Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Front Pharmacol ; 13: 888222, 2022.
Article in English | MEDLINE | ID: mdl-35721207

ABSTRACT

Stroke is the second leading cause of death worldwide following coronary heart disease. Despite significant efforts to find effective treatments to reduce neurological damage, many patients suffer from sequelae that impair their quality of life. For this reason, the search for new therapeutic options for the treatment of these patients is a priority. Glial cells, including microglia, astrocytes and oligodendrocytes, participate in crucial processes that allow the correct functioning of the neural tissue, being actively involved in the pathophysiological mechanisms of ischemic stroke. Although the exact mechanisms by which glial cells contribute in the pathophysiological context of stroke are not yet completely understood, they have emerged as potentially therapeutic targets to improve brain recovery. The endocannabinoid system has interesting immunomodulatory and protective effects in glial cells, and the pharmacological modulation of this signaling pathway has revealed potential neuroprotective effects in different neurological diseases. Therefore, here we recapitulate current findings on the potential promising contribution of the endocannabinoid system pharmacological manipulation in glial cells for the treatment of ischemic stroke.

2.
Sci Rep ; 12(1): 7895, 2022 05 12.
Article in English | MEDLINE | ID: mdl-35551497

ABSTRACT

Studies have shown that anodically grown TiO2 nanotubes (TNTs) exhibit excellent biocompatibility. However, TiO2 nanowires (TNWs) have received less attention. The objective of this study was to investigate the proliferation of osteoblast precursor cells on the surfaces of TNWs grown by electrochemical anodization of a Ti-35Nb-7Zr-5Ta (TNZT) alloy. TNT and flat TNZT surfaces were used as control samples. MC3T3-E1 cells were cultured on the surfaces of the samples for up to 5 days, and cell viability and proliferation were investigated using fluorescence microscopy, colorimetric assay, and scanning electron microscopy. The results showed lower cell proliferation rates on the TNW surface compared to control samples without significant differences in cell survival among experimental conditions. Contact angles measurements showed a good level of hydrophilicity for the TNWs, however, their relatively thin diameter and their high density may have affected cell proliferation. Although more research is necessary to understand all the parameters affecting biocompatibility, these TiO2 nanostructures may represent promising tools for the treatment of bone defects and regeneration of bone tissue, among other applications.


Subject(s)
Nanotubes , Nanowires , Alloys/chemistry , Cell Proliferation , Microscopy, Electron, Scanning , Nanotubes/chemistry , Osteoblasts , Surface Properties , Titanium/chemistry , Titanium/pharmacology
3.
J Neuroinflammation ; 19(1): 93, 2022 Apr 12.
Article in English | MEDLINE | ID: mdl-35413853

ABSTRACT

BACKGROUND: Friedreich's ataxia is a rare hereditary neurodegenerative disease caused by decreased levels of the mitochondrial protein frataxin. Similar to other neurodegenerative pathologies, previous studies suggested that astrocytes might contribute to the progression of the disease. To fully understand the mechanisms underlying neurodegeneration in Friedreich's ataxia, we investigated the reactivity status and functioning of cultured human astrocytes after frataxin depletion using an RNA interference-based approach and tested the effect of pharmacologically modulating the SHH pathway as a novel neuroprotective strategy. RESULTS: We observed loss of cell viability, mitochondrial alterations, increased autophagy and lipid accumulation in cultured astrocytes upon frataxin depletion. Besides, frataxin-deficient cells show higher expression of several A1-reactivity markers and release of pro-inflammatory cytokines. Interestingly, most of these defects were prevented by chronically treating the cells with the smoothened agonist SAG. Furthermore, in vitro culture of neurons with conditioned medium from frataxin-deficient astrocytes results in a reduction of neuronal survival, neurite length and synapse formation. However, when frataxin-deficient astrocytes were chronically treated with SAG, we did not observe these alterations in neurons. CONCLUSIONS: Our results demonstrate that the pharmacological activation of the SHH pathway could be used as a target to modulate astrocyte reactivity and neuron-glia interactions to prevent neurodegeneration in Friedreich's ataxia.


Subject(s)
Friedreich Ataxia , Neurodegenerative Diseases , Neurotoxicity Syndromes , Astrocytes/metabolism , Friedreich Ataxia/drug therapy , Friedreich Ataxia/genetics , Friedreich Ataxia/pathology , Humans , Iron-Binding Proteins , Mitochondria , Neurodegenerative Diseases/metabolism , Neurotoxicity Syndromes/metabolism , Frataxin
4.
EMBO Mol Med ; 12(12): e12025, 2020 12 07.
Article in English | MEDLINE | ID: mdl-33179866

ABSTRACT

Tau accumulation in the form of neurofibrillary tangles in the brain is a hallmark of tauopathies such as Alzheimer's disease (AD). Tau aggregates accumulate in brain regions in a defined spatiotemporal pattern and may induce the aggregation of native Tau in a prion-like manner. However, the underlying mechanisms of cell-to-cell spreading of Tau pathology are unknown and could involve encapsulation within exosomes, trans-synaptic passage, and tunneling nanotubes (TNTs). We have established a neuronal cell model to monitor both internalization of externally added fibrils, synthetic (K18) or Tau from AD brain extracts, and real-time conversion of microtubule-binding domain of Tau fused to a fluorescent marker into aggregates. We found that these endogenously formed deposits colabel with ubiquitin and p62 but are not recruited to macroautophagosomes, eventually escaping clearance. Furthermore, endogenous K18-seeded Tau aggregates spread to neighboring cells where they seed new deposits. Transfer of Tau aggregates depends on direct cell contact, and they are found inside TNTs connecting neuronal cells. We further demonstrate that contact-dependent transfer occurs in primary neurons and between neurons and astrocytes in organotypic cultures.


Subject(s)
Neurons/metabolism , Protein Aggregation, Pathological , tau Proteins/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Astrocytes , Brain/metabolism , Brain/pathology , Cell Line , Humans , Mice , Mice, Inbred C57BL , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , Neurons/pathology
5.
Int J Mol Sci ; 21(18)2020 Sep 11.
Article in English | MEDLINE | ID: mdl-32933002

ABSTRACT

Friedreich's ataxia is the most common hereditary ataxia for which there is no cure or approved treatment at present. However, therapeutic developments based on the understanding of pathological mechanisms underlying the disease have advanced considerably, with the implementation of cellular models that mimic the disease playing a crucial role. Human olfactory ecto-mesenchymal stem cells represent a novel model that could prove useful due to their accessibility and neurogenic capacity. Here, we isolated and cultured these stem cells from Friedreich´s ataxia patients and healthy donors, characterizing their phenotype and describing disease-specific features such as reduced cell viability, impaired aconitase activity, increased ROS production and the release of cytokines involved in neuroinflammation. Importantly, we observed a positive effect on patient-derived cells, when frataxin levels were restored, confirming the utility of this in vitro model to study the disease. This model will improve our understanding of Friedreich´s ataxia pathogenesis and will help in developing rationally designed therapeutic strategies.


Subject(s)
Friedreich Ataxia/metabolism , Olfactory Mucosa/metabolism , Reactive Oxygen Species/metabolism , Stem Cells/metabolism , Aconitate Hydratase/metabolism , Cell Survival/physiology , Cells, Cultured , Cytokines/metabolism , Humans , Inflammation/metabolism
6.
EMBO J ; 38(23): e101230, 2019 12 02.
Article in English | MEDLINE | ID: mdl-31625188

ABSTRACT

Tunneling nanotubes (TNTs) are actin-based transient tubular connections that allow direct communication between distant cells. TNTs play an important role in several physiological (development, immunity, and tissue regeneration) and pathological (cancer, neurodegeneration, and pathogens transmission) processes. Here, we report that the Wnt/Ca2+ pathway, an intracellular cascade that is involved in actin cytoskeleton remodeling, has a role in TNT formation and TNT-mediated transfer of cargoes. Specifically, we found that Ca2+ /calmodulin-dependent protein kinase II (CaMKII), a transducer of the Wnt/Ca2+ pathway, regulates TNTs in a neuronal cell line and in primary neurons. We identified the ß isoform of CaMKII as a key molecule in modulating TNT formation and transfer, showing that this depends on the actin-binding activity of the protein. Finally, we found that the transfer of vesicles and aggregated α-synuclein between primary neurons can be regulated by the activation of the Wnt/Ca2+ pathway. Our findings suggest that Wnt/Ca2+ pathway could be a novel promising target for therapies designed to impair TNT-mediated propagation of pathogens.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/physiology , Calcium/metabolism , Cell Communication , Cell Membrane/metabolism , Nanotubes/chemistry , Neurons/physiology , Wnt Proteins/metabolism , Actins/metabolism , Animals , Calcium Signaling , Cells, Cultured , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/cytology , Signal Transduction
7.
Acta Neuropathol ; 134(5): 789-808, 2017 11.
Article in English | MEDLINE | ID: mdl-28725967

ABSTRACT

Recent evidence suggests that disease progression in Parkinson's disease (PD) could occur by the spreading of α-synuclein (α-syn) aggregates between neurons. Here we studied the role of astrocytes in the intercellular transfer and fate of α-syn fibrils, using in vitro and ex vivo models. α-Syn fibrils can be transferred to neighboring cells; however, the transfer efficiency changes depending on the cell types. We found that α-syn is efficiently transferred from astrocytes to astrocytes and from neurons to astrocytes, but less efficiently from astrocytes to neurons. Interestingly, α-syn puncta are mainly found inside the lysosomal compartments of the recipient cells. However, differently from neurons, astrocytes are able to efficiently degrade fibrillar α-syn, suggesting an active role for these cells in clearing α-syn deposits. Astrocytes co-cultured with organotypic brain slices are able to take up α-syn fibrils from the slices. Altogether our data support a role for astrocytes in trapping and clearing α-syn pathological deposits in PD.


Subject(s)
Astrocytes/metabolism , Hippocampus/metabolism , Neurons/metabolism , Parkinson Disease/metabolism , alpha-Synuclein/metabolism , Animals , Astrocytes/pathology , Cells, Cultured , Coculture Techniques , Disease Progression , Hippocampus/pathology , Mice , Neurons/pathology , Parkinson Disease/pathology
8.
EMBO J ; 35(19): 2120-2138, 2016 10 04.
Article in English | MEDLINE | ID: mdl-27550960

ABSTRACT

Synucleinopathies such as Parkinson's disease are characterized by the pathological deposition of misfolded α-synuclein aggregates into inclusions throughout the central and peripheral nervous system. Mounting evidence suggests that intercellular propagation of α-synuclein aggregates may contribute to the neuropathology; however, the mechanism by which spread occurs is not fully understood. By using quantitative fluorescence microscopy with co-cultured neurons, here we show that α-synuclein fibrils efficiently transfer from donor to acceptor cells through tunneling nanotubes (TNTs) inside lysosomal vesicles. Following transfer through TNTs, α-synuclein fibrils are able to seed soluble α-synuclein aggregation in the cytosol of acceptor cells. We propose that donor cells overloaded with α-synuclein aggregates in lysosomes dispose of this material by hijacking TNT-mediated intercellular trafficking. Our findings thus reveal a possible novel role of TNTs and lysosomes in the progression of synucleinopathies.


Subject(s)
Amyloid/metabolism , Cell Communication , Lysosomes/metabolism , Nanotubes , Neurons/physiology , alpha-Synuclein/metabolism , Animals , Cells, Cultured , Coculture Techniques , Mice , Microscopy, Fluorescence
9.
Mol Ther ; 24(5): 877-89, 2016 05.
Article in English | MEDLINE | ID: mdl-26849417

ABSTRACT

Friedreich's ataxia is a predominantly neurodegenerative disease caused by recessive mutations that produce a deficiency of frataxin (FXN). Here, we have used a herpesviral amplicon vector carrying a gene encoding for brain-derived neurotrophic factor (BDNF) to drive its overexpression in neuronal cells and test for its effect on FXN-deficient neurons both in culture and in the mouse cerebellum in vivo. Gene transfer of BDNF to primary cultures of mouse neurons prevents the apoptosis which is triggered by the knockdown of FXN gene expression. This neuroprotective effect of BDNF is also observed in vivo in a viral vector-based knockdown mouse cerebellar model. The injection of a lentiviral vector carrying a minigene encoding for a FXN-specific short hairpin ribonucleic acid (shRNA) into the mouse cerebellar cortex triggers a FXN deficit which is accompanied by significant apoptosis of granule neurons as well as loss of calbindin in Purkinje cells. These pathological changes are accompanied by a loss of motor coordination of mice as assayed by the rota-rod test. Coinjection of a herpesviral vector encoding for BDNF efficiently prevents both the development of cerebellar neuropathology and the ataxic phenotype. These data demonstrate the potential therapeutic usefulness of neurotrophins like BDNF to protect FXN-deficient neurons from degeneration.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Friedreich Ataxia/prevention & control , Genetic Therapy/methods , Iron-Binding Proteins/genetics , Neurons/pathology , Animals , Apoptosis/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Cells, Cultured , Disease Models, Animal , Friedreich Ataxia/genetics , Gene Knockdown Techniques , Genetic Vectors/administration & dosage , Herpesviridae/genetics , Humans , Mice , Neurons/drug effects , Frataxin
10.
Neurobiol Dis ; 76: 1-12, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25554687

ABSTRACT

Friedreich's ataxia (FA) is a recessive, predominantly neurodegenerative disorder caused in most cases by mutations in the first intron of the frataxin (FXN) gene. This mutation drives the expansion of a homozygous GAA repeat that results in decreased levels of FXN transcription and frataxin protein. Frataxin (Fxn) is a ubiquitous mitochondrial protein involved in iron-sulfur cluster biogenesis, and a decrease in the levels of this protein is responsible for the symptoms observed in the disease. Although the pathological manifestations of FA are mainly observed in neurons of both the central and peripheral nervous system, it is not clear if changes in non-neuronal cells may also contribute to the pathogenesis of FA, as recently suggested for other neurodegenerative disorders. Therefore, the aims of this study were to generate and characterize a cell model of Fxn deficiency in human astrocytes (HAs) and to evaluate the possible involvement of non-cell autonomous processes in FA. To knockdown frataxin in vitro, we transduced HAs with a specific shRNA lentivirus (shRNA37), which produced a decrease in both frataxin mRNA and protein expression, along with mitochondrial superoxide production, and signs of p53-mediated cell cycle arrest and apoptotic cell death. To test for non-cell autonomous interactions we cultured wild-type mouse neurons in the presence of frataxin-deficient astrocyte conditioned medium, which provoked a delay in the maturation of these neurons, a decrease in neurite length and enhanced cell death. Our findings confirm a detrimental effect of frataxin silencing, not only for astrocytes, but also for neuron-glia interactions, underlining the need to take into account the role of non-cell autonomous processes in FA.


Subject(s)
Astrocytes/metabolism , Friedreich Ataxia/metabolism , Iron-Binding Proteins/metabolism , Animals , Apoptosis , Cell Cycle Checkpoints , Cell Death , Cell Survival , Cells, Cultured , Gene Knockdown Techniques , Humans , Iron-Binding Proteins/genetics , Mice , Mitochondria/metabolism , Neurons/physiology , Superoxides/metabolism , Frataxin
11.
Curr Pharm Des ; 20(29): 4707-22, 2014.
Article in English | MEDLINE | ID: mdl-24588829

ABSTRACT

The central nervous system (CNS) innate immune response includes an arsenal of molecules and receptors expressed by professional phagocytes, glial cells and neurons that is involved in host defence and clearance of toxic and dangerous cell debris. However, any uncontrolled innate immune responses within the CNS are widely recognized as playing a major role in the development of autoimmune disorders and neurodegeneration, with multiple sclerosis (MS) Alzheimer's disease (AD) being primary examples. Hence, it is important to identify the key regulatory mechanisms involved in the control of CNS innate immunity and which could be harnessed to explore novel therapeutic avenues. Neuroimmune regulatory proteins (NIReg) such as CD95L, CD200, CD47, sialic acid, complement regulatory proteins (CD55, CD46, fH, C3a), HMGB1, may control the adverse immune responses in health and diseases. In the absence of these regulators, when neurons die by apoptosis, become infected or damaged, microglia and infiltrating immune cells are free to cause injury as well as an adverse inflammatory response in acute and chronic settings. We will herein provide new emphasis on the role of the pair CD200-CD200R in MS and its experimental models: experimental autoimmune encephalomyelitis (EAE) and Theiler's virus induced demyelinating disease (TMEV-IDD). The interest of the cannabinoid system as inhibitor of inflammation prompt us to introduce our findings about the role of endocannabinoids (eCBs) in promoting CD200-CD200 receptor (CD200R) interaction and the benefits caused in TMEV-IDD. Finally, we also review the current data on CD200-CD200R interaction in AD, as well as, in the aging brain.


Subject(s)
Antigens, CD/metabolism , Antigens, Surface/metabolism , Brain/immunology , Encephalitis/immunology , Endocannabinoids/physiology , Immunity, Innate , Receptors, Cell Surface/metabolism , Aging/immunology , Alzheimer Disease/immunology , Encephalitis/therapy , Humans , Multiple Sclerosis/immunology , Orexin Receptors
12.
Glia ; 60(9): 1437-50, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22653796

ABSTRACT

The endocannabinoid anandamide (AEA) is released by macrophages and microglia on pathological neuroinflammatory conditions such as multiple sclerosis (MS). CD200 is a membrane glycoprotein expressed in neurons that suppresses immune activity via its receptor (CD200R) mainly located in macrophages/microglia. CD200-CD200R interactions contribute to the brain immune privileged status. In this study, we show that AEA protects neurons from microglia-induced neurotoxicity via CD200-CD200R interaction. AEA increases the expression of CD200R1 in LPS/IFN-γ activated microglia through the activation of CB(2) receptors. The neuroprotective effect of AEA disappears when microglial cells derive from CD200R1(-/-) mice. We also show that engagement of CD200R1 by CD200Fc decreased the production of the proinflammatory cytokines IL-1ß and IL-6, but increased IL-10 in activated microglia. In the chronic phases of Theiler's virus-induced demyelinating disease (TMEV-IDD) the expression of CD200 and CD200R1 was reduced in the spinal cord. AEA-treated animals up-regulated the expression of CD200 and CD200R1, restoring levels found in sham animals together with increased expression of IL-10 and reduced expression of IL-1ß and IL-6. Treated animals also improved their motor behavior. Because AEA up-regulated the expression of CD200R1 in microglia, but failed to enhance CD200 in neurons we suggest that AEA-induced up-regulation of CD200 in TMEV-IDD is likely due to IL-10 as this cytokine increases CD200 in neurons. Our findings provide a new mechanism of action of AEA to limit immune response in the inflamed brain.


Subject(s)
Antigens, CD/metabolism , Antigens, Surface/metabolism , Arachidonic Acids/therapeutic use , Brain/metabolism , Endocannabinoids/therapeutic use , Inflammation/drug therapy , Neuroprotective Agents/therapeutic use , Polyunsaturated Alkamides/therapeutic use , Receptors, Cell Surface/metabolism , Animals , Arachidonic Acids/pharmacology , Brain/drug effects , Brain/immunology , Cells, Cultured , Endocannabinoids/pharmacology , Inflammation/immunology , Inflammation/metabolism , Interleukin-1beta/biosynthesis , Interleukin-6/biosynthesis , Mice , Microglia/drug effects , Microglia/immunology , Microglia/metabolism , Neurons/drug effects , Neurons/immunology , Neurons/metabolism , Neuroprotective Agents/pharmacology , Orexin Receptors , Polyunsaturated Alkamides/pharmacology
13.
Neurosci Lett ; 508(2): 67-72, 2012 Feb 06.
Article in English | MEDLINE | ID: mdl-22206832

ABSTRACT

NSC-34 cells, a hybridoma cell line derived from the fusion of neuroblastoma cells with mice spinal cord cells, have been widely used as an in vitro model for the study of motor neuron diseases [i.e. amyotrophic lateral sclerosis (ALS)]. In the present study, they were used to characterize different elements of the cannabinoid signaling system, which have been reported to serve as targets for the neuroprotective action of different natural and synthetic cannabinoid compounds. Using RT-PCR, Western blotting and immunocytochemistry, we first identified the presence of the cannabinoid CB(1) receptor in these cells. As expected, CB(2) receptor is not expressed in this neuronal cell line, a result that is concordant with the idea that this receptor type is preferentially expressed in glial elements. Diacylglycerol-lipase (DAGL) and N-arachidonoylphosphatidylethanolamine-phospholipase D (NAPE-PLD), the enzymes that synthesize endocannabinoids, have also been detected in these cells using RT-PCR, and the same happened with the endocannabinoid-degrading enzymes fatty acid amide hydrolase (FAAH) and monoacylglycerol-lipase (MAGL). The presence of the CB(1) receptor in these cells supports the idea that this receptor may play a role in the regulation of cellular survival face to excitotoxic injury. Interestingly, the expression of CB(1) receptor (and also the FAAH enzyme) was strongly up-regulated after differentiation of these cells, as previously reported with glutamate receptors. No changes were found for NAPE-PLD, DAGL and MAGL. Assuming that glutamate toxicity is one of the major causes of neuronal damage in ALS and other motor neurons diseases, the differentiated NSC-34 cells might serve as a useful model for studying neuroprotection with cannabinoids in conditions of excitotoxic injury, mitochondrial malfunctioning and oxidative stress.


Subject(s)
Endocannabinoids/metabolism , Motor Neurons/metabolism , Receptor, Cannabinoid, CB1/metabolism , Amidohydrolases/genetics , Amidohydrolases/metabolism , Animals , Gene Expression , Hybridomas , Lipoprotein Lipase/genetics , Lipoprotein Lipase/metabolism , Mice , Monoacylglycerol Lipases/genetics , Monoacylglycerol Lipases/metabolism , Motor Neuron Disease/metabolism , Motor Neurons/enzymology , Phospholipase D/genetics , Phospholipase D/metabolism , Receptor, Cannabinoid, CB1/genetics
14.
J Neuroinflammation ; 8: 102, 2011 Aug 18.
Article in English | MEDLINE | ID: mdl-21851608

ABSTRACT

BACKGROUND: VCAM-1 represents one of the most important adhesion molecule involved in the transmigration of blood leukocytes across the blood-brain barrier (BBB) that is an essential step in the pathogenesis of MS. Several evidences have suggested the potential therapeutic value of cannabinoids (CBs) in the treatment of MS and their experimental models. However, the effects of endocannabinoids on VCAM-1 regulation are poorly understood. In the present study we investigated the effects of anandamide (AEA) in the regulation of VCAM-1 expression induced by Theiler's virus (TMEV) infection of brain endothelial cells using in vitro and in vivo approaches. METHODS: i) in vitro: VCAM-1 was measured by ELISA in supernatants of brain endothelial cells infected with TMEV and subjected to AEA and/or cannabinoid receptors antagonist treatment. To evaluate the functional effect of VCAM-1 modulation we developed a blood brain barrier model based on a system of astrocytes and brain endothelial cells co-culture. ii) in vivo: CB(1) receptor deficient mice (Cnr1(-/-)) infected with TMEV were treated with the AEA uptake inhibitor UCM-707 for three days. VCAM-1 expression and microglial reactivity were evaluated by immunohistochemistry. RESULTS: Anandamide-induced inhibition of VCAM-1 expression in brain endothelial cell cultures was mediated by activation of CB(1) receptors. The study of leukocyte transmigration confirmed the functional relevance of VCAM-1 inhibition by AEA. In vivo approaches also showed that the inhibition of AEA uptake reduced the expression of brain VCAM-1 in response to TMEV infection. Although a decreased expression of VCAM-1 by UCM-707 was observed in both, wild type and CB(1) receptor deficient mice (Cnr1(-/-)), the magnitude of VCAM-1 inhibition was significantly higher in the wild type mice. Interestingly, Cnr1(-/-) mice showed enhanced microglial reactivity and VCAM-1 expression following TMEV infection, indicating that the lack of CB(1) receptor exacerbated neuroinflammation. CONCLUSIONS: Our results suggest that CB(1) receptor dependent VCAM-1 inhibition is a novel mechanism for AEA-reduced leukocyte transmigration and contribute to a better understanding of the mechanisms underlying the beneficial role of endocannabinoid system in the Theiler's virus model of MS.


Subject(s)
Arachidonic Acids/pharmacology , Blood-Brain Barrier/metabolism , Endothelial Cells/drug effects , Leukocytes/physiology , Polyunsaturated Alkamides/pharmacology , Receptor, Cannabinoid, CB1/metabolism , Theilovirus/metabolism , Transendothelial and Transepithelial Migration/drug effects , Vascular Cell Adhesion Molecule-1/metabolism , Animals , Brain/cytology , Brain/drug effects , Brain/metabolism , Cannabinoid Receptor Modulators/pharmacology , Cell Adhesion/drug effects , Endocannabinoids , Endothelial Cells/cytology , Endothelial Cells/metabolism , Female , Humans , Leukocytes/cytology , Leukocytes/drug effects , Mice , Mice, Knockout , Microglia/metabolism , Theilovirus/genetics
15.
Brain Behav Immun ; 25(4): 736-49, 2011 May.
Article in English | MEDLINE | ID: mdl-21310228

ABSTRACT

Theiler's virus (TMEV) infection of the central nervous system (CNS) induces an immune-mediated demyelinating disease in susceptible mouse strains and serves as a relevant infection model for human multiple sclerosis (MS). The endocannabinoid system represents a novel therapeutic target for autoimmune and chronic inflammatory diseases due to its anti-inflammatory properties by regulating cytokine network. IL-12p70 and IL-23 are functionally related heterodimeric cytokines that play a crucial role in the pathogenesis of MS. In the present study we showed that the endocannabinoid anandamide (AEA) downregulated the gene expression of IL-12p70 and IL-23 forming subunits mRNAs in the spinal cord of TMEV-infected mice and ameliorated motor disturbances. This was accompanied by significant decreases on the serological levels of IL-12p70/IL-23 and more interestingly, of IL-17A. In contrast, serum levels of IL-10 resulted elevated. In addition, we studied the signalling pathways involved in the regulation of IL-12p70/IL-23 and IL-10 expression in TMEV-infected microglia and addressed the possible interactions of AEA with these pathways. AEA acted through the ERK1/2 and JNK pathways to downregulate IL-12p70 and IL-23 while upregulating IL-10. These effects were partially mediated by CB2 receptor activation. We also described an autocrine circuit of cross-talk between IL-12p70/IL-23 and IL-10, since endogenously produced IL-10 negatively regulates IL-12p70 and IL-23 cytokines in TMEV-infected microglia. This suggests that by altering the cytokine network, AEA could indirectly modify the type of immune responses within the CNS. Accordingly, pharmacological modulation of endocannabinoids might be a useful tool for treating neuroinflammatory diseases.


Subject(s)
Arachidonic Acids/pharmacology , Cannabinoid Receptor Modulators/pharmacology , Cardiovirus Infections/immunology , Endocannabinoids , Interleukins/immunology , Microglia/immunology , Nervous System Autoimmune Disease, Experimental/immunology , Polyunsaturated Alkamides/pharmacology , Adaptive Immunity/drug effects , Analysis of Variance , Animals , Cardiovirus Infections/drug therapy , Cardiovirus Infections/virology , Disease Models, Animal , Down-Regulation , Female , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-10/metabolism , Interleukin-12/genetics , Interleukin-12/immunology , Interleukin-12/metabolism , Interleukin-23/drug effects , Interleukin-23/genetics , Interleukin-23/metabolism , Interleukins/genetics , Interleukins/metabolism , Mice , Microglia/metabolism , Microglia/virology , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Nervous System Autoimmune Disease, Experimental/metabolism , Nervous System Autoimmune Disease, Experimental/virology , Neuroimmunomodulation/drug effects , Protein Subunits , RNA, Messenger/analysis , Receptor Cross-Talk , Signal Transduction , Statistics, Nonparametric , Theilovirus/immunology
16.
Glia ; 58(2): 135-47, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-19565660

ABSTRACT

The endocannabinoid system exhibits anti-inflammatory properties by regulating cytokine production. Anandamide (AEA) down-regulates proinflammatory cytokines in a viral model of multiple sclerosis (MS). However, little is known about the mechanisms by which AEA exerts these effects. Microglial cells are the main source of cytokines within the brain and the first barrier of defense against pathogens by acting as antigen presenting cells. IL-10 is a key physiological negative regulator of microglial activation. In this study we show that AEA enhances LPS/IFNgamma-induced IL-10 production in microglia by targeting CB(2) receptors through the activation of ERK1/2 and JNK MAPKs. AEA also inhibits NF-kappaB activation by interfering with the phosphorylation of IkappaBalpha, which may result in an increase of IL-10 production. Moreover, endogenously produced IL-10 negatively regulates IL-12 and IL-23 cytokines, which in its turn modify the pattern of expression of transcription factors involved in Th commitment of splenocytes. This suggests that by altering the cytokine network, AEA could indirectly modify the type of immune responses within the central nervous system (CNS). Accordingly, pharmacological modulation of AEA uptake and degradation might be a useful tool for treating neuroinflammatory diseases.


Subject(s)
Arachidonic Acids/metabolism , Interleukin-10/metabolism , Microglia/enzymology , Microglia/metabolism , Polyunsaturated Alkamides/metabolism , Receptor, Cannabinoid, CB2/metabolism , Animals , Cell Line , Cells, Cultured , Endocannabinoids , I-kappa B Proteins/metabolism , Interferon-gamma/toxicity , Interleukin-12/metabolism , Interleukin-23/metabolism , Lipopolysaccharides/toxicity , MAP Kinase Kinase 4/metabolism , MAP Kinase Signaling System , Mice , Mice, Inbred BALB C , Microglia/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Phosphorylation
17.
Biochem Pharmacol ; 77(1): 86-100, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-18848818

ABSTRACT

The endocannabinoid system represents a novel therapeutic target for autoimmune and chronic inflammatory diseases. IL-12 and IL-23 are functionally related heterodimeric cytokines that play a crucial role in the pathogenesis of multiple sclerosis (MS). In the present study we investigated the effects of the endocannabinoid anandamide (AEA) on the inducible expression of the biologically active cytokines IL-12p70 and IL-23, and their forming subunits, in activated microglial cells. We also studied the signalling pathways involved in the regulation of IL-12p70/IL-23 expression and addressed the possible interactions of AEA with these pathways. Here, we show that AEA was capable to inhibit the production of biologically active IL-12p70 and IL-23, and their subunits, by activated human and murine microglial cultures. Treatment of activated microglial cells with inhibitors of several mitogen-activated protein kinase (MAPK) reveals that AEA acts through the ERK1/2 and JNK pathways to down-regulate IL-12p70 and IL-23. These effects were partially mediated by CB2 receptor activation. Together, our results provide the first demonstration of a role of AEA in inhibiting IL-12p70/IL-23 axis in human and murine microglial cells via the CB2 receptor and suggest that the pharmacological manipulation of the endocannabinoid system is a potential tool for treating brain inflammatory and autoimmune diseases, like MS.


Subject(s)
Arachidonic Acids/pharmacology , Cannabinoids/pharmacology , Interleukin-12/physiology , Interleukin-23/physiology , Microglia/physiology , Polyunsaturated Alkamides/pharmacology , Receptor, Cannabinoid, CB2/physiology , Signal Transduction/physiology , Animals , Cells, Cultured , Endocannabinoids , Fetus , Humans , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Mice , Mice, Inbred BALB C , Microglia/drug effects , Signal Transduction/drug effects
18.
Eur Neuropsychopharmacol ; 18(12): 917-24, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18805677

ABSTRACT

Major depressive disorder (MDD) is a psychiatric condition characterized by hypercortisolism and variations in circulatory cytokines. Previously it has been reported that administration of selective serotonin reuptake inhibitors (SSRI) in MDD patients modify cortisol and cytokine levels but these studies only evaluated changes over a short time period. This work reports the long-term effects of administration of SSRI on the cortisol levels and pro-/anti-inflammatory cytokine profile in a group of MDD patients treated for 52 weeks. A total of 31 patients diagnosed with MDD received anti depressant treatment with SSRI. HDRS and BDI were administered over a year, and levels of interleukin IL-1beta, IL-10, IL-2, IFN-gamma, IL-4, IL-13, and 24-h urine cortisol were determined at weeks (W) 0, 5, 20, 36 and 52 of treatment. Before treatment we found high levels of cortisol, IL-4, IL-13 (Th2) and IL-10 in MDD patients when compared with healthy volunteers. At W20 psychiatric scales indicated a remission of the depressive episode concomitantly with increments in IL-2 and IL-1beta but without changes in cortisol. Towards the end of the treatment (W52) we observed a significant reduction (p<0.01) in cortisol levels, with an increment in IL-1beta and IFN-gamma and a decrease in Th2 cytokines. Our results suggest that depressed patients only reach a partial reestablishment of HPA axis function after the long-term administration of SSRI.


Subject(s)
Antidepressive Agents/therapeutic use , Cytokines/blood , Depressive Disorder, Major/blood , Depressive Disorder, Major/drug therapy , Selective Serotonin Reuptake Inhibitors/therapeutic use , Adult , Analysis of Variance , Enzyme-Linked Immunosorbent Assay , Female , Humans , Hydrocortisone/blood , Longitudinal Studies , Male , Psychiatric Status Rating Scales , Radioimmunoassay/methods , Retrospective Studies , Time Factors , Young Adult
19.
Eur J Neurosci ; 28(4): 633-41, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18657182

ABSTRACT

Cannabinoids have recently been approved as a treatment for pain in multiple sclerosis (MS). Increasing evidence from animal studies suggests that this class of compounds could also prove efficient to fight neurodegeneration, demyelination, inflammation and autoimmune processes occurring in this pathology. However, the use of cannabinoids is limited by their psychoactive effects. In this context, potentiation of the endogenous cannabinoid signalling could represent a substitute to the use of exogenously administrated cannabinoid ligands. Here, we studied the expression of different elements of the endocannabinoid system in a chronic model of MS in mice. We first studied the expression of the two cannabinoid receptors, CB(1) and CB(2), as well as the putative intracellular cannabinoid receptor peroxisome proliferator-activated receptor-alpha. We observed an upregulation of CB(2), correlated to the production of proinflammatory cytokines, at 60 days after the onset of the MS model. At this time, the levels of the endocannabinoid, 2-arachidonoylglycerol, and of the anti-inflammatory anandamide congener, palmithoylethanolamide, were enhanced, without changes in the levels of anandamide. These changes were not due to differences in the expression of the degradation enzymes, fatty acid amide hydrolase and monoacylglycerol lipase, or of biosynthetic enzymes, diacylglycerol lipase-alpha and N-acylphosphatidylethanolamine phospholipase-D at this time (60 days). Finally, the exogenous administration of palmitoylethanolamide resulted in a reduction of motor disability in the animals subjected to this model of MS, accompanied by an anti-inflammatory effect. This study overall highlights the potential therapeutic effects of endocannabinoids in MS.


Subject(s)
Antiviral Agents/therapeutic use , Cannabinoid Receptor Modulators/therapeutic use , Endocannabinoids , Multiple Sclerosis/drug therapy , Multiple Sclerosis/virology , Palmitic Acids/therapeutic use , Signal Transduction/physiology , Amides , Animals , Anti-Inflammatory Agents/therapeutic use , Cannabinoid Receptor Modulators/metabolism , Cytokines/genetics , Cytokines/immunology , Disease Models, Animal , Ethanolamines , Female , Humans , Mice , Motor Activity/physiology , Multiple Sclerosis/immunology , Multiple Sclerosis/physiopathology , PPAR alpha/genetics , PPAR alpha/metabolism , Pain/drug therapy , Palmitic Acids/metabolism , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/genetics , Receptor, Cannabinoid, CB2/metabolism , Rotarod Performance Test , Theilovirus/immunology
20.
Expert Opin Ther Targets ; 12(2): 185-95, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18208367

ABSTRACT

BACKGROUND: Cannabinoids have been shown to exert beneficial actions in different animal models of multiple sclerosis (MS). However, the use of cannabinoids compounds in human therapy is greatly limited by their psychoactivity. Thus, new hopes in MS therapy have arisen from the evidence for a cannabinoid receptor, termed CB2, which is devoid of psychoactive effects in animal models. OBJECTIVE: This review discusses the different mechanisms by which CB2 activation could induce therapeutic actions in MS. METHODS: Particular focus is given to the potential effects on inflammation/autoimmunity, remyelination and neuroprotection. CONCLUSION: This review discusses the importance of glial cells in sustaining these effects, as well as the putative secondary effects that would limit the use of CB2 agonists in the treatment of MS.


Subject(s)
Multiple Sclerosis/drug therapy , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Receptor, Cannabinoid, CB2/agonists , Receptor, Cannabinoid, CB2/metabolism , Animals , Humans , Multiple Sclerosis/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...