Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 11(1): 2974, 2021 02 03.
Article in English | MEDLINE | ID: mdl-33536542

ABSTRACT

In obesity, adipose tissue derived inflammation is associated with unfavorable metabolic consequences. Uremic inflammation is prevalent and contributes to detrimental outcomes. However, the contribution of adipose tissue inflammation in uremia has not been characterized. We studied the contribution of adipose tissue to uremic inflammation in-vitro, in-vivo and in human samples. Exposure to uremic serum resulted in activation of inflammatory pathways including NFκB and HIF1, upregulation of inflammatory cytokines/chemokines and catabolism with lipolysis, and lactate production. Also, co-culture of adipocytes with macrophages primed by uremic serum resulted in higher inflammatory cytokine expression than adipocytes exposed only to uremic serum. Adipose tissue of end stage renal disease subjects revealed increased macrophage infiltration compared to controls after BMI stratification. Similarly, mice with kidney disease recapitulated the inflammatory state observed in uremic patients and additionally demonstrated increased peripheral monocytes and inflammatory polarization of adipose tissue macrophages (ATMS). In contrast, adipose tissue in uremic IL-6 knock out mice showed reduced ATMS density compared to uremic wild-type controls. Differences in ATMS density highlight the necessary role of IL-6 in macrophage infiltration in uremia. Uremia promotes changes in adipocytes and macrophages enhancing production of inflammatory cytokines. We demonstrate an interaction between uremic activated macrophages and adipose tissue that augments inflammation in uremia.


Subject(s)
Adipocytes/immunology , Kidney Failure, Chronic/immunology , Macrophages/immunology , Obesity/complications , Uremia/immunology , 3T3-L1 Cells , Adipocytes/metabolism , Adipose Tissue/metabolism , Animals , Case-Control Studies , Cell Communication/immunology , Cells, Cultured , Coculture Techniques , Cytokines/metabolism , Humans , Inflammation/blood , Inflammation/immunology , Inflammation Mediators/metabolism , Kidney Failure, Chronic/blood , Kidney Failure, Chronic/metabolism , Lipolysis/immunology , Macrophages/metabolism , Male , Mice , Obesity/blood , Obesity/immunology , Obesity/metabolism , Primary Cell Culture , RAW 264.7 Cells , THP-1 Cells , Uremia/blood , Uremia/metabolism
2.
Mol Imaging Biol ; 22(2): 293-302, 2020 04.
Article in English | MEDLINE | ID: mdl-31292914

ABSTRACT

PURPOSE: Scintigraphic imaging of malignant glioblastoma (MG) continues to be challenging. We hypothesized that VPAC1 cell surface receptors can be targeted for positron emission tomography (PET) imaging of orthotopically implanted MG in a mouse model, using a VPAC1-specific peptide [64Cu]TP3805. PROCEDURES: The expression of VPAC1 in mouse GL261 and human U87 glioma cell lines was determined by western blot. The ability of [64Cu]TP3805 to bind to GL261 and U87 cells was studied by cell-binding. Receptor-blocking studies were performed to validate receptor specificity. GL261 tumors were implanted orthotopically in syngeneic T-bet knockout C57BL/6 mouse brain (N = 15) and allowed to grow for 2-3 weeks. Mice were injected i.v., first with ~ 150 µCi of 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) then 24 h later with ~ 200 µCi of [64Cu]TP3805. In another set of tumor-bearing mice, (N = 5), ionic [64Cu]Cl2 was injected as a control. Mice were imaged at a 2-h post-injection using an Inveon micro-PET/CT, sacrificed and % ID/g of [64Cu]TP3805 and [64Cu]Cl2 were calculated in a tumor, normal brain, and other tissues. For histologic tissue examination, 3-µm thick sections of the tumors and normal brain were prepared, digital autoradiography (DAR) was performed, and then the sections were H&E stained for histologic examination. RESULTS: Western blots showed a strong signal for VPAC1 on both cell lines. [64Cu]TP3805 cell-binding was 87 ± 1.5 %. Receptor-blocking reduced cell-binding to 24.3 ± 1.5 % (P < 0.01). PET imaging revealed remarkable accumulation of [64Cu]TP3805 in GL261 MG with a negligible background in the normal brain, as compared to [18F]FDG. Micro-PET/CT image analyses and tissue distribution showed that the brain tumor uptake for [64Cu]TP3805 was 8.2 ± 1.7 % ID/g and for [64Cu]Cl2 2.1 ± 0.5 % ID/g as compared to 1.0 ± 0.3 % ID/g and 1.4 ± 0.3 % ID/g for normal mouse brains, respectively. The high tumor/normal brain ratio for [64Cu]TP3805 (8.1 ± 1.1) allowed tumors to be visualized unequivocally. Histology and [64Cu]TP3805 DAR differentiated malignant tumors from healthy brain and confirmed PET findings. CONCLUSION: Targeting VPAC1 receptors using [64Cu]TP3805 for PET imaging of MG is a promising novel approach and calls for further investigation.


Subject(s)
Copper Radioisotopes/pharmacology , Glioblastoma/diagnostic imaging , Positron Emission Tomography Computed Tomography , Receptors, Vasoactive Intestinal Polypeptide, Type I/genetics , Animals , Brain/diagnostic imaging , Brain/metabolism , Brain Neoplasms/diagnostic imaging , Cell Line, Tumor , Fluorodeoxyglucose F18 , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Transplantation , Peptides/chemistry , Radiopharmaceuticals , Tissue Distribution
3.
Am J Clin Oncol ; 43(2): 82-86, 2020 02.
Article in English | MEDLINE | ID: mdl-31693508

ABSTRACT

OBJECTIVES: There is no study published regarding the benefit of radiation therapy (RT) in combination with immune checkpoint inhibitors (ICIs) for the treatment of metastatic renal cell cancer (mRCC). This report is part of an exploratory study aiming to determine the immunomodulatory activity of RT alone or in combination with pembrolizumab in solid tumors. MATERIALS AND METHODS: mRCC patients were treated with a combination of RT (8 Gy×1 or 4 Gy×5) followed by pembrolizumab with or without lead-in dose of pembrolizumab. Treatment response was measured based on the modified Response Evaluation Criteria in Solid Tumors criteria. Adverse events were monitored and graded. Pre-RT and post-RT tumor biopsies were obtained to evaluate programmed death-ligand 1 expression. Immune markers from peripheral blood before, during, and after treatment were analyzed using flow cytometry. RESULTS: Twelve mRCC patients who progressed on prior antiangiogenic therapy were enrolled. Half had 2 lines of prior therapy. Two patients (16.7%) had partial responses and were on study for 12.4 and 14.5 months. Three patients had stable disease for a period ranging from 4.2 to 10.4 months, whereas 7 patients had progressive disease. Median progression-free survival was 8.6 months and median overall survival was 32.3 months. Three patients had grade ≥3 events (hyperglycemia, thrombocytopenia, transaminitis). Biopsied tissue programmed death-ligand 1 expression and tumor-infiltrating lymphocytes were numerically higher in responders comparing to nonresponders (Modified Proportion Score 45% vs. 30.45%; tumor-infiltrating lymphocytes odds ratio 4.92). CONCLUSION: Combining RT with pembrolizumab in pretreated mRCC is well-tolerated and appears to have comparable efficacy with single-agent nivolumab.


Subject(s)
Adrenal Gland Neoplasms/therapy , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Carcinoma, Renal Cell/therapy , Chemoradiotherapy/methods , Kidney Neoplasms/pathology , Liver Neoplasms/therapy , Adrenal Gland Neoplasms/secondary , Adult , Aged , Aged, 80 and over , Alanine Transaminase , Angiogenesis Inhibitors/therapeutic use , Aspartate Aminotransferases , Carcinoma, Renal Cell/secondary , Female , Humans , Hyperglycemia/chemically induced , Liver Neoplasms/secondary , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Male , Middle Aged , Pilot Projects , Progression-Free Survival , Soft Tissue Neoplasms/secondary , Soft Tissue Neoplasms/therapy , Thrombocytopenia/chemically induced , Treatment Failure , Treatment Outcome
4.
Oncotarget ; 8(47): 81754-81775, 2017 Oct 10.
Article in English | MEDLINE | ID: mdl-29137220

ABSTRACT

The cyclin D1 gene encodes the regulatory subunit of a holoenzyme that drives cell autonomous cell cycle progression and proliferation. Herein we show cyclin D1 abundance is increased >30-fold in the stromal fibroblasts of patients with invasive breast cancer, associated with poor outcome. Cyclin D1 transformed hTERT human fibroblast to a cancer-associated fibroblast phenotype. Stromal fibroblast expression of cyclin D1 (cyclin D1Stroma) in vivo, enhanced breast epithelial cancer tumor growth, restrained apoptosis, and increased autophagy. Cyclin D1Stroma had profound effects on the breast tumor microenvironment increasing the recruitment of F4/80+ and CD11b+ macrophages and increasing angiogenesis. Cyclin D1Stroma induced secretion of factors that promoted expansion of stem cells (breast stem-like cells, embryonic stem cells and bone marrow derived stem cells). Cyclin D1Stroma resulted in increased secretion of proinflammatory cytokines (CCL2, CCL7, CCL11, CXCL1, CXCL5, CXCL9, CXCL12), CSF (CSF1, GM-CSF1) and osteopontin (OPN) (30-fold). OPN was induced by cyclin D1 in fibroblasts, breast epithelial cells and in the murine transgenic mammary gland and OPN was sufficient to induce stem cell expansion. These results demonstrate that cyclin D1Stroma drives tumor microenvironment heterocellular signaling, promoting several key hallmarks of cancer.

5.
Cell Rep ; 20(7): 1654-1666, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28813676

ABSTRACT

Glutamine-synthetase (GS), the glutamine-synthesizing enzyme from glutamate, controls important events, including the release of inflammatory mediators, mammalian target of rapamycin (mTOR) activation, and autophagy. However, its role in macrophages remains elusive. We report that pharmacologic inhibition of GS skews M2-polarized macrophages toward the M1-like phenotype, characterized by reduced intracellular glutamine and increased succinate with enhanced glucose flux through glycolysis, which could be partly related to HIF1α activation. As a result of these metabolic changes and HIF1α accumulation, GS-inhibited macrophages display an increased capacity to induce T cell recruitment, reduced T cell suppressive potential, and an impaired ability to foster endothelial cell branching or cancer cell motility. Genetic deletion of macrophagic GS in tumor-bearing mice promotes tumor vessel pruning, vascular normalization, accumulation of cytotoxic T cells, and metastasis inhibition. These data identify GS activity as mediator of the proangiogenic, immunosuppressive, and pro-metastatic function of M2-like macrophages and highlight the possibility of targeting this enzyme in the treatment of cancer metastasis.


Subject(s)
Enzyme Inhibitors/pharmacology , Glutamate-Ammonia Ligase/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Lung Neoplasms/drug therapy , Methionine Sulfoximine/pharmacology , Neovascularization, Pathologic/prevention & control , Animals , Cell Differentiation , Cell Line, Tumor , Cell Movement/drug effects , Endothelial Cells/drug effects , Endothelial Cells/immunology , Endothelial Cells/pathology , Glucose/metabolism , Glutamate-Ammonia Ligase/deficiency , Glutamine/metabolism , Glycolysis/drug effects , Glycolysis/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/immunology , Injections, Subcutaneous , Interleukin-10/genetics , Interleukin-10/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Macrophages/drug effects , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Knockout , Monocytes/drug effects , Monocytes/immunology , Monocytes/pathology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/pathology , Primary Cell Culture , Succinic Acid/metabolism , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/pathology
6.
Antioxid Redox Signal ; 26(8): 351-363, 2017 03 10.
Article in English | MEDLINE | ID: mdl-27758118

ABSTRACT

AIMS: Microglial cells are brain-resident macrophages engaged in surveillance and maintained in a constant state of relative inactivity. However, their involvement in autoimmune diseases indicates that in pathological conditions microglia gain an inflammatory phenotype. The mechanisms underlying this change in the microglial phenotype are still unclear. Since metabolism is an important modulator of immune cell function, we focused our attention on glutamine synthetase (GS), a modulator of the response to lipopolysaccharide (LPS) activation in other cell types, which is expressed by microglia. RESULTS: GS inhibition enhances release of inflammatory mediators of LPS-activated microglia in vitro, leading to perturbation of the redox balance and decreased viability of cocultured neurons. GS inhibition also decreases insulin-mediated glucose uptake in microglia. In vivo, microglia-specific GS ablation enhances expression of inflammatory markers upon LPS treatment. In the spinal cords from experimental autoimmune encephalomyelitis (EAE), GS expression levels and glutamine/glutamate ratios are reduced. INNOVATION: Recently, metabolism has been highlighted as mediator of immune cell function through the discovery of mechanisms that (behind these metabolic changes) modulate the inflammatory response. The present study shows for the first time a metabolic mechanism mediating microglial response to a proinflammatory stimulus, pointing to GS activity as a master modulator of immune cell function and thus unraveling a potential therapeutic target. CONCLUSIONS: Our study highlights a new role of GS in modulating immune response in microglia, providing insights into the pathogenic mechanisms associated with inflammation and new strategies of therapeutic intervention. Antioxid. Redox Signal. 26, 351-363.


Subject(s)
Glutamate-Ammonia Ligase/metabolism , Inflammation/metabolism , Microglia/metabolism , Animals , Biomarkers , Cell Survival , Cells, Cultured , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Enzyme Activation , Female , Gene Expression , Glucose/metabolism , Glutamate-Ammonia Ligase/antagonists & inhibitors , Glutamate-Ammonia Ligase/genetics , Insulin/metabolism , Lipopolysaccharides/immunology , Metabolome , Metabolomics/methods , Mice , Mice, Knockout , Microglia/immunology , Neurons/metabolism , Nitric Oxide , Reactive Oxygen Species/metabolism , Spinal Cord/metabolism , Spinal Cord/pathology
7.
PLoS Negl Trop Dis ; 9(8): e0004023, 2015.
Article in English | MEDLINE | ID: mdl-26292099

ABSTRACT

METHODOLOGY/PRINCIPAL FINDINGS: The experimental infection of dogs with TriGAS induced high levels of VNA in the serum, whereas wt RABV infection did not. Dogs infected with TriGAS developed antibodies against the virus including its glycoprotein, whereas dogs infected with DRV-NG11 only developed rabies antibodies that are presumably specific for the nucleoprotein, (N) and not the glycoprotein (G). We show that infection with TriGAS induces early activation of B cells in the draining lymph nodes and persistent activation of DCs and B cells in the blood. On the other hand, infection with DRV-NG11 fails to induce the activation of DCs and B cells and further reduces CD4 T cell production. Further, we show that intrathecal (IT) immunization of TriGAS not only induced high levels of VNA in the serum but also in the CSF while intramuscular (IM) immunization of TriGAS induced VNA only in the serum. In addition, high levels of total protein and WBC were detected in the CSF of IT immunized dogs, indicating the transient enhancement of blood-brain barrier (BBB) permeability, which is relevant to the passage of immune effectors from periphery into the CNS. CONCLUSIONS/SIGNIFICANCE: IM infection of dogs with TriGAS induced the production of serum VNA whereas, IT immunization of TriGAS in dogs induces high levels of VNA in the periphery as well as in the CSF and transiently enhances BBB permeability. In contrast, infection with wt DRV-NG11 resulted in the production of RABV-reactive antibodies but VNA and antibodies specific for G were absent. As a consequence, all of the dogs infected with wt DRV-NG11 succumbed to rabies. Thus the failure to activate protective immunity is one of the important features of RABV pathogenesis in dogs.


Subject(s)
Dog Diseases/immunology , Rabies virus/immunology , Rabies/veterinary , Animals , Antibodies, Viral/immunology , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Dog Diseases/prevention & control , Dog Diseases/virology , Dogs , Rabies/immunology , Rabies/prevention & control , Rabies/virology , Rabies Vaccines/administration & dosage , Rabies Vaccines/genetics , Rabies Vaccines/immunology , Rabies virus/genetics , Rabies virus/physiology , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology
8.
Neurol Neuroimmunol Neuroinflamm ; 2(4): e134, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26236760

ABSTRACT

Neuromyelitis optica (NMO) (and NMO spectrum disorder) is an autoimmune inflammatory disease of the CNS primarily affecting spinal cord and optic nerves. Reliable and sensitive biomarkers for onset, relapse, and progression in NMO are urgently needed because of the heterogeneous clinical presentation, severity of neurologic disability following relapses, and variability of therapeutic response. Detecting aquaporin-4 (AQP4) antibodies (AQP4-IgG or NMO-IgG) in serum supports the diagnosis of seropositive NMO. However, whether AQP4-IgG levels correlate with disease activity, severity, response to therapy, or long-term outcomes is unclear. Moreover, biomarkers for patients with seronegative NMO have yet to be defined and validated. Collaborative international studies hold great promise for establishing and validating biomarkers that are useful in therapeutic trials and clinical management. In this review, we discuss known and potential biomarkers for NMO.

9.
Biochim Biophys Acta ; 1847(8): 729-38, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25917893

ABSTRACT

The mitochondrial citrate-malate exchanger (CIC), a known target of acetylation, is up-regulated in activated immune cells and plays a key role in the production of inflammatory mediators. However, the role of acetylation in CIC activity is elusive. We show that CIC is acetylated in activated primary human macrophages and U937 cells and the level of acetylation is higher in glucose-deprived compared to normal glucose medium. Acetylation enhances CIC transport activity, leading to a higher citrate efflux from mitochondria in exchange with malate. Cytosolic citrate levels do not increase upon activation of cells grown in deprived compared to normal glucose media, indicating that citrate, transported from mitochondria at higher rates from acetylated CIC, is consumed at higher rates. Malate levels in the cytosol are lower in activated cells grown in glucose-deprived compared to normal glucose medium, indicating that this TCA intermediate is rapidly recycled back into the cytosol where it is used by the malic enzyme. Additionally, in activated cells CIC inhibition increases the NADP+/NADPH ratio in glucose-deprived cells; this ratio is unchanged in glucose-rich grown cells due to the activity of the pentose phosphate pathway. Consistently, the NADPH-producing isocitrate dehydrogenase level is higher in activated glucose-deprived as compared to glucose rich cells. These results demonstrate that, in the absence of glucose, activated macrophages increase CIC acetylation to enhance citrate efflux from mitochondria not only to produce inflammatory mediators but also to meet the NADPH demand through the actions of isocitrate dehydrogenase and malic enzyme.


Subject(s)
Carrier Proteins/metabolism , Citric Acid/metabolism , Macrophage Activation/physiology , Malates/metabolism , Mitochondria/metabolism , NADP/metabolism , Acetylation , Biological Transport , Blotting, Western , Cells, Cultured , Cytosol/drug effects , Cytosol/metabolism , Glucose/metabolism , Humans , Immunoprecipitation , Interferon-gamma/pharmacology , Macrophage Activation/drug effects , Mitochondria/drug effects , Sirtuin 3/metabolism , Tumor Necrosis Factor-alpha/pharmacology , U937 Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...