Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Front Immunol ; 13: 961129, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36110846

RESUMO

Background: Rheumatoid arthritis (RA) is the most common inflammatory arthropathy. Immune dysregulation was implicated in the pathogenesis of RA. Thus, the aim of the research was to determine the immune related biomarkers in RA. Methods: We downloaded the gene expression data of RA in GSE89408 and GSE45291 from Gene Expression Omnibus public database (GEO). Differentially expressed genes (DEGs) were identified between RA and control groups. Infiltrating immune cells related genes were obtained by ssGSEA and weighted gene co-expression network analysis (WGCNA). We performed functional enrichment analysis of differentially expressed immunity-related genes (DEIRGs) by "clusterProfiler" R package, key genes screening by protein-protein interaction (PPI) network of DEIRGs. And mice collagen-induced arthritis (CIA) model was employed to verify these key genes. Results: A total of 1,885 up-regulated and 1,899 down-regulated DEGs were identified in RA samples. The ssGSEA analysis showed that the infiltration of 25 cells was significantly different. 603 immune related genes were obtained by WGCNA, and 270 DEIRGs were obtained by taking the intersection of DEGs and immune related genes. Enrichment analyses indicated that DEIRGs were associated with immunity related biological processes. 4 candidate biomarkers (CCR7, KLRK1, TIGIT and SLAMF1) were identified from the PPI network of DEIRGs and literature research.In mice CIA model, the immunohistochemical stain showed SLAMF1 has a significantly high expression in diseased joints. And flow cytometry analysis shows the expression of SLAMF1 on CIA mice-derived CTL cells, Th, NK cells, NKT cells, classical dendritic cell (cDCs) and monocytes/macrophages was also significantly higher than corresponding immune cells from HC mice. Conclusion: Our study identified SMLAF1 as a key biomarker in the development and progression of RA, which might provide new insight for exploring the pathogenesis of RA.


Assuntos
Artrite Experimental , Artrite Reumatoide , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária , Animais , Artrite Experimental/genética , Artrite Reumatoide/genética , Biomarcadores , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Humanos , Camundongos , Subfamília K de Receptores Semelhantes a Lectina de Células NK/genética , Receptores CCR7/genética , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/genética
2.
J Immunol ; 208(6): 1417-1423, 2022 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-35217584

RESUMO

Homotypic signaling lymphocyte activation molecule (SLAM) receptor-ligand cell surface interactions between myeloid and lymphoid cells regulate innate and adaptive immune responses. In this article, we report that SLAMF1 is indispensable for host resistance to primary and vaccine-induced protection against fungal infection. Because vaccine immunity is dependent on cell-mediated immunity, we investigated the development of Ag-specific T cells. We studied the T cell-intrinsic and -extrinsic role of SLAMF1. We generated SLAMF1-/- TCR transgenic mice and analyzed the responses of adoptively transferred T cells. We also tracked endogenous Ag-specific T cells by using a tetramer. Intrinsic and extrinsic SLAMF1 signaling was dispensable for the development of antifungal Th1 and Th17 cells, which are requisite for the acquisition of vaccine-induced immunity. Despite intact T cell development, vaccinated SLAMF1-/- mice failed to control fungal infection. Failed accumulation of Ag-specific T cells in the lung on infection of vaccinated mice was due to uncontrolled early infection and inflammation, revealing a role for SLAMF1 in innate host immunity.


Assuntos
Micoses , Vacinas , Animais , Diferenciação Celular , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/genética , Células Th17
3.
Exp Oncol ; 43(4): 312-316, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34967540

RESUMO

BACKGROUND: SLAMF1/CD150 receptor is aberrantly expressed in malignant hematopoietic cells compared to ubiquitous expression in their normal analogues. However, the data about CD150 expression and function outside the hematopoietic system are limited. The aim of this pilot study was to examine the profile of mRNA expression of CD150 isoforms and the protein topology in highly and low malignant breast (BC) and prostate cancer (PC) cell lines. MATERIALS AND METHODS: The study was performed on BC T47D, MDA-MB-231, ВСС/Р and BC/ML cell lines and PC LNCap, Du-145 and PC-3 cell lines. The quantitative polymerase chain reaction was applied for study of CD150 isoforms mRNA expression and flow cytometry was used for determination of protein localization. RESULTS: Analyzed BC cell lines did not express CD150 on the cell surface membrane (csCD150-), but more than 45% of cells were positive for CD150 cytoplasmic reaction (cyCD150+). The cyCD150 expression level in T47D cells of luminal BC subtype was higher than in basal BC cell lines MDA-MB-231, ВСС/Р and BC/ML. The PC cell lines expressed CD150 both on the cell surface and in cytoplasm. The highest number of csCD150+ and cyCD150+ cells was revealed in less aggressive androgen responsive, non-metastatic LNCap cell line. All studied BC and PC cell lines expressed mRNA of canonical transmembrane mCD150 and novel nCD150 isoforms but with different pattern of prevalence. Soluble CD150 isoform was revealed at the low level only in BCC/P BC cell line and LNCap, PC-3 PC cell lines. CONCLUSIONS: We have shown that BC and PC cell lines differentially expressed multifunctional receptor CD150 at the mRNA and protein levels that may indicate its association with the degree of their malignancy.


Assuntos
Neoplasias da Mama , Neoplasias da Próstata/genética , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/genética , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Feminino , Humanos , Células MCF-7 , Masculino , Projetos Piloto , Isoformas de Proteínas/genética
4.
Viruses ; 13(4)2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33916225

RESUMO

Measles virus (MV) can cause severe acute diseases as well as long-lasting clinical deteriorations due to viral-induced immunosuppression and neuronal manifestation. How the virus enters the brain and manages to persist in neuronal tissue is not fully understood. Various mutations in the viral genes were found in MV strains isolated from patient brains. In this study, reverse genetics was used to introduce mutations in the fusion, matrix and polymerase genes of MV. The generated virus clones were characterized in cell culture and used to infect rat brain slice cultures. A mutation in the carboxy-terminal domain of the matrix protein (R293Q) promoted the production of progeny virions. This effect was observed in Vero cells irrespective of the expression of the signaling lymphocyte activation molecule (SLAM). Furthermore, a mutation in the fusion protein (I225M) induced syncytia formation on Vero cells in the absence of SLAM and promoted viral spread throughout the rat brain slices. In this study, a solid ex vivo model was established to elucidate the MV mutations contributing to neural manifestation.


Assuntos
Encéfalo/virologia , Vírus do Sarampo/genética , Mutação , Neurônios/virologia , Proteínas Virais/genética , Tropismo Viral/genética , Animais , Chlorocebus aethiops , Células HEK293 , Humanos , Técnicas In Vitro , Sarampo/virologia , Vírus do Sarampo/patogenicidade , Vírus do Sarampo/fisiologia , Ratos Endogâmicos Lew , Genética Reversa , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/genética , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/metabolismo , Células Vero , Proteínas Virais de Fusão/genética
5.
J Infect Dis ; 223(4): 667-672, 2021 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-32623457

RESUMO

Measles virus (MeV) binds, infects, and kills CD150+ memory T cells, leading to immune amnesia. Whether MeV targets innate, memory-like T cells is unknown. We demonstrate that human peripheral blood and hepatic mucosa-associated invariant T (MAIT) cells and invariant natural killer T cells express surprisingly high levels of CD150, more than other lymphocyte subsets. Furthermore, exposing MAIT cells to MeV results in their efficient infection and rapid apoptosis. This constitutes the first report of direct MAIT cell infection by a viral pathogen. Given MAIT cells' antimicrobial properties, their elimination by MeV may contribute to measles-induced immunosuppression and heightened vulnerability to unrelated infections.


Assuntos
Apoptose , Vírus do Sarampo/fisiologia , Células T Invariantes Associadas à Mucosa/fisiologia , Células T Invariantes Associadas à Mucosa/virologia , Feminino , Humanos , Interleucina-12/imunologia , Interleucina-18/imunologia , Leucócitos Mononucleares/imunologia , Masculino , Proteína Cofatora de Membrana/genética , Proteína Cofatora de Membrana/metabolismo , Células T Invariantes Associadas à Mucosa/imunologia , Células T Matadoras Naturais/imunologia , Células T Matadoras Naturais/metabolismo , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/genética , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/metabolismo , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo
6.
Br J Haematol ; 192(6): 1068-1072, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32578873

RESUMO

In a series of 349 patients with chronic lymphocytic leukaemia (CLL), we found lower levels of signalling lymphocytic activation molecule family member 1 (SLAMF1) expression in cases with highly complex karyotypes, as defined by the presence of five or more chromosomal abnormalities (CK5; P < 0·001) and with major chromosomal structural abnormalities (P < 0·001). SLAMF1 downregulation was significantly associated with advanced Binet Stage (P = 0·001), CD38 positivity (P < 0·001), high ß2 -microglobulin levels (P < 0·001), immunoglobulin heavy chain variable region gene (IGHV) unmutated status (P < 0·001), 11q deletion (P < 0·001), tumour protein p53 (TP53) disruption (P = 0·011) and higher risk CLL International Prognostic Index categories (P < 0·001). Multivariate analysis showed that downregulated SLAMF1 levels had independent negative prognostic impact on time-to-first treatment (P < 0·001) and overall survival (P < 0·001).


Assuntos
Aberrações Cromossômicas , Leucemia Linfocítica Crônica de Células B , Proteínas de Neoplasias , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária , Adulto , Idoso , Intervalo Livre de Doença , Feminino , Humanos , Leucemia Linfocítica Crônica de Células B/sangue , Leucemia Linfocítica Crônica de Células B/genética , Leucemia Linfocítica Crônica de Células B/mortalidade , Masculino , Pessoa de Meia-Idade , Proteínas de Neoplasias/sangue , Proteínas de Neoplasias/genética , Valor Preditivo dos Testes , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/sangue , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/genética , Taxa de Sobrevida
7.
PLoS Negl Trop Dis ; 14(9): e0008608, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32925918

RESUMO

The receptor Signaling Lymphocyte-Activation Molecule Family 1 (SLAMF1) controls susceptibility to Infection by the lethal Trypanosoma cruzi Y strain. To elucidate whether genetic diversity of the parasite was related with disease susceptibility, we further analyzed the role of SLAMF1 using 6 different Trypanosoma cruzi strains including Y. The interaction of SLAMF1 receptor with T. cruzi was evidenced by fluorescence microscopy, flow cytometry and quantitative PCR. All the strains, except VFRA, showed a decrease in parasite load in infected macrophages in Slamf1-/- compared to BALB/c. In macrophages gene expression NADPH oxidase (NOX2), and reactive oxygen species (ROS) production increased in Slamf1-/- compared to BALB/c in 5 out of 6 strains. However, Slamf1-/-macrophages infected with VFRA strain exhibited a divergent behavior, with higher parasite load, lower NOX2 expression and ROS production compared to BALB/c. Parasitological and immunological studies in vivo with Y strain showed that in the absence of SLAMF1 the immune response protected mice from the otherwise lethal Y infection favoring a proinflammatory response likely involving CD4, CD8, dendritic cells and classically activated macrophages. In the case of VFRA, no major changes were observed in the absence of SLAMF1. Thus, the results suggest that the T. cruzi affects SLAMF1-dependent ROS production, controlling parasite replication in macrophages and affecting survival in mice in a strain-dependent manner. Further studies will focus in the identification of parasite molecules involved in SLAMF1 interaction to explain the immunopathogenesis of the disease.


Assuntos
Macrófagos/parasitologia , Espécies Reativas de Oxigênio/metabolismo , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/genética , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/metabolismo , Trypanosoma cruzi/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular , Doença de Chagas/imunologia , Chlorocebus aethiops , Células Dendríticas/imunologia , Suscetibilidade a Doenças/imunologia , Células HEK293 , Coração/parasitologia , Humanos , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Miocárdio/patologia , NADPH Oxidase 2/genética , NADPH Oxidase 2/metabolismo , Carga Parasitária , Células Vero
8.
Curr Opin Virol ; 41: 38-45, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32344228

RESUMO

Like measles virus (MV), whose primary hosts are humans, non-human animal morbilliviruses use SLAM (signaling lymphocytic activation molecule) and PVRL4 (nectin-4) expressed on immune and epithelial cells, respectively, as receptors. PVRL4's amino acid sequence is highly conserved across species, while that of SLAM varies significantly. However, non-host animal SLAMs often function as receptors for different morbilliviruses. Uniquely, human SLAM is somewhat specific for MV, but canine distemper virus, which shows the widest host range among morbilliviruses, readily gains the ability to use human SLAM. The host range for morbilliviruses is also modulated by their ability to counteract the host's innate immunity, but the risk of cross-species transmission of non-human animal morbilliviruses to humans could occur if MV is successfully eradicated.


Assuntos
Infecções por Morbillivirus/veterinária , Infecções por Morbillivirus/virologia , Morbillivirus/fisiologia , Zoonoses Virais/transmissão , Animais , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Especificidade de Hospedeiro , Humanos , Morbillivirus/genética , Infecções por Morbillivirus/metabolismo , Infecções por Morbillivirus/transmissão , Receptores Virais/genética , Receptores Virais/metabolismo , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/genética , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/metabolismo , Zoonoses Virais/genética , Zoonoses Virais/metabolismo , Zoonoses Virais/virologia
9.
Microbiol Immunol ; 64(8): 578-583, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32215955

RESUMO

Morbilliviruses use the signaling lymphocyte activation molecule (SLAM) as a receptor to infect their hosts. Seals are almost the only animal species that show apparent infection with phocine distemper virus (PDV). Seal SLAM functioned as a PDV receptor. However, dolphin- and dog-SLAM molecules, but not human SLAM, were also fully functional PDV receptors. These data suggest that the host range of PDV is not simply determined by its SLAM usage. However, human nonsusceptibility to PDV infection may be at least partly attributable to the inability of PDV to use human SLAM as a receptor.


Assuntos
Vírus da Cinomose Canina/fisiologia , Vírus da Cinomose Focina/fisiologia , Morbillivirus/fisiologia , Receptores Virais/fisiologia , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/fisiologia , Animais , Linhagem Celular , Chlorocebus aethiops , Cinomose/virologia , Cães/virologia , Humanos , Phoca/virologia , Receptores Virais/genética , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/genética , Stenella/virologia , Células Vero
10.
J Alzheimers Dis ; 74(1): 139-149, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31985465

RESUMO

BACKGROUND: As cognitive function declines with age, identifying factors affecting the trajectory of cognitive decline is an indispensable step toward developing intervention strategies to improve the quality of the elderly life. OBJECTIVE: We performed a genome-wide association study (GWAS) focusing on memory function to explore single nucleotide polymorphisms (SNPs) associated with the rate of memory decline. METHODS: Seven hundred and nine eligible non-Hispanic Caucasians from the Alzheimer's Disease Neuroimaging Initiative (ADNI) were included for analysis after quality control. GWAS was performed with linear regression. We subsequently tested whether the associations remained significant in subgroup analysis and also examined the impact of SNPs on the longitudinal changes in other neuropsychological measures and amyloid pathology. RESULTS: We identified rs13374761-A in SLAMF1 gene associated with less memory decline (MAF = 0.071, ß= 0.0103, p = 4.14×10-8). Subgroup analysis showed stability of results across groups with different diagnosis at baseline. Rs13374761-A also had protective effects on global cognition (p = 0.024), episodic memory (p = 0.024), and semantic memory (p = 0.042), and exerts protection against a decrease in CSF Aß42 concentration (p = 0.0463) and an increase in Aß loading in cerebral cortex (p = 0.00666) among minor allele carriers. CONCLUSION: A novel variant in gene SLAMF1 affects the rate of memory decline in the aged population. Given the protective effect of this variant, SLAMF1 should be further investigated as a potential preventive and therapeutic target for monitoring cognition trajectories.


Assuntos
Disfunção Cognitiva/genética , Transtornos da Memória/genética , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/genética , Idoso , Idoso de 80 Anos ou mais , Peptídeos beta-Amiloides/líquido cefalorraquidiano , Apolipoproteínas E/genética , Angiopatia Amiloide Cerebral/genética , Angiopatia Amiloide Cerebral/patologia , Disfunção Cognitiva/psicologia , Progressão da Doença , Feminino , Estudo de Associação Genômica Ampla , Heterozigoto , Humanos , Estudos Longitudinais , Masculino , Transtornos da Memória/psicologia , Pessoa de Meia-Idade , Neuroimagem , Testes Neuropsicológicos , Fragmentos de Peptídeos/líquido cefalorraquidiano , Polimorfismo de Nucleotídeo Único , População Branca
11.
Cell Microbiol ; 22(4): e13164, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31953913

RESUMO

The strategies by which intracellular pathogenic bacteria manipulate innate immunity to establish chronicity are poorly understood. Here, we show that Brucella abortus outer membrane protein Omp25 specifically binds the immune cell receptor SLAMF1 in vitro. The Omp25-dependent engagement of SLAMF1 by B. abortus limits NF-κB translocation in dendritic cells (DCs) with no impact on Brucella intracellular trafficking and replication. This in turn decreases pro-inflammatory cytokine secretion and impairs DC activation. The Omp25-SLAMF1 axis also dampens the immune response without affecting bacterial replication in vivo during the acute phase of Brucella infection in a mouse model. In contrast, at the chronic stage of infection, the Omp25/SLAMF1 engagement is essential for Brucella persistence. Interaction of a specific bacterial protein with an immune cell receptor expressed on the DC surface at the acute stage of infection is thus a powerful mechanism to support microbe settling in its replicative niche and progression to chronicity.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Brucella abortus/imunologia , Células Dendríticas/microbiologia , Interações Hospedeiro-Patógeno/imunologia , Inflamação , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/metabolismo , Animais , Proteínas da Membrana Bacteriana Externa/genética , Brucella abortus/genética , Brucella abortus/patogenicidade , Células Dendríticas/imunologia , Feminino , Imunidade Inata , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Ligação Proteica , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/genética
12.
J Virol ; 94(2)2020 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-31619560

RESUMO

Measles virus (MeV) is an enveloped RNA virus bearing two envelope glycoproteins, the hemagglutinin (H) and fusion (F) proteins. Upon receptor binding, the H protein triggers conformational changes of the F protein, causing membrane fusion and subsequent virus entry. MeV may persist in the brain, infecting neurons and causing fatal subacute sclerosing panencephalitis (SSPE). Since neurons do not express either of the MeV receptors, signaling lymphocytic activation molecule (SLAM; also called CD150) and nectin-4, how MeV propagates in neurons is unknown. Recent studies have shown that specific substitutions in the F protein found in MeV isolates from SSPE patients are critical for MeV neuropathogenicity by rendering the protein unstable and hyperfusogenic. Recombinant MeVs possessing the F proteins with such substitutions can spread in primary human neurons and in the brains of mice and hamsters and induce cell-cell fusion in cells lacking SLAM and nectin-4. Here, we show that receptor-blind mutant H proteins that have decreased binding affinities to receptors can support membrane fusion mediated by hyperfusogenic mutant F proteins, but not the wild-type F protein, in cells expressing the corresponding receptors. The results suggest that weak interactions of the H protein with certain molecules (putative neuron receptors) trigger hyperfusogenic F proteins in SSPE patients. Notably, where cell-cell contacts are ensured, the weak cis interaction of the H protein with SLAM on the same cell surface also could trigger hyperfusogenic F proteins. Some enveloped viruses may exploit such cis interactions with receptors to infect target cells, especially in cell-to-cell transmission.IMPORTANCE Measles virus (MeV) may persist in the brain, causing incurable subacute sclerosing panencephalitis (SSPE). Because neurons, the main target in SSPE, do not express receptors for wild-type (WT) MeV, how MeV propagates in the brain is a key question for the disease. Recent studies have demonstrated that specific substitutions in the MeV fusion (F) protein are critical for neuropathogenicity. Here, we show that weak cis and trans interactions of the MeV attachment protein with receptors that are not sufficient to trigger the WT MeV F protein can trigger the mutant F proteins from neuropathogenic MeV isolates. Our study not only provides an important clue to understand MeV neuropathogenicity but also reveals a novel viral strategy to expand cell tropism.


Assuntos
Moléculas de Adesão Celular/metabolismo , Hemaglutininas Virais/metabolismo , Vírus do Sarampo/metabolismo , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/metabolismo , Panencefalite Esclerosante Subaguda/metabolismo , Proteínas Virais de Fusão/metabolismo , Animais , Moléculas de Adesão Celular/genética , Linhagem Celular , Cricetinae , Hemaglutininas Virais/genética , Humanos , Vírus do Sarampo/genética , Vírus do Sarampo/patogenicidade , Camundongos , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/genética , Panencefalite Esclerosante Subaguda/genética , Panencefalite Esclerosante Subaguda/patologia , Proteínas Virais de Fusão/genética
13.
Can J Microbiol ; 65(11): 783-794, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31238018

RESUMO

SLAM (CD150) and nectin-4 are the major morbillivirus receptors responsible for virus pathogenesis and host range expansion. Recently, morbillivirus infections have been reported in unnatural hosts, including endangered species, posing a threat to their conservation. To understand the host range expansion of morbilliviruses, we generated the full-length sequences of morbillivirus receptors (goat, sheep, and dog SLAM, and goat nectin-4) and tried to correlate their role in determining host tropism. A high level of amino acid identity was observed between the sequences of related species, and phylogenetic reconstruction showed that the receptor sequences of carnivores, marine mammals, and small ruminants grouped separately. Analysis of the ligand binding region (V region; amino acid residues 52-136) of SLAM revealed high amino acid identity between small ruminants and bovine SLAMs. Comparison of canine SLAM with ruminants and non-canids SLAM revealed appreciable changes, including charge alterations. Significant differences between feline SLAM and canine SLAM have been reported. The binding motifs of nectin-4 genes (FPAG motif and amino acid residues 60, 62, and 63) were found to be conserved in sheep, goat, and dog. The differences reported in the binding region may be responsible for the level of susceptibility or resistance of a species to a particular morbillivirus.


Assuntos
Mamíferos/genética , Infecções por Morbillivirus/veterinária , Morbillivirus/fisiologia , Receptores Virais/genética , Sequência de Aminoácidos , Animais , Gatos/genética , Bovinos/genética , Moléculas de Adesão Celular/química , Moléculas de Adesão Celular/genética , Cães/genética , Cabras/genética , Especificidade de Hospedeiro , Mamíferos/classificação , Mamíferos/virologia , Morbillivirus/genética , Infecções por Morbillivirus/genética , Infecções por Morbillivirus/metabolismo , Infecções por Morbillivirus/virologia , Filogenia , Receptores Virais/química , Alinhamento de Sequência , Análise de Sequência , Ovinos/genética , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/química , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/genética
14.
Virus Res ; 269: 197634, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31129173

RESUMO

Peste des petits ruminants virus (PPRV), a member of the genus Morbillivirus, in the family Paramyxoviridae expresses two membrane glycoproteins, the fusion (F) and haemagglutinin (H) glycoproteins which mediate virus-to-cell fusion and cell-to-cell fusion leading to the induction of syncytia in PPRV infected cells. In the context of the characterization of the virulent lineage IV strain PPRV Kurdistan 2011, isolated from wild goats from the Kurdistan region in Iraq, we observed that both PPRV Kurdistan 2011 and the PPRV Nigeria 75/1 vaccine strain led to induction of large syncytia in Vero-dogSLAM cells within 48 h whereas both failed to induce detectable cell-cell fusion events in two Vero cell lines of differing passage histories. We were unable to detect syncytium formation in transiently transfected cells expressing PPRV F or H alone whereas co-expression of F and H induced large syncytia - in Vero-dogSLAM cells only. In VeroMontpellier cells expressing PPRV F and H, fused cells were rarely detectable indicating that PPRV mediated cell fusion activity is impaired in this cell line. Surprisingly, on Vero-dogSLAM cells the vaccine strain grew to titers of 105.25 TCID50/ml, whereas infectious virus yield was about 200-fold higher on VeroMontpellier and Vero-76 cells. In contrast, the virulent Kurdistan 2011 strain grew to a maximum titer of 107.0 TCID50/ml on Vero-dogSLAM cells and only 104.5 TCID50/ml on normal Vero cells. This was as expected since Vero cells lacking the SLAM receptor for PPRV are regarded as not so permissive for infection. To elucidate the divergent productive replication behaviour of PPRV Nigeria 75/1 vaccine strain on Vero vs Vero-dogSLAM cells, we examined whether intracellular transport and/or maturation of the viral envelope glycoproteins F and H might be implicated with this phenomenon. The results indicate that F in contrast to the H glycoprotein matures inefficiently during intracellular transport in VeroMontpellier cells, thus leading to an absence of detectable syncytia formation. However, in the case of the PPRV Nigeria 75/1 vaccine strain this did not impair efficient virus assembly and release.


Assuntos
Vírus da Peste dos Pequenos Ruminantes/fisiologia , Proteínas Virais de Fusão/metabolismo , Montagem de Vírus , Replicação Viral , Animais , Transporte Biológico , Chlorocebus aethiops , Doenças das Cabras/virologia , Cabras/virologia , Hemaglutininas Virais/metabolismo , Iraque , Peste dos Pequenos Ruminantes/prevenção & controle , Vírus da Peste dos Pequenos Ruminantes/classificação , Vírus da Peste dos Pequenos Ruminantes/imunologia , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/genética , Células Vero
15.
Virol J ; 16(1): 30, 2019 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-30845967

RESUMO

BACKGROUND: Canine distemper virus (CDV), currently termed Canine morbillivirus, is an extremely contagious disease that affects dogs. It is identified as a multiple cell tropism pathogen, and its host range includes a vast array of species. As a member of Mononegavirales, CDV has a negative, single-stranded RNA genome, which encodes eight proteins. MAIN BODY: Regarding the molecular pathogenesis, the hemagglutinin protein (H) plays a crucial role both in the antigenic recognition and the viral interaction with SLAM and nectin-4, the host cells' receptors. These cellular receptors have been studied widely as CDV receptors in vitro in different cellular models. The SLAM receptor is located in lymphoid cells; therefore, the infection of these cells by CDV leads to immunosuppression, the severity of which can lead to variability in the clinical disease with the potential of secondary bacterial infection, up to and including the development of neurological signs in its later stage. CONCLUSION: Improving the understanding of the CDV molecules implicated in the determination of infection, especially the H protein, can help to enhance the biochemical comprehension of the difference between a wide range of CDV variants, their tropism, and different steps in viral infection. The regions of interaction between the viral proteins and the identified host cell receptors have been elucidated to facilitate this understanding. Hence, this review describes the significant molecular and cellular characteristics of CDV that contribute to viral pathogenesis.


Assuntos
Vírus da Cinomose Canina/genética , Vírus da Cinomose Canina/patogenicidade , Cinomose/virologia , Interações entre Hospedeiro e Microrganismos , Tropismo Viral , Animais , Modelos Animais de Doenças , Vírus da Cinomose Canina/fisiologia , Cães , Hemaglutininas Virais/genética , Especificidade de Hospedeiro , Humanos , Camundongos , Nectinas/genética , Receptores Virais/genética , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/genética , Proteínas Virais/genética , Zoonoses/virologia
16.
J Immunol ; 202(8): 2276-2286, 2019 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-30796181

RESUMO

Invariant NKT (iNKT) cells are innate lipid-reactive T cells that develop and differentiate in the thymus into iNKT1/2/17 subsets, akin to TH1/2/17 conventional CD4 T cell subsets. The factors driving the central priming of iNKT cells remain obscure, although strong/prolonged TCR signals appear to favor iNKT2 cell development. The Src homology 2 domain-containing phosphatase 1 (Shp1) is a protein tyrosine phosphatase that has been identified as a negative regulator of TCR signaling. In this study, we found that mice with a T cell-specific deletion of Shp1 had normal iNKT cell numbers and peripheral distribution. However, iNKT cell differentiation was biased toward the iNKT2/17 subsets in the thymus but not in peripheral tissues. Shp1-deficient iNKT cells were also functionally biased toward the production of TH2 cytokines, such as IL-4 and IL-13. Surprisingly, we found no evidence that Shp1 regulates the TCR and Slamf6 signaling cascades, which have been suggested to promote iNKT2 differentiation. Rather, Shp1 dampened iNKT cell proliferation in response to IL-2, IL-7, and IL-15 but not following TCR engagement. Our findings suggest that Shp1 controls iNKT cell effector differentiation independently of positive selection through the modulation of cytokine responsiveness.


Assuntos
Diferenciação Celular/imunologia , Células T Matadoras Naturais/imunologia , Proteína Tirosina Fosfatase não Receptora Tipo 6/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Transdução de Sinais/imunologia , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/imunologia , Animais , Diferenciação Celular/genética , Citocinas/genética , Citocinas/imunologia , Camundongos , Camundongos Knockout , Células T Matadoras Naturais/citologia , Proteína Tirosina Fosfatase não Receptora Tipo 6/genética , Receptores de Antígenos de Linfócitos T/genética , Transdução de Sinais/genética , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/genética
17.
J Virol ; 92(23)2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30232185

RESUMO

Morbilliviruses infect a broad range of mammalian hosts, including ruminants, carnivores, and humans. The recent eradication of rinderpest virus (RPV) and the active campaigns for eradication of the human-specific measles virus (MeV) have raised significant concerns that the remaining morbilliviruses may emerge in so-called vacated ecological niches. Seeking to assess the zoonotic potential of nonhuman morbilliviruses within human populations, we found that peste des petits ruminants virus (PPRV)-the small-ruminant morbillivirus-is restricted at the point of entry into human cells due to deficient interactions with human SLAMF1-the immune cell receptor for morbilliviruses. Using a structure-guided approach, we characterized a single amino acid change, mapping to the receptor-binding domain in the PPRV hemagglutinin (H) protein, which overcomes this restriction. The same mutation allowed escape from some cross-protective, human patient, anti-MeV antibodies, raising concerns that PPRV is a pathogen with zoonotic potential. Analysis of natural variation within human and ovine SLAMF1 also identified polymorphisms that could correlate with disease resistance. Finally, the mechanistic nature of the PPRV restriction was also investigated, identifying charge incompatibility and steric hindrance between PPRV H and human SLAMF1 proteins. Importantly, this research was performed entirely using surrogate virus entry assays, negating the requirement for in situ derivation of a human-tropic PPRV and illustrating alternative strategies for identifying gain-of-function mutations in viral pathogens.IMPORTANCE A significant proportion of viral pandemics occur following zoonotic transmission events, where animal-associated viruses jump species into human populations. In order to provide forewarnings of the emergence of these viruses, it is necessary to develop a better understanding of what determines virus host range, often at the genetic and structural levels. In this study, we demonstrated that the small-ruminant morbillivirus, a close relative of measles, is unable to use human receptors to enter cells; however, a change of a single amino acid in the virus is sufficient to overcome this restriction. This information will be important for monitoring this virus's evolution in the field. Of note, this study was undertaken in vitro, without generation of a fully infectious virus with this phenotype.


Assuntos
Anticorpos Antivirais/imunologia , Glicoproteínas/metabolismo , Mutação , Peste dos Pequenos Ruminantes/virologia , Vírus da Peste dos Pequenos Ruminantes/patogenicidade , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/metabolismo , Replicação Viral , Sequência de Aminoácidos , Animais , Chlorocebus aethiops , Glicoproteínas/química , Glicoproteínas/genética , Glicoproteínas/imunologia , Humanos , Modelos Teóricos , Mutagênese Sítio-Dirigida , Peste dos Pequenos Ruminantes/patologia , Peste dos Pequenos Ruminantes/transmissão , Vírus da Peste dos Pequenos Ruminantes/genética , Vírus da Peste dos Pequenos Ruminantes/imunologia , Conformação Proteica , Homologia de Sequência , Ovinos , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/química , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/genética , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/imunologia , Células Vero
18.
J Cell Biol ; 217(4): 1411-1429, 2018 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-29440514

RESUMO

Signaling lymphocytic activation molecule family 1 (SLAMF1) is an Ig-like receptor and a costimulatory molecule that initiates signal transduction networks in a variety of immune cells. In this study, we report that SLAMF1 is required for Toll-like receptor 4 (TLR4)-mediated induction of interferon ß (IFNß) and for killing of Gram-negative bacteria by human macrophages. We found that SLAMF1 controls trafficking of the Toll receptor-associated molecule (TRAM) from the endocytic recycling compartment (ERC) to Escherichia coli phagosomes. In resting macrophages, SLAMF1 is localized to ERC, but upon addition of E. coli, it is trafficked together with TRAM from ERC to E. coli phagosomes in a Rab11-dependent manner. We found that endogenous SLAMF1 protein interacted with TRAM and defined key interaction domains as amino acids 68 to 95 of TRAM as well as 15 C-terminal amino acids of SLAMF1. Interestingly, the SLAMF1-TRAM interaction was observed for human but not mouse proteins. Overall, our observations suggest that SLAMF1 is a new target for modulation of TLR4-TRAM-TRIF inflammatory signaling in human cells.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Endossomos/metabolismo , Ativação de Macrófagos , Macrófagos/metabolismo , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/metabolismo , Receptor 4 Toll-Like/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Proteínas Adaptadoras de Transporte Vesicular/genética , Proteínas Adaptadoras de Transporte Vesicular/imunologia , Animais , Endossomos/efeitos dos fármacos , Endossomos/imunologia , Endossomos/microbiologia , Escherichia coli/metabolismo , Escherichia coli/patogenicidade , Células HEK293 , Interações Hospedeiro-Patógeno , Humanos , Fator Regulador 3 de Interferon/metabolismo , Interferon beta/genética , Interferon beta/metabolismo , Lipopolissacarídeos/farmacologia , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/microbiologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fagossomos/metabolismo , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Proteínas Serina-Treonina Quinases/metabolismo , Transporte Proteico , Transdução de Sinais , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/genética , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/imunologia , Células THP-1 , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/imunologia , Proteínas rab de Ligação ao GTP/genética , Proteínas rab de Ligação ao GTP/metabolismo
19.
J Virol ; 92(6)2018 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-29298883

RESUMO

Measles virus (MV) usually causes acute infection but in rare cases persists in the brain, resulting in subacute sclerosing panencephalitis (SSPE). Since human neurons, an important target affected in the disease, do not express the known MV receptors (signaling lymphocyte activation molecule [SLAM] and nectin 4), how MV infects neurons and spreads between them is unknown. Recent studies have shown that many virus strains isolated from SSPE patients possess substitutions in the extracellular domain of the fusion (F) protein which confer enhanced fusion activity. Hyperfusogenic viruses with such mutations, unlike the wild-type MV, can induce cell-cell fusion even in SLAM- and nectin 4-negative cells and spread efficiently in human primary neurons and the brains of animal models. We show here that a hyperfusogenic mutant MV, IC323-F(T461I)-EGFP (IC323 with a fusion-enhancing T461I substitution in the F protein and expressing enhanced green fluorescent protein), but not the wild-type MV, spreads in differentiated NT2 cells, a widely used human neuron model. Confocal time-lapse imaging revealed the cell-to-cell spread of IC323-F(T461I)-EGFP between NT2 neurons without syncytium formation. The production of virus particles was strongly suppressed in NT2 neurons, also supporting cell-to-cell viral transmission. The spread of IC323-F(T461I)-EGFP was inhibited by a fusion inhibitor peptide as well as by some but not all of the anti-hemagglutinin antibodies which neutralize SLAM- or nectin-4-dependent MV infection, suggesting the presence of a distinct neuronal receptor. Our results indicate that MV spreads in a cell-to-cell manner between human neurons without causing syncytium formation and that the spread is dependent on the hyperfusogenic F protein, the hemagglutinin, and the putative neuronal receptor for MV.IMPORTANCE Measles virus (MV), in rare cases, persists in the human central nervous system (CNS) and causes subacute sclerosing panencephalitis (SSPE) several years after acute infection. This neurological complication is almost always fatal, and there is currently no effective treatment for it. Mechanisms by which MV invades the CNS and causes the disease remain to be elucidated. We have previously shown that fusion-enhancing substitutions in the fusion protein of MVs isolated from SSPE patients contribute to MV spread in neurons. In this study, we demonstrate that MV bearing the hyperfusogenic mutant fusion protein spreads between human neurons in a cell-to-cell manner. Spread of the virus was inhibited by a fusion inhibitor peptide and antibodies against the MV hemagglutinin, indicating that both the hemagglutinin and hyperfusogenic fusion protein play important roles in MV spread between human neurons. The findings help us better understand the disease process of SSPE.


Assuntos
Hemaglutininas Virais/metabolismo , Vírus do Sarampo/metabolismo , Sarampo/transmissão , Neurônios/metabolismo , Panencefalite Esclerosante Subaguda/transmissão , Proteínas Virais de Fusão/metabolismo , Animais , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Chlorocebus aethiops , Hemaglutininas Virais/genética , Humanos , Sarampo/genética , Sarampo/metabolismo , Sarampo/patologia , Vírus do Sarampo/genética , Vírus do Sarampo/patogenicidade , Neurônios/patologia , Neurônios/virologia , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/genética , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/metabolismo , Panencefalite Esclerosante Subaguda/genética , Panencefalite Esclerosante Subaguda/metabolismo , Panencefalite Esclerosante Subaguda/patologia , Células Vero , Proteínas Virais de Fusão/genética
20.
Future Oncol ; 14(4): 319-329, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29091475

RESUMO

Monoclonal antibodies (mAb) represent a new frontier to treat newly diagnosed and relapsed-refractory multiple myeloma (MM). Elotuzumab, an mAb targeted SLAM7 in the plasma cells and natural killer cells surface, is the first mAb approved for the treatment of relapsed-refractory MM in combination with lenalidomide and dexamethasone. This approval was the final result of several preclinical and Phase I-II clinical studies leading to ELOQUENT-2 Phase III trial that demonstrated that elotuzumab adds a significant and durable value to standard therapy, paved the way of this new treatment strategy for MM. In this review we will describe elotuzumab mechanisms of action, clinical pharmacology and clinical studies that have led to these developments.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Mieloma Múltiplo/tratamento farmacológico , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/antagonistas & inibidores , Anticorpos Monoclonais Humanizados/efeitos adversos , Ensaios Clínicos como Assunto , Dexametasona/uso terapêutico , Humanos , Células Matadoras Naturais/efeitos dos fármacos , Mieloma Múltiplo/genética , Mieloma Múltiplo/patologia , Recidiva , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA