Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Brain ; 144(4): 1214-1229, 2021 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-33871026

RESUMEN

Knowledge about converging disease mechanisms in the heterogeneous syndrome amyotrophic lateral sclerosis (ALS) is rare, but may lead to therapies effective in most ALS cases. Previously, we identified serum microRNAs downregulated in familial ALS, the majority of sporadic ALS patients, but also in presymptomatic mutation carriers. A 5-nucleotide sequence motif (GDCGG; D = G, A or U) was strongly enriched in these ALS-related microRNAs. We hypothesized that deregulation of protein(s) binding predominantly to this consensus motif was responsible for the ALS-linked microRNA fingerprint. Using microRNA pull-down assays combined with mass spectrometry followed by extensive biochemical validation, all members of the fragile X protein family, FMR1, FXR1 and FXR2, were identified to directly and predominantly interact with GDCGG microRNAs through their structurally disordered RGG/RG domains. Preferential association of this protein family with ALS-related microRNAs was confirmed by in vitro binding studies on a transcriptome-wide scale. Immunohistochemistry of lumbar spinal cord revealed aberrant expression level and aggregation of FXR1 and FXR2 in C9orf72- and FUS-linked familial ALS, but also patients with sporadic ALS. Further analysis of ALS autopsies and induced pluripotent stem cell-derived motor neurons with FUS mutations showed co-aggregation of FXR1 with FUS. Hence, our translational approach was able to take advantage of blood microRNAs to reveal CNS pathology, and suggests an involvement of the fragile X-related proteins in familial and sporadic ALS already at a presymptomatic stage. The findings may uncover disease mechanisms relevant to many patients with ALS. They furthermore underscore the systemic, extra-CNS aspect of ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , MicroARNs/sangre , MicroARNs/genética , Proteínas de Unión al ARN/metabolismo , Esclerosis Amiotrófica Lateral/genética , Proteína C9orf72/genética , Humanos , Proteína FUS de Unión a ARN/genética
2.
Hum Mol Genet ; 27(4): 706-715, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29315381

RESUMEN

Mutations in the mitochondrially located protein CHCHD10 cause motoneuron disease by an unknown mechanism. In this study, we investigate the mutations p.R15L and p.G66V in comparison to wild-type CHCHD10 and the non-pathogenic variant p.P34S in vitro, in patient cells as well as in the vertebrate in vivo model zebrafish. We demonstrate a reduction of CHCHD10 protein levels in p.R15L and p.G66V mutant patient cells to approximately 50%. Quantitative real-time PCR revealed that expression of CHCHD10 p.R15L, but not of CHCHD10 p.G66V, is already abrogated at the mRNA level. Altered secondary structure and rapid protein degradation are observed with regard to the CHCHD10 p.G66V mutant. In contrast, no significant differences in expression, degradation rate or secondary structure of non-pathogenic CHCHD10 p.P34S are detected when compared with wild-type protein. Knockdown of CHCHD10 expression in zebrafish to about 50% causes motoneuron pathology, abnormal myofibrillar structure and motility deficits in vivo. Thus, our data show that the CHCHD10 mutations p.R15L and p.G66V cause motoneuron disease primarily based on haploinsufficiency of CHCHD10.


Asunto(s)
Haploinsuficiencia/fisiología , Proteínas Mitocondriales/metabolismo , Enfermedad de la Neurona Motora/metabolismo , Animales , ADN Complementario/genética , ADN Complementario/metabolismo , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Haploinsuficiencia/genética , Humanos , Proteínas Mitocondriales/química , Proteínas Mitocondriales/genética , Enfermedad de la Neurona Motora/genética , Mutación/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Pez Cebra , Proteínas de Pez Cebra/química , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
3.
Cell Mol Life Sci ; 75(23): 4301-4319, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30030593

RESUMEN

Genetic and functional studies suggest diverse pathways being affected in the neurodegenerative disease amyotrophic lateral sclerosis (ALS), while knowledge about converging disease mechanisms is rare. We detected a downregulation of microRNA-1825 in CNS and extra-CNS system organs of both sporadic (sALS) and familial ALS (fALS) patients. Combined transcriptomic and proteomic analysis revealed that reduced levels of microRNA-1825 caused a translational upregulation of tubulin-folding cofactor b (TBCB). Moreover, we found that excess TBCB led to depolymerization and degradation of tubulin alpha-4A (TUBA4A), which is encoded by a known ALS gene. Importantly, the increase in TBCB and reduction of TUBA4A protein was confirmed in brain cortex tissue of fALS and sALS patients, and led to motor axon defects in an in vivo model. Our discovery of a microRNA-1825/TBCB/TUBA4A pathway reveals a putative pathogenic cascade in both fALS and sALS extending the relevance of TUBA4A to a large proportion of ALS cases.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Perfilación de la Expresión Génica , Predisposición Genética a la Enfermedad/genética , MicroARNs/genética , Proteínas Asociadas a Microtúbulos/genética , Tubulina (Proteína)/genética , Anciano , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Animales , Encéfalo/metabolismo , Encéfalo/patología , Células Cultivadas , Femenino , Células HEK293 , Humanos , Masculino , Ratones Endogámicos C57BL , Proteínas Asociadas a Microtúbulos/metabolismo , Persona de Mediana Edad , Tubulina (Proteína)/metabolismo
4.
Am J Pathol ; 186(8): 2152-2161, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27322773

RESUMEN

Aggregation of misfolded disease-related proteins is a hallmark of neurodegenerative diseases. Aggregate propagation accompanying disease progression has been demonstrated for different proteins (eg, for α-synuclein). Additional evidence supports aggregate cross-seeding activity for α-synuclein. For mutated superoxide dismutase 1 (SOD1), which causes familial amyotrophic lateral sclerosis (ALS), self-propagation of aggregation and cell-to-cell transmission have been demonstrated in vitro. However, there is a prominent lack of in vivo data concerning aggregation and cross-aggregation processes of SOD1. We analyzed the effect of α-synuclein and SOD1 seeds in cell culture using protein fragment complementation assay and intracerebral injection of α-synuclein and SOD1 seeds into SOD1(G93A) transgenic ALS mice. Survival of injected mice was determined, and SOD1 aggregates in the facial nuclei were quantified during disease course. We found that α-synuclein preformed fibrils increased the oligomerization rate of SOD1 in vivo and in vitro, whereas aggregated SOD1 did not exert any effect in both experimental setups. Notably, survival of ALS mice was not changed after inoculation of preformed fibrils. We conclude that misfolded α-synuclein can increase SOD1 aggregation and suppose that α-synuclein seeds are transported from the temporal cortex to the facial nuclei. However, unlike other proteins, the further enhancement of a self-aggregation process by additional SOD1 could not be confirmed in our models.


Asunto(s)
Esclerosis Amiotrófica Lateral/patología , Superóxido Dismutasa-1/metabolismo , alfa-Sinucleína/metabolismo , Esclerosis Amiotrófica Lateral/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Reacción en Cadena en Tiempo Real de la Polimerasa , Superóxido Dismutasa-1/genética
5.
Acta Neuropathol ; 132(3): 391-411, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-26910103

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a devastating progressive neurodegenerative disease affecting primarily the upper and lower motor neurons. A common feature of all ALS cases is a well-characterized neuroinflammatory reaction within the central nervous system (CNS). However, much less is known about the role of the peripheral immune system and its interplay with CNS resident immune cells in motor neuron degeneration. Here, we characterized peripheral monocytes in both temporal and spatial dimensions of ALS pathogenesis. We found the circulating monocytes to be deregulated in ALS regarding subtype constitution, function and gene expression. Moreover, we show that CNS infiltration of peripheral monocytes correlates with improved motor neuron survival in a genetic ALS mouse model. Furthermore, application of human immunoglobulins or fusion proteins containing only the human Fc, but not the Fab antibody fragment, increased CNS invasion of peripheral monocytes and delayed the disease onset. Our results underline the importance of peripheral monocytes in ALS pathogenesis and are in agreement with a protective role of monocytes in the early phase of the disease. The possibility to boost this beneficial function of peripheral monocytes by application of human immunoglobulins should be evaluated in clinical trials.


Asunto(s)
Esclerosis Amiotrófica Lateral/patología , Sistema Nervioso Central/metabolismo , Leucocitos Mononucleares/metabolismo , Monocitos/metabolismo , Sistema Mononuclear Fagocítico/metabolismo , Neuronas Motoras/patología , Médula Espinal/patología , Esclerosis Amiotrófica Lateral/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Ratones Endogámicos C57BL , Médula Espinal/metabolismo
6.
J Neurol Neuromedicine ; 1(7): 28-30, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27853754

RESUMEN

Alpha-synuclein and Cu, Zn superoxide dismutase (SOD1) are both aggregation-prone proteins that are associated with Parkinson's disease (PD) and amyotrophic lateral sclerosis (ALS), respectively. Recently, we showed that alpha-synuclein interacts with SOD1 in various cell types and tissues. Using a cell culture model, we also found that alpha-synuclein nucleates the polymerization of SOD1. Here, we discuss the current literature regarding their interaction and their co-localization in aggregates of human post-mortem tissue. Furthermore we comment on the reported alpha-synuclein-induced SOD1 polymerization in terms of cross-seeding effects in neurodegeneration.

7.
J Cell Biol ; 211(4): 897-911, 2015 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-26598621

RESUMEN

Transactive response DNA-binding protein 43 kD (TDP-43) is an aggregation-prone prion-like domain-containing protein and component of pathological intracellular aggregates found in most amyotrophic lateral sclerosis (ALS) patients. TDP-43 oligomers have been postulated to be released and subsequently nucleate TDP-43 oligomerization in recipient cells, which might be the molecular correlate of the systematic symptom spreading observed during ALS progression. We developed a novel protein complementation assay allowing quantification of TDP-43 oligomers in living cells. We demonstrate the exchange of TDP-43 between cell somata and the presence of TDP-43 oligomers in microvesicles/exosomes and show that microvesicular TDP-43 is preferentially taken up by recipient cells where it exerts higher toxicity than free TDP-43. Moreover, studies using microfluidic neuronal cultures suggest both anterograde and retrograde trans-synaptic spreading of TDP-43. Finally, we demonstrate TDP-43 oligomer seeding by TDP-43-containing material derived from both cultured cells and ALS patient brain lysate. Thus, using an innovative detection technique, we provide evidence for preferentially microvesicular uptake as well as both soma-to-soma "horizontal" and bidirectional "vertical" synaptic intercellular transmission and prion-like seeding of TDP-43.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Esclerosis Amiotrófica Lateral/metabolismo , Animales , Exosomas/metabolismo , Células HEK293 , Humanos , Ratones Endogámicos C57BL , Neuronas/metabolismo , Multimerización de Proteína , Transporte de Proteínas , Transmisión Sináptica
8.
Mol Neurodegener ; 10: 66, 2015 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-26643113

RESUMEN

BACKGROUND: Parkinson's disease (PD) and amyotrophic lateral sclerosis (ALS) are both neurodegenerative diseases leading to impaired execution of movement. α-Synuclein plays a central role in the pathogenesis of PD whereas Cu, Zn superoxide dismutase (SOD1) is a key player in a subset of familial ALS cases. Under pathological conditions both α-synuclein and SOD1 form oligomers and fibrils. In this study we investigated the possible molecular interaction of α-synuclein and SOD1 and its functional and pathological relevance. RESULTS: Using a protein-fragment complementation approach and co-IP, we found that α-synuclein and SOD1 physically interact in living cells, human erythrocytes and mouse brain tissue. Additionally, our data show that disease related mutations in α-synuclein (A30P, A53T) and SOD1 (G85R, G93A) modify the binding of α-synuclein to SOD1. Notably, α-synuclein accelerates SOD1 oligomerization independent of SOD1 activity. CONCLUSION: This study provides evidence for a novel interaction of α-synuclein and SOD1 that might be relevant for neurodegenerative diseases.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Encéfalo/metabolismo , Superóxido Dismutasa/metabolismo , alfa-Sinucleína/metabolismo , Animales , Humanos , Ratones Transgénicos , Mutación/genética , Enfermedad de Parkinson/metabolismo , Multimerización de Proteína , Superóxido Dismutasa-1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA