Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
JCI Insight ; 9(4)2024 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-38194268

RESUMO

Infection with chikungunya virus (CHIKV) causes disruption of draining lymph node (dLN) organization, including paracortical relocalization of B cells, loss of the B cell-T cell border, and lymphocyte depletion that is associated with infiltration of the LN with inflammatory myeloid cells. Here, we found that, during the first 24 hours of infection, CHIKV RNA accumulated in MARCO-expressing lymphatic endothelial cells (LECs) in both the floor and medullary LN sinuses. The accumulation of viral RNA in the LN was associated with a switch to an antiviral and inflammatory gene expression program across LN stromal cells, and this inflammatory response - including recruitment of myeloid cells to the LN - was accelerated by CHIKV-MARCO interactions. As CHIKV infection progressed, both floor and medullary LECs diminished in number, suggesting further functional impairment of the LN by infection. Consistent with this idea, antigen acquisition by LECs, a key function of LN LECs during infection and immunization, was reduced during pathogenic CHIKV infection.


Assuntos
Febre de Chikungunya , Vírus Chikungunya , Células Endoteliais/metabolismo , Imunização , Linfonodos , Animais
2.
mBio ; 15(2): e0299823, 2024 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-38170993

RESUMO

Cancers associated with the oncogenic gammaherpesviruses, Epstein-Barr virus and Kaposi sarcoma herpesvirus, are notable for their constitutive activation of the transcription factor signal transducer and activator of transcription 3 (STAT3). To better understand the role of STAT3 during gammaherpesvirus latency and the B cell response to infection, we used the model pathogen murine gammaherpesvirus 68 (MHV68). Genetic deletion of STAT3 in B cells of CD19cre/+Stat3f/f mice reduced peak MHV68 latency approximately sevenfold. However, infected CD19cre/+Stat3f/f mice exhibited disordered germinal centers and heightened virus-specific CD8 T cell responses compared to wild-type (WT) littermates. To circumvent the systemic immune alterations observed in the B cell-STAT3 knockout mice and more directly evaluate intrinsic roles for STAT3, we generated mixed bone marrow chimeric mice consisting of WT and STAT3 knockout B cells. We discovered a dramatic reduction in latency in STAT3 knockout B cells compared to their WT B cell counterparts in the same lymphoid organ. RNA sequencing of sorted germinal center B cells revealed that MHV68 infection shifts the gene signature toward proliferation and away from type I and type II IFN responses. Loss of STAT3 largely reversed the virus-driven transcriptional shift without impacting the viral gene expression program. STAT3 promoted B cell processes of the germinal center, including IL-21-stimulated downregulation of surface CD23 on B cells infected with MHV68 or EBV. Together, our data provide mechanistic insights into the role of STAT3 as a latency determinant in B cells for oncogenic gammaherpesviruses.IMPORTANCEThere are no directed therapies to the latency program of the human gammaherpesviruses, Epstein-Barr virus and Kaposi sarcoma herpesvirus. Activated host factor signal transducer and activator of transcription 3 (STAT3) is a hallmark of cancers caused by these viruses. We applied the murine gammaherpesvirus pathogen system to explore STAT3 function upon primary B cell infection in the host. Since STAT3 deletion in all CD19+ B cells of infected mice led to altered B and T cell responses, we generated chimeric mice with both normal and STAT3-deleted B cells. B cells lacking STAT3 failed to support virus latency compared to normal B cells from the same infected animal. Loss of STAT3 impaired B cell proliferation and differentiation and led to a striking upregulation of interferon-stimulated genes. These findings expand our understanding of STAT3-dependent processes that are key to its function as a pro-viral latency determinant for oncogenic gammaherpesviruses in B cells and may provide novel therapeutic targets.


Assuntos
Infecções por Vírus Epstein-Barr , Gammaherpesvirinae , Infecções por Herpesviridae , Herpesvirus Humano 8 , Rhadinovirus , Sarcoma de Kaposi , Animais , Humanos , Camundongos , Gammaherpesvirinae/genética , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/metabolismo , Herpesvirus Humano 8/metabolismo , Camundongos Endogâmicos C57BL , Rhadinovirus/genética , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Latência Viral/genética
3.
bioRxiv ; 2023 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-37873393

RESUMO

Infection with chikungunya virus (CHIKV) causes disruption of draining lymph node (dLN) organization, including paracortical relocalization of B cells, loss of the B cell-T cell border, and lymphocyte depletion that is associated with infiltration of the LN with inflammatory myeloid cells. Here, we find that during the first 24 h of infection, CHIKV RNA accumulates in MARCO-expressing lymphatic endothelial cells (LECs) in both the floor and medullary LN sinuses. The accumulation of viral RNA in the LN was associated with a switch to an antiviral and inflammatory gene expression program across LN stromal cells, and this inflammatory response, including recruitment of myeloid cells to the LN, was accelerated by CHIKV-MARCO interactions. As CHIKV infection progressed, both floor and medullary LECs diminished in number, suggesting further functional impairment of the LN by infection. Consistent with this idea, we find that antigen acquisition by LECs, a key function of LN LECs during infection and immunization, was reduced during pathogenic CHIKV infection.

4.
bioRxiv ; 2023 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-36993230

RESUMO

Cancers associated with the oncogenic gammaherpesviruses, Epstein-Barr virus and Kaposi sarcoma herpesvirus, are notable for their constitutive activation of the transcription factor STAT3. To better understand the role of STAT3 during gammaherpesvirus latency and immune control, we utilized murine gammaherpesvirus 68 (MHV68) infection. Genetic deletion of STAT3 in B cells of CD19cre/+Stat3f/f mice reduced peak latency approximately 7-fold. However, infected CD19cre/+Stat3f/f mice exhibited disordered germinal centers and heightened virus-specific CD8 T cell responses compared to WT littermates. To circumvent the systemic immune alterations observed in the B cell-STAT3 knockout mice and more directly evaluate intrinsic roles for STAT3, we generated mixed bone marrow chimeras consisting of WT and STAT3-knockout B cells. Using a competitive model of infection, we discovered a dramatic reduction in latency in STAT3-knockout B cells compared to their WT B cell counterparts in the same lymphoid organ. RNA sequencing of sorted germinal center B cells revealed that STAT3 promotes proliferation and B cell processes of the germinal center but does not directly regulate viral gene expression. Last, this analysis uncovered a STAT3-dependent role for dampening type I IFN responses in newly infected B cells. Together, our data provide mechanistic insight into the role of STAT3 as a latency determinant in B cells for oncogenic gammaherpesviruses.

5.
Cell Rep ; 42(2): 112126, 2023 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-36795561

RESUMO

To disseminate through the body, Zika virus (ZIKV) is thought to exploit the mobility of myeloid cells, in particular monocytes and dendritic cells. However, the timing and mechanisms underlying shuttling of the virus by immune cells remains unclear. To understand the early steps in ZIKV transit from the skin, at different time points, we spatially mapped ZIKV infection in lymph nodes (LNs), an intermediary site en route to the blood. Contrary to prevailing hypotheses, migratory immune cells are not required for the virus to reach the LNs or blood. Instead, ZIKV rapidly infects a subset of sessile CD169+ macrophages in the LNs, which release the virus to infect downstream LNs. Infection of CD169+ macrophages alone is sufficient to initiate viremia. Overall, our experiments indicate that macrophages that reside in the LNs contribute to initial ZIKV spread. These studies enhance our understanding of ZIKV dissemination and identify another anatomical site for potential antiviral intervention.


Assuntos
Infecção por Zika virus , Zika virus , Humanos , Macrófagos , Monócitos/patologia , Linfonodos/patologia
6.
iScience ; 25(12): 105510, 2022 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-36425759

RESUMO

The success of poxviruses as pathogens depends on their antagonism of host responses by multiple immunomodulatory proteins. The largest of these expressed by ectromelia virus (the agent of mousepox) is C15, one member of a well-conserved poxviral family previously shown to inhibit T cell activation. Here, we demonstrate by quantitative immunofluorescence imaging that C15 also limits contact between natural killer (NK) cells and infected cells in vivo. This corresponds to an inhibition in the number of total and degranulating NK cells, ex vivo and in vitro, with no detectable impact on NK cell cytokine production or the transcription of factors related to NK cell recruitment or activation. Thus, in addition to its previously identified capacity to antagonize CD4 T cell activation, C15 inhibits NK cell cytolytic function, which results in increased viral replication and dissemination in vivo. This work builds on a body of literature demonstrating the importance of early restriction of virus within the draining lymph node.

7.
JCI Insight ; 7(22)2022 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-36166305

RESUMO

Disseminated coccidioidomycosis (DCM) is caused by Coccidioides, pathogenic fungi endemic to the southwestern United States and Mexico. Illness occurs in approximately 30% of those infected, less than 1% of whom develop disseminated disease. To address why some individuals allow dissemination, we enrolled patients with DCM and performed whole-exome sequencing. In an exploratory set of 67 patients with DCM, 2 had haploinsufficient STAT3 mutations, and defects in ß-glucan sensing and response were seen in 34 of 67 cases. Damaging CLEC7A and PLCG2 variants were associated with impaired production of ß-glucan-stimulated TNF-α from PBMCs compared with healthy controls. Using ancestry-matched controls, damaging CLEC7A and PLCG2 variants were overrepresented in DCM, including CLEC7A Y238* and PLCG2 R268W. A validation cohort of 111 patients with DCM confirmed the PLCG2 R268W, CLEC7A I223S, and CLEC7A Y238* variants. Stimulation with a DECTIN-1 agonist induced DUOX1/DUOXA1-derived hydrogen peroxide [H2O2] in transfected cells. Heterozygous DUOX1 or DUOXA1 variants that impaired H2O2 production were overrepresented in discovery and validation cohorts. Patients with DCM have impaired ß-glucan sensing or response affecting TNF-α and H2O2 production. Impaired Coccidioides recognition and decreased cellular response are associated with disseminated coccidioidomycosis.


Assuntos
Coccidioidomicose , beta-Glucanas , Humanos , Fator de Necrose Tumoral alfa/genética , Peróxido de Hidrogênio , Coccidioidomicose/genética , Coccidioidomicose/epidemiologia , Coccidioidomicose/microbiologia , Coccidioides/genética
8.
EMBO J ; 40(22): e108966, 2021 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-34618370

RESUMO

Viremia in the vertebrate host is a major determinant of arboviral reservoir competency, transmission efficiency, and disease severity. However, immune mechanisms that control arboviral viremia are poorly defined. Here, we identify critical roles for the scavenger receptor MARCO in controlling viremia during arthritogenic alphavirus infections in mice. Following subcutaneous inoculation, arthritogenic alphavirus particles drain via the lymph and are rapidly captured by MARCO+ lymphatic endothelial cells (LECs) in the draining lymph node (dLN), limiting viral spread to the bloodstream. Upon reaching the bloodstream, alphavirus particles are cleared from the circulation by MARCO-expressing Kupffer cells in the liver, limiting viremia and further viral dissemination. MARCO-mediated accumulation of alphavirus particles in the draining lymph node and liver is an important host defense mechanism as viremia and viral tissue burdens are elevated in MARCO-/- mice and disease is more severe. In contrast to prior studies implicating a key role for lymph node macrophages in limiting viral dissemination, these findings exemplify a previously unrecognized arbovirus-scavenging role for lymphatic endothelial cells and improve our mechanistic understanding of viremia control during arthritogenic alphavirus infection.


Assuntos
Infecções por Alphavirus/virologia , Linfonodos/citologia , Receptores Imunológicos/metabolismo , Viremia/patologia , Alphavirus/patogenicidade , Animais , Febre de Chikungunya/genética , Febre de Chikungunya/virologia , Células Endoteliais/virologia , Interações Hospedeiro-Patógeno , Células de Kupffer/virologia , Linfonodos/virologia , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Camundongos Transgênicos , RNA Viral/metabolismo , Receptores Imunológicos/genética , Análise de Célula Única , Viremia/virologia
9.
Cell ; 184(5): 1214-1231.e16, 2021 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-33636133

RESUMO

Although enteric helminth infections modulate immunity to mucosal pathogens, their effects on systemic microbes remain less established. Here, we observe increased mortality in mice coinfected with the enteric helminth Heligmosomoides polygyrus bakeri (Hpb) and West Nile virus (WNV). This enhanced susceptibility is associated with altered gut morphology and transit, translocation of commensal bacteria, impaired WNV-specific T cell responses, and increased virus infection in the gastrointestinal tract and central nervous system. These outcomes were due to type 2 immune skewing, because coinfection in Stat6-/- mice rescues mortality, treatment of helminth-free WNV-infected mice with interleukin (IL)-4 mirrors coinfection, and IL-4 receptor signaling in intestinal epithelial cells mediates the susceptibility phenotypes. Moreover, tuft cell-deficient mice show improved outcomes with coinfection, whereas treatment of helminth-free mice with tuft cell-derived cytokine IL-25 or ligand succinate worsens WNV disease. Thus, helminth activation of tuft cell-IL-4-receptor circuits in the gut exacerbates infection and disease of a neurotropic flavivirus.


Assuntos
Coinfecção , Nematospiroides dubius/fisiologia , Transdução de Sinais , Infecções por Strongylida/patologia , Vírus do Nilo Ocidental/fisiologia , Animais , Linfócitos T CD8-Positivos/imunologia , Modelos Animais de Doenças , Suscetibilidade a Doenças , Mucosa Intestinal/parasitologia , Mucosa Intestinal/virologia , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/parasitologia , Neurônios/virologia , Receptores de Interleucina-4/metabolismo , Fator de Transcrição STAT6/genética , Índice de Gravidade de Doença , Infecções por Strongylida/parasitologia
10.
Immunity ; 54(2): 276-290.e5, 2021 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-33434494

RESUMO

The oropharyngeal mucosa serves as a perpetual pathogen entry point and a critical site for viral replication and spread. Here, we demonstrate that type 1 innate lymphoid cells (ILC1s) were the major immune force providing early protection during acute oral mucosal viral infection. Using intravital microscopy, we show that ILC1s populated and patrolled the uninfected labial mucosa. ILC1s produced interferon-γ (IFN-γ) in the absence of infection, leading to the upregulation of key antiviral genes, which were downregulated in uninfected animals upon genetic ablation of ILC1s or antibody-based neutralization of IFN-γ. Thus, tonic IFN-γ production generates increased oral mucosal viral resistance even before infection. Our results demonstrate barrier-tissue protection through tissue surveillance in the absence of rearranged-antigen receptors and the induction of an antiviral state during homeostasis. This aspect of ILC1 biology raises the possibility that these cells do not share true functional redundancy with other tissue-resident lymphocytes.


Assuntos
Interferon gama/metabolismo , Linfócitos/imunologia , Orofaringe/imunologia , Mucosa Respiratória/imunologia , Vírus Vaccinia/fisiologia , Vaccinia/imunologia , Animais , Células Cultivadas , Resistência à Doença , Humanos , Imunidade Inata , Interferon gama/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas com Domínio T/genética , Células Th1/imunologia
11.
Nat Immunol ; 22(1): 41-52, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33139915

RESUMO

Personalized cancer vaccines are a promising approach for inducing T cell immunity to tumor neoantigens. Using a self-assembling nanoparticle vaccine that links neoantigen peptides to a Toll-like receptor 7/8 agonist (SNP-7/8a), we show how the route and dose alter the magnitude and quality of neoantigen-specific CD8+ T cells. Intravenous vaccination (SNP-IV) induced a higher proportion of TCF1+PD-1+CD8+ T cells as compared to subcutaneous immunization (SNP-SC). Single-cell RNA sequencing showed that SNP-IV induced stem-like genes (Tcf7, Slamf6, Xcl1) whereas SNP-SC enriched for effector genes (Gzmb, Klrg1, Cx3cr1). Stem-like cells generated by SNP-IV proliferated and differentiated into effector cells upon checkpoint blockade, leading to superior antitumor response as compared to SNP-SC in a therapeutic model. The duration of antigen presentation by dendritic cells controlled the magnitude and quality of CD8+ T cells. These data demonstrate how to optimize antitumor immunity by modulating vaccine parameters for specific generation of effector or stem-like CD8+ T cells.


Assuntos
Antígenos de Neoplasias/imunologia , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/administração & dosagem , Fator 1-alfa Nuclear de Hepatócito/análise , Nanopartículas , Animais , Apresentação de Antígeno , Vacinas Anticâncer/imunologia , Células Dendríticas/imunologia , Feminino , Imunidade Inata , Camundongos , Camundongos Endogâmicos C57BL , Vacinação
12.
Cell Rep Med ; 1(3)2020 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-32699843

RESUMO

CD137 is a costimulatory receptor expressed on natural killer cells, T cells, and subsets of dendritic cells. An agonistic monoclonal antibody (mAb) against CD137 has been used to reduce tumor burden or reverse autoimmunity in animal models and clinical trials. Here, we show that mice treated with an agonistic anti-CD137 mAb have reduced numbers of germinal center (GC) B cells and follicular dendritic cells (FDCs) in lymphoid tissues, which impair antibody responses to multiple T-cell-dependent antigens, including infectious virus, viral proteins, and conjugated haptens. These effects are not due to enhanced apoptosis or impaired proliferation of B cells but instead correlate with changes in lymphoid follicle structure and GC B cell dispersal and are mediated by CD137 signaling in CD4+ and CD8+ T cells. Our experiments in mice suggest that agonistic anti-CD137 mAbs used in cancer and autoimmunity therapy may impair long-term antibody and B cell memory responses.


Assuntos
Anticorpos Monoclonais/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Células Dendríticas Foliculares/imunologia , Tecido Linfoide/imunologia , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia , Animais , Formação de Anticorpos/imunologia , Linfócitos B/imunologia , Linhagem Celular , Proliferação de Células/fisiologia , Centro Germinativo/imunologia , Células Matadoras Naturais/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias/imunologia
13.
PLoS Pathog ; 16(1): e1008292, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31999809

RESUMO

Humoral immune responses initiate in the lymph node draining the site of viral infection (dLN). Some viruses subvert LN B cell activation; however, our knowledge of viral hindrance of B cell responses of important human pathogens is lacking. Here, we define mechanisms whereby chikungunya virus (CHIKV), a mosquito-transmitted RNA virus that causes outbreaks of acute and chronic arthritis in humans, hinders dLN antiviral B cell responses. Infection of WT mice with pathogenic, but not acutely cleared CHIKV, induced MyD88-dependent recruitment of monocytes and neutrophils to the dLN. Blocking this influx improved lymphocyte accumulation, dLN organization, and CHIKV-specific B cell responses. Both inducible nitric oxide synthase (iNOS) and the phagocyte NADPH oxidase (Nox2) contributed to impaired dLN organization and function. Infiltrating monocytes expressed iNOS through a local IRF5- and IFNAR1-dependent pathway that was partially TLR7-dependent. Together, our data suggest that pathogenic CHIKV triggers the influx and activation of monocytes and neutrophils in the dLN that impairs virus-specific B cell responses.


Assuntos
Linfócitos B/imunologia , Febre de Chikungunya/imunologia , Fatores Reguladores de Interferon/imunologia , Monócitos/imunologia , Fator 88 de Diferenciação Mieloide/imunologia , NADPH Oxidase 2/imunologia , Neutrófilos/imunologia , Óxido Nítrico Sintase Tipo II/imunologia , Animais , Febre de Chikungunya/virologia , Vírus Chikungunya/fisiologia , Humanos , Fatores Reguladores de Interferon/genética , Linfonodos/imunologia , Linfonodos/virologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/genética , NADPH Oxidase 2/genética , Óxido Nítrico Sintase Tipo II/genética
14.
Methods Mol Biol ; 2023: 287-299, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31240685

RESUMO

This chapter provides methods for the propagation, purification, and titration of vaccinia virus (VACV) and the highly attenuated strain-modified vaccinia Ankara (MVA). Additionally, we provide information on VACV recombinants we have used for intravital imaging with multiphoton excitation.


Assuntos
Vírus Vaccinia/crescimento & desenvolvimento , Vírus Vaccinia/isolamento & purificação , Células Cultivadas , Humanos
15.
Methods Mol Biol ; 2023: 301-311, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31240686

RESUMO

Intravital multiphoton microscopy (MPM) allows the direct visualization of viral infections in real time as they occur in living animals. Here we describe the routes and considerations for murine infection with vaccinia virus (VACV) for imaging, and the preparation of the skin and inner lip (labial mucosa) of infected animals for MPM. Using different recombinant VACVs expressing fluorescent proteins in combination with transgenic fluorescent reporter mice, MPM imaging can be used to examine the movements, interactions, and functions of virus-infected cells or selected immune cell populations after infection.


Assuntos
Microscopia Intravital/métodos , Vírus Vaccinia/patogenicidade , Vaccinia/virologia , Animais , Imunidade nas Mucosas/fisiologia , Camundongos , Camundongos Transgênicos , Vaccinia/diagnóstico por imagem , Replicação Viral/fisiologia
16.
Nat Immunol ; 20(5): 602-612, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30886418

RESUMO

Despite intense interest in antiviral T cell priming, the routes by which virions move in lymph nodes (LNs) are imperfectly understood. Current models fail to explain how virus-infected cells rapidly appear within the LN interior after viral infection. To better understand virion trafficking in the LN, we determined the locations of virions and infected cells after administration to mice of vaccinia virus or Zika virus. Notably, many rapidly infected cells in the LN interior were adjacent to LN conduits. Through the use of confocal and electron microscopy, we clearly visualized virions within conduits. Functionally, CD8+ T cells rapidly and preferentially associated with vaccinia virus-infected cells in the LN paracortex, which led to T cell activation in the LN interior. These results reveal that it is possible for even large virions to flow through LN conduits and infect dendritic cells within the T cell zone to prime CD8+ T cells.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfonodos/imunologia , Ativação Linfocitária/imunologia , Vírion/imunologia , Animais , Linfócitos T CD8-Positivos/virologia , Feminino , Linfonodos/ultraestrutura , Linfonodos/virologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microscopia Confocal , Microscopia Eletrônica de Transmissão , Vírus Vaccinia/imunologia , Vírus Vaccinia/fisiologia , Vírion/fisiologia , Vírion/ultraestrutura , Viroses/imunologia , Viroses/virologia , Zika virus/imunologia , Zika virus/fisiologia
17.
Nat Microbiol ; 4(2): 216-225, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30420785

RESUMO

Cell motility is essential for viral dissemination1. Vaccinia virus (VACV), a close relative of smallpox virus, is thought to exploit cell motility as a means to enhance the spread of infection1. A single viral protein, F11L, contributes to this by blocking RhoA signalling to facilitate cell retraction2. However, F11L alone is not sufficient for VACV-induced cell motility, indicating that additional viral factors must be involved. Here, we show that the VACV epidermal growth factor homologue, VGF, promotes infected cell motility and the spread of viral infection. We found that VGF secreted from early infected cells is cleaved by ADAM10, after which it acts largely in a paracrine manner to direct cell motility at the leading edge of infection. Real-time tracking of cells infected in the presence of EGFR, MAPK, FAK and ADAM10 inhibitors or with VGF-deleted and F11-deleted viruses revealed defects in radial velocity and directional migration efficiency, leading to impaired cell-to-cell spread of infection. Furthermore, intravital imaging showed that virus spread and lesion formation are attenuated in the absence of VGF. Our results demonstrate how poxviruses hijack epidermal growth factor receptor-induced cell motility to promote rapid and efficient spread of infection in vitro and in vivo.


Assuntos
Movimento Celular , Interações Hospedeiro-Patógeno , Peptídeos/metabolismo , Transdução de Sinais , Vírus Vaccinia/fisiologia , Vaccinia/virologia , Proteína ADAM10/antagonistas & inibidores , Proteína ADAM10/metabolismo , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Efeito Citopatogênico Viral/genética , Inibidores Enzimáticos/farmacologia , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/metabolismo , Deleção de Genes , Células HeLa , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/metabolismo , Camundongos , Peptídeos/deficiência , Peptídeos/genética , Transdução de Sinais/efeitos dos fármacos , Vaccinia/metabolismo , Vaccinia/patologia , Vírus Vaccinia/genética , Vírus Vaccinia/crescimento & desenvolvimento , Vírus Vaccinia/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo
19.
J Immunol ; 201(4): 1222-1228, 2018 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-30012850

RESUMO

Probing the limits of CD8+ T cell immunosurveillance, we inserted the SIINFEKL peptide into influenza A virus (IAV)-negative strand gene segments. Although IAV genomic RNA is considered noncoding, there is a conserved, relatively long open reading frame present in segment 8, encoding a potential protein termed NEG8. The biosynthesis of NEG8 from IAV has yet to be demonstrated. Although we failed to detect NEG8 protein expression in IAV-infected mouse cells, cell surface Kb-SIINFEKL complexes are generated when SIINFEKL is genetically appended to the predicted C terminus of NEG8, as shown by activation of OT-I T cells in vitro and in vivo. Moreover, recombinant IAV encoding of SIINFEKL embedded in the negative strand of the neuraminidase-stalk coding sequence also activates OT-I T cells in mice. Together, our findings demonstrate both the translation of sequences on the negative strand of a single-stranded RNA virus and its relevance in antiviral immunosurveillance.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Vigilância Imunológica/imunologia , Vírus da Influenza A/genética , Vírus da Influenza A/imunologia , RNA Viral/imunologia , Animais , Células HeLa , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Infecções por Orthomyxoviridae/imunologia , Biossíntese de Proteínas/fisiologia , RNA Viral/genética
20.
JCI Insight ; 3(13)2018 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-29997290

RESUMO

Chikungunya virus (CHIKV) causes acute and chronic rheumatologic disease. Pathogenic CHIKV strains persist in joints of immunocompetent mice, while the attenuated CHIKV strain 181/25 is cleared by adaptive immunity. We analyzed the draining lymph node (dLN) to define events in lymphoid tissue that may contribute to CHIKV persistence or clearance. Acute 181/25 infection resulted in dLN enlargement and germinal center (GC) formation, while the dLN of mice infected with pathogenic CHIKV became highly disorganized and depleted of lymphocytes. Using CHIKV strains encoding ovalbumin-specific TCR epitopes, we found that lymphocyte depletion was not due to impaired lymphocyte proliferation. Instead, the accumulation of naive lymphocytes transferred from the vasculature to the dLN was reduced, which was associated with fewer high endothelial venule cells and decreased CCL21 production. Following NP-OVA immunization, NP-specific GC B cells in the dLN were decreased during pathogenic, but not attenuated, CHIKV infection. Our data suggest that pathogenic, persistent strains of CHIKV disable the development of adaptive immune responses within the dLN.


Assuntos
Imunidade Adaptativa , Febre de Chikungunya/imunologia , Vírus Chikungunya/patogenicidade , Linfonodos/imunologia , Animais , Linfócitos B , Proliferação de Células , Quimiocina CCL21 , Febre de Chikungunya/virologia , Modelos Animais de Doenças , Epitopos , Imunização , Linfonodos/patologia , Linfonodos/virologia , Ativação Linfocitária , Tecido Linfoide/imunologia , Tecido Linfoide/patologia , Camundongos , Camundongos Endogâmicos C57BL , Ovalbumina , Células Estromais/imunologia , Vênulas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...