Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 98
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nat Methods ; 19(1): 81-89, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34949807

RESUMO

CD4+ T cells are central mediators of adaptive and innate immune responses and constitute a major reservoir for human immunodeficiency virus (HIV) in vivo. Detailed investigations of resting human CD4+ T cells have been precluded by the absence of efficient approaches for genetic manipulation limiting our understanding of HIV replication and restricting efforts to find a cure. Here we report a method for rapid, efficient, activation-neutral gene editing of resting, polyclonal human CD4+ T cells using optimized cell cultivation and nucleofection conditions of Cas9-guide RNA ribonucleoprotein complexes. Up to six genes, including HIV dependency and restriction factors, were knocked out individually or simultaneously and functionally characterized. Moreover, we demonstrate the knock in of double-stranded DNA donor templates into different endogenous loci, enabling the study of the physiological interplay of cellular and viral components at single-cell resolution. Together, this technique allows improved molecular and functional characterizations of HIV biology and general immune functions in resting CD4+ T cells.


Assuntos
Linfócitos T CD4-Positivos/fisiologia , Sistemas CRISPR-Cas/genética , Edição de Genes/métodos , Infecções por HIV/genética , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/virologia , Proteína 9 Associada à CRISPR/genética , Movimento Celular/genética , Células Cultivadas , DNA , Técnicas de Inativação de Genes , Infecções por HIV/metabolismo , Humanos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Proteínas de Resistência a Myxovirus/genética , Proteínas de Resistência a Myxovirus/metabolismo , RNA Guia de Cinetoplastídeos , Proteína 1 com Domínio SAM e Domínio HD/genética , Transgenes , Fatores de Poliadenilação e Clivagem de mRNA/genética , Fatores de Poliadenilação e Clivagem de mRNA/metabolismo
2.
PLoS Pathog ; 17(4): e1009064, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33882114

RESUMO

Vaccines of outstanding efficiency, safety, and public acceptance are needed to halt the current SARS-CoV-2 pandemic. Concerns include potential side effects caused by the antigen itself and safety of viral DNA and RNA delivery vectors. The large SARS-CoV-2 spike (S) protein is the main target of current COVID-19 vaccine candidates but can induce non-neutralizing antibodies, which might cause vaccination-induced complications or enhancement of COVID-19 disease. Besides, encoding of a functional S in replication-competent virus vector vaccines may result in the emergence of viruses with altered or expanded tropism. Here, we have developed a safe single round rhabdovirus replicon vaccine platform for enhanced presentation of the S receptor-binding domain (RBD). Structure-guided design was employed to build a chimeric minispike comprising the globular RBD linked to a transmembrane stem-anchor sequence derived from rabies virus (RABV) glycoprotein (G). Vesicular stomatitis virus (VSV) and RABV replicons encoding the minispike not only allowed expression of the antigen at the cell surface but also incorporation into the envelope of secreted non-infectious particles, thus combining classic vector-driven antigen expression and particulate virus-like particle (VLP) presentation. A single dose of a prototype replicon vaccine complemented with VSV G, VSVΔG-minispike-eGFP (G), stimulated high titers of SARS-CoV-2 neutralizing antibodies in mice, equivalent to those found in COVID-19 patients, and protected transgenic K18-hACE2 mice from COVID-19-like disease. Homologous boost immunization further enhanced virus neutralizing activity. The results demonstrate that non-spreading rhabdovirus RNA replicons expressing minispike proteins represent effective and safe alternatives to vaccination approaches using replication-competent viruses and/or the entire S antigen.


Assuntos
Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Vacinas contra COVID-19/administração & dosagem , COVID-19/prevenção & controle , Imunização/métodos , SARS-CoV-2/imunologia , Vacinas de Partículas Semelhantes a Vírus/administração & dosagem , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , COVID-19/imunologia , COVID-19/virologia , Feminino , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL
3.
Nature ; 539(7628): 248-253, 2016 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-27783592

RESUMO

The ability of the adult mammalian brain to compensate for neuronal loss caused by injury or disease is very limited. Transplantation aims to replace lost neurons, but the extent to which new neurons can integrate into existing circuits is unknown. Here, using chronic in vivo two-photon imaging, we show that embryonic neurons transplanted into the visual cortex of adult mice mature into bona fide pyramidal cells with selective pruning of basal dendrites, achieving adult-like densities of dendritic spines and axonal boutons within 4-8 weeks. Monosynaptic tracing experiments reveal that grafted neurons receive area-specific, afferent inputs matching those of pyramidal neurons in the normal visual cortex, including topographically organized geniculo-cortical connections. Furthermore, stimulus-selective responses refine over the course of many weeks and finally become indistinguishable from those of host neurons. Thus, grafted neurons can integrate with great specificity into neocortical circuits that normally never incorporate new neurons in the adult brain.


Assuntos
Embrião de Mamíferos/citologia , Neocórtex/citologia , Vias Neurais , Neurônios/fisiologia , Neurônios/transplante , Córtex Visual/citologia , Vias Aferentes , Animais , Axônios/metabolismo , Diferenciação Celular , Rastreamento de Células , Espinhas Dendríticas/metabolismo , Vias Eferentes , Camundongos , Neocórtex/fisiologia , Neurônios/citologia , Terminações Pré-Sinápticas/metabolismo , Células Piramidais/citologia , Células Piramidais/fisiologia , Córtex Visual/fisiologia
4.
J Neurosci ; 40(46): 8870-8882, 2020 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-33051345

RESUMO

The emergence of genetic tools has provided new means of mapping functionality in central amygdala (CeA) neuron populations based on their molecular profiles, response properties, and importantly, connectivity patterns. While abundant evidence indicates that neuronal signals arrive in the CeA eliciting both aversive and appetitive behaviors, our understanding of the anatomy of the underlying long-range CeA network remains fragmentary. In this study, we combine viral tracings, electrophysiological, and optogenetic approaches to establish in male mice, a wiring chart between the insula cortex (IC), a major sensory input region of the lateral and capsular part of the CeA (CeL/C), and four principal output streams of this nucleus. We found that retrogradely labeled output neurons occupy discrete and likely strategic locations in the CeL/C, and that they are disproportionally controlled by the IC. We identified a direct line of connection between the IC and the lateral hypothalamus (LH), which engages numerous LH-projecting CeL/C cells whose activity can be strongly upregulated on firing of IC neurons. In comparison, CeL/C neurons projecting to the bed nucleus of the stria terminalis (BNST) are also frequently contacted by incoming IC axons, but the strength of this connection is weak. Our results provide a link between long-range inputs and outputs of the CeA and pave the way to a better understanding of how internal, external, and experience dependent information may impinge on action selection by the CeA.SIGNIFICANCE STATEMENT Our current knowledge of the circuit organization within the central amygdala (CeA), a critical regulator of emotional states, includes independent information about its long-range efferents and afferents. We do not know how incoming sensory information is appraised and routed through the CeA to the different output channels. We address this issue by using three different techniques to investigate how a sensory region, the insula cortex (IC), connects with the motor, physiological and autonomic output centers of the CeA. We uncover a strong connection between the IC and the lateral hypothalamus (LH) with a monosynaptic relay in the CeA and shed new light on the previously described functions of IC and CeA through direct projections to the LH.


Assuntos
Núcleo Central da Amígdala/fisiologia , Córtex Cerebral/fisiologia , Animais , Axônios/fisiologia , Fenômenos Eletrofisiológicos , Região Hipotalâmica Lateral/fisiologia , Técnicas In Vitro , Interneurônios/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Vias Neurais/fisiologia , Optogenética , Núcleos Septais/fisiologia
5.
Development ; 144(24): 4604-4615, 2017 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-29061639

RESUMO

The low-density lipoprotein receptor-related protein 4 (LRP4) is essential in muscle fibers for the establishment of the neuromuscular junction. Here, we show that LRP4 is also expressed by embryonic cortical and hippocampal neurons, and that downregulation of LRP4 in these neurons causes a reduction in density of synapses and number of primary dendrites. Accordingly, overexpression of LRP4 in cultured neurons had the opposite effect inducing more but shorter primary dendrites with an increased number of spines. Transsynaptic tracing mediated by rabies virus revealed a reduced number of neurons presynaptic to the cortical neurons in which LRP4 was knocked down. Moreover, neuron-specific knockdown of LRP4 by in utero electroporation of LRP4 miRNA in vivo also resulted in neurons with fewer primary dendrites and a lower density of spines in the developing cortex and hippocampus. Collectively, our results demonstrate an essential and novel role of neuronal LRP4 in dendritic development and synaptogenesis in the CNS.


Assuntos
Córtex Cerebral/metabolismo , Dendritos/metabolismo , Hipocampo/metabolismo , Receptores de LDL/metabolismo , Sinapses/metabolismo , Animais , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/embriologia , Técnicas de Inativação de Genes , Hipocampo/citologia , Hipocampo/embriologia , Proteínas Relacionadas a Receptor de LDL , Camundongos , Camundongos Endogâmicos C57BL , Raiva/patologia , Vírus da Raiva/crescimento & desenvolvimento , Receptores de LDL/genética
6.
J Neurosci ; 37(43): 10358-10371, 2017 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-28951448

RESUMO

Glycoprotein-deleted rabies virus-mediated monosynaptic tracing has become a standard method for neuronal circuit mapping, and is applied to virtually all parts of the rodent nervous system, including the spinal cord and primary sensory neurons. Here we identified two classes of unmyelinated sensory neurons (nonpeptidergic and C-fiber low-threshold mechanoreceptor neurons) resistant to direct and trans-synaptic infection from the spinal cord with rabies viruses that carry glycoproteins in their envelopes and that are routinely used for infection of CNS neurons (SAD-G and N2C-G). However, the same neurons were susceptible to infection with EnvA-pseudotyped rabies virus in tumor virus A receptor transgenic mice, indicating that resistance to retrograde infection was due to impaired virus adsorption rather than to deficits in subsequent steps of infection. These results demonstrate an important limitation of rabies virus-based retrograde tracing of sensory neurons in adult mice, and may help to better understand the molecular machinery required for rabies virus spread in the nervous system. In this study, mice of both sexes were used.SIGNIFICANCE STATEMENT To understand the neuronal bases of behavior, it is important to identify the underlying neural circuitry. Rabies virus-based monosynaptic tracing has been used to identify neuronal circuits in various parts of the nervous system. This has included connections between peripheral sensory neurons and their spinal targets. These connections form the first synapse in the somatosensory pathway. Here we demonstrate that two classes of unmyelinated sensory neurons, which account for >40% of dorsal root ganglia neurons, display resistance to rabies infection. Our results are therefore critical for interpreting monosynaptic rabies-based tracing in the sensory system. In addition, identification of rabies-resistant neurons might provide a means for future studies addressing rabies pathobiology.


Assuntos
Gânglios Espinais/química , Rede Nervosa/química , Técnicas de Rastreamento Neuroanatômico/métodos , Vírus da Raiva , Células Receptoras Sensoriais/química , Animais , Feminino , Gânglios Espinais/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Rede Nervosa/citologia , Células do Corno Posterior/química
8.
J Virol ; 90(4): 2031-8, 2016 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-26656686

RESUMO

UNLABELLED: Interferon beta (IFN-ß) is a key component of cellular innate immunity in mammals, and it constitutes the first line of defense during viral infection. Studies with cultured cells previously showed that almost all nucleated cells are able to produce IFN-ß to various extents, but information about the in vivo sources of IFN-ß remains incomplete. By applying immunohistochemistry and employing conditional-reporter mice that express firefly luciferase under the control of the IFN-ß promoter in either all or only distinct cell types, we found that astrocytes are the main producers of IFN-ß after infection of the brain with diverse neurotropic viruses, including rabies virus, Theiler's murine encephalomyelitis virus, and vesicular stomatitis virus. Analysis of a panel of knockout mouse strains revealed that sensing of viral components via both RIG-I-like helicases and Toll-like receptors contributes to IFN induction in the infected brain. A genetic approach to permanently mark rabies virus-infected cells in the brain showed that a substantial number of astrocytes became labeled and, therefore, must have been infected by the virus at least transiently. Thus, our results strongly indicate that abortive viral infection of astrocytes can trigger pattern recognition receptor signaling events which result in secretion of IFN-ß that confers antiviral protection. IMPORTANCE: Previous work indicated that astrocytes are the main producers of IFN after viral infection of the central nervous system (CNS), but it remained unclear how astrocytes might sense those viruses which preferentially replicate in neurons. We have now shown that virus sensing by both RIG-I-like helicases and Toll-like receptors is involved. Our results further demonstrate that astrocytes get infected in a nonproductive manner under these conditions, indicating that abortive infection of astrocytes plays a previously unappreciated role in the innate antiviral defenses of the CNS.


Assuntos
Astrócitos/imunologia , Encéfalo/imunologia , Encéfalo/virologia , Interferon beta/metabolismo , Vírus da Raiva/imunologia , Theilovirus/imunologia , Vesiculovirus/imunologia , Animais , Fusão Gênica Artificial , Astrócitos/virologia , Perfilação da Expressão Gênica , Genes Reporter , Imuno-Histoquímica , Luciferases/análise , Luciferases/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Imunológicos/metabolismo , Transdução de Sinais
9.
PLoS Pathog ; 10(4): e1004081, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24743923

RESUMO

RIG-I-like receptors (RLRs: RIG-I, MDA5 and LGP2) play a major role in the innate immune response against viral infections and detect patterns on viral RNA molecules that are typically absent from host RNA. Upon RNA binding, RLRs trigger a complex downstream signaling cascade resulting in the expression of type I interferons and proinflammatory cytokines. In the past decade extensive efforts were made to elucidate the nature of putative RLR ligands. In vitro and transfection studies identified 5'-triphosphate containing blunt-ended double-strand RNAs as potent RIG-I inducers and these findings were confirmed by next-generation sequencing of RIG-I associated RNAs from virus-infected cells. The nature of RNA ligands of MDA5 is less clear. Several studies suggest that double-stranded RNAs are the preferred agonists for the protein. However, the exact nature of physiological MDA5 ligands from virus-infected cells needs to be elucidated. In this work, we combine a crosslinking technique with next-generation sequencing in order to shed light on MDA5-associated RNAs from human cells infected with measles virus. Our findings suggest that RIG-I and MDA5 associate with AU-rich RNA species originating from the mRNA of the measles virus L gene. Corresponding sequences are poorer activators of ATP-hydrolysis by MDA5 in vitro, suggesting that they result in more stable MDA5 filaments. These data provide a possible model of how AU-rich sequences could activate type I interferon signaling.


Assuntos
RNA Helicases DEAD-box/metabolismo , Vírus do Sarampo/metabolismo , Sarampo/metabolismo , RNA Mensageiro/metabolismo , RNA Viral/metabolismo , Proteínas Virais/biossíntese , Linhagem Celular Tumoral , Proteína DEAD-box 58 , RNA Helicases DEAD-box/genética , Células HEK293 , Humanos , Helicase IFIH1 Induzida por Interferon , Sarampo/genética , Vírus do Sarampo/genética , RNA Mensageiro/genética , RNA Viral/genética , Receptores Imunológicos , Proteínas Virais/genética
10.
Proc Natl Acad Sci U S A ; 110(12): E1152-61, 2013 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-23487772

RESUMO

Identifying the connectome of adult-generated neurons is essential for understanding how the preexisting circuitry is refined by neurogenesis. Changes in the pattern of connectivity are likely to control the differentiation process of newly generated neurons and exert an important influence on their unique capacity to contribute to information processing. Using a monosynaptic rabies virus-based tracing technique, we studied the evolving presynaptic connectivity of adult-generated neurons in the dentate gyrus (DG) of the hippocampus and olfactory bulb (OB) during the first weeks of their life. In both neurogenic zones, adult-generated neurons first receive local connections from multiple types of GABAergic interneurons before long-range projections become established, such as those originating from cortical areas. Interestingly, despite fundamental similarities in the overall pattern of evolution of presynaptic connectivity, there were notable differences with regard to the development of cortical projections: although DG granule neuron input originating from the entorhinal cortex could be traced starting only from 3 to 5 wk on, newly generated neurons in the OB received input from the anterior olfactory nucleus and piriform cortex already by the second week. This early glutamatergic input onto newly generated interneurons in the OB was matched in time by the equally early innervations of DG granule neurons by glutamatergic mossy cells. The development of connectivity revealed by our study may suggest common principles for incorporating newly generated neurons into a preexisting circuit.


Assuntos
Giro Denteado/fisiologia , Neurônios/metabolismo , Bulbo Olfatório/fisiologia , Sinapses/metabolismo , Animais , Giro Denteado/citologia , Camundongos , Camundongos Transgênicos , Neurônios/citologia , Bulbo Olfatório/citologia , Vírus da Raiva
11.
Angew Chem Int Ed Engl ; 54(6): 1946-9, 2015 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-25522332

RESUMO

We report the development of dendritic siRNA nanostructures that are able to penetrate even difficult to transfect cells such as neurons with the help of a special receptor ligand. The nanoparticles elicit strong siRNA responses, despite the dendritic structure. An siRNA dendrimer directed against the crucial rabies virus (RABV) nucleoprotein (N protein) and phosphoprotein (P protein) allowed the suppression of the virus titer in neurons below the detection limit. The cell-penetrating siRNA dendrimers, which were assembled using click chemistry, open up new avenues toward finding novel molecules able to cure this deadly disease.


Assuntos
Dendrímeros , Nanoestruturas , RNA Interferente Pequeno/química , Cromatografia Líquida de Alta Pressão , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
12.
Hum Mol Genet ; 21(12): 2793-806, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22434821

RESUMO

ATP-binding cassette transporter A3 (ABCA3) is a lipid transporter active in lung alveolar epithelial type II cells (ATII) and is essential for their function as surfactant-producing cells. ABCA3 mutational defects cause respiratory distress in newborns and interstitial lung disease (ILD) in children. The molecular pathomechanisms are largely unknown; however, viral infections may initiate or aggravate ILDs. Here, we investigated the impact of the clinically relevant ABCA3 mutations, p.Q215K and p.E292V, by stable transfection of A549 lung epithelial cells. ABCA3 mutations strongly impaired expression of the ATII differentiation marker SP-C and the key epithelial cell adhesion proteins E-cadherin and zonula occludens-1. Concurrently, cells expressing ABCA3 mutation acquired mesenchymal features as observed by increased expression of SNAI1, MMP-2 and TGF-ß1, and elevated phosphorylation of Src. Infection with respiratory syncytial virus (RSV), the most common viral respiratory pathogen in small children, potentiated the observed mutational effects on loss of epithelial and acquisition of mesenchymal characteristics. In addition, RSV infection of cells harboring ABCA3 mutations resulted in a morphologic shift to a mesenchymal phenotype. We conclude that ABCA3 mutations, potentiated by RSV infection, induce loss of epithelial cell differentiation in ATII. Loss of key epithelial features may disturb the integrity of the alveolar epithelium, thereby comprising its functionality. We suggest the impairment of epithelial function as a mechanism by which ABCA3 mutations cause ILD.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Diferenciação Celular/genética , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Mutação , Vírus Sinciciais Respiratórios/fisiologia , Transportadores de Cassetes de Ligação de ATP/metabolismo , Western Blotting , Caderinas/genética , Caderinas/metabolismo , Linhagem Celular Tumoral , Criança , Células Epiteliais/patologia , Expressão Gênica , Interações Hospedeiro-Patógeno , Humanos , Recém-Nascido , Pulmão/metabolismo , Pulmão/patologia , Pulmão/virologia , Doenças Pulmonares Intersticiais/genética , Doenças Pulmonares Intersticiais/virologia , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Mesoderma/metabolismo , Mesoderma/patologia , Microscopia de Fluorescência , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/patologia , Alvéolos Pulmonares/virologia , Proteína C Associada a Surfactante Pulmonar/genética , Proteína C Associada a Surfactante Pulmonar/metabolismo , Síndrome do Desconforto Respiratório do Recém-Nascido/genética , Síndrome do Desconforto Respiratório do Recém-Nascido/virologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição da Família Snail , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo , Proteína da Zônula de Oclusão-1
13.
Cell Microbiol ; 15(2): 200-12, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23046288

RESUMO

Lyssavirus matrix proteins (M) support virus budding and have accessory functions that may contribute to host cell manipulation and adaptation to specific hosts. Here, we show that rabies virus (RABV) and European Bat Lyssavirus Type 1 (EBLV-1) M proteins differ in targeting and accumulation at cellular membranes. In contrast to RABV M, EBLV-1 M expressed from authentic EBLV-1 or chimeric RABV accumulated at the Golgi apparatus. Chimeric M proteins revealed that Golgi association depends on the integrity of the entire EBLV-1 M protein. Since RABV and EBLV-1 M differ in the use of cellular membranes for particle formation, differential membrane targeting and transport of M might determine the site of virus production. Moreover, both RABV and EBLV-1 M were for the first time detected within the nucleus and in Negri body-like inclusions bodies. Whereas nuclear M may imply hitherto unknown functions of lyssavirus M in host cell manipulation, the presence of M in inclusion bodies may correlate with regulatory functions of M in virus RNA synthesis. The data strongly support a model in which targeting of lyssavirus M proteins to distinctintracellular sites is a key determinant of diverse features in lyssavirus replication, host adaptation and pathogenesis.


Assuntos
Membrana Celular/metabolismo , Corpos de Inclusão/metabolismo , Lyssavirus/fisiologia , Vírus da Raiva/fisiologia , Infecções por Rhabdoviridae/veterinária , Proteínas da Matriz Viral/genética , Vírion/fisiologia , Animais , Linhagem Celular , Membrana Celular/ultraestrutura , Membrana Celular/virologia , Núcleo Celular/metabolismo , Núcleo Celular/ultraestrutura , Núcleo Celular/virologia , Quirópteros/virologia , Cricetinae , Complexo de Golgi/metabolismo , Complexo de Golgi/ultraestrutura , Complexo de Golgi/virologia , Corpos de Inclusão/ultraestrutura , Corpos de Inclusão/virologia , Microscopia Eletrônica , Transporte Proteico , Infecções por Rhabdoviridae/virologia , Especificidade da Espécie , Transfecção , Proteínas da Matriz Viral/metabolismo
14.
Cell Microbiol ; 15(2): 315-34, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23279019

RESUMO

The small matrix protein Z of arenaviruses has been identified as the main driving force to promote viral particle production at the plasma membrane. Although multiple functions of Z in the arenaviral life cycle have been uncovered, the mechanism of intracellular transport of Z to the site of virus budding is poorly understood and cellular motor proteins that mediate Z trafficking remain to be identified. In the present study, we report that the Z protein of the Old World arenavirus Lassa virus (LASV) interacts with the kinesin family member 13A (KIF13A), a plus-end-directed microtubule-dependent motor protein. Plasmid-driven overexpression of KIF13A results in relocalization of Z to the cell periphery, while functional blockage of endogenous KIF13A by overexpression of a dominant-negative mutant or KIF13A-specific siRNA causes a perinuclearaccumulation and decreased production of both Z-induced virus-like particles and infectious LASV. The interaction of KIF13A with Z proteins from both Old and New World arenaviruses suggests a conserved intracellular transport mechanism. In contrast, the intracellular distribution of the matrix proteins of prototypic members of the paramyxo- and rhabdovirus family is independent of KIF13A. In summary, our studies identify for the first time a molecular motor protein as a critical mediator for intracellular microtubule-dependent transport of arenavirus matrix proteins.


Assuntos
Proteínas de Transporte/metabolismo , Cinesinas/metabolismo , Vírus Lassa/fisiologia , Microtúbulos/metabolismo , Proteínas da Matriz Viral/metabolismo , Liberação de Vírus/fisiologia , Animais , Proteínas de Transporte/genética , Linhagem Celular , Membrana Celular/metabolismo , Membrana Celular/virologia , Chlorocebus aethiops , Expressão Gênica , Interações Hospedeiro-Patógeno , Humanos , Rim/patologia , Rim/virologia , Cinesinas/antagonistas & inibidores , Cinesinas/genética , Fígado/patologia , Fígado/virologia , Microtúbulos/virologia , Ligação Proteica , Transporte Proteico , RNA Interferente Pequeno/genética , Proteínas de Ligação a RNA , Células Vero , Proteínas da Matriz Viral/genética
15.
J Virol ; 86(2): 796-805, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22072748

RESUMO

Transcriptional induction of beta interferon (IFN-ß) through pattern recognition receptors is a key event in the host defense against invading viruses. Infection of cells by paramyxoviruses, like measles virus (MV) (genus Morbillivirus), is sensed predominantly by the ubiquitous cytoplasmic helicase RIG-I, recognizing viral 5'-triphosphate RNAs, and to some degree by MDA5. While MDA5 activation is effectively prevented by the MV V protein, the viral mechanisms for inhibition of MDA5-independent induction of IFN-ß remained obscure. Here, we identify the 186-amino-acid MV C protein, which shuttles between the nucleus and the cytoplasm, as a major viral inhibitor of IFN-ß transcription in human cells. Activation of the transcription factor IRF3 by upstream kinases and nuclear import of activated IRF3 were not affected in the presence of C protein, suggesting a nuclear target. Notably, C proteins of wild-type MV isolates, which are poor IFN-ß inducers, were found to comprise a canonical nuclear localization signal (NLS), whereas the NLSs of all vaccine strains, irrespective of their origins, were mutated. Site-directed mutagenesis of the C proteins from an MV wild-type isolate and from the vaccine virus strain Schwarz confirmed a correlation of nuclear localization and inhibition of IFN-ß transcription. A functional NLS and efficient nuclear accumulation are therefore critical for MV C to retain its potential to downregulate IFN-ß induction. We suggest that a defect in efficient nuclear import of C protein contributes to attenuation of MV vaccine strains.


Assuntos
Núcleo Celular/genética , Regulação para Baixo , Interferon beta/genética , Vírus do Sarampo/metabolismo , Sarampo/genética , Proteínas Virais/metabolismo , Sequência de Aminoácidos , Linhagem Celular , Núcleo Celular/metabolismo , Citoplasma/genética , Citoplasma/metabolismo , Humanos , Interferon beta/metabolismo , Sarampo/metabolismo , Sarampo/virologia , Vírus do Sarampo/química , Vírus do Sarampo/genética , Dados de Sequência Molecular , Transporte Proteico , Alinhamento de Sequência , Transcrição Gênica , Proteínas Virais/química , Proteínas Virais/genética
16.
Stem Cell Reports ; 18(12): 2418-2433, 2023 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-37995703

RESUMO

Although adult subependymal zone (SEZ) neural stem cells mostly generate GABAergic interneurons, a small progenitor population expresses the proneural gene Neurog2 and produces glutamatergic neurons. Here, we determined whether Neurog2 could respecify SEZ neural stem cells and their progeny toward a glutamatergic fate. Retrovirus-mediated expression of Neurog2 induced the glutamatergic lineage markers TBR2 and TBR1 in cultured SEZ progenitors, which differentiated into functional glutamatergic neurons. Likewise, Neurog2-transduced SEZ progenitors acquired glutamatergic neuron hallmarks in vivo. Intriguingly, they failed to migrate toward the olfactory bulb and instead differentiated within the SEZ or the adjacent striatum, where they received connections from local neurons, as indicated by rabies virus-mediated monosynaptic tracing. In contrast, lentivirus-mediated expression of Neurog2 failed to reprogram early SEZ neurons, which maintained GABAergic identity and migrated to the olfactory bulb. Our data show that NEUROG2 can program SEZ progenitors toward a glutamatergic identity but fails to reprogram their neuronal progeny.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos , Células-Tronco Neurais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Neurônios/metabolismo , Células-Tronco Neurais/metabolismo , Diferenciação Celular , Bulbo Olfatório/metabolismo , Neurogênese/fisiologia
17.
Neuron ; 111(15): 2367-2382.e6, 2023 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-37279750

RESUMO

The central nervous system regulates systemic immune responses by integrating the physiological and behavioral constraints faced by an individual. Corticosterone (CS), the release of which is controlled in the hypothalamus by the paraventricular nucleus (PVN), is a potent negative regulator of immune responses. Using the mouse model, we report that the parabrachial nucleus (PB), an important hub linking interoceptive afferent information to autonomic and behavioral responses, also integrates the pro-inflammatory cytokine IL-1ß signal to induce the CS response. A subpopulation of PB neurons, directly projecting to the PVN and receiving inputs from the vagal complex (VC), responds to IL-1ß to drive the CS response. Pharmacogenetic reactivation of these IL-1ß-activated PB neurons is sufficient to induce CS-mediated systemic immunosuppression. Our findings demonstrate an efficient brainstem-encoded modality for the central sensing of cytokines and the regulation of systemic immune responses.


Assuntos
Citocinas , Núcleos Parabraquiais , Animais , Camundongos , Corticosterona , Retroalimentação , Hipotálamo , Núcleo Hipotalâmico Paraventricular/fisiologia
18.
J Am Chem Soc ; 134(30): 12330-3, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22812910

RESUMO

Click chemistry of alkyne-modified RNA with different receptor ligand azides was used to prepare 3'-folate, 3'-cholesterol, and, as a new entity, 3'-anandamide-modified RNA in high yields and excellent purity. The anandamide-modified RNA shows surprisingly high transfection properties and enables the delivery of siRNA even into difficult-to-transfect RBL-2H3 cells which model neuronal uptake. Furthermore, the system was employed in human immune cells (BJAB), demonstrating silencing effects similar to those of a cationic, benchmark transfection reagent. In addition, the anandamide conjugates were found to be nontoxic. The reported chemistry and the described properties of the anandamide siRNA extend the possibilities of using siRNA-based gene silencing in neuronal and immune cells.


Assuntos
Ácidos Araquidônicos/química , Linfócitos B/metabolismo , Endocanabinoides/química , Inativação Gênica , Neurônios/metabolismo , Alcamidas Poli-Insaturadas/química , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/química , Animais , Sequência de Bases , Linhagem Celular , Colesterol/química , Química Click , Ácido Fólico/química , Células HeLa , Humanos , Luciferases/genética , RNA Interferente Pequeno/genética , Renilla/enzimologia
19.
J Virol ; 85(7): 3162-71, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21270162

RESUMO

Nuclear factor κB (NF-κB) transcription factors are involved in controlling numerous cellular processes, including inflammation, innate and adaptive immunity, and cell survival. Here we show that the immunosuppressive measles virus (MV; Morbillivirus genus, Paramyxoviridae) has evolved multiple functions to interfere with canonical NF-κB signaling in epithelial cells. The MV P, V, and C proteins, also involved in preventing host cell interferon responses, were found to individually suppress NF-κB-dependent reporter gene expression in response to activation of the tumor necrosis factor (TNF) receptor, RIG-I-like receptors, or Toll-like receptors. NF-κB activity was most efficiently suppressed in the presence of V, while expression of P or C resulted in moderate inhibition. As indicated by reporter gene assays involving overexpression of the IκB kinase (IKK) complex, which phosphorylates the inhibitor of κB to liberate NF-κB, V protein targets a downstream step in the signaling cascade. Coimmunoprecipitation experiments revealed that V specifically binds to the Rel homology domain of the NF-κB subunit p65 but not of p50. Notably, the short C-terminal domain of the V protein, which is also involved in binding STAT2, IRF7, and MDA5, was sufficient for the interaction and for preventing reporter gene activity. As observed by confocal microscopy, the presence of V abolished nuclear translocation of p65 upon TNF-α stimulation. Thus, MV V appears to prevent NF-κB-dependent gene expression by retaining p65 in the cytoplasm. These findings reveal NF-κB as a key target of MV and stress the importance of the V protein as the major viral immune-modulatory factor.


Assuntos
Evasão da Resposta Imune , Vírus do Sarampo/imunologia , Vírus do Sarampo/patogenicidade , NF-kappa B/antagonistas & inibidores , Fosfoproteínas/metabolismo , Fator de Transcrição RelA/metabolismo , Proteínas Virais/metabolismo , Linhagem Celular , Células Epiteliais/imunologia , Células Epiteliais/virologia , Genes Reporter , Hepatócitos/imunologia , Hepatócitos/virologia , Humanos , Imunoprecipitação , Ligação Proteica
20.
J Virol ; 85(2): 842-52, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21084487

RESUMO

The rabies virus (RV) phosphoprotein (P) is a type I interferon (IFN) antagonist preventing both transcriptional induction of IFN and IFN-mediated JAK/STAT signaling. In addition, P is an essential cofactor of the viral polymerase and is required for encapsidation of viral RNA into nucleoprotein during replication. By site-directed mutagenesis, we have identified a domain of P required for efficient inhibition of IFN induction. Phosphoproteins lacking amino acids (aa) 176 to 181, 182 to 186, or 176 to 186 were severely compromised in counteracting phosphorylation of IRF3 and IRF7 by TBK1 or IKKi while retaining the full capacity of preventing nuclear import of activated STATs and of supporting virus transcription and replication. Recombinant RV carrying the mutated phosphoproteins (the SAD ΔInd1, SAD ΔInd2, and SAD ΔInd1/2 viruses) activated IRF3 and beta IFN (IFN-ß) transcription in infected cells but still blocked STAT-mediated expression of IFN-stimulated genes. Due to a somewhat higher transcription rate, the SAD ΔInd1 virus activated IRF3 more efficiently than the SAD ΔInd2 virus. After intracerebral injection into mouse brains at high doses, the SAD ΔInd1 virus was completely apathogenic for wild-type (wt) mice, while the SAD ΔInd2 virus was partially attenuated and caused a slower progression of lethal rabies than wt RV. Neurovirulence of IFN-resistant RV thus correlates with the capacity of the virus to prevent activation of IRF3 and IRF7.


Assuntos
Fator Regulador 3 de Interferon/antagonistas & inibidores , Fator Regulador 7 de Interferon/antagonistas & inibidores , Interferon beta/antagonistas & inibidores , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Vírus da Raiva/imunologia , Vírus da Raiva/patogenicidade , Proteínas Estruturais Virais/genética , Proteínas Estruturais Virais/metabolismo , Animais , Encéfalo/patologia , Encéfalo/virologia , Linhagem Celular , Modelos Animais de Doenças , Feminino , Quinase I-kappa B , Masculino , Camundongos , Camundongos Transgênicos , Chaperonas Moleculares , Mutagênese Sítio-Dirigida , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases , Raiva/patologia , Raiva/virologia , Vírus da Raiva/genética , Análise de Sobrevida , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA