Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
J Neuroinflammation ; 21(1): 54, 2024 Feb 21.
Article in English | MEDLINE | ID: mdl-38383421

ABSTRACT

Parkinson's disease (PD) is a common age-related neurodegenerative disorder characterized by the aggregation of α-Synuclein (αSYN) building up intraneuronal inclusions termed Lewy pathology. Mounting evidence suggests that neuron-released αSYN aggregates could be central to microglial activation, which in turn mounts and orchestrates neuroinflammatory processes potentially harmful to neurons. Therefore, understanding the mechanisms that drive microglial cell activation, polarization and function in PD might have important therapeutic implications. Here, using primary microglia, we investigated the inflammatory potential of pure αSYN fibrils derived from PD patients. We further explored and characterized microglial cell responses to a chronic-type inflammatory stimulation combining PD patient-derived αSYN fibrils (FPD), Tumor necrosis factor-α (TNFα) and prostaglandin E2 (PGE2) (TPFPD). We showed that FPD hold stronger inflammatory potency than pure αSYN fibrils generated de novo. When combined with TNFα and PGE2, FPD polarizes microglia toward a particular functional phenotype departing from FPD-treated cells and featuring lower inflammatory cytokine and higher glutamate release. Whereas metabolomic studies showed that TPFPD-exposed microglia were closely related to classically activated M1 proinflammatory cells, notably with similar tricarboxylic acid cycle disruption, transcriptomic analysis revealed that TPFPD-activated microglia assume a unique molecular signature highlighting upregulation of genes involved in glutathione and iron metabolisms. In particular, TPFPD-specific upregulation of Slc7a11 (which encodes the cystine-glutamate antiporter xCT) was consistent with the increased glutamate response and cytotoxic activity of these cells toward midbrain dopaminergic neurons in vitro. Together, these data further extend the structure-pathological relationship of αSYN fibrillar polymorphs to their innate immune properties and demonstrate that PD-derived αSYN fibrils, TNFα and PGE2 act in concert to drive microglial cell activation toward a specific and highly neurotoxic chronic-type inflammatory phenotype characterized by robust glutamate release and iron retention.


Subject(s)
Neurotoxicity Syndromes , Parkinson Disease , Humans , Parkinson Disease/pathology , alpha-Synuclein/genetics , alpha-Synuclein/metabolism , Microglia/metabolism , Tumor Necrosis Factor-alpha/metabolism , Cues , Inflammation/metabolism , Dopaminergic Neurons/pathology , Neurotoxicity Syndromes/metabolism , Glutamates/metabolism , Iron/metabolism
2.
Cells ; 12(9)2023 05 07.
Article in English | MEDLINE | ID: mdl-37174736

ABSTRACT

Chlordecone (CLD) is an organochlorine pesticide (OCP) that is currently banned but still contaminates ecosystems in the French Caribbean. Because OCPs are known to increase the risk of Parkinson's disease (PD), we tested whether chronic low-level intoxication with CLD could reproduce certain key characteristics of Parkinsonism-like neurodegeneration. For that, we used culture systems of mouse midbrain dopamine (DA) neurons and glial cells, together with the nematode C. elegans as an in vivo model organism. We established that CLD kills cultured DA neurons in a concentration- and time-dependent manner while exerting no direct proinflammatory effects on glial cells. DA cell loss was not impacted by the degree of maturation of the culture. The use of fluorogenic probes revealed that CLD neurotoxicity was the consequence of oxidative stress-mediated insults and mitochondrial disturbances. In C. elegans worms, CLD exposure caused a progressive loss of DA neurons associated with locomotor deficits secondary to alterations in food perception. L-DOPA, a molecule used for PD treatment, corrected these deficits. Cholinergic and serotoninergic neuronal cells were also affected by CLD in C. elegans, although to a lesser extent than DA neurons. Noticeably, CLD also promoted the phosphorylation of the aggregation-prone protein tau (but not of α-synuclein) both in midbrain cell cultures and in a transgenic C. elegans strain expressing a human form of tau in neurons. In summary, our data suggest that CLD is more likely to promote atypical forms of Parkinsonism characterized by tau pathology than classical synucleinopathy-associated PD.


Subject(s)
Chlordecone , Parkinson Disease , Parkinsonian Disorders , Pesticides , Animals , Humans , Mice , Caenorhabditis elegans/metabolism , Chlordecone/metabolism , Pesticides/toxicity , Ecosystem , Parkinsonian Disorders/pathology , Parkinson Disease/metabolism , Dopaminergic Neurons/metabolism , Mesencephalon/pathology
4.
Cells ; 11(10)2022 05 13.
Article in English | MEDLINE | ID: mdl-35626675

ABSTRACT

To model α-Synuclein (αS) aggregation and neurodegeneration in Parkinson's disease (PD), we established cultures of mouse midbrain dopamine (DA) neurons and chronically exposed them to fibrils 91 (F91) generated from recombinant human αS. We found that F91 have an exquisite propensity to seed the aggregation of endogenous αS in DA neurons when compared to other neurons in midbrain cultures. Until two weeks post-exposure, somal aggregation in DA neurons increased with F91 concentrations (0.01-0.75 µM) and the time elapsed since the initiation of seeding, with, however, no evidence of DA cell loss within this time interval. Neither toxin-induced mitochondrial deficits nor genetically induced loss of mitochondrial quality control mechanisms promoted F91-mediated αS aggregation or neurodegeneration under these conditions. Yet, a significant loss of DA neurons (~30%) was detectable three weeks after exposure to F91 (0.5 µM), i.e., at a time point where somal aggregation reached a plateau. This loss was preceded by early deficits in DA uptake. Unlike αS aggregation, the loss of DA neurons was prevented by treatment with GDNF, suggesting that αS aggregation in DA neurons may induce a form of cell death mimicking a state of trophic factor deprivation. Overall, our model system may be useful for exploring PD-related pathomechanisms and for testing molecules of therapeutic interest for this disorder.


Subject(s)
Parkinson Disease , alpha-Synuclein , Animals , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Mesencephalon/metabolism , Mice , Parkinson Disease/metabolism , alpha-Synuclein/metabolism
6.
Cell Death Differ ; 26(3): 580-596, 2019 03.
Article in English | MEDLINE | ID: mdl-30006609

ABSTRACT

The precise contribution of astrocytes in neuroinflammatory process occurring in Parkinson's disease (PD) is not well characterized. In this study, using GRCx30CreERT2 mice that are conditionally inactivated for glucocorticoid receptor (GR) in astrocytes, we have examined the actions of astrocytic GR during dopamine neuron (DN) degeneration triggered by the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). The results show significantly augmented DN loss in GRCx30CreERT2 mutant mice in substantia nigra (SN) compared to controls. Hypertrophy of microglia but not of astrocytes was greatly enhanced in SN of these astrocytic GR mutants intoxicated with MPTP, indicating heightened microglial reactivity compared to similarly-treated control mice. In the SN of GR astrocyte mutants, specific inflammation-associated transcripts ICAM-1, TNF-α and Il-1ß as well as TNF-α protein levels were significantly elevated after MPTP neurotoxicity compared to controls. Interestingly, this paralleled increased connexin hemichannel activity and elevated intracellular calcium levels in astrocytes examined in acute midbrain slices from control and mutant mice treated with MPP+ . The increased connexin-43 hemichannel activity was found in vivo in MPTP-intoxicated mice. Importantly, treatment of MPTP-injected GRCx30CreERT2 mutant mice with TAT-Gap19 peptide, a specific connexin-43 hemichannel blocker, reverted both DN loss and microglial activation; in wild-type mice there was partial but significant survival effect. In the SN of post-mortem PD patients, a significant decrease in the number of astrocytes expressing nuclear GR was observed, suggesting the participation of astrocytic GR deregulation of inflammatory process in PD. Overall, these data provide mechanistic insights into GR-modulated processes in vivo, specifically in astrocytes, that contribute to a pro-inflammatory state and dopamine neurodegeneration in PD pathology.


Subject(s)
Astrocytes/metabolism , Connexins/metabolism , Dopaminergic Neurons/metabolism , Parkinson Disease/genetics , Animals , Humans , Male , Mice , Parkinson Disease/pathology
7.
Front Cell Neurosci ; 12: 31, 2018.
Article in English | MEDLINE | ID: mdl-29467625

ABSTRACT

It has long been proven that neurogenesis continues in the adult brains of mammals in the dentatus gyrus of the hippocampus due to the presence of neural stem cells. Although a large number of studies have been carried out to highlight the localization of vitamin D receptor in hippocampus, the expression of vitamin D receptor in neurogenic dentatus gyrus of hippocampus in Parkinson's disease (PD) and the molecular mechanisms triggered by vitamin D underlying the production of differentiated neurons from embryonic cells remain unknown. Thus, we performed a preclinical in vivo study by inducing PD in mice with MPTP and showed a reduction of glial fibrillary acidic protein (GFAP) and vitamin D receptor in the dentatus gyrus of hippocampus. Then, we performed an in vitro study by inducing embryonic hippocampal cell differentiation with vitamin D. Interestingly, vitamin D stimulates the expression of its receptor. Vitamin D receptor is a transcription factor that probably is responsible for the upregulation of microtubule associated protein 2 and neurofilament heavy polypeptide genes. The latter increases heavy neurofilament protein expression, essential for neurofilament growth. Notably N-cadherin, implicated in activity for dendritic outgrowth, is upregulated by vitamin D.

9.
Article in English | MEDLINE | ID: mdl-28261564

ABSTRACT

The human pathogen Legionella pneumophila must evade host cell death signaling to enable replication in lung macrophages and to cause disease. After bacterial growth, however, L. pneumophila is thought to induce apoptosis during egress from macrophages. The bacterial effector protein, SidF, has been shown to control host cell survival and death by inhibiting pro-apoptotic BNIP3 and BCL-RAMBO signaling. Using live-cell imaging to follow the L. pneumophila-macrophage interaction, we now demonstrate that L. pneumophila evades host cell apoptosis independent of SidF. In the absence of SidF, L. pneumophila was able to replicate, cause loss of mitochondria membrane potential, kill macrophages, and establish infections in lungs of mice. Consistent with this, deletion of BNIP3 and BCL-RAMBO did not affect intracellular L. pneumophila replication, macrophage death rates, and in vivo bacterial virulence. Abrogating mitochondrial cell death by genetic deletion of the effectors of intrinsic apoptosis, BAX, and BAK, or the regulator of mitochondrial permeability transition pore formation, cyclophilin-D, did not affect bacterial growth or the initial killing of macrophages. Loss of BAX and BAK only marginally limited the ability of L. pneumophila to efficiently kill all macrophages over extended periods. L. pneumophila induced killing of macrophages was delayed in the absence of capsase-11 mediated pyroptosis. Together, our data demonstrate that L. pneumophila evades host cell death responses independently of SidF during replication and can induce pyroptosis to kill macrophages in a timely manner.


Subject(s)
Bacterial Proteins/genetics , Flagellin/genetics , Legionella pneumophila/physiology , Legionnaires' Disease/microbiology , Macrophages/metabolism , Animals , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Bacterial Proteins/metabolism , Cell Death , Cell Survival , Female , Flagellin/metabolism , Gene Deletion , Host-Pathogen Interactions , Legionnaires' Disease/metabolism , Macrophages/microbiology , Membrane Proteins/metabolism , Mice , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Virulence Factors/genetics
10.
J Neuroinflammation ; 14(1): 60, 2017 03 21.
Article in English | MEDLINE | ID: mdl-28320442

ABSTRACT

BACKGROUND: Evidence from mice suggests that brain infiltrating immune cells contribute to neurodegeneration, and we previously identified a deleterious lymphocyte infiltration in Parkinson's disease mice. However, this remains controversial for monocytes, due to artifact-prone techniques used to distinguish them from microglia. Our aim was to reassess this open question, by taking advantage of the recent recognition that chemokine receptors CCR2 and CX3CR1 can differentiate between inflammatory monocytes and microglia, enabling to test whether CCR2+ monocytes infiltrate the brain during dopaminergic (DA) neurodegeneration and whether they contribute to neuronal death. This revealed unexpected insights into possible regulation of monocyte-attracting CCL2 induction. METHODS: We used acute 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mice and assessed monocyte infiltration by combining laser microdissection-guided chemokine RNA profiling of the substantia nigra (SN) with immunohistochemistry and CCR2-GFP reporter mice. To determine contribution to neuronal loss, we used CCR2-deletion and CCL2-overexpression, to reduce and increase CCR2+ monocyte infiltration, and CX3CR1-deletion to assess a potential implication in CCL2 regulation. RESULTS: Nigral chemokine profiling revealed early CCL2/7/12-CCR2 axis induction, suggesting monocyte infiltration in MPTP mice. CCL2 protein showed early peak induction in nigral astrocytes, while CCR2-GFP mice revealed early but limited nigral monocyte infiltration. However, blocking infiltration by CCR2 deletion did not influence DA neuronal loss. In contrast, transgenic astrocytic CCL2 over-induction increased CCR2+ monocyte infiltration and DA neuronal loss in MPTP mice. Surprisingly, CCL2 over-induction was also detected in MPTP intoxicated CX3CR1-deleted mice, which are known to present increased DA neuronal loss. Importantly, CX3CR1/CCL2 double-deletion suggested that increased neurotoxicity was driven by astrocytic CCL2 over-induction. CONCLUSIONS: We show that CCR2+ monocytes infiltrate the affected CNS, but at the level observed in acute MPTP mice, this does not contribute to DA neuronal loss. In contrast, the underlying astrocytic CCL2 induction seemed to be tightly controled, as already moderate CCL2 over-induction led to increased neurotoxicity in MPTP mice, likely due to the increased CCR2+ monocyte infiltration. Importantly, we found evidence suggesting that during DA neurodegeneration, this control was mediated by microglial CX3CR1 signaling, which protects against such neurotoxic CCL2 over-induction by astrocytes, thus hinting at an endogenous mechanism to limit neurotoxic effects of the CCL2-CCR2 axis.


Subject(s)
Astrocytes/metabolism , Cell Movement/drug effects , Chemokine CCL2/metabolism , MPTP Poisoning/pathology , Microglia/metabolism , Receptors, Interleukin-8A/deficiency , Animals , Astrocytes/drug effects , CD11b Antigen/metabolism , Calcium-Binding Proteins/metabolism , Cell Movement/genetics , Disease Models, Animal , Gene Expression Regulation/drug effects , Lipopolysaccharides/pharmacology , MPTP Poisoning/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins/metabolism , Microglia/drug effects , Monocytes/drug effects , Monocytes/metabolism , Receptors, CCR2/genetics , Receptors, CCR2/metabolism , Receptors, Interleukin-8A/genetics , Substantia Nigra/drug effects , Substantia Nigra/pathology , Time Factors , Tyrosine 3-Monooxygenase/metabolism
11.
Brain ; 140(1): 184-200, 2017 01.
Article in English | MEDLINE | ID: mdl-27818384

ABSTRACT

Alzheimer's disease is characterized by the combined presence of amyloid plaques and tau pathology, the latter being correlated with the progression of clinical symptoms. Neuroinflammatory changes are thought to be major contributors to Alzheimer's disease pathophysiology, even if their precise role still remains largely debated. Notably, to what extent immune responses contribute to cognitive impairments promoted by tau pathology remains poorly understood. To address this question, we took advantage of the THY-Tau22 mouse model that progressively develops hippocampal tau pathology paralleling cognitive deficits and reappraised the interrelationship between tau pathology and brain immune responses. In addition to conventional astroglial and microglial responses, we identified a CD8-positive T cell infiltration in the hippocampus of tau transgenic mice associated with an early chemokine response, notably involving CCL3. Interestingly, CD8-positive lymphocyte infiltration was also observed in the cortex of patients exhibiting frontemporal dementia with P301L tau mutation. To gain insights into the functional involvement of T cell infiltration in the pathophysiological development of tauopathy in THY-Tau22 mice, we chronically depleted T cells using anti-CD3 antibody. Such anti-CD3 treatment prevented hippocampal T cell infiltration in tau transgenic animals and reverted spatial memory deficits, in absence of tau pathology modulation. Altogether, these data support an instrumental role of hippocampal T cell infiltration in tau-driven pathophysiology and cognitive impairments in Alzheimer's disease and other tauopathies.


Subject(s)
Antibodies/therapeutic use , CD3 Complex/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cerebral Cortex/immunology , Chemokines/immunology , Cognitive Dysfunction/immunology , Hippocampus/immunology , Inflammation/immunology , Tauopathies/immunology , Aged , Animals , Cognitive Dysfunction/therapy , Disease Models, Animal , Humans , Inflammation/therapy , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Tauopathies/therapy
12.
Mediators Inflamm ; 2017: 2470950, 2017.
Article in English | MEDLINE | ID: mdl-29343884

ABSTRACT

Neutral sphingomyelinase is known to be implicated in growth arrest, differentiation, proliferation, and apoptosis. Although previous studies have reported the involvement of neutral sphingomyelinase in hippocampus physiopathology, its behavior in the hippocampus during Parkinson's disease remains undetected. In this study, we show an upregulation of inducible nitric oxide synthase and a downregulation of neutral sphingomyelinase in the hippocampus of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine- (MPTP-) induced mouse model of Parkinson's disease. Moreover, the stimulation of neutral sphingomyelinase activity with vitamin 1,25-dihydroxyvitamin D3 reduces specifically saturated fatty acid sphingomyelin by making sphingomyelin a less rigid molecule that might influence neurite plasticity. The possible biological relevance of the increase of neutral sphingomyelinase in Parkinson's disease is discussed.


Subject(s)
Hippocampus/enzymology , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/enzymology , Sphingomyelin Phosphodiesterase/metabolism , Animals , Calcitriol/pharmacology , Cell Line , Disease Models, Animal , Hippocampus/drug effects , Hippocampus/pathology , Inflammation Mediators/metabolism , MPTP Poisoning/enzymology , Male , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type II/metabolism , Parkinson Disease, Secondary/pathology , Sphingomyelins/metabolism
13.
Mediators Inflamm ; 2016: 3937057, 2016.
Article in English | MEDLINE | ID: mdl-27194825

ABSTRACT

Today a large number of studies are focused on clarifying the complexity and diversity of the pathogenetic mechanisms inducing Parkinson disease. We used 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), a neurotoxin that induces Parkinson disease, to evaluate the change of midbrain structure and the behavior of the anti-inflammatory factor e-cadherin, interleukin-6, tyrosine hydroxylase, phosphatase and tensin homolog, and caveolin-1. The results showed a strong expression of e-cadherin, variation of length and thickness of the heavy neurofilaments, increase of interleukin-6, and reduction of tyrosine hydroxylase known to be expression of dopamine cell loss, reduction of phosphatase and tensin homolog described to impair responses to dopamine, and reduction of caveolin-1 known to be expression of epithelial-mesenchymal transition and fibrosis. The possibility that the overexpression of the e-cadherin might be implicated in the anti-inflammatory reaction to MPTP treatment by influencing the behavior of the other analyzed molecules is discussed.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Cadherins/metabolism , Parkinson Disease/etiology , Parkinson Disease/metabolism , Animals , Interleukin-6/metabolism , Male , Mice, Inbred C57BL , Tyrosine 3-Monooxygenase/metabolism
14.
Neuron ; 90(4): 675-91, 2016 05 18.
Article in English | MEDLINE | ID: mdl-27196972

ABSTRACT

Parkinson disease (PD) is a multifactorial neurodegenerative disorder, the etiology of which remains largely unknown. Progressive impairment of voluntary motor control, which represents the primary clinical feature of the disease, is caused by a loss of midbrain substantia nigra dopamine (DA) neurons. We present here a synthetic overview of cell-autonomous mechanisms that are likely to participate in DA cell death in both sporadic and inherited forms of the disease. In particular, we describe how damage to vulnerable DA neurons may arise from cellular disturbances produced by protein misfolding and aggregation, disruption of autophagic catabolism, endoplasmic reticulum (ER) stress, mitochondrial dysfunction, or loss of calcium homeostasis. Where pertinent, we show how these mechanisms may mutually cooperate to promote neuronal death.


Subject(s)
Cell Death/physiology , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Mitochondria/metabolism , Parkinson Disease/metabolism , Animals , Humans , Substantia Nigra/metabolism
15.
Lancet Neurol ; 14(8): 855-866, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26050140

ABSTRACT

Progressive neuronal cell loss in a small subset of brainstem and mesencephalic nuclei and widespread aggregation of the α-synuclein protein in the form of Lewy bodies and Lewy neurites are neuropathological hallmarks of Parkinson's disease. Most cases occur sporadically, but mutations in several genes, including SNCA, which encodes α-synuclein, are associated with disease development. The discovery and development of therapeutic strategies to block cell death in Parkinson's disease has been limited by a lack of understanding of the mechanisms driving neurodegeneration. However, increasing evidence of multiple pivotal roles of α-synuclein in the pathogenesis of Parkinson's disease has led researchers to consider the therapeutic potential of several strategies aimed at reduction of α-synuclein toxicity. We critically assess the potential of experimental therapies targeting α-synuclein, and discuss steps that need to be taken for target validation and drug development.


Subject(s)
Drug Discovery , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , alpha-Synuclein/metabolism , Animals , Humans , alpha-Synuclein/chemistry , alpha-Synuclein/genetics
16.
Lancet Neurol ; 14(4): 388-405, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25792098

ABSTRACT

Increasing evidence suggests that Alzheimer's disease pathogenesis is not restricted to the neuronal compartment, but includes strong interactions with immunological mechanisms in the brain. Misfolded and aggregated proteins bind to pattern recognition receptors on microglia and astroglia, and trigger an innate immune response characterised by release of inflammatory mediators, which contribute to disease progression and severity. Genome-wide analysis suggests that several genes that increase the risk for sporadic Alzheimer's disease encode factors that regulate glial clearance of misfolded proteins and the inflammatory reaction. External factors, including systemic inflammation and obesity, are likely to interfere with immunological processes of the brain and further promote disease progression. Modulation of risk factors and targeting of these immune mechanisms could lead to future therapeutic or preventive strategies for Alzheimer's disease.


Subject(s)
Alzheimer Disease , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Brain Injuries/complications , Immunity, Innate , Inflammation Mediators/metabolism , Inflammation/metabolism , Microglia/immunology , Microglia/pathology , Obesity/complications , Alzheimer Disease/genetics , Alzheimer Disease/immunology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/prevention & control , Animals , Astrocytes/immunology , Astrocytes/pathology , Biomarkers/blood , Biomarkers/cerebrospinal fluid , Brain Injuries/metabolism , Clinical Trials as Topic , Disease Models, Animal , Disease Progression , Humans , Immunization , Inflammation/diagnosis , Inflammation/immunology , Inflammation Mediators/immunology , Locus Coeruleus/pathology , Nootropic Agents/administration & dosage , Obesity/metabolism , Phagocytosis , Protein Folding , Risk Factors , Severity of Illness Index
17.
Nat Commun ; 5: 5181, 2014 Oct 21.
Article in English | MEDLINE | ID: mdl-25333748

ABSTRACT

Mitochondrial dysfunction is a common feature of many neurodegenerative disorders, notably Parkinson's disease. Consequently, agents that protect mitochondria have strong therapeutic potential. Here, we sought to divert the natural strategy used by Borna disease virus (BDV) to replicate in neurons without causing cell death. We show that the BDV X protein has strong axoprotective properties, thereby protecting neurons from degeneration both in tissue culture and in an animal model of Parkinson's disease, even when expressed alone outside of the viral context. We also show that intranasal administration of a cell-permeable peptide derived from the X protein is neuroprotective. We establish that both the X protein and the X-derived peptide act by buffering mitochondrial damage and inducing enhanced mitochondrial filamentation. Our results open the way to novel therapies for neurodegenerative diseases by targeting mitochondrial dynamics and thus preventing the earliest steps of neurodegenerative processes in axons.


Subject(s)
Mitochondria/pathology , Neurodegenerative Diseases/prevention & control , Parkinson Disease/prevention & control , Peptides/chemistry , Viral Nonstructural Proteins/chemistry , Animals , Axons/metabolism , Axons/physiology , Disease Models, Animal , Female , HEK293 Cells , Humans , Male , Membrane Potential, Mitochondrial , Mice , Mice, Inbred C57BL , Microfluidics , Microscopy, Confocal , Microscopy, Fluorescence , Neurodegenerative Diseases/virology , Neurons/metabolism , Parkinson Disease/virology , Phosphorylation , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Rotenone/chemistry
18.
J Neuroinflammation ; 11: 86, 2014 May 08.
Article in English | MEDLINE | ID: mdl-24886419

ABSTRACT

BACKGROUND: Increasing evidence suggests that inflammation associated with microglial cell activation in the substantia nigra (SN) of patients with Parkinson disease (PD) is not only a consequence of neuronal degeneration, but may actively sustain dopaminergic (DA) cell loss over time. We aimed to study whether the intracellular chaperone heat shock protein 60 (Hsp60) could serve as a signal of CNS injury for activation of microglial cells. METHODS: Hsp60 mRNA expression in the mesencephalon and the striatum of C57/BL6 mice treated with MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) and the Hsp60/TH mRNA ratios in the SN of PD patients and aged-matched subjects were measured. To further investigate a possible link between the neuronal Hsp60 response and PD-related cellular stress, Hsp60 immunoblot analysis and quantification in cell lysates from SH-SY5Y after treatment with 100 µM MPP+ (1-methyl-4-phenylpyridinium) at different time points (6, 12, 24 and 48 hours) compared to control cells were performed. Additional MTT and LDH assay were used. We next addressed the question as to whether Hsp60 influences the survival of TH+ neurons in mesencephalic neuron-glia cultures treated either with MPP+ (1 µM), hHsp60 (10 µg/ml) or a combination of both. Finally, we measured IL-1ß, IL-6, TNF-α and NO-release by ELISA in primary microglial cell cultures following treatment with different hHsp60 preparations. Control cultures were exposed to LPS. RESULTS: In the mesencephalon and striatum of mice treated with MPTP and also in the SN of PD patients, we found that Hsp60 mRNA was up-regulated. MPP+, the active metabolite of MPTP, also caused an increased expression and release of Hsp60 in the human dopaminergic cell line SH-SY5Y. Interestingly, in addition to being toxic to DA neurons in primary mesencephalic cultures, exogenous Hsp60 aggravated the effects of MPP+. Yet, although we demonstrated that Hsp60 specifically binds to microglial cells, it failed to stimulate the production of pro-inflammatory cytokines or NO by these cells. CONCLUSIONS: Overall, our data suggest that Hsp60 is likely to participate in DA cell death in PD but via a mechanism unrelated to cytokine release.


Subject(s)
Chaperonin 60/metabolism , Corpus Striatum/pathology , Dopaminergic Neurons/metabolism , MPTP Poisoning/pathology , Mesencephalon/pathology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Cell Death/drug effects , Cells, Cultured , Chaperonin 60/genetics , Disease Models, Animal , Dopamine Agents/pharmacology , L-Lactate Dehydrogenase/metabolism , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Nitric Oxide/metabolism , Protein Binding/drug effects , RNA, Messenger/metabolism , Tyrosine 3-Monooxygenase/metabolism
19.
J Neuroinflammation ; 10: 82, 2013 Jul 11.
Article in English | MEDLINE | ID: mdl-23844828

ABSTRACT

BACKGROUND: Parkinson's disease (PD) is a neurodegenerative disorder characterized by a loss of dopaminergic neurons (DN) in the substantia nigra (SN). Several lines of evidence suggest that apoptotic cell death of DN is driven in part by non-cell autonomous mechanisms orchestrated by microglial cell-mediated inflammatory processes. Although the mechanisms and molecular network underlying this deleterious cross-talk between DN and microglial cells remain largely unknown, previous work indicates that, upon DN injury, activation of the ß2 integrin subunit CD11b is required for microglia-mediated DN cell death. Interestingly, during brain development, the CD11b integrin is also involved in microglial induction of neuronal apoptosis and has been shown to act in concert with the DAP12 immunoreceptor. Whether such a developmental CD11b/DAP12 pathway could be reactivated in a pathological context such as PD and play a role in microglia-induced DN cell death is a tantalizing hypothesis that we wished to test in this study. METHODS: To test the possibility that DAP12 could be involved in microglia-associated DN injury, we used both in vitro and in vivo toxin-based experimental models of PD recapitulating microglial-mediated non-cell autonomous mechanisms of DN cell death. In vitro, enriched mesencephalic neuronal/microglial co-cultures were exposed to the dopaminergic neurotoxin 1-methyl-4-phenylpyridinium (MPP+) whereas in vivo, mice were administrated with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) according to acute or subchronic mode. Mice deficient for DAP12 or CD11b were used to determine the pathological function of the CD11b/DAP12 pathway in our disease models. RESULTS: Our results show that DAP12 and CD11b partially contribute to microglia-induced DN cell death in vitro. Yet, in vivo, mice deficient for either of these factors develop similar neuropathological alterations as their wild-type counterparts in two different MPTP mouse models of PD. CONCLUSION: Overall, our data suggest that DAP12 and CD11b contribute to microglial-induced DN cell death in vitro but not in vivo in the MPTP mouse model of PD. Therefore, the CD11b/DAP12 pathway may not be considered as a promising therapeutic target for PD.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , CD11b Antigen/metabolism , Disease Models, Animal , Dopaminergic Neurons/metabolism , Microglia/metabolism , Parkinsonian Disorders/metabolism , Adaptor Proteins, Signal Transducing/deficiency , Animals , Cell Death/physiology , Cells, Cultured , Coculture Techniques , Dopaminergic Neurons/pathology , Gene Knock-In Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/pathology , Parkinsonian Disorders/pathology
20.
Sci Rep ; 3: 1393, 2013.
Article in English | MEDLINE | ID: mdl-23462811

ABSTRACT

In mammalians, toll-like receptors (TLR) signal-transduction pathways induce the expression of a variety of immune-response genes, including inflammatory cytokines. It is therefore plausible to assume that TLRs are mediators in glial cells triggering the release of cytokines that ultimately kill DA neurons in the substantia nigra in Parkinson disease (PD). Accordingly, recent data indicate that TLR4 is up-regulated by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) treatment in a mouse model of PD. Here, we wished to evaluate the role of TLR4 in the acute mouse MPTP model of PD: TLR4-deficient mice and wild-type littermates control mice were used for the acute administration way of MPTP or a corresponding volume of saline. We demonstrate that TLR4-deficient mice are less vulnerable to MPTP intoxication than wild-type mice and display a decreased number of Iba1+ and MHC II+ activated microglial cells after MPTP application, suggesting that the TLR4 pathway is involved in experimental PD.


Subject(s)
Parkinsonian Disorders/metabolism , Toll-Like Receptor 4/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/adverse effects , Animals , Cell Death/genetics , Corpus Striatum/metabolism , Dopamine/metabolism , Homovanillic Acid/metabolism , Mice , Mice, Knockout , Microglia/metabolism , Parkinsonian Disorders/genetics , Toll-Like Receptor 4/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...