Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 99
Filter
Add more filters

Publication year range
1.
Cell ; 152(4): 671-2, 2013 Feb 14.
Article in English | MEDLINE | ID: mdl-23415218

ABSTRACT

In this issue, Xiao et al. challenge the notion that cold temperatures promote longevity solely through thermodynamic effects. They show that low temperatures activate a cold-sensitive cation channel, TRPA-1, which triggers a complex signaling pathway in both neurons and nonneuronal cells to extend the lifespan of Caenorhabditis elegans.

2.
Nature ; 576(7785): 138-142, 2019 12.
Article in English | MEDLINE | ID: mdl-31748741

ABSTRACT

Haem is an essential prosthetic group of numerous proteins and a central signalling molecule in many physiologic processes1,2. The chemical reactivity of haem means that a network of intracellular chaperone proteins is required to avert the cytotoxic effects of free haem, but the constituents of such trafficking pathways are unknown3,4. Haem synthesis is completed in mitochondria, with ferrochelatase adding iron to protoporphyrin IX. How this vital but highly reactive metabolite is delivered from mitochondria to haemoproteins throughout the cell remains poorly defined3,4. Here we show that progesterone receptor membrane component 2 (PGRMC2) is required for delivery of labile, or signalling haem, to the nucleus. Deletion of PGMRC2 in brown fat, which has a high demand for haem, reduced labile haem in the nucleus and increased stability of the haem-responsive transcriptional repressors Rev-Erbα and BACH1. Ensuing alterations in gene expression caused severe mitochondrial defects that rendered adipose-specific PGRMC2-null mice unable to activate adaptive thermogenesis and prone to greater metabolic deterioration when fed a high-fat diet. By contrast, obese-diabetic mice treated with a small-molecule PGRMC2 activator showed substantial improvement of diabetic features. These studies uncover a role for PGRMC2 in intracellular haem transport, reveal the influence of adipose tissue haem dynamics on physiology and suggest that modulation of PGRMC2 may revert obesity-linked defects in adipocytes.


Subject(s)
Adipocytes/metabolism , Heme/metabolism , Membrane Proteins/metabolism , Receptors, Progesterone/metabolism , Animals , Homeostasis , Humans , Intracellular Space/metabolism , Male , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Molecular Chaperones/metabolism , Receptors, Progesterone/deficiency , Receptors, Progesterone/genetics , Transcription, Genetic
3.
Phytother Res ; 36(1): 448-461, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34862831

ABSTRACT

Docetaxel (DTX) is used against breast cancer despite its side effects such as toxicity and immunosuppression. Here we investigated the cytotoxic and immunomodulatory effects of the ethanol solution extract of propolis (EEP) in combination with DTX on MCF-7 breast cancer cells and on women's monocyte. The cytotoxic potential of EEP + DTX was assessed by MTT assay and the type of tumor cell death was evaluated by flow cytometry. The effects of EEP + DTX on the migration and invasion of MCF-7 cells were analyzed. Cytokine production by monocytes was assessed by ELISA and the expression of cell surface markers was evaluated by flow cytometry. We also assessed the fungicidal activity of monocytes against Candida albicans and the generation of reactive oxygen species (ROS). Finally, the impact of the supernatants of treated monocytes in the viability, migration, and invasiveness of tumor cells was assessed. EEP enhanced the cytotoxicity of DTX alone against MCF-7 cells by inducing necrosis and inhibiting their migratory ability. EEP + DTX exerted no cytotoxic effects on monocytes and stimulated HLA-DR expression, TNF-α, and IL-6 production, exerted an immunorestorative action in the fungicidal activity, and reduced the oxidative stress. Our findings have practical implications and reveal new insights for complementary medicine.


Subject(s)
Breast Neoplasms , Propolis , Breast Neoplasms/drug therapy , Docetaxel/pharmacology , Female , Humans , MCF-7 Cells , Monocytes , Propolis/pharmacology
4.
Biochemistry ; 59(19): 1793-1799, 2020 05 19.
Article in English | MEDLINE | ID: mdl-32364701

ABSTRACT

PHARC (polyneuropathy, hearing loss, cerebellar ataxia, retinitis pigmentosa, and cataract) is a human neurological disorder caused by deleterious mutations in the ABHD12 gene, which encodes an integral membrane lyso-phosphatidylserine (lyso-PS) lipase. Pharmacological or genetic disruption of ABHD12 leads to higher levels of lyso-PS lipids in human cells and the central nervous system (CNS) of mice. ABHD12 loss also causes rapid rewiring of PS content, resulting in selective increases in the level of arachidonoyl (C20:4) PS and decreases in the levels of other PS species. The biochemical basis for ABHD12-dependent PS remodeling and its pathophysiological significance remain unknown. Here, we show that genetic deletion of the lysophospholipid acyltransferase LPCAT3 blocks accumulation of brain C20:4 PS in mice lacking ABHD12 and concurrently produces hyper-increases in the level of lyso-PS in these animals. These lipid changes correlate with exacerbated auditory dysfunction and brain microgliosis in mice lacking both ABHD12 and LPCAT3. Taken together, our findings reveal that ABHD12 and LPCAT3 coordinately regulate lyso-PS and C20:4 PS content in the CNS and point to lyso-PS lipids as the likely bioactive metabolites contributing to PHARC-related neuropathologies.


Subject(s)
1-Acylglycerophosphocholine O-Acyltransferase/metabolism , Monoacylglycerol Lipases/metabolism , Nervous System Diseases/metabolism , Phosphatidylserines/metabolism , 1-Acylglycerophosphocholine O-Acyltransferase/deficiency , 1-Acylglycerophosphocholine O-Acyltransferase/genetics , Animals , Mice , Mice, Knockout , Molecular Structure , Monoacylglycerol Lipases/deficiency , Monoacylglycerol Lipases/genetics
5.
Brain Behav Immun ; 88: 920-924, 2020 08.
Article in English | MEDLINE | ID: mdl-32276028

ABSTRACT

The human genes for interleukin 13 (IL-13) and its receptor alpha 1 (IL-13Rα1) are in chromosomal regions associated with Parkinson's disease (PD). The interaction of IL-13 with its receptor increases the susceptibility of mouse dopaminergic neurons to oxidative stress. We identified two rare single SNPs in IL13 and IL13RA1 and measured their cytotoxic effects. rs148077750 is a missense leucine to proline substitution in IL13. It was found in individuals with early onset PD and no other known monogenic forms of the disease and is significantly linked with PD (Fisher's exact test: p-value = 0.01, odds ratio = 14.2). rs145868092 is a leucine to phenylalanine substitution in IL13RA1 affecting a residue critical for IL-13 binding. Both mutations increased the cytotoxic activity of IL-13 on human SH-SY5Y neurons exposed to sublethal doses of hydrogen peroxide, t-butyl hydroperoxide or RLS3, an inducer of ferroptosis. Our data show that both rs148077750 and rs145868092 conferred a gain-of-function that may increase the risk of developing PD.


Subject(s)
Interleukin-13 , Parkinson Disease , Animals , Interleukin-13/genetics , Interleukin-13 Receptor alpha1 Subunit/genetics , Mice , Oxidative Stress/genetics , Parkinson Disease/genetics , Polymorphism, Single Nucleotide
6.
Proc Natl Acad Sci U S A ; 114(36): 9737-9742, 2017 09 05.
Article in English | MEDLINE | ID: mdl-28827349

ABSTRACT

Changes in body temperature can profoundly affect survival. The dramatic longevity-enhancing effect of cold has long been known in organisms ranging from invertebrates to mammals, yet the underlying mechanisms have only recently begun to be uncovered. In the nematode Caenorhabditis elegans, this process is regulated by a thermosensitive membrane TRP channel and the DAF-16/FOXO transcription factor, but in more complex organisms the underpinnings of cold-induced longevity remain largely mysterious. We report that, in Drosophila melanogaster, variation in ambient temperature triggers metabolic changes in protein translation, mitochondrial protein synthesis, and posttranslational regulation of the translation repressor, 4E-BP (eukaryotic translation initiation factor 4E-binding protein). We show that 4E-BP determines Drosophila lifespan in the context of temperature changes, revealing a genetic mechanism for cold-induced longevity in this model organism. Our results suggest that the 4E-BP pathway, chiefly thought of as a nutrient sensor, may represent a master metabolic switch responding to diverse environmental factors.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Peptide Initiation Factors/metabolism , Aging/genetics , Aging/metabolism , Animals , Animals, Genetically Modified , Cold Temperature , Drosophila Proteins/deficiency , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Female , Gene Knockout Techniques , Genes, Insect , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Longevity/genetics , Longevity/physiology , Male , Mitochondrial Proteins/biosynthesis , Peptide Initiation Factors/deficiency , Peptide Initiation Factors/genetics , Protein Biosynthesis , Protein Processing, Post-Translational , Temperature
7.
Proc Natl Acad Sci U S A ; 114(36): 9731-9736, 2017 09 05.
Article in English | MEDLINE | ID: mdl-28827363

ABSTRACT

When food resources are scarce, endothermic animals can lower core body temperature (Tb). This phenomenon is believed to be part of an adaptive mechanism that may have evolved to conserve energy until more food becomes available. Here, we found in the mouse that the insulin-like growth factor 1 receptor (IGF-1R) controls this response in the central nervous system. Pharmacological or genetic inhibition of IGF-1R enhanced the reduction of temperature and of energy expenditure during calorie restriction. Full blockade of IGF-1R affected female and male mice similarly. In contrast, genetic IGF-1R dosage was effective only in females, where it also induced transient and estrus-specific hypothermia in animals fed ad libitum. These effects were regulated in the brain, as only central, not peripheral, pharmacological activation of IGF-1R prevented hypothermia during calorie restriction. Targeted IGF-1R knockout selectively in forebrain neurons revealed that IGF signaling also modulates calorie restriction-dependent Tb regulation in regions rostral of the canonical hypothalamic nuclei involved in controlling body temperature. In aggregate, these data identify central IGF-1R as a mediator of the integration of nutrient and temperature homeostasis. They also show that calorie restriction, IGF-1R signaling, and body temperature, three of the main regulators of metabolism, aging, and longevity, are components of the same pathway.


Subject(s)
Caloric Restriction/adverse effects , Hypothermia/physiopathology , Receptor, IGF Type 1/physiology , Aging/physiology , Animals , Energy Metabolism/physiology , Female , Gene Dosage , Homeostasis/physiology , Hypothermia/etiology , Hypothermia/prevention & control , Longevity/physiology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Podophyllotoxin/analogs & derivatives , Podophyllotoxin/pharmacology , Receptor, IGF Type 1/antagonists & inhibitors , Receptor, IGF Type 1/genetics , Sex Characteristics , Signal Transduction/physiology
8.
Alcohol Clin Exp Res ; 43(12): 2547-2558, 2019 12.
Article in English | MEDLINE | ID: mdl-31589333

ABSTRACT

BACKGROUND: Adolescence is a critical period for neural development, and alcohol exposure during adolescence can lead to an elevated risk for health consequences as well as alcohol use disorders. Clinical and experimental data suggest that chronic alcohol exposure may produce immunomodulatory effects that can lead to the activation of pro-inflammatory cytokine pathways as well as microglial markers. The present study evaluated, in brain and blood, the effects of adolescent alcohol exposure and withdrawal on microglia and on the most representative pro- and anti-inflammatory cytokines and major chemokines that can contribute to the establishing of a neuroinflammatory environment. METHODS: Wistar rats (males, n = 96) were exposed to ethanol (EtOH) vapors, or air control, for 5 weeks over adolescence (PD22-PD58). Brains and blood samples were collected at 3 time points: (i) after 35 days of vapor/air exposure (PD58); (ii) after 1 day of withdrawal (PD59), and (iii) 28 days after withdrawal (PD86). The ionized calcium-binding adapter molecule 1 (Iba-1) was used to index microglial activation, and cytokine/chemokine responses were analyzed using magnetic bead panels. RESULTS: After 35 days of adolescent vapor exposure, a significant increase in Iba-1 immunoreactivity was seen in amygdala, frontal cortex, hippocampus, and substantia nigra. However, Iba-1 density returned to control levels at both 1 day and 28 days of withdrawal except in the hippocampus where Iba-1 density was significantly lower than controls. In serum, adolescent EtOH exposure induced a reduction in IL-13 and an increase in fractalkine at day 35. After 1 day of withdrawal, IL-18 was reduced, and IP-10 was elevated, whereas both IP-10 and IL-10 were elevated at 28 days following withdrawal. In the frontal cortex, adolescent EtOH exposure induced an increase in IL-1ß at day 35, and 28 days of withdrawal, and IL-10 was increased after 28 days of withdrawal. CONCLUSION: These data demonstrate that EtOH exposure during adolescence produces significant microglial activation; however, inflammatory markers seen in the blood appear to differ from those observed in the brain.


Subject(s)
Brain/metabolism , Cytokines/metabolism , Ethanol/adverse effects , Substance Withdrawal Syndrome/metabolism , Age Factors , Animals , Calcium-Binding Proteins/metabolism , Cytokines/blood , Male , Microfilament Proteins/metabolism , Microglia/metabolism , Rats , Substance Withdrawal Syndrome/blood , Time Factors
9.
Proc Natl Acad Sci U S A ; 113(1): 26-33, 2016 Jan 05.
Article in English | MEDLINE | ID: mdl-26668358

ABSTRACT

Diacylglycerol lipases (DAGLα and DAGLß) convert diacylglycerol to the endocannabinoid 2-arachidonoylglycerol. Our understanding of DAGL function has been hindered by a lack of chemical probes that can perturb these enzymes in vivo. Here, we report a set of centrally active DAGL inhibitors and a structurally related control probe and their use, in combination with chemical proteomics and lipidomics, to determine the impact of acute DAGL blockade on brain lipid networks in mice. Within 2 h, DAGL inhibition produced a striking reorganization of bioactive lipids, including elevations in DAGs and reductions in endocannabinoids and eicosanoids. We also found that DAGLα is a short half-life protein, and the inactivation of DAGLs disrupts cannabinoid receptor-dependent synaptic plasticity and impairs neuroinflammatory responses, including lipopolysaccharide-induced anapyrexia. These findings illuminate the highly interconnected and dynamic nature of lipid signaling pathways in the brain and the central role that DAGL enzymes play in regulating this network.


Subject(s)
Arachidonic Acids/metabolism , Brain/drug effects , Diglycerides/metabolism , Endocannabinoids/metabolism , Enzyme Inhibitors/pharmacology , Glycerides/metabolism , Lipoprotein Lipase/antagonists & inhibitors , Neuronal Plasticity/drug effects , Animals , Brain/enzymology , Brain/metabolism , Enzyme Inhibitors/chemistry , Lipoprotein Lipase/metabolism , Male , Mice , Mice, Inbred C57BL , Receptors, Cannabinoid/metabolism , Signal Transduction/drug effects
10.
Phytother Res ; 33(3): 591-601, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30488503

ABSTRACT

Osteosarcoma (OSA) is a type of bone cancer showing an aggressive biological behavior with metastatic progression. Because propolis potential for the development of new antitumoral drugs has been indicated, we evaluated the chemical composition of Colombian propolis samples and the mechanisms involved in their cytotoxic effects on OSA cells. The chemical composition was analyzed by GC-MS and the DPPH free radical scavenging activity was measured. Cluster and principal components analysis were used to establish an association with their inhibitory concentration 50% (IC50 ). Cell viability was analyzed by MTT assay; apoptosis was determined by flow cytometry; mitochondrial membrane permeability and reactive oxygen species were evaluated by rhodamine 123 and DCFH-DA. Transwell assay was used to evaluate the invasiveness of propolis-treated cells. Samples were grouped: Cluster 1 contained diterpenes and benzophenones and showed the highest antiradical activity; Cluster 2 was characterized by triterpenes, fatty acid, and diterpenes. Usm contained diterpenes and triterpenes different of the other samples and Sil contained triterpenes and flavonoids. Apoptosis, mitochondrial membrane alteration, and suppression of cell invasion were the main mechanisms involved in the inhibition of OSA cells in vitro, suggesting the potential of Colombian propolis to discover new antitumor drugs.


Subject(s)
Apoptosis/drug effects , Bone Neoplasms/pathology , Osteosarcoma/pathology , Propolis/chemistry , Propolis/pharmacology , Animals , Antioxidants/chemistry , Antioxidants/pharmacology , Bone Neoplasms/metabolism , Cell Survival/drug effects , Colombia , Cytotoxins/chemistry , Cytotoxins/pharmacology , Diterpenes/chemistry , Diterpenes/pharmacology , Dog Diseases/metabolism , Dog Diseases/pathology , Dogs , Flavonoids/chemistry , Flavonoids/pharmacology , Gas Chromatography-Mass Spectrometry , Osteosarcoma/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Triterpenes/chemistry , Triterpenes/pharmacology , Tumor Cells, Cultured
11.
Cytokine ; 103: 114-120, 2018 03.
Article in English | MEDLINE | ID: mdl-28969943

ABSTRACT

The majority of Parkinson's disease (PD) cases are sporadic with only about 10% of PD patients having a family history of the disease suggesting that this neurodegenerative disorder is the result of both environmental and genetic factors. Both oxidative stress and neuroinflammation are thought to contribute to PD. Previously, we showed that the activation of interleukin 13 receptor alpha 1 (IL-13Rα1) increases the sensitivity of dopaminergic neurons to oxidative damage both in cultured cells and in animals. In this study, we investigated the pathways involved in the IL-13-mediated potentiation of oxidative stress-induced dopaminergic cell death using a combination of cell survival assays and Western blotting with appropriate antibodies. In addition, siRNA was used to examine the role of 4E-BP1 in this cell toxicity paradigm. We show that activation of both the Jak-Stat and PI3 kinase-mTOR pathways play key roles in the promotion of cell death by IL-13 in the presence of mild oxidative stress. The Jak 1/2 inhibitor ruxolitinib, the mTOR inhibitor rapamycin and the PI3 kinase inhibitor LY294002 all prevented the potentiation of cell death by IL-13. Moreover, 4E-BP1, a target of mTOR, appeared to mediate the protective effects of rapamycin. Together, these results indicate that multiple signaling pathways downstream of IL-13Rα1 activation play a role in the toxic effects of IL-13 in dopaminergic neurons in the presence of mild oxidative stress and suggest that any of these pathways might provide potential targets for the treatment of PD.


Subject(s)
Dopaminergic Neurons/metabolism , Interleukin-13/metabolism , Oxidative Stress , Signal Transduction , Animals , Cell Death/drug effects , Cell Line , Chromones/pharmacology , Dopaminergic Neurons/pathology , Interleukin-13/antagonists & inhibitors , Interleukin-13 Receptor alpha1 Subunit/metabolism , Janus Kinases/metabolism , Mice , Morpholines/pharmacology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Parkinson Disease/therapy , Phosphatidylinositol 3-Kinases/metabolism , STAT Transcription Factors/metabolism , TOR Serine-Threonine Kinases/metabolism
12.
J Neurosci ; 36(18): 5170-80, 2016 05 04.
Article in English | MEDLINE | ID: mdl-27147667

ABSTRACT

UNLABELLED: The proinflammatory cytokine IL-18 has central anorexigenic effects and was proposed to contribute to loss of appetite observed during sickness. Here we tested in the mouse the hypothesis that IL-18 can decrease food intake by acting on neurons of the bed nucleus of the stria terminalis (BST), a component of extended amygdala recently shown to influence feeding via its projections to the lateral hypothalamus (LH). We found that both subunits of the heterodimeric IL-18 receptor are highly expressed in the BST and that local injection of recombinant IL-18 (50 ng/ml) significantly reduced c-fos activation and food intake for at least 6 h. Electrophysiological experiments performed in BST brain slices demonstrated that IL-18 strongly reduces the excitatory input on BST neurons through a presynaptic mechanism. The effects of IL-18 are cell-specific and were observed in Type III but not in Type I/II neurons. Interestingly, IL-18-sensitve Type III neurons were recorded in the juxtacapsular BST, a region that contains BST-LH projecting neurons. Reducing the excitatory input on Type III GABAergic neurons, IL-18 can increase the firing of glutamatergic LH neurons through a disinhibitory mechanism. Imbalance between excitatory and inhibitory activity in the LH can induce changes in food intake. Effects of IL-18 were mediated by the IL-18R because they were absent in neurons from animals null for IL-18Rα (Il18ra(-/-)), which lack functional IL-18 receptors. In conclusion, our data show that IL-18 may inhibit feeding by inhibiting the activity of BST Type III GABAergic neurons. SIGNIFICANCE STATEMENT: Loss of appetite during sickness is a common and often debilitating phenomenon. Although proinflammatory cytokines are recognized as mediators of these anorexigenic effects, their mechanism and sites of action remain poorly understood. Here we show that interleukin 18, an anorexigenic cytokine, can act on neurons of the bed nucleus of the stria terminalis to reduce food intake via the IL-18 receptor. The findings identify a site and a mode of action that indicate targets for the treatment of cachexia or other eating disorders.


Subject(s)
Feeding Behavior/physiology , Interleukin-18/physiology , Septal Nuclei/physiology , Animals , Electrophysiological Phenomena/physiology , Hypothalamic Area, Lateral/physiology , Interleukin-18/biosynthesis , Interleukin-18/genetics , Interleukin-18 Receptor alpha Subunit/genetics , Interleukin-18 Receptor alpha Subunit/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/physiology , Recombinant Proteins/pharmacology , Synapses/drug effects , gamma-Aminobutyric Acid/physiology
13.
J Neuroinflammation ; 14(1): 88, 2017 04 21.
Article in English | MEDLINE | ID: mdl-28427412

ABSTRACT

BACKGROUND: The majority of Parkinson's disease (PD) cases are sporadic and idiopathic suggesting that this neurodegenerative disorder is the result of both environmental and genetic factors. Stress and neuroinflammation are among the factors being investigated for their possible contributions to PD. Experiments in rodents showed that severe chronic stress can reduce the number of dopaminergic neurons in the substantia nigra pars compacta (SNc); the same cells that are lost in PD. These actions are at least in part mediated by increased oxidative stress. Here, we tested the hypothesis that the interleukin-13 receptor alpha 1 (IL-13Rα1), a cytokine receptor whose activation increases the vulnerability of dopaminergic neurons to oxidative damage, participates in the stress-dependent damage of these neurons. METHODS: Mice were subject to daily sessions of 8 h (acute) stress for 16 weeks (5 days a week), a procedure previously showed to induce loss of dopaminergic neurons in the SNc. The source and the kinetics of interleukin-13 (IL-13), the endogenous ligand of IL-13Rα1, were evaluated 0, 1, 3, 6, and 8 h and at 16 weeks of stress. Identification of IL-13 producing cell-type was performed by immunofluorescent and by in situ hybridization experiments. Markers of oxidative stress, microglia activation, and the number of dopaminergic neurons in IL-13Rα1 knock-out animals (Il13ra1 Y/ - ) and their wild-type littermates (Il13ra1 Y/+ ) were evaluated at 16 weeks of stress and at 20 weeks, following a 4 week non-stressed period and compared to non-stressed mice. RESULTS: IL-13 was expressed in microglial cells within the SN and in a fraction of the tyrosine hydroxylase-positive neurons in the SNc. IL-13 levels were elevated during daily stress and peaked at 6 h. 16 weeks of chronic restraint stress significantly reduced the number of SNc dopaminergic neurons in Il13ra1 Y/+ mice. Neuronal loss at 16 weeks was significantly lower in Il13ra1 Y/- mice. However, the loss of dopaminergic neurons measured at 20 weeks, after 4 weeks of non-stress following the 16 weeks of stress, was similar in Il13ra1 Y/+ and Il13ra1 Y/- mice. CONCLUSIONS: IL-13, a cytokine previously demonstrated to increase the susceptibility of SNc dopaminergic neurons to oxidative stress, is elevated in the SN by restraint stress. Lack of IL-13Rα1 did not prevent nor halted but delayed neuronal loss in the mouse model of chronic restraint stress. IL-13/IL-13Rα1 may represent a target to reduce the rate of DA neuronal loss that can occur during severe chronic restraint stress.


Subject(s)
Dopaminergic Neurons/metabolism , Interleukin-13 Receptor alpha1 Subunit/deficiency , Oxidative Stress/physiology , Stress, Psychological/metabolism , Animals , Cell Count/methods , Dopaminergic Neurons/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Stress, Psychological/pathology , Substantia Nigra/metabolism , Substantia Nigra/pathology
15.
Mediators Inflamm ; 2017: 7582437, 2017.
Article in English | MEDLINE | ID: mdl-28811681

ABSTRACT

Osteopontin (OPN) is a molecule that is common in central nervous system (CNS) pathologies, which participates in the activation, migration, and survival of inflammatory cells. However, the mechanisms by which OPN modulates inflammatory pathways are not clear. To understand the role of OPN in CNS viral infections, we used a lethal mouse model of West Nile virus (WNV), characterized by the injection of high doses of the Eg101 strain of WNV, causing the increase of OPN levels in the brain since early time points. To measure the impact of OPN in neuropathogenesis and resistance, we compared C57BI/6 WT with mice lacking the OPN gene (OPN KO). OPN KO presented a significantly higher mortality compared to WT mice, detectable since day 5 pi. Our data suggests that OPN expression at early time points may provide protection against viral spread in the CNS by negatively controlling the type I IFN-sensitive, caspase 1-dependent inflammasome, while promoting an alternative caspase 8-associated pathway, to control the apoptosis of infected cells during WNV infection in the CNS. Overall, we conclude that the expression of OPN maintains a critical threshold in the innate immune response that controls apoptosis and lethal viral spread in early CNS infection.


Subject(s)
Central Nervous System/metabolism , Inflammasomes/metabolism , Osteopontin/metabolism , West Nile Fever/metabolism , West Nile Fever/pathology , Animals , Brain/immunology , Brain/metabolism , Cell Death/immunology , Central Nervous System/immunology , Inflammasomes/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteopontin/genetics , West Nile Fever/immunology
16.
Immunol Invest ; 45(5): 420-38, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27105208

ABSTRACT

Bloodstream infections caused by Candida species are responsible for high morbidity and mortality, and diabetes mellitus (DM) is an important underlying disease in candidemia episodes. Although DM patients show an enhanced proinflammatory profile, they are highly susceptible to mycobacterial and mycotic infections. Attempting to understand this paradox, we investigated if imbalanced macrophage and dendritic cell (DC) activations could be associated to high incidence and/or severity of Candida albicans infection in the hypoinsulinemia-hyperglycemia (HH) milieu. HH alloxan-induced mice were infected with C. albicans and peritoneal aderent phagocytes were co-cultured with or without lipopolyssaccharide or heat-killed C. albicans, and the production of cytotoxic metabolites, cytokines, and chemokines was evaluated. We also evaluated the surface expression of MHC-II and CD86 in splenic DCs. Our findings showed that both uninfected and C. albicans-infected HH mice showed less production of CCL2 and reduced expression of CD86 by peritoneal phagocytes and splenic DCs, respectively.


Subject(s)
Candida albicans/immunology , Candidiasis/microbiology , Dendritic Cells/immunology , Diabetes Mellitus, Experimental/immunology , Macrophages/immunology , Alloxan/toxicity , Animals , B7-2 Antigen/metabolism , Brazil , Cells, Cultured , Chemokine CCL2/metabolism , Coculture Techniques , Dendritic Cells/metabolism , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/microbiology , Genes, MHC Class II/immunology , Macrophages/metabolism , Male , Mice
18.
Brain Behav Immun ; 37: 45-53, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24316258

ABSTRACT

OBJECTIVE: The proinflammatory cytokine interleukin-18 (IL-18) putatively modulates food intake and energy metabolism, but the effects of IL-18 in high-fat diet fed animals are unknown. Whether IL-18 alters basal metabolic rate or metabolic processes of living is unknown. Here, we tested the hypothesis that IL-18 modulates weight gain, energy intake, whole-body energy expenditure, and utilization of lipid as a fuel substrate in high-fat diet fed mice. METHODS: Food intake, whole-body metabolism, and motor activity of IL-18 knockout mice were compared to those of wildtype littermates; anorectic effects of intracerebroventricular IL-18 administration were compared between IL-18 receptor knockout, IL-18/IL-18R knockout and wildtype mice. RESULTS: Chow-reared IL-18 knockout mice were overweight at 6 months of age and then gained excess weight on both low-fat and high-fat diets, ate more high-fat diet, and showed reduced whole-body energy expenditure and increased respiratory exchange ratios. Reductions in energy expenditure of IL-18 knockout mice were seen across fasting vs. feeding conditions, low- vs. high-fat diets, high vs. low levels of physical activity and times of day, suggesting actions on basal metabolic rate. The circadian amplitude of energy expenditure, but not respiratory exchange ratio, food intake, or motor activity, also was blunted in IL-18 knockout mice. Central IL-18 administration reduced high-fat diet intake in wildtype mice, but not in mice lacking the IL-18 receptor. CONCLUSION: The loss-of-function results support the hypothesis that endogenous IL-18 suppresses appetite and promote energy expenditure and lipid fuel substrate utilization not only during sickness, but also in healthy adults consuming high-fat diets.


Subject(s)
Interleukin-18/physiology , Receptors, Interleukin-18/physiology , Animals , Diet, High-Fat , Eating , Energy Metabolism , Female , Interleukin-18/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/physiology , Mutation , Receptors, Interleukin-18/genetics , Weight Gain
19.
Brain Behav Immun ; 40: 85-94, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24603356

ABSTRACT

Interleukin (IL)-18 is a cytokine previously demonstrated to participate in neuroinflammatory processes. Since the components of the IL-18 receptor complex are expressed in neurons throughout the brain, IL-18 is also believed to directly influence neuronal function. Here we tested this hypothesis on mouse hippocampal neurons by measuring the effects of IL-18 on three pathways previously shown to be regulated by this cytokine in non-neuronal cells: the MAPK pathways, p38 and ERK1/2 MAPKs, STAT3 and NF-κB. Experiments were carried out in vitro using the immortalized hippocampal neuronal line HT-22 or in vivo following i.c.v. injection with recombinant mouse IL-18. We showed that IL-18 did not activate NF-κB in HT-22 cells whereas it induced a rapid (within 15min) activation of the MAPK pathways. Moreover, we demonstrated that IL-18 treatment enhanced P-STAT3 (Tyr705)/STAT3 ratio in the nucleus of HT-22 cells after 30-60min of exposure. A similar increase in P-STAT3 (Tyr705)/STAT3 ratio was observed in the whole hippocampus one hour after i.c.v. injection. These data demonstrate that IL-18 can act directly on neuronal cells affecting the STAT3 pathway; therefore, possibly regulating the expression of specific genes within the hippocampus. This effect may help to explain some of the IL-18-induced effects on synaptic plasticity and functionality within the hippocampal system.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Hippocampus/metabolism , Interleukin-18/metabolism , NF-kappa B/metabolism , Neurons/metabolism , STAT3 Transcription Factor/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cells, Cultured , Hippocampus/drug effects , Interleukin-18/pharmacology , Male , Mice , Mice, Inbred C57BL , Receptors, Interleukin-18/metabolism , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Signal Transduction
20.
J Immunol ; 189(12): 5498-502, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23169588

ABSTRACT

Inflammation and its mediators, including cytokines and reactive oxygen species, are thought to contribute to neurodegeneration. In the mouse brain, we found that IL-13Rα1 was expressed in the dopaminergic (DA) neurons of the substantia nigra pars compacta, which are preferentially lost in human Parkinson's disease. Mice deficient for Il13ra1 exhibited resistance to loss of DA neurons in a model of chronic peripheral inflammation using bacterial LPS. IL-13, as well as IL-4, potentiated the cytotoxic effects of t-butyl hydroperoxide and hydrogen peroxide on mouse DA MN9D cells. Collectively, our data indicate that expression of IL-13Rα1 on DA neurons can increase their susceptibility to oxidative stress-mediated damage, thereby contributing to their preferential loss. In humans, Il13ra1 lies on the X chromosome within the PARK12 locus of susceptibility to Parkinson's disease, suggesting that IL-13Rα1 may have a role in the pathogenesis of this neurodegenerative disease.


Subject(s)
Dopaminergic Neurons/immunology , Dopaminergic Neurons/metabolism , Interleukin-13 Receptor alpha1 Subunit/biosynthesis , Lipopolysaccharides/toxicity , Oxidative Stress/immunology , Animals , Cell Death/genetics , Cell Death/immunology , Chronic Disease , Disease Models, Animal , Dopaminergic Neurons/pathology , Genetic Diseases, X-Linked/genetics , Genetic Predisposition to Disease/etiology , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interleukin-13 Receptor alpha1 Subunit/deficiency , Interleukin-13 Receptor alpha1 Subunit/genetics , Lipopolysaccharides/administration & dosage , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress/genetics , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/pathology
SELECTION OF CITATIONS
SEARCH DETAIL