Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Cell ; 154(1): 103-17, 2013 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-23827677

RESUMEN

Many neurodegenerative diseases are characterized by the accumulation of insoluble protein aggregates, including neurofibrillary tangles comprised of tau in Alzheimer's disease and Lewy bodies composed of α-synuclein in Parkinson's disease. Moreover, different pathological proteins frequently codeposit in disease brains. To test whether aggregated α-synuclein can directly cross-seed tau fibrillization, we administered preformed α-synuclein fibrils assembled from recombinant protein to primary neurons and transgenic mice. Remarkably, we discovered two distinct strains of synthetic α-synuclein fibrils that demonstrated striking differences in the efficiency of cross-seeding tau aggregation, both in neuron cultures and in vivo. Proteinase K digestion revealed conformational differences between the two synthetic α-synuclein strains and also between sarkosyl-insoluble α-synuclein extracted from two subgroups of Parkinson's disease brains. We speculate that distinct strains of pathological α-synuclein likely exist in neurodegenerative disease brains and may underlie the tremendous heterogeneity of synucleinopathies.


Asunto(s)
Neuronas/metabolismo , Enfermedad de Parkinson/patología , alfa-Sinucleína/metabolismo , Proteínas tau/metabolismo , Amiloide/química , Amiloide/metabolismo , Animales , Células Cultivadas , Embrión de Mamíferos/metabolismo , Humanos , Masculino , Ratones , Ratones Transgénicos , Enfermedad de Parkinson/metabolismo , Proteínas Recombinantes/metabolismo , alfa-Sinucleína/química
2.
J Neuroinflammation ; 21(1): 93, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38622654

RESUMEN

The neuroinflammatory process in synucleinopathies of the aging population such as Parkinson's disease (PD) and dementia with Lewy bodies (DLB) involves microglial activation as well as infiltration of the CNS by T cells and natural killer T cells (NKTs). To evaluate the potential of targeting NKT cells to modulate neuroinflammation, we treated α-syn transgenic (tg) mice (e.g.: Thy1 promoter line 61) with an antibody against CD1d, which is a glycoprotein expressed in antigen presenting cells (APCs). CD1d-presented lipid antigens activate NKT cells through the interaction with T cell receptor in NKTs, resulting in the production of cytokines. Thus, we hypothesized that blocking the APC-NKT interaction with an anti-CD1d antibody might reduce neuroinflammation and neurodegeneration in models of DLB/PD. Treatment with the anti-CD1d antibody did not have effects on CD3 (T cells), slightly decreased CD4 and increased CD8 lymphocytes in the mice. Moreover, double labeling studies showed that compared to control (IgG) treated α-syn tg mice, treatment with anti-CD1d decreased numbers of CD3/interferon γ (IFN γ)-positive cells, consistent with NKTs. Further double labeling studies showed that CD1d-positive cells co-localized with the astrocytes marker GFAP and that anti-CD1d antibody reduced this effect. While in control α-syn tg mice CD3 positive cells were near astrocytes, this was modified by the treatment with the CD1d antibody. By qPCR, levels of IFN γ, CCL4, and interleukin-6 were increased in the IgG treated α-syn tg mice. Treatment with CD1d antibody blunted this cytokine response that was associated with reduced astrocytosis and microgliosis in the CNS of the α-syn tg mice treated with CD1d antibody. Flow cytometric analysis of immune cells in α-syn tg mice revealed that CD1d-tet + T cells were also increased in the spleen of α-syn tg mice, which treatment with the CD1d antibody reduced. Reduced neuroinflammation in the anti-CD1d-treated α-syn tg mice was associated with amelioration of neurodegenerative pathology. These results suggest that reducing infiltration of NKT cells with an antibody against CD1d might be a potential therapeutical approach for DLB/PD.


Asunto(s)
Enfermedad de Parkinson , alfa-Sinucleína , Ratones , Animales , alfa-Sinucleína/genética , Cuerpos de Lewy/patología , Enfermedades Neuroinflamatorias , Enfermedad de Parkinson/patología , Ratones Transgénicos , Inmunoterapia/métodos , Citocinas , Inmunoglobulina G
3.
Brain Behav Immun ; 123: 254-269, 2024 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-39284516

RESUMEN

Synucleinopathies are age-related neurological disorders characterized by the abnormal accumulation of α-synuclein (α-syn) in neuronal and non-neuronal cells. It has been proposed that microglial cells play an important role in synucleinopathy neuroinflammation, as well as homeostatically, such as in the clearance of α-syn aggregates in the brain. Here, we examined the effects of microglia on the pathogenesis of synucleinopathies by cell depletion in a mouse model of synucleinopathies. For this purpose, we treated non-transgenic (Non-tg) and α-synuclein transgenic (α-syn-tg) mice with pexidartinib (PLX3397), a tyrosine kinase inhibitor of colony-stimulating factor 1 receptor (CSF-1R). Neuropathological and immunoblot analysis confirmed that Iba-1 immunoreactive microglial cells were decreased by 95% following PLX3397 treatment in Non-tg and α-syn-tg mice. The level of total α-syn in the Triton X-insoluble fraction of brain homogenate was significantly decreased by microglial depletion in the α-syn-tg mice, while the level of Triton X-soluble human α-syn was not affected. Furthermore, the number of p-α-syn immunoreactive inclusions was reduced in α-syn-tg mice treated with PLX3397. Microglial depletion also ameliorated neuronal and synaptic degeneration in α-syn-tg mice, thereby resulted partially improving the motor behavioral deficit in α-syn-tg mice. Moreover, we demonstrated that microglia that survived post-PLX3397 treatment (PLX-resistant microglia) have lower expressions of CSF-1R, and microglial transcriptome analysis further elucidated that PLX-resistant microglia have unique morphology and transcriptomic signatures relative to vehicle-treated microglia of both genotypes; these include differences in definitive microglial functions such as their immune response, cell mobility, cell-cell communications, and regulation of neural homeostasis. Therefore, we suggest that microglia play a critical role in the pathogenesis of synucleinopathies, and that modulation of microglial status might be an effective therapeutic strategy for synucleinopathies.

4.
J Neuroinflammation ; 17(1): 214, 2020 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-32680537

RESUMEN

BACKGROUND: α-Synuclein (α-syn) is a pre-synaptic protein which progressively accumulates in neuronal and non-neuronal cells in neurodegenerative diseases such as Parkinson's disease (PD), dementia with Lewy bodies (DLB), and multiple system atrophy. Recent evidence suggests that aberrant immune activation may be involved in neurodegeneration in PD/DLB. While previous studies have often focused on the microglial responses, less is known about the role of the peripheral immune system in these disorders. METHODS: To understand the involvement of the peripheral immune system in PD/DLB, we evaluated T cell populations in the brains of α-syn transgenic (tg) mice (e.g., Thy1 promoter line 61) and DLB patients. RESULTS: Immunohistochemical analysis showed perivascular and parenchymal infiltration by CD3+/CD4+ helper T cells, but not cytotoxic T cells (CD3+/CD8+) or B cells (CD20+), in the neocortex, hippocampus, and striatum of α-syn tg mice. CD3+ cells were found in close proximity to the processes of activated astroglia, particularly in areas of the brain with significant astrogliosis, microgliosis, and expression of pro-inflammatory cytokines. In addition, a subset of CD3+ cells co-expressed interferon γ. Flow cytometric analysis of immune cells in the brains of α-syn tg mice revealed that CD1d-tet+ T cells were also increased in the brains of α-syn tg mice suggestive of natural killer T cells. In post-mortem DLB brains, we similarly detected increased numbers of infiltrating CD3+/CD4+ T cells in close proximity with blood vessels. CONCLUSION: These results suggest that infiltrating adaptive immune cells play an important role in neuroinflammation and neurodegeneration in synucleinopathies and that modulating peripheral T cells may be a viable therapeutic strategy for PD/DLB.


Asunto(s)
Inmunidad Adaptativa/fisiología , Encéfalo/metabolismo , Enfermedad por Cuerpos de Lewy/metabolismo , Linfocitos T/metabolismo , alfa-Sinucleína/metabolismo , Anciano , Anciano de 80 o más Años , Animales , Encéfalo/inmunología , Encéfalo/patología , Femenino , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Enfermedad por Cuerpos de Lewy/inmunología , Enfermedad por Cuerpos de Lewy/patología , Masculino , Ratones , Ratones Transgénicos , Linfocitos T/inmunología , Linfocitos T/patología , alfa-Sinucleína/inmunología
5.
Hippocampus ; 27(3): 285-302, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27997993

RESUMEN

Tauopathies are neurodegenerative disorders characterized by abnormal intracellular aggregates of tau protein, and include Alzheimer's disease, corticobasal degeneration, frontotemporal dementia, and traumatic brain injury. Glutamate metabolism is altered in neurodegenerative disorders manifesting in higher or lower concentrations of glutamate, its transporters or receptors. Previously, glutamate chemical exchange saturation transfer (GluCEST) magnetic resonance imaging (MRI) demonstrated that glutamate levels are reduced in regions of synapse loss in the hippocampus of a mouse model of late-stage tauopathy. We performed a longitudinal GluCEST imaging experiment paired with a cross-sectional study of histologic markers of tauopathy to determine whether (1) early GluCEST changes are associated with synapse loss before volume loss occurs in the hippocampus, and whether (2) subhippocampal dynamics in GluCEST are associated with histopathologic events related to glutamate alterations in tauopathy. Live imaging of the hippocampus in three serial slices was performed without exogenous contrast agents, and subregions were segmented based on a k-means cluster model. Subregions of the hippocampus were analyzed (cornu ammonis CA1, CA3, dentate gyrus DG, and ventricle) in order to associate local MRI-observable changes in glutamate with histological measures of glial cell proliferation (GFAP), synapse density (synaptophysin, VGlut1) and glutamate receptor (NMDA-NR1) levels. Early differences in GluCEST between healthy and tauopathy mice were measured in the CA1 and DG subregions (30% reduction, P ≤ 0.001). Synapse density was also significantly reduced in every subregion of the hippocampus in tauopathy mice by 6 months. Volume was not significantly reduced in any subregion until 13 months. Further, a gradient in glutamate levels was observed in vivo along hippocampal axes that became polarized as tauopathy progressed. Dynamics in hippocampal glutamate levels throughout lifetime were most closely correlated with combined changes in synaptophysin and GFAP, indicating that GluCEST imaging may be a surrogate marker of glutamate concentration in glial cells and at the synaptic level. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Ácido Glutámico/metabolismo , Hipocampo/metabolismo , Hipocampo/patología , Tauopatías/metabolismo , Tauopatías/patología , Animales , Proliferación Celular/fisiología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Proteína Ácida Fibrilar de la Glía/metabolismo , Hipocampo/diagnóstico por imagen , Humanos , Inmunohistoquímica , Estudios Longitudinales , Ratones Transgénicos , Proteínas del Tejido Nervioso/metabolismo , Neuroglía/metabolismo , Neuroglía/patología , Tamaño de los Órganos , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapsis/metabolismo , Sinapsis/patología , Sinaptofisina/metabolismo , Tauopatías/diagnóstico por imagen , Proteína 1 de Transporte Vesicular de Glutamato/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo
6.
Acta Neuropathol ; 129(2): 221-37, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25534024

RESUMEN

Filamentous tau pathologies are hallmark lesions of several neurodegenerative tauopathies including Alzheimer's disease (AD) and corticobasal degeneration (CBD) which show cell type-specific and topographically distinct tau inclusions. Growing evidence supports templated transmission of tauopathies through functionally interconnected neuroanatomical pathways suggesting that different self-propagating strains of pathological tau could account for the diverse manifestations of neurodegenerative tauopathies. Here, we describe the rapid and distinct cell type-specific spread of pathological tau following intracerebral injections of CBD or AD brain extracts enriched in pathological tau (designated CBD-Tau and AD-Tau, respectively) in young human mutant P301S tau transgenic (Tg) mice (line PS19) ~6-9 months before they show onset of mutant tau transgene-induced tau pathology. At 1 month post-injection of CBD-Tau, tau inclusions developed predominantly in oligodendrocytes of the fimbria and white matter near the injection sites with infrequent intraneuronal tau aggregates. In contrast, injections of AD-Tau in young PS19 mice induced tau pathology predominantly in neuronal perikarya with little or no oligodendrocyte involvement 1 month post-injection. With longer post-injection survival intervals of up to 6 months, CBD-Tau- and AD-Tau-induced tau pathology spread to different brain regions distant from the injection sites while maintaining the cell type-specific pattern noted above. Finally, CA3 neuron loss was detected 3 months post-injection of AD-Tau but not CBD-Tau. Thus, AD-Tau and CBD-Tau represent specific pathological tau strains that spread differentially and may underlie distinct clinical and pathological features of these two tauopathies. Hence, these strains could become targets to develop disease-modifying therapies for CBD and AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Tauopatías/metabolismo , Tauopatías/patología , Proteínas tau/metabolismo , Animales , Western Blotting , Trasplante de Tejido Encefálico , Síndrome de Down/metabolismo , Ensayo de Inmunoadsorción Enzimática , Humanos , Inmunohistoquímica , Ratones Transgénicos , Neuroglía/metabolismo , Neuroglía/patología , Neuronas/metabolismo , Neuronas/patología , Proteínas tau/genética
7.
Acta Neuropathol ; 130(3): 349-62, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26150341

RESUMEN

Filamentous tau inclusions are hallmarks of Alzheimer's disease (AD) and other neurodegenerative tauopathies. An increasing number of studies implicate the cell-to-cell propagation of tau pathology in the progression of tauopathies. We recently showed (Iba et al., J Neurosci 33:1024-1037, 2013) that inoculation of preformed synthetic tau fibrils (tau PFFs) into the hippocampus of young transgenic (Tg) mice (PS19) overexpressing human P301S mutant tau induced robust tau pathology in anatomically connected brain regions including the locus coeruleus (LC). Since Braak and colleagues hypothesized that the LC is the first brain structure to develop tau lesions and since LC has widespread connections throughout the CNS, LC neurons could be the critical initiators of the stereotypical spreading of tau pathology through connectome-dependent transmission of pathological tau in AD. Here, we report that injections of tau PFFs into the LC of PS19 mice induced propagation of tau pathology to major afferents and efferents of the LC. Notably, tau pathology propagated along LC efferent projections was localized not only to axon terminals but also to neuronal perikarya, suggesting transneuronal transfer of templated tau pathology to neurons receiving LC projections. Further, brainstem neurons giving rise to major LC afferents also developed perikaryal tau pathology. Surprisingly, while tangle-bearing neurons degenerated in the LC ipsilateral to the injection site starting 6 months post-injection, no neuron loss was seen in the contralateral LC wherein tangle-bearing neurons gradually cleared tau pathology by 6-12 months post-injection. However, the spreading pattern of tau pathology observed in our LC-injected mice is different from that in AD brains since hippocampus and entorhinal cortex, which are affected in early stages of AD, were largely spared of tau inclusions in our model. Thus, while our study tested critical aspects of the Braak hypothesis of tau pathology spread, this novel mouse model provides unique opportunities to elucidate mechanisms underlying the selective vulnerability of neurons to acquire tau pathology and succumb to or resist tau-mediated neurodegeneration.


Asunto(s)
Locus Coeruleus/patología , Neuronas/patología , Tauopatías/patología , Vías Aferentes/metabolismo , Vías Aferentes/patología , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Vías Eferentes/metabolismo , Vías Eferentes/patología , Escherichia coli , Femenino , Humanos , Hipotálamo/metabolismo , Hipotálamo/patología , Inmunohistoquímica , Locus Coeruleus/metabolismo , Masculino , Ratones Transgénicos , Mutación , Tauopatías/metabolismo , Tálamo/metabolismo , Tálamo/patología , Tirosina 3-Monooxigenasa/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo
8.
J Neurosci ; 33(3): 1024-37, 2013 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-23325240

RESUMEN

Tauopathies, including Alzheimer's disease (AD) and frontotemporal lobar degeneration with tau pathologies, are neurodegenerative diseases characterized by neurofibrillary tangles (NFTs) comprising filamentous tau protein. Although emerging evidence suggests that tau pathology may be transmitted, we demonstrate here that synthetic tau fibrils are sufficient to transmit tau inclusions in a mouse model. Specifically, intracerebral inoculation of young PS19 mice overexpressing mutant human tau (P301S) with synthetic preformed fibrils (pffs) assembled from recombinant full-length tau or truncated tau containing four microtubule binding repeats resulted in rapid induction of NFT-like inclusions that propagated from injected sites to connected brain regions in a time-dependent manner. Interestingly, injection of tau pffs into either hippocampus or striatum together with overlaying cortex gave rise to distinct pattern of spreading. Moreover, unlike tau pathology that spontaneously develops in old PS19 mice, the pff-induced tau inclusions more closely resembled AD NFTs because they were Thioflavin S positive, acetylated, and more resistant to proteinase K digestion. Together, our study demonstrates that synthetic tau pffs alone are capable of inducing authentic NFT-like tau aggregates and initiating spreading of tau pathology in a tauopathy mouse model.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Ovillos Neurofibrilares/metabolismo , Tauopatías/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer/patología , Animales , Encéfalo/patología , Modelos Animales de Enfermedad , Ratones , Ratones Transgénicos , Ovillos Neurofibrilares/patología , Fosforilación , Tauopatías/patología
9.
Neuroimage ; 101: 185-92, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25003815

RESUMEN

Glutamate is the primary excitatory neurotransmitter in the brain, and is implicated in neurodegenerative diseases such as Alzheimer's disease (AD) and several other tauopathies. The current method for measuring glutamate in vivo is proton magnetic resonance spectroscopy ((1)H MRS), although it has poor spatial resolution and weak sensitivity to glutamate changes. In this study, we sought to measure the effect of tau pathology on glutamate levels throughout the brain of a mouse model of tauopathy using a novel magnetic resonance imaging (MRI) technique. We employed glutamate chemical exchange saturation transfer (GluCEST) imaging, which has been previously validated as a complimentary method for measuring glutamate levels with several important advantages over conventional (1)H MRS. We hypothesized that the regional changes in glutamate levels would correlate with histological measurements of pathology including pathological tau, synapse and neuron loss. Imaging and spectroscopy were carried out on tau transgenic mice with the P301S mutation (PS19, n=9) and their wild-type littermates (WT, n=8), followed by immunohistochemistry of their brain tissue. GluCEST imaging resolution allowed for sub-hippocampal analysis of glutamate. Glutamate was significantly decreased by 29% in the CA sub-region of the PS19 hippocampus, and by 15% in the thalamus, where synapse loss was also measured. Glutamate levels and synapse density remained high in the dentate gyrus sub-region of the hippocampus, where neurogenesis is known to occur. The further development of GluCEST imaging for preclinical applications will be valuable, as therapies are being tested in mouse models of tauopathy.


Asunto(s)
Ácido Glutámico/metabolismo , Hipocampo/metabolismo , Imagen por Resonancia Magnética/métodos , Sinapsis/patología , Tauopatías/metabolismo , Tálamo/metabolismo , Animales , Giro Dentado/metabolismo , Giro Dentado/patología , Modelos Animales de Enfermedad , Hipocampo/patología , Ratones , Ratones Transgénicos , Neurogénesis/fisiología , Espectroscopía de Protones por Resonancia Magnética , Tauopatías/patología , Tálamo/patología
10.
bioRxiv ; 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-39372781

RESUMEN

One of the unifying pathological hallmarks of Parkinson's disease (PD) and dementia with Lewy bodies (DLB) is the presence of misfolded, aggregated, and often phosphorylated forms of the protein α-synuclein in neurons. α-Synuclein pathology appears in select populations of neurons throughout various cortical and subcortical regions, and little is currently known about why some neurons develop pathology while others are spared. Here, we utilized subcellular-resolution imaging-based spatial transcriptomics (IST) in a transgenic mouse model that overexpresses wild-type human α-synuclein (α-syn-tg) to evaluate patterns of selective neuronal vulnerability to α-synuclein pathology. By performing post-IST immunofluorescence for α-synuclein phosphorylated at Ser129 (pSyn), we identified cell types in the cortex and hippocampus that were vulnerable or resistant to developing pSyn pathology. Next, we investigated the transcriptional underpinnings of the observed selective vulnerability using a set of custom probes to detect genes involved in α-synuclein processing and toxicity. We identified expression of the kinase:substrate pair Plk2 , which phosphorylates α-synuclein at Ser129, and human SNCA ( hSNCA ), as underlying the selective vulnerability to pSyn pathology. Finally, we performed differential gene expression analysis, comparing non-transgenic cells to pSyn - and pSyn + α-syn-tg cells to reveal gene expression changes downstream of hSNCA overexpression and pSyn pathology, which included pSyn-dependent alterations in mitochondrial and endolysosomal genes. This study provides a comprehensive use case of IST, yielding new biological insights into the formation of α-synuclein pathology and its downstream effects in a PD/DLB mouse model.

11.
J Neurosci ; 32(11): 3601-11, 2012 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-22423084

RESUMEN

Neurodegenerative tauopathies, such as Alzheimer's disease (AD), are characterized by insoluble deposits of hyperphosphorylated tau protein within brain neurons. Increased phosphorylation and decreased solubility has been proposed to diminish normal tau stabilization of microtubules (MTs), thereby leading to neuronal dysfunction. Earlier studies have provided evidence that small molecule MT-stabilizing drugs that are used in the treatment of cancer may have utility in the treatment of tauopathies. However, it has not been established whether treatment with a small molecule MT-stabilizing compound will provide benefit in a transgenic model with pre-existing tau pathology, as would be seen in human patients with clinical symptoms. Accordingly, we describe here an interventional study of the brain-penetrant MT-stabilizing agent, epothilone D (EpoD), in aged PS19 mice with existing tau pathology and related behavioral deficits. EpoD treatment reduced axonal dystrophy and increased axonal MT density in the aged PS19 mice, which led to improved fast axonal transport and cognitive performance. Moreover, the EpoD-treated PS19 mice had less forebrain tau pathology and increased hippocampal neuronal integrity, with no dose-limiting side effects. These data reveal that brain-penetrant MT-stabilizing drugs hold promise for the treatment of AD and related tauopathies, and that EpoD could be a candidate for clinical testing.


Asunto(s)
Envejecimiento/efectos de los fármacos , Enfermedad de Alzheimer/tratamiento farmacológico , Trastornos del Conocimiento/tratamiento farmacológico , Epotilonas/uso terapéutico , Tauopatías/tratamiento farmacológico , Moduladores de Tubulina/uso terapéutico , Envejecimiento/patología , Envejecimiento/psicología , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/psicología , Animales , Axones/efectos de los fármacos , Axones/patología , Trastornos del Conocimiento/patología , Trastornos del Conocimiento/psicología , Epotilonas/farmacología , Humanos , Masculino , Ratones , Ratones Transgénicos , Microtúbulos/efectos de los fármacos , Microtúbulos/patología , Síndromes de Neurotoxicidad/tratamiento farmacológico , Síndromes de Neurotoxicidad/patología , Síndromes de Neurotoxicidad/psicología , Tauopatías/patología , Tauopatías/psicología , Moduladores de Tubulina/farmacología , Proteínas tau/genética
12.
J Parkinsons Dis ; 13(2): 255-270, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36847016

RESUMEN

BACKGROUND: Synucleinopathies are a group of neurodegenerative disorders that are pathologically characterized by intracellular aggregates called Lewy bodies. Lewy bodies are primarily composed of α-synuclein (asyn) protein, which is mostly phosphorylated at serine 129 (pS129) when aggregated and therefore used as a marker for pathology. Currently commercial antibodies against pS129 asyn stain aggregates well but in healthy brains cross react with other proteins, thus making it difficult to specifically detect physiological pS129 asyn. OBJECTIVE: To develop a staining procedure that detects endogenous and physiological relevant pS129 asyn with high specificity and low background. METHODS: We used the fluorescent and brightfield in situ proximity ligation assay (PLA) to specifically detect pS129 asyn in cell culture, mouse, and human brain sections. RESULTS: The pS129 asyn PLA specifically stained physiological and soluble pS129 asyn in cell culture, mouse brain sections, and human brain tissue without significant cross-reactivity or background signal. However, this technique was not successful in detecting Lewy bodies in human brain tissue. CONCLUSION: We successfully developed a novel PLA method that can, in the future, be used on in vitro and in vivo samples as a tool to explore and better understand the cellular localization and function of pS129 asyn in health and disease.


Asunto(s)
Enfermedad de Parkinson , Sinucleinopatías , Animales , Humanos , Ratones , alfa-Sinucleína/metabolismo , Cuerpos de Lewy/metabolismo , Enfermedad de Parkinson/diagnóstico , Fosforilación , Sinucleinopatías/metabolismo
13.
Sci Transl Med ; 15(695): eabq6089, 2023 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-37163617

RESUMEN

Alterations in the p38 mitogen-activated protein kinases (MAPKs) play an important role in the pathogenesis of dementia with Lewy bodies (DLB) and Parkinson's disease (PD). Activation of the p38α MAPK isoform and mislocalization of the p38γ MAPK isoform are associated with neuroinflammation and synaptic degeneration in DLB and PD. Therefore, we hypothesized that p38α might be associated with neuronal p38γ distribution and synaptic dysfunction in these diseases. To test this hypothesis, we treated in vitro cellular and in vivo mouse models of DLB/PD with SKF-86002, a compound that attenuates inflammation by inhibiting p38α/ß, and then investigated the effects of this compound on p38γ and neurodegenerative pathology. We found that inhibition of p38α reduced neuroinflammation and ameliorated synaptic, neurodegenerative, and motor behavioral deficits in transgenic mice overexpressing human α-synuclein. Moreover, treatment with SKF-86002 promoted the redistribution of p38γ to synapses and reduced the accumulation of α-synuclein in mice overexpressing human α-synuclein. Supporting the potential value of targeting p38 in DLB/PD, we found that SKF-86002 promoted the redistribution of p38γ in neurons differentiated from iPS cells derived from patients with familial PD (carrying the A53T α-synuclein mutation) and healthy controls. Treatment with SKF-86002 ameliorated α-synuclein-induced neurodegeneration in these neurons only when microglia were pretreated with this compound. However, direct treatment of neurons with SKF-86002 did not affect α-synuclein-induced neurotoxicity, suggesting that SKF-86002 treatment inhibits α-synuclein-induced neurotoxicity mediated by microglia. These findings provide a mechanistic connection between p38α and p38γ as well as a rationale for targeting this pathway in DLB/PD.


Asunto(s)
Proteína Quinasa 14 Activada por Mitógenos , Enfermedad de Parkinson , Humanos , Ratones , Animales , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/patología , alfa-Sinucleína/metabolismo , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Enfermedades Neuroinflamatorias , Neuronas/metabolismo , Ratones Transgénicos
14.
J Neurosci ; 31(29): 10648-65, 2011 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-21775608

RESUMEN

The temporal structuring of multiple events is essential for the purposeful regulation of behavior. We investigated the role of the lateral prefrontal cortex (LPFC) in transforming external signals of multiple sensory modalities into information suitable for monitoring successive events across behavioral phases until an intended action is prompted and then initiated. We trained monkeys to receive a succession of 1 s visual, auditory, or tactile sensory signals separated by variable intervals and to then release a key as soon as the fourth signal appeared. Thus, the animals had to monitor and update information about the progress of the task upon receiving each signal preceding the key release in response to the fourth signal. We found that the initial, short-latency responses of LPFC neurons reflected primarily the sensory modality, rather than the phase or progress of the task. However, a task phase-selective response developed within 500 ms of signal reception, and information about the task phase was maintained throughout the presentation of successive cues. The task phase-selective activity was updated with the appearance of each cue until the planned action was initiated. The phase-selective activity of individual neurons reflected not merely a particular phase of the task but also multiple successive phases. Furthermore, we found combined representations of task phase and sensory modality in the activity of individual LPFC neurons. These properties suggest how information representing multiple phases of behavioral events develops in the LPFC to provide a basis for the temporal regulation of behavior.


Asunto(s)
Mapeo Encefálico , Recuerdo Mental/fisiología , Corteza Prefrontal/fisiología , Tiempo de Reacción/fisiología , Estimulación Acústica , Potenciales de Acción/fisiología , Análisis de Varianza , Animales , Conducta Animal , Señales (Psicología) , Femenino , Macaca fascicularis , Masculino , Neuronas/fisiología , Estimulación Luminosa , Corteza Prefrontal/citología , Desempeño Psicomotor/fisiología , Factores de Tiempo , Tacto
15.
J Neurosci ; 31(40): 14436-49, 2011 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-21976528

RESUMEN

Because overactivation of the hypothalamic-pituitary-adrenal (HPA) axis occurs in Alzheimer's disease (AD), dysregulation of stress neuromediators may play a mechanistic role in the pathophysiology of AD. However, the effects of stress on tau phosphorylation are poorly understood, and the relationship between corticosterone and corticotropin-releasing factor (CRF) on both ß-amyloid (Aß) and tau pathology remain unclear. Therefore, we first established a model of chronic stress, which exacerbates Aß accumulation in Tg2576 mice and then extended this stress paradigm to a tau transgenic mouse model with the P301S mutation (PS19) that displays tau hyperphosphorylation, insoluble tau inclusions and neurodegeneration. We show for the first time that both Tg2576 and PS19 mice demonstrate a heightened HPA stress profile in the unstressed state. In Tg2576 mice, 1 month of restraint/isolation (RI) stress increased Aß levels, suppressed microglial activation, and worsened spatial and fear memory compared with nonstressed mice. In PS19 mice, RI stress promoted tau hyperphosphorylation, insoluble tau aggregation, neurodegeneration, and fear-memory impairments. These effects were not mimicked by chronic corticosterone administration but were prevented by pre-stress administration of a CRF receptor type 1 (CRF(1)) antagonist. The role for a CRF(1)-dependent mechanism was further supported by the finding that mice overexpressing CRF had increased hyperphosphorylated tau compared with wild-type littermates. Together, these results implicate HPA dysregulation in AD neuropathogenesis and suggest that prolonged stress may increase Aß and tau hyperphosphorylation. These studies also implicate CRF in AD pathophysiology and suggest that pharmacological manipulation of this neuropeptide may be a potential therapeutic strategy for AD.


Asunto(s)
Trastornos del Conocimiento/patología , Modelos Animales de Enfermedad , Degeneración Nerviosa/patología , Receptores de Hormona Liberadora de Corticotropina/biosíntesis , Estrés Psicológico/patología , Tauopatías/patología , Animales , Enfermedad Crónica , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/metabolismo , Femenino , Sistema Hipotálamo-Hipofisario/patología , Sistema Hipotálamo-Hipofisario/fisiología , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Transgénicos , Degeneración Nerviosa/etiología , Degeneración Nerviosa/metabolismo , Sistema Hipófiso-Suprarrenal/patología , Sistema Hipófiso-Suprarrenal/fisiología , Estrés Psicológico/complicaciones , Estrés Psicológico/metabolismo , Tauopatías/etiología , Tauopatías/metabolismo , Proteínas tau/biosíntesis
16.
Exp Mol Med ; 54(4): 447-454, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35396576

RESUMEN

The coronavirus (COVID-19) pandemic, caused by severe acute respiratory system coronavirus 2 (SARS-CoV-2), has created significant challenges for scientists seeking to understand the pathogenic mechanisms of SARS-CoV-2 infection and to identify the best therapies for infected patients. Although ACE2 is a known receptor for the virus and has been shown to mediate viral entry into the lungs, accumulating reports highlight the presence of neurological symptoms resulting from infection. As ACE2 expression is low in the central nervous system (CNS), these neurological symptoms are unlikely to be caused by ACE2-virus binding. In this review, we will discuss a proposed interaction between SARS-CoV-2 and Toll-like receptor 2 (TLR2) in the CNS. TLR2 is an innate immune receptor that recognizes exogenous microbial components but has also been shown to interact with multiple viral components, including the envelope (E) protein of SARS-CoV-2. In addition, TLR2 plays an important role in the pathogenesis of neurodegenerative diseases such as Alzheimer's disease (AD) and Parkinson's disease (PD). Based on these observations, we hypothesize that TLR2 may play a critical role in the response to SARS-CoV-2 infiltration in the CNS, thereby resulting in the induction or acceleration of AD and PD pathologies in patients.


Asunto(s)
Enfermedad de Alzheimer , COVID-19 , Enfermedades Neurodegenerativas , Enfermedad de Parkinson , Enzima Convertidora de Angiotensina 2 , Sistema Nervioso Central , Humanos , SARS-CoV-2 , Receptor Toll-Like 2
17.
Mol Neurodegener ; 17(1): 60, 2022 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-36064424

RESUMEN

BACKGROUND: Although ɑ-synuclein (ɑ-syn) spreading in age-related neurodegenerative diseases such as Parkinson's disease (PD) and Dementia with Lewy bodies (DLB) has been extensively investigated, the role of aging in the manifestation of disease remains unclear. METHODS: We explored the role of aging and inflammation in the pathogenesis of synucleinopathies in a mouse model of DLB/PD initiated by intrastriatal injection of ɑ-syn preformed fibrils (pff). RESULTS: We found that aged mice showed more extensive accumulation of ɑ-syn in selected brain regions and behavioral deficits that were associated with greater infiltration of T cells and microgliosis. Microglial inflammatory gene expression induced by ɑ-syn-pff injection in young mice had hallmarks of aged microglia, indicating that enhanced age-associated pathologies may result from inflammatory synergy between aging and the effects of ɑ-syn aggregation. Based on the transcriptomics analysis projected from Ingenuity Pathway Analysis, we found a network that included colony stimulating factor 2 (CSF2), LPS related genes, TNFɑ and poly rl:rC-RNA as common regulators. CONCLUSIONS: We propose that aging related inflammation (eg: CSF2) influences outcomes of pathological spreading of ɑ-syn and suggest that targeting neuro-immune responses might be important in developing treatments for DLB/PD.


Asunto(s)
Enfermedad de Parkinson , Sinucleinopatías , Animales , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Inflamación/metabolismo , Ratones , Enfermedad de Parkinson/metabolismo , alfa-Sinucleína/metabolismo
18.
NPJ Vaccines ; 7(1): 1, 2022 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-35013319

RESUMEN

Accumulation of misfolded proteins such as amyloid-ß (Aß), tau, and α-synuclein (α-Syn) in the brain leads to synaptic dysfunction, neuronal damage, and the onset of relevant neurodegenerative disorder/s. Dementia with Lewy bodies (DLB) and Parkinson's disease (PD) are characterized by the aberrant accumulation of α-Syn intracytoplasmic Lewy body inclusions and dystrophic Lewy neurites resulting in neurodegeneration associated with inflammation. Cell to cell propagation of α-Syn aggregates is implicated in the progression of PD/DLB, and high concentrations of anti-α-Syn antibodies could inhibit/reduce the spreading of this pathological molecule in the brain. To ensure sufficient therapeutic concentrations of anti-α-Syn antibodies in the periphery and CNS, we developed four α-Syn DNA vaccines based on the universal MultiTEP platform technology designed especially for the elderly with immunosenescence. Here, we are reporting on the efficacy and immunogenicity of these vaccines targeting three B-cell epitopes of hα-Syn aa85-99 (PV-1947D), aa109-126 (PV-1948D), aa126-140 (PV-1949D) separately or simultaneously (PV-1950D) in a mouse model of synucleinopathies mimicking PD/DLB. All vaccines induced high titers of antibodies specific to hα-Syn that significantly reduced PD/DLB-like pathology in hα-Syn D line mice. The most significant reduction of the total and protein kinase resistant hα-Syn, as well as neurodegeneration, were observed in various brain regions of mice vaccinated with PV-1949D and PV-1950D in a sex-dependent manner. Based on these preclinical data, we selected the PV-1950D vaccine for future IND enabling preclinical studies and clinical development.

19.
J Neurosci ; 30(41): 13861-6, 2010 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-20943926

RESUMEN

Neurons in the brains of those with Alzheimer's disease (AD) and many frontotemporal dementias (FTDs) contain neurofibrillary tangles comprised of hyperphosphorylated tau protein. Tau normally stabilizes microtubules (MTs), and tau misfolding could lead to a loss of this function with consequent MT destabilization and neuronal dysfunction. Accordingly, a possible therapeutic strategy for AD and related "tauopathies" is treatment with a MT-stabilizing anti-cancer drug such as paclitaxel. However, paclitaxel and related taxanes have poor blood-brain barrier permeability and thus are unsuitable for diseases of the brain. We demonstrate here that the MT-stabilizing agent, epothilone D (EpoD), is brain-penetrant and we subsequently evaluated whether EpoD can compensate for tau loss-of-function in PS19 tau transgenic mice that develop forebrain tau inclusions, axonal degeneration and MT deficits. Treatment of 3-month-old male PS19 mice with low doses of EpoD once weekly for a 3 month period significantly improved CNS MT density and axonal integrity without inducing notable side-effects. Moreover, EpoD treatment reduced cognitive deficits that were observed in the PS19 mice. These results suggest that certain brain-penetrant MT-stabilizing agents might provide a viable therapeutic strategy for the treatment of AD and FTDs.


Asunto(s)
Axones/efectos de los fármacos , Encéfalo/efectos de los fármacos , Cognición/efectos de los fármacos , Epotilonas/farmacología , Microtúbulos/efectos de los fármacos , Neuronas/efectos de los fármacos , Tauopatías/tratamiento farmacológico , Animales , Axones/metabolismo , Axones/patología , Encéfalo/metabolismo , Encéfalo/patología , Encéfalo/fisiopatología , Epotilonas/uso terapéutico , Femenino , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Transgénicos , Microscopía Electrónica , Microtúbulos/metabolismo , Microtúbulos/patología , Destreza Motora/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Fosforilación , Distribución Aleatoria , Prueba de Desempeño de Rotación con Aceleración Constante , Tauopatías/metabolismo , Tauopatías/patología , Tauopatías/fisiopatología , Moduladores de Tubulina/farmacología , Moduladores de Tubulina/uso terapéutico , Proteínas tau/metabolismo
20.
Am J Pathol ; 177(4): 1977-88, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20802182

RESUMEN

Senile plaques formed by ß-amyloid peptides (Aß) and neurofibrillary tangles (NFTs) formed by hyperphosphorylated tau, a microtubule-associated protein, are the hallmark lesions of Alzheimer's disease (AD) in addition to loss of neurons. While several transgenic (Tg) mouse models have recapitulated aspects of AD-like Aß and tau pathologies, a spatiotemporal mapping paradigm for progressive NFT accumulation is urgently needed to stage disease progression in AD mouse models. Braak and co-workers developed an effective and widely used NFT staging paradigm for human AD brains. The creation of a Braak-like spatiotemporal staging scheme for tau pathology in mouse models would facilitate mechanistic studies of AD-like tau pathology. Such a scheme would also enhance the reproducibility of preclinical AD therapeutic studies. Thus, we developed a novel murine model of Aß and tau pathologies and devised a spatiotemporal scheme to stage the emergence and accumulation of NFTs with advancing age. Notably, the development of NFTs followed a spatiotemporal Braak-like pattern similar to that observed in authentic AD. More significantly, the presence of Aß accelerated NFT formation and enhanced tau amyloidosis; however, tau pathology did not have the same effect on Aß pathology. This novel NFT staging scheme provides new insights into the mechanisms of tau pathobiology, and we speculate that this scheme will prove useful for other basic and translational studies of AD mouse models.


Asunto(s)
Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Amiloidosis/etiología , Encéfalo/patología , Ovillos Neurofibrilares/patología , Neuronas/patología , Proteínas tau/metabolismo , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/genética , Amiloidosis/metabolismo , Amiloidosis/patología , Animales , Peso Corporal , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Ensayo de Inmunoadsorción Enzimática , Humanos , Técnicas para Inmunoenzimas , Ratones , Ratones Transgénicos , Ovillos Neurofibrilares/metabolismo , Neuronas/metabolismo , Fenotipo , Placa Amiloide/metabolismo , Placa Amiloide/patología , Tasa de Supervivencia
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda