Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 29
Filtrer
Plus de filtres










Base de données
Gamme d'année
2.
Cytokine ; 174: 156457, 2024 02.
Article de Anglais | MEDLINE | ID: mdl-38056248

RÉSUMÉ

The level of IL-2 increases markedly in serum and central nervous system (CNS) of patients with multiple sclerosis (MS) and animals with experimental allergic encephalomyelitis (EAE). However, mechanisms by which IL-2 is induced under autoimmune demyelinating conditions are poorly understood. The present study underlines the importance of IL-12p40 homodimer (p402), the so-called biologically inactive molecule, in inducing the expression of IL-2 in mouse BV-2 microglial cells, primary mouse and human microglia, mouse peritoneal macrophages, RAW264.7 macrophages, and T cells. Interestingly, we found that p402 and IL-12p70 (IL-12), but not IL-23, dose-dependently induced the production of IL-2 and the expression of IL-2 mRNA in microglial cells. Similarly, p402 also induced the activation of IL-2 promoter in microglial cells and RAW264.7 cells. Among various stimuli tested, p402 was the most potent stimulus followed by IFN-γ, bacterial lipopolysaccharide, HIV-1 gp120, and IL-12 in inducing the activation of IL-2 promoter in microglial cells. Moreover, p402, but not IL-23, increased NFATc2 mRNA expression and the transcriptional activity of NFAT. Furthermore, induction of IL-2 mRNA expression by over-expression of p40, but not by p19, cDNA indicated that p40, but not p19, is responsible for the induction of IL-2 mRNA in microglia. Finally, by using primary microglia from IL to 12 receptor ß1 deficient (IL-12Rß1-/-) and IL-12 receptor ß2 deficient (IL-12Rß2-/-) mice, we demonstrate that p402 induces the expression of IL-2 via IL-12Rß1, but not IL-12Rß2. In experimental autoimmune encephalomyelitis, an animal model of MS, neutralization of p402 by mAb a3-1d led to decrease in clinical symptoms and reduction in IL-2 in T cells and microglia. These results delineate a new biological function of p402, which is missing in the so-called autoimmune cytokine IL-23, and raise the possibility of controlling increased IL-2 and the disease process of MS via neutralization of p402.


Sujet(s)
Encéphalomyélite auto-immune expérimentale , Sclérose en plaques , Humains , Animaux , Souris , Interleukine-12/métabolisme , Microglie/métabolisme , Interleukine-2/métabolisme , Macrophages/métabolisme , ARN messager/génétique , ARN messager/métabolisme , Interleukine-23
3.
Nat Commun ; 14(1): 4360, 2023 07 19.
Article de Anglais | MEDLINE | ID: mdl-37468519

RÉSUMÉ

Chemotherapy-induced cardiac damage remains a leading cause of death amongst cancer survivors. Anthracycline-induced cardiotoxicity is mediated by severe mitochondrial injury, but little is known about the mechanisms by which cardiomyocytes adaptively respond to the injury. We observed the translocation of selected mitochondrial tricarboxylic acid (TCA) cycle dehydrogenases to the nucleus as an adaptive stress response to anthracycline-cardiotoxicity in human induced pluripotent stem cell-derived cardiomyocytes and in vivo. The expression of nuclear-targeted mitochondrial dehydrogenases shifts the nuclear metabolic milieu to maintain their function both in vitro and in vivo. This protective effect is mediated by two parallel pathways: metabolite-induced chromatin accessibility and AMP-kinase (AMPK) signaling. The extent of chemotherapy-induced cardiac damage thus reflects a balance between mitochondrial injury and the protective response initiated by the nuclear pool of mitochondrial dehydrogenases. Our study identifies nuclear translocation of mitochondrial dehydrogenases as an endogenous adaptive mechanism that can be leveraged to attenuate cardiomyocyte injury.


Sujet(s)
Cardiopathies , Cellules souches pluripotentes induites , Humains , Cardiotoxicité/métabolisme , Cardiopathies/métabolisme , Cellules souches pluripotentes induites/métabolisme , Antibiotiques antinéoplasiques/pharmacologie , Anthracyclines/pharmacologie , Inhibiteurs de la topoisomérase-II/pharmacologie , Oxidoreductases/métabolisme , Myocytes cardiaques/métabolisme , Doxorubicine/pharmacologie
4.
Sci Rep ; 12(1): 16488, 2022 10 01.
Article de Anglais | MEDLINE | ID: mdl-36182964

RÉSUMÉ

Blood-brain barrier (BBB) dysfunction is emerging as a key pathogenic factor in the progression of Alzheimer's disease (AD), where increased microvascular endothelial permeability has been proposed to play an important role. However, the molecular mechanisms leading to increased brain microvascular permeability in AD are not fully understood. We studied brain endothelial permeability in female APPswe/PS1∆E9 (APP/PS1) mice which constitute a transgenic mouse model of amyloid-beta (Aß) amyloidosis and found that permeability increases with aging in the areas showing the greatest amyloid plaque deposition. We performed an unbiased bulk RNA-sequencing analysis of brain endothelial cells (BECs) in female APP/PS1 transgenic mice. We observed that upregulation of interferon signaling gene expression pathways in BECs was among the most prominent transcriptomic signatures in the brain endothelium. Immunofluorescence analysis of isolated BECs from female APP/PS1 mice demonstrated higher levels of the Type I interferon-stimulated gene IFIT2. Immunoblotting of APP/PS1 BECs showed downregulation of the adherens junction protein VE-cadherin. Stimulation of human brain endothelial cells with interferon-ß decreased the levels of the adherens junction protein VE-cadherin as well as tight junction proteins Occludin and Claudin-5 and increased barrier leakiness. Depletion of the Type I interferon receptor in human brain endothelial cells prevented interferon-ß-induced VE-cadherin downregulation and restored endothelial barrier integrity. Our study suggests that Type I interferon signaling contributes to brain endothelial dysfunction in AD.


Sujet(s)
Maladie d'Alzheimer , Interféron de type I , Maladie d'Alzheimer/métabolisme , Peptides bêta-amyloïdes/métabolisme , Animaux , Barrière hémato-encéphalique/métabolisme , Encéphale/métabolisme , Claudine-5/métabolisme , Modèles animaux de maladie humaine , Cellules endothéliales/métabolisme , Endothélium/métabolisme , Femelle , Humains , Interféron de type I/métabolisme , Interféron bêta/métabolisme , Souris , Souris transgéniques , Occludine/métabolisme , Plaque amyloïde/anatomopathologie , ARN/métabolisme , Récepteur à l'interféron alpha-bêta/métabolisme , Protéines de la jonction serrée/métabolisme
6.
Elife ; 92020 01 16.
Article de Anglais | MEDLINE | ID: mdl-31944177

RÉSUMÉ

Blood vessels are lined by endothelial cells engaged in distinct organ-specific functions but little is known about their characteristic gene expression profiles. RNA-Sequencing of the brain, lung, and heart endothelial translatome identified specific pathways, transporters and cell-surface markers expressed in the endothelium of each organ, which can be visualized at http://www.rehmanlab.org/ribo. We found that endothelial cells express genes typically found in the surrounding tissues such as synaptic vesicle genes in the brain endothelium and cardiac contractile genes in the heart endothelium. Complementary analysis of endothelial single cell RNA-Seq data identified the molecular signatures shared across the endothelial translatome and single cell transcriptomes. The tissue-specific heterogeneity of the endothelium is maintained during systemic in vivo inflammatory injury as evidenced by the distinct responses to inflammatory stimulation. Our study defines endothelial heterogeneity and plasticity and provides a molecular framework to understand organ-specific vascular disease mechanisms and therapeutic targeting of individual vascular beds.


Blood vessels supply nutrients, oxygen and other key molecules to all of the organs in the body. Cells lining the blood vessels, called endothelial cells, regulate which molecules pass from the blood to the organs they supply. For example, brain endothelial cells prevent toxic molecules from getting into the brain, and lung endothelial cells allow immune cells into the lungs to fight off bacteria or viruses.Determining which genes are switched on in the endothelial cells of major organs might allow scientists to determine what endothelial cells do in the brain, heart, and lung, and how they differ; or help scientists deliver drugs to a particular organ. If endothelial cells from different organs switch on different groups of genes, each of these groups of genes can be thought of as a 'genetic signature' that identifies endothelial cells from a specific organ.Now, Jambusaria et al. show that brain, heart, and lung endothelial cells have distinct genetic signatures. The experiments used mice that had been genetically modified to have tags on their endothelial cells. These tags made it possible to isolate RNA ­ a molecule similar to DNA that contains the information about which genes are active ­ from endothelial cells without separating the cells from their tissue of origin. Next, RNA from endothelial cells in the heart, brain and lung was sequenced and analyzed.The results show that each endothelial cell type has a distinct genetic signature under normal conditions and infection-like conditions. Unexpectedly, the experiments also showed that genes that were thought to only be switched on in the cells of specific tissues are also on in the endothelial cells lining the blood vessels of the tissue. For example, genes switched on in brain cells are also active in brain endothelial cells, and genes allowing heart muscle cells to pump are also on in the endothelial cells of the heart blood vessels.The endothelial cell genetic signatures identified by Jambusaria et al. can be used as "postal codes" to target drugs to a specific organ via the endothelial cells that feed it. It might also be possible to use these genetic signatures to build organ-specific blood vessels from stem cells in the laboratory. Future work will try to answer why endothelial cells serving the heart and brain use genes from these organs.


Sujet(s)
Endothélium vasculaire/cytologie , Homéostasie , Inflammation/anatomopathologie , Animaux , Encéphale/métabolisme , Endothélium vasculaire/métabolisme , Endothélium vasculaire/anatomopathologie , Expression des gènes , Humains , Poumon/métabolisme , Souris , Souris de lignée C57BL , Myocarde/métabolisme , ARN messager/génétique
7.
PLoS One ; 15(1): e0227916, 2020.
Article de Anglais | MEDLINE | ID: mdl-31961892

RÉSUMÉ

BACKGROUND: Colorectal cancer remains a deadly cancer due to metastatic disease. To understand the molecular mechanisms of metastasis in colon cancer, we investigated whether the copper chaperone antioxidant-1 (Atox1) protein plays a role in this process. Recent findings indicate that Atox1 protein has transcription factor activities and plays a vital role in cell proliferation in cancer cells. However, the role of Atox1 in metastasis has not been examined. METHODS: Atox1 expression was determined by immunofluorescence in a tissue microarray generated from a spectrum of CRC patients. Subcellular fractionation of colon cancer cell lines SW480 and SW620 cells was used to examine the cellular location of Atox1 in the face of activin A, a cytokine that stimulates colon cancer metastasis. Atox1 expression was genetically manipulated and cellular migration measured through trans-well assay and proliferation measured by colony formation assays. RESULTS: Here we demonstrate that in patients with metastatic colon cancer, there is a significant increase in the expression of nuclear Atox1. Interestingly, the metastatic CRC cell line SW620 has increased nuclear localization of Atox1 compared to its related non-metastatic cell line SW480. Further, inhibition of endogenous Atox1 by siRNA in SW620 decreased colony formation and reactive oxygen species generation via decreased expression of Atox1 targets cyclin D1 and NADPH oxidase subunit p47 phox, respectively. Additionally, overexpression of nuclear-targeted but not copper binding domain-mutated Atox1 in SW480 cells increased colony formation and cell migration that was further augmented by activin A stimulation, a known enhancer of colon cancer metastasis. CONCLUSIONS: Our findings suggest that nuclear Atox1 might be a new therapeutic target as well as a new biomarker for metastatic colorectal cancer.


Sujet(s)
Activines/métabolisme , Carcinomes , Mouvement cellulaire , Tumeurs du côlon , Protéines de transport du cuivre/physiologie , Chaperons moléculaires/physiologie , Carcinomes/métabolisme , Carcinomes/anatomopathologie , Lignée cellulaire tumorale , Prolifération cellulaire , Tumeurs du côlon/métabolisme , Tumeurs du côlon/anatomopathologie , Humains
8.
J Neuroimmune Pharmacol ; 14(3): 503-518, 2019 09.
Article de Anglais | MEDLINE | ID: mdl-31119595

RÉSUMÉ

Glial cell line-derived neurotrophic factor (GDNF) has potent neurotrophic effects and is known to promote the dopaminergic (DA) neuronal survival in cellular and animal models of Parkinson's disease (PD). However, long-term ectopic GDNF delivery is associated with long lasting adverse side effects in PD patients. Therefore, finding safer and effective ways to elevate endogenous GDNF levels is an active area of research. This study underlines the importance of sodium benzoate (NaB), a metabolite of commonly-used spice cinnamon, a food-additive and an FDA-approved drug against hyperammonemia, in stimulating GDNF in primary mouse and human astrocytes. Presence of cAMP response element (CRE) in the Gdnf gene promoter, recruitment of CREB to the Gdnf promoter by NaB and abrogation of NaB-mediated GDNF expression by siRNA knockdown of CREB suggest that NaB induces the transcription of Gdnf via CREB. Finally, oral administration of NaB and cinnamon itself increased the level of GDNF in vivo in the substantia nigra pars compacta (SNpc) of normal as well as MPTP-intoxicated mice. Accordingly, cinnamon and NaB treatment protected tyrosine hydroxylase positive neurons in the SNpc and fibers in the striatum, normalized striatal neurotransmitters, and improved locomotor activities in MPTP-intoxicated Gfapcre mice, but not GdnfΔastro mice lacking GDNF in astrocytes. These findings highlight the importance of astroglial GDNF in cinnamon- and NaB-mediated protection of the nigrostriatum in MPTP mouse model of PD and suggest possible therapeutic potential of cinnamon and NaB in PD patients. Graphical abstract Cinnamon metabolite sodium benzoate (NaB) activates cAMP-response element-binding (CREB) via protein kinase A (PKA) in astrocytes. Activated CREB then binds to cAMP-response element (CRE) present in GDNF gene promoter to stimulate the transcription of GDNF in astrocytes. This astrocytic GDNF leads to nigral trophism and protects dopaminergic neurons from MPTP insult.


Sujet(s)
Antiparkinsoniens/usage thérapeutique , Astrocytes/métabolisme , Cinnamomum zeylanicum/métabolisme , Corps strié/effets des médicaments et des substances chimiques , Facteur neurotrophique dérivé des cellules gliales/physiologie , Syndromes parkinsoniens/traitement médicamenteux , Benzoate de sodium/pharmacologie , Substantia nigra/effets des médicaments et des substances chimiques , Animaux , Antiparkinsoniens/pharmacologie , Biotransformation , Corps strié/métabolisme , Corps strié/anatomopathologie , Protéine de liaison à l'élément de réponse à l'AMP cyclique/métabolisme , Évaluation préclinique de médicament , Comportement d'exploration , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Facteur neurotrophique dérivé des cellules gliales/biosynthèse , Facteur neurotrophique dérivé des cellules gliales/génétique , Humains , Intoxication au MPTP/traitement médicamenteux , Intoxication au MPTP/anatomopathologie , Souris , Souris de lignée C57BL , Syndromes parkinsoniens/anatomopathologie , Pars compacta/effets des médicaments et des substances chimiques , Pars compacta/métabolisme , Pars compacta/anatomopathologie , Écorce , Régions promotrices (génétique)/génétique , Test du rotarod , Substantia nigra/métabolisme , Substantia nigra/anatomopathologie , Facteurs de transcription/métabolisme , Régulation positive/effets des médicaments et des substances chimiques
9.
Sci Rep ; 7(1): 5569, 2017 07 17.
Article de Anglais | MEDLINE | ID: mdl-28717230

RÉSUMÉ

Advanced colorectal cancer (CRC) remains a critical health care challenge worldwide. Various TGF-ß superfamily members are important in colorectal cancer metastasis, but their signaling effects and predictive value have only been assessed in isolation. Here, we examine cross-regulation and combined functions of the two most prominent TGF-ß superfamily members activin and TGF-ß in advanced colorectal cancer. In two clinical cohorts we observed by immune-based assay that combined serum and tissue activin and TGF-ß ligand levels predicts outcome in CRC patients and is superior to single ligand assessment. While TGF-ß growth suppression is independent of activin, TGF-ß treatment leads to increased activin secretion in colon cancer cells and TGF-ß induced cellular migration is dependent on activin, indicating pathway cross-regulation and functional interaction in vitro. mRNA expression of activin and TGF-ß pathway members were queried in silico using the TCGA data set. Coordinated ligand and receptor expression is common in solid tumors for activin and TGF-ß pathway members. In conclusion, activin and TGF-ß are strongly connected signaling pathways that are important in advanced CRC. Assessing activin and TGF-ß signaling as a unit yields important insights applicable to future diagnostic and therapeutic interventions.


Sujet(s)
Activines/génétique , Activines/métabolisme , Tumeurs colorectales/métabolisme , Facteur de croissance transformant bêta/métabolisme , Activines/sang , Lignée cellulaire tumorale , Mouvement cellulaire , Prolifération cellulaire , Tumeurs colorectales/sang , Tumeurs colorectales/anatomopathologie , Femelle , Régulation de l'expression des gènes tumoraux , Réseaux de régulation génique , Humains , Mâle , Stadification tumorale , Pronostic , Transduction du signal , Analyse de survie , Facteur de croissance transformant bêta/sang , Régulation positive
11.
Oncotarget ; 8(23): 37377-37393, 2017 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-28418896

RÉSUMÉ

Colorectal cancer (CRC) remains a common and deadly cancer due to metastatic disease. Activin and TGFB (TGFß) signaling are growth suppressive pathways that exert non-canonical pro-metastatic effects late in CRC carcinogenesis. We have recently shown that activin downregulates p21 via ubiquitination and degradation associated with enhanced cellular migration independent of SMADs. To investigate the mechanism of metastatic activin signaling, we examined activated NFkB signaling and activin ligand expression in CRC patient samples and found a strong correlation. We hypothesize that activation of the E3 ubiquitin ligase MDM2 by NFkB leads to p21 degradation in response to activin treatment. To dissect the link between activin and pro-carcinogenic NFkB signaling and downstream targets, we found that activin but not TGFB induced activation of NFkB leading to increased MDM2 ubiquitin ligase via PI3K. Further, overexpression of wild type p65 NFkB increased MDM2 expression while the NFkB inhibitors NEMO-binding domain (NBD) and Bay11-7082 blocked the activin-induced increase in MDM2. In conclusion, in colon cancer cell migration, activin utilizes NFkB to induce MDM2 activity leading to the degradation of p21 in a PI3K dependent mechanism. This provides new mechanistic knowledge linking activin and NFkB signaling in advanced colon cancer which is applicable to targeted therapeutic interventions.


Sujet(s)
Activines/métabolisme , Tumeurs du côlon/métabolisme , Tumeurs du côlon/anatomopathologie , Tumeurs colorectales/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Carcinogenèse , Lignée cellulaire tumorale , Mouvement cellulaire , Tumeurs colorectales/anatomopathologie , Femelle , Régulation de l'expression des gènes tumoraux , Humains , Mâle , Adulte d'âge moyen , Facteur de transcription NF-kappa B/génétique , Nitriles/pharmacologie , Phosphatidylinositol 3-kinases/métabolisme , Protéines proto-oncogènes c-mdm2/métabolisme , Transduction du signal , Sulfones/pharmacologie , Ubiquitin-protein ligases/métabolisme
12.
Sci Rep ; 6: 26273, 2016 05 20.
Article de Anglais | MEDLINE | ID: mdl-27197561

RÉSUMÉ

BRCA1-associated RING domain protein 1 (BARD1) stabilizes BRCA1 protein by forming a heterodimeric RING-RING complex, and impacts function of BRCA1, including homologous recombination (HR) repair. Although colon cancer cells usually express wild type BRCA1, presence of an oncogenic BARD1 splice variant (SV) in select cancers may render BRCA1 dysfunctional and allow cells to become sensitive to HR targeting therapies. We previously reported association of loss of full-length (FL) BARD1 with poor prognosis in colon cancer as well as expression of various BARD1 SVs with unknown function. Here we show that loss of BARD1 function through the expression of a BARD1 SV, BARD1ß, results in a more malignant phenotype with decreased RAD51 foci formation, reduced BRCA1 E3 ubiquitin ligase activity, and decreased nuclear BRCA1 protein localization. BARD1ß sensitizes colon cancer cells to poly ADP ribose polymerase 1 (PARP-1) inhibition even in a FL BRCA1 background. These results suggest that expression of BARD1ß may serve as a future biomarker to assess suitability of colon cancers for HR targeting with PARP-1 inhibitors in treatment of advanced colon cancer.


Sujet(s)
Tumeurs du côlon/traitement médicamenteux , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Protéines suppresseurs de tumeurs/génétique , Ubiquitin-protein ligases/génétique , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Protéine BRCA1/génétique , Lignée cellulaire tumorale , Tumeurs du côlon/génétique , Recombinaison homologue , Humains , Irinotécan/pharmacologie , Irinotécan/usage thérapeutique , Oxaliplatine/pharmacologie , Oxaliplatine/usage thérapeutique , Inhibiteurs de poly(ADP-ribose) polymérases/usage thérapeutique , Épissage des protéines , Protéines suppresseurs de tumeurs/métabolisme , Ubiquitin-protein ligases/métabolisme , Ubiquitination
13.
PLoS One ; 9(8): e103606, 2014.
Article de Anglais | MEDLINE | ID: mdl-25089827

RÉSUMÉ

Alzheimer's disease (AD), the leading cause of dementia in the aging population, is characterized by the presence of neuritic plaques, neurofibrillary tangles and extensive neuronal apoptosis. Neuritic plaques are mainly composed of aggregates of amyloid-ß (Aß) protein while neurofibrillary tangles are composed of the hyperphosphorylated tau protein. Despite intense investigations, no effective therapy is currently available to halt the progression of this disease. Here, we have undertaken a novel approach to attenuate apoptosis and tau phosphorylation in cultured neuronal cells and in a transgenic animal model of AD. RNS60 is a 0.9% saline solution containing oxygenated nanobubbles that is generated by subjecting normal saline to Taylor-Couette-Poiseuille (TCP) flow under elevated oxygen pressure. In our experiments, fibrillar Aß1-42, but not the reverse peptide Aß42-1, induced apoptosis and cell death in human SHSY5Y neuronal cells. RNS60, but not NS (normal saline), RNS10.3 (TCP-modified saline without excess oxygen) or PNS60 (saline containing excess oxygen without TCP modification), attenuated Aß(1-42)-induced cell death. RNS60 inhibited neuronal cell death via activation of the type 1A phosphatidylinositol-3 (PI-3) kinase-Akt-BAD pathway. Furthermore, RNS60 also decreased Aß(1-42)-induced tau phosphorylation via (PI-3 kinase-Akt)-mediated inhibition of GSK-3ß. Similarly, RNS60 treatment suppressed neuronal apoptosis, attenuated Tau phosphorylation, inhibited glial activation, and reduced the burden of Aß in the hippocampus and protected memory and learning in 5XFAD transgenic mouse model of AD. Therefore, RNS60 may be a promising pharmaceutical candidate in halting or delaying the progression of AD.


Sujet(s)
Maladie d'Alzheimer/anatomopathologie , Maladie d'Alzheimer/physiopathologie , Apoptose/effets des médicaments et des substances chimiques , Mémoire/effets des médicaments et des substances chimiques , Neurones/anatomopathologie , Chlorure de sodium/pharmacologie , Protéines tau/métabolisme , Amyloïde/effets des médicaments et des substances chimiques , Amyloïde/métabolisme , Peptides bêta-amyloïdes/toxicité , Animaux , Modèles animaux de maladie humaine , Régulation négative/effets des médicaments et des substances chimiques , Hippocampe/effets des médicaments et des substances chimiques , Hippocampe/anatomopathologie , Hippocampe/physiopathologie , Humains , Souris , Souris transgéniques , Névroglie/effets des médicaments et des substances chimiques , Névroglie/métabolisme , Névroglie/anatomopathologie , Neurones/effets des médicaments et des substances chimiques , Neurones/enzymologie , Phosphatidylinositol 3-kinases/métabolisme , Phosphorylation/effets des médicaments et des substances chimiques , Plaque amyloïde/anatomopathologie , Protéines proto-oncogènes c-akt/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Apprentissage spatial/effets des médicaments et des substances chimiques
14.
Immunology ; 141(4): 549-63, 2014 Apr.
Article de Anglais | MEDLINE | ID: mdl-24224652

RÉSUMÉ

Interleukin-12 (IL-12) p70 and IL-23 are bioactive cytokines and their biological functions are becoming clear. Increased expression of IL-7 in the central nervous system as well as in peripheral immune cells is associated with multiple sclerosis and experimental allergic encephalomyelitis. Here, we describe the induction of IL-7 in primary mouse and human microglia, BV-2 microglial cells, mouse peritoneal macrophages and astrocytes by IL-12p70. Interestingly, IL-12 strongly induced the expression of IL-7 whereas IL-23 and other p40 family members remained weak inducers of IL-7 in these cell types. Consistently, IL-12, but not IL-23 and other p40 family members, induced IL-7 promoter-driven luciferase activity in microglial cells. Among various stimuli tested, IL-12 emerged as the most potent stimulus followed by bacterial lipopolysaccharide and HIV-1 gp120 in inducing the activation of IL-7 promoter in microglial cells. Furthermore, increase in IL-7 mRNA expression by over-expression of IL-12p35 subunit, but not p40 and IL-23 p19 subunit, confirm that p35, but not p40 and p19, is responsible for the induction of IL-7. Finally, by using primary microglia from IL-12 receptor ß1-deficient (IL-12Rß1(-/-)) and IL-12Rß2(-/-) mice, we demonstrate that IL-12 induces the expression of IL-7 in microglia and macrophages via both IL-12Rß2 and IL-12Rß1. These studies delineate a novel biological function of IL-12 that is absent in IL-23 and other p40 family members.


Sujet(s)
Encéphalomyélite auto-immune expérimentale/métabolisme , Interleukine-12/métabolisme , Interleukine-23/métabolisme , Interleukine-7/métabolisme , Macrophages péritonéaux/métabolisme , Microglie/métabolisme , Sclérose en plaques/métabolisme , Animaux , Astrocytes/immunologie , Astrocytes/métabolisme , Cellules cultivées , Encéphalomyélite auto-immune expérimentale/génétique , Encéphalomyélite auto-immune expérimentale/immunologie , Femelle , Gènes rapporteurs , Protéine d'enveloppe gp120 du VIH/immunologie , Protéine d'enveloppe gp120 du VIH/métabolisme , Humains , Interleukine-12/génétique , Interleukine-12/immunologie , Sous-unité p35 de l'interleukine-12/immunologie , Sous-unité p35 de l'interleukine-12/métabolisme , Sous-unité p40 de l'interleukine-12/immunologie , Sous-unité p40 de l'interleukine-12/métabolisme , Interleukine-23/génétique , Interleukine-23/immunologie , Interleukine-7/génétique , Interleukine-7/immunologie , Lipopolysaccharides/pharmacologie , Luciferases/biosynthèse , Luciferases/génétique , Macrophages péritonéaux/effets des médicaments et des substances chimiques , Macrophages péritonéaux/immunologie , Souris , Souris de lignée C57BL , Souris knockout , Microglie/effets des médicaments et des substances chimiques , Microglie/immunologie , Sclérose en plaques/génétique , Sclérose en plaques/immunologie , Culture de cellules primaires , Régions promotrices (génétique) , ARN messager/métabolisme , Récepteurs à l'interleukine-12/déficit , Récepteurs à l'interleukine-12/génétique , Protéines recombinantes/immunologie , Protéines recombinantes/métabolisme , Transduction du signal , Transfection , Régulation positive
15.
J Neuroimmune Pharmacol ; 8(3): 739-55, 2013 Jun.
Article de Anglais | MEDLINE | ID: mdl-23475543

RÉSUMÉ

This study underlines the importance of cinnamon, a widely-used food spice and flavoring material, and its metabolite sodium benzoate (NaB), a widely-used food preservative and a FDA-approved drug against urea cycle disorders in humans, in increasing the levels of neurotrophic factors [e.g., brain-derived neurotrophic factor (BDNF) and neurotrophin-3 (NT-3)] in the CNS. NaB, but not sodium formate (NaFO), dose-dependently induced the expression of BDNF and NT-3 in primary human neurons and astrocytes. Interestingly, oral administration of ground cinnamon increased the level of NaB in serum and brain and upregulated the levels of these neurotrophic factors in vivo in mouse CNS. Accordingly, oral feeding of NaB, but not NaFO, also increased the level of these neurotrophic factors in vivo in the CNS of mice. NaB induced the activation of protein kinase A (PKA), but not protein kinase C (PKC), and H-89, an inhibitor of PKA, abrogated NaB-induced increase in neurotrophic factors. Furthermore, activation of cAMP response element binding (CREB) protein, but not NF-κB, by NaB, abrogation of NaB-induced expression of neurotrophic factors by siRNA knockdown of CREB and the recruitment of CREB and CREB-binding protein to the BDNF promoter by NaB suggest that NaB exerts its neurotrophic effect through the activation of CREB. Accordingly, cinnamon feeding also increased the activity of PKA and the level of phospho-CREB in vivo in the CNS. These results highlight a novel neutrophic property of cinnamon and its metabolite NaB via PKA - CREB pathway, which may be of benefit for various neurodegenerative disorders.


Sujet(s)
Cinnamomum zeylanicum/métabolisme , Facteurs de croissance nerveuse/biosynthèse , Maladies neurodégénératives/traitement médicamenteux , Maladies neurodégénératives/métabolisme , Benzoate de sodium/usage thérapeutique , Régulation positive/physiologie , Animaux , Cellules cultivées , Foetus , Humains , Souris , Souris de lignée C57BL , Extraits de plantes/isolement et purification , Extraits de plantes/pharmacologie , Extraits de plantes/usage thérapeutique , Benzoate de sodium/pharmacologie , Régulation positive/effets des médicaments et des substances chimiques
16.
ASN Neuro ; 5(1): e00105, 2013 Feb 06.
Article de Anglais | MEDLINE | ID: mdl-23289514

RÉSUMÉ

MS (multiple sclerosis) is the most prevalent autoimmune disease of the CNS (central nervous system) historically characterized as an inflammatory and demyelinating disease. More recently, extensive neuronal pathology has lead to its classification as a neurodegenerative disease as well. While the immune system initiates the autoimmune response it remains unclear how it orchestrates neuronal damage. In our previous studies, using in vitro cultured embryonic neurons, we demonstrated that MBP (myelin basic protein)-specific encephalitogenic CD4 T-cells induce early neuronal damage. In an extension of those studies, here we show that polarized CD4 Th1 and Th17 cells as wells as CD8 T-cells and NK (natural killer) cells induce microtubule destabilization within neurites in a contact-independent manner. Owing to the cytotoxic potential of these immune cells, we isolated the luminal components of lytic granules and determined that they were sufficient to drive microtubule destabilization. Since lytic granules contain cytolytic proteins, we determined that the induction of microtubule destabilization occurred prior to signs of apoptosis. Furthermore, we determined that microtubule destabilization was largely restricted to axons, sparing dendrites. This study demonstrated that lymphocytes with cytolytic activity have the capacity to directly drive MAD (microtubule axonal destabilization) in a bystander manner that is independent of neuronal death.


Sujet(s)
Axones/physiologie , Encéphalomyélite auto-immune expérimentale/anatomopathologie , Lymphocytes/métabolisme , Microtubules/métabolisme , Neurones/physiologie , Animaux , Protéines bactériennes/génétique , Lymphocytes T CD4+ , Mort cellulaire/immunologie , Cellules cultivées , Milieux de culture conditionnés/pharmacologie , Cytokines/métabolisme , Cytokines/pharmacologie , Embryon de mammifère , Encéphalomyélite auto-immune expérimentale/immunologie , Granzymes/déficit , Méthode TUNEL , Cellules tueuses naturelles , Protéines luminescentes/génétique , Lymphocytes/immunologie , Souris , Souris de lignée C57BL , Souris transgéniques , Microscopie confocale , Modèles biologiques , Protéine basique de la myéline/génétique , Neurones/cytologie , Perforine/déficit , Lymphocytes auxiliaires Th1 , Cellules Th17/métabolisme
17.
J Biol Chem ; 287(35): 29529-42, 2012 Aug 24.
Article de Anglais | MEDLINE | ID: mdl-22753407

RÉSUMÉ

Chronic inflammation involving activated microglia and astroglia is becoming a hallmark of many human diseases, including neurodegenerative disorders. Although NF-κB is a multifunctional transcription factor, it is an important target for controlling inflammation as the transcription of many proinflammatory molecules depends on the activation of NF-κB. Here, we have undertaken a novel approach to attenuate NF-κB activation and associated inflammation in activated glial cells. RNS60 is a 0.9% saline solution containing charge-stabilized nanostructures that are generated by subjecting normal saline to Taylor-Couette-Poiseuille (TCP) flow under elevated oxygen pressure. RNS60, but not normal saline, RNS10.3 (TCP-modified saline without excess oxygen), and PNS60 (saline containing excess oxygen without TCP modification) were found to inhibit the production of nitric oxide (NO) and the expression of inducible NO synthase in activated microglia. Similarly, RNS60 also inhibited the expression of inducible NO synthase in activated astroglia. Inhibition of NF-κB activation by RNS60 suggests that RNS60 exerts its anti-inflammatory effect through the inhibition of NF-κB. Interestingly, RNS60 induced the activation of type IA phosphatidylinositol (PI) 3-kinase and Akt and rapidly up-regulated IκBα, a specific endogenous inhibitor of NF-κB. Inhibition of PI 3-kinase and Akt by either chemical inhibitors or dominant-negative mutants abrogated the RNS60-mediated up-regulation of IκBα. Furthermore, we demonstrate that RNS60 induced the activation of cAMP-response element-binding protein (CREB) via the PI 3-kinase-Akt pathway and that RNS60 up-regulated IκBα via CREB. These results describe a novel anti-inflammatory property of RNS60 via type IA PI 3-kinase-Akt-CREB-mediated up-regulation of IκBα, which may be of therapeutic benefit in neurodegenerative disorders.


Sujet(s)
Facteur de transcription NF-kappa B/métabolisme , Protéines de tissu nerveux/métabolisme , Oxygène/pharmacologie , Chlorure de sodium/pharmacologie , Animaux , Lignée cellulaire , Protéine de liaison à l'élément de réponse à l'AMP cyclique/métabolisme , Activation enzymatique/effets des médicaments et des substances chimiques , Activation enzymatique/physiologie , Régulation de l'expression des gènes codant pour des enzymes/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes codant pour des enzymes/physiologie , Humains , Protéines I-kappa B/métabolisme , Inflammation/métabolisme , Souris , Microglie , Inhibiteur alpha de NF-KappaB , Maladies neurodégénératives/traitement médicamenteux , Maladies neurodégénératives/métabolisme , Monoxyde d'azote/biosynthèse , Nitric oxide synthase type II/biosynthèse , Phosphatidylinositol 3-kinases/génétique , Phosphatidylinositol 3-kinases/métabolisme , Protéines proto-oncogènes c-akt/génétique , Protéines proto-oncogènes c-akt/métabolisme
18.
PLoS One ; 7(6): e38113, 2012.
Article de Anglais | MEDLINE | ID: mdl-22723850

RÉSUMÉ

Neuroinflammation and oxidative stress underlie the pathogenesis of various neurodegenerative disorders. Here we demonstrate that sodium phenylbutyrate (NaPB), an FDA-approved therapy for reducing plasma ammonia and glutamine in urea cycle disorders, can suppress both proinflammatory molecules and reactive oxygen species (ROS) in activated glial cells. Interestingly, NaPB also decreased the level of cholesterol but involved only intermediates, not the end product of cholesterol biosynthesis pathway for these functions. While inhibitors of both geranylgeranyl transferase (GGTI) and farnesyl transferase (FTI) inhibited the activation of NF-κB, inhibitor of GGTI, but not FTI, suppressed the production of ROS. Accordingly, a dominant-negative mutant of p21(rac), but not p21(ras), attenuated the production of ROS from activated microglia. Inhibition of both p21(ras) and p21(rac) activation by NaPB in microglial cells suggests that NaPB exerts anti-inflammatory and antioxidative effects via inhibition of these small G proteins. Consistently, we found activation of both p21(ras) and p21(rac)in vivo in the substantia nigra of acute 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of Parkinson's disease. Oral administration of NaPB reduced nigral activation of p21(ras) and p21(rac), protected nigral reduced glutathione, attenuated nigral activation of NF-κB, inhibited nigral expression of proinflammatory molecules, and suppressed nigral activation of glial cells. These findings paralleled dopaminergic neuronal protection, normalized striatal neurotransmitters, and improved motor functions in MPTP-intoxicated mice. Consistently, FTI and GGTI also protected nigrostriata in MPTP-intoxicated mice. Furthermore, NaPB also halted the disease progression in a chronic MPTP mouse model. These results identify novel mode of action of NaPB and suggest that NaPB may be of therapeutic benefit for neurodegenerative disorders.


Sujet(s)
Antioxydants/pharmacologie , Neurones dopaminergiques/effets des médicaments et des substances chimiques , Neurones dopaminergiques/métabolisme , Maladie de Parkinson/métabolisme , Phénylbutyrates/pharmacologie , Animaux , Antioxydants/administration et posologie , Cytokines/métabolisme , Modèles animaux de maladie humaine , Évolution de la maladie , Activation enzymatique/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Humains , Médiateurs de l'inflammation/métabolisme , Mâle , Acide mévalonique/pharmacologie , Souris , Souris de lignée C57BL , Microglie/effets des médicaments et des substances chimiques , Microglie/métabolisme , Activité motrice/effets des médicaments et des substances chimiques , Facteur de transcription NF-kappa B/métabolisme , Névroglie/effets des médicaments et des substances chimiques , Névroglie/métabolisme , Nitric oxide synthase type II/génétique , Nitric oxide synthase type II/métabolisme , Protéine oncogène p21(ras)/métabolisme , Maladie de Parkinson/génétique , Phénylbutyrates/administration et posologie , Espèces réactives de l'oxygène/métabolisme
19.
J Neuroimmune Pharmacol ; 7(3): 544-56, 2012 Sep.
Article de Anglais | MEDLINE | ID: mdl-22661311

RÉSUMÉ

Parkinson's disease (PD) is the most common human neurodegenerative disorder affecting movement, balance, flexibility, and coordination. Despite intense investigation, no effective therapy is available to stop the onset PD or halt its progression. The primate model of PD is considered to be one of the best available models for human PD. Since neuroinflammation plays an important role in the pathogenesis of PD and NF-κB, a proinflammatory transcription factor, participates in the transcription of many proinflammatory molecules, this study evaluates the ability of a peptide corresponding to the NF-κB essential modifier (NEMO)-binding domain (NBD) of IκB kinase (IKK)α or IKKß to protect dopaminergic neurons in hemiparkinsonian monkeys. First, we found that NF-κB was activated within the substantia nigra pars compacta of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-intoxicated hemiparkinsonian monkeys. However, intramuscular injection of wild type NBD (wtNBD) peptide reduced nigral activation of NF-κB and expression of inducible nitric oxide synthase, protected both the nigrostriatal axis and neurotransmitters, and improved motor functions in hemiparkinsonian monkeys. These findings were specific as mutated NBD peptide did not exhibit such effects. These results may help in the translation of NF-κB-based therapy to PD clinics.


Sujet(s)
Facteur de transcription NF-kappa B/métabolisme , Syndromes parkinsoniens/traitement médicamenteux , Syndromes parkinsoniens/métabolisme , Peptides/usage thérapeutique , Séquence d'acides aminés , Animaux , Femelle , I-kappa B Kinase/génétique , I-kappa B Kinase/métabolisme , Macaca mulatta , Données de séquences moléculaires , Facteur de transcription NF-kappa B/génétique , Syndromes parkinsoniens/génétique , Peptides/génétique , Peptides/métabolisme , Facteur de nécrose tumorale alpha/génétique , Facteur de nécrose tumorale alpha/métabolisme
20.
J Neurosci ; 30(38): 12676-89, 2010 Sep 22.
Article de Anglais | MEDLINE | ID: mdl-20861373

RÉSUMÉ

Glial activation plays an important role in the pathogenesis of various neurodegenerative disorders including Alzheimer's disease. However, molecular mechanisms by which activated glia could kill neurons are poorly understood. The present study underlines the importance of neutral sphingomyelinase (N-SMase) in mediating the damaging effect of fibrillar amyloid-ß 1-42 (Aß1-42) peptide-activated astroglia on neurons. In transwell experiments, soluble products released from activated primary human astroglia induced the activation of neutral sphingomyelinase (N-SMase), production of ceramide, and cell death in primary human neurons. Protection of neurons from cytotoxic effects of activated astroglia by antisense knockdown of N-SMase, but not acidic sphingomyelinase (A-SMase), suggests that soluble products released from activated astroglia kill neurons via N-SMase but not A-SMase. Next we examined the role of N-SMase in the activation of human astroglia. Interestingly, knockdown of N-SMase, but not A-SMase, by either antisense oligonucleotides or chemical inhibitor, prevented the induction of proinflammatory molecules [tumor necrosis factor-α, inducible nitric oxide synthase, interleukin-1ß (IL-1ß), and IL-6] and the activation of nuclear factor-κB in Aß1-42-activated astroglia. Subsequently, fibrillar Aß peptides also induced the activation of N-SMase and ceramide in vivo in mouse cortex. Most importantly, antisense knockdown of N-SMase, but not A-SMase, decreased the activation of astroglia and protected neurons from fibrillar Aß toxicity in vivo in the cortex. Together, it is apparent that both the activation of astroglia by Aß and that the cytotoxicity of activated astroglia on neurons depend on N-SMase.


Sujet(s)
Peptides bêta-amyloïdes/métabolisme , Astrocytes/anatomopathologie , Neurones/anatomopathologie , Fragments peptidiques/métabolisme , Sphingomyeline phosphodiesterase/métabolisme , Peptides bêta-amyloïdes/pharmacologie , Animaux , Astrocytes/métabolisme , Mort cellulaire , Cellules cultivées , Céramides/métabolisme , Techniques de coculture , Test de retard de migration électrophorétique , Humains , Immunohistochimie , Souris , Neurones/métabolisme , Fragments peptidiques/pharmacologie , RT-PCR
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE