Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.161
Filter
1.
Horm Res Paediatr ; 95(6): 619-630, 2022.
Article in English | MEDLINE | ID: mdl-36446332

ABSTRACT

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.


Subject(s)
Insulin-Like Growth Factor II , Insulin-Like Growth Factor I , Laron Syndrome , Animals , Humans , Insulin-Like Growth Factor I/deficiency , Insulin-Like Growth Factor I/history , Insulin-Like Growth Factor I/physiology , Insulin-Like Growth Factor I/therapeutic use , Laron Syndrome/drug therapy , Laron Syndrome/genetics , Laron Syndrome/history , Laron Syndrome/physiopathology , Peptide Hormones , Protein Processing, Post-Translational , Signal Transduction , Somatomedins/deficiency , Somatomedins/history , Somatomedins/physiology , Insulin-Like Growth Factor II/deficiency , Insulin-Like Growth Factor II/history , Insulin-Like Growth Factor II/physiology , Insulin-Like Growth Factor II/therapeutic use
2.
Cells ; 11(20)2022 10 17.
Article in English | MEDLINE | ID: mdl-36291127

ABSTRACT

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.


Subject(s)
Carrier Proteins , Cellular Senescence , Insulin-Like Growth Factor I , Laron Syndrome , Thioredoxins , Animals , Mice , Carrier Proteins/metabolism , Cellular Senescence/drug effects , Cellular Senescence/physiology , Glucose/metabolism , Growth Hormone/metabolism , Insulin-Like Growth Factor I/pharmacology , Insulin-Like Growth Factor I/physiology , Laron Syndrome/metabolism , Mitochondrial Proteins/metabolism , Thioredoxins/metabolism , Humans , Fibroblasts/drug effects , 3T3-L1 Cells
3.
Front Endocrinol (Lausanne) ; 13: 852382, 2022.
Article in English | MEDLINE | ID: mdl-35370981

ABSTRACT

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.


Subject(s)
Gastrointestinal Microbiome , Insulin-Like Growth Factor I , Carcinogenesis , Gastrointestinal Microbiome/physiology , Humans , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor I/physiology , Male , Phosphatidylinositol 3-Kinases , Prostate
4.
Growth Horm IGF Res ; 62: 101440, 2022 02.
Article in English | MEDLINE | ID: mdl-34814007

ABSTRACT

Patients with growth hormone deficiency (GHD) have many clinical features in common with Cushing's syndrome (glucocorticoid excess) - notably visceral obesity, insulin resistance, muscle myopathy and increased vascular mortality. Within key metabolic tissues, 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1) converts cortisone to the active glucocorticoid, cortisol (11-dehydrocorticosterone and corticosterone in rodents respectively), and thus amplifies local glucocorticoid action. We hypothesize that 11ß-HSD1 expression is negatively regulated by growth hormone (GH), and that GHD patients have elevated 11ß-HSD1 within key metabolic tissues (leading to increased intracellular cortisol generation) which contributes to the clinical features of this disease. To identify the impact of GH excess/resistance on 11ß-HSD1 in vivo, we measured mRNA expression in key metabolic tissues of giant mice expressing the bovine GH (bGH) gene, dwarf mice with a disrupted GH receptor (GHRKO) gene and mice expressing a gene encoding a GH receptor antagonist (GHA). Additionally, we assessed urine steroid markers of 11ß-HSD1 activity in both GHRKO and bGH animals. 11ß-HSD1 expression was decreased in gastrocnemius muscle (0.43-fold, p < 0.05), subcutaneous adipose (0.53-fold, p < 0.05) and epididymal adipose tissue (0.40-fold, p < 0.05), but not liver, in bGH mice compared to WT controls. This was paralleled by an increased percentage of 11-DHC (inactive glucocorticoid) present in the urine of bGH mice compared to WT controls (2.5-fold, p < 0.01) - consistent with decreased systemic 11ß-HSD1 activity. By contrast, expression of 11ß-HSD1 was increased in the liver of GHRKO (2.7-fold, p < 0.05) and GHA mice (2.0-fold, p < 0.05) compared to WT controls, but not gastrocnemius muscle, subcutaneous adipose tissue or epididymal adipose tissue. In summary, we have demonstrated a negative relationship between GH action and 11ß-HSD1 expression which appears to be tissue specific. These data provide evidence that increased intracellular cortisol production within key tissues may contribute to metabolic disease in GHD patients.


Subject(s)
Human Growth Hormone , Insulin Resistance , 11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , 11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Animals , Cattle , Glucocorticoids , Growth Hormone/physiology , Humans , Hydrocortisone/metabolism , Insulin-Like Growth Factor I/physiology , Mice
5.
Int J Mol Sci ; 22(20)2021 Oct 15.
Article in English | MEDLINE | ID: mdl-34681810

ABSTRACT

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.


Subject(s)
Diabetes Mellitus, Type 2/complications , Obesity/complications , Urinary Bladder Neoplasms/etiology , Animals , Biomarkers, Tumor/genetics , Diabetes Mellitus, Type 2/epidemiology , Diabetes Mellitus, Type 2/genetics , Humans , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/physiology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/physiology , Obesity/epidemiology , Obesity/genetics , Risk Factors , Signal Transduction/genetics , Urinary Bladder Neoplasms/epidemiology , Urinary Bladder Neoplasms/genetics
6.
Int J Mol Sci ; 22(17)2021 Aug 31.
Article in English | MEDLINE | ID: mdl-34502376

ABSTRACT

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.


Subject(s)
Cachexia/metabolism , Insulin-Like Growth Factor Binding Protein 3/metabolism , Insulin-Like Growth Factor I/metabolism , Cachexia/physiopathology , Growth Hormone/metabolism , Human Growth Hormone/metabolism , Humans , Hypothalamus/metabolism , Inflammation/physiopathology , Insulin/metabolism , Insulin-Like Growth Factor Binding Protein 3/physiology , Insulin-Like Growth Factor I/physiology , Muscular Atrophy/metabolism , Muscular Atrophy/physiopathology
7.
Neural Plast ; 2021: 5511010, 2021.
Article in English | MEDLINE | ID: mdl-34306061

ABSTRACT

As a general sensory disorder, hearing loss was a major concern worldwide. Autophagy is a common cellular reaction to stress that degrades cytoplasmic waste through the lysosome pathway. Autophagy not only plays major roles in maintaining intracellular homeostasis but is also involved in the development and pathogenesis of many diseases. In the auditory system, several studies revealed the link between autophagy and hearing protection. In this review, we aimed to establish the correlation between autophagy and hair cells (HCs) from the aspects of ototoxic drugs, aging, and acoustic trauma and discussed whether autophagy could serve as a potential measure in the protection of HCs.


Subject(s)
Autophagy , Hearing Loss, Sensorineural/prevention & control , Aging/genetics , Aging/physiology , Animals , Autophagy/drug effects , Cisplatin/toxicity , Cochlea/blood supply , Cochlea/growth & development , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/pathology , Hearing Loss, Noise-Induced , Hearing Loss, Sensorineural/etiology , Hearing Loss, Sensorineural/genetics , Hearing Loss, Sensorineural/metabolism , Humans , Insulin-Like Growth Factor I/physiology , Ischemia/physiopathology , Mice , Mice, Knockout , MicroRNAs/genetics , Oxidative Stress , Resveratrol/therapeutic use , Sleep Deprivation/complications
8.
Commun Biol ; 4(1): 775, 2021 06 23.
Article in English | MEDLINE | ID: mdl-34163008

ABSTRACT

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.


Subject(s)
Cardiomyopathies/prevention & control , DNA-Cytosine Methylases/physiology , Diabetes Mellitus, Type 1/complications , Insulin-Like Growth Factor I/physiology , Oxidative Stress/drug effects , Suppressor of Cytokine Signaling 1 Protein/physiology , Suppressor of Cytokine Signaling 3 Protein/physiology , Vitamin B 12/pharmacology , Adaptor Proteins, Signal Transducing/physiology , Animals , Diabetes Mellitus, Type 1/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Signal Transduction/drug effects
9.
FASEB J ; 35(5): e21563, 2021 05.
Article in English | MEDLINE | ID: mdl-33818810

ABSTRACT

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.


Subject(s)
Biomarkers/metabolism , Estradiol/pharmacology , Fallopian Tubes/physiology , Gene Expression Regulation, Developmental/drug effects , Insulin-Like Growth Factor I/physiology , Oviducts/physiology , Single-Cell Analysis/methods , Animals , Estrogens/pharmacology , Fallopian Tubes/cytology , Fallopian Tubes/drug effects , Female , Gene Expression Profiling , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oviducts/cytology , Oviducts/drug effects , Receptors, Progesterone/physiology
10.
Otolaryngol Head Neck Surg ; 165(6): 757-758, 2021 12.
Article in English | MEDLINE | ID: mdl-33781112

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal, Humanized/adverse effects , Graves Ophthalmopathy/drug therapy , Hearing Loss/chemically induced , Insulin-Like Growth Factor I/physiology , Receptor, IGF Type 1/antagonists & inhibitors , Animals , Antibodies, Monoclonal, Humanized/therapeutic use , Humans , Mice , Otolaryngologists
11.
Psychoneuroendocrinology ; 127: 105191, 2021 05.
Article in English | MEDLINE | ID: mdl-33706042

ABSTRACT

BACKGROUND: Insulin-like growth factor (IGF) binding protein (IGFBP)-3 and cyclic Glycine-Proline (cGP) regulate circulating IGF-1 function that is associated with cognition. The association between IGF-1 function and Alzheimer's disease (AD) remains inconclusive. This study evaluated the changes of IGFBPs and cGP, and their effects on the bioavailability and function of IGF-1 in human brain of AD cases. METHODS: Using biological and mathematic analysis we measured the concentrations of total, bound and unbound forms of IGF-1, IGFBPs and cGP in the inferior-frontal gyrus and middle-frontal gyrus of human AD (n = 15) and control cases (n = 15). The association between the changes of total concentration of these peptides and total protein concentration in brain tissues were also analyzed. RESULTS: The unbound bioavailable IGF-1 was lower whereas the bound cGP and IGFBP-3 were higher in AD than the control cases. Total protein that was lower in AD than control cases, was negatively associated with cGP concentration of control cases and with IGFBP-3 concentration of AD cases. CONCLUSIONS: The results provide direct evidence for IGF-1 deficiency in AD brain due to lower bioavailable IGF-1. The increase of bound IGFBP-3 impaired autocrine regulation. The increase of bound cGP is an autocrine response to improve the bioavailability and function of IGF-1 in AD brain. AVAILABILITY OF DATA AND MATERIAL: All data generated or analysed during this study are included in this published article. Additional datasets analysed during the current study available from the corresponding author on reasonable request.


Subject(s)
Alzheimer Disease , Brain , Insulin-Like Growth Factor I , Alzheimer Disease/physiopathology , Brain/metabolism , Humans , Insulin-Like Growth Factor I/physiology
13.
Ann Endocrinol (Paris) ; 82(2): 107-111, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33727117

ABSTRACT

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.


Subject(s)
Acromegaly/veterinary , Cat Diseases , Dog Diseases , Acromegaly/diagnosis , Acromegaly/therapy , Animals , Cat Diseases/diagnosis , Cat Diseases/therapy , Cats , Dog Diseases/diagnosis , Dog Diseases/therapy , Dogs , Dopamine Agonists/therapeutic use , Growth Hormone/metabolism , Hypophysectomy , Insulin Resistance , Insulin-Like Growth Factor I/physiology , Radiotherapy , Somatostatin/analogs & derivatives
14.
J Pediatr Endocrinol Metab ; 34(3): 301-309, 2021 Mar 26.
Article in English | MEDLINE | ID: mdl-33618443

ABSTRACT

OBJECTIVES: Childhood obesity is a serious medical condition with alarmingly high rates worldwide. There is controversy regarding the relationship between insulin-like growth factor-1 (IGF-1) and pediatric obesity. We investigated the relationship between IGF-1, insulin resistance and metabolic profile with childhood pre-obesity/obesity. METHODS: The study involved 201 children aged 7-15 years, divided in three groups according to their nutritional status (International Obesity Task Force criteria): normal-weight (n=84), pre-obese (n=82), obese (n=35). Laboratory IGF-1, insulin, fasting blood glucose (FBG), lipid profile, alanine-aminotransferase (ALT), uric acid (UA), anthropometric and body composition parameters were analyzed. Body mass index and IGF-1 standard deviation score (SDS), waist-to-height ratio (WtHR) and Homeostatic Model Assessment for Insulin Resistance (HOMA-IR) score were calculated. RESULTS: Pre-obese/obese children had significantly higher IGF-1 SDS, FBG, insulin, HOMA-IR, UA, ALT, triglycerides, and lower high-density lipoprotein cholesterol (HDL-c); obese group had higher WtHR and low-density lipoprotein cholesterol (LDL-c) compared to controls (p<0.05). In obese group, IGF-1 SDS was positively correlated with fat free/muscle mass, total body water (p<0.05) and negatively correlated with LDL-c (p<0.05). In pre-obese/obese HOMA-IR and insulin were positively correlated with age, total body fat (TBF) (p<0.05) and negatively correlated with HDL-c (pre-obese) (p<0.05). Multivariate ordinal logistic regression analyses showed that IGF-1 SDS (OR=1.94; 95%CI: 1.21-3.11), TBF (OR=1.37; 95%CI: 1.21-1.54) were predictors of nutritional status (p<0.001). FBG (OR=42.39; 95%CI: 2.31-77.2) and UA (OR=1.03; 95%CI: 1.01-1.05) were predictors of IR (p<0.001). CONCLUSIONS: IGF-1 SDS and TBF were predictors of nutritional status. Further studies are required to clarify the role of IGF-1 in pathophysiology of obesity and its comorbidities.


Subject(s)
Insulin Resistance , Insulin-Like Growth Factor I/physiology , Pediatric Obesity/metabolism , Adolescent , Blood Glucose/analysis , Body Composition , Child , Female , Humans , Insulin-Like Growth Factor I/analysis , Lipids/blood , Logistic Models , Male
15.
Pediatr Res ; 90(3): 559-564, 2021 09.
Article in English | MEDLINE | ID: mdl-33096541

ABSTRACT

BACKGROUND: Apoptosis that occurs after hypoxia/reoxygenation (H/R) has an important role in the pathogenesis of necrotizing enterocolitis (NEC). Telomerase activity, showing the regeneration capacity, may also be important in the recovery process. Therefore, we aimed to investigate the effects of insulin-like growth factor-1 (IGF-1) and erythropoietin (EPO) on apoptosis and telomerase activity in an H/R model. METHODS: Young mice were divided into four groups each containing ten Balb/c mice. Group 1 (H/R) were exposed to H/R; group 2 and group 3 were pretreated with IGF-1 and EPO, respectively, for 7 days before H/R. Group 4 served as control. Intestinal injury was evaluated by histological scoring and assessment of apoptosis was performed by terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) test. Proapoptotic and antiapoptotic gene expressions and telomerase activity were analyzed by real-time PCR. RESULTS: IGF-1- and EPO-treated animals had decreased histological damage and apoptosis, confirmed by TUNEL test and caspase activity. Telomerase activity was increased in these animals in addition to increased expression of antiapoptotic genes. However, proapoptotic gene expressions were not statistically different. CONCLUSIONS: The protective effects of IGF-1 and EPO in H/R damage may be through increased expression of antiapoptotic genes and increased telomerase activity, especially for IGF-1. IMPACT: This is a comprehensive study measuring various variables, namely IGF-1, EPO, apoptosis, apoptotic and antiapoptotic genes, and telomerase activity in the NEC model. The intestinal protective effects of IGF-1 and EPO in H/R damage may occur through increased expression of antiapoptotic genes and increased telomerase activity. To the best of our knowledge, telomerase activity has not been investigated in the NEC model before. Regarding our results, novel strategies may be implemented for the early definitive diagnosis, robust preventive measures, and effective treatment modalities for NEC.


Subject(s)
Apoptosis/physiology , Enterocolitis, Necrotizing/prevention & control , Erythropoietin/physiology , Insulin-Like Growth Factor I/physiology , Telomerase/metabolism , Animals , Disease Models, Animal , Mice , Mice, Inbred BALB C
16.
J Endocrinol ; 248(1): 17-30, 2021 01.
Article in English | MEDLINE | ID: mdl-33112794

ABSTRACT

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.


Subject(s)
Cell Proliferation , Estradiol/physiology , Hydrogen Sulfide/metabolism , Insulin-Like Growth Factor I/physiology , Myocytes, Smooth Muscle/physiology , Animals , Aorta/metabolism , Estrogen Receptor alpha/metabolism , Female , Male , Mice, Knockout , Protein Phosphatase 2/metabolism , Receptor, IGF Type 1/metabolism
17.
J Endocrinol ; 248(1): 31-44, 2021 01.
Article in English | MEDLINE | ID: mdl-33112796

ABSTRACT

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.


Subject(s)
Fatty Liver/etiology , Growth Hormone/physiology , Hepatocytes/physiology , Insulin-Like Growth Factor I/physiology , Liver/metabolism , Animals , Female , Lipid Metabolism , Male , Mice , Receptors, Somatotropin/physiology , Sex Characteristics , Somatotrophs/metabolism
18.
Front Endocrinol (Lausanne) ; 11: 554994, 2020.
Article in English | MEDLINE | ID: mdl-33123086

ABSTRACT

The Caenorhabditis elegans genome possesses homologs of about two-thirds of all human disease genes. Based on its physiological aging characteristics and superiority, the use of C. elegans as a model system for studies on aging, age-related diseases, mechanisms of longevity, and drug screening has been widely acknowledged in recent decades. Lifespan increasing mutations in C. elegans were found to delay aging by impinging several signaling pathways and related epigenetic modifications, including the insulin/IGF-1 signaling (IIS), AMP-activated protein kinase (AMPK), and mechanistic target of rapamycin (mTOR) pathways. Interestingly, dietary restriction (DR) has been shown to increase the lifespan of numerous metazoans and protect them from multiple age-related pathologies. However, the underlying molecular mechanisms are unclear. In recent decades, C. elegans has been used as a unique model system for high-throughput drug screening. Here, we review C. elegans mutants exhibiting increased in lifespan and age-dependent changes under DR, as well as the utility of C. elegans for drug screening. Thus, we provide evidence for the use of this model organism in research on the prevention of aging.


Subject(s)
Aging/genetics , Caenorhabditis elegans/genetics , Mutation , AMP-Activated Protein Kinases/physiology , Animals , Caenorhabditis elegans/drug effects , Caloric Restriction , Drug Evaluation, Preclinical , Epigenesis, Genetic , Insulin-Like Growth Factor I/physiology , Models, Animal , Signal Transduction , TOR Serine-Threonine Kinases/physiology
19.
Mol Cell Endocrinol ; 518: 111003, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32919021

ABSTRACT

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.


Subject(s)
Human Growth Hormone/physiology , Insulin-Like Growth Factor I/physiology , Neoplasms/pathology , Adult , Cell Cycle/genetics , Child , Disease Progression , Humans , Neoplasms/genetics , Oncogenes/physiology , Signal Transduction/physiology
20.
Curr Opin Otolaryngol Head Neck Surg ; 28(5): 286-290, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32796270

ABSTRACT

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.


Subject(s)
Ear Diseases/therapy , Insulin-Like Growth Factor I/physiology , Ear Diseases/diagnosis , Ear Diseases/etiology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...