Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
Add more filters

Publication year range
1.
J Surg Oncol ; 125(6): 1042-1052, 2022 May.
Article in English | MEDLINE | ID: mdl-35099823

ABSTRACT

BACKGROUND: Stimulator of interferon (IFN) genes (STING) is a protein that promotes type I IFN production essential for T-cell activation. In this study, we aim to characterize STING expression comprehensively using The Cancer Genome Atlas (TCGA) database, cell lines, and patient tumor samples stained with immunohistochemistry. METHODS: Two cohorts were evaluated comprising 721 non-small cell lung cancer (NSCLC) patients and 55 NSCLC cell lines for STING and cyclic GMP-AMP synthase (cGAS) expression using immunohistochemistry. Moreover, an independent cohort of n = 499 patients from the TCGA database was analyzed. Methylation was evaluated on STING and cGAS in five STING-negative NSCLC cell lines. RESULTS: STING RNA expression positively correlates with T cell function and development genes, negatively correlates with cell proliferation and associated with increased survival (5-year-overall survival [OS] 47.3% vs. 38.8%, p = 0.033). STING protein expression is significantly higher in adenocarcinoma (AC) and is lost with increasing stages of AC. STING-positivity is significantly higher in mutant EGFR and KRAS tumors. STING-positive NSCLC patients identified with immunohistochemistry (H-score > 50) have increased survival (median OS: 58 vs. 35 months, p = 0.02). Treatment of STING-negative cell lines with a demethylating agent restores STING expression. CONCLUSIONS: STING is ubiquitously expressed in NSCLC and associated with T cell function genes, AC histology, EGFR, and KRAS mutations and improved overall survival.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Membrane Proteins/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , ErbB Receptors/metabolism , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Membrane Proteins/genetics , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Prognosis , Proto-Oncogene Proteins p21(ras)
2.
Transpl Int ; 31(10): 1164-1177, 2018 10.
Article in English | MEDLINE | ID: mdl-29722117

ABSTRACT

We have previously reported that co-transplantation of the kidney with vascularized donor thymus from α-1,3-galactosyltransferase gene knockout pigs with an anti-CD154 with rituximab-based regimen led to improved xenograft survival in baboons with donor-specific unresponsiveness. However, nephrotic syndrome emerged as a complication in which the glomeruli showed mild mesangial expansion with similarities to minimal change disease (MCD) in humans. Since MCD is associated with CD80 expression in glomeruli and elevated urinary excretion, we evaluated a potential role for CD80 in xenograft nephropathy. Study 1 confirmed high urinary CD80 excretion in nephrotic animals with renal xenografts showing CD80 expression in glomeruli. In Study 2, baboons receiving xenografts received CTLA4-Ig once a week from the second postoperative week or no CTLA4-Ig. The non-CTLA4-Ig group developed severe proteinuria with modest mesangial expansion with high urinary excretion of CD80 and documented CD80 expression in glomerular podocytes. All of the recipients in non-CTLA4-Ig groups had to be euthanized before POD 60. In contrast, CTLA4-Ig group showed a marked reduction in proteinuria and survived significantly longer, up to 193 days. These results demonstrate that anti-CD80 targeted therapy represents a promising strategy for reduction of proteinuria following renal xeno-transplantation with improved survival.


Subject(s)
B7-1 Antigen/metabolism , Gene Expression Regulation , Kidney Glomerulus/immunology , Kidney Transplantation , Podocytes/immunology , Proteinuria/immunology , Abatacept/immunology , Animals , Animals, Genetically Modified , CD40 Ligand/immunology , CTLA-4 Antigen/immunology , Galactosyltransferases/genetics , Immunoglobulin G/immunology , Kidney/metabolism , Kidney Diseases/immunology , Kidney Diseases/surgery , Nephrosis , Nephrosis, Lipoid , Papio , Swine , Transplantation, Heterologous , Urinalysis
3.
Med Sci Monit ; 23: 1208-1216, 2017 Mar 09.
Article in English | MEDLINE | ID: mdl-28275222

ABSTRACT

BACKGROUND Immunotherapy targeting the programmed death-1 (PD-1)/programmed death ligand-1 (PD-L1) checkpoint has shown the good outcomes in non-small cell lung cancer (NSCLC). We investigated PD-1 and PD-L1 protein expression and their correlation with tumor-infiltrating lymphocytes (TILs), and association with survival in NSCLC. MATERIAL AND METHODS The expression of PD-1 (NAT105, Cell Marque) and PD-L1 (28-8, Dako) protein was assessed in 55 NSCLC cell lines by immunohistochemistry (IHC). PD-1 (NAT105, Cell Marque) and PD-L1 (22C3, Dako) protein expression was evaluated by IHC, and TIL percentage was scored, in 139 surgically resected specimens from patients with NSCLC. RESULTS PD-1 was not expressed on NSCLC cell lines. PD-L1 was expressed on 20 NSCLC cell lines (36.4%). A total of 60 patient samples (43.2%) were positive for PD-1 on the TILs, and 25 (18.0%) were positive for PD-L1 on tumor cells. High expression of PD-1 on tumor cells was significantly correlated with higher expression of PD-L1 (P=0.026) and a higher percentage of TILs (P<0.001). In the Cox regression model, the odds ratio for PD-1 was 2.828 (95% CI: 1.325-11.165; P=0.013) and 8.579 (95% CI: 4.148-22.676; P<0.001) when PD-L1 and TILs were positive. Patients whose tumor cells were PD-L1 negative had a tendency for longer relapse-free survival (RFS) than patients who were PD-L1 positive (1.85 years, 95% CI: 0.77-2.93 vs. 0.97 years, 95% CI: 0.71-1.23; P=0.054). CONCLUSIONS PD-1 was expressed on TILs in tumor tissues in NSCLC patients. PD-L1 was expressed on both TILs and tumor tissues. PD-1 expression was correlated with PD-L1 on tumor cells and TILs. Patients who were PD-L1 positive tended to experience progression after surgery.


Subject(s)
B7-H1 Antigen/metabolism , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Programmed Cell Death 1 Receptor/metabolism , Aged , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Disease-Free Survival , Female , Humans , Immunohistochemistry , Logistic Models , Lung Neoplasms/pathology , Male , Middle Aged , Multivariate Analysis
4.
Cancer Sci ; 107(9): 1193-7, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27297395

ABSTRACT

Immunotherapy has recently become widely used in lung cancer. Many oncologists are focused on cytotoxic T lymphocyte antigen-4 (CTLA-4), programmed cell death ligand-1 (PD-L1) and programmed cell death-1 (PD-1). Immunotherapy targeting the PD-1/PD-L1 checkpoints has shown promising efficacy in non-small cell lung cancer (NSCLC), but questions remain to be answered. Among them is whether the simultaneous inhibition of other checkpoints could improve outcomes. Lymphocyte-activation gene-3 (LAG-3) is another vital checkpoint that may have a synergistic interaction with PD-1/PD-L1. Here we review the LAG-3 function in cancer, clinical trials with agents targeting LAG-3 and the correlation of LAG-3 with other checkpoints.


Subject(s)
Antigens, CD/metabolism , Immunomodulation , Neoplasms/immunology , Neoplasms/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antigens, CD/genetics , B7-H1 Antigen/metabolism , CTLA-4 Antigen/metabolism , Disease Susceptibility , Drug Discovery , Gene Expression , Humans , Immunomodulation/genetics , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/genetics , Programmed Cell Death 1 Receptor/metabolism , Signal Transduction/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Lymphocyte Activation Gene 3 Protein
5.
FASEB J ; 28(8): 3339-50, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24755741

ABSTRACT

Reduced AMP kinase (AMPK) activity has been shown to play a key deleterious role in increased hepatic gluconeogenesis in diabetes, but the mechanism whereby this occurs remains unclear. In this article, we document that another AMP-dependent enzyme, AMP deaminase (AMPD) is activated in the liver of diabetic mice, which parallels with a significant reduction in AMPK activity and a significant increase in intracellular glucose accumulation in human HepG2 cells. AMPD activation is induced by a reduction in intracellular phosphate levels, which is characteristic of insulin resistance and diabetic states. Increased gluconeogenesis is mediated by reduced TORC2 phosphorylation at Ser171 by AMPK in these cells, as well as by the up-regulation of the rate-limiting enzymes PEPCK and G6Pc. The mechanism whereby AMPD controls AMPK activation depends on the production of a specific AMP downstream metabolite through AMPD, uric acid. In this regard, humans have higher uric acid levels than most mammals due to a mutation in uricase, the enzyme involved in uric acid degradation in most mammals, that developed during a period of famine in Europe 1.5 × 10(7) yr ago. Here, working with resurrected ancestral uricases obtained from early hominids, we show that their expression on HepG2 cells is enough to blunt gluconeogenesis in parallel with an up-regulation of AMPK activity. These studies identify a key role AMPD and uric acid in mediating hepatic gluconeogenesis in the diabetic state, via a mechanism involving AMPK down-regulation and overexpression of PEPCK and G6Pc. The uricase mutation in the Miocene likely provided a survival advantage to help maintain glucose levels under conditions of near starvation, but today likely has a role in the pathogenesis of diabetes.


Subject(s)
AMP Deaminase/physiology , Gluconeogenesis/physiology , Liver/metabolism , Starvation/physiopathology , Uric Acid/metabolism , AMP Deaminase/antagonists & inhibitors , AMP Deaminase/genetics , AMP-Activated Protein Kinases/physiology , Animals , Diabetes Mellitus, Experimental/metabolism , Europe , Gene Expression Regulation, Enzymologic , Gluconeogenesis/drug effects , Glucose-6-Phosphatase/biosynthesis , Hep G2 Cells , History, Ancient , Hominidae/physiology , Humans , Insulin/metabolism , Insulin Resistance , Insulin Secretion , Liver/enzymology , Male , Mechanistic Target of Rapamycin Complex 2 , Mice , Mice, Inbred C57BL , Models, Biological , Multiprotein Complexes/physiology , Phosphates/metabolism , Phosphates/pharmacology , Phosphoenolpyruvate Carboxykinase (ATP)/biosynthesis , Recombinant Fusion Proteins/metabolism , Selection, Genetic , Specific Pathogen-Free Organisms , Starvation/history , TOR Serine-Threonine Kinases/physiology , Transduction, Genetic , Urate Oxidase/genetics , Urate Oxidase/history , Urate Oxidase/metabolism , Uric Acid/pharmacology
6.
Proc Natl Acad Sci U S A ; 109(11): 4320-5, 2012 Mar 13.
Article in English | MEDLINE | ID: mdl-22371574

ABSTRACT

Fructose intake from added sugars correlates with the epidemic rise in obesity, metabolic syndrome, and nonalcoholic fatty liver disease. Fructose intake also causes features of metabolic syndrome in laboratory animals and humans. The first enzyme in fructose metabolism is fructokinase, which exists as two isoforms, A and C. Here we show that fructose-induced metabolic syndrome is prevented in mice lacking both isoforms but is exacerbated in mice lacking fructokinase A. Fructokinase C is expressed primarily in liver, intestine, and kidney and has high affinity for fructose, resulting in rapid metabolism and marked ATP depletion. In contrast, fructokinase A is widely distributed, has low affinity for fructose, and has less dramatic effects on ATP levels. By reducing the amount of fructose for metabolism in the liver, fructokinase A protects against fructokinase C-mediated metabolic syndrome. These studies provide insights into the mechanisms by which fructose causes obesity and metabolic syndrome.


Subject(s)
Fructokinases/metabolism , Metabolic Syndrome/enzymology , Animals , Energy Metabolism/drug effects , Feeding Behavior/drug effects , Fructose/administration & dosage , Fructose/metabolism , Fructose/pharmacology , Isoenzymes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout
7.
J Am Soc Nephrol ; 25(11): 2526-38, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24876114

ABSTRACT

Diabetes is associated with activation of the polyol pathway, in which glucose is converted to sorbitol by aldose reductase. Previous studies focused on the role of sorbitol in mediating diabetic complications. However, in the proximal tubule, sorbitol can be converted to fructose, which is then metabolized largely by fructokinase, also known as ketohexokinase, leading to ATP depletion, proinflammatory cytokine expression, and oxidative stress. We and others recently identified a potential deleterious role of dietary fructose in the generation of tubulointerstitial injury and the acceleration of CKD. In this study, we investigated the potential role of endogenous fructose production, as opposed to dietary fructose, and its metabolism through fructokinase in the development of diabetic nephropathy. Wild-type mice with streptozotocin-induced diabetes developed proteinuria, reduced GFR, and renal glomerular and proximal tubular injury. Increased renal expression of aldose reductase; elevated levels of renal sorbitol, fructose, and uric acid; and low levels of ATP confirmed activation of the fructokinase pathway. Furthermore, renal expression of inflammatory cytokines with macrophage infiltration was prominent. In contrast, diabetic fructokinase-deficient mice demonstrated significantly less proteinuria, renal dysfunction, renal injury, and inflammation. These studies identify fructokinase as a novel mediator of diabetic nephropathy and document a novel role for endogenous fructose production, or fructoneogenesis, in driving renal disease.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/metabolism , Fructokinases/metabolism , Fructose/biosynthesis , Fructose/metabolism , Kidney Tubules, Proximal/enzymology , Animals , Blood Glucose/metabolism , Body Weight , Cell Line, Transformed , Chemokines/metabolism , Cytokines/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetic Nephropathies/pathology , Humans , Kidney Cortex/enzymology , Kidney Cortex/pathology , Kidney Glomerulus/cytology , Kidney Glomerulus/pathology , Kidney Tubules, Proximal/pathology , Macrophages/metabolism , Macrophages/pathology , Mice, Inbred C57BL , Mice, Knockout , Polymers/metabolism
8.
Kidney Int ; 86(2): 294-302, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24336030

ABSTRACT

The epidemic of chronic kidney disease in Nicaragua (Mesoamerican nephropathy) has been linked with recurrent dehydration. Here we tested whether recurrent dehydration may cause renal injury by activation of the polyol pathway, resulting in the generation of endogenous fructose in the kidney that might subsequently induce renal injury via metabolism by fructokinase. Wild-type and fructokinase-deficient mice were subjected to recurrent heat-induced dehydration. One group of each genotype was provided water throughout the day and the other group was hydrated at night, after the dehydration. Both groups received the same total hydration in 24 h. Wild-type mice that received delayed hydration developed renal injury, with elevated serum creatinine, increased urinary NGAL, proximal tubular injury, and renal inflammation and fibrosis. This was associated with activation of the polyol pathway, with increased renal cortical sorbitol and fructose levels. Fructokinase-knockout mice with delayed hydration were protected from renal injury. Thus, recurrent dehydration can induce renal injury via a fructokinase-dependent mechanism, likely from the generation of endogenous fructose via the polyol pathway. Access to sufficient water during the dehydration period can protect mice from developing renal injury. These studies provide a potential mechanism for Mesoamerican nephropathy.


Subject(s)
Dehydration/enzymology , Fructokinases/metabolism , Kidney/injuries , Aldehyde Reductase/metabolism , Animals , Blood Pressure , Dehydration/complications , Disease Models, Animal , Fructokinases/deficiency , Fructokinases/genetics , Kidney/pathology , Kidney/physiopathology , Kidney Cortex/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Renal Insufficiency, Chronic/enzymology , Renal Insufficiency, Chronic/etiology , Uric Acid/metabolism
9.
Hepatology ; 58(5): 1632-43, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23813872

ABSTRACT

UNLABELLED: Fructose intake from added sugars has been implicated as a cause of nonalcoholic fatty liver disease. Here we tested the hypothesis that fructose may interact with a high-fat diet to induce fatty liver, and to determine if this was dependent on a key enzyme in fructose metabolism, fructokinase. Wild-type or fructokinase knockout mice were fed a low-fat (11%), high-fat (36%), or high-fat (36%) and high-sucrose (30%) diet for 15 weeks. Both wild-type and fructokinase knockout mice developed obesity with mild hepatic steatosis and no evidence of hepatic inflammation on a high-fat diet compared to a low-fat diet. In contrast, wild-type mice fed a high-fat and high-sucrose diet developed more severe hepatic steatosis with low-grade inflammation and fibrosis, as noted by increased CD68, tumor necrosis factor alpha, monocyte chemoattractant protein-1, alpha-smooth muscle actin, and collagen I and TIMP1 expression. These changes were prevented in the fructokinase knockout mice. CONCLUSION: An additive effect of high-fat and high-sucrose diet on the development of hepatic steatosis exists. Further, the combination of sucrose with high-fat diet may induce steatohepatitis. The protection in fructokinase knockout mice suggests a key role for fructose (from sucrose) in this development of steatohepatitis. These studies emphasize the important role of fructose in the development of fatty liver and nonalcoholic steatohepatitis.


Subject(s)
Diet, High-Fat , Fatty Liver/etiology , Fructokinases/physiology , Sucrose/administration & dosage , Animals , Energy Intake , Fructose/metabolism , Liver/metabolism , Liver/pathology , Mice , Mice, Inbred C57BL , Weight Gain
10.
Pediatr Nephrol ; 28(9): 1803-12, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23689904

ABSTRACT

BACKGROUND: Minimal change disease (MCD) is the most common cause of nephrotic syndrome in children and is associated with the expression of CD80 in podocytes and the increased excretion of CD80 in urine. We hypothesized that serum from patients with MCD might stimulate CD80 expression in cultured podocytes. METHODS: Sera and peripheral blood mononuclear cells (PBMCs) were collected from subjects with MCD in relapse and remission and from normal controls. Immortalized human podocytes were incubated with culture media containing patient sera or supernatants from patient and control PBMC cultures. CD80 expression was measured by quantitative PCR and western blot analysis. RESULTS: Sera collected from patients with MCD in relapse, but not in remission, significantly increased CD80 expression (mean ± standard deviation: 1.8 ± 0.7 vs. 0.8 ± 0.2; p < 0.004) and CD80 protein secretion by podocytes (p < 0.05 between relapse and normal controls). No such CD80 increase was observed when podocytes were incubated with supernatants of PBMC cultures from patients in relapse. CONCLUSIONS: Sera from MCD patients in relapse, but not in remission, stimulated CD80 expression in cultured podocytes. Identifying this factor in sera could provide insights into the pathogenesis of this disorder. No role in CD80 expression by podocytes was found for cytokines released by PBMCs.


Subject(s)
B7-1 Antigen/biosynthesis , Nephrosis, Lipoid/metabolism , Podocytes/metabolism , Adolescent , Anti-Inflammatory Agents/therapeutic use , Blotting, Western , Cells, Cultured , Child , Child, Preschool , Cytokines/metabolism , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Female , Glyceraldehyde 3-Phosphate Dehydrogenase (NADP+)/metabolism , Humans , Kidney Function Tests , Male , Monocytes/metabolism , Nephrosis, Lipoid/blood , Nephrosis, Lipoid/drug therapy , Prednisone/therapeutic use , RNA/biosynthesis , RNA/genetics , RNA/isolation & purification , Real-Time Polymerase Chain Reaction , Recurrence , Serum , Young Adult
11.
bioRxiv ; 2023 Sep 07.
Article in English | MEDLINE | ID: mdl-36993762

ABSTRACT

The Warburg Effect is characterized by accelerated glycolytic metabolism and lactate production and under fully aerobic conditions is a hallmark of cancer cells. Recently, we have demonstrated the role of endogenous, glucose-derived lactate as an oncometabolite which regulates gene expression in the estrogen receptor positive (ER+) MCF7 cell line cultivated in glucose media. Presently, with the addition of a triple negative breast cancer (TNBC) cell line, MDA-MB-231, we further confirm the effect of lactate on gene expression patterns and extend results to include lactate effects on protein expression. As well, we report effects of lactate on the expression of E-cadherin and vimentin, proteins associated with epithelial-to-mesenchymal transition (EMT). Endogenous lactate regulates the expression of multiple genes involved in carcinogenesis. In MCF7 cells, lactate increased the expression of EGFR, VEGF, HIF-1a, KRAS, MIF, mTOR, PIK3CA, TP53, and CDK4 as well as decreased the expression of ATM, BRCA1, BRCA2, E2F1, MET, MYC, and RAF mainly after 48h of exposure. On the other hand, in the MDA-MB-231 cell line, lactate increased the expressions of PIK3CA, VEGF, EGFR, mTOR, HIF-1α, ATM, E2F1, TP53 and decreased the expressions of BRCA1, BRCA2, CDK4, CDK6, MET, MIF, MYC, and RAF after 48h of exposure. In response to endogenous lactate, changes in protein expression of representative genes corroborated changes in mRNA expressions. Finally, lactate exposure decreased E-cadherin protein expression in MCF7 cells and increased vimentin expression in MDA-MB-231 cells. Furthermore, by genetically silencing LDHA in MCF7 cells, we show suppression of protein expression of EGFR and HIF-1α, while full protein expression occurred under glucose and glucose + exogenous lactate exposure. Hence, endogenous, glucose-derived lactate, and not glucose, elicited changes in gene and protein expression levels. In this study, we demonstrate that endogenous lactate produced under aerobic conditions (Warburg Effect) elicits important changes in gene and protein expression in both ER+ and TNBC cell lines. The widespread regulation of multiple genes by lactate and involves those involved in carcinogenesis including DNA repair, cell growth, proliferation, angiogenesis, and metastasis. Furthermore, lactate affected the expression of two relevant EMT biomarkers, E-cadherin and vimentin, which could contribute to the complex process of EMT and a shift towards a more mesenchymal phenotype in the two cancer cell lines studied.

12.
Am J Physiol Renal Physiol ; 302(9): F1151-60, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22338086

ABSTRACT

Nicorandil is an orally available drug that can act as a nitric oxide donor, an antioxidant, and an ATP-dependent K channel activator. We hypothesized that it may have a beneficial role in treating diabetic nephropathy. We administered nicorandil to a model of advanced diabetic nephropathy (the streptozotocin-induced diabetes in mice lacking endothelial nitric oxide synthase, eNOSKO); controls included diabetic eNOS KO mice without nicorandil and nondiabetic eNOS KO mice treated with either nicorandil or vehicle. Mice were treated for 8 wk. Histology, blood pressure, and renal function were determined. Additional studies involved examining the effects of nicorandil on cultured human podocytes. Here, we found that nicorandil did not affect blood glucose levels, blood pressure, or systemic endothelial function, but significantly reduced proteinuria and glomerular injury (mesangiolysis and glomerulosclerosis). Nicorandil protected against podocyte loss and podocyte oxidative stress. Studies in cultured podocytes showed that nicorandil likely protects against glucose-mediated oxidant stress via the ATP-dependent K channel as opposed to its NO-stimulating effects. In conclusion, nicorandil may be beneficial in diabetic nephropathy by preserving podocyte function. We recommend clinical trials to determine whether nicorandil may benefit diabetic nephropathy or other conditions associated with podocyte dysfunction.


Subject(s)
Antioxidants/therapeutic use , Diabetic Nephropathies/drug therapy , Nicorandil/therapeutic use , Nitric Oxide Donors/therapeutic use , Nitric Oxide Synthase Type III/deficiency , Severity of Illness Index , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , Blood Pressure/drug effects , Blood Pressure/physiology , Cells, Cultured , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/complications , Diabetic Nephropathies/etiology , Diabetic Nephropathies/physiopathology , Disease Models, Animal , Disease Progression , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nicorandil/pharmacology , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase Type III/genetics , Oxidative Stress/drug effects , Oxidative Stress/physiology , Podocytes/cytology , Podocytes/drug effects , Podocytes/metabolism , Reactive Oxygen Species/metabolism , Streptozocin/adverse effects
13.
Nephrol Dial Transplant ; 27(1): 81-9, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21617192

ABSTRACT

BACKGROUND: Recent studies suggest that CD80 (also known as B7.1) is expressed on podocytes in minimal-change disease (MCD) and may have a role in mediating proteinuria. CD80 expression is known to be induced by Toll-like receptor (TLR) ligands in dendritic cells. We therefore evaluated the ability of TLR to induce CD80 in human cultured podocytes. METHODS: Conditionally immortalized human podocytes were evaluated for TLR expression. Based on high expression of TLR3, we evaluated the effect of polyinosinic-polycytidylic acid (polyIC), a TLR3 ligand, to induce CD80 expression in vitro. RESULTS: TLR1-6 and 9 messenger RNA (mRNA) were expressed in podocytes. Among TLR ligands 1-9, CD80 mRNA expression was significantly induced by polyIC and lipopolysaccharide (TLR4 ligand) with the greatest stimulation by polyIC (6.8 ± 0.7 times at 6 h, P < 0.001 versus control). PolyIC induced increased expression of Cathepsin L, decreased synaptopodin expression and resulted in actin reorganization which suggested a similar injury pattern as observed with lipopolyssaccharide. PolyIC induced type I and type II interferon signaling, nuclear factor kappa B (NF-κB) activation and the induction of CD80 expression. Knockdown of CD80 protected against actin reorganization and reduced synaptopodin expression in response to polyIC. Dexamethasone, a corticosteroid commonly used to treat MCD, also blocked both basal and polyIC-stimulated CD80 expression, as did inhibition of NF-κB. CONCLUSIONS: Activation of TLR3 on cultured human podocytes induces CD80 expression and phenotypic change via an NF-κB-dependent mechanism and is partially blocked by dexamethasone. These studies provide a mechanism by which viral infections may cause proteinuria.


Subject(s)
B7-1 Antigen/metabolism , Dendritic Cells/metabolism , NF-kappa B/metabolism , Podocytes/metabolism , Toll-Like Receptor 3/metabolism , Anti-Inflammatory Agents/pharmacology , B7-1 Antigen/antagonists & inhibitors , B7-1 Antigen/genetics , Blotting, Western , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/drug effects , Dexamethasone/pharmacology , Fluorescent Antibody Technique , Humans , Lipopolysaccharides/pharmacology , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , Podocytes/cytology , Podocytes/drug effects , Poly I-C/pharmacology , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Signal Transduction , Toll-Like Receptor 3/genetics
14.
J Biol Chem ; 285(41): 31694-703, 2010 Oct 08.
Article in English | MEDLINE | ID: mdl-20639513

ABSTRACT

Expression of aquaporin-1 (AQP1) and -2 (AQP2) channels in the kidney are critical for the maintenance of water homeostasis and the operation of the urinary concentrating mechanism. Hypertonic stress induced in inner medullary (IMCD3) cells by addition of NaCl to the medium substantially up-regulated the mRNA and protein expression of AQP1, suggesting that its activation occurs at a transcriptional and a translational levels. In contrast, no up-regulation of AQP1 was observed when these cells were exposed to the same tonicity by addition of urea. To explore the transcriptional activation of aqp1 under hypertonic stress, we examined the role of the transcription factor associated with hypertonicity, TonEBP. Treatment of IMCD3 cells with the TonEBP inhibitor rottlerin or silencing its expression with specific shRNA technology led to a substantial reduction in AQP1 expression under hypertonic conditions. Moreover, we defined a conserved TonEBP binding site located 811 bp upstream of the aqp1 exon that is essential for its expression. Single site-directed mutation of this TonE site led to a 54 ± 5% (p < 0.01) decrease in AQP1 luciferase-driven activity under hypertonic stress. TonEBP mutant mice display marked decrement in the expression of AQP1 in the inner medulla. In conclusion, these data demonstrate that TonEBP is necessary for the regulation of AQP1 expression in the inner medulla of the kidney under hypertonic conditions.


Subject(s)
Aquaporin 1/biosynthesis , Kidney Medulla/metabolism , Response Elements/physiology , Transcription Factors/metabolism , Up-Regulation/physiology , Acetophenones/pharmacology , Animals , Aquaporin 1/genetics , Aquaporin 2/genetics , Aquaporin 2/metabolism , Benzopyrans/pharmacology , Cell Line , Enzyme Inhibitors/pharmacology , Gene Silencing , Mice , Mice, Mutant Strains , Mutagenesis, Site-Directed , Mutation , Osmotic Pressure/drug effects , Osmotic Pressure/physiology , Protein Biosynthesis/drug effects , Protein Biosynthesis/physiology , Sodium Chloride/pharmacology , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription, Genetic/drug effects , Transcription, Genetic/physiology , Up-Regulation/drug effects , Urea/pharmacology
15.
Am J Physiol Renal Physiol ; 300(5): F1255-61, 2011 May.
Article in English | MEDLINE | ID: mdl-21325494

ABSTRACT

ANG II plays a major role in renal water and sodium regulation. In the immortalized mouse renal collecting duct principal cells (mpkCCD(cl4)) cell line, we treated cells with ANG II and examined aquaporin-2 (AQP2) protein expression, trafficking, and mRNA levels, by immunoblotting, immunofluorescence, and RT-PCR. After 24-h incubation, ANG II-induced AQP2 protein expression was observed at the concentration of 10(-10) M and increased in a dose-dependent manner. ANG II (10(-7) M) increased AQP2 protein expression and mRNA levels at 0.5, 1, 2, 6, and 24 h. Immunofluorescence studies showed that ANG II increased the apical membrane targeting of AQP2 from 30 min to 6 h. Next, the signaling pathways underlying the ANG II-induced AQP2 expression were investigated. The PKC inhibitor Ro 31-8220 (5 × 10(-6) M) and the PKA inhibitor H89 (10(-5) M) blocked ANG II-induced AQP2 expression, respectively. Calmodulin inhibitor W-7 markedly reduced ANG II- and/or dDAVP-stimulated AQP2 expression. ANG II (10(-9) M) and/or dDAVP (10(-10) M) stimulated AQP2 protein levels and cAMP accumulation, which was completely blocked by pretreatment with the vasopressin V2 receptor (V2R) antagonist SR121463B (10(-8) M). Pretreatment with the angiotensin AT(1) receptor (AT1R) antagonist losartan (3 × 10(-6) M) blocked ANG II (10(-9) M)-stimulated AQP2 protein expression and cAMP accumulation, and partially blocked dDAVP (10(-10) M)- and dDAVP+ANG II-induced AQP2 protein expression and cAMP accumulation. In conclusion, ANG II regulates AQP2 protein, trafficking, and gene expression in renal collecting duct principal cells. ANG II-induced AQP2 expression involves cAMP, PKC, PKA, and calmodulin signaling pathways via V2 and AT(1) receptors.


Subject(s)
Angiotensin II/metabolism , Aquaporin 2/metabolism , Kidney Tubules, Collecting/metabolism , Analysis of Variance , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Aquaporin 2/genetics , Blotting, Western , Calmodulin/metabolism , Cell Line , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Deamino Arginine Vasopressin/pharmacology , Fluorescent Antibody Technique , Hormone Antagonists/pharmacology , Kidney Tubules, Collecting/cytology , Kidney Tubules, Collecting/drug effects , Mice , Protein Kinase C/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Transport , RNA, Messenger/metabolism , Receptor, Angiotensin, Type 1/metabolism , Receptors, Vasopressin/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Time Factors
16.
Proc Natl Acad Sci U S A ; 105(41): 15797-802, 2008 Oct 14.
Article in English | MEDLINE | ID: mdl-18840681

ABSTRACT

We reported that the multiple PDZ protein 1 (MUPP1) is an osmotic response protein in kidney cells. This up-regulation was found to be necessary for the maintenance of tight epithelial properties in these cells. We investigated whether an interaction with one or more members of the claudin family is responsible for this observation. In response to hypertonicity, the up-regulation of claudin-4 (Cldn4) expression, and not other claudins, was initially identified in inner medullary collecting duct (IMCD3) cells by gene array and further verified by quantitative PCR and Western blotting. In kidney tissues, Cldn4 expression was substantial in the papilla and absent in the cortex. Furthermore, Cldn4 expression significantly increased in the papilla of mice after 36 h of thirsting. Cldn4 immunofluorescence in hypertonically stressed cells revealed colocalization with MUPP1 at the tight junctions. Interaction between Cldn4 and MUPP1 was also demonstrated by coimmunoprecipitation of both proteins from IMCD3 cells chronically adapted to hypertonicity. In IMCD3 cells stably silenced for MUPP1 expression under hypertonic conditions, a significant decrement in Cldn4 expression was observed that was restored after inhibition of lysosome activity. Immunofluorescence detection identified that in these MUPP1-silenced cells Cldn4 was mistargeted to the lysosomes. Functionally, silencing Cldn4 expression in IMCD3 cells resulted in a decrease in the transepithelial resistance to the same degree as observed when MUPP1 expression was silenced, suggesting that MUPP1 contributes to the maintenance of a tight epithelium in the medulla of the kidney under hypertonic stress by correctly localizing Cldn4 to the tight junctions.


Subject(s)
Carrier Proteins/metabolism , Kidney Medulla/cytology , Membrane Proteins/genetics , Osmotic Pressure , Tight Junctions/metabolism , Animals , Cells, Cultured , Claudin-4 , Kidney Tubules, Collecting , Mice , Protein Binding , Tissue Distribution , Up-Regulation/genetics
17.
Kidney Int ; 78(3): 296-302, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20485332

ABSTRACT

Controversy exists as to whether minimal change disease (MCD) and focal segmental glomerulosclerosis (FSGS) represent different diseases or are manifestations within the same disease spectrum. Urinary excretion of CD80 (also known as B7.1) is elevated in patients with MCD and hence we tested whether urinary CD80 excretion might distinguish between patients with MCD from those with FSGS. Urinary CD80 was measured in 17 patients with biopsy-proven MCD and 22 with proven FSGS using a commercially available enzyme-linked immunosorbent assay and its molecular size determined by western blot analysis. A significant increase in urinary CD80, normalized to urinary creatinine, was found in patients with MCD in relapse compared to those in remission or those with FSGS. No significant differences were seen when CD80 urinary excretion from MCD patients in remission were compared to those with FSGS. In seven of eight MCD patients in relapse, CD80 was found in glomeruli by immunohistochemical analysis of their biopsy specimen. No CD80 was found in glomeruli of two patients with FSGS and another MCD patient in remission. Thus, our study supports the hypothesis that MCD and FSGS represent two different diseases rather than a continuum of one disease. Urinary CD80 excretion may be a useful marker to differentiate between MCD and FSGS.


Subject(s)
B7-1 Antigen/urine , Glomerulosclerosis, Focal Segmental/immunology , Nephrosis, Lipoid/urine , Adolescent , Adult , Aged, 80 and over , Biomarkers/urine , Biopsy , Blotting, Western , Child , Child, Preschool , Creatinine/urine , Enzyme-Linked Immunosorbent Assay , Female , Glomerulosclerosis, Focal Segmental/pathology , Glomerulosclerosis, Focal Segmental/urine , Humans , Kidney Glomerulus/immunology , Kidney Glomerulus/pathology , Male , Middle Aged , Nephrosis, Lipoid/immunology , Nephrosis, Lipoid/pathology , Recurrence , Remission Induction , Urinary Tract/immunology , Urinary Tract/pathology , Young Adult
18.
Nephrol Dial Transplant ; 25(6): 1865-9, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20064950

ABSTRACT

BACKGROUND: Recent studies suggest that uric acid is a mediator of diabetic nephropathy. We hypothesized that elevated serum uric acid levels are a strong predictor of albuminuria in patients with type 1 diabetes. METHODS: We analyzed data from the Coronary Artery Calcification in Type 1 Diabetes study, a prospective observational study. A stepwise logistic regression model was applied to predict the development of micro- or macroalbuminuria after 6 years of follow-up in 324 participants who had no evidence of micro- or macroalbuminuria at baseline. A P-value <0.1 was used as the criteria for entry into and removal from the model. RESULTS: The following factors were selected in the stepwise multivariate model as predictors of micro- or macroalbuminuria at the 6-year follow-up visit: baseline serum uric acid levels, HbA(1c) and pre-albuminuria. For every 1-mg/dl increase in serum uric acid levels at baseline, there was an 80% increased risk of developing micro- or macroalbuminuria at 6 years (odds ratio 1.8; 95% confidence interval 1.2, 2.8; P = 0.005). Additional covariates considered in the stepwise model were sex, age, duration of diabetes, angiotensin-converting enzyme inhibitor or angiotensin II receptor blocker treatment, waist circumference, waist/hip ratio, body mass index, systolic and diastolic blood pressure, smoking, serum creatinine, cystatin C, high-density lipoprotein cholesterol and triglycerides. CONCLUSION: Elevated serum uric acid levels are a strong predictor of the development of albuminuria in patients with type 1 diabetes.


Subject(s)
Albuminuria/etiology , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/complications , Diabetic Nephropathies/etiology , Uric Acid/blood , Adult , Biomarkers/blood , Cohort Studies , Female , Humans , Logistic Models , Male , Middle Aged , Models, Biological , Prospective Studies , Risk Factors
19.
Cancer Res ; 80(22): 5051-5062, 2020 11 15.
Article in English | MEDLINE | ID: mdl-32973083

ABSTRACT

Small cell lung cancer (SCLC) remains a recalcitrant disease where limited therapeutic options have not improved overall survival, and approved targeted therapies are lacking. Amplification of the tyrosine kinase receptor FGFR1 (fibroblast growth factor receptor 1) is one of the few actionable alterations found in the SCLC genome. However, efforts to develop targeted therapies for FGFR1-amplified SCLC are hindered by critical gaps in knowledge around the molecular origins and mediators of FGFR1-driven signaling as well as the physiologic impact of targeting FGFR1. Here we show that increased FGFR1 promotes tumorigenic progression in precancerous neuroendocrine cells and is required for SCLC development in vivo. Notably, Fgfr1 knockout suppressed tumor development in a mouse model lacking the retinoblastoma-like protein 2 (Rbl2) tumor suppressor gene but did not affect a model with wild-type Rbl2. In support of a functional interaction between these two genes, loss of RBL2 induced FGFR1 expression and restoration of RBL2 repressed it, suggesting a novel role for RBL2 as a regulator of FGFR1 in SCLC. Additionally, FGFR1 activated phospholipase C gamma 1 (PLCG1), whereas chemical inhibition of PLCG1 suppressed SCLC growth, implicating PLCG1 as an effector of FGFR1 signaling in SCLC. Collectively, this study uncovers mechanisms underlying FGFR1-driven SCLC that involve RBL2 upstream and PLCG1 downstream, thus providing potential biomarkers for anti-FGFR1 therapy. SIGNIFICANCE: This study identifies RBL2 and PLCG1 as critical components of amplified FGFR1 signaling in SCLC, thus representing potential targets for biomarker analysis and therapeutic development in this disease.


Subject(s)
Lung Neoplasms/pathology , Neoplasm Proteins/metabolism , Phospholipase C gamma/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Retinoblastoma-Like Protein p130/metabolism , Small Cell Lung Carcinoma/pathology , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Disease Progression , Female , Gene Deletion , Genes, Regulator , Genes, Retinoblastoma , Humans , Lung Neoplasms/etiology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Mice , Mice, Knockout , Neoplasm Proteins/genetics , Neuroendocrine Cells/metabolism , Neuroendocrine Cells/pathology , Phospholipase C gamma/antagonists & inhibitors , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , Receptor, Fibroblast Growth Factor, Type 1/genetics , Retinoblastoma-Like Protein p130/genetics , Small Cell Lung Carcinoma/etiology , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/metabolism
20.
Cancer Metab ; 8: 16, 2020.
Article in English | MEDLINE | ID: mdl-32670573

ABSTRACT

Obesity and metabolic syndrome are strongly associated with cancer, and these disorders may share a common mechanism. Recently, fructose has emerged as a driving force to develop obesity and metabolic syndrome. Thus, we assume that fructose may be the mechanism to explain why obesity and metabolic syndrome are linked with cancer. Clinical and experimental evidence showed that fructose intake was associated with cancer growth and that fructose transporters are upregulated in various malignant tumors. Interestingly, fructose metabolism can be driven under low oxygen conditions, accelerates glucose utilization, and exhibits distinct effects as compared to glucose, including production of uric acid and lactate as major byproducts. Fructose promotes the Warburg effect to preferentially downregulate mitochondrial respiration and increases aerobic glycolysis that may aid metastases that initially have low oxygen supply. In the process, uric acid may facilitate carcinogenesis by inhibiting the TCA cycle, stimulating cell proliferation by mitochondrial ROS, and blocking fatty acid oxidation. Lactate may also contribute to cancer growth by suppressing fat oxidation and inducing oncogene expression. The ability of fructose metabolism to directly stimulate the glycolytic pathway may have been protective for animals living with limited access to oxygen, but may be deleterious toward stimulating cancer growth and metastasis for humans in modern society. Blocking fructose metabolism may be a novel approach for the prevention and treatment of cancer.

SELECTION OF CITATIONS
SEARCH DETAIL