Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 7.324
1.
Sci Transl Med ; 16(745): eadh1763, 2024 May.
Article En | MEDLINE | ID: mdl-38691618

An abdominal aortic aneurysm (AAA) is a life-threatening cardiovascular disease. We identified plasma insulin-like growth factor 1 (IGF1) as an independent risk factor in patients with AAA by correlating plasma IGF1 with risk. Smooth muscle cell- or fibroblast-specific knockout of Igf1r, the gene encoding the IGF1 receptor (IGF1R), attenuated AAA formation in two mouse models of AAA induced by angiotensin II infusion or CaCl2 treatment. IGF1R was activated in aortic aneurysm samples from human patients and mice with AAA. Systemic administration of IGF1C, a peptide fragment of IGF1, 2 weeks after disease development inhibited AAA progression in mice. Decreased AAA formation was linked to competitive inhibition of IGF1 binding to its receptor by IGF1C and modulation of downstream alpha serine/threonine protein kinase (AKT)/mammalian target of rapamycin signaling. Localized application of an IGF1C-loaded hydrogel was developed to reduce the side effects observed after systemic administration of IGF1C or IGF1R antagonists in the CaCl2-induced AAA mouse model. The inhibitory effect of the IGF1C-loaded hydrogel administered at disease onset on AAA formation was further evaluated in a guinea pig-to-rat xenograft model and in a sheep-to-minipig xenograft model of AAA formation. The therapeutic efficacy of IGF1C for treating AAA was tested through extravascular delivery in the sheep-to-minipig model with AAA established for 2 weeks. Percutaneous injection of the IGF1C-loaded hydrogel around the AAA resulted in improved vessel flow dynamics in the minipig aorta. These findings suggest that extravascular administration of IGF1R antagonists may have translational potential for treating AAA.


Aortic Aneurysm, Abdominal , Disease Models, Animal , Insulin-Like Growth Factor I , Receptor, IGF Type 1 , Animals , Receptor, IGF Type 1/metabolism , Receptor, IGF Type 1/antagonists & inhibitors , Humans , Aortic Aneurysm, Abdominal/pathology , Aortic Aneurysm, Abdominal/drug therapy , Aortic Aneurysm, Abdominal/metabolism , Aortic Aneurysm, Abdominal/prevention & control , Insulin-Like Growth Factor I/metabolism , Male , Swine , Mice , Signal Transduction/drug effects , Mice, Inbred C57BL , Rats
2.
Cell Death Dis ; 15(5): 374, 2024 May 29.
Article En | MEDLINE | ID: mdl-38811543

High workload-induced cellular stress can cause pancreatic islet ß cell death and dysfunction, or ß cell failure, a hallmark of type 2 diabetes mellitus. Thus, activation of molecular chaperones and other stress-response genes prevents ß cell failure. To this end, we have shown that deletion of the glucose-regulated protein 94 (GRP94) in Pdx1+ pancreatic progenitor cells led to pancreas hypoplasia and reduced ß cell mass during pancreas development in mice. Here, we show that GRP94 was involved in ß cell adaption and compensation (or failure) in islets from leptin receptor-deficient (db/db) mice in an age-dependent manner. GRP94-deficient cells were more susceptible to cell death induced by various diabetogenic stress conditions. We also identified a new client of GRP94, insulin-like growth factor-1 receptor (IGF-1R), a critical factor for ß cell survival and function that may mediate the effect of GRP94 in the pathogenesis of diabetes. This study has identified essential functions of GRP94 in ß cell failure related to diabetes.


Insulin-Secreting Cells , Receptor, IGF Type 1 , Animals , Mice , Cell Death , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/genetics , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Receptor, IGF Type 1/metabolism , Receptor, IGF Type 1/genetics , Receptors, Leptin/metabolism , Receptors, Leptin/genetics
3.
BMC Biol ; 22(1): 127, 2024 May 30.
Article En | MEDLINE | ID: mdl-38816743

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.


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
4.
Front Immunol ; 15: 1392956, 2024.
Article En | MEDLINE | ID: mdl-38817600

Thyroid eye disease (TED) is a disfiguring autoimmune disease characterized by changes in the orbital tissues and is caused by abnormal thyroid function or thyroid-related antibodies. It is the ocular manifestation of Graves' disease. The expression of thyroid-stimulating hormone receptor (TSHR) and the insulin-like growth factor-1 receptor (IGF-1 R) on the cell membrane of orbital fibroblasts (OFs) is responsible for TED pathology. Excessive inflammation is caused when these receptors in the orbit are stimulated by autoantibodies. CD34+ fibrocytes, found in the peripheral blood and orbital tissues of patients with TED, express immune checkpoints (ICs) like MHC II, B7, and PD-L1, indicating their potential role in presenting antigens and regulating the immune response in TED pathogenesis. Immune checkpoint inhibitors (ICIs) have significantly transformed cancer treatment. However, it can also lead to the occurrence of TED in some instances, suggesting the abnormality of ICs in TED. This review will examine the overall pathogenic mechanism linked to the immune cells of TED and then discuss the latest research findings on the immunomodulatory role of ICs in the development and pathogenesis of TED. This will offer fresh perspectives on the study of pathogenesis and the identification of potential therapeutic targets.


Graves Ophthalmopathy , Immune Checkpoint Inhibitors , Humans , Graves Ophthalmopathy/immunology , Graves Ophthalmopathy/etiology , Graves Ophthalmopathy/pathology , Animals , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Proteins/metabolism , Immune Checkpoint Proteins/genetics , Autoantibodies/immunology , Receptor, IGF Type 1/immunology , Receptor, IGF Type 1/metabolism , Receptors, Thyrotropin/immunology , Receptors, Thyrotropin/metabolism
5.
Int Immunopharmacol ; 134: 112245, 2024 Jun 15.
Article En | MEDLINE | ID: mdl-38749334

Gastric cancer (GC) has posed a great threat to the lives of people around the world. To date, safer and more cost-effective therapy for GC is lacking. Traditional Chinese medicine (TCM) may provide some new options for this. Guiqi Baizhu Formula (GQBZF), a classic TCM formula, has been extensively used to treat GC, while its bioactive components and therapeutic mechanisms remain unclear. In this study, we evaluated the underlying mechanisms of GQBZF in treating GC by integrative approach of chemical bioinformatics. GQBZF lyophilized powder (0.0625 mg/mL, 0.125 mg/mL) significantly attenuated the expression of p-IGF1R, PI3K, p-PDK1, p-VEGFR2 to inhibit the proliferation, migration and induce apoptosis of gastric cancer cells, which was consistent with the network pharmacology. Additionally, atractylenolide Ⅰ, quercetin, glycyrol, physcione and aloe-emodin, emodin, kaempferol, licoflavone A were found to be the key compounds of GQBZF regulating IGF1R and VEGFR2, respectively. And among which, glycyrol and emodin were determined as key active compounds against GC by farther vitro experiments and LC/MS. Meanwhile, we also found that glycyrol inhibited MKN-45 cells proliferation and enhanced apoptosis, which might be related to the inhibition of IGF1R/PI3K/PDK1, and emodin could significantly attenuate the MKN-45 cells migration, which might be related to the inhibition of VEGFR2-related signaling pathway. These results were verified again by molecular dynamics simulation and binding interaction pattern. In summary, this study suggested that GQBZF and its key active components (glycyrol and emodin) can suppress IGF1R/PI3K/PDK1 and VEGFR2-related signaling pathway, thereby inhibiting tumor cell proliferation and migration and inducing apoptosis. These findings provided an important strategy for developing new agents and facilitated clinical use of GQBZF against GC.


Apoptosis , Cell Movement , Cell Proliferation , Computational Biology , Drugs, Chinese Herbal , Receptor, IGF Type 1 , Stomach Neoplasms , Vascular Endothelial Growth Factor Receptor-2 , Stomach Neoplasms/drug therapy , Stomach Neoplasms/metabolism , Humans , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Apoptosis/drug effects , Cell Movement/drug effects , Receptor, IGF Type 1/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Computational Biology/methods , Signal Transduction/drug effects , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Somatomedin/metabolism , Network Pharmacology , Antineoplastic Agents, Phytogenic/pharmacology
6.
Genes (Basel) ; 15(5)2024 May 12.
Article En | MEDLINE | ID: mdl-38790245

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.


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
7.
Int J Mol Sci ; 25(10)2024 May 14.
Article En | MEDLINE | ID: mdl-38791406

The allosteric inhibition of insulin-like growth factor receptor 1 kinase (IGF1RK) is a potential strategy to overcome selectivity barriers for targeting receptor tyrosine kinases. We constructed structural models of a series of 12 indole-butyl-amine derivatives that have been reported as allosteric inhibitors of IGF1RK. We further studied the dynamics and interactions of each inhibitor in the allosteric pocket via all-atom explicit-solvent molecular dynamics (MD) simulations. We discovered that a bulky carbonyl substitution at the R1 indole ring is structurally unfavorable for inhibitor binding in the IGF1RK allosteric pocket. Moreover, we found that the most potent derivative (termed C11) acquires a distinct conformation: forming an allosteric pocket channel with better shape complementarity and interactions with the receptor. In addition to a hydrogen-bonding interaction with V1063, the cyano derivative C11 forms a stable hydrogen bond with M1156, which is responsible for its unique binding conformation in the allosteric pocket. Our findings show that the positioning of chemical substituents with different pharmacophore features at the R1 indole ring influences molecular interactions and binding conformations of indole-butyl-amine derivatives and, hence, dramatically affects their potencies. Our results provide a structural framework for the design of allosteric inhibitors with improved affinities and specificities against IGF1RK.


Molecular Dynamics Simulation , Protein Kinase Inhibitors , Receptor, IGF Type 1 , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Allosteric Regulation , Receptor, IGF Type 1/antagonists & inhibitors , Receptor, IGF Type 1/chemistry , Receptor, IGF Type 1/metabolism , Humans , Hydrogen Bonding , Allosteric Site , Indoles/chemistry , Indoles/pharmacology , Protein Binding , Structure-Activity Relationship , Models, Molecular
8.
BMC Immunol ; 25(1): 31, 2024 May 11.
Article En | MEDLINE | ID: mdl-38734625

BACKGROUND: Thyroid eye disease (TED) is an inflammatory process involving lymphocyte-mediated immune response and orbital tissue damage. The anti-insulin-like growth factor-1 receptor (IGF-1R) antibodies produced by B lymphocytes are involved in the activation of orbital fibroblasts and the inflammatory process of orbital tissue damage in TED. The purpose of this study was to explore the role of IGF-1R in the mechanistic connection between orbital fibroblasts and B lymphocytes in TED. METHODS: Orbital fibroblasts sampled from orbital connective tissues and peripheral B lymphocytes isolated from peripheral blood, which were obtained from 15 patients with TED and 15 control patients, were co-cultured at a ratio of 1:20. The level of IGF-1R expression in orbital fibroblasts was evaluated by flow cytometry and confocal microscopy. Transient B lymphocyte depletion was induced with anti-CD20 monoclonal antibody rituximab, while the IGF-1R pathway was blocked by the IGF-1R binding protein. The expression levels of interleukin-6 (IL-6) and regulated upon activation, normal T cell expressed and secreted (RANTES) in the co-culture model were quantified via ELISA. RESULTS: IGF-1R expression was significantly elevated in TED orbital fibroblasts compared to that of controls. A 24-h co-culture of orbital fibroblasts with peripheral B lymphocytes induced elevated expression levels of IL-6 and RANTES in each group (TED patients and controls), with the highest levels occurring in TED patients (T + T group). Rituximab and IGF-1R binding protein significantly inhibited increased levels of IL-6 and RANTES in the co-culture model of TED patients. CONCLUSIONS: IGF-1R may mediate interaction between orbital fibroblasts and peripheral B lymphocytes; thus, blocking IGF-1R may reduce the local inflammatory response in TED. Rituximab-mediated B lymphocyte depletion played a role in inhibiting inflammatory responses in this in vitro co-culture model, providing a theoretical basis for the clinical application of anti-CD20 monoclonal antibodies in TED.


B-Lymphocytes , Coculture Techniques , Fibroblasts , Graves Ophthalmopathy , Receptor, IGF Type 1 , Humans , Graves Ophthalmopathy/metabolism , Graves Ophthalmopathy/immunology , Fibroblasts/metabolism , Receptor, IGF Type 1/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Female , Male , Middle Aged , Adult , Rituximab/pharmacology , Rituximab/therapeutic use , Orbit/metabolism , Orbit/immunology , Lymphocyte Depletion , Interleukin-6/metabolism , Cells, Cultured , Chemokine CCL5/metabolism , Cell Communication , Aged
9.
Mol Cancer ; 23(1): 91, 2024 May 07.
Article En | MEDLINE | ID: mdl-38715012

BACKGROUND: Recent evidence has demonstrated that abnormal expression and regulation of circular RNA (circRNAs) are involved in the occurrence and development of a variety of tumors. The aim of this study was to investigate the effects of circ_PPAPDC1A in Osimertinib resistance in NSCLC. METHODS: Human circRNAs microarray analysis was conducted to identify differentially expressed (DE) circRNAs in Osimertinib-acquired resistance tissues of NSCLC. The effect of circ_PPAPDC1A on cell proliferation, invasion, migration, and apoptosis was assessed in both in vitro and in vivo. Dual-luciferase reporter assay, RT-qPCR, Western-blot, and rescue assay were employed to confirm the interaction between circ_PPAPDC1A/miR-30a-3p/IGF1R axis. RESULTS: The results revealed that circ_PPAPDC1A was significantly upregulated in Osimertinib acquired resistance tissues of NSCLC. circ_PPAPDC1A reduced the sensitivity of PC9 and HCC827 cells to Osimertinib and promoted cell proliferation, invasion, migration, while inhibiting apoptosis in Osimertinib-resistant PC9/OR and HCC829/OR cells, both in vitro and in vivo. Silencing circ_PPAPDC1A partially reversed Osimertinib resistance. Additionally, circ_PPAPDC1A acted as a competing endogenous RNA (ceRNA) by targeting miR-30a-3p, and Insulin-like Growth Factor 1 Receptor (IGF1R) was identified as a functional gene for miR-30a-3p in NSCLC. Furthermore, the results confirmed that circ_PPAPDC1A/miR-30a-3p/IGF1R axis plays a role in activating the PI3K/AKT/mTOR signaling pathway in NSCLC with Osimertinib resistance. CONCLUSIONS: Therefore, for the first time we identified that circ_PPAPDC1A was significantly upregulated and exerts an oncogenic role in NSCLC with Osimertinib resistance by sponging miR-30a-3p to active IGF1R/PI3K/AKT/mTOR pathway. circ_PPAPDC1A may serve as a novel diagnostic biomarker and therapeutic target for NSCLC patients with Osimertinib resistance.


Acrylamides , Aniline Compounds , Carcinoma, Non-Small-Cell Lung , Cell Proliferation , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Lung Neoplasms , MicroRNAs , RNA, Circular , Receptor, IGF Type 1 , Signal Transduction , Humans , MicroRNAs/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/metabolism , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Drug Resistance, Neoplasm/genetics , Acrylamides/pharmacology , RNA, Circular/genetics , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Aniline Compounds/pharmacology , Cell Line, Tumor , Animals , Mice , Apoptosis , Cell Movement/genetics , Xenograft Model Antitumor Assays , Male , Female , Indoles , Pyrimidines
10.
Mol Pharm ; 21(5): 2176-2186, 2024 May 06.
Article En | MEDLINE | ID: mdl-38625027

The blood-brain barrier (BBB) is instrumental in clearing toxic metabolites from the brain, such as amyloid-ß (Aß) peptides, and in delivering essential nutrients to the brain, like insulin. In Alzheimer's disease (AD) brain, increased Aß levels are paralleled by decreased insulin levels, which are accompanied by insulin signaling deficits at the BBB. Thus, we investigated the impact of insulin-like growth factor and insulin receptor (IGF1R and IR) signaling on Aß and insulin trafficking at the BBB. Following intravenous infusion of an IGF1R/IR kinase inhibitor (AG1024) in wild-type mice, the BBB trafficking of 125I radiolabeled Aß peptides and insulin was assessed by dynamic SPECT/CT imaging. The brain efflux of [125I]iodo-Aß42 decreased upon AG1024 treatment. Additionally, the brain influx of [125I]iodoinsulin, [125I]iodo-Aß42, [125I]iodo-Aß40, and [125I]iodo-BSA (BBB integrity marker) was decreased, increased, unchanged, and unchanged, respectively, upon AG1024 treatment. Subsequent mechanistic studies were performed using an in vitro BBB cell model. The cell uptake of [125I]iodoinsulin, [125I]iodo-Aß42, and [125I]iodo-Aß40 was decreased, increased, and unchanged, respectively, upon AG1024 treatment. Further, AG1024 reduced the phosphorylation of insulin signaling kinases (Akt and Erk) and the membrane expression of Aß and insulin trafficking receptors (LRP-1 and IR-ß). These findings reveal that insulin signaling differentially regulates the BBB trafficking of Aß peptides and insulin. Moreover, deficits in IGF1R and IR signaling, as observed in the brains of type II diabetes and AD patients, are expected to increase Aß accumulation while decreasing insulin delivery to the brain, which has been linked to the progression of cognitive decline in AD.


Amyloid beta-Peptides , Blood-Brain Barrier , Insulin , Signal Transduction , Animals , Male , Mice , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain/metabolism , Insulin/metabolism , Iodine Radioisotopes , Mice, Inbred C57BL , Peptide Fragments/metabolism , Receptor, IGF Type 1/metabolism , Receptor, Insulin/metabolism , Single Photon Emission Computed Tomography Computed Tomography/methods , Tyrphostins/pharmacology
11.
Gene ; 918: 148503, 2024 Aug 05.
Article En | MEDLINE | ID: mdl-38670398

The study is based on the complexity of Insulin like growth factor receptor (IGF1R) signaling and its regulation by noncoding RNAs (ncRNAs). IGF1R signaling is an important cascade in Alzheimer's disease (AD); however, its regulation and roles are poorly understood. Due to the presence of ß-arrestin and GPCR Receptor Kinase binding sites, this protein has been termed a 'functional hybrid', as it can take part in both kinase and GPCR signaling pathways, further adding to its complexity. The objective of this study is to understand the underlying ncRNA regulation controlling IGF1R and GPCRs in AD to find commonalities in the network. We found through data mining that 45 GPCRs were reportedly deregulated in AD and built clusters based on GO/KEGG pathways to show shared functionality with IGF1R. Eight miRs were further discovered that could coregulate IGF1R and GPCRs. We validated their expression in an AD cell model and probed for common lncRNAs downstream that could regulate these miRs. Seven such candidates were identified and further validated. A combined network comprising IGF1R with nine GPCRs, eight miRs, and seven lncRNAs was created to visualize the interconnectivity within pathways. Betweenness centrality analysis showed a cluster of NEAT1, hsa-miR-15a-5p, hsa-miR-16-5p, and IGF1R to be crucial form a competitive endogenous RNA-based (ceRNA) tetrad that could relay information within the network, which was further validated by cell-based studies. NEAT1 emerged as a master regulator that could alter the levels of IGF1R and associated GPCRs. This combined bioinformatics and experimental study for the first time explored the regulation of IGF1R through ncRNAs from the perspective of neurodegeneration.


Alzheimer Disease , MicroRNAs , RNA, Long Noncoding , Receptor, IGF Type 1 , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Humans , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Signal Transduction/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Gene Expression Regulation , Receptors, Somatomedin/genetics , Receptors, Somatomedin/metabolism , Gene Regulatory Networks
12.
Neurotherapeutics ; 21(3): e00342, 2024 Apr.
Article En | MEDLINE | ID: mdl-38493057

Novel therapeutics for the treatment of ischemic stroke remains to be the unmet clinical needs. Previous studies have indicated that salvianolic acid A (SAA) is a promising candidate for the treatment of the brain diseases. However, SAA has poor absolute bioavailability and does not efficiently cross the intact blood-brain barrier (BBB), which limit its efficacy. To this end we developed a brain-targeted liposomes for transporting SAA via the BBB by incorporating the liposomes to a transport receptor, insulin-like growth factor-1 receptor (IGF1R). The liposomes were prepared by ammonium sulfate gradients loading method. The prepared SAA-loaded liposomes (Lipo/SAA) were modified with IGF1R monoclonal antibody to generate IGF1R antibody-conjugated Lipo/SAA (IGF1R-targeted Lipo/SAA). The penetration of IGF1R-targeted Lipo/SAA into the brain was confirmed by labeling with Texas Red, and their efficacy were evaluate using middle cerebral artery occlusion (MCAO) model. The results showed that IGF1R-targeted Lipo/SAA are capable of transporting SAA across the BBB into the brain, accumulation in brain tissue, and sustained releasing SAA for several hours. Administration o IGF1R-targeted Lipo/SAA notably reduced infarct size and neuronal damage, improved neurological function and inhibited cerebral inflammation, which had much higher efficiency than no-targeted SAA.


Ischemic Stroke , Liposomes , Animals , Ischemic Stroke/drug therapy , Male , Caffeic Acids/administration & dosage , Caffeic Acids/chemistry , Caffeic Acids/pharmacology , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Receptor, IGF Type 1/metabolism , Mice , Lactates/administration & dosage , Lactates/chemistry , Infarction, Middle Cerebral Artery/drug therapy , Drug Delivery Systems/methods , Rats, Sprague-Dawley , Rats , Brain/metabolism , Brain/drug effects
13.
Talanta ; 274: 125914, 2024 Jul 01.
Article En | MEDLINE | ID: mdl-38537356

Progress in medical sciences aims for tailored therapy of civilization diseases like diabetes. Preclinical screening of new medicines superior to insulin should include the verification of their affinity to the membrane receptors naturally stimulated by this hormone: insulin receptor isoforms A and B and insulin-like growth factor receptor. Considering that the affinity constants obtained using different experimental conditions are incomparable, it is essential to develop a robust and reliable method to analyze these interactions. The versatile SPR platform developed in this study enables the evaluation of the bioactivity of hypoglycaemic molecules. Thanks to the comprehensive characterization of miscellaneous aspects of the analytical platform, including the design of the SPR biosensor receptor layer, ensuring interaction specificity, as well as the quality control of the standards used (human insulin, HI; long-acting insulin analog: glargine, Gla), the feasibility of the method of equilibrium and kinetic constants determination for insulin-like targets was confirmed. SPR assays constructed in the direct format using IR-A, IR-B, and IGF1-R receptor proteins show high sensitivities and low detection limits towards insulin and glargine detection in the range of 18.3-53.3 nM with no signs of mass transport limitations. The improved analytical performance and stability of SPR biosensors favor the acquisition of good-quality kinetic data, while preservation of receptors activity after binding to long-chain carboxymethyldextran, combined with spontaneous regeneration, results in stability and long shelf life of the biosensor, which makes it useful for label-free insulin analogs biosensing and thus extensive screening in diabetic drugs discovery.


High-Throughput Screening Assays , Hypoglycemic Agents , Receptor, Insulin , Surface Plasmon Resonance , Humans , Hypoglycemic Agents/chemistry , Surface Plasmon Resonance/methods , Receptor, Insulin/metabolism , High-Throughput Screening Assays/methods , Insulin Glargine/chemistry , Biosensing Techniques/methods , Insulin/metabolism , Insulin/analysis , Receptor, IGF Type 1/metabolism
14.
Sci Adv ; 10(9): eadk8123, 2024 Mar.
Article En | MEDLINE | ID: mdl-38427732

Besides recent advances in neonatal care, preterm newborns still develop sex-biased behavioral alterations. Preterms fail to receive placental insulin-like growth factor-1 (IGF-1), a major fetal growth hormone in utero, and low IGF-1 serum levels correlate with preterm poor neurodevelopmental outcomes. Here, we mimicked IGF-1 deficiency of preterm newborns in mice by perinatal administration of an IGF-1 receptor antagonist. This resulted in sex-biased brain microstructural, functional, and behavioral alterations, resembling those of ex-preterm children, which we characterized performing parallel mouse/human behavioral tests. Pharmacological enhancement of GABAergic tonic inhibition by the U.S. Food and Drug Administration-approved drug ganaxolone rescued functional/behavioral alterations in mice. Establishing an unprecedented mouse model of prematurity, our work dissects the mechanisms at the core of abnormal behaviors and identifies a readily translatable therapeutic strategy for preterm brain disorders.


Brain Diseases , Insulin-Like Growth Factor I , United States , Child , Humans , Infant, Newborn , Pregnancy , Female , Animals , Mice , Receptor, IGF Type 1 , Placenta , Infant, Premature , Brain Diseases/drug therapy
15.
Front Immunol ; 15: 1356321, 2024.
Article En | MEDLINE | ID: mdl-38420122

Cancer immunotherapy has made impressive advances in improving the outcome of patients affected by malignant diseases. Nonetheless, some limitations still need to be tackled to more efficiently and safely treat patients, in particular for those affected by solid tumors. One of the limitations is related to the immunosuppressive tumor microenvironment (TME), which impairs anti-tumor immunity. Efforts to identify targets able to turn the TME into a milieu more auspicious to current immuno-oncotherapy is a real challenge due to the high redundancy of the mechanisms involved. However, the insulin-like growth factor 1 receptor (IGF1R), an attractive drug target for cancer therapy, is emerging as an important immunomodulator and regulator of key immune cell functions. Here, after briefly summarizing the IGF1R signaling pathway in cancer, we review its role in regulating immune cells function and activity, and discuss IGF1R as a promising target to improve anti-cancer immunotherapy.


Neoplasms , Tumor Microenvironment , Humans , Immunotherapy , Neoplasms/therapy , Drug Delivery Systems , Molecular Targeted Therapy , Receptor, IGF Type 1
16.
Sci Rep ; 14(1): 4331, 2024 02 21.
Article En | MEDLINE | ID: mdl-38383709

Insulin and insulin-like growth factor 1 (IGF-1) are metabolic hormones with known effects on CD4+ T cells through insulin receptor (IR) and IGF-1 receptor (IGF-1R) signaling. Here, we describe specific and distinct roles for these hormones and receptors. We have found that IGF-1R, but not IR, expression is increased following CD4+ T cell activation or following differentiation toward Th17 cells. Although both insulin and IGF-1 increase the metabolism of CD4+ T cells, insulin has a more potent effect. However, IGF-1 has a unique role and acts specifically on Th17 cells to increase IL-17 production and Th17 cell metabolism. Furthermore, IGF-1 decreases mitochondrial membrane potential and mitochondrial reactive oxygen species (mROS) in Th17 cells, providing a cytoprotective effect. Interestingly, both IR and IGF-1R are required for this effect of IGF-1 on mitochondria, which suggests that the hybrid IR/IGF-1R may be required for mediating the effect of IGF-1 on mitochondrial membrane potential and mROS production.


Insulin-Like Growth Factor I , Insulin , Insulin-Like Growth Factor I/metabolism , Insulin/metabolism , Receptor, IGF Type 1/metabolism , Receptor, Insulin/metabolism , Signal Transduction , Mitochondria/metabolism , CD4-Positive T-Lymphocytes/metabolism
17.
Geroscience ; 46(3): 3481-3501, 2024 Jun.
Article En | MEDLINE | ID: mdl-38388918

Cerebrovascular fragility and cerebral microhemorrhages (CMH) contribute to age-related cognitive impairment, mobility defects, and vascular cognitive impairment and dementia, impairing healthspan and reducing quality of life in the elderly. Insulin-like growth factor 1 (IGF-1) is a key vasoprotective growth factor that is reduced during aging. Circulating IGF-1 deficiency leads to the development of CMH and other signs of cerebrovascular dysfunction. Here our goal was to understand the contribution of IGF-1 signaling on vascular smooth muscle cells (VSMCs) to the development of CMH and associated gait defects. We used an inducible VSMC-specific promoter and an IGF-1 receptor (Igf1r) floxed mouse line (Myh11-CreERT2 Igf1rf/f) to knockdown Igf1r. Angiotensin II in combination with L-NAME-induced hypertension was used to elicit CMH. We observed that VSMC-specific Igf1r knockdown mice had accelerated development of CMH, and subsequent associated gait irregularities. These phenotypes were accompanied by upregulation of a cluster of pro-inflammatory genes associated with VSMC maladaptation. Collectively our findings support an essential role for VSMCs as a target for the vasoprotective effects of IGF-1, and suggest that VSMC dysfunction in aging may contribute to the development of CMH.


Hypertension , Muscle, Smooth, Vascular , Receptor, IGF Type 1 , Aged , Animals , Humans , Mice , Gait , Hypertension/genetics , Hypertension/complications , Insulin-Like Growth Factor I/metabolism , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Receptor, IGF Type 1/genetics , Gait Disorders, Neurologic/genetics
18.
Signal Transduct Target Ther ; 9(1): 31, 2024 Feb 12.
Article En | MEDLINE | ID: mdl-38342897

Pancreatic ductal adenocarcinoma (PDAC) is a lethal disease, characterized by an intense desmoplastic reaction that compresses blood vessels and limits nutrient supplies. PDAC aggressiveness largely relies on its extraordinary capability to thrive and progress in a challenging tumor microenvironment. Dysregulation of the onco-suppressor miR-15a has been extensively documented in PDAC. Here, we identified the transcription factor Fos-related antigen-2 (Fra-2) as a miR-15a target mediating the adaptive mechanism of PDAC to nutrient deprivation. We report that the IGF1 signaling pathway was enhanced in nutrient deprived PDAC cells and that Fra-2 and IGF1R were significantly overexpressed in miR-15a downmodulated PDAC patients. Mechanistically, we discovered that miR-15a repressed IGF1R expression via Fra-2 targeting. In miR-15a-low context, IGF1R hyperactivated mTOR, modulated the autophagic flux and sustained PDAC growth in nutrient deprivation. In a genetic mouse model, Mir15aKO PDAC showed Fra-2 and Igf1r upregulation and mTOR activation in response to diet restriction. Consistently, nutrient restriction improved the efficacy of IGF1R inhibition in a Fra-2 dependent manner. Overall, our results point to a crucial role of Fra-2 in the cellular stress response due to nutrient restriction typical of pancreatic cancer and support IGF1R as a promising and vulnerable target in miR-15a downmodulated PDAC.


Carcinoma, Pancreatic Ductal , MicroRNAs , Pancreatic Neoplasms , Humans , Animals , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Fos-Related Antigen-2 , Cell Line, Tumor , Pancreatic Neoplasms/pathology , Carcinoma, Pancreatic Ductal/pathology , TOR Serine-Threonine Kinases , Tumor Microenvironment , Receptor, IGF Type 1/genetics
19.
Sci Rep ; 14(1): 3565, 2024 02 12.
Article En | MEDLINE | ID: mdl-38347000

Gout is a common autoinflammatory joint diseases characterized by deposition of monosodium urate (MSU) crystals which trigger an innate immune response mediated by inflammatory cytokines. IGF1R is one of the loci associated with both urate levels and gout susceptibility in GWAS to date, and IGF-1-IGF-1R signaling is implicated in urate control. We investigate the role of IGF-1/IGF1R signaling in the context of gouty inflammation. Also, we test the gout and urate-associated IGF1R rs6598541 polymorphism for association with the inflammatory capacity of mononuclear cells. For this, freshly isolated human peripheral blood mononuclear cells (PBMCs) were exposed to recombinant IGF-1 or anti-IGF1R neutralizing antibody in the presence or absence of solubilized urate, stimulated with LPS/MSU crystals. Also, the association of rs6598541 with IGF1R and protein expression and with ex vivo cytokine production levels after stimulation with gout specific stimuli was tested. Urate exposure was not associated with IGF1R expression in vitro or in vivo. Modulation of IGF1R did not alter urate-induced inflammation. Developing urate-induced trained immunity in vitro was not influenced in cells challenged with IGF-1 recombinant protein. Moreover, the IGF1R rs6598541 SNP was not associated with cytokine production. Our results indicate that urate-induced inflammatory priming is not regulated by IGF-1/IGF1R signaling in vitro. IGF1R rs6598541 status was not asociated with IGF1R expression or cytokine production in primary human PBMCs. This study suggests that the role of IGF1R in gout is tissue-specific and may be more relevant in the control of urate levels rather than in inflammatory signaling in gout.


Gout , Hyperuricemia , Humans , Uric Acid/metabolism , Hyperuricemia/complications , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Leukocytes, Mononuclear/metabolism , Genome-Wide Association Study , Gout/genetics , Gout/complications , Inflammation/metabolism , Cytokines/metabolism , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism
20.
Reprod Biol ; 24(1): 100850, 2024 Mar.
Article En | MEDLINE | ID: mdl-38262267

Gestational diabetes mellitus (GDM) is a prevalent metabolic disturbance in pregnancy. This article investigated the correlations between serum IGF1R and ATG7 with insulin resistance (IR) in GDM patients. Firstly, 100 GDM patients and 100 healthy pregnant women were selected as study subjects. The levels of serum IGF1, IGF1R, and ATG7 and their correlations with the insulin resistance index homeostasis model assessment of insulin resistance (HOMA-IR) were measured and analyzed by ELISA and Pearson. Additionally, in mouse pancreatic ß cells, IGF1R, ATG7, Beclin-1, and LC3-II/LC3-I levels, cell viability/apoptosis, and insulin level were assessed by western blot, CCK-8, flow cytometry, and ELISA. The GDM group exhibited obviously raised serum IGF1 level and diminished serum IGF1R/ATG7 levels. The IGF1 level was positively correlated with HOMA-IR, while IGF1R/ATG7 levels were negatively correlated with HOMA-IR in GDM patients. Collectively, IGF1R stimulated cell viability, suppressed apoptosis, amplified insulin secretion, and increased ATG7 expression to induce cell autophagy, which could be partially averted by ATG7 silencing.


Diabetes, Gestational , Insulin Resistance , Insulin-Secreting Cells , Animals , Mice , Pregnancy , Humans , Female , Diabetes, Gestational/metabolism , Insulin Secretion , Insulin-Secreting Cells/chemistry , Insulin-Secreting Cells/metabolism , Blood Glucose/analysis , Blood Glucose/metabolism , Insulin , Receptor, IGF Type 1/metabolism
...