Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.051
Filter
1.
Nat Commun ; 15(1): 4909, 2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38851766

ABSTRACT

Tobacco smoking (TS) is implicated in lung cancer (LC) progression through the development of metabolic syndrome. However, direct evidence linking metabolic syndrome to TS-mediated LC progression remains to be established. Our findings demonstrate that 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone and benzo[a]pyrene (NNK and BaP; NB), components of tobacco smoke, induce metabolic syndrome characteristics, particularly hyperglycemia, promoting lung cancer progression in male C57BL/6 J mice. NB enhances glucose uptake in tumor-associated macrophages by increasing the expression and surface localization of glucose transporter (GLUT) 1 and 3, thereby leading to transcriptional upregulation of insulin-like growth factor 2 (IGF2), which subsequently activates insulin receptor (IR) in LC cells in a paracrine manner, promoting its nuclear import. Nuclear IR binds to nucleophosmin (NPM1), resulting in IR/NPM1-mediated activation of the CD274 promoter and expression of programmed death ligand-1 (PD-L1). Restricting glycolysis, depleting macrophages, or blocking PD-L1 inhibits NB-mediated LC progression. Analysis of patient tissues and public databases reveals elevated levels of IGF2 and GLUT1 in tumor-associated macrophages, as well as tumoral PD-L1 and phosphorylated insulin-like growth factor 1 receptor/insulin receptor (pIGF-1R/IR) expression, suggesting potential poor prognostic biomarkers for LC patients. Our data indicate that paracrine IGF2/IR/NPM1/PD-L1 signaling, facilitated by NB-induced dysregulation of glucose levels and metabolic reprogramming of macrophages, contributes to TS-mediated LC progression.


Subject(s)
B7-H1 Antigen , Benzo(a)pyrene , Disease Progression , Hyperglycemia , Insulin-Like Growth Factor II , Lung Neoplasms , Mice, Inbred C57BL , Nuclear Proteins , Nucleophosmin , Receptor, Insulin , Animals , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/genetics , Male , Humans , Receptor, Insulin/metabolism , Receptor, Insulin/genetics , Mice , B7-H1 Antigen/metabolism , B7-H1 Antigen/genetics , Hyperglycemia/metabolism , Benzo(a)pyrene/toxicity , Insulin-Like Growth Factor II/metabolism , Insulin-Like Growth Factor II/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Nitrosamines/toxicity , Tumor-Associated Macrophages/metabolism , Cell Line, Tumor , Paracrine Communication , Gene Expression Regulation, Neoplastic , Smoking/adverse effects , Macrophages/metabolism
2.
Age Ageing ; 53(5)2024 May 01.
Article in English | MEDLINE | ID: mdl-38752921

ABSTRACT

OBJECTIVE: To investigate longitudinal associations between variations in the co-expression-based brain insulin receptor polygenic risk score and frailty, as well as change in frailty across follow-up. METHODS: This longitudinal study included 1605 participants from the Helsinki Birth Cohort Study. Biologically informed expression-based polygenic risk scores for the insulin receptor gene network, which measure genetic variation in the function of the insulin receptor, were calculated for the hippocampal (hePRS-IR) and the mesocorticolimbic (mePRS-IR) regions. Frailty was assessed in at baseline in 2001-2004, 2011-2013 and 2017-2018 by applying a deficit accumulation-based frailty index. Analyses were carried out by applying linear mixed models and logistical regression models adjusted for adult socioeconomic status, birthweight, smoking and their interactions with age. RESULTS: The FI levels of women were 1.19%-points (95% CI 0.12-2.26, P = 0.029) higher than in men. Both categorical and continuous hePRS-IR in women were associated with higher FI levels than in men at baseline (P < 0.05). In women with high hePRS-IR, the rate of change was steeper with increasing age compared to those with low or moderate hePRS-IR (P < 0.05). No associations were detected between mePRS-IR and frailty at baseline, nor between mePRS-IR and the increase in mean FI levels per year in either sex (P > 0.43). CONCLUSIONS: Higher variation in the function of the insulin receptor gene network in the hippocampus is associated with increasing frailty in women. This could potentially offer novel targets for future drug development aimed at frailty and ageing.


Subject(s)
Frailty , Receptor, Insulin , Humans , Male , Female , Frailty/genetics , Frailty/diagnosis , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Aged , Longitudinal Studies , Middle Aged , Gene Regulatory Networks , Finland/epidemiology , Frail Elderly/statistics & numerical data , Age Factors , Risk Factors , Aged, 80 and over , Aging/genetics , Sex Factors , Hippocampus/metabolism , Multifactorial Inheritance , Geriatric Assessment/methods , Brain/metabolism , Antigens, CD
3.
Genes (Basel) ; 15(5)2024 May 12.
Article in English | MEDLINE | ID: mdl-38790245

ABSTRACT

Insulin receptor signaling promotes cell differentiation, proliferation, and growth which are essential for oocyte maturation, embryo implantation, endometrial decidualization, and placentation. The dysregulation of insulin signaling in women with metabolic syndromes including diabetes exhibits poor pregnancy outcomes that are poorly understood. We utilized the Cre/LoxP system to target the tissue-specific conditional ablation of insulin receptor (Insr) and insulin-like growth factor-1 receptor (Igf1r) using an anti-Mullerian hormone receptor 2 (Amhr2) Cre-driver which is active in ovarian granulosa and uterine stromal cells. Our long-term goal is to examine insulin-dependent molecular mechanisms that underlie diabetic pregnancy complications, and our conditional knockout models allow for such investigation without confounding effects of ligand identity, source and cross-reactivity, or global metabolic status within dams. Puberty occurred with normal timing in all conditional knockout models. Estrous cycles progressed normally in Insrd/d females but were briefly stalled in diestrus in Igf1rd/d and double receptor (DKO) mice. The expression of vital ovulatory genes (Lhcgr, Pgr, Ptgs2) was not significantly different in 12 h post-hCG superovulated ovaries in knockout mice. Antral follicles exhibited an elevated apoptosis of granulosa cells in Igf1rd/d and DKO mice. However, the distribution of ovarian follicle subtypes and subsequent ovulations was normal in all insulin receptor mutants compared to littermate controls. While ovulation was normal, all knockout lines were subfertile suggesting that the loss of insulin receptor signaling in the uterine stroma elicits implantation and decidualization defects responsible for subfertility in Amhr2-Cre-derived insulin receptor mutants.


Subject(s)
Mice, Knockout , Ovary , Receptor, IGF Type 1 , Receptor, Insulin , Animals , Female , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Mice , Ovary/metabolism , Ovary/pathology , Infertility, Female/genetics , Infertility, Female/metabolism , Infertility, Female/pathology , Ovulation/genetics , Granulosa Cells/metabolism , Granulosa Cells/pathology , Pregnancy , Signal Transduction/genetics
4.
BMC Biol ; 22(1): 127, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38816743

ABSTRACT

BACKGROUND: Optimal size at birth dictates perinatal survival and long-term risk of developing common disorders such as obesity, type 2 diabetes and cardiovascular disease. The imprinted Grb10 gene encodes a signalling adaptor protein capable of inhibiting receptor tyrosine kinases, including the insulin receptor (Insr) and insulin-like growth factor type 1 receptor (Igf1r). Grb10 restricts fetal growth such that Grb10 knockout (KO) mice are at birth some 25-35% larger than wild type. Using a mouse genetic approach, we test the widely held assumption that Grb10 influences growth through interaction with Igf1r, which has a highly conserved growth promoting role. RESULTS: Should Grb10 interact with Igf1r to regulate growth Grb10:Igf1r double mutant mice should be indistinguishable from Igf1r KO single mutants, which are around half normal size at birth. Instead, Grb10:Igf1r double mutants were intermediate in size between Grb10 KO and Igf1r KO single mutants, indicating additive effects of the two signalling proteins having opposite actions in separate pathways. Some organs examined followed a similar pattern, though Grb10 KO neonates exhibited sparing of the brain and kidneys, whereas the influence of Igf1r extended to all organs. An interaction between Grb10 and Insr was similarly investigated. While there was no general evidence for a major interaction for fetal growth regulation, the liver was an exception. The liver in Grb10 KO mutants was disproportionately overgrown with evidence of excess lipid storage in hepatocytes, whereas Grb10:Insr double mutants were indistinguishable from Insr single mutants or wild types. CONCLUSIONS: Grb10 acts largely independently of Igf1r or Insr to control fetal growth and has a more variable influence on individual organs. Only the disproportionate overgrowth and excess lipid storage seen in the Grb10 KO neonatal liver can be explained through an interaction between Grb10 and the Insr. Our findings are important for understanding how positive and negative influences on fetal growth dictate size and tissue proportions at birth.


Subject(s)
Fetal Development , GRB10 Adaptor Protein , Mice, Knockout , Receptor, IGF Type 1 , Receptor, Insulin , Animals , GRB10 Adaptor Protein/genetics , GRB10 Adaptor Protein/metabolism , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Mice , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Fetal Development/genetics , Genomic Imprinting , Female , Male , Insulin-Like Peptides
5.
Zoolog Sci ; 41(2): 230-243, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38587918

ABSTRACT

The insulin/insulin-like growth factor-like signaling (IIS) pathway is highly conserved across metazoans and regulates numerous physiological functions, including development, metabolism, fecundity, and lifespan. The insulin receptor (InR), a crucial membrane receptor in the IIS pathway, is known to be ubiquitously expressed in various tissues, albeit at generally low levels, and its subcellular localization remains incompletely characterized. In this study, we employed CRISPR-mediated mutagenesis in the fruit fly Drosophila to create knock-in alleles of InR tagged with fluorescent proteins (InR::mCherry or InR::EYFP). By inserting the coding sequence of the fluorescent proteins mCherry or EYFP near the end of the coding sequence of the endogenous InR gene, we could trace the natural InR protein through their fluorescence. As an example, we investigated epithelial cells of the male accessory gland (AG), an internal reproductive organ, and identified two distinct patterns of InR::mCherry localization. In young AG, InR::mCherry accumulated on the basal plasma membrane between cells, whereas in mature AG, it exhibited intracellular localization as multiple puncta, indicating endocytic recycling of InR during cell growth. In the AG senescence accelerated by the mutation of Diuretic hormone 31 (Dh31), the presence of InR::mCherry puncta was more pronounced compared to the wild type. These findings raise expectations for the utility of the newly created InR::mCherry/EYFP alleles for studying the precise expression levels and subcellular localization of InR. Furthermore, this fluorescently tagged allele approach can be extended to investigate other membrane receptors with low abundance, facilitating the direct examination of their true expression and localization.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , Male , Animals , Drosophila melanogaster/physiology , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Alleles , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila
6.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167173, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38631410

ABSTRACT

Here, we show that insulin induces palmitoylation turnover of Caveolin-2 (Cav-2) in adipocytes. Acyl protein thioesterases-1 (APT1) catalyzes Cav-2 depalmitoylation, and zinc finger DHHC domain-containing protein palmitoyltransferase 21 (ZDHHC21) repalmitoylation of the depalmitoylated Cav-2 for the turnover, thereby controlling insulin receptor (IR)-Cav-2-insulin receptor substrate-1 (IRS-1)-Akt-driven signaling. Insulin-induced palmitoylation turnover of Cav-2 facilitated glucose uptake and fat storage through induction of lipogenic genes. Cav-2-, APT1-, and ZDHHC21-deficient adipocytes, however, showed increased induction of lipolytic genes and glycerol release. In addition, white adipose tissues from insulin sensitive and resistant obese patients exhibited augmented expression of LYPLA1 (APT1) and ZDHHC20 (ZDHHC20). Our study identifies the specific enzymes regulating Cav-2 palmitoylation turnover, and reveals a new mechanism by which insulin-mediated lipid metabolism is controlled in adipocytes.


Subject(s)
Adipocytes , Caveolin 2 , Insulin Receptor Substrate Proteins , Insulin , Lipid Metabolism , Lipoylation , Receptor, Insulin , Humans , Adipocytes/metabolism , Animals , Insulin Receptor Substrate Proteins/metabolism , Insulin Receptor Substrate Proteins/genetics , Mice , Caveolin 2/metabolism , Caveolin 2/genetics , Receptor, Insulin/metabolism , Receptor, Insulin/genetics , Insulin/metabolism , Obesity/metabolism , Obesity/genetics , Thiolester Hydrolases/metabolism , Thiolester Hydrolases/genetics , Acyltransferases/metabolism , Acyltransferases/genetics , Signal Transduction , Insulin Resistance , 3T3-L1 Cells , Male
7.
Int J Mol Sci ; 25(6)2024 Mar 08.
Article in English | MEDLINE | ID: mdl-38542117

ABSTRACT

Rabson-Mendenhall syndrome (RMS) is a rare autosomal recessive disorder characterized by severe insulin resistance, resulting in early-onset diabetes mellitus. We report the first case of RMS in a Paraguayan patient. The patient is a 6-year-old girl who presented with hypertrichosis, acanthosis nigricans, nephrocalcinosis, and elevated levels of glucose and insulin that served as diagnostic indicators for RMS. Genetic testing by next-generation sequencing (NGS) revealed two pathogenic variants in exons 2 and 19 of the INSR gene: c.332G>T (p.Gly111Val) and c.3485C>T (p.Ala1162Val), in combined heterozygosis. The novel INSR c. 332G>T variant leads to the substitution of glycine to valine at position 111 in the protein, and multiple in silico software programs predicted it as pathogenic. The c.3485C>T variant leads to the substitution of alanine to valine at position 1162 in the protein previously described for insulin resistance and RMS. The management of RMS is particularly challenging in children, and the use of metformin is often limited by its side effects. The patient was managed with nutritional measures due to the early age of onset. This report expands the knowledge of RMS to the Paraguayan population and adds a novel pathogenic variant to the existing literature.


Subject(s)
Donohue Syndrome , Insulin Resistance , Child , Female , Humans , Donohue Syndrome/diagnosis , Insulin Resistance/genetics , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Mutation , Valine/genetics , Antigens, CD/genetics
8.
Discov Med ; 36(181): 372-384, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38409842

ABSTRACT

BACKGROUND: Allergic asthma (AA) is a prevalent chronic airway inflammation disease. In this study, this study aims to investigate the biological functions and potential regulatory mechanisms of the insulin receptor (INSR) in the progression of AA. METHODS: BALB/c mice (n = 48) were randomly divided into the following groups: control group, AA group, AA+Lentivirus (Lv)-vector short hairpin RNA (shRNA) group, AA+Lv-vector group, AA+Lv-INSR shRNA group, and AA+Lv-INSR group. The pulmonary index was calculated. mRNA and protein expression levels of INSR, signal transducer and activator of transcription 3 (STAT3), Janus kinase 2 (JAK2), phosphorylated-STAT3 (p-STAT3), phosphorylated-JAK2 (p-JAK2), alpha-smooth muscle actin (α-SMA), febrile neutropenia (FN), mucin 5AC (MUC5AC), and mucin 5B (MUC5B) were examined using reverse-transcription quantitative PCR (RT-qPCR) and western blot assays. Positive expressions of INSR, retinoic acid-related orphan receptor gamma-t (RORγt), and forkhead box protein P3 (Foxp3) were quantified by immunohistochemistry. Fluorescence intensities of α-SMA and FN were detected by immunofluorescence. Pathological morphology was observed through hematoxylin-eosin (H&E) staining, Masson staining, and Periodic Acid-Schiff (PAS) staining. Contents of immunoglobulin E (IgE), interleukin-6 (IL-6), eotaxin, interleukin-4 (IL-4), interleukin-13 (IL-13), interferon-γ (IFN-γ), interleukin-17 (IL-17), and interleukin-10 (IL-10) were quantified using enzyme-linked immunosorbent assay (ELISA). The percentage of T helper 17 (Th17) and regulatory T (Treg) cells was determined through flow cytometry. RESULTS: Compared to the control group, expression levels of INSR, p-STAT3, p-JAK2, α-SMA, FN, MUC5AC, MUC5B, RORγt, and Foxp3, as well as IgE, IL-6, eotaxin, IL-4, IL-13, and IL-17 contents, pulmonary index, glycogen-positive area (%), and Th17 cell percentage significantly increased (p < 0.05). Additionally, pulmonary histopathological deterioration and collagen deposition were aggravated, while Treg cell percentage and IFN-γ and IL-10 contents remarkably decreased (p < 0.05). The overexpression of INSR further exacerbated the progression of allergic asthma, but the down-regulation of INSR reversed the trends of the above indicators. CONCLUSIONS: The down-regulation of INSR alleviates airway hyperviscosity, inflammatory infiltration, and airway remodeling, restoring Th17/Treg immune balance in AA mice by inactivating the STAT3 pathway.


Subject(s)
Asthma , Interleukin-10 , Pulmonary Disease, Chronic Obstructive , Mice , Animals , Interleukin-17/genetics , Interleukin-17/metabolism , Interleukin-4/genetics , Interleukin-4/metabolism , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology , Interleukin-13/genetics , Interleukin-13/metabolism , Interleukin-6/metabolism , Down-Regulation , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Asthma/metabolism , Asthma/pathology , Immunoglobulin E/genetics , Immunoglobulin E/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , RNA, Small Interfering
9.
Eur J Nutr ; 63(4): 1163-1175, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38358514

ABSTRACT

PURPOSE: The present study aimed at evaluating possible synergistic effects between two risk factors for cognitive decline and neurodegenerative disorders, i.e. iron overload and exposure to a hypercaloric/hyperlipidic diet, on cognition, insulin resistance, and hippocampal GLUT1, GLUT3, Insr mRNA expression, and AKT phosporylation. METHODS: Male Wistar rats were treated with iron (30 mg/kg carbonyl iron) or vehicle (5% sorbitol in water) from 12 to 14th post-natal days. Iron-treated rats received a standard laboratory diet or a high fat diet from weaning to adulthood (9 months of age). Recognition and emotional memory, peripheral blood glucose and insulin levels were evaluated. Glucose transporters (GLUT 1 and GLUT3) and insulin signaling were analyzed in the hippocampus of rats. RESULTS: Both iron overload and exposure to a high fat diet induced memory deficits. Remarkably, the association of iron with the high fat diet induced more severe cognitive deficits. Iron overload in the neonatal period induced higher insulin levels associated with significantly higher HOMA-IR, an index of insulin resistance. Long-term exposure to a high fat diet resulted in higher fasting glucose levels. Iron treatment induced changes in Insr and GLUT1 expression in the hippocampus. At the level of intracellular signaling, both iron treatment and the high fat diet decreased AKT phosphorylation. CONCLUSION: The combination of iron overload with exposure to a high fat diet only led to synergistic deleterious effect on emotional memory, while the effects induced by iron and by the high fat diet on AKT phosphorylation were comparable. These findings indicate that there is, at least to some extent, an additive effect of iron combined with the diet. Further studies investigating the mechanisms associated to deleterious effects on cognition and susceptibility for the development of age-associated neurodegenerative disorders are warranted.


Subject(s)
Animals, Newborn , Diet, High-Fat , Glucose Transporter Type 1 , Hippocampus , Insulin Resistance , Iron Overload , Memory Disorders , Rats, Wistar , Animals , Male , Diet, High-Fat/adverse effects , Iron Overload/complications , Iron Overload/metabolism , Memory Disorders/etiology , Hippocampus/metabolism , Hippocampus/drug effects , Rats , Glucose Transporter Type 1/metabolism , Glucose Transporter Type 1/genetics , Glucose Transporter Type 3/metabolism , Glucose Transporter Type 3/genetics , Receptor, Insulin/metabolism , Receptor, Insulin/genetics , Proto-Oncogene Proteins c-akt/metabolism , Blood Glucose/metabolism , Insulin/blood , Signal Transduction
10.
Int J Food Sci Nutr ; 75(4): 396-406, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38389245

ABSTRACT

Magnesium may have a significant impact on the development of cancer. However, the relationship between magnesium intake and the risk of colorectal cancer (CRC) is unclear. Therefore, we evaluated the association between magnesium intake and the risk of CRC, and we investigated how the insulin receptor (INSR) rs1799817 variant impacts this relationship. Data from 1,420 CRC patients and 2,840 controls from the Korean National Cancer Centre were analysed. A higher intake of magnesium was associated with a reduced risk of CRC in the total population (odds ratio (OR) = 0.65, 95% confidence interval (CI) = 0.52-0.81). We found that G + carriers of INSR rs1799817 with higher magnesium intake had a significantly lower risk of CRC (p for interaction = 0.003). Our findings indicated that high magnesium intake could be associated with a decreased risk of CRC, and this association could be modified by the INSR rs1799817 variant.


Subject(s)
Colorectal Neoplasms , Magnesium , Receptor, Insulin , Humans , Colorectal Neoplasms/genetics , Receptor, Insulin/genetics , Male , Case-Control Studies , Female , Middle Aged , Republic of Korea , Magnesium/administration & dosage , Aged , Risk Factors , Polymorphism, Single Nucleotide , Antigens, CD/genetics , Asian People/genetics , Genetic Predisposition to Disease , Adult , Odds Ratio
11.
J Alzheimers Dis ; 99(s2): S281-S297, 2024.
Article in English | MEDLINE | ID: mdl-38393902

ABSTRACT

Background: A strong body of evidence suggests that cerebrovascular pathologies augment the onset and progression of Alzheimer's disease (AD). One distinctive aspect of this cerebrovascular dysfunction is the degeneration of brain pericytes-often overlooked supporting cells of blood-brain barrier endothelium. Objective: The current study investigates the influence of pericytes on gene and protein expressions in the blood-brain barrier endothelium, which is expected to facilitate the identification of pathophysiological pathways that are triggered by pericyte loss and lead to blood-brain barrier dysfunction in AD. Methods: Bioinformatics analysis was conducted on the RNA-Seq expression counts matrix (GSE144474), which compared solo-cultured human blood-brain barrier endothelial cells against endothelial cells co-cultured with human brain pericytes in a non-contact model. We constructed a similar cell culture model to verify protein expression using western blots. Results: The insulin resistance and ferroptosis pathways were found to be enriched. Western blots of the insulin receptor and heme oxygenase expressions were consistent with those observed in RNA-Seq data. Additionally, we observed more than 5-fold upregulation of several genes associated with neuroprotection, including insulin-like growth factor 2 and brain-derived neurotrophic factor. Conclusions: Results suggest that pericyte influence on blood-brain barrier endothelial gene expression confers protection from insulin resistance, iron accumulation, oxidative stress, and amyloid deposition. Since these are conditions associated with AD pathophysiology, they imply mechanisms by which pericyte degeneration could contribute to disease progression.


Subject(s)
Alzheimer Disease , Blood-Brain Barrier , Endothelial Cells , Pericytes , Pericytes/metabolism , Pericytes/pathology , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Humans , Alzheimer Disease/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Endothelial Cells/metabolism , Coculture Techniques , Brain/metabolism , Brain/pathology , Cells, Cultured , Receptor, Insulin/metabolism , Receptor, Insulin/genetics , Gene Expression Regulation , Insulin Resistance/physiology
12.
Article in English | MEDLINE | ID: mdl-38267766

ABSTRACT

Alzheimer's disease (AD) is an irreversible and neurodegenerative disorder. Its etiology is not clear, but the involvement of genetic components plays a central role in the onset of the disease. In the present study, the expression of 10 genes (APP, PS1 and PS2, APOE, APBA2, LRP1, GRIN2B, INSR, GJB1, and IDE) involved in the main pathways related to AD were analyzed in auditory cortices and cerebellum from 29 AD patients and 29 healthy older adults. Raw analysis revealed tissue-specific changes in genes LRP1, INSR, and APP. A correlation analysis showed a significant effect also tissue-specific AD in APP, GRIN2B, INSR, and LRP1. Furthermore, the E4 allele of the APOE gene revealed a significant correlation with change expression tissue-specific in ABPA2, APP, GRIN2B, LRP1, and INSR genes. To assess the existence of a correction between changes in target gene expression and a probability of AD in each tissue (auditory cortices and cerebellum) an analysis of the effect of expressions was realized and showed that the reduction in the expression of the APP in auditory cortex and GRIN2B cerebellum had a significant effect in increasing the probability of AD, in the same logic, our result also suggesting that increased expression of the LRP1 and INSR genes had a significant effect on increasing the probability of AD. Our results showed tissue-specific gene expression alterations associated with AD and certainly opened new perspectives to characterize factors involved in gene regulation and to obtain possible biomarkers for AD.


Subject(s)
Alzheimer Disease , Antigens, CD , Low Density Lipoprotein Receptor-Related Protein-1 , Humans , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Male , Female , Aged , Low Density Lipoprotein Receptor-Related Protein-1/genetics , Cerebellum/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Auditory Cortex/metabolism , Amyloid beta-Protein Precursor/genetics , Aged, 80 and over , Apolipoproteins E/genetics , Gene Expression/genetics , Case-Control Studies
13.
Arch Endocrinol Metab ; 68: e210305, 2024 Jan 29.
Article in English | MEDLINE | ID: mdl-38289143

ABSTRACT

Mutations in the insulin receptor (INSR) gene may present with variable clinical phenotypes. We report herein a novel heterozygous INSR mutation in an adolescent girl with type A insulin resistance syndrome and her mother.The index case was a 12-year-old girl without obesity who presented with excessive hair growth, especially in the chest and back area, and hyperpigmentation on the back of the neck (acanthosis nigricans). Acanthosis nigricans was first observed at the age of 11 years. On physical examination, the patient had acanthosis nigricans and hypertrichosis with no acne. Systolic and diastolic blood pressure measurement was within the normal range for age and sex. Laboratory tests revealed fasting hyperglycemia, fasting and postprandial hyperinsulinemia, elevated HbA1c level, and biochemical hyperandrogenemia. Fasting plasma lipids were normal. A diagnosis of type A insulin resistance syndrome was considered, and INSR gene mutation analysis was performed. Next generation sequence analysis was performed with the use of primers containing exon/exon-intron junctions in the INSR gene, and a novel heterozygous c.3486_3503delGAGAAACTGCATGGTCGC/p.Arg1163_Ala1168del change was detected in exon 19 of the INSR gene. In segregation analysis, the same variant was detected in the patient's mother, who had a milder clinical phenotype.We reported a novel, heterozygous, p.Arg1163_Ala1168del mutation in exon 19 of the INSR gene in a patient with type A insulin resistance syndrome, expanding the mutation database. The same mutation was associated with variable phenotypical severity in two subjects within the same family.


Subject(s)
Acanthosis Nigricans , Diabetes Mellitus , Insulin Resistance , Child , Female , Humans , Acanthosis Nigricans/genetics , Antigens, CD , Diabetes Mellitus/genetics , Insulin Resistance/genetics , Mothers , Mutation/genetics , Receptor, Insulin/genetics
14.
Clin Endocrinol (Oxf) ; 100(3): 284-293, 2024 03.
Article in English | MEDLINE | ID: mdl-38172081

ABSTRACT

OBJECTIVE: Insulin receptor substract 1 (IRS1) protein is an important signal transduction adapter for extracellular signal transduction from insulin-like growth factor-1 receptor and its family members to IRS1 downstream proteins. IRS1 has been reported to be involved in tumourigenesis and metastasis in some of solid tumors. Investigating the role of IRS1 in thyroid cancer can help to screen high risk patients at the initial diagnosis. DESIGN, PATIENTS AND MEASUREMENTS: Immunohistochemical assay was used to detect the expression levels of IRS1 in 131 metastatic thyroid cancer tissues. Wound healing, cell invasion and colony formation assays were used to study the functions of IRS1 in vitro. RNA sequencing (RNA-seq) and Western blot analysis analyses were performed to examine the underlying regulation mechanisms of IRS1 in thyroid cancer cells. RESULTS: IRS1 was highly expressed in thyroid cancers and its expression was positively associated with distant metastasis and advanced clinical stages. In vitro studies demonstrated that IRS1 is an important mediator of migration, invasion and colony formation of thyroid cancer cells. RNA-seq showed that IRS1 promoted the metastasis of thyroid cancer by regulating epithelial-mesenchymal transition and phosphoinositide 3-kinase (PI3K)/AKT pathway. CONCLUSIONS: IRS1 overexpression contributes to the aggressiveness of thyroid cancer and is expected to be a stratified marker and a potential therapeutic target for thyroid cancer.


Subject(s)
Phosphatidylinositol 3-Kinase , Thyroid Neoplasms , Humans , Phosphatidylinositol 3-Kinase/genetics , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Cell Proliferation/genetics , Cell Line, Tumor , Cell Movement/genetics , Thyroid Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/metabolism
15.
Nutrition ; 120: 112333, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38271759

ABSTRACT

OBJECTIVE: The aim was to investigate the intergenerational inheritance induced by a high-fat diet on sensitivity to insulin and leptin in the hypothalamic control of satiety in second-generation offspring, which were fed a control diet. METHODS: Progenitor rats were fed a high-fat or a control diet for 59 d until weaning. The first-generation and second-generation offspring were fed the control diet until 90 d of age. Body mass and adiposity index of the progenitors fed the high-fat diet and the second-generation offspring from progenitors fed the high-fat diet were evaluated as were the gene expression of DNA methyltransferase 3a, angiotensin-converting enzyme type 2, angiotensin II type 2 receptor, insulin and leptin signaling pathway (insulin receptor, leptin receptor, insulin receptor substrate 2, protein kinase B, signal transducer and transcriptional activator 3, pro-opiomelanocortin, and neuropeptide Agouti-related protein), superoxide dismutase activity, and the concentration of carbonyl protein and satiety-regulating neuropeptides, pro-opiomelanocortin and neuropeptide Agouti-related protein, in the hypothalamus. RESULTS: The progenitor group fed a high-fat diet showed increased insulin resistance and reduced insulin-secreting beta-cell function and reduced food intake, without changes in caloric intake. The second-generation offspring from progenitors fed a high-fat diet, compared with second-generation offspring from progenitors fed a control diet group, had decreased insulin-secreting beta-cell function and increased food and caloric intake, insulin resistance, body mass, and adiposity index. Furthermore, second-generation offspring from progenitors fed a high-fat diet had increased DNA methyltransferase 3a, neuropeptide Agouti-related protein, angiotensin II type 1 receptor, and nicotinamide adenine dinucleotide phosphate oxidase p47phox gene expression, superoxide dismutase activity, and neuropeptide Agouti-related protein concentration in the hypothalamus. In addition, there were reduced in gene expression of the insulin receptor, leptin receptor, insulin receptor substrate 2, pro-opiomelanocortin, angiotensin II type 2 receptor, angiotensin-converting enzyme type 2, and angiotensin-(1-7) receptor and pro-opiomelanocortin concentration in the second-generation offspring from progenitors fed the high-fat diet. CONCLUSIONS: Overall, progenitors fed a high-fat diet induced changes in the hypothalamic control of satiety of the second-generation offspring from progenitors fed the high-fat diet through intergenerational inheritance. These changes led to hyperphagia, alterations in the hypothalamic pathways of insulin, and leptin and adiposity index increase, favoring the occurrence of different cardiometabolic disorders in the second-generation offspring from progenitors fed the high-fat diet fed only with the control diet.


Subject(s)
Insulin Resistance , Neuropeptides , Rats , Animals , Leptin/metabolism , Insulin/metabolism , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Diet, High-Fat/adverse effects , Agouti-Related Protein/metabolism , Insulin Receptor Substrate Proteins/metabolism , Receptor, Angiotensin, Type 2/metabolism , Pro-Opiomelanocortin/genetics , Pro-Opiomelanocortin/metabolism , Receptors, Leptin/genetics , DNA Methyltransferase 3A , Rats, Sprague-Dawley , Obesity/genetics , Obesity/metabolism , Hyperphagia/complications , Hypothalamus/metabolism , Neuropeptides/metabolism , Superoxide Dismutase/metabolism , Angiotensins/metabolism
16.
J Biol Chem ; 300(2): 105631, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38199575

ABSTRACT

Integrins are cell adhesion receptors that dimerize to mediate cell-cell interactions and regulate processes, including proliferation, inflammation, and tissue repair. The role of integrins in regulating insulin signaling is incompletely understood. We have previously shown that binding of the integrin ligand milk fat globule epidermal growth factor like 8 (MFGE8) to the αvß5 integrin promotes termination of insulin receptor signaling in mice. Upon ligation of MFGE8, integrin ß5 complexes with the insulin receptor beta (IRß) in skeletal muscle, resulting in dephosphorylation of IRß and reduction of insulin-stimulated glucose uptake. Here, we investigate the mechanism by which the interaction between ß5 and IRß impacts IRß phosphorylation status. We show in in vitro and in vivo in skeletal muscle in mice that antibody-mediated blockade of the ß5 integrin inhibits and recombinant MFGE8 promotes PTP1B binding to and dephosphorylation of IRß resulting in increased or reduced insulin-stimulated glucose uptake, respectively. The ß5-PTP1B complex is recruited by MFGE8 to IRß leading to termination of canonical insulin signaling. ß5 blockade enhances insulin-stimulated glucose uptake in wildtype but not Ptp1b KO mice indicating that PTP1B functions downstream of MFGE8 in modulating insulin receptor signaling. Furthermore, in a human cohort, we report serum MFGE8 levels correlate with indices of insulin resistance. These data provide mechanistic insights into the role of MFGE8 and ß5 in regulating insulin signaling.


Subject(s)
Insulin , Receptor, Insulin , Animals , Humans , Mice , Antigens, Surface/metabolism , Glucose/metabolism , Insulin/metabolism , Integrin beta Chains , Milk Proteins/metabolism , Receptor, Insulin/genetics , Mice, Inbred C57BL , Male , Cell Line
17.
Cancer Res ; 84(3): 351-352, 2024 02 01.
Article in English | MEDLINE | ID: mdl-38095504

ABSTRACT

Although there has been a long-standing connection between hyperinsulinemia and cancer development, there is a lack of understanding of the role of the insulin receptor on cells that can become cancerous. In a recent issue of Cell Metabolism, Zhang and colleagues, using a diet-induced obesity mouse model, identified a direct function of insulin receptors on pancreatic acinar cells expressing a KRASG12D mutation in promoting obesity-associated pancreatic cancer. Furthermore, insulin receptor signaling from hyperinsulinemia promoted the secretion of digestive enzymes that contributed to acinar to ductal metaplasia. These findings highlight an important connection between obesity, diabetes, and pancreatic tumor development and suggest potential strategies for obesity-associated cancer prevention targeting the insulin receptor signaling pathways.


Subject(s)
Carcinoma, Pancreatic Ductal , Hyperinsulinism , Pancreatic Neoplasms , Mice , Animals , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Carcinoma, Pancreatic Ductal/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Obesity/metabolism , Acinar Cells/metabolism , Hyperinsulinism/complications , Hyperinsulinism/metabolism
18.
EBioMedicine ; 99: 104906, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38061240

ABSTRACT

BACKGROUND: In spite of new treatments, the incidence of type 2 diabetes (T2D) and its morbidities continue to rise. The key feature of T2D is resistance of adipose tissue and other organs to insulin. Approaches to overcome insulin resistance are limited due to a poor understanding of the mechanisms and inaccessibility of drugs to relevant intracellular targets. We previously showed in mice and humans that CD248, a pre/adipocyte cell surface glycoprotein, acts as an adipose tissue sensor that mediates the transition from healthy to unhealthy adipose, thus promoting insulin resistance. METHODS: Molecular mechanisms by which CD248 regulates insulin signaling were explored using in vivo insulin clamp studies and biochemical analyses of cells/tissues from CD248 knockout (KO) and wild-type (WT) mice with diet-induced insulin resistance. Findings were validated with human adipose tissue specimens. FINDINGS: Genetic deletion of CD248 in mice, overcame diet-induced insulin resistance with improvements in glucose uptake and lipolysis in white adipose tissue depots, effects paralleled by increased adipose/adipocyte GLUT4, phosphorylated AKT and GSK3ß, and reduced ATGL. The insulin resistance of the WT mice could be attributed to direct interaction of the extracellular domains of CD248 and the insulin receptor (IR), with CD248 acting to block insulin binding to the IR. This resulted in dampened insulin-mediated autophosphorylation of the IR, with reduced downstream signaling/activation of intracellular events necessary for glucose and lipid homeostasis. INTERPRETATION: Our discovery of a cell-surface CD248-IR complex that is accessible to pharmacologic intervention, opens research avenues toward development of new agents to prevent/reverse insulin resistance. FUNDING: Funded by Canadian Institutes of Health Research (CIHR), Natural Sciences and Engineering Research Council of Canada (NSERC), Canada Foundations for Innovation (CFI), the Swedish Diabetes Foundation, Family Ernfors Foundation and Novo Nordisk Foundation.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin Resistance , Humans , Mice , Animals , Insulin/metabolism , Insulin Resistance/genetics , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Diabetes Mellitus, Type 2/metabolism , Mice, Knockout , Canada , Adipose Tissue/metabolism , Obesity/metabolism , Antigens, Neoplasm/metabolism , Antigens, CD/genetics , Antigens, CD/metabolism
19.
Pest Manag Sci ; 80(2): 648-660, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37756442

ABSTRACT

BACKGROUND: Insulin signalling pathways play crucial roles in regulating growth and development in insects, but their effects on the growth and development of Arachnids, such as spiders, have rarely been studied. As a valuable pest natural enemy in agricultural fields, the molecular mechanisms of insulin signalling pathway-mediated growth and development of the wolf spider, Pardosa pseudoannulata, are of particular interest. RESULTS: In this study, we identified and characterized six insulin signalling pathway genes - InR, InR2, IRS1, PI3K1, PI3K2, and PDK - in Pardosa pseudoannulata. Real-time quantitative polymerase chain reaction results were used to analyse the relative expression levels of the six genes in different developmental instars and tissues, and in response to starvation treatment. In addition, the function of the insulin receptor substrate (IRS1) gene was investigated using RNA interference technology, which found that IRS1 significantly influenced nutrient content, developmental duration, body weight, and gonad development. CONCLUSION: This study revealed the roles of six key insulin signalling pathway genes in Pardosa pseudoannulata, and in particular the importance of the IRS1 gene in regulating growth and development in the spider. The results lay the foundation for further research on the internal regulation mechanisms of growth and development in Araneae species, and also provide a reference for the artificial breeding of spiders. © 2023 Society of Chemical Industry.


Subject(s)
Animals, Poisonous , Insulins , Spiders , Animals , RNA Interference , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Receptor, Insulin/pharmacology , Spiders/genetics , Growth and Development , Insulins/genetics , Insulins/metabolism , Insulins/pharmacology
20.
Exp Cell Res ; 434(1): 113862, 2024 01 01.
Article in English | MEDLINE | ID: mdl-38036052

ABSTRACT

Breast cancer is the most common cancer in women. The upregulation of insulin-like growth factor (IGF) system observed in certain types of breast cancers was linked to growth, metastasis, and survival resulting in multiple strategies designed to target the type I IGF receptor (IGF-1R) in breast cancer. These attempts failed to prove beneficial and it has been suggested that insulin receptor (IR) could also play an important role in breast cancer biology. To better understand the IR's role in breast cancer cells, the receptor was deleted from MCF-7L cells using CRISPR technology, and fluorescence-assisted cell sorting was used to obtain clone 35 (CL35). It was found that CL35 activated signaling pathways upon insulin stimulation despite the absence of IR expression. We hypothesized that CL35 used a surrogate receptor for sustained growth and development. IGF-1R was able to activate insulin signaling and growth in CL35. Thus, insulin may play a central role in regulating breast cancer growth due to its ability to activate all the receptors of the IGF family. These findings argue that dual targeting of IR and IGF-IR may be required to inhibit breast cancer growth.


Subject(s)
Breast Neoplasms , Receptor, Insulin , Female , Humans , Breast Neoplasms/metabolism , Cell Line, Tumor , Insulin/pharmacology , Insulin-Like Growth Factor I/metabolism , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Receptors, Somatomedin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...