Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
PLoS One ; 18(9): e0286256, 2023.
Article in English | MEDLINE | ID: mdl-37713409

ABSTRACT

FAM3C/ILEI is an important factor in epithelial-to-mesenchymal transition (EMT) induction, tumor progression and metastasis. Overexpressed in many cancers, elevated ILEI levels and secretion correlate with poor patient survival. Although ILEI's causative role in invasive tumor growth and metastasis has been demonstrated in several cellular tumor models, there are no available transgenic mice to study these effects in the context of a complex organism. Here, we describe the generation and initial characterization of a Tet-ON inducible Fam3c/ILEI transgenic mouse strain. We find that ubiquitous induction of ILEI overexpression (R26-ILEIind) at weaning age leads to a shortened lifespan, reduced body weight and microcytic hypochromic anemia. The anemia was reversible at a young age within a week upon withdrawal of ILEI induction. Vav1-driven overexpression of the ILEIind transgene in all hematopoietic cells (Vav-ILEIind) did not render mice anemic or lower overall fitness, demonstrating that no intrinsic mechanisms of erythroid development were dysregulated by ILEI and that hematopoietic ILEI hyperfunction did not contribute to death. Reduced serum iron levels of R26-ILEIind mice were indicative for a malfunction in iron uptake or homeostasis. Accordingly, the liver, the main organ of iron metabolism, was severely affected in moribund ILEI overexpressing mice: increased alanine transaminase and aspartate aminotransferase levels indicated liver dysfunction, the liver was reduced in size, showed increased apoptosis, reduced cellular iron content, and had a fibrotic phenotype. These data indicate that high ILEI expression in the liver might reduce hepatoprotection and induce liver fibrosis, which leads to liver dysfunction, disturbed iron metabolism and eventually to death. Overall, we show here that the novel Tet-ON inducible Fam3c/ILEI transgenic mouse strain allows tissue specific timely controlled overexpression of ILEI and thus, will serve as a versatile tool to model the effect of elevated ILEI expression in diverse tissue entities and disease conditions, including cancer.


Subject(s)
Anemia , Longevity , Mice , Animals , Longevity/genetics , Liver Cirrhosis/genetics , Anemia/genetics , Iron , Mice, Transgenic
2.
Eur J Cancer ; 138: 1-10, 2020 10.
Article in English | MEDLINE | ID: mdl-32818762

ABSTRACT

BACKGROUND: RAS mutations are the only validated biomarkers in metastatic colorectal cancer (mCRC) for anti-epidermal growth factor receptor (EGFR) therapy. Limited clinical information is available on AXL expression, marker of epithelial to mesenchymal transition, in mCRC. METHODS: AXL was retrospectively assessed by immunohistochemistry in 307 patients. RAS wild-type (WT) patients (N = 136) received first-line anti-EGFR-based therapy; RAS mutant patients (N = 171) received anti-angiogenic-based regimens. Preclinical experiments were performed using human RAS WT CRC cell lines and xenograft models. AXL RNA levels were assessed in a cohort of patients with available samples at baseline and at progression to anti-EGFR treatment and in the GSE5851 dataset. RESULTS: AXL was expressed in 55/307 tumour tissues, correlating with worse survival in the overall population (AXL-positive, 23.7 months; AXL-negative, 30.8 months; HR, 1.455, P = 0.032) and in RAS WT patients (AXL-positive, 23.0 months; AXL-negative, 35.8 months; HR,1.780, P = 0.032). Progression-free survival (PFS) in the RAS WT cohort was shorter in the AXL-positive cohort (6.2 months versus 12.1 months; HR, 1.796, P = 0.013). Three-dimensional cultures obtained from a patient following anti-EGFR therapy resulted AXL-positive, showing resistance to anti-EGFR drugs and sensitivity to AXL inhibition. AXL transfection in CRC cell lines induced AXL overexpression and resistance to the EGFR blockade. At progression to cetuximab, 2/10 SW48-tumour xenograft mice showed AXL expression. Consistently, AXL RNA levels increased in 5/7 patients following anti-EGFR therapy. Moreover, in the GSE5851 dataset higher AXL RNA levels correlated with worse PFS with cetuximab in KRAS-exon2 WT chemorefractory patients. CONCLUSIONS: AXL is a marker of poor prognosis in mCRC with consistent clinical and preclinical evidences of involvement in primary and acquired resistance to anti-EGFR drugs in RAS WT patients.


Subject(s)
Colorectal Neoplasms/drug therapy , ErbB Receptors/antagonists & inhibitors , Genes, ras , Proto-Oncogene Proteins/analysis , Receptor Protein-Tyrosine Kinases/analysis , Animals , Biomarkers , Cell Line, Tumor , Colorectal Neoplasms/chemistry , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm , Humans , Mice , Neoplasm Metastasis , Xenograft Model Antitumor Assays , Axl Receptor Tyrosine Kinase
3.
Mol Cancer Ther ; 18(4): 845-855, 2019 04.
Article in English | MEDLINE | ID: mdl-30824612

ABSTRACT

The EPHA2 tyrosine kinase receptor is implicated in tumor progression and targeted therapies resistance. We evaluated EPHA2 as a potential resistance marker to the antiepidermal growth factor receptor (EGFR) monoclonal antibody cetuximab in colorectal cancer. We studied activation of EPHA2 in a panel of human colorectal cancer cell lines sensitive or resistant to anti-EGFR drugs. The in vitro and in vivo effects of ALW-II-41-27 (an EPHA2 inhibitor) and/or cetuximab treatment were tested. Formalin-fixed paraffin-embedded tumor specimens from 82 RAS wild-type (WT) metastatic colorectal cancer patients treated with FOLFIRI + cetuximab as first-line therapy in the CAPRI-GOIM trial were assessed for EPHA2 expression by immunohistochemistry and correlated with treatment efficacy. EPHA2 was differentially activated in colorectal cancer cell lines. Combined treatment with ALW-II-41-27 plus cetuximab reverted primary and acquired resistance to cetuximab, causing cell growth inhibition, inducing apoptosis and cell-cycle G1-G2 arrest. In tumor xenograft models, upon progression to cetuximab, ALW-II-41-27 addition significantly inhibited tumor growth. EPHA2 protein expression was detected in 55 of 82 tumor samples, frequently expressed in less-differentiated and left-sided tumors. High levels of EPHA2 significantly correlated with worse progression-free survival [8.6 months; confidence interval (CI) 95%, 6.4-10.8; vs. 12.3 months; CI 95%, 10.4-14.2; P = 0.03] and with increased progression rate (29% vs. 9%, P = 0.02). A specific EPHA2 inhibitor reverts in vitro and in vivo primary and acquired resistance to anti-EGFR therapy. EPHA2 levels are significantly associated with worse outcome in patients treated with FOLFIRI + cetuximab. These results highlight EPHA2 as a potential therapeutic target in metastatic colorectal cancer.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Cetuximab/pharmacology , Cetuximab/therapeutic use , Colorectal Neoplasms/drug therapy , Drug Resistance, Neoplasm , Ephrin-A2/metabolism , Adult , Aged , Aged, 80 and over , Animals , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Benzamides/pharmacology , Biomarkers, Tumor/metabolism , Camptothecin/analogs & derivatives , Camptothecin/therapeutic use , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cetuximab/administration & dosage , Colorectal Neoplasms/pathology , Ephrin-A2/antagonists & inhibitors , ErbB Receptors/antagonists & inhibitors , Female , Fluorouracil/therapeutic use , Humans , Leucovorin/therapeutic use , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Progression-Free Survival , RNA Interference , Receptor, EphA2 , Signal Transduction/drug effects , Transfection , Xenograft Model Antitumor Assays
4.
Exp Clin Endocrinol Diabetes ; 127(8): 550-556, 2019 Sep.
Article in English | MEDLINE | ID: mdl-30075479

ABSTRACT

Obesity and type 2 diabetes mellitus have reached an epidemic level, thus novel treatment concepts need to be identified. Myostatin, a myokine known for restraining skeletal muscle growth, has been associated with the development of insulin resistance and type 2 diabetes mellitus. Yet, little is known about the regulation of myostatin in human obesity and insulin resistance. We aimed to investigate the regulation of myostatin in obesity and uncover potential associations between myostatin, metabolic markers and insulin resistance/sensitivity indices. Circulating active myostatin concentration was measured in the serum of twenty-eight severely obese non-diabetic patients compared to a sex and age matched lean and overweight control group (n=22). Insulin resistance/sensitivity was assessed in the obese group. Skeletal muscle and adipose tissue specimens from the obese group were collected during elective bariatric surgery. Adipose tissue samples from lean and overweight subjects were collected during elective abdominal surgery. Myostatin concentration was increased in obese compared to lean individuals, while myostatin adipose tissue expression did not differ. Muscle myostatin gene expression strongly correlated with expression of metabolic genes such as IRS1, PGC1α, SREBF1. Circulating myostatin concentration correlated positively with insulin resistance indices and negatively with insulin sensitivity indices. The best correlation was obtained for the oral glucose insulin sensitivity index. Our results point to an interesting correlation between myostatin and insulin resistance/sensitivity in humans, and emphasize its need for further evaluation as a pharmacological target in the prevention and treatment of obesity-associated metabolic complications.


Subject(s)
Diabetes Mellitus, Type 2/blood , Insulin Resistance , Myostatin/blood , Obesity/blood , Up-Regulation , Adolescent , Adult , Aged , Female , Humans , Male , Middle Aged , Muscle Proteins/biosynthesis , Muscle, Skeletal/metabolism
5.
Obes Res Clin Pract ; 12(3): 277-285, 2018.
Article in English | MEDLINE | ID: mdl-29519755

ABSTRACT

OBJECTIVE: Osteopontin (OPN, Spp1) is a protein upregulated in white adipose tissue (WAT) of obese subjects. Deletion of OPN protects mice from high-fat diet-induced WAT inflammation and insulin resistance. However, the alterations mediated by loss of OPN in WAT before the obesogenic challenge have not yet been investigated. Therefore, we hypothesised that the lack of OPN might enhance the pro-adipogenic micro environment before obesity driven inflammation. METHODS: OPN deficiency was tested in visceral (V) and subcutaneous (SC) WAT from WT and Spp1-/- female mice. Gene expression for hypoxia, inflammation and adipogenesis was checked in WT vs. Spp1-/- mice (n=15). Adipocytes progenitor cells (APC) were isolated by fluorescence cell sorting and role of OPN deficiency in adipogenesis was investigated by cell images and RT-PCR. RESULTS: We show that Spp1-/- maintained normal body and fat-pad weights, although hypoxia and inflammation markers were significantly reduced. In contrast, expression of genes involved in adipogenesis was increased in WAT from Spp1-/- mice. Strikingly, APC from Spp1-/- were diminished but differentiated more efficiently to adipocytes than those from control mice. CONCLUSIONS: APC from SC-WAT of lean OPN-deficient mice display an enhanced capacity for differentiating to adipocytes. These alterations may explain the healthy expansion of WAT in the OPN-deficient model which is associated with reduced inflammation and insulin resistance.


Subject(s)
Adipocytes/cytology , Adipogenesis , Adipose Tissue, White/cytology , Osteopontin/deficiency , Stem Cells/cytology , Adipocytes/metabolism , Adipose Tissue, White/metabolism , Animals , Diet, High-Fat , Disease Models, Animal , Female , Gene Expression , Inflammation/pathology , Inflammation/physiopathology , Mice , Osteopontin/genetics , Osteopontin/metabolism , Stem Cells/metabolism , Thinness/genetics , Thinness/metabolism
6.
Sci Rep ; 7(1): 14661, 2017 11 07.
Article in English | MEDLINE | ID: mdl-29116096

ABSTRACT

Aquaglyceroporins (AQPs) allow the movement of glycerol that is required for triglyceride formation in hepatic stellate cells (HSC), as key cellular source of fibrogenesis in the liver. The genetic polymorphism I148M of the patatin-like phospholipase domain-containing 3 (PNPLA3) is associated with hepatic steatosis and its progression to steatohepatitis (NASH), fibrosis and cancer. We aimed to explore the role of AQP3 for HSC activation and unveil its potential interactions with PNPLA3. HSC were isolated from human liver, experiments were performed in primary HSC and human HSC line LX2. AQP3 was the only aquaglyceroporin present in HSC and its expression decreased during activation. The PPARγ agonist, rosiglitazone, recovered AQP3 expression also in PNPLA3 I148M carrying HSC. When PNPLA3 was silenced, AQP3 expression increased. In liver sections from patients with NASH, the decreased amount of AQP3 was proportional to the severity of fibrosis and presence of the PNPLA3 I148M variant. In PNPLA3 I148M cells, the blockade of JNK pathway upregulated AQP3 in synergism with PPARγ. In conclusion, we demonstrated profound reduction of AQP3 in HSC carrying the PNPLA3 I148M variant in parallel to decreased PPARγ activation, which could be rescued by rosiglitazone and blockade of JNK.


Subject(s)
Aquaporin 3/metabolism , Hepatic Stellate Cells/metabolism , Lipase/metabolism , MAP Kinase Signaling System , Membrane Proteins/metabolism , PPAR gamma/metabolism , Amino Acid Substitution , Cell Line , Down-Regulation , Hepatic Stellate Cells/drug effects , Humans , Lipase/genetics , Lipogenesis , Membrane Proteins/genetics , PPAR gamma/antagonists & inhibitors , Rosiglitazone/pharmacology
7.
Biochem Biophys Res Commun ; 490(1): 51-54, 2017 08 12.
Article in English | MEDLINE | ID: mdl-28595905

ABSTRACT

Aquaporins (AQPs) are trans-membrane proteins which allow the movement of water and glycerol required by hepatic stellate cells (HSC) for triglyceride formation and lipid storage. Adiponectin (ADPQ) is a hormone produced by the adipose tissue, which is known to increase AQP3 expression. Since ADPQ receptor signals via the nuclear receptor PPAR we aimed to explore the role of this pathway in AQP3 regulation by ADPQ in HSC. AQP3 and CPT1α expression increased only after ADPQ but not rosiglitazone stimulation. In LX2 cells co-transfected with plasmids expressing PPARα or PPARγ coupled to a luciferase reporter gene, only PPARα increased luciferase activity after ADPQ stimulation. Collectively, our findings demonstrate that ADPQ increases AQP3 expression through PPARα-mediated signaling in HSC.


Subject(s)
Adiponectin/metabolism , Aquaporin 3/metabolism , Hepatic Stellate Cells/metabolism , PPAR gamma/metabolism , Cells, Cultured , Humans
8.
J Gastroenterol Hepatol ; 32(1): 253-260, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27083512

ABSTRACT

BACKGROUND AND AIM: Obesity is a major risk factor for liver fibrosis and tightly associated with low levels of adiponectin. Adiponectin has antifibrogenic activity protecting from liver fibrosis, which is mainly driven by activated hepatic stellate cells (HSC). Aquaporins are transmembrane proteins that allow the movement of water and, in case of aquaglyceroporins (AQPs), of glycerol that is needed in quiescent HSC for lipogenesis. Expression of various AQPs in liver is altered by obesity; however, the mechanisms through which obesity influences HSCs activation and AQPs expression remain unclear. This study aimed to identify obesity-associated factors that are related to HSC AQPs expression activation and lipid storage. METHODS: Correlations between serum adipokine levels and hepatic AQPs gene expression were analyzed from a cohort of obese patients. AQP and fibrotic gene expression was determined in a HSC line (LX2) and in a hepatocyte cell line (HepG2) after stimulation with adiponectin using quantitative real-time polymerase chain reaction. RESULTS: We found that serum adiponectin significantly correlated with liver AQP3, AQP7, AQP9 gene expressions. In vitro, adiponectin induced upregulation of AQP3 gene and AQP3 protein expression in human HSCs, but not in hepatocytes, while AQP7, AQP9 remained undetectable. Accordingly, HSC stimulated with adiponectin increased glycerol uptake, lipogenic gene expression, and lipid storage while downregulating activation/fibrosis markers. CONCLUSIONS: These findings demonstrate that adiponectin is a potent inhibitor of HSC activation and induces AQPs expression. Thus, low serum levels of adiponectin could be a mechanism how obesity affects the functional state of HSC, thereby contributing to obesity-associated liver fibrosis.


Subject(s)
Adiponectin/physiology , Aquaglyceroporins/genetics , Aquaglyceroporins/metabolism , Gene Expression Regulation/genetics , Gene Expression/genetics , Hepatic Stellate Cells/metabolism , Adiponectin/deficiency , Adult , Body Mass Index , Female , Hep G2 Cells , Humans , Lipogenesis/genetics , Liver Cirrhosis/etiology , Liver Cirrhosis/prevention & control , Male , Obesity/genetics , Obesity/metabolism
9.
Mol Metab ; 5(11): 1131-1137, 2016 11.
Article in English | MEDLINE | ID: mdl-27818939

ABSTRACT

OBJECTIVE: Recent findings point towards an important role of local macrophage proliferation also in obesity-induced adipose tissue inflammation that underlies insulin resistance and type 2 diabetes. Osteopontin (OPN) is an inflammatory cytokine highly upregulated in adipose tissue (AT) of obese and has repeatedly been shown to be functionally involved in adipose-tissue inflammation and metabolic sequelae. In the present work, we aimed at unveiling both the role of OPN in human monocyte and macrophage proliferation as well as the impact of OPN deficiency on local macrophage proliferation in a mouse model for diet-induced obesity. METHODS: The impact of recombinant OPN on viability, apoptosis, and proliferation was analyzed in human peripheral blood monocytes and derived macrophages. Wild type (WT) and OPN knockout mice (SPP1KO) were compared with respect to in vivo adipose tissue macrophage and in vitro bone marrow-derived macrophage (BMDM) proliferation. RESULTS: OPN not only enhanced survival and decreased apoptosis of human monocytes but also induced proliferation similar to macrophage colony stimulating factor (M-CSF). Even in fully differentiated monocyte-derived macrophages, OPN induced a proliferative response. Moreover, proliferation of adipose tissue macrophages in obese mice was detectable in WT but virtually absent in SPP1KO. In BMDM, OPN also induced proliferation while OPN as well as M-CSF-induced proliferation was similar in WT and SPP1KO. CONCLUSIONS: These data confirm that monocytes and macrophages not only are responsive to OPN and migrate to sites of inflammation but also they survive and proliferate more in the presence of OPN, a mechanism also strongly confirmed in vivo. Therefore, secreted OPN appears to be an essential player in AT inflammation, not only by driving monocyte chemotaxis and macrophage differentiation but also by facilitating local proliferation of macrophages.


Subject(s)
Adipose Tissue/cytology , Cell Proliferation , Macrophages , Obesity , Osteopontin/physiology , Animals , Diabetes Mellitus, Type 2 , Humans , Inflammation , Insulin Resistance , Mice , Mice, Knockout
10.
Immunol Lett ; 179: 85-94, 2016 11.
Article in English | MEDLINE | ID: mdl-27639826

ABSTRACT

Obesity causes insulin resistance via a chronic low-grade inflammation. This inflammation is characterized by elevated pro-inflammatory markers and macrophage accumulation in the adipose tissue (AT). AT inflammation is a key factor causing insulin resistance and thus type 2 diabetes, both linked to atherosclerotic cardiovascular disease. Osteopontin (OPN), a well-known inflammatory cytokine, is involved in obesity-linked complications including AT inflammation, insulin resistance, atherosclerosis and CVD. During inflammation, OPN is proteolytically cleaved by matrix metalloproteinases or thrombin leading to increased OPN activity. Therefore, OPN provides a new interesting target for immunological prevention and treatment of obesity-associated diseases. The aim of our study was to evaluate peptide-based vaccines against integrin binding sites of OPN and to examine whether these active immunotherapies are functional in reducing metabolic tissue inflammation, insulin resistance, and atherosclerosis in a cardio-metabolic (Ldlr-/- mice) and a diet-induced obesity model (WT mice). However, atherosclerosis, insulin resistance and AT inflammation were not diminished after treatment with OPN-derived peptides in murine models. Lack of efficacy was based on a failure to induce antibodies capable to bind epitopes in the context of functional OPN protein. In conclusion, our data point to unexpected challenges in the immunotherapeutic targeting of adhesive motives, such as RGD containing sequences, on endogenous proteins.


Subject(s)
Binding Sites/immunology , Heart Diseases/metabolism , Integrins/metabolism , Metabolic Diseases/metabolism , Osteopontin/immunology , Osteopontin/metabolism , Peptide Fragments/immunology , Animals , Antibodies/blood , Antibodies/immunology , Atherosclerosis/etiology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Biomarkers , Cross Reactions/immunology , Disease Models, Animal , Heart Diseases/blood , Heart Diseases/etiology , Heart Diseases/therapy , Immunization , Inflammation/etiology , Inflammation/metabolism , Insulin Resistance , Integrins/chemistry , Male , Metabolic Diseases/blood , Metabolic Diseases/etiology , Metabolic Diseases/therapy , Mice , Mice, Knockout , Obesity/metabolism , Osteopontin/chemistry , Peptide Fragments/administration & dosage , Protein Binding , Receptors, LDL/deficiency
11.
Transl Res ; 178: 63-73.e2, 2016 12.
Article in English | MEDLINE | ID: mdl-27490454

ABSTRACT

Osteopontin (OPN) is a multifunctional protein involved in several inflammatory processes and pathogeneses including obesity-related disorders and cancer. OPN binds to a variety of integrin receptors and CD44 resulting in a proinflammatory stimulus. Therefore, OPN constitutes a novel interesting target to develop new therapeutic strategies, which counteract OPN's proinflammatory properties. We established a humanized SPP1 (hSPP1) mouse model and evaluated its suitability as a model for obesity and insulin resistance. Unchallenged hSPP1 animals did not significantly differ in body weight and gross behavioral properties compared to wild-type (WT) animals. High-fat diet-challenged hSPP1 similarly developed obesity and inflammation, whereas insulin resistance was markedly changed. However, OPN expression profile in tissues was significantly altered in hSPP1 compared to WT depending on the diet. In conclusion, we developed a versatile humanized model to study the action of OPN in vivo and to develop strategies that target human OPN in a variety of pathologies.


Subject(s)
Obesity/immunology , Obesity/metabolism , Osteopontin/metabolism , Adipocytes/pathology , Adipose Tissue/pathology , Amino Acid Sequence , Animals , Blood Glucose/metabolism , Body Weight , Diet, High-Fat/adverse effects , Disease Models, Animal , Gene Expression Regulation , Genotype , Homologous Recombination/genetics , Humans , Hypertrophy , Inflammation/pathology , Insulin Resistance , Liver/pathology , Male , Mice, Inbred C57BL , Mice, Transgenic , Obesity/blood , Obesity/genetics , Osteopontin/chemistry , Osteopontin/genetics
12.
Obesity (Silver Spring) ; 24(7): 1489-98, 2016 07.
Article in English | MEDLINE | ID: mdl-27221527

ABSTRACT

OBJECTIVE: Macrophages are the main drivers of obesity-induced adipose tissue (AT) inflammation that causes insulin resistance. Macrophages polarize toward different inflammatory (M1) or protective (M2) phenotypes. Osteopontin (OPN) is an inflammatory cytokine highly expressed in AT in obesity and known to be involved in chronic inflammatory processes. It was hypothesized that OPN polarizes macrophages into a proinflammatory phenotype. METHODS: AT macrophages (ATMs) of OPN-deficient (Spp1(-/-) ) and wild-type C57BL/6 (WT) mice with obesity and bone marrow-derived macrophages (BMDMs) of Spp1(-/-) and WT mice as well as human monocyte-derived macrophages (MDMs) polarized in the presence of OPN were investigated. RESULTS: While ATM infiltration was lower in Spp1(-/-) upon high-fat diet, Spp1(-/-) ATMs expressed more M1 and less M2 markers but less tumor necrosis factor-α compared with WT. There was no effect of OPN deficiency on BMDM polarization. In human MDMs, the presence of OPN during polarization ambiguously altered M1/M2-related marker expression and diminished LPS-induced inflammatory cytokine production. Strikingly, phagocytic activity was elevated by the presence of OPN during polarization in both human MDMs and murine BMDMs. CONCLUSIONS: In contradiction to our hypothesis, data indicated that OPN does not induce inflammatory macrophages but was a signal to induce phagocytosis, which may be required due to increased adipocyte death in obesity.


Subject(s)
Macrophages/physiology , Obesity/physiopathology , Osteopontin/metabolism , Adipocytes/physiology , Adipose Tissue/metabolism , Animals , Diet, High-Fat , Inflammation/physiopathology , Insulin Resistance , Male , Mice , Mice, Inbred C57BL , Phagocytosis/physiology , Phenotype , Tumor Necrosis Factor-alpha/metabolism
13.
Biochem Pharmacol ; 81(2): 279-88, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-20959116

ABSTRACT

The diabetic phenotype caused by the deletion of insulin receptor substrate-2 (Irs-2) in mice displays a sexual dimorphism. Whereas the majority of male Irs-2(-/-) mice are overtly diabetic by 12 weeks of age, female Irs-2(-/-) animals develop mild obesity and progress less rapidly to diabetes. Here we investigated ß-cell function and lipolysis as potential explanations for the gender-related differences in this model. Glucose-stimulated insulin secretion was enhanced in islets from male null mice as compared to male WT whereas this response in female Irs-2(-/-) islets was identical to that of female controls. The ability of α(2)-adrenoceptor (α(2)-AR) agonists to inhibit insulin secretion was attenuated in male Irs2 null mice. Consistent with this, the expression of the α(2A)-AR was reduced in male Irs-2(-/-) islets. The response of male Irs-2(-/-) islets to forskolin was enhanced, owing to increased production of cAMP. Basal lipolysis was increased in male Irs-2(-/-) but decreased in female Irs-2(-/-) mice, concordant with the observation that adipose tissue is sparse in males whereas female Irs2 null mice are mildly obese. Adipocytes from both male and female Irs-2(-/-) were resistant to the anti-lipolytic effects of insulin but female Irs-2(-/-) fat cells were additionally resistant to the catabolic effects of beta-adrenergic agonists. This catecholamine resistance was associated with impaired generation of cAMP. Consequently, targets of cAMP-dependent protein kinase (PKA) which mediate lipolysis were not phosphorylated in adipose tissue of female Irs-2(-/-) mice. Our findings suggest that IRS-2 deficiency in mice alters the expression and/or sensitivity of components of adrenergic signaling.


Subject(s)
Adrenergic alpha-2 Receptor Agonists/pharmacology , Diabetes Mellitus/genetics , Diabetes Mellitus/metabolism , Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/metabolism , Sex Characteristics , Adenylyl Cyclases , Adipocytes/metabolism , Animals , Cells, Cultured , Colforsin/pharmacology , Cyclic AMP/metabolism , Female , Gene Expression Regulation/physiology , Insulin/metabolism , Insulin/pharmacology , Islets of Langerhans/physiology , Lipolysis/physiology , Male , Mice , Mice, Knockout , Receptors, Adrenergic, alpha-2/metabolism , Signal Transduction , Sterol Esterase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...