Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2021 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-34462748

RESUMO

Hematopoiesis is finely regulated to enable timely production of the right numbers and types of mature immune cells to maintain tissue homeostasis. Dysregulated hematopoiesis may compromise antiviral immunity and/or exacerbate immunopathogenesis. Herein, we report an essential role of UBXN3B in maintenance of hematopoietic homeostasis and restriction of immunopathogenesis during respiratory viral infection. Ubxn3b deficient ( Ubxn3b -/- ) mice are highly vulnerable to SARS-CoV-2 and influenza A infection, characterized by more severe lung immunopathology, lower virus-specific IgG, significantly fewer B cells, but more myeloid cells than Ubxn3b +/+ littermates. This aberrant immune compartmentalization is recapitulated in uninfected Ubxn3b -/- mice. Mechanistically, UBXN3B controls precursor B-I (pre-BI) transition to pre-BII and subsequent proliferation in a cell-intrinsic manner, by maintaining BLNK protein stability and pre-BCR signaling. These results reveal an essential role of UBXN3B for the early stage of B cell development.

2.
PLoS Negl Trop Dis ; 15(6): e0009442, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34115766

RESUMO

Dengue virus (DENV) is a flavivirus that causes marked human morbidity and mortality worldwide, and is transmitted to humans by Aedes aegypti mosquitoes. Habitat expansion of Aedes, mainly due to climate change and increasing overlap between urban and wild habitats, places nearly half of the world's population at risk for DENV infection. After a bloodmeal from a DENV-infected host, the virus enters the mosquito midgut. Next, the virus migrates to, and replicates in, other tissues, like salivary glands. Successful viral transmission occurs when the infected mosquito takes another blood meal on a susceptible host and DENV is released from the salivary gland via saliva into the skin. During viral dissemination in the mosquito and transmission to a new mammalian host, DENV interacts with a variety of vector proteins, which are uniquely important during each phase of the viral cycle. Our study focuses on the interaction between DENV particles and protein components in the A. aegypti vector. We performed a mass spectrometry assay where we identified a set of A. aegypti salivary gland proteins which potentially interact with the DENV virion. Using dsRNA to silence gene expression, we analyzed the role of these proteins in viral infectivity. Two of these candidates, a synaptosomal-associated protein (AeSNAP) and a calcium transporter ATPase (ATPase) appear to play a role in viral replication both in vitro and in vivo, observing a ubiquitous expression of these proteins in the mosquito. These findings suggest that AeSNAP plays a protective role during DENV infection of mosquitoes and that ATPase protein is required for DENV during amplification within the vector.


Assuntos
Aedes/genética , Aedes/virologia , ATPases Transportadoras de Cálcio/metabolismo , Vírus da Dengue/fisiologia , Mosquitos Vetores/genética , Mosquitos Vetores/virologia , Animais , ATPases Transportadoras de Cálcio/genética , Linhagem Celular , Clonagem Molecular , Dengue/transmissão , Dengue/virologia , Regulação Enzimológica da Expressão Gênica , Técnicas de Silenciamento de Genes , Polimorfismo de Nucleotídeo Único , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Glândulas Salivares/virologia
3.
Viruses ; 12(11)2020 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-33147869

RESUMO

Emerging and re-emerging arthritogenic alphaviruses, such as Chikungunya virus (CHIKV) and O'nyong nyong virus, cause acute and chronic crippling arthralgia associated with inflammatory immune responses. Approximately 50% of CHIKV-infected patients suffer from rheumatic manifestations that last 6 months to years. However, the physiological functions of individual immune signaling pathways in the pathogenesis of alphaviral arthritis remain poorly understood. Here, we report that a deficiency in CXCL10, which is a chemoattractant for monocytes/macrophages/T cells, led to the same viremia as wild-type animals, but fewer immune infiltrates and lower viral loads in footpads at the peak of arthritic disease (6-8 days post infection). Macrophages constituted the largest immune cell population in footpads following infection, and were significantly reduced in Cxcl10-/- mice. The viral RNA loads in neutrophils and macrophages were reduced in Cxcl10-/- compared to wild-type mice. In summary, our results demonstrate that CXCL10 signaling promotes the pathogenesis of alphaviral disease and suggest that CXCL10 may be a therapeutic target for mitigating alphaviral arthritis.


Assuntos
Infecções por Alphavirus/imunologia , Alphavirus/patogenicidade , Artrite Infecciosa/imunologia , Quimiocina CXCL10/imunologia , Transdução de Sinais/imunologia , Alphavirus/genética , Infecções por Alphavirus/fisiopatologia , Animais , Artrite Infecciosa/virologia , Quimiocina CXCL10/genética , Modelos Animais de Doenças , Feminino , Macrófagos/imunologia , Macrófagos/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/imunologia , Neutrófilos/virologia , Carga Viral , Viremia/imunologia
4.
Viruses ; 12(5)2020 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-32397176

RESUMO

Mosquito-borne Zika virus (ZIKV) can cause congenital microcephaly and Guillain-Barré syndrome, among other symptoms. Specific treatments and vaccines for ZIKV are not currently available. To further understand the host factors that support ZIKV replication, we used mass spectrometry to characterize mammalian proteins that associate with the ZIKV NS1 protein and identified the TRiC/CCT complex as an interacting partner. Furthermore, the suppression of CCT2, one of the critical components of the TRiC/CCT complex, inhibited ZIKV replication in both mammalian cells and mosquitoes. These results highlight an important role for the TRiC/CCT complex in ZIKV infection, suggesting that the TRiC/CCT complex may be a promising therapeutic target.


Assuntos
Aedes/virologia , Chaperonina com TCP-1/metabolismo , Proteínas de Insetos/metabolismo , Mosquitos Vetores/virologia , Proteínas não Estruturais Virais/metabolismo , Replicação Viral , Infecção por Zika virus/virologia , Zika virus/fisiologia , Aedes/genética , Aedes/metabolismo , Animais , Chaperonina com TCP-1/genética , Feminino , Interações Hospedeiro-Patógeno , Humanos , Proteínas de Insetos/genética , Mosquitos Vetores/genética , Mosquitos Vetores/metabolismo , Ligação Proteica , Proteínas não Estruturais Virais/genética , Zika virus/genética , Infecção por Zika virus/genética , Infecção por Zika virus/metabolismo
5.
PLoS One ; 13(8): e0200385, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30089112

RESUMO

The innate immune system plays a critical role in the initial antiviral response. However, the timing and duration of these responses must be tightly regulated during infection to ensure appropriate immune cell activation and anti-viral defenses. Here we demonstrate that during antiviral response, a negative regulator miR-221 was also induced in an ELF4-dependent manner. We further show that ELF4 promotes miR-221 expression through direct binding to its promoter. Overexpression and knockdown assay show that miR-221 can negatively regulate IFNß production in time of virus infection. RNA-seq analysis of miR-221 overexpressed cells revealed multiple candidate targets. Taken together, our study identified a novel negative microRNA regulator of innate antiviral response, which is dependent on ELF4.


Assuntos
Imunidade Inata/genética , MicroRNAs/metabolismo , Infecções por Rhabdoviridae/imunologia , Animais , Antagomirs/metabolismo , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Regulação para Baixo , Células HEK293 , Herpesvirus Humano 1/fisiologia , Humanos , Interferon beta/genética , Interferon beta/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , MicroRNAs/antagonistas & inibidores , MicroRNAs/genética , Regiões Promotoras Genéticas , Proteínas Serina-Treonina Quinases/genética , Infecções por Rhabdoviridae/patologia , Infecções por Rhabdoviridae/virologia , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Vesiculovirus/genética , Vesiculovirus/imunologia
6.
Nat Commun ; 9(1): 2714, 2018 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-30006528

RESUMO

Plasmodium species produce an ortholog of the cytokine macrophage migration inhibitory factor, PMIF, which modulates the host inflammatory response to malaria. Using a novel RNA replicon-based vaccine, we show the impact of PMIF immunoneutralization on the host response and observed improved control of liver and blood-stage Plasmodium infection, and complete protection from re-infection. Vaccination against PMIF delayed blood-stage patency after sporozoite infection, reduced the expression of the Th1-associated inflammatory markers TNF-α, IL-12, and IFN-γ during blood-stage infection, augmented Tfh cell and germinal center responses, increased anti-Plasmodium antibody titers, and enhanced the differentiation of antigen-experienced memory CD4 T cells and liver-resident CD8 T cells. Protection from re-infection was recapitulated by the adoptive transfer of CD8 or CD4 T cells from PMIF RNA immunized hosts. Parasite MIF inhibition may be a useful approach to promote immunity to Plasmodium and potentially other parasite genera that produce MIF orthologous proteins.


Assuntos
Imunidade Adaptativa/efeitos dos fármacos , Anticorpos Antiprotozoários/biossíntese , Fatores Inibidores da Migração de Macrófagos/antagonistas & inibidores , Vacinas Antimaláricas/administração & dosagem , Malária/prevenção & controle , Proteínas de Protozoários/antagonistas & inibidores , Vacinas de DNA/administração & dosagem , Transferência Adotiva , Animais , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/parasitologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/parasitologia , Feminino , Expressão Gênica , Centro Germinativo/efeitos dos fármacos , Centro Germinativo/imunologia , Centro Germinativo/parasitologia , Memória Imunológica/efeitos dos fármacos , Interferon gama/genética , Interferon gama/imunologia , Interleucina-12/genética , Interleucina-12/imunologia , Fatores Inibidores da Migração de Macrófagos/genética , Fatores Inibidores da Migração de Macrófagos/imunologia , Malária/imunologia , Malária/parasitologia , Vacinas Antimaláricas/biossíntese , Camundongos , Camundongos Endogâmicos BALB C , Plasmodium berghei/efeitos dos fármacos , Plasmodium berghei/genética , Plasmodium berghei/imunologia , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , Proteínas de Protozoários/genética , Proteínas de Protozoários/imunologia , RNA de Protozoário/genética , RNA de Protozoário/imunologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia , Vacinas de DNA/biossíntese
7.
J Immunol ; 201(1): 193-201, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29760192

RESUMO

Circadian rhythms coordinate an organism's activities and biological processes to the optimal time in the 24-h daylight cycle. We previously demonstrated that male C57BL/6 mice develop sepsis more rapidly when the disease is induced in the nighttime versus the daytime. In this report, we elucidate the mechanism of this diurnal difference. Sepsis was induced via cecal ligation and puncture (CLP) at zeitgeber time (ZT)-19 (2 am) or ZT-7 (2 pm). Like the males used in our prior study, female C57BL/6 mice had a worse outcome when CLP was induced at ZT-19 versus ZT-7, and these effects persisted when we pooled the data from both sexes. In contrast, mice with a mutated Period 2 (Per2) gene had a similar outcome when CLP was induced at ZT-19 versus ZT-7. Bone marrow chimeras reconstituted with C57BL/6 immune cells exhibited a worse outcome when sepsis was induced at ZT-19 versus ZT-7, whereas chimeras with Per2-mutated immune cells did not. Next, murine macrophages were subjected to serum shock to synchronize circadian rhythms and exposed to bacteria cultured from the mouse cecum at 4-h intervals for 48 h. We observed that IL-6 production oscillated with a 24-h period in C57BL/6 cells exposed to cecal bacteria. Interestingly, we observed a similar pattern when cells were exposed to the TLR2 agonist lipoteichoic acid. Furthermore, TLR2-knockout mice exhibited a similar sepsis phenotype when CLP was induced at ZT-19 versus ZT-7. Together, these data suggest that circadian rhythms in immune cells mediate diurnal variations in murine sepsis severity via a TLR2-dependent mechanism.


Assuntos
Ritmo Circadiano/fisiologia , Macrófagos Peritoneais/imunologia , Sepse/imunologia , Sepse/patologia , Receptor 2 Toll-Like/metabolismo , Animais , Ceco/cirurgia , Feminino , Interleucina-6/biossíntese , Leucócitos/imunologia , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Circadianas Period/genética , Ácidos Teicoicos/farmacologia , Fatores de Tempo , Receptor 2 Toll-Like/agonistas
8.
Clin Infect Dis ; 66(7): 1133-1139, 2018 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-29228208

RESUMO

The cause of Lyme disease, Borrelia burgdorferi, was discovered in 1983. A 2-tiered testing protocol was established for serodiagnosis in 1994, involving an enzyme immunoassay (EIA) or indirect fluorescence antibody, followed (if reactive) by immunoglobulin M and immunoglobulin G Western immunoblots. These assays were prepared from whole-cell cultured B. burgdorferi, lacking key in vivo expressed antigens and expressing antigens that can bind non-Borrelia antibodies. Additional drawbacks, particular to the Western immunoblot component, include low sensitivity in early infection, technical complexity, and subjective interpretation when scored by visual examination. Nevertheless, 2-tiered testing with immunoblotting remains the benchmark for evaluation of new methods or approaches. Next-generation serologic assays, prepared with recombinant proteins or synthetic peptides, and alternative testing protocols, can now overcome or circumvent many of these past drawbacks. This article describes next-generation serodiagnostic testing for Lyme disease, focusing on methods that are currently available or near-at-hand.


Assuntos
Anticorpos Antibacterianos/sangue , Doença de Lyme/diagnóstico , Testes Sorológicos/métodos , Antígenos de Bactérias/imunologia , Proteínas de Bactérias/imunologia , Borrelia burgdorferi/imunologia , Ensaio de Imunoadsorção Enzimática , Europa (Continente) , Humanos , Técnicas Imunoenzimáticas , Imunoglobulina G/sangue , Imunoglobulina M/sangue , Proteínas Recombinantes , Sensibilidade e Especificidade , Testes Sorológicos/tendências , Estados Unidos
9.
Nature ; 546(7660): 667-670, 2017 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-28636595

RESUMO

Rotavirus, a leading cause of severe gastroenteritis and diarrhoea in young children, accounts for around 215,000 deaths annually worldwide. Rotavirus specifically infects the intestinal epithelial cells in the host small intestine and has evolved strategies to antagonize interferon and NF-κB signalling, raising the question as to whether other host factors participate in antiviral responses in intestinal mucosa. The mechanism by which enteric viruses are sensed and restricted in vivo, especially by NOD-like receptor (NLR) inflammasomes, is largely unknown. Here we uncover and mechanistically characterize the NLR Nlrp9b that is specifically expressed in intestinal epithelial cells and restricts rotavirus infection. Our data show that, via RNA helicase Dhx9, Nlrp9b recognizes short double-stranded RNA stretches and forms inflammasome complexes with the adaptor proteins Asc and caspase-1 to promote the maturation of interleukin (Il)-18 and gasdermin D (Gsdmd)-induced pyroptosis. Conditional depletion of Nlrp9b or other inflammasome components in the intestine in vivo resulted in enhanced susceptibility of mice to rotavirus replication. Our study highlights an important innate immune signalling pathway that functions in intestinal epithelial cells and may present useful targets in the modulation of host defences against viral pathogens.


Assuntos
Células Epiteliais/imunologia , Células Epiteliais/virologia , Inflamassomos/metabolismo , Intestinos/citologia , Receptores Acoplados a Proteínas G/metabolismo , Infecções por Rotavirus/imunologia , Infecções por Rotavirus/virologia , Rotavirus/imunologia , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Caspase 1/metabolismo , RNA Helicases DEAD-box/metabolismo , Células Epiteliais/metabolismo , Feminino , Imunidade Inata , Inflamassomos/química , Inflamassomos/genética , Interleucina-18/imunologia , Mucosa Intestinal/metabolismo , Intestinos/imunologia , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas de Ligação a Fosfato , Piroptose , RNA de Cadeia Dupla/metabolismo , Receptores Acoplados a Proteínas G/deficiência , Receptores Acoplados a Proteínas G/imunologia , Rotavirus/crescimento & desenvolvimento
10.
Cell Rep ; 19(3): 558-568, 2017 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-28423319

RESUMO

Tyro3, Axl, and Mertk (TAM) receptors are candidate entry receptors for infection with the Zika virus (ZIKV), an emerging flavivirus of global public health concern. To investigate the requirement of TAM receptors for ZIKV infection, we used several routes of viral inoculation and compared viral replication in wild-type versus Axl-/-, Mertk-/-, Axl-/-Mertk-/-, and Axl-/-Tyro3-/- mice in various organs. Pregnant and non-pregnant mice treated with interferon-α-receptor (IFNAR)-blocking (MAR1-5A3) antibody and infected subcutaneously with ZIKV showed no reliance on TAMs for infection. In the absence of IFNAR-blocking antibody, adult female mice challenged intravaginally with ZIKV showed no difference in mucosal viral titers. Similarly, in young mice that were infected with ZIKV intracranially or intraperitoneally, ZIKV replication occurred in the absence of TAM receptors, and no differences in cell tropism were observed. These findings indicate that, in mice, TAM receptors are not required for ZIKV entry and infection.


Assuntos
Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Infecção por Zika virus/metabolismo , Zika virus/fisiologia , Animais , Animais Recém-Nascidos , Feminino , Feto/virologia , Injeções Intraperitoneais , Camundongos , Placenta/virologia , Gravidez , Tropismo , Vagina/virologia , Replicação Viral , c-Mer Tirosina Quinase , Receptor Tirosina Quinase Axl
11.
PLoS Pathog ; 13(2): e1006187, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28152074

RESUMO

UBXN proteins likely participate in the global regulation of protein turnover, and we have shown that UBXN1 interferes with RIG-I-like receptor (RLR) signaling by interacting with MAVS and impeding its downstream effector functions. Here we demonstrate that over-expression of multiple UBXN family members decreased lentivirus and retrovirus production by several orders-of-magnitude in single cycle assays, at the level of long terminal repeat-driven transcription, and three family members, UBXN1, N9, and N11 blocked the canonical NFκB pathway by binding to Cullin1 (Cul1), inhibiting IκBα degradation. Multiple regions of UBXN1, including its UBA domain, were critical for its activity. Elimination of UBXN1 resulted in early murine embryonic lethality. shRNA-mediated knockdown of UBXN1 enhanced human immunodeficiency virus type 1 (HIV) production up to 10-fold in single cycle assays. In primary human fibroblasts, knockdown of UBXN1 caused prolonged degradation of IκBα and enhanced NFκB signaling, which was also observed after CRISPR-mediated knockout of UBXN1 in mouse embryo fibroblasts. Knockout of UBXN1 significantly up- and down-regulated hundreds of genes, notably those of several cell adhesion and immune signaling pathways. Reduction in UBXN1 gene expression in Jurkat T cells latently infected with HIV resulted in enhanced HIV gene expression, consistent with the role of UBXN1 in modulating the NFκB pathway. Based upon co-immunoprecipitation studies with host factors known to bind Cul1, models are presented as to how UBXN1 could be inhibiting Cul1 activity. The ability of UBXN1 and other family members to negatively regulate the NFκB pathway may be important for dampening the host immune response in disease processes and also re-activating quiescent HIV from latent viral reservoirs in chronically infected individuals.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/imunologia , Infecções por HIV/imunologia , Transdução de Sinais/imunologia , Animais , Técnicas de Inativação de Genes , HIV-1/imunologia , Humanos , Imunoprecipitação , Células Jurkat , Lentivirus/imunologia , Camundongos , Camundongos Knockout , Microscopia Confocal , Inibidor de NF-kappaB alfa/imunologia , NF-kappa B/imunologia , Retroviridae/imunologia
12.
Nat Commun ; 8: 14401, 2017 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-28195158

RESUMO

The insect immune deficiency (IMD) pathway resembles the tumour necrosis factor receptor network in mammals and senses diaminopimelic-type peptidoglycans present in Gram-negative bacteria. Whether unidentified chemical moieties activate the IMD signalling cascade remains unknown. Here, we show that infection-derived lipids 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphoglycerol (POPG) and 1-palmitoyl-2-oleoyl diacylglycerol (PODAG) stimulate the IMD pathway of ticks. The tick IMD network protects against colonization by three distinct bacteria, that is the Lyme disease spirochete Borrelia burgdorferi and the rickettsial agents Anaplasma phagocytophilum and A. marginale. Cell signalling ensues in the absence of transmembrane peptidoglycan recognition proteins and the adaptor molecules Fas-associated protein with a death domain (FADD) and IMD. Conversely, biochemical interactions occur between x-linked inhibitor of apoptosis protein (XIAP), an E3 ubiquitin ligase, and the E2 conjugating enzyme Bendless. We propose the existence of two functionally distinct IMD networks, one in insects and another in ticks.


Assuntos
Artrópodes/imunologia , Síndromes de Imunodeficiência/imunologia , Síndromes de Imunodeficiência/veterinária , Ixodes/imunologia , Lipídeos/efeitos adversos , Lipídeos/imunologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Anaplasma marginale/imunologia , Anaplasma marginale/patogenicidade , Anaplasma phagocytophilum/imunologia , Anaplasma phagocytophilum/patogenicidade , Animais , Artrópodes/metabolismo , Borrelia burgdorferi/imunologia , Borrelia burgdorferi/patogenicidade , Proteínas de Transporte , Modelos Animais de Doenças , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Escherichia coli/genética , Proteína de Domínio de Morte Associada a Fas , Inativação Gênica , Células HEK293 , Humanos , Ixodes/metabolismo , Doença de Lyme/imunologia , Fosfatidilgliceróis/imunologia , RNA Interferente Pequeno/metabolismo , Proteínas Recombinantes , Transdução de Sinais , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Enzimas de Conjugação de Ubiquitina/genética , Enzimas de Conjugação de Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/metabolismo
13.
J Virol ; 91(1)2017 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-27795421

RESUMO

CD8+ T cells are crucial components of immunity and play a vital role in recovery from West Nile virus (WNV) infection. Here, we identify a previously unrecognized function of interleukin-17A (IL-17A) in inducing cytotoxic-mediator gene expression and promoting CD8+ T cell cytotoxicity against WNV infection in mice. We find that IL-17A-deficient (Il17a-/-) mice are more susceptible to WNV infection and develop a higher viral burden than wild-type (WT) mice. Interestingly, the CD8+ T cells isolated from Il17a-/- mice are less cytotoxic and express lower levels of cytotoxic-mediator genes, which can be restored by supplying recombinant IL-17A in vitro and in vivo Importantly, treatment of WNV-infected mice with recombinant IL-17A, as late as day 6 postinfection, significantly reduces the viral burden and increases survival, suggesting a therapeutic potential for IL-17A. In conclusion, we report a novel function of IL-17A in promoting CD8+ T cell cytotoxicity, which may have broad implications in other microbial infections and cancers. IMPORTANCE: Interleukin-17A (IL-17A) and CD8+ T cells regulate diverse immune functions in microbial infections, malignancies, and autoimmune diseases. IL-17A is a proinflammatory cytokine produced by diverse cell types, while CD8+ T cells (known as cytotoxic T cells) are major cells that provide immunity against intracellular pathogens. Previous studies have demonstrated a crucial role of CD8+ T cells in recovery from West Nile virus (WNV) infection. However, the role of IL-17A during WNV infection remains unclear. Here, we demonstrate that IL-17A protects mice from lethal WNV infection by promoting CD8+ T cell-mediated clearance of WNV. In addition, treatment of WNV-infected mice with recombinant IL-17A reduces the viral burden and increases survival of mice, suggesting a potential therapeutic. This novel IL-17A-CD8+ T cell axis may also have broad implications for immunity to other microbial infections and cancers, where CD8+ T cell functions are crucial.


Assuntos
Citotoxicidade Imunológica/efeitos dos fármacos , Interleucina-17/farmacologia , Linfócitos T Citotóxicos/efeitos dos fármacos , Febre do Nilo Ocidental/tratamento farmacológico , Vírus do Nilo Ocidental/efeitos dos fármacos , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/imunologia , Encéfalo/virologia , Feminino , Expressão Gênica , Humanos , Interleucina-17/genética , Interleucina-17/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Neurônios/imunologia , Neurônios/virologia , Cultura Primária de Células , Receptores de Interleucina-17/genética , Receptores de Interleucina-17/imunologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/farmacologia , Análise de Sobrevida , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/virologia , Resultado do Tratamento , Carga Viral/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Febre do Nilo Ocidental/imunologia , Febre do Nilo Ocidental/mortalidade , Febre do Nilo Ocidental/virologia , Vírus do Nilo Ocidental/genética , Vírus do Nilo Ocidental/crescimento & desenvolvimento
14.
Am J Reprod Immunol ; 77(2)2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27966815

RESUMO

Recent studies have linked antenatal infection with Zika virus (ZIKV) with major adverse fetal and neonatal outcomes, including microcephaly. There is a growing consensus for the existence of a congenital Zika syndrome (CZS). Previous studies have indicated that non-placental macrophages play a key role in the replication of dengue virus (DENV), a closely related flavivirus. As the placenta provides the conduit for vertical transmission of certain viruses, and placental Hofbauer cells (HBCs) are fetal-placental macrophages located adjacent to fetal capillaries, it is not surprising that several recent studies have examined infection of HBCs by ZIKV. In this review, we describe congenital abnormalities associated with ZIKV infection, the role of HBCs in the placental response to infection, and evidence for the susceptibility of HBCs to ZIKV infection. We conclude that HBCs may contribute to the spread of ZIKV in placenta and promote vertical transmission of ZIKV, ultimately compromising fetal and neonatal development and function. Current evidence strongly suggests that further studies are warranted to dissect the specific molecular mechanism through which ZIKV infects HBCs and its potential impact on the development of CZS.


Assuntos
Anormalidades Congênitas/imunologia , Macrófagos/imunologia , Placenta/imunologia , Infecção por Zika virus/imunologia , Zika virus/fisiologia , Animais , Vírus da Dengue/fisiologia , Feminino , Desenvolvimento Fetal , Humanos , Transmissão Vertical de Doenças Infecciosas , Macrófagos/virologia , Microcirculação , Placenta/virologia , Gravidez , Replicação Viral , Infecção por Zika virus/transmissão
15.
Biochim Biophys Acta ; 1860(9): 1898-909, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27241849

RESUMO

BACKGROUND: Dengue virus (DENV) is a mosquito-borne flavivirus that causes significant human disease and mortality in the tropics and subtropics. By examining the effects of virus infection on gene expression, and interactions between virus and vector, new targets for prevention of infection and novel treatments may be identified in mosquitoes. We previously performed a microarray analysis of the Aedes aegypti transcriptome during infection with DENV and found that mosquito ubiquitin protein Ub3881 (AAEL003881) was specifically and highly down-regulated. Ubiquitin proteins have multiple functions in insects, including marking proteins for proteasomal degradation, regulating apoptosis and mediating innate immune signaling. METHODS: We used qRT-PCR to quantify gene expression and infection, and RNAi to reduce Ub3881 expression. Mosquitoes were infected with DENV through blood feeding. We transfected DENV protein expression constructs to examine the effect of Ub3881 on protein degradation. We used site-directed mutagenesis and transfection to determine what amino acids are involved in Ub3881-mediated protein degradation. Immunofluorescence, Co-immunoprecipitation and Western blotting were used to examine protein interactions and co-localization. RESULTS: The overexpression of Ub3881, but not related ubiquitin proteins, decreased DENV infection in mosquito cells and live Ae. aegypti. The Ub3881 protein was demonstrated to be involved in DENV envelope protein degradation and reduce the number of infectious virions released. CONCLUSIONS: We conclude that Ub3881 has several antiviral functions in the mosquito, including specific viral protein degradation. GENERAL SIGNIFICANCE: Our data highlights Ub3881 as a target for future DENV prevention strategies in the mosquito transmission vector.


Assuntos
Aedes/metabolismo , Vírus da Dengue/metabolismo , Dengue/metabolismo , Dengue/virologia , Ubiquitina/metabolismo , Proteínas do Envelope Viral/metabolismo , Vírion/metabolismo , Aedes/genética , Animais , Apoptose/genética , Linhagem Celular , Dengue/genética , Dengue/prevenção & controle , Vírus da Dengue/genética , Regulação para Baixo/genética , Expressão Gênica/genética , Imunidade Inata/genética , Imunoprecipitação/métodos , Insetos Vetores/genética , Insetos Vetores/metabolismo , Complexo de Endopeptidases do Proteassoma/genética , Transcriptoma/genética , Proteínas do Envelope Viral/genética , Vírion/genética
16.
PLoS Pathog ; 11(10): e1005202, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26491875

RESUMO

Dengue virus (DENV) is a mosquito-borne flavivirus that causes serious human disease and mortality worldwide. There is no specific antiviral therapy or vaccine for DENV infection. Alterations in gene expression during DENV infection of the mosquito and the impact of these changes on virus infection are important events to investigate in hopes of creating new treatments and vaccines. We previously identified 203 genes that were ≥5-fold differentially upregulated during flavivirus infection of the mosquito. Here, we examined the impact of silencing 100 of the most highly upregulated gene targets on DENV infection in its mosquito vector. We identified 20 genes that reduced DENV infection by at least 60% when silenced. We focused on one gene, a putative cysteine rich venom protein (SeqID AAEL000379; CRVP379), whose silencing significantly reduced DENV infection in Aedes aegypti cells. Here, we examine the requirement for CRVP379 during DENV infection of the mosquito and investigate the mechanisms surrounding this phenomenon. We also show that blocking CRVP379 protein with either RNAi or specific antisera inhibits DENV infection in Aedes aegypti. This work identifies a novel mosquito gene target for controlling DENV infection in mosquitoes that may also be used to develop broad preventative and therapeutic measures for multiple flaviviruses.


Assuntos
Aedes/virologia , Vírus da Dengue/patogenicidade , Dengue/metabolismo , Proteínas de Insetos/metabolismo , Insetos Vetores/virologia , Animais , Venenos de Artrópodes/metabolismo , Western Blotting , Cisteína , Ensaio de Imunoadsorção Enzimática , Imunofluorescência , Inativação Gênica , Insetos Vetores/metabolismo , Reação em Cadeia da Polimerase , RNA Interferente Pequeno , Transfecção
17.
Cell Microbiol ; 16(8): 1133-45, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24612118

RESUMO

Anaplasma phagocytophilum, which causes granulocytic anaplasmosis in humans and animals, is a tick-transmitted obligate intracellular bacterium that mediates its own uptake into neutrophils and non-phagocytic cells. Invasins of obligate intracellular pathogens are attractive targets for protecting against or curing infection because blocking the internalization step prevents survival of these organisms. The complement of A. phagocytophilum invasins is incompletely defined. Here, we report the significance of a novel A. phagocytophilum invasion protein, AipA. A. phagocytophilum induced aipA expression during transmission feeding of infected ticks on mice. The bacterium upregulated aipA transcription when it transitioned from its non-infectious reticulate cell morphotype to its infectious dense-cored morphotype during infection of HL-60 cells. AipA localized to the bacterial surface and was expressed during in vivo infection. Of the AipA regions predicted to be surface-exposed, only residues 1 to 87 (AipA1-87 ) were found to be essential for host cell invasion. Recombinant AipA1-87 protein bound to and competitively inhibited A. phagocytophilum infection of mammalian cells. Antiserum specific for AipA1-87 , but not other AipA regions, antagonized infection. Additional blocking experiments using peptide-specific antisera narrowed down the AipA invasion domain to residues 9 to 21. An antisera combination targeting AipA1-87 together with two other A. phagocytophilum invasins, OmpA and Asp14, nearly abolished infection of host cells. This study identifies AipA as an A. phagocytophilum surface protein that is critical for infection, demarcates its invasion domain, and establishes a rationale for targeting multiple invasins to protect against granulocytic anaplasmosis.


Assuntos
Adesinas Bacterianas/biossíntese , Anaplasma phagocytophilum/patogenicidade , Anaplasmose/microbiologia , Proteínas da Membrana Bacteriana Externa/biossíntese , Ehrlichiose/patologia , Adesinas Bacterianas/genética , Adesinas Bacterianas/imunologia , Anaplasma phagocytophilum/imunologia , Anaplasmose/imunologia , Anaplasmose/patologia , Animais , Anticorpos Monoclonais/imunologia , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/imunologia , Células CHO , Linhagem Celular Tumoral , Cricetulus , Ehrlichiose/imunologia , Ehrlichiose/microbiologia , Células HL-60 , Humanos , Soros Imunes/imunologia , Glicoproteínas de Membrana/imunologia , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana/biossíntese , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Camundongos , Carrapatos , Regulação para Cima
18.
PLoS One ; 8(8): e71388, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23940747

RESUMO

Research on syphilis, a sexually transmitted infection caused by the non-cultivatable spirochete Treponema pallidum, has been hampered by the lack of an inbred animal model. We hypothesized that Toll-like receptor (TLR)-dependent responses are essential for clearance of T. pallidum and, consequently, compared infection in wild-type (WT) mice and animals lacking MyD88, the adaptor molecule required for signaling by most TLRs. MyD88-deficient mice had significantly higher pathogen burdens and more extensive inflammation than control animals. Whereas tissue infiltrates in WT mice consisted of mixed mononuclear and plasma cells, infiltrates in MyD88-deficient animals were predominantly neutrophilic. Although both WT and MyD88-deficient mice produced antibodies that promoted uptake of treponemes by WT macrophages, MyD88-deficient macrophages were deficient in opsonophagocytosis of treponemes. Our results demonstrate that TLR-mediated responses are major contributors to the resistance of mice to syphilitic disease and that MyD88 signaling and FcR-mediated opsonophagocytosis are linked to the macrophage-mediated clearance of treponemes.


Assuntos
Síndromes de Imunodeficiência/imunologia , Síndromes de Imunodeficiência/patologia , Inflamação/genética , Fator 88 de Diferenciação Mieloide/genética , Sífilis/imunologia , Treponema pallidum/imunologia , Animais , Carga Bacteriana , Feminino , Síndromes de Imunodeficiência/genética , Inflamação/imunologia , Inflamação/patologia , Macrófagos/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fagocitose/genética , Fagocitose/imunologia , Doenças da Imunodeficiência Primária , Sífilis/genética , Sífilis/patologia , Treponema pallidum/crescimento & desenvolvimento
19.
Viruses ; 5(7): 1664-81, 2013 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-23881275

RESUMO

The West Nile virus (WNV) is an emerging infection of biodefense concern and there are no available treatments or vaccines. Here we used a high-throughput method based on a novel gene expression analysis, RNA-Seq, to give a global picture of differential gene expression by primary human macrophages of 10 healthy donors infected in vitro with WNV. From a total of 28 million reads per sample, we identified 1,514 transcripts that were differentially expressed after infection. Both predicted and novel gene changes were detected, as were gene isoforms, and while many of the genes were expressed by all donors, some were unique. Knock-down of genes not previously known to be associated with WNV resistance identified their critical role in control of viral infection. Our study distinguishes both common gene pathways as well as novel cellular responses. Such analyses will be valuable for translational studies of susceptible and resistant individuals--and for targeting therapeutics--in multiple biological settings.


Assuntos
Resistência à Doença/genética , Perfilação da Expressão Gênica , Macrófagos/imunologia , Macrófagos/virologia , RNA/biossíntese , RNA/genética , Vírus do Nilo Ocidental/imunologia , Adulto , Feminino , Técnicas de Silenciamento de Genes , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Adulto Jovem
20.
Cell Rep ; 3(4): 1057-70, 2013 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-23545497

RESUMO

RNA viruses are sensed by RIG-I-like receptors (RLRs), which signal through a mitochondria-associated adaptor molecule, MAVS, resulting in systemic antiviral immune responses. Although RLR signaling is essential for limiting RNA virus replication, it must be stringently controlled to prevent damage from inflammation. We demonstrate here that among all tested UBX-domain-containing protein family members, UBXN1 exhibits the strongest inhibitory effect on RNA-virus-induced type I interferon response. UBXN1 potently inhibits RLR- and MAVS-induced, but not TLR3-, TLR4-, or DNA-virus-induced innate immune responses. Depletion of UBXN1 enhances virus-induced innate immune responses, including those resulting from RNA viruses such as vesicular stomatitis, Sendai, West Nile, and dengue virus infection, repressing viral replication. Following viral infection, UBXN1 is induced, binds to MAVS, interferes with intracellular MAVS oligomerization, and disrupts the MAVS/TRAF3/TRAF6 signalosome. These findings underscore a critical role of UBXN1 in the modulation of a major antiviral signaling pathway.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Antivirais/farmacologia , Imunidade Inata/efeitos dos fármacos , Interferon Tipo I/farmacologia , Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/genética , Linhagem Celular Tumoral , Proteína DEAD-box 58 , RNA Helicases DEAD-box/metabolismo , Células HEK293 , Humanos , Ligação Proteica , Interferência de RNA , Vírus de RNA/imunologia , Vírus de RNA/fisiologia , RNA Interferente Pequeno/metabolismo , Receptores Imunológicos , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA