Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 577
Filter
1.
Front Immunol ; 12: 672519, 2021.
Article in English | MEDLINE | ID: mdl-33995414

ABSTRACT

Inflammatory arthritis is burdened by an increased risk of metabolic disorders. Cytokines and other mediators in inflammatory diseases lead to insulin resistance, diabetes and hyperlipidemia. Accumulating evidence in the field of immunometabolism suggests that the cause-effect relationship between arthritis and metabolic abnormalities might be bidirectional. Indeed, the immune response can be modulated by various factors such as environmental agents, bacterial products and hormones. Insulin is produced by pancreatic cells and regulates glucose, fat metabolism and cell growth. The action of insulin is mediated through the insulin receptor (IR), localized on the cellular membrane of hepatocytes, myocytes and adipocytes but also on the surface of T cells, macrophages, and dendritic cells. In murine models, the absence of IR in T-cells coincided with reduced cytokine production, proliferation, and migration. In macrophages, defective insulin signaling resulted in enhanced glycolysis affecting the responses to pathogens. In this review, we focalize on the bidirectional cause-effect relationship between impaired insulin signaling and arthritis analyzing how insulin signaling may be involved in the aberrant immune response implicated in arthritis and how inflammatory mediators affect insulin signaling. Finally, the effect of glucose-lowering agents on arthritis was summarized.


Subject(s)
Arthritis/immunology , Insulin/immunology , Signal Transduction/physiology , Animals , Arthritis/metabolism , Humans , Insulin/metabolism , Receptor, Insulin/immunology , Receptor, Insulin/metabolism
2.
Domest Anim Endocrinol ; 74: 106510, 2021 01.
Article in English | MEDLINE | ID: mdl-32652390

ABSTRACT

Prolonged hyperinsulinemia is thought to be the cause of equine endocrinopathic laminitis, a common and crippling disease of the foot, for which there are no pharmacologic treatments other than pain relief. It has been suggested that insulin causes its effects on the lamellae by activating IGF-1 receptors (IGF-1R), as insulin receptors (InsR) are scarce in this tissue, whereas IGF-1R are abundant and become downregulated after prolonged insulin infusion. As a first step toward confirming this mechanism and beginning to develop a therapeutic anti-IGF-1R monoclonal antibody (mAb) for horses, it was necessary to identify available human IGF-1R mAbs that would recognize equine receptors. Four IGF-1R mAbs were tested using soluble equine IGF-1R, with ELISA and flow cytometry. Frozen equine lamellar and liver tissue was also used in radioligand binding assays. The results demonstrated that only one of the mAbs tested (mAb1) was able to compete effectively with IGF-1 for binding to its receptors in equine lamellar tissue, with an IC50 of 5 to 159 ng/mL. None of the 4 mAbs were able to bind to equine hepatic InsR. This study has generated valuable structure-activity information and has identified a prototype anti-IGF-1R mAb suitable for further development.


Subject(s)
Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacology , Horse Diseases/drug therapy , Receptor, IGF Type 1/antagonists & inhibitors , Receptor, IGF Type 1/immunology , Animals , Antibodies, Monoclonal/chemistry , Foot Diseases/drug therapy , Foot Diseases/etiology , Foot Diseases/veterinary , Horses , Humans , Hyperinsulinism/complications , Hyperinsulinism/veterinary , Liver/chemistry , Receptor, Insulin/antagonists & inhibitors , Receptor, Insulin/immunology , Structure-Activity Relationship
3.
Diabetes ; 69(11): 2481-2489, 2020 11.
Article in English | MEDLINE | ID: mdl-32816962

ABSTRACT

Loss-of-function mutations in both alleles of the human insulin receptor gene (INSR) cause extreme insulin resistance (IR) and usually death in childhood, with few effective therapeutic options. Bivalent antireceptor antibodies can elicit insulin-like signaling by mutant INSR in cultured cells, but whether this translates into meaningful metabolic benefits in vivo, wherein the dynamics of insulin signaling and receptor recycling are more complex, is unknown. To address this, we adopted a strategy to model human insulin receptoropathy in mice, using Cre recombinase delivered by adeno-associated virus to knockout endogenous hepatic Insr acutely in floxed Insr mice (liver insulin receptor knockout [L-IRKO] + GFP), before adenovirus-mediated add back of wild-type (WT) or mutant human INSR Two murine anti-INSR monoclonal antibodies, previously shown to be surrogate agonists for mutant INSR, were then tested by intraperitoneal injections. As expected, L-IRKO + GFP mice showed glucose intolerance and severe hyperinsulinemia. This was fully corrected by add back of WT but not with either D734A or S350L mutant INSR. Antibody injection improved glucose tolerance in D734A INSR-expressing mice and reduced hyperinsulinemia in both S350L and D734A INSR-expressing animals. It did not cause hypoglycemia in WT INSR-expressing mice. Antibody treatment also downregulated both WT and mutant INSR protein, attenuating its beneficial metabolic effects. Anti-INSR antibodies thus improve IR in an acute model of insulin receptoropathy, but these findings imply a narrow therapeutic window determined by competing effects of antibodies to stimulate receptors and induce their downregulation.


Subject(s)
Antibodies , Disease Models, Animal , Hyperglycemia/therapy , Insulin/immunology , Receptor, Insulin/immunology , Receptor, Insulin/metabolism , Animals , Blood Glucose , Gene Expression Regulation , Liver/metabolism , Male , Mice , Mice, Knockout , RNA, Messenger/genetics , RNA, Messenger/metabolism , Random Allocation , Receptor, Insulin/genetics
4.
J Exp Med ; 217(8)2020 08 03.
Article in English | MEDLINE | ID: mdl-32478834

ABSTRACT

Adipose tissue (AT) regulatory T cells (T regs) control inflammation and metabolism. Diet-induced obesity causes hyperinsulinemia and diminishes visceral AT (VAT) T reg number and function, but whether these two phenomena were mechanistically linked was unknown. Using a T reg-specific insulin receptor (Insr) deletion model, we found that diet-induced T reg dysfunction is driven by T reg-intrinsic insulin signaling. Compared with Foxp3cre mice, after 13 wk of high-fat diet, Foxp3creInsrfl/fl mice exhibited improved glucose tolerance and insulin sensitivity, effects associated with lower AT inflammation and increased numbers of ST2+ T regs in brown AT, but not VAT. Similarly, Foxp3creInsrfl/fl mice were protected from the metabolic effects of aging, but surprisingly had reduced VAT T regs and increased VAT inflammation compared with Foxp3cre mice. Thus, in both diet- and aging-associated hyperinsulinemia, excessive Insr signaling in T regs leads to undesirable metabolic outcomes. Ablation of Insr signaling in T regs represents a novel approach to mitigate the detrimental effects of hyperinsulinemia on immunoregulation of metabolic syndrome.


Subject(s)
Aging/immunology , Diet, High-Fat/adverse effects , Intra-Abdominal Fat/immunology , Metabolic Syndrome/immunology , Receptor, Insulin/deficiency , T-Lymphocytes, Regulatory/immunology , Aging/genetics , Aging/pathology , Animals , Gene Deletion , Intra-Abdominal Fat/pathology , Metabolic Syndrome/chemically induced , Metabolic Syndrome/genetics , Metabolic Syndrome/pathology , Mice , Mice, Transgenic , Receptor, Insulin/immunology , T-Lymphocytes, Regulatory/pathology
5.
Life Sci ; 256: 117910, 2020 Sep 01.
Article in English | MEDLINE | ID: mdl-32504753

ABSTRACT

AIMS: Insulin (Ins) covalently modified by catecholestrogens (CEs) was commonly found in diabetic patients who have developed insulin resistance. Estrogenization of insulin altered its molecular function and effect carbohydrates metabolisms in these patients. Insulin resistance is a common phenomenon in diabetes but the exact mechanism remains unknown. In this study, binding specificity and affinity of autoantibodies against estrogenized insulin (4-hydroxyestradiol-insulin; 4-OHE2-Ins) were assayed in the serum of type 1 diabetes (T1D) patients in order to explain the phenomena behind insulin resistance. MATERIALS AND METHODS: Specificity and affinity of autoantibodies from the sera of 66 T1D patients and 41 controls were analyzed by direct binding, competition ELISA and quantitative precipitin titration. Insulin was also estimated in the serum of T1D patients by ELISA. KEY FINDING: Estrogenized insulin (4-OHE2-Ins) exhibited high affinity and specificity to T1D autoantibodies in comparison to Ins (p < .05) or 4-OHE2 (p < .001). Estrogenization of insulin alters its interaction with the insulin receptor (IR). The affinity constant of 4-OHE2-Ins with the T1D autoantibodies was found to be 1.41 × 10-7 M. SIGNIFICANCE: Estrogenization of insulin by catecholestrogen makes these molecules highly antigenic and produced high-affinity autoantibodies in T1D patients. As a result, patients develop insulin resistance and presented this molecule as a potential biomarker for T1D.


Subject(s)
Autoantibodies/immunology , Diabetes Mellitus, Type 1/drug therapy , Estrogens, Catechol/chemistry , Hypoglycemic Agents/chemistry , Insulin/chemistry , Adult , Autoantibodies/metabolism , Biomarkers/metabolism , Blood Glucose/analysis , Blood Specimen Collection , Competitive Bidding , Enzyme-Linked Immunosorbent Assay , Estrogens, Catechol/therapeutic use , Female , Humans , Hypoglycemic Agents/therapeutic use , Insulin/therapeutic use , Insulin Resistance , Male , Middle Aged , Receptor, Insulin/immunology , Receptor, Insulin/metabolism , Sensitivity and Specificity
6.
Acta Trop ; 209: 105552, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32485167

ABSTRACT

Insulin signaling pathway is an ancient and highly conserved pathway known to play critical roles in cell growth, control and metabolic regulation. In this study, we identified and characterized two insulin receptor genes (TsIR-1316 and TsIR-4810) from Taenia solium. TsIR-1316 was grouped with E. multilocularis insulin receptor (EmIR-1) and TsIR-4810 was closer to Taenia pisiformis insulin-like growth factor receptor (TpIR) on the same branch with a very high bootstrap value. TsIR-1316 was located on the integument of larvae and adult worms, as well as the ovary of adults and eggs. Alternatively, TsIR-4810 was located in the parenchyma and reproductive organs of the adult worms. By using in vitro cultivation systems with Cysticercus pisiformis as a model, we demonstrated that anti-TsIRs-LBD antibodies could effectively block the insulin signaling pathway, resulting in reduced phosphorylation of the insulin receptor as well as lower levels of glucose uptake and glycogen synthesis. The rabbits immunized with TsIR-1316-LBD, TsIR-4810-LBD and TsIR-1316-LBD + TsIR-4810-LBD produced protection against infection of T. pisiformis as demonstrated by a 94.6%, 96% and 80% reduction of establishment of larvae, respectively. These data suggested that TsIR-1316-LBD and TsIR-4810-LBD are promising vaccine candidates or novel drug targets against swine cysticercosis.


Subject(s)
Cysticercosis/prevention & control , Receptor, Insulin/immunology , Taenia solium/immunology , Vaccines/immunology , Animals , Cysticercosis/drug therapy , Female , Rabbits , Receptor, Insulin/antagonists & inhibitors , Receptor, Insulin/genetics , Receptor, Insulin/physiology , Swine , Swine Diseases/prevention & control
7.
Horm Metab Res ; 51(11): 723-728, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31683342

ABSTRACT

Insulin autoimmune syndrome (IAS) and type B insulin resistance syndrome (B-IRS) are rare autoimmune dysglycemia syndromes, but their treatment and prognosis are different. This study aimed to provide a basis for the clinical differential diagnosis of IAS and B-IRS. This was a retrospective study of the medical records of all patients diagnosed with IAS or B-IRS between January 2006 and March 2018 at the Chinese PLA General Hospital. Demographic, clinical, biochemistry, treatment, and follow-up data were examined. There were several different biochemical parameters between IAS (n=13) and B-IRS (n=6): white blood count (WBC, 7.05±3.06 vs. 2.70±0.73×109/l, p=0.004), platelet (249±56.6 vs. 111±68.0×109/l, p<0.001), serum creatine (59.0±17.8 vs. 43.1±7.05 µmol/l, p=0.013), serum albumin (42.3±5.17 vs. 33.6±3.40 g/l, p=0.002), triglyceride (median, 1.33 (1.01, 1.93) vs. 0.56 (0.50, 0.79) mmol/l, p=0.002), plasma IgG (1183±201 vs. 1832±469 mg/ml, p=0.018), IgA (328±140 vs. 469±150 mg/ml, p=0.018), and C3 (128±23.4 vs. 45.3±13.5 mg/l, p<0.001). Fasting insulin in the IAS and B-IRS patients was high (299-4708 vs. 118-851 mU/l, p=0.106), and there was a difference in 2 h oral glucose tolerance test insulin (4217-8343 mU/l vs. 274-1143 mU/l, p=0.012). Glycated hemoglobin (HbA1c) in the B-IRS patients was higher than in IAS patients (114±14.4. vs. 40.6±8.89 mmol/mol, p<0.001). Serum insulin-like growth factor-1 (IGF-1) was lower in all B-IRS patients (25±0.00 vs. 132±52.7 ng/ml, p<0.001). Although IAS and B-IRS are autoimmune hyperinsulinemic dysglycemic syndromes, several clinical parameters (body mass index, HbA1c, WBC, platelet, albumin, triglyceride, IgG, C3, and IGF-1) are different between these two syndromes.


Subject(s)
Autoantibodies/blood , Autoimmune Diseases/diagnosis , Biomarkers/blood , Insulin Resistance , Metabolic Syndrome/diagnosis , Receptor, Insulin/immunology , Autoimmune Diseases/blood , Autoimmune Diseases/immunology , Diagnosis, Differential , Female , Follow-Up Studies , Humans , Male , Metabolic Syndrome/blood , Metabolic Syndrome/immunology , Middle Aged , Prognosis , Retrospective Studies
8.
J Immunol Methods ; 465: 20-26, 2019 02.
Article in English | MEDLINE | ID: mdl-30500329

ABSTRACT

Translation across species of immunoassay results is often challenging due to the lack of cross-species reactivity of antibodies. In order to investigate the biology of insulin and IGF1 receptors, we generated new versatile monoclonal assay antibodies using the extracellular domain of the insulin/IGF1 hybrid receptor as the bait protein in the Adimab yeast antibody discovery platform and as the antigen in a rabbit monoclonal antibody platform. The resulting antibody clones were screened for receptor specificity as well as cross-species reactivity to both tissue and cell line derived samples. Using these strategies, we were able to identify highly specific insulin receptor monoclonal antibodies that lack cross-reactivity to the IGF1 receptor using the Adimab platform and a highly specific IGF1 receptor monoclonal antibody that lacks cross-reactivity to the insulin receptor using the rabbit antibody platform. Unlike earlier monoclonal antibodies reported in the literature, these antibodies show cross-species reactivity to the extracellular domains of mouse, rat, pig, and human receptors, indicating that they bind conserved epitopes. Furthermore, the antibodies work well in several different assay formats, including ELISA, flow cytometry, and immunoprecipitation, and therefore provide new tools to study insulin and IGF1 receptor biology with translation across several species and experimental model systems.


Subject(s)
Antibodies, Monoclonal/immunology , Receptor, IGF Type 1/immunology , Receptor, Insulin/immunology , Animals , Antibodies, Monoclonal/chemistry , Cross Reactions , HCT116 Cells , Humans , Mice , Rabbits , Rats , Species Specificity , Swine
9.
Cell Death Dis ; 9(10): 1043, 2018 10 11.
Article in English | MEDLINE | ID: mdl-30310051

ABSTRACT

Immune escape due to immunosuppressive microenvironments, such as those associated with regulatory T (Treg) cells is highly associated with initial occurrence and development of solid tumors or hematologic malignancies. Here, we employed high-throughput transcriptome screening to demonstrate immunosuppression-associated increases in the long noncoding (lnc) RNA lnc-insulin receptor precursor (INSR), which was corrected with INSR expression in CD4+ T cells extracted from the bone marrow of patients with childhood acute T lymphoblastic leukemia. Loss-of-function and gain-of-function assays in vitro and in vivo revealed that membrane-localized and cytoplasm-localized lnc-INSR promoted Treg distribution and decreased the percentage of cytotoxic T lymphocytes, which induced tumor growth. Through direct binding with INSR, lnc-INSR blocked the INSR ubiquitination site, causing abnormal activation of INSR and the phosphatidylinositide 3-kinase/AKT-signaling pathway. These results indicated that lnc-INSR might promote immune suppression by enhancing Treg-cell differentiation and serve as valuable therapeutic targets in the immunosuppressive tumor microenvironment.


Subject(s)
Antigens, CD/immunology , Bone Marrow/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , RNA, Long Noncoding/immunology , Receptor, Insulin/immunology , Tumor Microenvironment/genetics , Adolescent , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cell Proliferation/physiology , Child , Humans , Immunosuppression Therapy/methods , Jurkat Cells , Male , Mice , Mice, Inbred NOD , Mice, SCID , Phosphatidylinositol 3-Kinase/immunology , Proto-Oncogene Proteins c-akt/immunology , Signal Transduction/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Regulatory/immunology , Transcriptome/immunology
10.
Int J Mol Sci ; 19(10)2018 Oct 09.
Article in English | MEDLINE | ID: mdl-30304851

ABSTRACT

There is a pressing need to develop vaccines for schistosomiasis given the current heavy dependency on praziquantel as the only available drug for treatment. We previously showed the ligand domain of the Schistosoma japonicum insulin receptor 1 and 2 (rSjLD1 and 2) fusion proteins conferred solid protection in mice against challenge infection with S. japonicum. To improve vaccine efficacy, we compared the immunogenicity and protective efficacy of rSjLD1 on its own and in combination with S. japonicum triose-phosphate isomerase (SjTPI), formulated with either of two adjuvants (QuilA and montanide ISA 720VG) in murine vaccine trials against S. japonicum challenge. The level of protection was higher in mice vaccinated only with rSjLD1 formulated with either adjuvant; rSjTPI or the rSjTPI-rSjLD1 combination resulted in a lower level of protection. Mirroring our previous results, there were significant reductions in the number of female worms (30⁻44%), faecal eggs (61⁻68%), liver eggs (44⁻56%), intestinal eggs (46⁻48%) and mature intestinal eggs (58⁻63%) in the rSjLD1-vaccinated mice compared with the adjuvant only groups. At 6-weeks post-cercarial challenge, a significantly increased production of interferon gamma (IFNγ) in rSjLD1-stimulated splenic CD4⁺ T cells was observed in the rSjLD1-vaccinated mice suggesting a Th1-type response is associated with the generated level of protective efficacy.


Subject(s)
Immunity , Receptor, Insulin/immunology , Recombinant Proteins/immunology , Schistosoma japonicum/immunology , Schistosomiasis japonica/immunology , Schistosomiasis japonica/prevention & control , Animals , Antibodies, Protozoan/immunology , Cross Reactions/immunology , Cytokines/metabolism , Disease Models, Animal , Female , Fluorescent Antibody Technique , Glucose/metabolism , Immunization , Immunoglobulin G/immunology , Male , Mice , Protozoan Vaccines/immunology , Schistosomiasis japonica/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
11.
Diabetes Care ; 41(11): 2353-2360, 2018 11.
Article in English | MEDLINE | ID: mdl-30201849

ABSTRACT

OBJECTIVE: Type B insulin resistance due to autoantibodies against the insulin receptor is characterized by diabetes refractory to massive doses of insulin, severe hypercatabolism, hyperandrogenism, and a high mortality rate. We analyzed the efficacy of combined immunosuppressive therapy in the management of this extreme form of diabetes. RESEARCH DESIGN AND METHODS: We performed a prospective cohort study including patients with confirmed insulin receptor autoantibodies, monitored for median 72 months (25th, 75th interquartile range 25, 88), and treated with rituximab, high-dose pulsed steroids, and cyclophosphamide until remission, followed by maintenance therapy with azathioprine. Remission was defined as the amelioration of the hyperglycemia and discontinuation of insulin and/or normalization of hyperandrogenemia. RESULTS: All data are given as median (25th, 75th interquartile range). Twenty-two patients aged 42 (25, 57) years, 86.4% women, fulfilled inclusion criteria. At baseline, fasting glucose was 307 (203, 398) mg/dL, HbA1c was 11.8% (9.7, 13.6), total testosterone (women) was 126 (57, 571) ng/dL (normal 8-60), and daily insulin requirement was 1,775 (863, 2,700) units. After 5 (4, 6.3) months, 86.4% (19 of 22) of patients achieved remission, documented by discontinuation of insulin in all patients, normal fasting glucose of 80 (76, 92) mg/dL, HbA1c of 5.5% (5.2, 6), and testosterone (women) of 28 (20, 47) ng/dL. During follow-up of 72 (25, 88) months, 13.6% (3 of 22) of patients developed disease recurrence, occurring 24 (22, 36) months after initial remission, which responded to repeated therapy. None of the patients died. CONCLUSIONS: Combined immunosuppressive therapy has changed the natural history of this disease, from 54% mortality to a curable form of diabetes and, as such, should be recommended in patients with type B insulin resistance.


Subject(s)
Cyclophosphamide/administration & dosage , Diabetes Mellitus, Type 1/drug therapy , Immunosuppressive Agents/administration & dosage , Insulin Resistance , Insulin/administration & dosage , Adult , Antigens, CD/immunology , Autoantibodies/blood , Azathioprine/therapeutic use , Cohort Studies , Cyclophosphamide/adverse effects , Dexamethasone/administration & dosage , Dexamethasone/adverse effects , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Drug Therapy, Combination , Female , Follow-Up Studies , Humans , Hyperglycemia/blood , Hyperglycemia/drug therapy , Hyperglycemia/immunology , Immunosuppressive Agents/adverse effects , Insulin/adverse effects , Insulin Resistance/immunology , Maintenance Chemotherapy , Male , Methylprednisolone/administration & dosage , Methylprednisolone/adverse effects , Middle Aged , Receptor, Insulin/immunology , Remission Induction/methods , Rituximab/administration & dosage , Rituximab/adverse effects , Severity of Illness Index , Syndrome
12.
Cell Metab ; 28(6): 922-934.e4, 2018 12 04.
Article in English | MEDLINE | ID: mdl-30174303

ABSTRACT

T cells represent a critical effector of cell-mediated immunity. Activated T cells engage in metabolic reprogramming during effector differentiation to accommodate dynamic changes in energy demands. Here, we show that the hormone, insulin, and downstream signaling through its insulin receptor shape adaptive immune function through modulating T cell metabolism. T cells lacking insulin receptor expression (LckCre+ Insrfl/fl) show reduced antigen-specific proliferation and compromised production of pro-inflammatory cytokines. In vivo, T cell-specific insulin receptor deficiency reduces T cell-driven colonic inflammation. In a model of severe influenza infection with A/PR8 (H1N1), lack of insulin receptor on T cells curtails antigen-specific immunity to influenza viral antigens. Mechanistically, insulin receptor signaling reinforces a metabolic program that supports T cell nutrient uptake and associated glycolytic and respiratory capacities. These data highlight insulin receptor signaling as an important node integrating immunometabolic pathways to drive optimal T cell effector function in health and disease.


Subject(s)
Antigens, CD/immunology , Immunity, Cellular/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza, Human/immunology , Lymphocyte Activation/immunology , Receptor, Insulin/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes/immunology , Animals , Antigens, CD/genetics , Cytokines/immunology , Cytokines/metabolism , Glycolysis/immunology , Humans , Inflammation/immunology , Inflammation/virology , Insulin/metabolism , Lymph Nodes , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections , Receptor, Insulin/genetics , Signal Transduction , Spleen , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism
13.
J Crohns Colitis ; 12(12): 1459-1474, 2018 Nov 28.
Article in English | MEDLINE | ID: mdl-30137286

ABSTRACT

BACKGROUND AND AIMS: Epithelial expression of the insulin receptor in the colon has previously been reported to correlate with extent of colonic inflammation. However, the impact of insulin signalling in the intestinal mucosa is still unknown. Here, we investigated the effects of inactivating the epithelial insulin receptor in the intestinal tract, in an experimental model of inflammation-induced colorectal cancer. METHODS: The mice were generated by utilizing the intestinal- and epithelial-specific villin promoter and the Cre-Lox technology. All mice included in the cohorts were generated by crossing [vil-Cre-INSR+/-] × [INSRfl/fl] to obtain [vil-Cre-INSR-/-], and their floxed littermates [INSRfl/fl] served as the control group. For the intervention study, phosphate-buffered saline with or without insulin was instilled rectally in anaesthetized wild-type mice with chemically induced colitis. RESULTS: We found higher endoscopic colitis scores together with potentiated colonic tumorigenesis in the knockout mice. Furthermore, we showed that topically administered insulin in inflamed colons of wild-type mice reduced inflammation-induced weight loss and improved remission in a dose-dependent manner. Mice receiving rectal insulin enemas exhibited lower colitis endoscopic scores and reduced cyclooxygenase 2 mRNA expression, and developed significantly fewer and smaller tumours compared with the control group receiving phosphate-buffered saline only. CONCLUSIONS: Rectal insulin therapy could potentially be a novel treatment, targeting the epithelial layer to enhance mucosal healing in ulcerated areas. Our findings open up new possibilities for combination treatments to synergize with the existing anti-inflammatory therapies.


Subject(s)
Colitis , Colorectal Neoplasms , Inflammation , Insulin/administration & dosage , Intestinal Mucosa , Administration, Rectal , Animals , Colitis/drug therapy , Colitis/etiology , Colitis/immunology , Colitis/pathology , Colorectal Neoplasms/etiology , Colorectal Neoplasms/pathology , Colorectal Neoplasms/prevention & control , Crossing Over, Genetic , Disease Models, Animal , Endoscopy/methods , Hypoglycemic Agents/administration & dosage , Inflammation/drug therapy , Inflammation/pathology , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Mice , Microfilament Proteins/genetics , Receptor, Insulin/immunology
14.
Front Immunol ; 9: 1037, 2018.
Article in English | MEDLINE | ID: mdl-29868002

ABSTRACT

In this historical perspective, written in honor of Dr. William E. Paul, we describe the initial discovery of one of the dominant substrates for tyrosine phosphorylation stimulated by IL-4. We further describe how this "IL-4-induced phosphorylated substrate" (4PS) was characterized as a member of the insulin receptor substrate (IRS) family of large adaptor proteins that link IL-4 and insulin receptors to activation of the phosphatidyl-inositol 3' kinase pathway as well as other downstream signaling pathways. The relative contribution of the 4PS/IRS pathway to the early models of IL-4-induced proliferation and suppression of apoptosis are compared to our more recent understanding of the complex interplay between positive and negative regulatory pathways emanating from members of the IRS family that impact allergic responses.


Subject(s)
Receptor, Insulin/immunology , Receptors, Interleukin-13/metabolism , Receptors, Interleukin-4/metabolism , Animals , Cell Division , History, 20th Century , History, 21st Century , Humans , Insulin/metabolism , Insulin Receptor Substrate Proteins/immunology , Insulin Receptor Substrate Proteins/metabolism , Interleukin-13/metabolism , Interleukin-4/metabolism , Mice , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Receptor, Insulin/history , Receptors, Interleukin-13/immunology , Receptors, Interleukin-4/immunology , Signal Transduction , Tyrosine/metabolism , U937 Cells
15.
PLoS One ; 13(6): e0197478, 2018.
Article in English | MEDLINE | ID: mdl-29874257

ABSTRACT

BACKGROUND: The manufacture of insulin analogs requires sophisticated production procedures which can lead to differences in the structure, purity, and/or other physiochemical properties of resultant products that can affect their biologic activity. Here, we sought to compare originator and non-originator copies of insulin glargine for innate immune activity and mechanisms leading to differences in these response profiles in an in vitro model of human immunity. METHODS: An endothelial/dendritic cell-based innate immune model was used to study antigen-presenting cell activation, cytokine secretion, and insulin receptor signalling pathways induced by originator and non-originator insulin glargine products. Mechanistic studies included signalling pathway blockade with specific inhibitors, analysis of the products in a Toll-like receptor reporter cell line assay, and natural insulin removal from the products by immunopurification. FINDINGS: All insulin glargine products elicited at least a minor innate immune response comparable to natural human insulin, but some lots of a non-originator copy product induced the elevated secretion of the cytokines, IL-8 and IL-6. In studies aimed at addressing the mechanisms leading to differential cytokine production by these products, we found (1) the inflammatory response was not mediated by bacterial contaminants, (2) the innate response was driven by the native insulin receptor through the MAPK pathway, and (3) the removal of insulin glargine significantly reduced their capacity to induce innate activity. No evidence of product aggregates was detected, though the presence of some high molecular weight proteins argues for the presence of insulin glargine dimers or others contaminants in these products. CONCLUSION: The data presented here suggests some non-originator insulin glargine product lots drive heightened in vitro human innate activity and provides preliminary evidence that changes in the biochemical composition of non-originator insulin glargine products (dimers, impurities) might be responsible for their greater immunostimulatory potential.


Subject(s)
Dendritic Cells/drug effects , Endothelial Cells/drug effects , Insulin Glargine/immunology , Insulin/pharmacology , Antigens, CD/immunology , Dendritic Cells/immunology , Humans , Immunity, Innate/drug effects , Immunity, Innate/immunology , Insulin/analogs & derivatives , Insulin/chemistry , Insulin/immunology , Insulin Glargine/chemistry , Insulin, Long-Acting/immunology , Insulin, Long-Acting/pharmacology , Interleukin-6/immunology , Interleukin-8/immunology , Receptor, Insulin/immunology
16.
Diabetologia ; 61(7): 1662-1675, 2018 07.
Article in English | MEDLINE | ID: mdl-29700562

ABSTRACT

AIMS/HYPOTHESIS: Bi-allelic loss-of-function mutations in the INSR gene (encoding the insulin receptor [INSR]) commonly cause extreme insulin resistance and early mortality. Therapeutic options are limited, but anti-INSR antibodies have been shown to activate two mutant receptors, S323L and F382V. This study evaluates four well-characterised murine anti-INSR monoclonal antibodies recognising distinct epitopes (83-7, 83-14, 18-44, 18-146) as surrogate agonists for potential targeted treatment of severe insulin resistance arising from insulin receptoropathies. METHODS: Ten naturally occurring mutant human INSRs with defects affecting different aspects of receptor function were modelled and assessed for response to insulin and anti-INSR antibodies. A novel 3T3-L1 adipocyte model of insulin receptoropathy was generated, permitting conditional knockdown of endogenous mouse Insr by lentiviral expression of species-specific short hairpin (sh)RNAs with simultaneous expression of human mutant INSR transgenes. RESULTS: All expressed mutant INSR bound to all antibodies tested. Eight mutants showed antibody-induced autophosphorylation, while co-treatment with antibody and insulin increased maximal phosphorylation compared with insulin alone. After knockdown of mouse Insr and expression of mutant INSR in 3T3-L1 adipocytes, two antibodies (83-7 and 83-14) activated signalling via protein kinase B (Akt) preferentially over signalling via extracellular signal-regulated kinase 1/2 (ERK1/2) for seven mutants. These antibodies stimulated glucose uptake via P193L, S323L, F382V and D707A mutant INSRs, with antibody response greater than insulin response for D707A. CONCLUSIONS/INTERPRETATION: Anti-INSR monoclonal antibodies can activate selected naturally occurring mutant human insulin receptors, bringing closer the prospect of novel therapy for severe insulin resistance caused by recessive mutations.


Subject(s)
Adipocytes/drug effects , Antibodies/pharmacology , Glucose/metabolism , Hypoglycemic Agents/pharmacology , Insulin Resistance , Insulin/pharmacology , Receptor, Insulin/agonists , 3T3-L1 Cells , Adipocytes/immunology , Adipocytes/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, CD/metabolism , CHO Cells , Cricetulus , Humans , Insulin Resistance/genetics , Mice , Mutation , Phosphorylation , Receptor, Insulin/genetics , Receptor, Insulin/immunology , Receptor, Insulin/metabolism , Signal Transduction/drug effects
17.
Intern Med ; 57(16): 2359-2363, 2018 Aug 15.
Article in English | MEDLINE | ID: mdl-29526960

ABSTRACT

Type B insulin resistance syndrome is a rare autoimmune disease and no effective therapy has yet been established. On the other hand, it is known that Saibokuto, one type of Japanese Kampo medicine, may have beneficial effects on various symptoms associated with this disease and it is therefore occasionally prescribed for various immune disorders. We herein describe a case of type B insulin resistance syndrome in which anti-insulin receptor antibody disappeared and the patient's glycemic control markedly improved after the administration of Saibokuto. At first, we administered various anti-oral diabetic drugs and insulin therapy, but the patient's glycemic control became further aggravated. In addition, Helicobacter pylori eradication therapy was not effective, although its benefit has been reported. Interestingly, after the patient started taking Saibokuto, her glycemic control markedly improved. In addition, the patient's plasma insulin levels markedly decreased and anti-insulin receptor antibody became negative after taking Saibokuto. Taken together, there is a possibility that Saibokuto may one of the options for type B insulin resistance syndrome therapy.


Subject(s)
Antibodies/blood , Autoimmune Diseases/drug therapy , Insulin Resistance , Medicine, Kampo , Receptor, Insulin/immunology , Autoimmune Diseases/blood , Blood Glucose , Female , Humans , Insulin/therapeutic use , Male , Syndrome
18.
MAbs ; 10(5): 796-802, 2018 07.
Article in English | MEDLINE | ID: mdl-29589989

ABSTRACT

Loss-of-function mutations of the ß-cell ATP-sensitive potassium channels (KATP) cause the most common and severe form of congenital hyperinsulinism (KATPHI), a disorder of ß-cell function characterized by severe hypoglycemia. Children with KATPHI are typically unresponsive to medical therapy and require pancreatectomy for intractable hypoglycemia. We tested the hypothesis that inhibition of insulin receptor signaling may prevent hypoglycemia in KATPHI. To test this hypothesis, we examined the effect of an antibody allosteric inhibitor of the insulin receptor, XMetD, on fasting plasma glucose in a mouse model of KATPHI (SUR-1-/- mice). SUR-1-/- and wild-type mice received twice weekly intraperitoneal injections of either XMetD or control antibody for 8 wks. Treatment with XMetD significantly decreased insulin sensitivity, and increased hepatic glucose output and fasting plasma glucose. These findings support the potential use of insulin receptor antagonists as a therapeutic approach to control the hypoglycemia in congenital hyperinsulinism.


Subject(s)
Antibodies, Monoclonal/pharmacology , Disease Models, Animal , Hyperinsulinism/metabolism , Hypoglycemia/prevention & control , KATP Channels/metabolism , Receptor, Insulin/immunology , Animals , Antibodies, Monoclonal/immunology , Blood Glucose/metabolism , Fasting/blood , Humans , Hyperinsulinism/genetics , Hypoglycemia/genetics , Hypoglycemia/immunology , Insulin/blood , KATP Channels/genetics , Male , Mice, Inbred C57BL , Mice, Knockout
19.
Mol Metab ; 10: 87-99, 2018 04.
Article in English | MEDLINE | ID: mdl-29453154

ABSTRACT

OBJECTIVE: Insulin resistance is a key feature of Type 2 Diabetes (T2D), and improving insulin sensitivity is important for disease management. Allosteric modulation of the insulin receptor (IR) with monoclonal antibodies (mAbs) can enhance insulin sensitivity and restore glycemic control in animal models of T2D. METHODS: A novel human mAb, IRAB-A, was identified by phage screening using competition binding and surface plasmon resonance assays with the IR extracellular domain. Cell based assays demonstrated agonist and sensitizer effects of IRAB-A on IR and Akt phosphorylation, as well as glucose uptake. Lean and diet-induced obese mice were used to characterize single-dose in vivo pharmacological effects of IRAB-A; multiple-dose IRAB-A effects were tested in obese mice. RESULTS: In vitro studies indicate that IRAB-A exhibits sensitizer and agonist properties distinct from insulin on the IR and is translated to downstream signaling and function; IRAB-A bound specifically and allosterically to the IR and stabilized insulin binding. A single dose of IRAB-A given to lean mice rapidly reduced fed blood glucose for approximately 2 weeks, with concomitant reduced insulin levels suggesting improved insulin sensitivity. Phosphorylated IR (pIR) from skeletal muscle and liver were increased by IRAB-A; however, phosphorylated Akt (pAkt) levels were only elevated in skeletal muscle and not liver vs. control; immunochemistry analysis (IHC) confirmed the long-lived persistence of IRAB-A in skeletal muscle and liver. Studies in diet-induced obese (DIO) mice with IRAB-A reduced fed blood glucose and insulinemia yet impaired glucose tolerance and led to protracted insulinemia during a meal challenge. CONCLUSION: Collectively, the data suggest IRAB-A acts allosterically on the insulin receptor acting non-competitively with insulin to both activate the receptor and enhance insulin signaling. While IRAB-A produced a decrease in blood glucose in lean mice, the data in DIO mice indicated an exacerbation of insulin resistance; these data were unexpected and suggested the interplay of complex unknown pharmacology. Taken together, this work suggests that IRAB-A may be an important tool to explore insulin receptor signaling and pharmacology.


Subject(s)
Allosteric Site , Antibodies, Monoclonal/pharmacology , Hypoglycemic Agents/pharmacology , Receptor, Insulin/agonists , 3T3 Cells , Allosteric Regulation , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Blood Glucose/metabolism , Cell Line, Tumor , Humans , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/immunology , Insulin/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Receptor, Insulin/chemistry , Receptor, Insulin/immunology , Signal Transduction
20.
Am J Med ; 131(1): 102-106, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28822702

ABSTRACT

BACKGROUND: Type 2 diabetes mellitus is characterized by relative insulin deficiency and insulin resistance. Features suggesting severe insulin resistance include acanthosis nigricans, hyperandrogenism, weight loss, and recurrent hospital admissions for diabetic ketoacidosis. In rare circumstances, hyperglycemia persists despite administration of massive doses of insulin. In these cases, it is important to consider autoimmune etiologies for insulin resistance, such as type B insulin resistance and insulin antibody-mediated extreme insulin resistance, which carry high morbidity and mortality if untreated. Encouragingly, immunomodulatory regimens have recently been published that induce remission at high rates. METHODS/RESULTS: We describe 3 cases of extreme insulin resistance mediated by anti-insulin receptor autoantibodies or insulin autoantibodies. All cases were effectively treated with an immunomodulatory regimen. CONCLUSION: Although cases of extreme insulin resistance are rare, it is important to be aware of autoimmune causes, recognize suggestive signs and symptoms, and pursue appropriate diagnostic evaluation. Prompt treatment with immunomodulators is key to restoring euglycemia in patients with autoimmune etiologies of insulin resistance.


Subject(s)
Antibodies/immunology , Diabetes Mellitus, Type 2/immunology , Insulin Resistance/immunology , Insulin/therapeutic use , Receptor, Insulin/immunology , Azathioprine/administration & dosage , Azathioprine/therapeutic use , Cyclophosphamide/administration & dosage , Cyclophosphamide/therapeutic use , Dexamethasone/administration & dosage , Dexamethasone/therapeutic use , Drug Therapy, Combination , Female , Glucocorticoids/administration & dosage , Glucocorticoids/therapeutic use , Humans , Immunologic Factors/administration & dosage , Immunologic Factors/therapeutic use , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/therapeutic use , Insulin/administration & dosage , Insulin/immunology , Male , Middle Aged , Rituximab/administration & dosage , Rituximab/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...