Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Neuropathol Appl Neurobiol ; 48(2): e12778, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34823271

RESUMO

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.


Assuntos
Encéfalo/metabolismo , Proteínas de Ligação a DNA/metabolismo , Degeneração Lobar Frontotemporal/complicações , Neurônios/metabolismo , Tauopatias/complicações , Proteínas tau/metabolismo , Idoso , Idoso de 80 Anos ou mais , Encéfalo/patologia , Feminino , Degeneração Lobar Frontotemporal/metabolismo , Degeneração Lobar Frontotemporal/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Neurônios/patologia , Tauopatias/metabolismo , Tauopatias/patologia
2.
EMBO Rep ; 21(10): e50197, 2020 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-32761777

RESUMO

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.


Assuntos
Demência Frontotemporal , Degeneração Lobar Frontotemporal , Animais , Degeneração Lobar Frontotemporal/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , Proteínas de Membrana , Camundongos , Camundongos Knockout , Mutação , Proteínas do Tecido Nervoso , Progranulinas/genética
3.
Neuropathol Appl Neurobiol ; 47(7): 931-941, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-33763863

RESUMO

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.


Assuntos
Degeneração Corticobasal/patologia , Árvores de Decisões , Aprendizado de Máquina , Emaranhados Neurofibrilares/patologia , Paralisia Supranuclear Progressiva/patologia , Idoso , Idoso de 80 Anos ou mais , Encéfalo/patologia , Degeneração Corticobasal/diagnóstico , Diagnóstico Diferencial , Feminino , Humanos , Corpos de Inclusão/patologia , Masculino , Pessoa de Meia-Idade , Degeneração Neural/patologia , Emaranhados Neurofibrilares/metabolismo , Paralisia Supranuclear Progressiva/diagnóstico , Proteínas tau/metabolismo
4.
Brain ; 143(7): 2255-2271, 2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32572497

RESUMO

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.


Assuntos
Encéfalo/metabolismo , Lisossomos/metabolismo , Proteínas de Membrana/metabolismo , Bainha de Mielina/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Oligodendroglia/metabolismo , Animais , Feminino , Degeneração Lobar Frontotemporal/genética , Humanos , Masculino , Proteínas de Membrana/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/deficiência
5.
Brain ; 143(6): 1905-1919, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32504082

RESUMO

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.


Assuntos
Demência Frontotemporal/genética , Demência Frontotemporal/terapia , Proteínas de Membrana/genética , Proteínas do Tecido Nervoso/genética , Animais , Proteínas de Ligação a DNA/metabolismo , Feminino , Expressão Gênica/genética , Haplótipos , Células HeLa , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Masculino , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação/genética , Fibras Nervosas Mielinizadas/patologia , Proteínas do Tecido Nervoso/metabolismo , Polimorfismo de Nucleotídeo Único/genética , Transcriptoma/genética
6.
Adv Exp Med Biol ; 1281: 219-242, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33433878

RESUMO

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.


Assuntos
Degeneração Lobar Frontotemporal , Peptídeos e Proteínas de Sinalização Intercelular , Biologia , Degeneração Lobar Frontotemporal/genética , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Lisossomos/genética , Mutação , Progranulinas/genética
7.
J Neurochem ; 143(2): 236-243, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28640985

RESUMO

The frontotemporal lobar degeneration (FTLD) protein progranulin (PGRN) is essential for proper lysosomal function. PGRN localizes in the lysosomal compartment within the cell. Prosaposin (PSAP), the precursor of lysosomal saposin activators (saposin A, B, C, D), physically interacts with PGRN. Previously, we have shown that PGRN and PSAP facilitate each other's lysosomal trafficking. Here, we report that the interaction between PSAP and PGRN requires the linker region of saposin B and C (BC linker). PSAP protein with the BC linker mutated, fails to interact with PGRN and deliver PGRN to lysosomes in the biosynthetic and endocytic pathways. On the other hand, PGRN interacts with PSAP through multiple granulin motifs. Granulin D and E bind to PSAP with similar affinity as full-length PGRN. Read the Editorial Comment for this article on page 154.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Saposinas/genética , Saposinas/metabolismo , Sequência de Aminoácidos , Animais , Animais Recém-Nascidos , Células COS , Chlorocebus aethiops , Células HEK293 , Humanos , Camundongos , Camundongos Knockout , Progranulinas , Ligação Proteica/fisiologia , Ratos
8.
J Biol Chem ; 289(28): 19670-80, 2014 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-24872421

RESUMO

The sequential processing of single pass transmembrane proteins via ectodomain shedding followed by intramembrane proteolysis is involved in a wide variety of signaling processes, as well as maintenance of membrane protein homeostasis. Here we report that the recently identified frontotemporal lobar degeneration risk factor TMEM106B undergoes regulated intramembrane proteolysis. We demonstrate that TMEM106B is readily processed to an N-terminal fragment containing the transmembrane and intracellular domains, and this processing is dependent on the activities of lysosomal proteases. The N-terminal fragment is further processed into a small, rapidly degraded intracellular domain. The GxGD aspartyl proteases SPPL2a and, to a lesser extent, SPPL2b are responsible for this intramembrane cleavage event. Additionally, the TMEM106B paralog TMEM106A is also lysosomally localized; however, it is not a specific substrate of SPPL2a or SPPL2b. Our data add to the growing list of proteins that undergo intramembrane proteolysis and may shed light on the regulation of the frontotemporal lobar degeneration risk factor TMEM106B.


Assuntos
Ácido Aspártico Endopeptidases/metabolismo , Membrana Celular/metabolismo , Degeneração Lobar Frontotemporal , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteólise , Animais , Ácido Aspártico Endopeptidases/genética , Membrana Celular/genética , Células HEK293 , Humanos , Lisossomos/genética , Lisossomos/metabolismo , Proteínas de Membrana/genética , Camundongos , Proteínas do Tecido Nervoso/genética , Estrutura Terciária de Proteína , Fatores de Risco
9.
J Neurochem ; 134(1): 125-34, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25827682

RESUMO

Microglia-mediated neuroinflammation has been reported as a common feature of familial and sporadic forms of Parkinson's disease (PD), and a growing body of evidence indicates that onset and progression of PD correlates with the extent of neuroinflammatory responses involving Interferon γ (IFNγ). Transforming growth factor ß1 (TGFß1) has been shown to be a major player in the regulation of microglia activation states and functions and, thus, might be a potential therapeutic agent by shaping microglial activation phenotypes during the course of neurodegenerative diseases such as PD. In this study, we demonstrate that TGFß1 is able to block IFNγ-induced microglia activation by attenuating STAT1 phosphorylation and IFNγRα expression. Moreover, we identified a set of genes involved in microglial IFNγ signaling transduction that were significantly down-regulated upon TGFß1 treatment, resulting in decreased sensitivity of microglia toward IFNγ stimuli. Interestingly, genes mediating negative regulation of IFNγ signaling, such as SOCS2 and SOCS6, were up-regulated after TGFß1 treatment. Finally, we demonstrate that TGFß1 is capable of protecting midbrain dopaminergic (mDA) neurons from IFNγ-driven neurotoxicity in mixed neuron-glia cultures derived from embryonic day 14 (E14) midbrain tissue. Together, these data underline the importance of TGFß1 as a key immunoregulatory factor for microglia by silencing IFNγ-mediated microglia activation and, thereby, rescuing mDA neurons from IFNγ-induced neurotoxicity. Interferon γ (IFNγ) is a potent pro-inflammatory factor that triggers the activation of microglia and the subsequent release of neurotoxic factors. Transforming growth factor ß1 (TGFß1) is able to inhibit the IFNγ-mediated activation of microglia, which is characterized by the release of nitric oxide (NO) and tumor necrosis factor α (TNFα). By decreasing the expression of IFNγ-induced genes as well as the signaling receptor IFNγR1, TGFß1 reduces the responsiveness of microglia towards IFNγ. In mixed neuron-glia cultures, TGFß1 protects midbrain dopaminergic (mDA) neurons from IFNγ-induced neurotoxicity.


Assuntos
Neurônios Dopaminérgicos/metabolismo , Interferon gama/antagonistas & inibidores , Interferon gama/toxicidade , Microglia/metabolismo , Fármacos Neuroprotetores/farmacologia , Fator de Crescimento Transformador beta1/farmacologia , Animais , Animais Recém-Nascidos , Células Cultivadas , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/patologia , Mesencéfalo/efeitos dos fármacos , Mesencéfalo/metabolismo , Mesencéfalo/patologia , Camundongos , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Microglia/patologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia
10.
Aging Cell ; 23(5): e14109, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38372175

RESUMO

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.


Assuntos
Envelhecimento , Encéfalo , Transcriptoma , Animais , Envelhecimento/genética , Envelhecimento/metabolismo , Transcriptoma/genética , Camundongos , Encéfalo/metabolismo , Masculino , Camundongos Endogâmicos C57BL
11.
Glia ; 61(2): 287-300, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23065670

RESUMO

Microglia are the immune cells of the central nervous system (CNS) and play important roles under physiological and pathophysiological conditions. Activation of microglia has been reported for a variety of CNS diseases and is believed to be involved in inflammation-mediated neurodegeneration. Loss of TGFß1 results in increased microgliosis and neurodegeneration in mice which indicates that TGFß1 is an important regulator of microglial functions in vivo. Here, we addressed the role of endogenous TGFß signaling for microglia in vitro. We clearly demonstrate active TGFß signaling in primary microglia and further introduce Klf10 as a new TGFß target gene in microglia. Moreover, we provide evidence that microglia express and release TGFß1 that acts in an autocrine manner to activate microglial TGFß/Smad signaling in vitro. Using microarrays, we identified TGFß-regulated genes in microglia that are involved in TGFß1 processing, its extracellular storage as well as activation of latent TGFß. Finally, we demonstrate that pharmacological inhibition of microglial TGFß signaling resulted in upregulation of the proinflammatory markers IL6 and iNOS and downregulation of the alternative activation markers Arg1 and Ym1 in vitro. Together, these data clearly show that endogenous TGFß1 and autocrine TGFß signaling is important for microglial quiescence in vitro and further suggest the upregulation of TGFß1 in neurodegenerative diseases as a mechanism to regulate microglia functions and silence neuroinflammation.


Assuntos
Microglia/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Animais Recém-Nascidos , Encéfalo/citologia , Células Cultivadas , Citocinas/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Análise de Sequência com Séries de Oligonucleotídeos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais , Proteínas Smad/metabolismo , Fatores de Tempo , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/farmacologia
13.
Invest Ophthalmol Vis Sci ; 64(13): 47, 2023 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-37906058

RESUMO

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.


Assuntos
Fator de Crescimento Transformador beta , Vitreorretinopatia Proliferativa , Animais , Ratos , Fosforilação , Transição Epitelial-Mesenquimal , Vitreorretinopatia Proliferativa/tratamento farmacológico , Silibina
14.
J Neuroinflammation ; 9: 210, 2012 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-22947253

RESUMO

BACKGROUND: Microglia are the resident immune cells of the central nervous system and are accepted to be involved in a variety of neurodegenerative diseases. Several studies have demonstrated that microglia, like peripheral macrophages, exhibit two entirely different functional activation states, referred to as classical (M1) and alternative (M2) activation. TGFß is one of the most important anti-inflammatory cytokines and its effect on inhibiting microglia or macrophage classical activation has been extensively studied. However, the role of TGFß during alternative activation of microglia has not been described yet. METHODS: To investigate the role of TGFß in IL4-induced microglia alternative activation, both, BV2 as well as primary microglia from new born C57BL/6 mice were used. Quantitative RT-PCR and western blots were performed to detect mRNA and protein levels of the alternative activation markers Arginase1 (Arg1) and Chitinase 3-like 3 (Ym1) after treatment with IL4, TGFß or both. Endogenous TGFß release after IL4 treatment was evaluated using the mink lung epithelial cell (MLEC) assay and a direct TGFß2 ELISA. TGFß receptor type I inhibitor and MAPK inhibitor were applied to address the involvement of TGFß signalling and MAPK signalling in IL4-induced alternative activation of microglia. RESULTS: TGFß enhances IL4-induced microglia alternative activation by strongly increasing the expression of Arg1 and Ym1. This synergistic effect on Arg1 induction is almost completely blocked by the application of the MAPK inhibitor, PD98059. Further, treatment of primary microglia with IL4 increased the expression and secretion of TGFß2, suggesting an involvement of endogenous TGFß in IL4-mediated microglia activation process. Moreover, IL4-mediated induction of Arg1 and Ym1 is impaired after blocking the TGFß receptor I indicating that IL4-induced microglia alternative activation is dependent on active TGFß signalling. Interestingly, treatment of primary microglia with TGFß alone results in up regulation of the IL4 receptor alpha, indicating that TGFß increases the sensitivity of microglia for IL4 signals. CONCLUSIONS: Taken together, our data reveal a new role for TGFß during IL4-induced alternative activation of microglia and consolidate the essential functions of TGFß as an anti-inflammatory molecule and immunoregulatory factor for microglia.


Assuntos
Interleucina-4/farmacologia , Microglia/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta/metabolismo , Regulação para Cima/efeitos dos fármacos , Análise de Variância , Animais , Animais Recém-Nascidos , Arginase/genética , Arginase/metabolismo , Encéfalo/citologia , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Inibidores Enzimáticos/farmacologia , Ensaio de Imunoadsorção Enzimática , Lectinas/genética , Lectinas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Quinases Ativadas por Mitógeno/metabolismo , RNA Mensageiro/metabolismo , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Fator de Crescimento Transformador beta/genética , beta-N-Acetil-Hexosaminidases/genética , beta-N-Acetil-Hexosaminidases/metabolismo
16.
Brain Commun ; 4(1): fcab310, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35169707

RESUMO

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.

17.
Front Public Health ; 10: 922325, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36299748

RESUMO

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.


Assuntos
Oftalmologia , Estudantes de Medicina , Humanos , Aprendizagem Baseada em Problemas/métodos , Oftalmologia/educação , China , Universidades
18.
Ophthalmic Res ; 45(4): 180-90, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21051916

RESUMO

PURPOSE: Our aim was to establish a rat model of proliferative vitreoretinopathy (PVR) induced by macrophages and investigate whether macrophages can be a cell origin of fibroblast-like cells present in PVR. METHODS: One eye of each rat received an intravitreal injection of macrophages. Clinical examination was performed to evaluate the development of PVR. Histological study was carried out to observe the pathological progression. Immunohistochemical staining with vimentin (VIM), glial fibrillary acidic protein (GFAP), α-smooth-muscle actin (α-SM actin), cytokeratin (CK) and CD68 characterized the cell types within the PVR membranes. The distribution, morphological change of prelabeled macrophages, as well as their colocalization with CD68, VIM, GFAP, α-SM actin and CK, were observed on days 3, 14 and 28 after injection. RESULTS: In response to intravitreal injection of macrophages, 90% of the experimental rats developed PVR from postoperative day 7. The histological progression of PVR was characterized by the sequential appearance of inflammatory cell invasion, fibroblast proliferation and scar formation. The dominating cells comprising the proliferative membranes at the advanced stage were fibroblasts. Injected macrophages retained round shape and positive staining with CD68 on day 3. On day 28, they acquired elongated/spindle shape combined with intense staining of VIM but absence of CD68, GFAP, α-SM actin and CK, and became the primary constituent of fibrocellular membranes. CONCLUSIONS: Macrophages effectively and reproducibly induce the development of proliferative fibrocellular membranes in rats. In this PVR model, macrophages acquire fibroblast-like cell phenotype and contribute to fibrocellular membranes directly, suggesting that macrophages may be a cell origin of fibroblast-like cells involved in PVR.


Assuntos
Transdiferenciação Celular/fisiologia , Modelos Animais de Doenças , Fibroblastos/patologia , Macrófagos Peritoneais/patologia , Vitreorretinopatia Proliferativa/patologia , Actinas/metabolismo , Animais , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Fibroblastos/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Injeções Intravítreas , Queratinas/metabolismo , Macrófagos Peritoneais/metabolismo , Fenótipo , Ratos , Ratos Sprague-Dawley , Vimentina/metabolismo , Vitreorretinopatia Proliferativa/metabolismo
19.
Brain Pathol ; 31(3): e12945, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33709463

RESUMO

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.


Assuntos
Células de Purkinje , Animais , Modelos Animais de Doenças , Camundongos
20.
PLoS One ; 14(7): e0212382, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31291241

RESUMO

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.


Assuntos
Glucosilceramidase/metabolismo , Progranulinas/deficiência , Animais , Linhagem Celular , Modelos Animais de Doenças , Feminino , Degeneração Lobar Frontotemporal/genética , Degeneração Lobar Frontotemporal/metabolismo , Glucosilceramidase/genética , Células HEK293 , Haploinsuficiência , Humanos , Lisossomos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Lipofuscinoses Ceroides Neuronais/genética , Lipofuscinoses Ceroides Neuronais/metabolismo , Progranulinas/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA