Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Aging Cell ; 23(5): e14109, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38372175

RESUMEN

Brain aging is associated with cognitive decline, memory loss and many neurodegenerative disorders. The mammalian brain has distinct structural regions that perform specific functions. However, our understanding in gene expression and cell types within the context of the spatial organization of the mammalian aging brain is limited. Here we generated spatial transcriptomic maps of young and old mouse brains. We identified 27 distinguished brain spatial domains, including layer-specific subregions that are difficult to dissect individually. We comprehensively characterized spatial-specific changes in gene expression in the aging brain, particularly for isocortex, the hippocampal formation, brainstem and fiber tracts, and validated some gene expression differences by qPCR and immunohistochemistry. We identified aging-related genes and pathways that vary in a coordinated manner across spatial regions and parsed the spatial features of aging-related signals, providing important clues to understand genes with specific functions in different brain regions during aging. Combined with single-cell transcriptomics data, we characterized the spatial distribution of brain cell types. The proportion of immature neurons decreased in the DG region with aging, indicating that the formation of new neurons is blocked. Finally, we detected changes in information interactions between regions and found specific pathways were deregulated with aging, including classic signaling WNT and layer-specific signaling COLLAGEN. In summary, we established a spatial molecular atlas of the aging mouse brain (http://sysbio.gzzoc.com/Mouse-Brain-Aging/), which provides important resources and novel insights into the molecular mechanism of brain aging.


Asunto(s)
Envejecimiento , Encéfalo , Transcriptoma , Animales , Envejecimiento/genética , Envejecimiento/metabolismo , Transcriptoma/genética , Ratones , Encéfalo/metabolismo , Masculino , Ratones Endogámicos C57BL
3.
Invest Ophthalmol Vis Sci ; 64(13): 47, 2023 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-37906058

RESUMEN

Purpose: The purpose of this study was to investigate the effects of silibinin on epithelial-mesenchymal transition (EMT) of retinal pigment epithelial (RPE) and proliferative vitreoretinopathy (PVR) formation, as well as its underlying molecular mechanism. Methods: Cellular morphological change and EMT molecular markers were evaluated by using phase contrast imaging, qPCR, and Western blot (WB) to investigate the impact of silibinin on the EMT of ARPE-19 cells. Scratch assay and transwell assay were used to study the effect of silibinin on cell migration. An intravitreally injected RPE-induced rat PVR model was used to assess the effect of silibinin on PVR in vivo. RNA-seq was applied to study the molecular mechanism of silibinin-mediated PVR prevention. Results: Silibinin inhibited TGFß1-induced EMT and migration of RPE in a dose-dependent manner in vitro. Moreover, silibinin prevented proliferative membrane formation in an intravitreal injected RPE-induced rat PVR model. In line with these findings, RNA-seq revealed a global suppression of TGFß1-induced EMT and migration-related genes by silibinin in RPEs. Mechanistically, silibinin reduced TGFß1-induced phosphorylation levels of Smad3 and Stat3, and Smad3 nuclear translocation in RPE. Conclusions: Silibinin inhibits the EMT of RPE cells in vitro and prevents the formation of PVR membranes in vivo. Mechanistically, silibinin inhibits Smad3 phosphorylation and suppresses Smad3 nuclear translocation through the inhibition of Stat3 phosphorylation. These findings suggest that silibinin may serve as a potential treatment for PVR.


Asunto(s)
Factor de Crecimiento Transformador beta , Vitreorretinopatía Proliferativa , Animales , Ratas , Fosforilación , Transición Epitelial-Mesenquimal , Vitreorretinopatía Proliferativa/tratamiento farmacológico , Silibina
4.
Front Public Health ; 10: 922325, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36299748

RESUMEN

Objectives: The purpose of this study was to explore whether team-based learning (TBL) was more effective than traditional didactic lectures (TDLs) in improving medical students' problem-solving and study skills in the clinical course of ophthalmology. In addition, we were also concerned about Chinese students' satisfaction with TBL. Methods: Our study program involved 275 students of the 5-year clinical medicine program from Central South China University, of which 140 were enrolled in a modified TBL course. A questionnaire that included closed-ended and open-ended items was distributed to students immediately following the completion of the TBL session, and 108 valid questionnaires were collected. Descriptive statistics were used to analyze quantitative data. The effects of the TBL module on students' performance were measured between the groups using a one-way between-group analysis of variance (ANOVA) test by the individual readiness assurance test (IRAT), the group readiness assurance test (GRAT), and final examination scores (FESs), compared with a class without the TBL session. Results: With our modified TBL strategy, 140 students achieved a mean test score of 72.65 on test questions that assessed their knowledge of ophthalmology compared to 135 students who achieved a mean score of 70.8 using the TDL method (p = 0.3434). The performance in a pre-class quiz was significantly better in the GRAT compared to the IRAT. In comparison to the TDL session, the modified TBL was preferred and acceptable by most medical students. Conclusions: By applying the modified TBL to ophthalmology, students improved their performance, self-study, and teamwork, and their class engagement and satisfaction were enhanced. However, TBL should be further optimized and developed to enhance educational outcomes.


Asunto(s)
Oftalmología , Estudiantes de Medicina , Humanos , Aprendizaje Basado en Problemas/métodos , Oftalmología/educación , China , Universidades
5.
Brain Commun ; 4(1): fcab310, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35169707

RESUMEN

Haploinsufficiency of the progranulin protein is a leading cause of frontotemporal lobar degeneration. Accumulating evidence support a crucial role of progranulin in the lysosome. Progranulin comprises 7.5 granulin repeats and is known to traffic to lysosomes via direct interactions with prosaposin or sortilin. Within the lysosome, progranulin gets processed into granulin peptides. Here, we report that sortilin and prosaposin independently regulate lysosomal trafficking of progranulin in vivo. The deletion of either prosaposin or sortilin alone results in a significant decrease in the ratio of granulin peptides versus full-length progranulin in mouse brain lysates. This decrease is further augmented by the deficiency of both prosaposin and sortilin. A concomitant increase in the levels of secreted progranulin in the serum was observed. Interestingly, while the deletion of both prosaposin and sortilin totally abolishes lysosomal localization of progranulin in neurons, it has a limited effect on lysosomal trafficking of progranulin in microglia, suggesting the existence of a novel sortilin and prosaposin independent pathway mediating progranulin lysosomal trafficking. In summary, our studies shed light on the regulation of lysosomal trafficking and processing of progranulin in vivo.

6.
Neuropathol Appl Neurobiol ; 48(2): e12778, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34823271

RESUMEN

AIMS: Accumulating evidence suggests that patients with frontotemporal lobar degeneration (FTLD) can have pathologic accumulation of multiple proteins, including tau and TDP-43. This study aimed to determine the frequency and characteristics of concurrent tau pathology in FTLD with TDP-43 pathology (FTLD-TDP). METHODS: The study included 146 autopsy-confirmed cases of FTLD-TDP and 55 cases of FTLD-TDP with motor neuron disease (FTLD-MND). Sections from the basal forebrain were screened for tau pathology with phosphorylated-tau immunohistochemistry. For cases with tau pathology on the screening section, additional brain sections were studied to establish a diagnosis. Genetic analysis of C9orf72, GRN and MAPT was performed on select cases. RESULTS: We found 72 cases (36%) with primary age-related tauopathy (PART), 85 (42%) with ageing-related tau astrogliopathy (ARTAG), 45 (22%) with argyrophilic grain disease (AGD) and 2 cases (1%) with corticobasal degeneration (CBD). Patients with ARTAG or AGD were significantly older than those without these comorbidities. One of the patients with FTLD-TDP and CBD had C9orf72 mutation and relatively mild tau pathology, consistent with incidental CBD. CONCLUSION: The coexistence of TDP-43 and tau pathologies was relatively common, particularly PART and ARTAG. Although rare, patients with FTLD can have multiple neurodegenerative proteinopathies. The absence of TDP-43-positive astrocytic plaques may suggest that CBD and FTLD-TDP were independent disease processes in the two patients with both tau and TDP-43 pathologies. It remains to be determined if mixed cases represent a unique disease process or two concurrent disease processes in an individual.


Asunto(s)
Encéfalo/metabolismo , Proteínas de Unión al ADN/metabolismo , Degeneración Lobar Frontotemporal/complicaciones , Neuronas/metabolismo , Tauopatías/complicaciones , Proteínas tau/metabolismo , Anciano , Anciano de 80 o más Años , Encéfalo/patología , Femenino , Degeneración Lobar Frontotemporal/metabolismo , Degeneración Lobar Frontotemporal/patología , Humanos , Masculino , Persona de Mediana Edad , Neuronas/patología , Tauopatías/metabolismo , Tauopatías/patología
7.
Neuropathol Appl Neurobiol ; 47(7): 931-941, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-33763863

RESUMEN

AIMS: This study aimed to clarify the different topographical distribution of tau pathology between progressive supranuclear palsy (PSP) and corticobasal degeneration (CBD) and establish a machine learning-based decision tree classifier. METHODS: Paraffin-embedded sections of the temporal cortex, motor cortex, caudate nucleus, globus pallidus, subthalamic nucleus, substantia nigra, red nucleus, and midbrain tectum from 1020 PSP and 199 CBD cases were assessed by phospho-tau immunohistochemistry. The severity of tau lesions (i.e., neurofibrillary tangle, coiled body, tufted astrocyte or astrocytic plaque, and tau threads) was semi-quantitatively scored in each region. Hierarchical cluster analysis was performed using tau pathology scores. A decision tree classifier was made with tau pathology scores using 914 cases. Cross-validation was done using 305 cases. An additional ten cases were used for a validation study. RESULTS: Cluster analysis displayed two distinct clusters; the first cluster included only CBD, and the other cluster included all PSP and six CBD cases. We built a decision tree, which used only seven decision nodes. The scores of tau threads in the caudate nucleus were the most decisive factor for predicting CBD. In a cross-validation, 302 out of 305 cases were correctly diagnosed. In the pilot validation study, three investigators made a correct diagnosis in all cases using the decision tree. CONCLUSION: Regardless of the morphology of astrocytic tau lesions, semi-quantitative tau pathology scores in select brain regions are sufficient to distinguish PSP and CBD. The decision tree simplifies neuropathologic differential diagnosis of PSP and CBD.


Asunto(s)
Degeneración Corticobasal/patología , Árboles de Decisión , Aprendizaje Automático , Ovillos Neurofibrilares/patología , Parálisis Supranuclear Progresiva/patología , Anciano , Anciano de 80 o más Años , Encéfalo/patología , Degeneración Corticobasal/diagnóstico , Diagnóstico Diferencial , Femenino , Humanos , Cuerpos de Inclusión/patología , Masculino , Persona de Mediana Edad , Degeneración Nerviosa/patología , Ovillos Neurofibrilares/metabolismo , Parálisis Supranuclear Progresiva/diagnóstico , Proteínas tau/metabolismo
8.
Brain Pathol ; 31(3): e12945, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33709463

RESUMEN

TMEM106B has been recently implicated in multiple neurodegenerative diseases. Here, Rademakers et al. report a late-onset cerebellar Purkinje cell loss and progressive decline in motor function and gait deficits in a conventional Tmem106b-/- mouse model. By using high-power microscopy and bulk RNA sequencing, the authors further identify lysosomal and immune dysfunction as potential underlying mechanisms of the Purkinje cell loss.


Asunto(s)
Células de Purkinje , Animales , Modelos Animales de Enfermedad , Ratones
9.
Adv Exp Med Biol ; 1281: 219-242, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33433878

RESUMEN

It has been more than a decade since heterozygous loss-of-function mutations in the progranulin gene (GRN) were first identified as an important genetic cause of frontotemporal lobar degeneration (FTLD). Due to the highly diverse biological functions of the progranulin (PGRN) protein, encoded by GRN, multiple possible disease mechanisms have been proposed. Early work focused on the neurotrophic properties of PGRN and its role in the inflammatory response. However, since the discovery of homozygous GRN mutations in patients with a lysosomal storage disorder, investigation into the possible roles of PGRN and its proteolytic cleavage products granulins, in lysosomal function and dysfunction, has taken center stage. In this chapter, we summarize the GRN mutational spectrum and its associated phenotypes followed by an in-depth discussion on the possible disease mechanisms implicated in FTLD-GRN. We conclude with key outstanding questions which urgently require answers to ensure safe and successful therapy development for GRN mutation carriers.


Asunto(s)
Degeneración Lobar Frontotemporal , Péptidos y Proteínas de Señalización Intercelular , Biología , Degeneración Lobar Frontotemporal/genética , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Lisosomas/genética , Mutación , Progranulinas/genética
10.
EMBO Rep ; 21(10): e50197, 2020 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-32761777

RESUMEN

Progranulin (PGRN) and transmembrane protein 106B (TMEM106B) are important lysosomal proteins implicated in frontotemporal lobar degeneration (FTLD) and other neurodegenerative disorders. Loss-of-function mutations in progranulin (GRN) are a common cause of FTLD, while TMEM106B variants have been shown to act as disease modifiers in FTLD. Overexpression of TMEM106B leads to lysosomal dysfunction, while loss of Tmem106b ameliorates lysosomal and FTLD-related pathologies in young Grn-/- mice, suggesting that lowering TMEM106B might be an attractive strategy for therapeutic treatment of FTLD-GRN. Here, we generate and characterize older Tmem106b-/- Grn-/- double knockout mice, which unexpectedly show severe motor deficits and spinal cord motor neuron and myelin loss, leading to paralysis and premature death at 11-12 months. Compared to Grn-/- , Tmem106b-/- Grn-/- mice have exacerbated FTLD-related pathologies, including microgliosis, astrogliosis, ubiquitin, and phospho-Tdp43 inclusions, as well as worsening of lysosomal and autophagic deficits. Our findings confirm a functional interaction between Tmem106b and Pgrn and underscore the need to rethink whether modulating TMEM106B levels is a viable therapeutic strategy.


Asunto(s)
Demencia Frontotemporal , Degeneración Lobar Frontotemporal , Animales , Degeneración Lobar Frontotemporal/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Proteínas de la Membrana , Ratones , Ratones Noqueados , Mutación , Proteínas del Tejido Nervioso , Progranulinas/genética
11.
Brain ; 143(7): 2255-2271, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32572497

RESUMEN

TMEM106B encodes a lysosomal membrane protein and was initially identified as a risk factor for frontotemporal lobar degeneration. Recently, a dominant D252N mutation in TMEM106B was shown to cause hypomyelinating leukodystrophy. However, how TMEM106B regulates myelination is still unclear. Here we show that TMEM106B is expressed and localized to the lysosome compartment in oligodendrocytes. TMEM106B deficiency in mice results in myelination defects with a significant reduction of protein levels of proteolipid protein (PLP) and myelin oligodendrocyte glycoprotein (MOG), the membrane proteins found in the myelin sheath. The levels of many lysosome proteins are significantly decreased in the TMEM106B-deficient Oli-neu oligodendroglial precursor cell line. TMEM106B physically interacts with the lysosomal protease cathepsin D and is required to maintain proper cathepsin D levels in oligodendrocytes. Furthermore, we found that TMEM106B deficiency results in lysosome clustering in the perinuclear region and a decrease in lysosome exocytosis and cell surface PLP levels. Moreover, we found that the D252N mutation abolished lysosome enlargement and lysosome acidification induced by wild-type TMEM106B overexpression. Instead, it stimulates lysosome clustering near the nucleus as seen in TMEM106B-deficient cells. Our results support that TMEM106B regulates myelination through modulation of lysosome function in oligodendrocytes.


Asunto(s)
Encéfalo/metabolismo , Lisosomas/metabolismo , Proteínas de la Membrana/metabolismo , Vaina de Mielina/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Oligodendroglía/metabolismo , Animales , Femenino , Degeneración Lobar Frontotemporal/genética , Humanos , Masculino , Proteínas de la Membrana/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/deficiencia
12.
Brain ; 143(6): 1905-1919, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32504082

RESUMEN

Genetic variants that define two distinct haplotypes at the TMEM106B locus have been implicated in multiple neurodegenerative diseases and in healthy brain ageing. In frontotemporal dementia (FTD), the high expressing TMEM106B risk haplotype was shown to increase susceptibility for FTD with TDP-43 inclusions (FTD-TDP) and to modify disease penetrance in progranulin mutation carriers (FTD-GRN). To elucidate the biological function of TMEM106B and determine whether lowering TMEM106B may be a viable therapeutic strategy, we performed brain transcriptomic analyses in 8-month-old animals from our recently developed Tmem106b-/- mouse model. We included 10 Tmem106b+/+ (wild-type), 10 Tmem106b+/- and 10 Tmem106-/- mice. The most differentially expressed genes (153 downregulated and 60 upregulated) were identified between Tmem106b-/- and wild-type animals, with an enrichment for genes implicated in myelination-related cellular processes including axon ensheathment and oligodendrocyte differentiation. Co-expression analysis also revealed that the most downregulated group of correlated genes was enriched for myelination-related processes. We further detected a significant loss of OLIG2-positive cells in the corpus callosum of Tmem106b-/- mice, which was present already in young animals (21 days) and persisted until old age (23 months), without worsening. Quantitative polymerase chain reaction revealed a reduction of differentiated but not undifferentiated oligodendrocytes cellular markers. While no obvious changes in myelin were observed at the ultrastructure levels in unchallenged animals, treatment with cuprizone revealed that Tmem106b-/- mice are more susceptible to cuprizone-induced demyelination and have a reduced capacity to remyelinate, a finding which we were able to replicate in a newly generated Tmem106b CRISPR/cas9 knock-out mouse model. Finally, using a TMEM106B HeLa knock-out cell line and primary cultured oligodendrocytes, we determined that loss of TMEM106B leads to abnormalities in the distribution of lysosomes and PLP1. Together these findings reveal an important function for TMEM106B in myelination with possible consequences for therapeutic strategies aimed at lowering TMEM106B levels.


Asunto(s)
Demencia Frontotemporal/genética , Demencia Frontotemporal/terapia , Proteínas de la Membrana/genética , Proteínas del Tejido Nervioso/genética , Animales , Proteínas de Unión al ADN/metabolismo , Femenino , Expresión Génica/genética , Haplotipos , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación/genética , Fibras Nerviosas Mielínicas/patología , Proteínas del Tejido Nervioso/metabolismo , Polimorfismo de Nucleótido Simple/genética , Transcriptoma/genética
13.
PLoS One ; 14(7): e0212382, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31291241

RESUMEN

Mutation in the GRN gene, encoding the progranulin (PGRN) protein, shows a dose-dependent disease correlation, wherein haploinsufficiency results in frontotemporal lobar degeneration (FTLD) and complete loss results in neuronal ceroid lipofuscinosis (NCL). Although the exact function of PGRN is unknown, it has been increasingly implicated in lysosomal physiology. Here we report that PGRN interacts with the lysosomal enzyme, glucocerebrosidase (GCase), and is essential for proper GCase activity. GCase activity is significantly reduced in tissue lysates from PGRN-deficient mice. This is further evidence that reduced lysosomal hydrolase activity may be a pathological mechanism in cases of GRN-related FTLD and NCL.


Asunto(s)
Glucosilceramidasa/metabolismo , Progranulinas/deficiencia , Animales , Línea Celular , Modelos Animales de Enfermedad , Femenino , Degeneración Lobar Frontotemporal/genética , Degeneración Lobar Frontotemporal/metabolismo , Glucosilceramidasa/genética , Células HEK293 , Haploinsuficiencia , Humanos , Lisosomas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Lipofuscinosis Ceroideas Neuronales/genética , Lipofuscinosis Ceroideas Neuronales/metabolismo , Progranulinas/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
14.
Mol Neurodegener ; 13(1): 32, 2018 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-29929528

RESUMEN

BACKGROUND: Loss of function mutations in progranulin (GRN) are a major cause of frontotemporal dementia (FTD). Progranulin is a secreted glycoprotein that localizes to lysosomes and is critical for proper lysosomal function. Heterozygous GRN mutation carriers develop FTD with TDP-43 pathology and exhibit signs of lysosomal dysfunction in the brain, with increased levels of lysosomal proteins and lipofuscin accumulation. Homozygous GRN mutation carriers develop neuronal ceroid lipofuscinosis (NCL), an earlier-onset lysosomal storage disorder caused by severe lysosomal dysfunction. Multiple genome-wide association studies have shown that risk of FTD in GRN mutation carriers is modified by polymorphisms in TMEM106B, which encodes a lysosomal membrane protein. Risk alleles of TMEM106B may increase TMEM106B levels through a variety of mechanisms. Brains from FTD patients with GRN mutations exhibit increased TMEM106B expression, and protective TMEM106B polymorphisms are associated with decreased TMEM106B expression. Together, these data raise the possibility that reduction of TMEM106B levels may protect against the pathogenic effects of progranulin haploinsufficiency. METHODS: We crossed Tmem106b +/- mice with Grn +/- mice, which model the progranulin haploinsufficiency of GRN mutation carriers and develop age-dependent social deficits and lysosomal abnormalities in the brain. We tested whether partial Tmem106b reduction could normalize the social deficits and lysosomal abnormalities of Grn +/- mice. RESULTS: Partial reduction of Tmem106b levels did not correct the social deficits of Grn +/- mice. Tmem106b reduction also failed to normalize most lysosomal abnormalities of Grn +/- mice, except for ß-glucuronidase activity, which was suppressed by Tmem106b reduction and increased by progranulin insufficiency. CONCLUSIONS: These data do not support the hypothesis that Tmem106b reduction protects against the pathogenic effects of progranulin haploinsufficiency, but do show that Tmem106b reduction normalizes some lysosomal phenotypes in Grn +/- mice.


Asunto(s)
Demencia Frontotemporal/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Proteínas de la Membrana/genética , Proteínas del Tejido Nervioso/genética , Animales , Demencia Frontotemporal/patología , Granulinas , Haploinsuficiencia , Heterocigoto , Ratones , Ratones Mutantes , Mutación , Polimorfismo de Nucleótido Simple , Progranulinas
15.
Methods Mol Biol ; 1806: 269-288, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29956282

RESUMEN

Accumulating evidence suggests that progranulin is essential for proper lysosomal function. Progranulin is a lysosomal resident protein and sortilin has been demonstrated to be the lysosomal trafficking receptor for progranulin. Here we describe the methods used to study the interaction between progranulin and sortilin, as well as the critical role of sortilin in mediating the lysosomal delivery of progranulin.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Lisosomas/metabolismo , Biología Molecular/métodos , Progranulinas/metabolismo , Fosfatasa Alcalina/metabolismo , Animales , Encéfalo/metabolismo , Células COS , Chlorocebus aethiops , Células HEK293 , Humanos , Progranulinas/sangre , Unión Proteica , Transporte de Proteínas , Proteínas Recombinantes de Fusión/metabolismo
16.
Acta Neuropathol Commun ; 6(1): 42, 2018 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-29855382

RESUMEN

Loss-of-function mutations in progranulin (GRN) and a non-coding (GGGGCC)n hexanucleotide repeat expansions in C9ORF72 are the two most common genetic causes of frontotemporal lobar degeneration with aggregates of TAR DNA binding protein 43 (FTLD-TDP). TMEM106B encodes a type II transmembrane protein with unknown function. Genetic variants in TMEM106B associated with reduced TMEM106B levels have been identified as disease modifiers in individuals with GRN mutations and C9ORF72 expansions. Recently, loss of Tmem106b has been reported to protect the FTLD-like phenotypes in Grn-/- mice. Here, we generated Tmem106b-/- mice and examined whether loss of Tmem106b could rescue FTLD-like phenotypes in an AAV mouse model of C9ORF72-repeat induced toxicity. Our results showed that neither partial nor complete loss of Tmem106b was able to rescue behavioral deficits induced by the expression of (GGGGCC)66 repeats (66R). Loss of Tmem106b also failed to ameliorate 66R-induced RNA foci, dipeptide repeat protein formation and pTDP-43 pathological burden. We further found that complete loss of Tmem106b increased astrogliosis, even in the absence of 66R, and failed to rescue 66R-induced neuronal cell loss, whereas partial loss of Tmem106b significantly rescued the neuronal cell loss but not neuroinflammation induced by 66R. Finally, we showed that overexpression of 66R did not alter expression of Tmem106b and other lysosomal genes in vivo, and subsequent analyses in vitro found that transiently knocking down C9ORF72, but not overexpression of 66R, significantly increased TMEM106B and other lysosomal proteins. In summary, reducing Tmem106b levels failed to rescue FTLD-like phenotypes in a mouse model mimicking the toxic gain-of-functions associated with overexpression of 66R. Combined with the observation that loss of C9ORF72 and not 66R overexpression was associated with increased levels of TMEM106B, this work suggests that the protective TMEM106B haplotype may exert its effect in expansion carriers by counteracting lysosomal dysfunction resulting from a loss of C9ORF72.


Asunto(s)
Proteína C9orf72/genética , Expansión de las Repeticiones de ADN/genética , Degeneración Lobar Frontotemporal/genética , Degeneración Lobar Frontotemporal/terapia , Regulación de la Expresión Génica/genética , Proteínas de la Membrana/deficiencia , Proteínas Supresoras de Tumor/deficiencia , Animales , Proteína C9orf72/metabolismo , Línea Celular Transformada , Condicionamiento Psicológico/fisiología , Modelos Animales de Enfermedad , Conducta Exploratoria , Miedo/psicología , Degeneración Lobar Frontotemporal/psicología , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Glicerofosfatos , Humanos , Relaciones Interpersonales , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción Genética , Proteínas Supresoras de Tumor/genética
17.
Lancet Neurol ; 17(6): 548-558, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29724592

RESUMEN

BACKGROUND: Loss-of-function mutations in GRN cause frontotemporal lobar degeneration (FTLD). Patients with GRN mutations present with a uniform subtype of TAR DNA-binding protein 43 (TDP-43) pathology at autopsy (FTLD-TDP type A); however, age at onset and clinical presentation are variable, even within families. We aimed to identify potential genetic modifiers of disease onset and disease risk in GRN mutation carriers. METHODS: The study was done in three stages: a discovery stage, a replication stage, and a meta-analysis of the discovery and replication data. In the discovery stage, genome-wide logistic and linear regression analyses were done to test the association of genetic variants with disease risk (case or control status) and age at onset in patients with a GRN mutation and controls free of neurodegenerative disorders. Suggestive loci (p<1 × 10-5) were genotyped in a replication cohort of patients and controls, followed by a meta-analysis. The effect of genome-wide significant variants at the GFRA2 locus on expression of GFRA2 was assessed using mRNA expression studies in cerebellar tissue samples from the Mayo Clinic brain bank. The effect of the GFRA2 locus on progranulin concentrations was studied using previously generated ELISA-based expression data. Co-immunoprecipitation experiments in HEK293T cells were done to test for a direct interaction between GFRA2 and progranulin. FINDINGS: Individuals were enrolled in the current study between Sept 16, 2014, and Oct 5, 2017. After quality control measures, statistical analyses in the discovery stage included 382 unrelated symptomatic GRN mutation carriers and 1146 controls free of neurodegenerative disorders collected from 34 research centres located in the USA, Canada, Australia, and Europe. In the replication stage, 210 patients (67 symptomatic GRN mutation carriers and 143 patients with FTLD without GRN mutations pathologically confirmed as FTLD-TDP type A) and 1798 controls free of neurodegenerative diseases were recruited from 26 sites, 20 of which overlapped with the discovery stage. No genome-wide significant association with age at onset was identified in the discovery or replication stages, or in the meta-analysis. However, in the case-control analysis, we replicated the previously reported TMEM106B association (rs1990622 meta-analysis odds ratio [OR] 0·54, 95% CI 0·46-0·63; p=3·54 × 10-16), and identified a novel genome-wide significant locus at GFRA2 on chromosome 8p21.3 associated with disease risk (rs36196656 meta-analysis OR 1·49, 95% CI 1·30-1·71; p=1·58 × 10-8). Expression analyses showed that the risk-associated allele at rs36196656 decreased GFRA2 mRNA concentrations in cerebellar tissue (p=0·04). No effect of rs36196656 on plasma and CSF progranulin concentrations was detected by ELISA; however, co-immunoprecipitation experiments in HEK293T cells did suggest a direct binding of progranulin and GFRA2. INTERPRETATION: TMEM106B-related and GFRA2-related pathways might be future targets for treatments for FTLD, but the biological interaction between progranulin and these potential disease modifiers requires further study. TMEM106B and GFRA2 might also provide opportunities to select and stratify patients for future clinical trials and, when more is known about their potential effects, to inform genetic counselling, especially for asymptomatic individuals. FUNDING: National Institute on Aging, National Institute of Neurological Disorders and Stroke, Canadian Institutes of Health Research, Italian Ministry of Health, UK National Institute for Health Research, National Health and Medical Research Council of Australia, and the French National Research Agency.


Asunto(s)
Degeneración Lobar Frontotemporal/genética , Predisposición Genética a la Enfermedad/genética , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Mutación/genética , Progranulinas/genética , Edad de Inicio , Anciano , Estudios de Casos y Controles , Cerebelo/metabolismo , Femenino , Degeneración Lobar Frontotemporal/metabolismo , Estudio de Asociación del Genoma Completo , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Progranulinas/metabolismo , ARN Mensajero/metabolismo
18.
Neurotox Res ; 33(4): 856-867, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29188488

RESUMEN

Microglia are the resident immune cells of the central nervous system (CNS) and respond to a variety of endogenous and exogenous stimuli in order to restore cell and tissue homeostasis. Lipopolysaccharide (LPS) is one of these exogenous stimuli, constitutes a major component of the outer membrane of Gram-negative bacteria, and binds to the microglial pattern recognition receptor Toll-like receptor 4 (TLR4). LPS-induced microglia activation is believed to promote neurodegeneration by release of neurotoxic factors such as interleukin-1ß, tumor necrosis factor α, or nitric oxide. In the present study, we investigated whether the physical presence of microglia is required to promote neurotoxicity and whether microglia-derived factors are essential. Interestingly, we observed that dopaminergic (mDA) neuron survival was only affected in mixed neuron-glia cultures containing microglia but not in neuron-enriched cultures. Moreover, we clearly demonstrate that microglia-conditioned medium (MCM) after LPS treatment increased mDA neuron survival, process numbers as well as process length. The observed protective effects of MCM was rather caused by microglia-derived factors and only partially dependent on the increase in reactive astrocytes. These results indicate that LPS-induced microglia activation does not necessarily have detrimental effects on mDA neurons and further support the hypothesis that activated microglia support neuron survival by release of neurotrophic and neuroprotective factors.


Asunto(s)
Supervivencia Celular/efectos de los fármacos , Neuronas Dopaminérgicas/efectos de los fármacos , Lipopolisacáridos/farmacología , Microglía/efectos de los fármacos , Animales , Células Cultivadas , Medios de Cultivo Condicionados/farmacología , Dopamina/metabolismo , Ratones , Neuroglía/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo
20.
Mol Neurodegener ; 12(1): 62, 2017 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-28835281

RESUMEN

BACKGROUND: Mutations resulting in progranulin (PGRN) haploinsufficiency cause frontotemporal lobar degeneration with TDP-43-positive inclusions (FTLD-TDP), a devastating neurodegenerative disease. PGRN is localized to the lysosome and important for proper lysosome function. However, the metabolism of PGRN in the lysosome is still unclear. RESULTS: Here, we report that PGRN is processed into ~10 kDa peptides intracellularly in multiple cell types and tissues and this processing is dependent on lysosomal activities. PGRN endocytosed from the extracellular space is also processed in a similar manner. We further demonstrated that multiple cathepsins are involved in PGRN processing and cathepsin L cleaves PGRN in vitro. CONCLUSIONS: Our data support that PGRN is processed in the lysosome through the actions of cathepsins.


Asunto(s)
Catepsinas/metabolismo , Degeneración Lobar Frontotemporal/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Lisosomas/metabolismo , Mutación/genética , Animales , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Degeneración Lobar Frontotemporal/genética , Granulinas , Haploinsuficiencia/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Ratones Noqueados , Progranulinas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA