Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Front Pharmacol ; 15: 1379965, 2024.
Article in English | MEDLINE | ID: mdl-38576478

ABSTRACT

Almost all individuals with Down's syndrome (DS) show the characteristic neuropathological features of Alzheimer's disease (AD) by the age of 40, yet not every individual with DS experiences symptoms of AD later in life. Similar to neurotypical developing subjects, AD in people with DS lasts for a long preclinical phase in which biomarkers follow a predictable order of changes. Hence, a prolonged asymptomatic period precedes the onset of dementia, underscoring the importance of identifying new biomarkers for the early detection and monitoring of cognitive decline in individuals with DS. Blood-based biomarkers may offer an alternative non-invasive strategy for the detection of peripheral biological alterations paralleling nervous system pathology in an early phase of the AD continuum. In the last few years, a strong neurobiological link has been demonstrated between the deficit of transforming growth factor-ß1 (TGF-ß1) levels, an anti-inflammatory cytokine endowed with neuroprotective activity, and early pro-inflammatory processes in the AD brain. In this clinical prospective observational study, we found significant lower plasma TGF-ß1 concentrations at the first neuropsychological evaluation (baseline = T0) both in young adult DS individuals (19-35 years) and older DS subjects without AD (35-60 years) compared to age- and sex-matched healthy controls. Interestingly, we found that the lower TGF-ß1 plasma concentrations at T0 were strongly correlated with the following cognitive decline at 12 months. In addition, in young individuals with DS, we found, for the first time, a negative correlation between low TGF-ß1 concentrations and high TNF-α plasma concentrations, a pro-inflammatory cytokine that is known to be associated with cognitive impairment in DS individuals with AD. Finally, adopting an ex vivo approach, we found that TGF-ß1 concentrations were reduced in parallel both in the plasma and in the peripheral blood mononuclear cells (PBMCs) of DS subjects, and interestingly, therapeutic concentrations of fluoxetine (FLX) applied to cultured PBMCs (1 µM for 24 h) were able to rescue TGF-ß1 concentrations in the culture media from DS PBMCs, suggesting that FLX, a selective serotonin reuptake inhibitor (SSRI) endowed with neuroprotective activity, might rescue TGF-ß1 concentrations in DS subjects at higher risk to develop cognitive decline.

2.
Adv Healthc Mater ; 11(20): e2201203, 2022 10.
Article in English | MEDLINE | ID: mdl-35856921

ABSTRACT

Extracellular vesicles (EVs) are emerging as powerful players in cell-to-cell communication both in healthy and diseased brain. In Parkinson's disease (PD)-characterized by selective dopaminergic neuron death in ventral midbrain (VMB) and degeneration of their terminals in striatum (STR)-astrocytes exert dual harmful/protective functions, with mechanisms not fully elucidated. Here, this study shows that astrocytes from the VMB-, STR-, and VMB/STR-depleted brains release a population of small EVs  in a region-specific manner. Interestingly, VMB-astrocytes secreted the highest rate of EVs, which is further exclusively increased in response to CCL3, a chemokine that promotes robust dopaminergic neuroprotection in different PD models. The neuroprotective potential of nigrostriatal astrocyte-EVs is investigated in differentiated versus undifferentiated SH-SY5Y cells exposed to oxidative stress and mitochondrial toxicity. EVs from both VMB- and STR-astrocytes counteract H2 O2 -induced caspase-3 activation specifically in differentiated cells, with EVs from CCL3-treated astrocytes showing a higher protective effect. High resolution respirometry further reveals that nigrostriatal astrocyte-EVs rescue neuronal mitochondrial complex I function impaired by the neurotoxin MPP+ . Notably, only EVs from VMB-astrocyte fully restore ATP production, again specifically in differentiated SH-SY5Y. These results highlight a regional diversity in the nigrostriatal system for the secretion and activities of astrocyte-EVs, with neuroprotective implications for PD.


Subject(s)
Extracellular Vesicles , Neuroblastoma , Parkinson Disease , Humans , Astrocytes/metabolism , Parkinson Disease/metabolism , Neurotoxins/metabolism , Neurotoxins/pharmacology , Caspase 3/metabolism , Neuroblastoma/metabolism , Dopaminergic Neurons/metabolism , Mitochondria , Cell Death , Extracellular Vesicles/metabolism , Dopamine/pharmacology , Adenosine Triphosphate/metabolism
3.
Article in English | MEDLINE | ID: mdl-34501904

ABSTRACT

Reliability, accuracy, and timeliness of diagnostic testing for SARS-CoV-2 infection have allowed adequate public health management of the disease, thus notably helping the timely mapping of viral spread within the community. Furthermore, the most vulnerable populations, such as people with intellectual disability and dementia, represent a high-risk group across multiple dimensions, including a higher prevalence of pre-existing conditions, lower health maintenance, and a propensity for rapid community spread. This led to an urgent need for reliable in-house rapid testing to be performed prior to hospital admission. In the present study, we describe a pooling procedure in which oropharyngeal and nasopharyngeal swabs for SARS-CoV-2 detection (performed prior to hospital admission using rapid RT-PCR assay) are pooled together at the time of sample collection. Sample pooling (groups of 2-4 samples per tube) allowed us to significantly reduce response times, consumables, and personnel costs while maintaining the same test sensitivity.


Subject(s)
COVID-19 , Intellectual Disability , Hospitals , Humans , Intellectual Disability/diagnosis , Intellectual Disability/epidemiology , Real-Time Polymerase Chain Reaction , Reproducibility of Results , SARS-CoV-2 , Sensitivity and Specificity
4.
J Musculoskelet Neuronal Interact ; 20(4): 610-613, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33265090

ABSTRACT

Spinal muscular atrophy (SMA) refers to a group of genetic neuromuscular disorders affecting lower motor neurons causative of numerous phenotypes. To date, according to the age of onset, maximum muscular activity achieved, and life expectation four types of SMA are recognized, all caused by mutations in the SMN1 gene with SMN2 copy number influencing disease severity. Herein, we describe the case of a 31-year-old young male with normal psychomotor development who has experienced fatigue, cramps, and muscle fasciculations in the lower limbs for a period of 2 months. Based on electrophysiological and clinical findings we performed SMA genetic, clinical exome and RNA expression of candidate genes which led us to suggest SMN1-SMN2 genes [(2+0) and (0+0)] combination as possibly being implicated in the phenotype.


Subject(s)
Muscular Atrophy, Spinal/genetics , Survival of Motor Neuron 1 Protein/genetics , Adult , Electromyography , Genotype , Humans , Male , Mutation , Survival of Motor Neuron 2 Protein/economics
5.
Biomolecules ; 10(9)2020 09 16.
Article in English | MEDLINE | ID: mdl-32948090

ABSTRACT

Extracellular vesicles (EVs) are naturally occurring membranous structures secreted by normal and diseased cells, and carrying a wide range of bioactive molecules. In the central nervous system (CNS), EVs are important in both homeostasis and pathology. Through receptor-ligand interactions, direct fusion, or endocytosis, EVs interact with their target cells. Accumulating evidence indicates that EVs play crucial roles in the pathogenesis of many neurodegenerative disorders (NDs), including Parkinson's disease (PD). PD is the second most common ND, characterized by the progressive loss of dopaminergic (DAergic) neurons within the Substantia Nigra pars compacta (SNpc). In PD, EVs are secreted by both neurons and glial cells, with either beneficial or detrimental effects, via a complex program of cell-to-cell communication. The functions of EVs in PD range from their etiopathogenetic relevance to their use as diagnostic tools and innovative carriers of therapeutics. Because they can cross the blood-brain barrier, EVs can be engineered to deliver bioactive molecules (e.g., small interfering RNAs, catalase) within the CNS. This review summarizes the latest findings regarding the role played by EVs in PD etiology, diagnosis, prognosis, and therapy, with a particular focus on their use as novel PD nanotherapeutics.


Subject(s)
Brain/metabolism , Extracellular Vesicles/metabolism , Nanoparticles/metabolism , Neurodegenerative Diseases/metabolism , Parkinson Disease/metabolism , Brain/pathology , Cell Communication/drug effects , Humans , Inflammation/drug therapy , Inflammation/metabolism , Models, Biological , Nanoparticles/therapeutic use , Neurodegenerative Diseases/drug therapy , Parkinson Disease/drug therapy , Signal Transduction/drug effects
6.
J Clin Med ; 9(6)2020 Jun 21.
Article in English | MEDLINE | ID: mdl-32575923

ABSTRACT

Glial cells are fundamental players in the central nervous system (CNS) development and homeostasis, both in health and disease states. In Parkinson's disease (PD), a dysfunctional glia-neuron crosstalk represents a common final pathway contributing to the chronic and progressive death of dopaminergic (DAergic) neurons of the substantia nigra pars compacta (SNpc). Notably, glial cells communicating with each other by an array of molecules, can acquire a "beneficial" or "destructive" phenotype, thereby enhancing neuronal death/vulnerability and/or exerting critical neuroprotective and neuroreparative functions, with mechanisms that are actively investigated. An important way of delivering messenger molecules within this glia-neuron cross-talk consists in the secretion of extracellular vesicles (EVs). EVs are nano-sized membranous particles able to convey a wide range of molecular cargoes in a controlled way, depending on the specific donor cell and the microenvironmental milieu. Given the dual role of glia in PD, glia-derived EVs may deliver molecules carrying various messages for the vulnerable/dysfunctional DAergic neurons. Here, we summarize the state-of-the-art of glial-neuron interactions and glia-derived EVs in PD. Also, EVs have the ability to cross the blood brain barrier (BBB), thus acting both within the CNS and outside, in the periphery. In these regards, this review discloses the emerging applications of EVs, with a special focus on glia-derived EVs as potential carriers of new biomarkers and nanotherapeutics for PD.

7.
Front Aging Neurosci ; 12: 24, 2020.
Article in English | MEDLINE | ID: mdl-32226376

ABSTRACT

Astrocyte (As) bidirectional dialog with neurons plays a fundamental role in major homeostatic brain functions, particularly providing metabolic support and antioxidant self-defense against reactive oxygen (ROS) and nitrogen species (RNS) via the activation of NF-E2-related factor 2 (Nrf2), a master regulator of oxidative stress. Disruption of As-neuron crosstalk is chiefly involved in neuronal degeneration observed in Parkinson's disease (PD), the most common movement disorder characterized by the selective degeneration of dopaminergic (DAergic) cell bodies of the substantia nigra (SN) pars compacta (SNpc). Ventral midbrain (VM)-As are recognized to exert an important role in DAergic neuroprotection via the expression of a variety of factors, including wingless-related MMTV integration site 1 (Wnt1), a principal player in DAergic neurogenesis. However, whether As, by themselves, might fulfill the role of chief players in DAergic neurorestoration of aged PD mice is presently unresolved. Here, we used primary postnatal mouse VM-As as a graft source for unilateral transplantation above the SN of aged 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mice after the onset of motor symptoms. Spatio-temporal analyses documented that the engrafted cells promoted: (i) a time-dependent nigrostriatal rescue along with increased high-affinity synaptosomal DA uptake and counteraction of motor deficit, as compared to mock-grafted counterparts; and (ii) a restoration of the impaired microenvironment via upregulation of As antioxidant self-defense through the activation of Nrf2/Wnt/ß-catenin signaling, suggesting that grafting As has the potential to switch the SN neurorescue-unfriendly environment to a beneficial antioxidant/anti-inflammatory prosurvival milieu. These findings highlight As-derived factors/mechanisms as the crucial key for successful therapeutic outcomes in PD.

8.
Aging Cell ; 19(3): e13101, 2020 03.
Article in English | MEDLINE | ID: mdl-32050297

ABSTRACT

A common hallmark of age-dependent neurodegenerative diseases is an impairment of adult neurogenesis. Wingless-type mouse mammary tumor virus integration site (Wnt)/ß-catenin (WßC) signalling is a vital pathway for dopaminergic (DAergic) neurogenesis and an essential signalling system during embryonic development and aging, the most critical risk factor for Parkinson's disease (PD). To date, there is no known cause or cure for PD. Here we focus on the potential to reawaken the impaired neurogenic niches to rejuvenate and repair the aged PD brain. Specifically, we highlight WßC-signalling in the plasticity of the subventricular zone (SVZ), the largest germinal region in the mature brain innervated by nigrostriatal DAergic terminals, and the mesencephalic aqueduct-periventricular region (Aq-PVR) Wnt-sensitive niche, which is in proximity to the SNpc and harbors neural stem progenitor cells (NSCs) with DAergic potential. The hallmark of the WßC pathway is the cytosolic accumulation of ß-catenin, which enters the nucleus and associates with T cell factor/lymphoid enhancer binding factor (TCF/LEF) transcription factors, leading to the transcription of Wnt target genes. Here, we underscore the dynamic interplay between DAergic innervation and astroglial-derived factors regulating WßC-dependent transcription of key genes orchestrating NSC proliferation, survival, migration and differentiation. Aging, inflammation and oxidative stress synergize with neurotoxin exposure in "turning off" the WßC neurogenic switch via down-regulation of the nuclear factor erythroid-2-related factor 2/Wnt-regulated signalosome, a key player in the maintenance of antioxidant self-defense mechanisms and NSC homeostasis. Harnessing WßC-signalling in the aged PD brain can thus restore neurogenesis, rejuvenate the microenvironment, and promote neurorescue and regeneration.


Subject(s)
Aging/metabolism , Lateral Ventricles/metabolism , Neurogenesis , Parkinson Disease/metabolism , Wnt Signaling Pathway , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Astrocytes/metabolism , Dopaminergic Neurons/metabolism , Humans , Microglia/metabolism , Neural Stem Cells/metabolism , Neuronal Plasticity , Parkinsonian Disorders/metabolism
9.
Stem Cells ; 36(8): 1179-1197, 2018 08.
Article in English | MEDLINE | ID: mdl-29575325

ABSTRACT

During aging-one the most potent risk factors for Parkinson's disease (PD)-both astrocytes and microglia undergo functional changes that ultimately hamper homoeostasis, defense, and repair of substantia nigra pars compacta (SNpc) midbrain dopaminergic (mDA) neurons. We tested the possibility of rejuvenating the host microenvironment and boosting SNpc DA neuronal plasticity via the unilateral transplantation of syngeneic neural stem/progenitor cells (NSCs) in the SNpc of aged mice with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced experimental PD. Transplanted NSCs within the aged SNpc engrafted and migrated in large proportions to the tegmental aqueduct mDA niche, with 30% acquiring an astroglial phenotype. Both graft-derived exogenous (ex-Astro) and endogenous astrocytes (en-Astro) expressed Wnt1. Both ex-Astro and en-Astro were key triggers of Wnt/ß-catenin signaling in SNpc-mDA neurons and microglia, which was associated with mDA neurorescue and immunomodulation. At the aqueduct-ventral tegmental area level, NSC grafts recapitulated a genetic Wnt1-dependent mDA developmental program, inciting the acquisition of a mature Nurr1+ TH+ neuronal phenotype. Wnt/ß-catenin signaling antagonism abolished mDA neurorestoration and immune modulatory effects of NSC grafts. Our work implicates an unprecedented therapeutic potential for somatic NSC grafts in the restoration of mDA neuronal function in the aged Parkinsonian brain. Stem Cells 2018;36:1179-1197.


Subject(s)
Aging/pathology , Astrocytes/pathology , Brain/pathology , Neural Stem Cells/transplantation , Parkinson Disease/pathology , Parkinson Disease/therapy , Wnt Signaling Pathway , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Astrocytes/metabolism , Cell Death , Cell Differentiation/genetics , Cell Lineage , Cell Proliferation , Cell Survival , Disease Models, Animal , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Down-Regulation/genetics , Genes, Developmental , Inflammation/genetics , Inflammation/pathology , Inflammation Mediators/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Microglia/metabolism , Microglia/pathology , Neural Stem Cells/cytology , Oxidative Stress/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Substantia Nigra/pathology , Synaptosomes/metabolism , Tyrosine 3-Monooxygenase/metabolism , Wnt Signaling Pathway/genetics
10.
Front Aging Neurosci ; 10: 12, 2018.
Article in English | MEDLINE | ID: mdl-29483868

ABSTRACT

Neuroinflammatory processes are recognized key contributory factors in Parkinson's disease (PD) physiopathology. While the causes responsible for the progressive loss of midbrain dopaminergic (mDA) neuronal cell bodies in the subtantia nigra pars compacta are poorly understood, aging, genetics, environmental toxicity, and particularly inflammation, represent prominent etiological factors in PD development. Especially, reactive astrocytes, microglial cells, and infiltrating monocyte-derived macrophages play dual beneficial/harmful effects, via a panel of pro- or anti-inflammatory cytokines, chemokines, neurotrophic and neurogenic transcription factors. Notably, with age, microglia may adopt a potent neurotoxic, pro-inflammatory "primed" (M1) phenotype when challenged with inflammatory or neurotoxic stimuli that hamper brain's own restorative potential and inhibit endogenous neurorepair mechanisms. In the last decade we have provided evidence for a major role of microglial crosstalk with astrocytes, mDA neurons and neural stem progenitor cells (NSCs) in the MPTP- (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-) mouse model of PD, and identified Wnt/ß-catenin signaling, a pivotal morphogen for mDA neurodevelopment, neuroprotection, and neuroinflammatory modulation, as a critical actor in glia-neuron and glia-NSCs crosstalk. With age however, Wnt signaling and glia-NSC-neuron crosstalk become dysfunctional with harmful consequences for mDA neuron plasticity and repair. These findings are of importance given the deregulation of Wnt signaling in PD and the emerging link between most PD related genes, Wnt signaling and inflammation. Especially, in light of the expanding field of microRNAs and inflammatory PD-related genes as modulators of microglial-proinflammatory status, uncovering the complex molecular circuitry linking PD and neuroinflammation will permit the identification of new druggable targets for the cure of the disease. Here we summarize recent findings unveiling major microglial inflammatory and oxidative stress pathways converging in the regulation of Wnt/ß-catenin signaling, and reciprocally, the ability of Wnt signaling pathways to modulate microglial activation in PD. Unraveling the key factors and conditons promoting the switch of the proinflammatory M1 microglia status into a neuroprotective and regenerative M2 phenotype will have important consequences for neuroimmune interactions and neuronal outcome under inflammatory and/or neurodegenerative conditions.

11.
Int J Mol Sci ; 18(12)2017 Dec 13.
Article in English | MEDLINE | ID: mdl-29236052

ABSTRACT

Parkinson's disease (PD) is the most prevalent central nervous system (CNS) movement disorder and the second most common neurodegenerative disease overall. PD is characterized by the progressive loss of dopaminergic (DAergic) neurons in the substantia nigra pars compacta (SNpc) within the midbrain, accumulation of alpha-synuclein (α-SYN) in Lewy bodies and neurites and excessive neuroinflammation. The neurodegenerative processes typically begin decades before the appearance of clinical symptoms. Therefore, the diagnosis is achievable only when the majority of the relevant DAergic neurons have already died and for that reason available treatments are only palliative at best. The causes and mechanism(s) of this devastating disease are ill-defined but complex interactions between genetic susceptibility and environmental factors are considered major contributors to the etiology of PD. In addition to the role of classical gene mutations in PD, the importance of regulatory elements modulating gene expression has been increasingly recognized. One example is the critical role played by microRNAs (miRNAs) in the development and homeostasis of distinct populations of neurons within the CNS and, in particular, in the context of PD. Recent reports demonstrate how distinct miRNAs are involved in the regulation of PD genes, whereas profiling approaches are unveiling variations in the abundance of certain miRNAs possibly relevant either to the onset or to the progression of the disease. In this review, we provide an overview of the miRNAs recently found to be implicated in PD etiology, with particular focus on their potential relevance as PD biomarkers, as well as their possible use in PD targeted therapy.


Subject(s)
MicroRNAs/metabolism , Parkinson Disease/diagnosis , Antagomirs/therapeutic use , Biomarkers/blood , Biomarkers/cerebrospinal fluid , Biomarkers/metabolism , Exosomes/metabolism , Humans , MicroRNAs/analysis , MicroRNAs/antagonists & inhibitors , Neuroprotective Agents/therapeutic use , Parkinson Disease/pathology , Parkinson Disease/therapy , Protein Deglycase DJ-1/antagonists & inhibitors , Protein Deglycase DJ-1/genetics , Protein Deglycase DJ-1/metabolism , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , alpha-Synuclein/antagonists & inhibitors , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
12.
Stem Cells ; 32(8): 2147-63, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24648001

ABSTRACT

Wnt/ß-catenin signaling is required for specification and neurogenesis of midbrain dopaminergic (mDA) neurons, the pivotal neuronal population that degenerates in Parkinson's disease (PD), and in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD. Wnt/ß-catenin signaling plays a vital role in adult neurogenesis but whether it might engage DA neurogenesis/neurorepair in the affected PD brain is yet unresolved. Recently, the adult midbrain aqueduct periventricular regions (Aq-PVRs) were shown to harbor multipotent clonogenic neural stem/progenitor cells (mNPCs) with DA potential in vitro, but restrictive mechanisms in vivo are believed to limit their DA regenerative capacity. Using in vitro mNPC culture systems we herein demonstrate that aging is one most critical factor restricting mNPC neurogenic potential via dysregulation of Wnt/ß-catenin signaling. Coculture paradigms between young/aged (Y/A) mNPCs and Y/A astrocytes identified glial age and a decline of glial-derived factors including Wnts as key determinants of impaired neurogenic potential, whereas Wnt activation regimens efficiently reversed the diminished proliferative, neuronal, and DA differentiation potential of A-mNPCs. Next, in vivo studies in wild (Wt) and transgenic ß-catenin reporter mice uncovered Wnt/ß-catenin signaling activation and remarkable astrocyte remodeling of Aq-PVR in response to MPTP-induced DA neuron death. Spatio-temporal analyses unveiled ß-catenin signaling in predopaminergic (Nurr1(+)/TH(-)) and imperiled or rescuing DAT(+) neurons during MPTP-induced DA neuron injury and self-repair. Aging inhibited Wnt signaling, whereas ß-catenin activation in situ with a specific GSK-3ß antagonist promoted a significant degree of DA neurorestoration associated with reversal of motor deficit, with implications for neurorestorative approaches in PD.


Subject(s)
Aging/metabolism , Neural Stem Cells/metabolism , Neurogenesis/physiology , Parkinson Disease/metabolism , Wnt Signaling Pathway/physiology , Animals , Blotting, Western , Coculture Techniques , Disease Models, Animal , Dopaminergic Neurons/metabolism , Immunohistochemistry , Male , Mesencephalon/metabolism , Mesencephalon/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuroglia/metabolism , Parkinson Disease/physiopathology , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
13.
J Mol Cell Biol ; 6(1): 13-26, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24431301

ABSTRACT

During the past three decades, the Wingless-type MMTV integration site (Wnt) signaling cascade has emerged as an essential system regulating multiple processes in developing and adult brain. Accumulating evidence points to a dysregulation of Wnt signaling in major neurodegenerative pathologies including Parkinson's disease (PD), a common neurodegenerative disorder characterized by the progressive loss of midbrain dopaminergic (mDA) neurons and deregulated activation of astrocytes and microglia. This review highlights the emerging link between Wnt signaling and key inflammatory pathways during mDA neuron damage/repair in PD progression. In particular, we summarize recent evidence documenting that aging and neurotoxicant exposure strongly antagonize Wnt/ß-catenin signaling in mDA neurons and subventricular zone (SVZ) neuroprogenitors via astrocyte-microglial interactions. Dysregulation of the crosstalk between Wnt/ß-catenin signaling and anti-oxidant/anti-inflammatory pathways delineate novel mechanisms driving the decline of SVZ plasticity with age and the limited nigrostriatal dopaminergic self-repair in PD. These findings hold a promise in developing therapies that target Wnt/ß-catenin signaling to enhance endogenous restoration and neuronal outcome in age-dependent diseases, such as PD.


Subject(s)
Dopaminergic Neurons/physiology , Nerve Regeneration/genetics , Neuroimmunomodulation/genetics , Parkinson Disease/metabolism , Brain/metabolism , Brain/pathology , Cell Survival , Cellular Microenvironment , Models, Biological , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Parkinson Disease/genetics
14.
Eur J Neurosci ; 37(10): 1550-63, 2013 May.
Article in English | MEDLINE | ID: mdl-23461676

ABSTRACT

Parkinson's disease (PD) is a common neurodegenerative disorder characterized by progressive loss of dopaminergic (DAergic) neuronal cell bodies in the substantia nigra pars compacta and gliosis. The cause and mechanisms underlying the demise of nigrostriatal DAergic neurons are ill-defined, but interactions between genes and environmental factors are recognized to play a critical role in modulating the vulnerability to PD. Current evidence points to reactive glia as a pivotal factor in PD pathophysiology, playing both protective and destructive roles. Here, the contribution of reactive astrocytes and their ability to modulate DAergic neurodegeneration, neuroprotection and neurorepair in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) rodent model of PD will be discussed in the light of novel emerging evidence implicating wingless-type mouse mammary tumor virus integration site (Wnt)/ß-catenin signaling as a strong candidate in MPTP-induced nigrostriatal DAergic plasticity. In this work, we highlight an intrinsic Wnt1/frizzled-1/ß-catenin tone that critically contributes to the survival and protection of adult midbrain DAergic neurons, with potential implications for drug design or drug action in PD. The dynamic interplay between astrocyte-derived factors and neurogenic signals in MPTP-induced nigrostriatal DAergic neurotoxicity and repair will be summarized, together with recent findings showing a critical role of glia-neural stem/progenitor cell (NPC) interactions aimed at overcoming neurodegeneration and inducing neurorestoration. Understanding the intrinsic plasticity of nigrostriatal DAergic neurons and deciphering the signals facilitating the crosstalk between astrocytes, microglia, DAergic neurons and NPCs may have major implications for the role of stem cell technology in PD, and for identifying potential therapeutic targets to induce endogenous neurorepair.


Subject(s)
Astrocytes/metabolism , MPTP Poisoning/metabolism , Neurons/metabolism , Wnt Signaling Pathway , Animals , MPTP Poisoning/pathology , Mice
15.
J Neurosci ; 33(4): 1462-85, 2013 Jan 23.
Article in English | MEDLINE | ID: mdl-23345222

ABSTRACT

Aging and exposure to environmental toxins including MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) are strong risk factors for developing Parkinson's disease (PD), a common neurologic disorder characterized by selective degeneration of midbrain dopaminergic (DAergic) neurons and astrogliosis. Aging and PD impair the subventricular zone (SVZ), one of the most important brain regions for adult neurogenesis. Because inflammation and oxidative stress are the hallmarks of aging and PD, we investigated the nature, timing, and signaling mechanisms contributing to aging-induced SVZ stem/neuroprogenitor cell (NPC) inhibition in aging male mice and attempted to determine to what extent manipulation of these pathways produces a functional response in the outcome of MPTP-induced DAergic toxicity. We herein reveal an imbalance of Nrf2-driven antioxidant/anti-inflammatory genes, such as Heme oxygenase1 in the SVZ niche, starting by middle age, amplified upon neurotoxin treatment and associated with an exacerbated proinflammatory SVZ microenvironment converging to dysregulate the Wingless-type MMTV integration site (Wnt)/ß-catenin signaling, a key regulatory pathway for adult NPCs. In vitro experiments using coculture paradigms uncovered aged microglial proinflammatory mediators as critical inhibitors of NPC proliferative potential. We also found that interruption of PI3K (phosphatidylinositol3-kinase)/Akt and the Wnt/Fzd/ß-catenin signaling cascades, which switch glycogen synthase kinase 3ß (GSK-3ß) activation on and off, were causally related to the impairment of SVZ-NPCs. Moreover, a synergy between dysfunctional microglia of aging mice and MPTP exposure further inhibited astrocyte proneurogenic properties, including the expression of key Wnts components. Last, pharmacological activation/antagonism studies in vivo and in vitro suggest the potential that aged SVZ manipulation is associated with DAergic functional recovery.


Subject(s)
Aging/metabolism , Mesencephalon/metabolism , Neural Stem Cells/metabolism , Parkinsonian Disorders/metabolism , Signal Transduction/physiology , Animals , Blotting, Western , Cells, Cultured , Coculture Techniques , Immunohistochemistry , In Situ Nick-End Labeling , Male , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/metabolism , Neuroglia/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Real-Time Polymerase Chain Reaction , Wnt Signaling Pathway/physiology , beta Catenin/metabolism
16.
J Neurosci ; 32(6): 2062-85, 2012 Feb 08.
Article in English | MEDLINE | ID: mdl-22323720

ABSTRACT

In Parkinson's disease (PD), neurogenesis is impaired in the subventricular zone (SVZ) of postmortem human PD brains, in primate nonhuman and rodent models of PD. The vital role of Wingless-type MMTV integration site (Wnt)/ß-catenin signaling in the modulation of neurogenesis, neuroprotection, and synaptic plasticity coupled to our recent findings uncovering an active role for inflammation and Wnt/ß-catenin signaling in MPTP-induced loss and repair of nigrostriatal dopaminergic (DAergic) neurons prompted us to study the impact of neuroinflammation and the Wnt/ß-catenin pathway in the response of SVZ neuroprogenitors (NPCs) in MPTP-treated mice. In vivo experiments, using bromodeoxyuridine and cell-specific markers, and ex vivo time course analyses documented an inverse correlation between the reduced proliferation of NPCs and the generation of new neuroblasts with the phase of maximal exacerbation of microglia reaction, whereas a shift in the microglia proinflammatory phenotype correlated with a progressive NPC recovery. Ex vivo and in vitro experiments using microglia-NPC coculture paradigms pointed to NADPH-oxidase (gpPHOX(91)), a major source of microglial ROS, and reactive nitrogen species as candidate inhibitors of NPC neurogenic potential via the activation of glycogen synthase 3 (pGSK-3ß(Tyr216)), leading to loss of ß-catenin, a chief downstream transcriptional effector. Accordingly, MPTP/MPP(+) (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) caused ß-catenin downregulation and pGSK-3ß(Tyr216) overexpression, whereas manipulation of Wnt/ß-catenin signaling with RNA interference-mediated GSK-3ß knockdown or GSK-3ß antagonism reversed MPTP-induced neurogenic impairment ex vivo/in vitro or in vivo. Reciprocally, pharmacological modulation of inflammation prevented ß-catenin downregulation and restored neurogenesis, suggesting the possibility to modulate this endogenous system with potential consequences for DAergic neuroprotection and self-repair.


Subject(s)
Inflammation Mediators/administration & dosage , MPTP Poisoning/metabolism , Neuronal Plasticity/physiology , Parkinson Disease/metabolism , Receptor Cross-Talk/physiology , Stem Cells/metabolism , Wnt Signaling Pathway/physiology , beta Catenin/antagonists & inhibitors , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/administration & dosage , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/therapeutic use , Animals , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Gene Knockdown Techniques/methods , Inflammation Mediators/physiology , MPTP Poisoning/drug therapy , MPTP Poisoning/pathology , Male , Mice , Mice, Inbred C57BL , Neurogenesis/drug effects , Neurogenesis/physiology , Neuronal Plasticity/drug effects , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/therapeutic use , Parkinson Disease/drug therapy , Parkinson Disease/pathology , Receptor Cross-Talk/drug effects , Stem Cells/drug effects , Stem Cells/pathology , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism
17.
Rejuvenation Res ; 14(4): 411-24, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21793734

ABSTRACT

Aging represents a major risk factor for the development and progression of Parkinson disease (PD), a chronic degenerative disorder characterized by the selective loss of dopaminergic (DAergic) neurons in the subtantia nigra pars compacta (SNpc). Emerging evidence highlights the glia as a pivotal factor in PD etiology, and epidemiological studies indicate that certain nonsteroidal antiinflammatory drugs (NSAIDs) may prevent or delay the progression of PD. Given that the exaggerated inflammatory response observed in old age may play a critical role in exacerbating DAergic vulnerability, we hypothesize here that switching the harmful glial response to inflammation and oxidative stress might increase the ability of the SN to resist inflammatory attacks. To this end, we developed a treatment in which we combined the effects of nitric oxide (NO) with nonsteroidal antiinflammatory activity by using HCT1026, a NO-donating derivative of flurbiprofen that has a safe profile and additional immunomodulatory properties. Young and aged mice fed with control or HCT1026 (30 mg kg(-1) day(-1)) diet were exposed to a single systemic injection of a subtoxic dose (0.2 mg kg(-1)) of lipopolysaccharide (LPS), one of the most extensively used glial activators. HCT1026 efficiently reversed the age-dependent increase of microglial activation in response to LPS to levels measured in younger mice. In aged mice, LPS induced a progressive loss of DAergic neurons with no recovery for their entire life span, whereas younger mice or aged mice fed with HCT1026 were resistant to systemic LPS-induced DAergic neurodegeneration, underscoring glia as a key pharmacological target for DAergic neuroprotection.


Subject(s)
Aging/pathology , Dopaminergic Neurons/pathology , Inflammation/pathology , Microglia/pathology , Nerve Degeneration/pathology , Nerve Regeneration , Substantia Nigra/pathology , Aging/drug effects , Animals , Dopaminergic Neurons/drug effects , Flurbiprofen/analogs & derivatives , Flurbiprofen/pharmacology , Inflammation/complications , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Nerve Degeneration/complications , Nerve Regeneration/drug effects , Nitrosation/drug effects , Oxidative Stress/drug effects , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Substantia Nigra/drug effects , Tyrosine 3-Monooxygenase/metabolism
18.
Mol Neurodegener ; 6: 49, 2011 Jul 13.
Article in English | MEDLINE | ID: mdl-21752258

ABSTRACT

BACKGROUND: Dopamine-synthesizing (dopaminergic, DA) neurons in the ventral midbrain (VM) constitute a pivotal neuronal population controlling motor behaviors, cognitive and affective brain functions, which generation critically relies on the activation of Wingless-type MMTV integration site (Wnt)/ß-catenin pathway in their progenitors. In Parkinson's disease, DA cell bodies within the substantia nigra pars compacta (SNpc) progressively degenerate, with causes and mechanisms poorly understood. Emerging evidence suggests that Wnt signaling via Frizzled (Fzd) receptors may play a role in different degenerative states, but little is known about Wnt signaling in the adult midbrain. Using in vitro and in vivo model systems of DA degeneration, along with functional studies in both intact and SN lesioned mice, we herein highlight an intrinsic Wnt1/Fzd-1/ß-catenin tone critically contributing to the survival and protection of adult midbrain DA neurons. RESULTS: In vitro experiments identifie Fzd-1 receptor expression at a mRNA and protein levels in dopamine transporter (DAT) expressing neurons, and demonstrate the ability of exogenous Wnt1 to exert robust neuroprotective effects against Caspase-3 activation, the loss of tyrosine hydroxylase-positive (TH+) neurons and [3H] dopamine uptake induced by different DA-specific insults, including serum and growth factor deprivation, 6-hydroxydopamine and MPTP/MPP+. Co-culture of DA neurons with midbrain astrocytes phenocopies Wnt1 neuroprotective effects, whereas RNA interference-mediated knockdown of Wnt1 in midbrain astrocytes markedly reduces astrocyte-induced TH+ neuroprotection. Likewise, silencing ß-catenin mRNA or knocking down Fzd-1 receptor expression in mesencephalic neurons counteract astrocyte-induced TH+ neuroprotection. In vivo experiments document Fzd-1 co-localization with TH+ neurons within the intact SNpc and blockade of Fzd/ß-catenin signaling by unilateral infusion of a Fzd/ß-catenin antagonist within the SN induces reactive astrocytosis and acutely inhibits TH+ neuron survival in ipsilateral SNpc, an effect efficiently prevented by pharmacological activation of ß-catenin signaling within the SNpc. CONCLUSION: These results defining a novel Wnt1/Fzd-1/ß-catenin astrocyte-DA autoprotective loop provide a new mechanistic inside into the regulation of pro-survival processes, with potentially relevant consequences for drug design or drug action in Parkinson's disease.

19.
J Neuroinflammation ; 7: 83, 2010 Nov 23.
Article in English | MEDLINE | ID: mdl-21092260

ABSTRACT

BACKGROUND: Current evidence suggests a role of neuroinflammation in the pathogenesis of Parkinson's disease (PD) and in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) model of basal ganglia injury. Reportedly, nonsteroidal anti-inflammatory drugs (NSAIDs) mitigate DAergic neurotoxicity in rodent models of PD. Consistent with these findings, epidemiological analysis indicated that certain NSAIDs may prevent or delay the progression of PD. However, a serious impediment of chronic NSAID therapy, particularly in the elderly, is gastric, renal and cardiac toxicity. Nitric oxide (NO)-donating NSAIDs, have a safer profile while maintaining anti-inflammatory activity of parent compounds. We have investigated the oral activity of the NO-donating derivative of flurbiprofen, [2-fluoro-α-methyl (1,1'-biphenyl)-4-acetic-4-(nitrooxy)butyl ester], HCT1026 (30 mg kg(-1) daily in rodent chow) in mice exposed to the parkinsonian neurotoxin MPTP. METHODS: Ageing mice were fed with a control, flurbiprofen, or HCT1026 diet starting ten days before MPTP administration and continuing for all the experimental period. Striatal high affinity synaptosomal dopamine up-take, motor coordination assessed with the rotarod, tyrosine hydroxylase (TH)- and dopamine transporter (DAT) fiber staining, stereological cell counts, immunoblotting and gene expression analyses were used to assess MPTP-induced nigrostriatal DAergic toxicity and glial activation 1-40 days post-MPTP. RESULTS: HCT1026 was well tolerated and did not cause any measurable toxic effect, whereas flurbiprofen fed mice showed severe gastrointestinal side-effects. HCT1026 efficiently counteracted motor impairment and reversed MPTP-induced decreased synaptosomal [3H]dopamine uptake, TH- and DAT-stained fibers in striatum and TH+ neuron loss in substantia nigra pars compacta (SNpc), as opposed to age-matched mice fed with a control diet. These effects were associated to a significant decrease in reactive macrophage antigen-1 (Mac-1)-positive microglial cells within the striatum and ventral midbrain, decreased expression of iNOS, Mac-1 and NADPH oxidase (PHOX), and downregulation of 3-Nitrotyrosine, a peroxynitrite finger print, in SNpc DAergic neurons. CONCLUSIONS: Oral treatment with HCT1026 has a safe profile and a significant efficacy in counteracting MPTP-induced dopaminergic (DAergic) neurotoxicity, motor impairment and microglia activation in ageing mice. HCT1026 provides a novel promising approach towards the development of effective pharmacological neuroprotective strategies against PD.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/immunology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Corpus Striatum/pathology , Motor Activity/drug effects , Nitric Oxide/metabolism , Parkinson Disease/physiopathology , Substantia Nigra/pathology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Corpus Striatum/metabolism , Disease Models, Animal , Dopamine/metabolism , Dopamine Agents , Flurbiprofen/administration & dosage , Flurbiprofen/analogs & derivatives , Flurbiprofen/immunology , Flurbiprofen/pharmacology , Flurbiprofen/therapeutic use , Humans , Inflammation/drug therapy , Inflammation/immunology , Inflammation/pathology , MPTP Poisoning , Macrophage-1 Antigen/metabolism , Male , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type II/metabolism , Oxidoreductases/metabolism , Parkinson Disease/drug therapy , Parkinson Disease/pathology , Rotarod Performance Test , Substantia Nigra/metabolism , Tyrosine/analogs & derivatives , Tyrosine/metabolism
20.
CNS Neurol Disord Drug Targets ; 9(3): 349-72, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20438439

ABSTRACT

Parkinson's disease (PD) is a common neurodegenerative disorder characterized by the presence of tremor, muscle rigidity, slowness of voluntary movements and postural instability. One of the pathological hallmarks of PD is loss of dopaminergic (DAergic) neurons in the subtantia nigra pars compacta (SNpc). The cause and mechanisms underlying the demise of nigrostriatal DAergic neurons are not fully understood, but interactions between genes and environmental factors are recognized to play a critical role in modulating the vulnerability to PD. Current evidence points to reactive glia as a pivotal factor in PD, but whether astroglia activation may protect or exacerbate DAergic neuron loss is the subject of much debate. Astrocytes and microglia are the key players in neuroinflammatory responses, secreting an array of pro- and anti-inflammatory cytokines, anti-oxidants and neurotrophic factors. These mediators act as double-edged swords, exerting both detrimental and neuroprotective effects. Here, the contribution of astrocytes and microglia in mediating the effects of both genetic and environmental factors, including hormones, endotoxins and neurotoxins, and their ability to influence DAergic neurodegeneration, neuroprotection and neurorepair will be discussed. Approaches capable to regulate glial-associated oxidative stress and mitochondrial damage, by decreasing inflammatory burden, restoring mitochondrial function and DAergic neuron metabolism, might hold great promise for therapeutic interventions. Therapies that support astrocyte function, replacing astrocytes either modified or unmodified in culture, may represent novel approaches targeting astrocytes to promote DAergic neurorescue. Dissecting the molecular determinants of glia-neuron crosstalk will give us the possibility to test novel strategies to promote restoration of injured nigrostriatal DAergic neurons.


Subject(s)
Neuroglia/drug effects , Neuroglia/pathology , Parkinson Disease/drug therapy , Parkinson Disease/pathology , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Humans , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Neuroglia/metabolism , Parkinson Disease/genetics , Parkinson Disease/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...