Cell Endocrinol ; 518: 111003, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32919021

RESUMEN

Emerging evidence links the growth hormone (GH)-insulin-like growth factor-1 (IGF1) endocrine axis to cancer development. While this putative correlation is of major translational relevance, most clinical and epidemiological reports to date found no causal linkage between GH therapy and enhanced cancer risk. Thus, it is generally agreed that GH therapy constitutes a safe pharmacological intervention. The present review focuses on a number of issues in the area of GH-IGF1 action in cancer development. Emphasis is given to the idea that GH and IGF1 do not conform to the definition of oncogenic factors. Specifically, these hormones, even at high pharmacological doses, are unable to induce malignant transformation. However, the GH-IGF1 axis is capable of 'pushing' already transformed cells through the various phases of the cell cycle. Viral and cellular oncogenes require an intact IGF1 signaling pathway in order to elicit transformation; in other words, oncogenic agents adopt the IGF1 pathway. This universal mechanism of action of oncogenes has broad implications in oncology. Our review provides an in-depth analysis of the interplay between the GH-IGF1 axis and cancer genes, including tumor suppressors p53 and BRCA1. Finally, the safety of GH therapy in both children and adults needs further long-term follow-up studies.


Asunto(s)
Hormona de Crecimiento Humana/fisiología , Factor I del Crecimiento Similar a la Insulina/fisiología , Neoplasias/patología , Adulto , Ciclo Celular/genética , Niño , Progresión de la Enfermedad , Humanos , Neoplasias/genética , Oncogenes/fisiología , Transducción de Señal/fisiología
14.
Curr Opin Otolaryngol Head Neck Surg ; 28(5): 286-290, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32796270

RESUMEN

PURPOSE OF REVIEW: Insulin-like growth factor 1 (IGF-1) is a hormone necessary for the development, growth, and maintenance of various organs, and has been used as a therapeutic agent in clinical settings. This review aimed to illustrate its role in the auditory systems and its potential use as a therapeutic in the field of otology. RECENT FINDINGS: Previous animal studies have indicated the critical role of IGF-1 in the development and maintenance of the auditory system, especially in the cochlea. A clinical study demonstrated a close relationship between the serum level of IGF-1 and the progression of age-related hearing impairment, suggesting its importance in the maintenance of hearing in humans. More recently, its effect on the regeneration of cochlear synapses has been reported using explant cultures, which could explain the course of hearing recovery in patients who underwent topical IGF-1 application for the treatment of sudden sensorineural hearing loss. SUMMARY: Recent advances in experimental and clinical investigations have revealed the importance of IGF-1 in the maintenance of the auditory function. On the basis of broad targets, its clinical application will expand to the field of otology in the future.


Asunto(s)
Enfermedades del Oído/terapia , Factor I del Crecimiento Similar a la Insulina/fisiología , Enfermedades del Oído/diagnóstico , Enfermedades del Oído/etiología , Humanos
15.
Growth Horm IGF Res ; 53-54: 101330, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32693362

RESUMEN

IGF-I was originally discovered as a GH-dependent growth factor stimulating longitudinal growth. Currently, however, it has become evident that the biological activities of IGF-I extend well beyond those of a simple growth factor and impact such processes as insulin sensitivity, aging, cancer and cardiovascular disease. The vast majority of IGF-I is tightly bound to IGF-binding proteins (IGFBPs), which renders IGF-I unable to stimulate the IGF-I receptor (IGF-IR) in vivo. This binding means that liberation of IGF-I from the IGFBPs is an important step controlling IGF-I action. In this context, IGFBP-cleaving enzymes appear to play a key role. Enzymatic cleavage of the IGFBPs markedly lowers their ligand affinity, and as a consequence, IGF-I becomes liberated and hence available for stimulation of the IGF-IR. Two of the best-characterized IGFBP-cleaving enzymes are pregnancy-associated plasma protein-A (PAPP-A) and its paralog PAPP-A2. The two enzymes (often referred to as pappalysins) regulate the liberation of IGF-I in a highly controlled manner. PAPP-A is believed to act predominantly in tissues, serving to liberate IGF-I at the cell surface in close proximity to the IGF-IR. In keeping with this notion, mice lacking PAPP-A exhibit reduced body size, despite having normal circulating IGF-I concentrations. In contrast, human findings indicate that altered PAPP-A2 activity changes circulating IGF-I concentrations, although PAPP-A2 is also present in high concentrations in tissues. Thus, PAPP-A2 appears to impact circulating, as well as tissue, IGF-I activity. The enzymatic activity of PAPP-A and PAPP-A2 was recently discovered to be regulated by the protein Stanniocalcin-2 (STC2). By binding to the enzymatic sites of PAPP-A and PAPP-A2, STC2 inhibits their activity. To date, the majority of findings demonstrating the ability of pappalysins and STC2 to regulate IGF-I action are from preclinical studies. However, clinical studies are now beginning to emerge. In this review, we will summarize our data on STC2, PAPP-A and PAPP-A2 in humans. These results indicate that pappalysins and STC2 constitute an important IGF-I activity-regulating system that warrants further investigation.


Asunto(s)
Glicoproteínas/metabolismo , Factor I del Crecimiento Similar a la Insulina/fisiología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteína Plasmática A Asociada al Embarazo/metabolismo , Animales , Femenino , Humanos , Ratones , Embarazo
16.
Reprod Fertil Dev ; 32(10): 929-940, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32586423

RESUMEN

The critical role of insulin-like growth factor (IGF) 1 in promoting Sertoli cell proliferation invivo and invitro has been established, but its downstream signalling mechanisms remain unknown. In addition to mitogenic effects, a role for IGF1 in mediating cholesterol biosynthesis within testes has been implied. The aims of this study were to investigate the roles of: (1) phosphatidylinositol 3-kinase/Akt/mammalian target of rapamycin (mTOR) signalling in IGF1-mediated Sertoli cell proliferation; and (2) IGF1 in mediating cholesterol biosynthesis in Sertoli cells. Primary cultures of Sertoli cells were prepared from 1-week-old porcine testes. On Day 3 of culture, Sertoli cells were treated with 300ng mL-1 IGF1, alone or in combination with inhibitors of IGF1 receptor (2µM picropodophyllotoxin), Akt (1µM wortmannin) or mTOR (200nM rapamycin). Cells were cultured for 30min and phosphorylation levels of Akt, mTOR and p70 ribosomal protein S6 kinase (p70S6K) were determined by immunoblotting. Cell proliferation and quantitative polymerase chain reaction assays were conducted using cells cultured for 24h. IGF1 increased phosphorylation of Akt, mTOR and p70S6K and cell proliferation, and these effects were inhibited by inhibitors of IGF1R, Akt and mTOR. Furthermore, IGF1 upregulated the expression of cholesterol biosynthetic genes (3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR), 3-hydroxy-3-methylglutaryl-CoA synthase (HMGCS1) and cytochrome P450, family 5, subfamily A, polypeptide 1 (CYP5A1)), but not sterol regulatory element-binding transcription factor 1 (SREBF1). Increased phosphorylation of p70S6K, a major downstream target of mTOR, and upregulated expression of genes involved in cholesterol biosynthesis are indicative of the key role played by IGF1 in regulating the synthesis of cholesterol, the precursor for steroid hormones.


Asunto(s)
Proliferación Celular/fisiología , Factor I del Crecimiento Similar a la Insulina/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Células de Sertoli/fisiología , Transducción de Señal/fisiología , Serina-Treonina Quinasas TOR/metabolismo , Animales , Células Cultivadas , Colesterol/biosíntesis , Colesterol/genética , Expresión Génica/efectos de los fármacos , Factor I del Crecimiento Similar a la Insulina/farmacología , Masculino , Modelos Animales , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Receptor IGF Tipo 1/antagonistas & inhibidores , Receptor IGF Tipo 1/fisiología , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Transducción de Señal/efectos de los fármacos , Sus scrofa , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Testículo/citología
17.
Front Immunol ; 11: 684, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32425932

RESUMEN

Background: Colorectal cancer (CRC) is frequently associated with dysbiosis of the gut microbiome which, together with a compromised gut barrier, can result in perioperative endotoxin leakage into the circulation. Constant local and systemic inflammatory activity is suggested to facilitate metastases formation. Previous studies have pointed to the capacity of a colostrum preparation to neutralize endotoxins within the gastrointestinal tract which could ameliorate associated inflammatory responses and tumor recurrence in affected patients. This study aimed to examine the effects of the colostrum preparation, KMP01D, on the inflammatory activity of patient-derived immune cells. Methods: The effects of KMP01D on pro-/anti-inflammatory cytokine responses and apoptosis were examined ex vivo using immune cells from CRC patients (stages I-IV, n = 48). The expression of CD14, CD68, Toll-like receptor (TLR)4, and insulin-like growth factor (IGF)-1 was also analyzed. Results: KMP01D increased interleukin (IL)-10 and IL-13 anti-inflammatory cytokine expression in patient-derived peripheral blood mononuclear cells (PBMCs). Interestingly, KMP01D also decreased the secretion of IL-1ß, IL-6, interferon (IFN)-γ, tumor necrosis factor (TNF)-α, IL-12 inflammatory cytokines, and IGF-1 in these cells. Moreover, CD14 and TLR4 expression involved in endotoxin signaling was downregulated in PBMCs and tumor-derived cells. Apoptosis of immune cells and tumor-derived cells was likewise enhanced with KMP01D. Addition of vitamin D3 as a cofactor demonstrated enhanced anti-inflammatory effects. Conclusions: KMP01D demonstrated beneficial ex vivo effects on inflammatory cytokine responses in PBMCs and enhanced apoptosis of immune cells from CRC patients. In line with previous clinical trials, we present new evidence endorsing KMP01D as a treatment strategy to regulate stage-dependent local and systemic inflammation in CRC patients.


Asunto(s)
Antiinflamatorios/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Calostro , Monocitos/efectos de los fármacos , Anciano , Apoptosis , Colecalciferol/farmacología , Neoplasias Colorrectales/inmunología , Citocinas/biosíntesis , Microbioma Gastrointestinal/fisiología , Humanos , Inmunoglobulinas Intravenosas/farmacología , Factor I del Crecimiento Similar a la Insulina/fisiología , Persona de Mediana Edad , Monocitos/inmunología , Estudios Prospectivos
18.
Life Sci ; 249: 117503, 2020 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-32142767

RESUMEN

AIMS: To investigate the role and mechanism of insulin-like growth factor 1(IGF-1)-mediated EMT on multiple myeloma (MM) growth and metastasis. MATERIALS AND METHODS: The expression data from GEO datasets were utilized to explore the expression levels of IGF-1 and epithelial-mesenchymal transition (EMT) markers in MM. Western blotting and flow cytometry analysis were performed to detect the protein levels of EMT markers as well as key components of the PI3K/Akt pathway. Cell proliferation ability was assessed using colony formation assay and EdU incorporation assays. Transwell migration and invasion assays were performed to assess cell metastasis properties. Vimentin was knocked down by using electro-transfection with small interfering RNA (siRNA) to detect the effect of IGF-1-mediated EMT on MM cell growth and metastasis. KEY FINDINGS: First of all, the analysis of GEO database revealed that IGF-1 was excessively expressed and closely correlated with the expression of the EMT markers in MM patients. Furthermore, we demonstrated that IGF-1 enhanced the acquisition of mesenchymal features in a time-dependent manner. Additionally, in vitro studies revealed that IGF-1-mediated mesenchymal phenotype promoted MM migration, invasion and colony formation. Finally, the mechanism study showed PI3K/Akt signaling pathway was involved in the IGF-1-induced EMT in MM cells. SIGNIFICANCE: IGF-1-induced mesenchymal phenotype contributed to MM progression via the PI3K/Akt pathway regulation.


Asunto(s)
Transición Epitelial-Mesenquimal/fisiología , Factor I del Crecimiento Similar a la Insulina/fisiología , Mieloma Múltiple/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Biomarcadores/metabolismo , Línea Celular Tumoral , Proliferación Celular/fisiología , Progresión de la Enfermedad , Regulación hacia Abajo , Humanos , Mieloma Múltiple/metabolismo , Metástasis de la Neoplasia , Transducción de Señal , Regulación hacia Arriba , Vimentina/metabolismo
19.
Artículo en Inglés | MEDLINE | ID: mdl-32194500

RESUMEN

The insulin-like growth factor (IGF) family plays key roles in growth and development. In the cornea, IGF family members have been implicated in proliferation, differentiation, and migration, critical events that maintain a smooth refracting surface that is essential for vision. The IGF family is composed of multiple ligands, receptors, and ligand binding proteins. Expression of IGF type 1 receptor (IGF-1R), IGF type 2 receptor (IGF-2R), and insulin receptor (INSR) in the cornea has been well characterized, including the presence of the IGF-1R and INSR hybrid (Hybrid-R) in the corneal epithelium. Recent data also indicates that each of these receptors display unique intracellular localization. Thus, in addition to canonical ligand binding at the plasma membrane and the initiation of downstream signaling cascades, IGF-1R, INSR, and Hybrid-R also function to regulate mitochondrial stability and nuclear gene expression. IGF-1 and IGF-2, two of three principal ligands, are polypeptide growth factors that function in all cellular layers of the cornea. Unlike IGF-1 and IGF-2, the hormone insulin plays a unique role in the cornea, different from many other tissues in the body. In the corneal epithelium, insulin is not required for glucose uptake, due to constitutive activation of the glucose transporter, GLUT1. However, insulin is needed for the regulation of metabolism, circadian rhythm, autophagy, proliferation, and migration after wounding. There is conflicting evidence regarding expression of the six IGF-binding proteins (IGFBPs), which function primarily to sequester IGF ligands. Within the cornea, IGFBP-2 and IGFBP-3 have identified roles in tissue homeostasis. While IGFBP-3 regulates growth control and intracellular receptor localization in the corneal epithelium, both IGFBP-2 and IGFBP-3 function in corneal fibroblast differentiation and myofibroblast proliferation, key events in stromal wound healing. IGFBP-2 has also been linked to cellular overgrowth in pterygium. There is a clear role for IGF family members in regulating tissue homeostasis in the cornea. This review summarizes what is known regarding the function of IGF and related proteins in corneal development, during wound healing, and in the pathophysiology of disease. Finally, we highlight key areas of research that are in need of future study.


Asunto(s)
Córnea/fisiología , Enfermedades de la Córnea/etiología , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/fisiología , Factor I del Crecimiento Similar a la Insulina/fisiología , Insulinas/fisiología , Cicatrización de Heridas/fisiología , Animales , Córnea/crecimiento & desarrollo , Córnea/patología , Enfermedades de la Córnea/patología , Lesiones de la Cornea/fisiopatología , Humanos , Transducción de Señal/fisiología , Cicatrización de Heridas/genética
20.
Front Neuroendocrinol ; 57: 100821, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32006533

RESUMEN

The pleiotropic peptide insulin-like growth factor 1 (IGF-I) regulates human body homeostasis and cell growth. IGF-I activates two major signaling pathways, namely phosphoinositide-3-kinase (PI3K)/protein kinase B (PKB/Akt) and Ras/extracellular signal-regulated kinase (ERK), which contribute to brain development, metabolism and function as well as to neuronal maintenance and survival. In this review, we discuss the general and tissue-specific effects of the IGF-I pathways. In addition, we present a comprehensive overview examining the role of IGF-I in neurodegenerative diseases, such as spinal and muscular atrophy, amyotrophic lateral sclerosis, and polyglutamine diseases. In each disease, we analyze the disturbances of the IGF-I pathway, the modification of the disease protein by IGF-I signaling, and the therapeutic strategies based on the use of IGF-I developed to date. Lastly, we highlight present and future considerations in the use of IGF-I for the treatment of these disorders.


Asunto(s)
Factor I del Crecimiento Similar a la Insulina/fisiología , Neuronas Motoras/metabolismo , Enfermedades Neurodegenerativas/fisiopatología , Péptidos , Transducción de Señal/fisiología , Esclerosis Amiotrófica Lateral/fisiopatología , Animales , Glutamina/genética , Humanos , Factor I del Crecimiento Similar a la Insulina/genética , Sistema de Señalización de MAP Quinasas/fisiología , Atrofia Muscular Espinal/fisiopatología , Enfermedades Neurodegenerativas/tratamiento farmacológico , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas ras/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA