Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
Cell Metab ; 36(5): 1030-1043.e7, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38670107

ABSTRACT

The mechanisms of hepatic stellate cell (HSC) activation and the development of liver fibrosis are not fully understood. Here, we show that deletion of a nuclear seven transmembrane protein, TM7SF3, accelerates HSC activation in liver organoids, primary human HSCs, and in vivo in metabolic-dysfunction-associated steatohepatitis (MASH) mice, leading to activation of the fibrogenic program and HSC proliferation. Thus, TM7SF3 knockdown promotes alternative splicing of the Hippo pathway transcription factor, TEAD1, by inhibiting the splicing factor heterogeneous nuclear ribonucleoprotein U (hnRNPU). This results in the exclusion of the inhibitory exon 5, generating a more active form of TEAD1 and triggering HSC activation. Furthermore, inhibiting TEAD1 alternative splicing with a specific antisense oligomer (ASO) deactivates HSCs in vitro and reduces MASH diet-induced liver fibrosis. In conclusion, by inhibiting TEAD1 alternative splicing, TM7SF3 plays a pivotal role in mitigating HSC activation and the progression of MASH-related fibrosis.


Subject(s)
DNA-Binding Proteins , Liver Cirrhosis , TEA Domain Transcription Factors , Transcription Factors , TEA Domain Transcription Factors/metabolism , Animals , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Liver Cirrhosis/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Humans , Mice , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Alternative Splicing , Mice, Inbred C57BL , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Hepatic Stellate Cells/metabolism , Male , Fatty Liver/metabolism , Fatty Liver/pathology , Fatty Liver/genetics , Mice, Knockout
2.
iScience ; 25(11): 105270, 2022 Nov 18.
Article in English | MEDLINE | ID: mdl-36304109

ABSTRACT

The seven-transmembrane superfamily member 3 protein (TM7SF3) is a p53-regulated homeostatic factor that attenuates cellular stress and the unfolded protein response. Here we show that TM7SF3 localizes to nuclear speckles; eukaryotic nuclear bodies enriched in splicing factors. This unexpected location for a trans -membranal protein enables formation of stable complexes between TM7SF3 and pre-mRNA splicing factors including DHX15, LARP7, HNRNPU, RBM14, and HNRNPK. Indeed, TM7SF3 regulates alternative splicing of >330 genes, mainly at the 3'end of introns by directly modulating the activity of splicing factors such as HNRNPK. These effects are observed both in cell lines and primary human pancreatic islets. Accordingly, silencing of TM7SF3 results in differential expression of 1465 genes (about 7% of the human genome); with 844 and 621 genes being up- or down-regulated, respectively. Our findings implicate TM7SF3, as a resident protein of nuclear speckles and suggest a role for seven-transmembrane proteins as regulators of alternative splicing.

3.
Biochem Soc Trans ; 50(1): 135-149, 2022 02 28.
Article in English | MEDLINE | ID: mdl-35015084

ABSTRACT

Galectin-8 (Gal-8) belongs to a family of animal lectins that modulate cell adhesion, cell proliferation, apoptosis, and immune responses. Recent studies have shown that mammalian Gal-8 induces in an autocrine and paracrine manner, the expression and secretion of cytokines and chemokines such as RANKL, IL-6, IL-1ß, SDF-1, and MCP-1. This involves Gal-8 binding to receptor complexes that include MRC2/uPAR/LRP1, integrins, and CD44. Receptors ligation triggers FAK, ERK, Akt, and the JNK signaling pathways, leading to induction of NF-κB that promotes cytokine expression. Indeed, immune-competent Gal-8 knockout (KO) mice express systemic lower levels of cytokines and chemokines while the opposite is true for Gal-8 transgenic animals. Cytokine and chemokine secretion, induced by Gal-8, promotes the migration of cancer cells toward cells expressing this lectin. Accordingly, Gal-8 KO mice experience reduced tumor size and smaller and fewer metastatic lesions when injected with cancer cells. These observations suggest the existence of a 'vicious cycle' whereby Gal-8 expression and secretion promotes the secretion of cytokines and chemokines that further promote Gal-8 expression. This 'vicious cycle' could enhance the development of a 'cytokine storm' which is a key contributor to the poor prognosis of COVID-19 patients.


Subject(s)
Cytokines/immunology , Galectins/immunology , Immunity , Animals , COVID-19/immunology , Humans , Signal Transduction
4.
J Cell Sci ; 134(8)2021 04 15.
Article in English | MEDLINE | ID: mdl-33722978

ABSTRACT

The mechanisms underlying the cellular response to extracellular matrices (ECMs) that consist of multiple adhesive ligands are still poorly understood. Here, we address this topic by monitoring specific cellular responses to two different extracellular adhesion molecules - the main integrin ligand fibronectin and galectin-8, a lectin that binds ß-galactoside residues  - as well as to mixtures of the two proteins. Compared with cell spreading on fibronectin, cell spreading on galectin-8-coated substrates resulted in increased projected cell area, more-pronounced extension of filopodia and, yet, the inability to form focal adhesions and stress fibers. These differences can be partially reversed by experimental manipulations of small G-proteins of the Rho family and their downstream targets, such as formins, the Arp2/3 complex and Rho kinase. We also show that the physical adhesion of cells to galectin-8 was stronger than adhesion to fibronectin. Notably, galectin-8 and fibronectin differently regulate cell spreading and focal adhesion formation, yet act synergistically to upregulate the number and length of filopodia. The physiological significance of the coherent cellular response to a molecularly complex matrix is discussed. This article has an associated First Person interview with the first author of the paper.


Subject(s)
Adhesives , Fibronectins , Cell Adhesion , Galectins , Pseudopodia
5.
J Med Chem ; 63(20): 11573-11584, 2020 10 22.
Article in English | MEDLINE | ID: mdl-32809817

ABSTRACT

Galectin-8 is a ß-galactoside-recognizing protein having an important role in the regulation of bone remodeling and cancer progression and metastasis. Methyl ß-d-galactopyranoside malonyl aromatic esters have been designed to target and engage with particular amino acid residues of the galectin-8N extended carbohydrate-binding site. The chemically synthesized compounds had in vitro binding affinity toward galectin-8N in the range of 5-33 µM, as evaluated by isothermal titration calorimetry. This affinity directly correlated with the compounds' ability to inhibit galectin-8-induced expression of chemokines and proinflammatory cytokines in the SUM159 breast cancer cell line. X-ray crystallographic structure determination revealed that these monosaccharide-based compounds bind galectin-8N by engaging its unique arginine (Arg59) and simultaneously cross-linking to another arginine (Arg45) located across the carbohydrate-binding site. This structure-based drug design approach has led to the discovery of novel monosaccharide galactose-based antagonists, with the strongest-binding compound (Kd 5.72 µM) holding 7-fold tighter than the disaccharide lactose.


Subject(s)
Drug Design , Galactosides/chemical synthesis , Galectins/antagonists & inhibitors , Binding Sites , Cell Line, Tumor , Computer Simulation , Cytokines/genetics , Female , Galactosides/chemistry , Galactosides/pharmacology , Humans , Molecular Docking Simulation , Molecular Structure , Protein Binding , Thermodynamics
6.
Sci Rep ; 10(1): 7375, 2020 04 30.
Article in English | MEDLINE | ID: mdl-32355198

ABSTRACT

Secreted animal lectins of the galectin family are key players in cancer growth and metastasis. Here we show that galectin-8 (gal-8) induces the expression and secretion of cytokines and chemokines such as SDF-1 and MCP-1 in a number of cell types. This involves gal-8 binding to a uPAR/LRP1/integrin complex that activates JNK and the NFkB pathway. Cytokine and chemokine secretion, induced by gal-8, promotes migration of cancer cells toward cells treated with this lectin. Indeed, immune-competent gal-8 knockout (KO) mice express systemic lower levels of cytokines and chemokines while the opposite is true for gal-8 transgenic animals. Accordingly, gal-8 KO mice experience reduced tumor size and smaller and fewer metastatic lesions when injected with cancer cells. These results suggest the existence of a 'vicious cycle' whereby gal-8 secreted by the tumor microenvironment, promotes secretion of chemoattractants at the metastatic niche that promote further recruitment of tumor cells to that site. This study further implicate gal-8 in control of cancer progression and metastasis through its effects on the production of immunoregulatory cytokines.


Subject(s)
Cell Movement , Chemokine CXCL12/metabolism , Galectins/metabolism , Neoplasm Proteins/metabolism , Neoplasms, Experimental/metabolism , Animals , Chemokine CCL2/genetics , Chemokine CXCL12/genetics , Galectins/genetics , Mice , Mice, Knockout , Neoplasm Metastasis , Neoplasm Proteins/genetics , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology
8.
ChemMedChem ; 13(16): 1664-1672, 2018 08 20.
Article in English | MEDLINE | ID: mdl-29926535

ABSTRACT

Galectin-8 is a ß-galactoside-recognising protein that has a role in the regulation of bone remodelling and is an emerging new target for tackling diseases with associated bone loss. We have designed and synthesised methyl 3-O-[1-carboxyethyl]-ß-d-galactopyranoside (compound 6) as a ligand to target the N-terminal domain of galectin-8 (galectin-8N). Our design involved molecular dynamics (MD) simulations that predicted 6 to mimic the interactions made by the galactose ring as well as the carboxylic acid group of 3'-O-sialylated lactose (3'-SiaLac), with galectin-8N. Isothermal titration calorimetry (ITC) determined that the binding affinity of galectin-8N for 6 was 32.8 µm, whereas no significant affinity was detected for the C-terminal domain of galectin-8 (galectin-8C). The crystal structure of the galectin-8N-6 complex validated the predicted binding conformation and revealed the exact protein-ligand interactions that involve evolutionarily conserved amino acids of galectin and also those unique to galectin-8N for recognition. Overall, we have initiated and demonstrated a rational ligand design campaign to develop a monosaccharide-based scaffold as a binder of galectin-8.


Subject(s)
Galactosides/metabolism , Galectins/metabolism , Arginine/chemistry , Binding Sites , Calorimetry , Crystallography, X-Ray , Galactosides/chemical synthesis , Galactosides/chemistry , Galectins/chemistry , Humans , Ligands , Molecular Dynamics Simulation , Protein Binding , Protein Domains
13.
FASEB J ; 32(5): 2366-2380, 2018 05.
Article in English | MEDLINE | ID: mdl-29259034

ABSTRACT

Mice overexpressing galectin-8 [gal-8 transgenic (Tg)], a secreted mammalian lectin, exhibit enhanced bone turnover and reduced bone mass, similar to cases of postmenopausal osteoporosis. Here, we show that gal-8 knockout (KO) mice have increased bone mass accrual at a young age but exhibit accelerated bone loss during adulthood. These phenotypes can be attributed to a gal-8-mediated increase in receptor activator of NF-κB ligand (RANKL) expression that promotes osteoclastogenesis, combined with direct inhibition of osteoblast differentiation, evident by reduced bone morphogenetic protein (BMP) signaling, reduced phosphorylation of receptor regulated mothers against decapentaplegic homolog (R-SMAD) and reduced expression of osteoblast differentiation markers osterix, osteocalcin, runt-related transcription factor 2 (RUNX2), dentin matrix acidic phosphoprotein-1 (DMP1), and alkaline phosphatase. At the same time, gal-8 promotes expression of estrogen receptor α (ESR1). Accordingly, the rate of bone loss is accelerated in ovariectomized, estrogen-deficient gal-8 Tg mice, whereas gal-8 KO mice, having low levels of ESR1, are refractory to ovariectomy. Finally, gal-8 mRNA positively correlates with the mRNA levels of osteoclastogenic markers RANKL, tartrate-resistant acid phosphatase, and cathepsin K in human femurs. Collectively, these findings identify gal-8 as a new physiologic player in the regulation of bone mass.-Vinik, Y., Shatz-Azoulay, H., Hiram-Bab, S., Kandel, L., Gabet, Y., Rivkin, G., Zick, Y. Ablation of the mammalian lectin galectin-8 induces bone defects in mice.


Subject(s)
Femur/metabolism , Galectins/metabolism , Osteoporosis/metabolism , Animals , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Femur/pathology , Galectins/genetics , Humans , Mice , Mice, Knockout , Osteoporosis/genetics , Osteoporosis/pathology , RANK Ligand/genetics , RANK Ligand/metabolism
14.
Diabetes ; 66(7): 1879-1889, 2017 07.
Article in English | MEDLINE | ID: mdl-28424159

ABSTRACT

Cellular stress and proinflammatory cytokines induce phosphorylation of insulin receptor substrate (IRS) proteins at Ser sites that inhibit insulin and IGF-I signaling. We therefore examined the effects of mutation of five "inhibitory" Ser phosphorylation sites on IRS2 function in transgenic mice that overexpress, selectively in pancreatic ß-cells, either wild-type (WT) or a mutated IRS2 protein (IRS25A). Islets size, number, and mRNA levels of catalase and superoxide dismutase were increased, whereas those of nitric oxide synthase were decreased, in 7- to 10-week-old IRS25A-ß mice compared with IRS2WT-ß mice. However, glucose homeostasis and insulin secretion in IRS25A-ß mice were impaired when compared with IRS2WT-ß mice or to nontransgenic mice. This was associated with reduced mRNA levels of Glut2 and islet ß-cell transcription factors such as Nkx6.1 and MafA Similarly, components mediating the unfolded protein response were decreased in islets of IRS25A-ß mice in accordance with their decreased insulin secretion. The beneficial effects of IRS25A on ß-cell proliferation and ß-cell transcription factors were evident only in 5- to 8-day-old mice. These findings suggest that elimination of inhibitory Ser phosphorylation sites of IRS2 exerts short-term beneficial effects in vivo; however, their sustained elimination leads to impaired ß-cell function.


Subject(s)
Feedback, Physiological , Insulin Receptor Substrate Proteins/genetics , Insulin/metabolism , RNA, Messenger/metabolism , Animals , Blood Glucose/metabolism , Catalase/genetics , Catalase/metabolism , Cell Proliferation/genetics , Glucose Transporter Type 2/genetics , Glucose Transporter Type 2/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Insulin-Secreting Cells , Islets of Langerhans/pathology , Maf Transcription Factors, Large/genetics , Maf Transcription Factors, Large/metabolism , Mice , Mice, Transgenic , Mutation , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Organ Size , Phosphorylation , Signal Transduction , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism
15.
Cell Death Differ ; 24(1): 132-143, 2017 01.
Article in English | MEDLINE | ID: mdl-27740623

ABSTRACT

Earlier reported small interfering RNA (siRNA) high-throughput screens, identified seven-transmembrane superfamily member 3 (TM7SF3) as a novel inhibitor of pancreatic ß-cell death. Here we show that TM7SF3 maintains protein homeostasis and promotes cell survival through attenuation of ER stress. Overexpression of TM7SF3 inhibits caspase 3/7 activation. In contrast, siRNA-mediated silencing of TM7SF3 accelerates ER stress and activation of the unfolded protein response (UPR). This involves inhibitory phosphorylation of eukaryotic translation initiation factor 2α activity and increased expression of activating transcription factor-3 (ATF3), ATF4 and C/EBP homologous protein, followed by induction of apoptosis. This process is observed both in human pancreatic islets and in a number of cell lines. Some of the effects of TM7SF3 silencing are evident both under basal conditions, in otherwise untreated cells, as well as under different stress conditions induced by thapsigargin, tunicamycin or a mixture of pro-inflammatory cytokines (tumor necrosis factor alpha, interleukin-1 beta and interferon gamma). Notably, TM7SF3 is a downstream target of p53: activation of p53 by Nutlin increases TM7SF3 expression in a time-dependent manner, although silencing of p53 abrogates this effect. Furthermore, p53 is found in physical association with the TM7SF3 promoter. Interestingly, silencing of TM7SF3 promotes p53 activity, suggesting the existence of a negative-feedback loop, whereby p53 promotes expression of TM7SF3 that acts to restrict p53 activity. Our findings implicate TM7SF3 as a novel p53-regulated pro-survival homeostatic factor that attenuates the development of cellular stress and the subsequent induction of the UPR.


Subject(s)
Membrane Glycoproteins/metabolism , Tumor Suppressor Protein p53/metabolism , Activating Transcription Factor 3/metabolism , Activating Transcription Factor 4/metabolism , Animals , Apoptosis/drug effects , CCAAT-Enhancer-Binding Proteins/metabolism , Caspase 3/metabolism , Caspase 7/metabolism , Cell Line , Endoplasmic Reticulum Stress/drug effects , HEK293 Cells , Hep G2 Cells , Humans , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/genetics , Mice , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Promoter Regions, Genetic , Protein Binding , Thapsigargin/toxicity , Transcription Factor CHOP/metabolism , Tunicamycin/toxicity , Unfolded Protein Response/drug effects , eIF-2 Kinase/antagonists & inhibitors , eIF-2 Kinase/metabolism
16.
Sci Rep ; 6: 39556, 2016 12 21.
Article in English | MEDLINE | ID: mdl-28000747

ABSTRACT

Glycosphingolipids are ubiquitous cell surface molecules undertaking fundamental cellular processes. Lacto-N-tetraose (LNT) and lacto-N-neotetraose (LNnT) are the representative core structures for lacto- and neolacto-series glycosphingolipids. These glycolipids are the carriers to the blood group antigen and human natural killer antigens mainly found on blood cells, and are also principal components in human milk, contributing to infant health. The ß-galactoside recognising galectins mediate various cellular functions of these glycosphingolipids. We report crystallographic structures of the galectin-8 N-terminal domain (galectin-8N) in complex with LNT and LNnT. We reveal the first example in which the non-reducing end of LNT binds to the primary binding site of a galectin, and provide a structure-based rationale for the significant ten-fold difference in binding affinities of galectin-8N toward LNT compared to LNnT, such a magnitude of difference not being observed for any other galectin. In addition, the LNnT complex showed that the unique Arg59 has ability to adopt a new orientation, and comparison of glycerol- and lactose-bound galectin-8N structures reveals a minimum atomic framework for ligand recognition. Overall, these results enhance our understanding of glycosphingolipids interactions with galectin-8N, and highlight a structure-based rationale for its significantly different affinity for components of biologically relevant glycosphingolipids.


Subject(s)
Galectins/chemistry , Glycosphingolipids/chemistry , Arginine/chemistry , Binding Sites , Crystallography, X-Ray , Humans , Lactose/chemistry , Molecular Dynamics Simulation , Oligosaccharides/chemistry , Protein Binding , Protein Conformation , Protein Domains , Software
17.
Biochem Biophys Res Commun ; 465(4): 851-6, 2015 Oct 02.
Article in English | MEDLINE | ID: mdl-26319551

ABSTRACT

High-throughput siRNA screening was employed to identify novel genes that regulate cytokine-induced death of pancreatic ß-cells. One of the 'hits' was Nedd4 family interacting protein 1 (Ndfip1), an adaptor and activator of Nedd4-family ubiquitin ligases. Silencing of Ndfip1 inhibited cytokine-induced apoptosis of mouse and human pancreatic islets and promoted glucose-stimulated insulin secretion. These effects were associated with an increase in the cellular content of JunB, a potent inhibitor of ER stress and apoptosis. Silencing of Ndfip1 also increased the expression of ATF4, IRE-1α, and the spliced form of XBP that govern the unfolded protein response (UPR) and relieve cytokine-induced ER stress, while overexpression of Ndfip1 exerted opposite effects. These findings implicate Ndfip1 in the degradation of JunB; inhibition of the UPR and insulin secretion; and promotion of cytokine-induced death of pancreatic ß-cells.


Subject(s)
Apoptosis/physiology , Carrier Proteins/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Membrane Proteins/metabolism , Animals , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Cell Line , Cells, Cultured , Cytokines/metabolism , Endoplasmic Reticulum Stress , High-Throughput Screening Assays , Humans , Insulin/metabolism , Insulin Secretion , Intercellular Signaling Peptides and Proteins , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Proteolysis , RNA, Small Interfering/genetics , Transcription Factors/metabolism , Unfolded Protein Response
18.
Elife ; 4: e05914, 2015 May 08.
Article in English | MEDLINE | ID: mdl-25955862

ABSTRACT

Skeletal integrity is maintained by the co-ordinated activity of osteoblasts, the bone-forming cells, and osteoclasts, the bone-resorbing cells. In this study, we show that mice overexpressing galectin-8, a secreted mammalian lectin of the galectins family, exhibit accelerated osteoclasts activity and bone turnover, which culminates in reduced bone mass, similar to cases of postmenopausal osteoporosis and cancerous osteolysis. This phenotype can be attributed to a direct action of galectin-8 on primary cultures of osteoblasts that secrete the osteoclastogenic factor RANKL upon binding of galectin-8. This results in enhanced differentiation into osteoclasts of the bone marrow cells co-cultured with galectin-8-treated osteoblasts. Secretion of RANKL by galectin-8-treated osteoblasts can be attributed to binding of galectin-8 to receptor complexes that positively (uPAR and MRC2) and negatively (LRP1) regulate galectin-8 function. Our findings identify galectins as new players in osteoclastogenesis and bone remodeling, and highlight a potential regulation of bone mass by animal lectins.


Subject(s)
Bone Marrow Cells/metabolism , Bone Resorption/genetics , Bone and Bones/metabolism , Galectins/genetics , Osteoblasts/metabolism , Osteoclasts/metabolism , RANK Ligand/genetics , Animals , Bone Density/genetics , Bone Marrow Cells/cytology , Bone Resorption/metabolism , Bone Resorption/pathology , Bone and Bones/cytology , Coculture Techniques , Female , Galectins/metabolism , Gene Expression Regulation , Low Density Lipoprotein Receptor-Related Protein-1 , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Transgenic , Osteoblasts/cytology , Osteoclasts/cytology , Osteogenesis/genetics , Primary Cell Culture , Protein Binding , RANK Ligand/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, LDL/genetics , Receptors, LDL/metabolism , Receptors, Urokinase Plasminogen Activator/genetics , Receptors, Urokinase Plasminogen Activator/metabolism , Signal Transduction , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
19.
J Biol Chem ; 288(8): 5682-93, 2013 Feb 22.
Article in English | MEDLINE | ID: mdl-23275337

ABSTRACT

Selective serotonin reuptake inhibitors (SSRIs) are antidepressants used for the treatment of mood and anxiety disorders. Here, we demonstrate that incubation (2 h) of murine islets or Min6 ß cell line with the SSRIs paroxetine, fluoxetine, or sertraline inhibited insulin-induced Tyr phosphorylation of insulin receptor substrate (IRS)-2 protein and the activation of its downstream targets Akt and the ribosomal protein S6 kinase-1 (S6K1). Inhibition was dose-dependent with half-maximal effects at ∼15-20 µM. It correlated with a rapid dephosphorylation and activation of the IRS kinase GSK3ß. Introduction of GSK3ß siRNAs eliminated the inhibitory effects of the SSRIs. Inhibition of IRS-2 action by 30 µM SSRI was associated with a marked inhibition of glucose-stimulated insulin secretion from murine and human pancreatic islets. Secretion induced by basic secretagogues (KCl and Arg) was not affected by these drugs. Prolonged treatment (16 h) of Min6 cells with sertraline resulted in the induction of inducible nitric oxide synthase; activation of endoplasmic reticulum stress, and the initiation of the unfolded protein response, manifested by enhanced transcription of ATF4 and C/EBP homologous protein. This triggered an apoptotic process, manifested by enhanced caspase 3/7 activity, which resulted in ß cell death. These findings implicate SSRIs as inhibitors of IRS protein function and insulin action through the activation of GSK3ß. They further suggest that SSRIs inhibit insulin secretion; induce the unfolded protein response; activate an apoptotic process, and trigger ß cell death. Given that SSRIs promote insulin resistance while inhibiting insulin secretion, these drugs might accelerate the transition from an insulin-resistant state to overt diabetes.


Subject(s)
Insulin-Secreting Cells/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology , Animals , Apoptosis , Cell Death , Cell Line , Diabetes Mellitus/chemically induced , Diabetes Mellitus/metabolism , Fluoxetine/pharmacology , Humans , Insulin/metabolism , Insulin Receptor Substrate Proteins/metabolism , Insulin Resistance , Insulin-Secreting Cells/cytology , Male , Mice , Mice, Inbred C57BL , Paroxetine/pharmacology , Sertraline/pharmacology , Signal Transduction , Unfolded Protein Response
20.
Nanotoxicology ; 6: 804-12, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22007682

ABSTRACT

Upon exposure, TiO(2) nanoparticles (NPs) have been recovered in internal organs such as the liver, and are proposed to cause cellular/organ dysfunction, particularly in the liver and lungs. We hypothesized that despite being considered "inert" as bulk material, TiO(2) NPs may impair insulin responses in liver-derived cells, either indirectly by inflammatory activation of macrophages, and/or by directly interfering with insulin signaling. Using qRT-PCR and conditioned medium (CM) approaches, we show that exposure to TiO(2) NPs activates macrophages' expression of TNF-α, IL-6, IL-8, IL-1α and IL-1ß and the resulting CM induces insulin resistance in Fao cells. Furthermore, direct exposure of Fao cells to TiO(2) results in activation of the stress kinases JNK and p38MAP kinase, and in induction of insulin resistance at the signaling and metabolic levels. Collectively, our findings provide a proof-of-concept for the ability of man-made NPs to induce insulin resistance in liver-derived cells, an endocrine abnormality underlying some of the most common human diseases.


Subject(s)
Insulin Resistance , Liver/drug effects , Macrophage Activation , Metal Nanoparticles/toxicity , Titanium/toxicity , Animals , Blotting, Western , Cell Line, Tumor , Culture Media, Conditioned , Glycogen/biosynthesis , Insulin/metabolism , Rats , Real-Time Polymerase Chain Reaction , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...