Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Pharmacol Res ; 201: 107098, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38325728

RESUMO

Neuronal death is one of the key pathologies in Alzheimer's disease (AD). How neuronal death begins in AD is far from clear, so clarifying this process may help develop effective therapies. This study collected single-cell RNA sequencing data of 85 AD samples and 83 control samples, covering the prefrontal cortex, internal olfactory cortex, superior parietal lobe, superior frontal gyrus, caudal internal olfactory cortex, somatosensory cortex, hippocampus, superior frontal cortex and peripheral blood mononuclear cells. Additionally, spatial transcriptomic data of coronal sections from 6 AppNL-G-F AD mice and 6 control C57Bl/6 J mice were acquired. The main single-cell and spatial transcriptomics results were experimentally validated in wild type and 5 × FAD mice. We found that the microglia subpopulation Mic_PTPRG can communicate with specific types of neurons (especially excitatory ExNeu_PRKN_VIRMA and inhibitory InNeu_PRKN_VIRMA neuronal subpopulations) and cause them to express PTPRG during AD progression. Within neurons, PTPRG binds and upregulates the m6A methyltransferase VIRMA, thus inhibiting translation of PRKN mRNA to prevent the clearance of damaged mitochondria in neurons through suppressing mitophagy. As the disease progresses, the energy and nutrient metabolic pathways in neurons are reprogrammed, leading to their death. Consistently, we determined that PTPTRG can physically interact with VIRMA in mouse brains and PRKN is significantly upregulated in 5 × FAD mouse brain. Altogether, our findings demonstrate that PTPRG activates the m6A methyltransferase VIRMA to block mitophagy-mediated neuronal death in AD, which is a potential pathway, through which microglia and neuronal PTPRG modify neuronal connections in the brain during AD progression.


Assuntos
Doença de Alzheimer , Animais , Camundongos , Doença de Alzheimer/genética , Leucócitos Mononucleares , Mitofagia , Perfilação da Expressão Gênica , Metiltransferases , Camundongos Endogâmicos C57BL
2.
Int J Mol Sci ; 24(5)2023 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-36902031

RESUMO

RNA-binding motif 8A (RBM8A) is a core component of the exon junction complex (EJC) that binds pre-mRNAs and regulates their splicing, transport, translation, and nonsense-mediated decay (NMD). Dysfunction in the core proteins has been linked to several detriments in brain development and neuropsychiatric diseases. To understand the functional role of Rbm8a in brain development, we have generated brain-specific Rbm8a knockout mice and used next-generation RNA-sequencing to identify differentially expressed genes (DEGs) in mice with heterozygous, conditional knockout (cKO) of Rbm8a in the brain at postnatal day 17 (P17) and at embryonic day 12. Additionally, we analyzed enriched gene clusters and signaling pathways within the DEGs. At the P17 time point, between the control and cKO mice, about 251 significant DEGs were identified. At E12, only 25 DEGs were identified in the hindbrain samples. Bioinformatics analyses have revealed many signaling pathways related to the central nervous system (CNS). When E12 and P17 results were compared, three DEGs, Spp1, Gpnmb, and Top2a, appeared to peak at different developmental time points in the Rbm8a cKO mice. Enrichment analyses suggested altered activity in pathways affecting cellular proliferation, differentiation, and survival. The results support the hypothesis that loss of Rbm8a causes decreased cellular proliferation, increased apoptosis, and early differentiation of neuronal subtypes, which may lead ultimately to an altered neuronal subtype composition in the brain.


Assuntos
Encéfalo , Transcriptoma , Animais , Camundongos , Camundongos Knockout , Encéfalo/metabolismo , Proteínas de Ligação a RNA/metabolismo , Transdução de Sinais
3.
Mol Ther ; 29(11): 3274-3292, 2021 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-33892188

RESUMO

With the development of CRISPR-Cas9-mediated gene-editing technologies, correction of disease-causing mutations has become possible. However, current gene-correction strategies preclude mutation repair in post-mitotic cells of human tissues, and a unique repair strategy must be designed and tested for each and every mutation that may occur in a gene. We have developed a novel gene-correction strategy, co-opting regulation bypass repair (CRBR), which can repair a spectrum of mutations in mitotic or post-mitotic cells and tissues. CRBR utilizes the non-homologous end joining (NHEJ) pathway to insert a coding sequence (CDS) and transcription/translation terminators targeted upstream of any CDS mutation and downstream of the transcriptional promoter. CRBR results in simultaneous co-option of the endogenous regulatory region and bypass of the genetic defect. We validated the CRBR strategy for human gene therapy by rescuing a mouse model of Wolcott-Rallison syndrome (WRS) with permanent neonatal diabetes caused by either a large deletion or a nonsense mutation in the PERK (EIF2AK3) gene. Additionally, we integrated a CRBR GFP-terminator cassette downstream of the human insulin promoter in cadaver pancreatic islets of Langerhans, which resulted in insulin promoter regulated expression of GFP, demonstrating the potential utility of CRBR in human tissue gene repair.


Assuntos
Sistemas CRISPR-Cas , Edição de Genes/métodos , Doenças Genéticas Inatas/genética , Doenças Genéticas Inatas/terapia , Terapia Genética , Animais , Linhagem Celular , Feminino , Expressão Gênica , Técnicas de Silenciamento de Genes , Ordem dos Genes , Marcação de Genes , Genes Reporter , Marcadores Genéticos , Terapia Genética/métodos , Vetores Genéticos/genética , Humanos , Masculino , Camundongos , Mutação , RNA Guia de Cinetoplastídeos , eIF-2 Quinase/genética
4.
Int J Mol Sci ; 23(18)2022 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-36142288

RESUMO

The exon junction complex (EJC) plays a crucial role in regulating gene expression at the levels of alternative splicing, translation, mRNA localization, and nonsense-mediated decay (NMD). The EJC is comprised of three core proteins: RNA-binding motif 8A (RBM8A), Mago homolog (MAGOH), eukaryotic initiation factor 4A3 (eIF4A3), and a peripheral EJC factor, metastatic lymph node 51 (MLN51), in addition to other peripheral factors whose structural integration is activity-dependent. The physiological and mechanistic roles of the EJC in contribution to molecular, cellular, and organismal level function continue to be explored for potential insights into genetic or pathological dysfunction. The EJC's specific role in the cell cycle and its implications in cancer and neurodevelopmental disorders prompt enhanced investigation of the EJC as a potential target for these diseases. In this review, we highlight the current understanding of the EJC's position in the cell cycle, its relation to cancer and developmental diseases, and potential avenues for therapeutic targeting.


Assuntos
Neoplasias , Transtornos do Neurodesenvolvimento , Ciclo Celular/genética , Fator de Iniciação 4A em Eucariotos/genética , Fator de Iniciação 4A em Eucariotos/metabolismo , Éxons/genética , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/genética , Transtornos do Neurodesenvolvimento/genética , Proteínas Nucleares/genética , Splicing de RNA , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo
5.
Mol Ther ; 28(1): 217-234, 2020 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-31551137

RESUMO

Adult mammalian brains have largely lost neuroregeneration capability except for a few niches. Previous studies have converted glial cells into neurons, but the total number of neurons generated is limited and the therapeutic potential is unclear. Here, we demonstrate that NeuroD1-mediated in situ astrocyte-to-neuron conversion can regenerate a large number of functional new neurons after ischemic injury. Specifically, using NeuroD1 adeno-associated virus (AAV)-based gene therapy, we were able to regenerate one third of the total lost neurons caused by ischemic injury and simultaneously protect another one third of injured neurons, leading to a significant neuronal recovery. RNA sequencing and immunostaining confirmed neuronal recovery after cell conversion at both the mRNA level and protein level. Brain slice recordings found that the astrocyte-converted neurons showed robust action potentials and synaptic responses at 2 months after NeuroD1 expression. Anterograde and retrograde tracing revealed long-range axonal projections from astrocyte-converted neurons to their target regions in a time-dependent manner. Behavioral analyses showed a significant improvement of both motor and cognitive functions after cell conversion. Together, these results demonstrate that in vivo cell conversion technology through NeuroD1-based gene therapy can regenerate a large number of functional new neurons to restore lost neuronal functions after injury.


Assuntos
Astrócitos/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Isquemia Encefálica/terapia , Reprogramação Celular/genética , Terapia Genética/métodos , Neurônios/metabolismo , Potenciais de Ação , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Dependovirus/genética , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Transgênicos , Degeneração Neural/terapia , Neuroglia/metabolismo , Ratos , Ratos Sprague-Dawley , Resultado do Tratamento
6.
Cells ; 13(6)2024 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-38534343

RESUMO

The role of RNA Binding Motif Protein 8a (RBM8A), an exon junction complex (EJC) component, in neurodevelopmental disorders has been increasingly studied for its crucial role in regulating multiple levels of gene expression. It regulates mRNA splicing, translation, and mRNA degradation and influences embryonic development. RBM8A protein is expressed in both neurons and astrocytes, but little is known about RBM8A's specific role in glial fibrillary acid protein (GFAP)-positive astrocytes. To address the role of RBM8A in astrocytes, we generated a conditional heterozygous knockout (KO) mouse line of Rbm8a in astrocytes using a GFAP-cre line. We confirmed a decreased expression of RBM8A in astrocytes of heterozygous conditional KO mice via RT-PCR and Sanger sequencing, as well as qRT-PCR, immunohistochemistry, and Western blot. Interestingly, these mice exhibit significantly increased movement and mobility, alongside sex-specific altered anxiety in the open field test (OFT) and elevated plus maze (OPM) tests. These tests, along with the rotarod test, suggest that these mice have normal motor coordination but hyperactive phenotypes. In addition, the haploinsufficiency of Rbm8a in astrocytes leads to a sex-specific change in astrocyte density in the dentate gyrus. This study further reveals the contribution of Rbm8a deletion to CNS pathology, generating more insights via the glial lens of an Rbm8a model of neurodevelopmental disorder.


Assuntos
Astrócitos , Neurônios , Masculino , Feminino , Camundongos , Animais , Astrócitos/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Neurônios/metabolismo , Camundongos Knockout , Éxons , Locomoção , Proteínas de Ligação a RNA/metabolismo
7.
J Virol ; 86(7): 3474-85, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22258249

RESUMO

Ubiquitin is important for the budding of many retroviruses and other enveloped viruses, but the precise role of ubiquitin in virus budding remains unclear. Here, we characterized the ubiquitination of the matrix (M) protein of a paramyxovirus, parainfluenza virus 5 (PIV5). The PIV5 M protein (but not the PIV5 nucleocapsid protein) was found to be targeted for monoubiquitination in transfected mammalian cells. Major sites of ubiquitin attachment identified by mass spectrometry analysis were lysine residues at amino acid positions 79/80, 130, and 247. The cumulative mutation of lysine residues 79, 80, and 130 to arginines led to an altered pattern of M protein ubiquitination and impaired viruslike particle (VLP) production. However, the cumulative mutation of lysine residues 79, 80, 130, and 247 to arginines restored M protein ubiquitination and VLP production, suggesting that ubiquitin is attached to alternative sites on the M protein when the primary ones have been removed. Additional lysine residues were targeted for mutagenesis based on the UbiPred algorithm. An M protein with seven lysine residues changed to arginines exhibited altered ubiquitination and poor VLP production. A recombinant virus encoding an M protein with seven lysines mutated was generated, and this virus exhibited a 6-fold-reduced maximum titer, with the defect being attributed mainly to the budding of noninfectious particles. The recombinant virus was assembly deficient, as judged by the redistribution of viral M and hemagglutinin-neuraminidase proteins in infected cells. Similar assembly defects were observed for the wild-type (wt) virus after treatment with a proteasome inhibitor. Collectively, these findings suggest that the monoubiquitination of the PIV5 M protein is important for proper virus assembly and for the budding of infectious particles.


Assuntos
Vírus da Parainfluenza 5/fisiologia , Infecções por Rubulavirus/virologia , Ubiquitina/metabolismo , Proteínas da Matriz Viral/metabolismo , Montagem de Vírus , Motivos de Aminoácidos , Sequência de Aminoácidos , Linhagem Celular , Humanos , Dados de Sequência Molecular , Vírus da Parainfluenza 5/química , Vírus da Parainfluenza 5/genética , Ubiquitina/genética , Ubiquitinação , Proteínas da Matriz Viral/química , Proteínas da Matriz Viral/genética , Liberação de Vírus
8.
Aging Dis ; 2023 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-38270116

RESUMO

AAV-PHP.eB depends on endothelial cells to highly transduce the central nervous system (CNS) and is widely used for intravenous gene therapy. However, the transduction profile and therapeutic efficiency after endothelial cell injury such as ischemic stroke is largely unknown. In this study, we tested the transduction profiles of AAV-PHP.eB and developed intravenous NeuroD1 gene therapy to treat ischemic stroke in mice. We found that AAV-PHP.eB-GFP control virus crossed the BBB and infected brain cells efficiently in normal brain. However, after stroke, AAV-PHP.eB-GFP control virus was highly restricted in the blood vessels. Surprisingly, after switching to therapeutic vector AAV-PHP.eB-NeuroD1-GFP, the viral vector successfully crossed blood vessels and infected brain cells. Using Tie2-cre transgenic mice, we demonstrated that NeuroD1 regulated endothelial gene expression to modulate AAV-PHP.eB transduction. Following the changes of signaling pathways in endothelial cells, NeuroD1 effectively protected BBB integrity, attenuated neuroinflammation, inhibited neuron apoptosis and rescued motor deficits after ischemic stroke. Moreover, NeuroD1 over-expression in brain cells further promoted neural regeneration. These results indicate that intravenous gene therapy using AAV-PHP.eB for ischemic stroke differs from intracranial gene therapy and NeuroD1 intravenous delivery using AAV-PHP.eB efficiently rescue both vascular damage and neuronal loss, providing an advancing therapeutic treatment for stroke.

9.
J Virol ; 85(5): 2050-9, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21147917

RESUMO

Paramyxovirus matrix (M) proteins organize virus assembly, linking viral glycoproteins and viral ribonucleoproteins together at virus assembly sites on cellular membranes. Using a yeast two-hybrid screening approach, we identified 14-3-3 as a binding partner for the M protein of parainfluenza virus 5 (PIV5). Binding in both transfected and PIV5-infected cells was confirmed by coimmunoprecipitation and was mapped to a C-terminal region within the M protein, namely, 366-KTKSLP-371. This sequence resembles known 14-3-3 binding sites, in which the key residue for binding is a phosphorylated serine residue. Mutation of S369 within the PIV5 M protein disrupted 14-3-3 binding and improved the budding of both virus-like particles (VLPs) and recombinant viruses, suggesting that 14-3-3 binding impairs virus budding. 14-3-3 protein overexpression reduced the budding of VLPs. Using (33)P labeling, phosphorylated M protein was detected in PIV5-infected cells, and this phosphorylation was nearly absent in cells infected with a recombinant virus harboring an S369A mutation within the M protein. Assembly of the M protein into clusters and filaments at infected cell surfaces was enhanced in cells infected with a recombinant virus defective in 14-3-3 binding. These findings support a model in which a portion of M protein within PIV5-infected cells is phosphorylated at residue S369, binds the 14-3-3 protein, and is held away from sites of virus budding.


Assuntos
Proteínas 14-3-3/metabolismo , Regulação para Baixo , Vírus da Parainfluenza 5/fisiologia , Infecções por Rubulavirus/metabolismo , Proteínas da Matriz Viral/metabolismo , Vírion/fisiologia , Montagem de Vírus , Proteínas 14-3-3/genética , Sequência de Aminoácidos , Linhagem Celular , Humanos , Dados de Sequência Molecular , Vírus da Parainfluenza 5/química , Vírus da Parainfluenza 5/genética , Fosforilação , Ligação Proteica , Infecções por Rubulavirus/genética , Infecções por Rubulavirus/virologia , Alinhamento de Sequência , Proteínas da Matriz Viral/química , Proteínas da Matriz Viral/genética , Vírion/química , Vírion/genética
10.
Cancer Biol Med ; 2021 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-33755378

RESUMO

OBJECTIVE: Glioblastoma (GBM) is the most prevalent and aggressive adult primary cancer in the central nervous system. Therapeutic approaches for GBM treatment are under intense investigation, including the use of emerging immunotherapies. Here, we propose an alternative approach to treat GBM through reprogramming proliferative GBM cells into non-proliferative neurons. METHODS: Retroviruses were used to target highly proliferative human GBM cells through overexpression of neural transcription factors. Immunostaining, electrophysiological recording, and bulk RNA-seq were performed to investigate the mechanisms underlying the neuronal conversion of human GBM cells. An in vivo intracranial xenograft mouse model was used to examine the neuronal conversion of human GBM cells. RESULTS: We report efficient neuronal conversion from human GBM cells by overexpressing single neural transcription factor Neurogenic differentiation 1 (NeuroD1), Neurogenin-2 (Neurog2), or Achaete-scute homolog 1 (Ascl1). Subtype characterization showed that the majority of Neurog2- and NeuroD1-converted neurons were glutamatergic, while Ascl1 favored GABAergic neuron generation. The GBM cell-converted neurons not only showed pan-neuronal markers but also exhibited neuron-specific electrophysiological activities. Transcriptome analyses revealed that neuronal genes were activated in glioma cells after overexpression of neural transcription factors, and different signaling pathways were activated by different neural transcription factors. Importantly, the neuronal conversion of GBM cells was accompanied by significant inhibition of GBM cell proliferation in both in vitro and in vivo models. CONCLUSIONS: These results suggest that GBM cells can be reprogrammed into different subtypes of neurons, leading to a potential alternative approach to treat brain tumors using in vivo cell conversion technology.

11.
Front Cell Dev Biol ; 9: 720078, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34490268

RESUMO

Neural circuits underlying brain functions are vulnerable to damage, including ischemic injury, leading to neuronal loss and gliosis. Recent technology of direct conversion of endogenous astrocytes into neurons in situ can simultaneously replenish the neuronal population and reverse the glial scar. However, whether these newly reprogrammed neurons undergo normal development, integrate into the existing neuronal circuit, and acquire functional properties specific for this circuit is not known. We investigated the effect of NeuroD1-mediated in vivo direct reprogramming on visual cortical circuit integration and functional recovery in a mouse model of ischemic injury. After performing electrophysiological extracellular recordings and two-photon calcium imaging of reprogrammed cells in vivo and mapping the synaptic connections formed onto these cells ex vivo, we discovered that NeuroD1 reprogrammed neurons were integrated into the cortical microcircuit and acquired direct visual responses. Furthermore, following visual experience, the reprogrammed neurons demonstrated maturation of orientation selectivity and functional connectivity. Our results show that NeuroD1-reprogrammed neurons can successfully develop and integrate into the visual cortical circuit leading to vision recovery after ischemic injury.

12.
Front Cell Neurosci ; 14: 594170, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33250718

RESUMO

Injuries in the central nervous system (CNS) often causes neuronal loss and glial scar formation. We have recently demonstrated NeuroD1-mediated direct conversion of reactive glial cells into functional neurons in adult mouse brains. Here, we further investigate whether such direct glia-to-neuron conversion technology can reverse glial scar back to neural tissue in a severe stab injury model of the mouse cortex. Using an adeno-associated virus (AAV)-based gene therapy approach, we ectopically expressed a single neural transcription factor NeuroD1 in reactive astrocytes in the injured areas. We discovered that the reactive astrocytes were efficiently converted into neurons both before and after glial scar formation, and the remaining astrocytes proliferated to repopulate themselves. The astrocyte-converted neurons were highly functional, capable of firing action potentials and establishing synaptic connections with other neurons. Unexpectedly, the expression of NeuroD1 in reactive astrocytes resulted in a significant reduction of toxic A1 astrocytes, together with a significant decrease of reactive microglia and neuroinflammation. Furthermore, accompanying the regeneration of new neurons and repopulation of new astrocytes, new blood vessels emerged and blood-brain-barrier (BBB) was restored. These results demonstrate an innovative neuroregenerative gene therapy that can directly reverse glial scar back to neural tissue, opening a new avenue for brain repair after injury.

13.
Nat Commun ; 11(1): 1105, 2020 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-32107381

RESUMO

Huntington's disease (HD) is caused by Huntingtin (Htt) gene mutation resulting in the loss of striatal GABAergic neurons and motor functional deficits. We report here an in vivo cell conversion technology to reprogram striatal astrocytes into GABAergic neurons in both R6/2 and YAC128 HD mouse models through AAV-mediated ectopic expression of NeuroD1 and Dlx2 transcription factors. We found that the astrocyte-to-neuron (AtN) conversion rate reached 80% in the striatum and >50% of the converted neurons were DARPP32+ medium spiny neurons. The striatal astrocyte-converted neurons showed action potentials and synaptic events, and projected their axons to the targeted globus pallidus and substantia nigra in a time-dependent manner. Behavioral analyses found that NeuroD1 and Dlx2-treated R6/2 mice showed a significant extension of life span and improvement of motor functions. This study demonstrates that in vivo AtN conversion may be a disease-modifying gene therapy to treat HD and other neurodegenerative disorders.


Assuntos
Astrócitos/fisiologia , Técnicas de Reprogramação Celular/métodos , Corpo Estriado/patologia , Neurônios GABAérgicos/fisiologia , Terapia Genética/métodos , Doença de Huntington/terapia , Potenciais de Ação/fisiologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Técnicas de Observação do Comportamento , Comportamento Animal , Corpo Estriado/citologia , Dependovirus/genética , Modelos Animais de Doenças , Expressão Ectópica do Gene , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Células HEK293 , Proteínas de Homeodomínio , Humanos , Proteína Huntingtina/genética , Doença de Huntington/genética , Doença de Huntington/patologia , Longevidade , Camundongos , Camundongos Transgênicos , Técnicas de Patch-Clamp , Técnicas Estereotáxicas , Fatores de Transcrição
14.
Front Cell Dev Biol ; 8: 591883, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33425896

RESUMO

Spinal cord injury (SCI) often leads to impaired motor and sensory functions, partially because the injury-induced neuronal loss cannot be easily replenished through endogenous mechanisms. In vivo neuronal reprogramming has emerged as a novel technology to regenerate neurons from endogenous glial cells by forced expression of neurogenic transcription factors. We have previously demonstrated successful astrocyte-to-neuron conversion in mouse brains with injury or Alzheimer's disease by overexpressing a single neural transcription factor NeuroD1. Here we demonstrate regeneration of spinal cord neurons from reactive astrocytes after SCI through AAV NeuroD1-based gene therapy. We find that NeuroD1 converts reactive astrocytes into neurons in the dorsal horn of stab-injured spinal cord with high efficiency (~95%). Interestingly, NeuroD1-converted neurons in the dorsal horn mostly acquire glutamatergic neuronal subtype, expressing spinal cord-specific markers such as Tlx3 but not brain-specific markers such as Tbr1, suggesting that the astrocytic lineage and local microenvironment affect the cell fate after conversion. Electrophysiological recordings show that the NeuroD1-converted neurons can functionally mature and integrate into local spinal cord circuitry by displaying repetitive action potentials and spontaneous synaptic responses. We further show that NeuroD1-mediated neuronal conversion can occur in the contusive SCI model with a long delay after injury, allowing future studies to further evaluate this in vivo reprogramming technology for functional recovery after SCI. In conclusion, this study may suggest a paradigm shift from classical axonal regeneration to neuronal regeneration for spinal cord repair, using in vivo astrocyte-to-neuron conversion technology to regenerate functional new neurons in the gray matter.

15.
BMC Dev Biol ; 8: 4, 2008 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-18208612

RESUMO

BACKGROUND: Extracellular domains of the Notch family of signalling receptors contain many EGF repeat domains, as do their major ligands. Some EGF repeats are modified by O-fucosylation, and most have no identified role in ligand binding. RESULTS: Using a binding assay with purified proteins in vitro, it was determined that, in addition to binding to Delta, the ligand binding region of Notch bound to EGF repeats 22-27 of Notch, but not to other EGF repeat regions of Notch. EGF repeats 22-27 of Drosophila Notch overlap the genetically-defined 'Abruptex' region, and competed with Delta for binding to proteins containing the ligand-binding domain. Delta differed from the Abruptex domain in showing markedly enhanced binding at acid pH. Both Delta and the Abruptex region are heavily modified by protein O-fucosylation, but the split mutation of Drosophila Notch, which affects O-fucosylation of EGF repeat 14, did not affect binding of Notch to either Delta or the Abruptex region. CONCLUSION: The Abruptex region may serve as a barrier to Notch activation by competing for the ligand-binding domain of Notch.


Assuntos
Drosophila melanogaster/metabolismo , Proteínas de Membrana/metabolismo , Receptores Notch/química , Receptores Notch/metabolismo , Animais , Fucose/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Peptídeos e Proteínas de Sinalização Intracelular , Ligantes , Proteínas de Membrana/química , Proteínas Mutantes/metabolismo , Mutação/genética , Ligação Proteica , Estrutura Terciária de Proteína , Receptores Fc/metabolismo , Sequências Repetitivas de Aminoácidos
16.
Oncogene ; 23(2): 457-64, 2004 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-14724574

RESUMO

Telomerase is a therapeutic target for cancer. Human telomerase reverse transcriptase (hTERT), the catalytic subunit of the telomerase, is transcriptionaly upregulated exclusively in about 90% of cancer cells. Previous studies have demonstrated that hTERT promoter can control the expression of exogenous genes to the telomerase-positive cancer cells, thus hTERT promoter is an excellent candidate for generating cancer-specific oncolytic adenovirus. In this study, we devised a novel oncolytic adenovirus (Ad.TERT) by replacing the normal E1A regulatory elements with hTERT promoter. Ad.TERT displays cancer-specific E1A expression, virus replication and cytolysis in in vitro experiments. In animal experiments, intratumoral administration of Ad.TERT demonstrates potent antitumoral efficacy at least in two xenograft models (Bcap37 and BEL7404). Ad.TERT was targeted by the telomerase activity in cancer cells and has potent antitumoral efficacy in vivo, and since telomerase activity is a wide-ranged tumor marker, Ad.TERT could be a powerful therapeutic agent for a variety of cancers.


Assuntos
Adenoviridae/genética , Adenoviridae/fisiologia , Proteínas E1A de Adenovirus/genética , Proteínas E1A de Adenovirus/metabolismo , Antineoplásicos , Neoplasias/enzimologia , Neoplasias/patologia , Telomerase/genética , Telomerase/metabolismo , Animais , Western Blotting , Morte Celular , Proteínas de Ligação a DNA , Feminino , Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Mutagênese Sítio-Dirigida , Transplante de Neoplasias , Neoplasias/genética , Neoplasias/virologia , Proibitinas , Regiões Promotoras Genéticas/genética , Células Tumorais Cultivadas , Replicação Viral
17.
J Interferon Cytokine Res ; 24(4): 219-30, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15144568

RESUMO

To improve the therapeutic effect of ONYX015, an E1B55kD-deleted replication-competent adenovirus, ZD55 was constructed and armed with the therapeutic gene hTRAIL to form ZD55-hTRAIL, which was used for cancer therapy and which we call Targeting Gene-ViroTherapy. In vitro experiments with SW620, HCT116, and HT29 colorectal carcinoma cell lines demonstrated that they were all sensitive to ZD55-hTRAIL, and especially sensitive to ZD55-hTRAIL plus 5-fluorouracil (5-FU) treatment. In the SW620 xenograft tumor model, various treatment groups showed marked differences at week 11, with the tumor volume for the phosphate-buffered saline (PBS) treatment group >1700 mm3, for 5-FU > 1300 mm3, for ONYX015 1051.3 mm3, for ZD55-hTRAIL 600.05 mm3, and for ZD55-hTRAIL plus 5-FU 230.2 mm3. At the end of week 14, tumor-bearing mice in the other groups almost all died, whereas all the mice in the combined treatment group were alive, with one mouse tumor free. By transmission electron microscopy (TEM) assay, most tumor cells treated with ONYX015 or with ZD55-hTRAIL singly or in combination with 5-FU were lysed due to viral propagation. RT-PCR analysis and immunohistochemistry examination revealed that hTRAIL was expressed in ZD55-hTRAIL-treated SW620 tumor tissue. Furthermore, no detectable hepatoxicity was found by serum enzyme level analysis. These results suggest that ZD55-hTRAIL alone or in combination with 5-FU may have potential clinical implications.


Assuntos
Adenoviridae/genética , Antimetabólitos Antineoplásicos/uso terapêutico , Neoplasias Colorretais/terapia , Fluoruracila/uso terapêutico , Terapia Genética , Adenoviridae/metabolismo , Animais , Proteínas Reguladoras de Apoptose , Linhagem Celular , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/virologia , Sinergismo Farmacológico , Fluoruracila/farmacologia , Vetores Genéticos/química , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Vetores Genéticos/farmacologia , Humanos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/uso terapêutico , Camundongos , Camundongos Nus , Ligante Indutor de Apoptose Relacionado a TNF , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Virology ; 397(1): 155-66, 2010 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-19932912

RESUMO

Paramyxovirus matrix (M) proteins organize virus assembly, functioning as adapters that link together viral ribonucleoprotein complexes and viral glycoproteins at infected cell plasma membranes. M proteins may also function to recruit and manipulate host factors to assist virus budding, similar to retroviral Gag proteins. By yeast two-hybrid screening, angiomotin-like 1 (AmotL1) was identified as a host factor that interacts with the M protein of parainfluenza virus 5 (PIV5). AmotL1-M protein interaction was observed in yeast, in transfected mammalian cells, and in virus-infected cells. Binding was mapped to a 83-amino acid region derived from the C-terminal portion of AmotL1. Overexpression of M-binding AmotL1-derived polypeptides potently inhibited production of PIV5 VLPs and impaired virus budding. Expression of these polypeptides moderately inhibited production of mumps VLPs, but had no effect on production of Nipah VLPs. siRNA-mediated depletion of AmotL1 protein reduced PIV5 budding, suggesting that this interaction is beneficial to paramyxovirus infection.


Assuntos
Proteínas de Membrana/metabolismo , Mapeamento de Interação de Proteínas , Respirovirus/fisiologia , Proteínas da Matriz Viral/metabolismo , Montagem de Vírus , Angiomotinas , Interações Hospedeiro-Patógeno , Humanos , Ligação Proteica , Técnicas do Sistema de Duplo-Híbrido
19.
Mol Ther ; 11(4): 531-41, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15771956

RESUMO

Virotherapy with oncolytic viruses is a highly promising approach for cancer therapy. To improve further the therapeutic effect of oncolytic viruses, therapeutic genes have been incorporated into these types of vectors. In this study, we have inserted hTRAIL (approved gene symbol TNFSF10) into the ZD55 vector, which was based on deletion of the adenoviral E1B 55-kDa gene and could replicate in and lyse p53-deficient tumors. Our data shows that infection of colorectal carcinoma cells with ZD55-hTRAIL resulted in tumor cell death that was much greater than that induced by ZD55 vector or replication-defective adenovirus expressing hTRAIL. In contrast to these, ZD55-hTRAIL did not induce any cytopathic effect in normal cells. Treatment of established tumor with ZD55-hTRAIL resulted in dramatic inhibition of tumor growth in an animal model of colorectal carcinoma. However, when the established tumors were treated by coadministration of ZD55-hTRAIL and Ad-k5, we observed complete eradication of the established tumors in all animals treated with the combined therapy. This strong anti-tumor activity was due to the fact that two genes may act with compensative (or synergic) effect through different mechanisms to kill tumors. Therefore, targeting dual gene-virotherapy may be one of the best strategies for cancer therapy if two suitable genes are chosen.


Assuntos
Adenoviridae/genética , Carcinoma/terapia , Neoplasias Colorretais/terapia , Terapia Genética/métodos , Glicoproteínas de Membrana/genética , Fragmentos de Peptídeos/genética , Plasminogênio/genética , Fator de Necrose Tumoral alfa/genética , Proteínas E1B de Adenovirus/genética , Animais , Apoptose , Proteínas Reguladoras de Apoptose , Carcinoma/genética , Neoplasias Colorretais/genética , Vetores Genéticos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Ligante Indutor de Apoptose Relacionado a TNF , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/genética , Replicação Viral/genética
20.
J Biol Chem ; 277(50): 48677-84, 2002 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-12324475

RESUMO

Two antiapoptotic types of genes, iap and p35, were found in baculoviruses. P35 is a 35-kDa protein that can suppress apoptosis induced by virus infection or by diverse stimuli in vertebrates or invertebrates. iap homologues were identified in insects and mammals. Recently, we have identified sl-p49, a novel apoptosis suppressor gene and the first homologue of p35, in the genome of the Spodoptera littoralis nucleopolyhedrovirus. Here we show that sl-p49 encodes a 49-kDa protein, confirmed its primary structure that displays 48.8% identity to P35, and performed computer-assisted modeling of P49 based on the structure of P35. We demonstrated that P49 is able to inhibit insect and human effector caspases, which requires P49 cleavage at Asp(94). Finally we identified domains important for P49's antiapoptotic function that include a reactive site loop (RSL) protruding from a beta-barrel domain. RSL begins at an amphipathic alpha1 helix, traverses the beta-sheet central region, exposing Asp(94) at the apex, and rejoins the beta-barrel. Our model predicted seven alpha-helical motifs, three of them unique to P49. alpha-Helical motifs alpha(1), alpha(2), and alpha(4') were required for P49 function. The high structural homology between P49 and P35 suggests that these molecules bear a scaffold common to baculovirus "apoptotic suppressor" proteins. P49 may serve as a novel tool to analyze the contribution of different components of the caspase chain in the apoptotic response in organisms not related phylogenetically.


Assuntos
Apoptose/fisiologia , Nucleopoliedrovírus/metabolismo , Spodoptera/virologia , Proteínas Virais/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Linhagem Celular , Primers do DNA , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese , Conformação Proteica , Homologia de Sequência de Aminoácidos , Proteínas Virais/química , Proteínas Virais/genética , Proteínas Virais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA