Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 11(1): 10400, 2021 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-34002008

RESUMO

The lateral hypothalamus (LH) is critically involved in the regulation of homeostatic energy balance. Some neurons in the LH express receptors for leptin (LepRb), a hormone known to increase energy expenditure and decrease energy intake. However, the neuroanatomical inputs to LepRb-expressing LH neurons remain unknown. We used rabies virus tracing technology to map these inputs, but encountered non-specific tracing. To optimize this technology for a minor cell population (LepRb is not ubiquitously expressed in LH), we used LepRb-Cre mice and assessed how different titers of the avian tumor virus receptor A (TVA) helper virus affected rabies tracing efficiency and specificity. We found that rabies expression is dependent on TVA receptor expression, and that leakiness of TVA receptors is dependent on the titer of TVA virus used. We concluded that a titer of 1.0-3.0 × 107 genomic copies per µl of the TVA virus is optimal for rabies tracing. Next, we successfully applied modified rabies virus tracing technology to map inputs to LepRb-expressing LH neurons. We discovered that other neurons in the LH itself, the periventricular hypothalamic nucleus (Pe), the posterior hypothalamic nucleus (PH), the bed nucleus of the stria terminalis (BNST), and the paraventricular hypothalamic nucleus (PVN) are the most prominent input areas to LepRb-expressing LH neurons.


Assuntos
Conectoma/métodos , Hipotálamo/diagnóstico por imagem , Imagem Molecular/métodos , Neurônios/metabolismo , Receptores para Leptina/análise , Animais , Proteínas Aviárias/genética , Feminino , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Vírus Auxiliares/genética , Hipotálamo/citologia , Hipotálamo/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Microscopia de Fluorescência , Vírus da Raiva/genética , Receptores para Leptina/metabolismo , Receptores Virais/genética , Núcleos Septais/citologia , Núcleos Septais/diagnóstico por imagem , Núcleos Septais/metabolismo , Técnicas Estereotáxicas
2.
Elife ; 92020 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-32940600

RESUMO

The insular cortex (IC) plays key roles in emotional and regulatory brain functions and is affected across psychiatric diseases. However, the brain-wide connections of the mouse IC have not been comprehensively mapped. Here, we traced the whole-brain inputs and outputs of the mouse IC across its rostro-caudal extent. We employed cell-type-specific monosynaptic rabies virus tracings to characterize afferent connections onto either excitatory or inhibitory IC neurons, and adeno-associated viral tracings to label excitatory efferent axons. While the connectivity between the IC and other cortical regions was highly bidirectional, the IC connectivity with subcortical structures was often unidirectional, revealing prominent cortical-to-subcortical or subcortical-to-cortical pathways. The posterior and medial IC exhibited resembling connectivity patterns, while the anterior IC connectivity was distinct, suggesting two major functional compartments. Our results provide insights into the anatomical architecture of the mouse IC and thus a structural basis to guide investigations into its complex functions.


Assuntos
Mapeamento Encefálico , Córtex Cerebral/anatomia & histologia , Camundongos/anatomia & histologia , Neurônios/citologia , Animais , Feminino , Masculino
3.
Neuron ; 104(5): 972-986.e6, 2019 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-31761708

RESUMO

How neural circuits develop in the human brain has remained almost impossible to study at the neuronal level. Here, we investigate human cortical neuron development, plasticity, and function using a mouse/human chimera model in which xenotransplanted human cortical pyramidal neurons integrate as single cells into the mouse cortex. Combined neuronal tracing, electrophysiology, and in vivo structural and functional imaging of the transplanted cells reveal a coordinated developmental roadmap recapitulating key milestones of human cortical neuron development. The human neurons display a prolonged developmental timeline, indicating the neuron-intrinsic retention of juvenile properties as an important component of human brain neoteny. Following maturation, human neurons in the visual cortex display tuned, decorrelated responses to visual stimuli, like mouse neurons, demonstrating their capacity for physiological synaptic integration in host cortical circuits. These findings provide new insights into human neuronal development and open novel avenues for the study of human neuronal function and disease. VIDEO ABSTRACT.


Assuntos
Neurogênese/fisiologia , Células Piramidais/citologia , Células Piramidais/fisiologia , Células Piramidais/transplante , Animais , Diferenciação Celular/fisiologia , Xenoenxertos , Humanos , Camundongos , Córtex Visual/citologia , Córtex Visual/fisiologia
4.
Cell Rep ; 27(7): 2092-2104.e10, 2019 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-31091448

RESUMO

Guanylate-binding protein (GBP) 5 is an interferon (IFN)-inducible cellular factor reducing HIV-1 infectivity by an incompletely understood mechanism. Here, we show that this activity is shared by GBP2, but not by other members of the human GBP family. GBP2/5 decrease the activity of the cellular proprotein convertase furin, which mediates conversion of the HIV-1 envelope protein (Env) precursor gp160 into mature gp120 and gp41. Because this process primes HIV-1 Env for membrane fusion, viral particles produced in the presence of GBP2/5 are poorly infectious due to increased incorporation of non-functional gp160. Furin activity is critical for the processing of envelope glycoproteins of many viral pathogens. Consistently, GBP2/5 also inhibit Zika, measles, and influenza A virus replication and decrease infectivity of viral particles carrying glycoproteins of Marburg and murine leukemia viruses. Collectively, our results show that GPB2/5 exert broad antiviral activity by suppressing the activity of the virus-dependency factor furin.


Assuntos
Furina/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Proteína gp120 do Envelope de HIV/metabolismo , Proteína gp41 do Envelope de HIV/metabolismo , HIV-1/metabolismo , Furina/genética , Proteínas de Ligação ao GTP/genética , Células HEK293 , Proteína gp120 do Envelope de HIV/genética , Proteína gp41 do Envelope de HIV/genética , HIV-1/genética , Humanos , Vírus da Influenza A/genética , Vírus da Influenza A/metabolismo , Vírus do Sarampo/genética , Vírus do Sarampo/metabolismo , Zika virus/genética , Zika virus/metabolismo
5.
Physiol Rep ; 6(14): e13807, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-30047252

RESUMO

The DMH is known to regulate brown adipose tissue (BAT) thermogenesis via projections to sympathetic premotor neurons in the raphe pallidus, but there is evidence that the periaqueductal gray (PAG) is also an important relay in the descending pathways regulating thermogenesis. The anatomical projections from the DMH to the PAG subdivisions and their function are largely elusive, and may differ per anterior-posterior level from bregma. We here aimed to investigate the anatomical projections from the DMH to the PAG along the entire anterior-posterior axis of the PAG, and to study the role of these projections in thermogenesis in Wistar rats. Anterograde channel rhodopsin viral tracing showed that the DMH projects especially to the dorsal and lateral PAG. Retrograde rabies viral tracing confirmed this, but also indicated that the PAG receives a diffuse input from the DMH and adjacent hypothalamic subregions. We aimed to study the role of the identified DMH to PAG projections in thermogenesis in conscious rats by specifically activating them using a combination of canine adenovirus-2 (CAV2Cre) and Cre-dependent designer receptor exclusively activated by designer drugs (DREADD) technology. Chemogenetic activation of DMH to PAG projections increased BAT temperature and core body temperature, but we cannot exclude the possibility that at least some thermogenic effects were mediated by adjacent hypothalamic subregions due to difficulties in specifically targeting the DMH and distinct subdivisions of the PAG because of diffuse virus expression. To conclude, our study shows the complexity of the anatomical and functional connection between the hypothalamus and the PAG, and some technical challenges in studying their connection.


Assuntos
Regulação da Temperatura Corporal , Hipotálamo Médio/anatomia & histologia , Substância Cinzenta Periaquedutal/anatomia & histologia , Animais , Hipotálamo Médio/fisiologia , Masculino , Vias Neurais/anatomia & histologia , Vias Neurais/fisiologia , Substância Cinzenta Periaquedutal/fisiologia , Ratos , Ratos Wistar
6.
Nat Biotechnol ; 36(1): 81-88, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29251729

RESUMO

Genetic engineering by viral infection of single cells is useful to study complex systems such as the brain. However, available methods for infecting single cells have drawbacks that limit their applications. Here we describe 'virus stamping', in which viruses are reversibly bound to a delivery vehicle-a functionalized glass pipette tip or magnetic nanoparticles in a pipette-that is brought into physical contact with the target cell on a surface or in tissue, using mechanical or magnetic forces. Different single cells in the same tissue can be infected with different viruses and an individual cell can be simultaneously infected with different viruses. We use rabies, lenti, herpes simplex, and adeno-associated viruses to drive expression of fluorescent markers or a calcium indicator in target cells in cell culture, mouse retina, human brain organoid, and the brains of live mice. Virus stamping provides a versatile solution for targeted single-cell infection of diverse cell types, both in vitro and in vivo.


Assuntos
Encéfalo/virologia , Nanopartículas de Magnetita/administração & dosagem , Análise de Célula Única/métodos , Vírus/genética , Animais , Engenharia Genética/tendências , Humanos , Nanopartículas de Magnetita/química , Camundongos , Organoides/metabolismo , Organoides/virologia , Retina/metabolismo , Retina/virologia , Distribuição Tecidual , Viroses/genética , Viroses/metabolismo , Replicação Viral/genética
7.
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.
Nat Commun ; 8: 15143, 2017 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-28489068

RESUMO

Consuming a calorically dense diet stimulates microglial reactivity in the mediobasal hypothalamus (MBH) in association with decreased number of appetite-curbing pro-opiomelanocortin (POMC) neurons; whether the reduction in POMC neuronal function is secondary to the microglial activation is unclear. Here we show that in hypercaloric diet-induced obese mice, persistently activated microglia in the MBH hypersecrete TNFα that in turn stimulate mitochondrial ATP production in POMC neurons, promoting mitochondrial fusion in their neurites, and increasing POMC neuronal firing rates and excitability. Specific disruption of the gene expressions of TNFα downstream signals TNFSF11A or NDUFAB1 in the MBH of diet-induced obese mice reverses mitochondrial elongation and reduces obesity. These data imply that in a hypercaloric environment, persistent elevation of microglial reactivity and consequent TNFα secretion induces mitochondrial stress in POMC neurons that contributes to the development of obesity.


Assuntos
Hipotálamo Médio/metabolismo , Microglia/metabolismo , Mitocôndrias/metabolismo , Neurônios/metabolismo , Obesidade/metabolismo , Pró-Opiomelanocortina , Estresse Fisiológico , Fator de Necrose Tumoral alfa/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Complexo I de Transporte de Elétrons/genética , Regulação da Expressão Gênica , Camundongos , Dinâmica Mitocondrial , Neuritos/metabolismo , Ligante RANK/genética , Transdução de Sinais
9.
Nat Commun ; 7: 13275, 2016 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-27848954

RESUMO

Oligodendrocyte damage is a central event in the pathogenesis of the common neuroinflammatory condition, multiple sclerosis (MS). Where and how oligodendrocyte damage is initiated in MS is not completely understood. Here, we use a combination of light and electron microscopy techniques to provide a dynamic and highly resolved view of oligodendrocyte damage in neuroinflammatory lesions. We show that both in MS and in its animal model structural damage is initiated at the myelin sheaths and only later spreads to the oligodendrocyte cell body. Early myelin damage itself is characterized by the formation of local myelin out-foldings-'myelinosomes'-, which are surrounded by phagocyte processes and promoted in their formation by anti-myelin antibodies and complement. The presence of myelinosomes in actively demyelinating MS lesions suggests that oligodendrocyte damage follows a similar pattern in the human disease, where targeting demyelination by therapeutic interventions remains a major open challenge.


Assuntos
Esclerose Múltipla/patologia , Bainha de Mielina/patologia , Oligodendroglia/patologia , Animais , Anticorpos/metabolismo , Proteínas do Sistema Complemento/metabolismo , Doenças Desmielinizantes/patologia , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/patologia , Humanos , Imageamento Tridimensional , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Bainha de Mielina/ultraestrutura , Oligodendroglia/ultraestrutura , Proteínas Opsonizantes/metabolismo , Organelas/metabolismo , Organelas/ultraestrutura
10.
Cold Spring Harb Protoc ; 2015(12): pdb.prot089409, 2015 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-26631127

RESUMO

This protocol describes the recovery of replication-competent rabies viruses (RV) such as SAD L16 or SAD L16-eGFP. It is suggested that at least three parallel transfection experiments are performed to increase the success rate (in three 3.5-cm(2) dishes or in three wells in a six-well plate). The entire protocol takes 10 d, and successful rescue can be obtained after 6 d. An additional procedure for the recovery of G-gene-deleted viruses is also included. Please note that appropriate biosafety measures are needed.


Assuntos
Deleção de Genes , Glicoproteínas/deficiência , Vírus da Raiva/genética , Vírus da Raiva/fisiologia , Genética Reversa/métodos , Proteínas do Envelope Viral/deficiência , Replicação Viral , Antígenos Virais/genética , DNA Complementar/genética , DNA Viral/genética , Glicoproteínas/genética , Proteínas do Envelope Viral/genética
11.
Cold Spring Harb Protoc ; 2015(12): pdb.prot089417, 2015 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-26631128

RESUMO

G-deleted fluorescent rabies virus (RV) pseudotyped with RV G proteins, SAD ΔG eGFP (RV CVS-G), can be used as single-round vectors for efficient retrograde labeling of neurons. For these experiments, as well as for monosynaptic tracing, which involves pseudotyping in situ, the use of the CVS strain G is recommended because of its high tropism for neurons. Pseudotype virus stocks generated by transfection of pCAGGS-G (or in MG139-on cells) contain the G protein of the vaccine strain SAD L16, which is broader in its tropism, and infects astrocytes, glia, and oligodendrocytes. We also describe a procedure for pseudotyping with ASLV Env A, which uses a cell-line expressing a version of the EnvA protein that is incorporated efficiently into the RV envelope (EnvARG(RGct)).


Assuntos
Antígenos Virais/metabolismo , Deleção de Genes , Glicoproteínas/metabolismo , Neurônios/virologia , Vírus da Raiva/genética , Sinapses/virologia , Proteínas do Envelope Viral/metabolismo , Linhagem Celular , Genes Reporter , Glicoproteínas/deficiência , Humanos , Proteínas Luminescentes/análise , Proteínas Luminescentes/genética , Neurônios/química , Imagem Óptica/métodos , Proteínas Recombinantes/análise , Proteínas Recombinantes/genética , Coloração e Rotulagem/métodos , Sinapses/química , Proteínas do Envelope Viral/deficiência
12.
Elife ; 42015 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-26609812

RESUMO

The cytosolic antiviral innate immune sensor RIG-I distinguishes 5' tri- or diphosphate containing viral double-stranded (ds) RNA from self-RNA by an incompletely understood mechanism that involves ATP hydrolysis by RIG-I's RNA translocase domain. Recently discovered mutations in ATPase motifs can lead to the multi-system disorder Singleton-Merten Syndrome (SMS) and increased interferon levels, suggesting misregulated signaling by RIG-I. Here we report that SMS mutations phenocopy a mutation that allows ATP binding but prevents hydrolysis. ATPase deficient RIG-I constitutively signals through endogenous RNA and co-purifies with self-RNA even from virus infected cells. Biochemical studies and cryo-electron microscopy identify a 60S ribosomal expansion segment as a dominant self-RNA that is stably bound by ATPase deficient RIG-I. ATP hydrolysis displaces wild-type RIG-I from this self-RNA but not from 5' triphosphate dsRNA. Our results indicate that ATP-hydrolysis prevents recognition of self-RNA and suggest that SMS mutations lead to unintentional signaling through prolonged RNA binding.


Assuntos
Trifosfato de Adenosina/metabolismo , RNA Helicases DEAD-box/metabolismo , RNA Viral/metabolismo , Linhagem Celular , Proteína DEAD-box 58 , Humanos , Hidrólise , Receptores Imunológicos , Especificidade por Substrato
13.
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
14.
Science ; 339(6120): 690-3, 2013 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-23328395

RESUMO

The retinoic acid-inducible gene I (RIG-I)-like receptor (RLR) melanoma differentiation-associated protein 5 (MDA5) senses cytoplasmic viral RNA and activates antiviral innate immunity. To reveal how paramyxoviruses counteract this response, we determined the crystal structure of the MDA5 adenosine 5'-triphosphate (ATP)-hydrolysis domain in complex with the viral inhibitor V protein. The V protein unfolded the ATP-hydrolysis domain of MDA5 via a ß-hairpin motif and recognized a structural motif of MDA5 that is normally buried in the conserved helicase fold. This leads to disruption of the MDA5 ATP-hydrolysis site and prevention of RNA-bound MDA5 filament formation. The structure explains why V proteins inactivate MDA5, but not RIG-I, and mutating only two amino acids in RIG-I induces robust V protein binding. Our results suggest an inhibition mechanism of RLR signalosome formation by unfolding of receptor and inhibitor.


Assuntos
RNA Helicases DEAD-box/química , RNA Helicases DEAD-box/metabolismo , Vírus da Parainfluenza 5 , RNA de Cadeia Dupla/metabolismo , Proteínas Virais/química , Proteínas Virais/metabolismo , Trifosfato de Adenosina/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Cristalografia por Raios X , Proteína DEAD-box 58 , RNA Helicases DEAD-box/genética , Células HEK293 , Humanos , Hidrólise , Imunidade Inata , Helicase IFIH1 Induzida por Interferon , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Vírus da Parainfluenza 5/imunologia , Ligação Proteica , Dobramento de Proteína , Estrutura Terciária de Proteína , Receptores Imunológicos , Transdução de Sinais , Sus scrofa , Proteínas Virais/genética
15.
Cell Rep ; 2(5): 1375-86, 2012 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-23142666

RESUMO

Cellular resolution three-dimensional (3D) visualization of defined, fluorescently labeled long-range neuronal networks in the uncut adult mouse brain has been elusive. Here, a virus-based strategy is described that allowed fluorescent labeling of centrifugally projecting neuronal populations in the ventral forebrain and their directly, monosynaptically connected bulbar interneurons upon a single stereotaxic injection into select neuronal populations. Implementation of improved tissue clearing combined with light-sheet fluorescence microscopy permitted imaging of the resulting connectivity maps in a single whole-brain scan. Subsequent 3D reconstructions revealed the exact distribution of the diverse neuronal ensembles monosynaptically connected with distinct bulbar interneuron populations. Moreover, rehydratation of brains after light-sheet fluorescence imaging enabled the immunohistochemical identification of synaptically connected neurons. Thus, this study describes a method for identifying monosynaptic connectivity maps from distinct, virally labeled neuronal populations that helps in better understanding of information flow in neural systems.


Assuntos
Encéfalo/metabolismo , Imageamento Tridimensional/métodos , Microscopia de Fluorescência/métodos , Rede Nervosa/anatomia & histologia , Animais , Encéfalo/anatomia & histologia , Dependovirus/genética , Dependovirus/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Interneurônios/patologia , Luz , Camundongos , Bulbo Olfatório/anatomia & histologia , Vírus da Raiva/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo
16.
Berl Munch Tierarztl Wochenschr ; 125(5-6): 219-27, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22712419

RESUMO

The glycoprotein G of lyssaviruses is the major determinant of virus pathogenicity and serves as a target for immunological responses to virus infections. However, assessment of the exact contribution of lyssavirus G proteins to observed differences in the pathogenicity of lyssavirus species is challenging, since the direct comparison of natural lyssaviruses does not allow specific ascription to individual virus proteins or domains. Here we describe the generation and characterization of recombinant rabies viruses (RABV) that express chimeric G proteins comprising of a RABV cytoplasma domain fused to transmembrane and ectodomain G sequences of a virulent RABV (challenge virus standard; CVS-11) or two European bat lyssaviruses (EBLV- and EBLV-2). These "envelope-switched" recombinant viruses were recovered from cDNAs. Similar growth kinetics and protein expression in neuroblastoma cell cultures and successful targeting of primary neurons showed that the chimeric G proteins were able to replace the authentic G protein in a RABV based virus vector. Inoculation of six week old CD-1 mice by the intracranial (i. c.) route of infection further demonstrated that all recombinant viruses were able to spread in the brain and to induce disease. The "envelope-switched" RABV therefore represent an important tool to further investigate the influence of lyssavirus ectodomains on virus tropism, and pathogenicity.


Assuntos
Glicoproteínas/genética , Lyssavirus/genética , Lyssavirus/metabolismo , Vírus da Raiva/genética , Vírus da Raiva/patogenicidade , Proteínas Virais/genética , Replicação Viral/genética , Animais , Antígenos Virais/metabolismo , Linhagem Celular , Células Cultivadas , Córtex Cerebral/virologia , Regulação da Expressão Gênica , Hipocampo/virologia , Imuno-Histoquímica , Camundongos , Neurônios/virologia , Nucleoproteínas/genética , Nucleoproteínas/metabolismo , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Infecções por Rhabdoviridae/mortalidade , Infecções por Rhabdoviridae/virologia
17.
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
18.
Eur J Cell Biol ; 91(1): 17-23, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21397980

RESUMO

The rabies virus (RV) phosphoprotein P is a multifunctional protein involved in viral RNA synthesis and in counteracting host innate immune responses. We have previously shown that RV P gene expression levels can be regulated by using picornavirus internal ribosome entry site (IRES) elements. Here we exploited a particular feature of the foot-and-mouth disease virus (FMDV) IRES, namely, preferential initiation at a downstream initiation codon, to address the role of N-terminally truncated RV phosphoproteins usually generated in RV-infected cells through ribosomal leaky scanning. Recombinant RVs in which P synthesis was directed by the poliovirus or FMDV IRES produced full-length P (P1) or a truncated form (P2), as the dominant product, respectively. While the P2 overexpressing virus showed attenuated growth in interferon-incompetent cells, it was superior to the P1 overexpressing virus in preventing expression of host interferon-stimulated genes. This indicates that in RV infected cells the availability of the truncated P2 protein is critical for viral resistance to interferon.


Assuntos
Engenharia Genética/métodos , Genoma Viral , Interferons/biossíntese , Fosfoproteínas/genética , Biossíntese de Proteínas , Isoformas de Proteínas/genética , Vírus da Raiva/genética , Raiva/virologia , Proteínas Estruturais Virais/genética , Animais , Linhagem Celular , Códon de Iniciação/genética , Cricetinae , RNA Polimerases Dirigidas por DNA/genética , Vírus da Febre Aftosa/química , Vírus da Febre Aftosa/genética , Genes Reporter , Interferons/imunologia , Luciferases/análise , Chaperonas Moleculares , Iniciação Traducional da Cadeia Peptídica/genética , Fosfoproteínas/química , Fosfoproteínas/imunologia , Fosfoproteínas/metabolismo , Plasmídeos , Poliovirus/química , Poliovirus/genética , Isoformas de Proteínas/química , Isoformas de Proteínas/imunologia , Isoformas de Proteínas/metabolismo , Raiva/imunologia , Vírus da Raiva/imunologia , Vírus da Raiva/metabolismo , Ribossomos/genética , Ribossomos/metabolismo , Transfecção , Proteínas Virais/genética , Proteínas Estruturais Virais/química , Proteínas Estruturais Virais/imunologia , Proteínas Estruturais Virais/metabolismo
19.
Adv Virus Res ; 79: 91-114, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21601044

RESUMO

Rabies is among the longest known and most dangerous and feared infectious diseases for humans and animals and still is responsible for tenth of thousands of human deaths per year. The rabies virus (RABV) is a rather atypical member of the Rhabdoviridae family as it has completely adapted during evolution to warm-blooded hosts and is directly transmitted between them, whereas most other rhabdoviruses are transmitted by insect vectors. The virus is also unique with respect to its extremely broad host species range and a very narrow host organ range, namely its strict neurotropism. It is becoming increasingly clear that the host innate immune system, particularly the type I interferon system, and the viral counteractions profoundly shape this virus-host relationship. In the past few years, exciting new insight was obtained on how viruses are sensed by innate immune receptors, how the downstream signaling networks for activation of interferon are working, and how viruses can interfere with the system. While RABV 5'-triphosphate RNAs were identified as the major pathogen-associated molecular pattern sensed by cytoplasmic RIG-I-like receptors (RLR), the RABV phosphoprotein (P) has emerged as a potent multifunctional antagonist able to counteract the signaling cascades leading to transcriptional activation of interferon genes as well as interferon signaling pathways, thereby limiting expression of antiviral and immune-stimulatory genes.


Assuntos
Interações Hospedeiro-Patógeno , Evasão da Resposta Imune , Interferons/antagonistas & inibidores , Interferons/imunologia , Vírus da Raiva/imunologia , Vírus da Raiva/patogenicidade , Animais , Humanos , Chaperonas Moleculares , Fosfoproteínas/imunologia , Fosfoproteínas/metabolismo , Proteínas Estruturais Virais/imunologia , Proteínas Estruturais Virais/metabolismo , Fatores de Virulência/imunologia , Fatores de Virulência/metabolismo
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA