Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 116
Filtrar
1.
J Feline Med Surg ; 26(8): 1098612X241264731, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39212546

RESUMO

OBJECTIVES: Vaccinations should only be given to healthy cats, and deworming before vaccination is generally recommended; however, so far, no study has investigated the influence of intestinal parasitic infection on the immune response in kittens. The aim of this prospective study was to compare the antibody response to feline panleukopenia virus (FPV) vaccination in kittens with and without intestinal parasites. METHODS: Overall, 74 healthy kittens were included. Of these, 17 had intestinal parasites (12/17 Toxocara cati, 6/17 Cystoisospora felis, 1/17 Capillaria species). Both kittens with and without (n = 57) parasites received two primary kitten vaccinations with modified live FPV vaccines in a 4-week interval starting at the age of 8-12 weeks. Anti-FPV antibodies were determined at the beginning of the study (week 0) and at week 8 (4 weeks after the second vaccination) by haemagglutination inhibition. A ⩾four-fold titre increase (week 8 vs week 0) was defined as a response to vaccination. Comparison of the immune response in the kittens with and without intestinal parasites was performed using Pearson's χ2 test. RESULTS: Pre-vaccination antibodies were present in 4/17 (23.5%) kittens with intestinal parasites and in 24/57 (42.1%) without parasites. A ⩾four-fold titre increase was seen in 13/17 (76.5%) kittens with parasites compared with 32/57 (56.1%) kittens without parasites. There was neither a significant difference in pre-vaccination antibodies (P = 0.17), nor in vaccination response (P = 0.13) between kittens with and without parasites. CONCLUSIONS AND RELEVANCE: The results indicate that asymptomatic intestinal infections with endoparasites do not interfere with the immune response to kitten vaccination series. Parasitic infection (at least with T cati, C felis and Capillaria species) is therefore not a reason to postpone important vaccinations.


Assuntos
Anticorpos Antivirais , Vírus da Panleucopenia Felina , Panleucopenia Felina , Enteropatias Parasitárias , Vacinas Virais , Animais , Gatos , Vírus da Panleucopenia Felina/imunologia , Panleucopenia Felina/prevenção & controle , Panleucopenia Felina/imunologia , Enteropatias Parasitárias/veterinária , Enteropatias Parasitárias/prevenção & controle , Enteropatias Parasitárias/imunologia , Anticorpos Antivirais/sangue , Vacinas Virais/imunologia , Vacinas Virais/administração & dosagem , Masculino , Vacinação/veterinária , Feminino , Doenças do Gato/prevenção & controle , Doenças do Gato/imunologia , Doenças do Gato/parasitologia , Doenças do Gato/virologia , Estudos Prospectivos , Infecções por Caliciviridae/veterinária , Infecções por Caliciviridae/prevenção & controle , Infecções por Caliciviridae/imunologia
2.
Virol Sin ; 39(4): 675-684, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38997087

RESUMO

Norovirus (NoV) infection is a major cause of gastroenteritis worldwide. The virus poses great challenges in developing vaccines with broad immune protection due to its genetic and antigenic diversity. To date, there are no approved NoV vaccines for clinical use. Here, we aimed to develop a broad-acting quadrivalent NoV vaccine based on a chimpanzee adenovirus vector, AdC68, carrying the major capsid protein (VP1) of noroviral GI and GII genotypes. Compared to intramuscular (i.m.), intranasal (i.n.), or other prime-boost immunization regimens (i.m. â€‹+ â€‹i.m., i.m. â€‹+ â€‹i.n., i.n. â€‹+ â€‹i.m.), AdC68-GI.1-GII.3 (E1)-GII.4-GII.17 (E3), administered via i.n. â€‹+ â€‹i.n. induced higher titers of serum IgG antibodies and higher IgA antibodies in bronchoalveolar lavage fluid (BALF) and saliva against the four homologous VP1s in mice. It also significantly stimulated the production of blocking antibodies against the four genotypes. In response to re-stimulation with virus-like particles (VLP)-GI.1, VLP-GII.3, VLP-GII.4, and VLP-GII.17, the quadrivalent vaccine administered according to the i.n. â€‹+ â€‹i.n. regimen effectively triggered specific cell-mediated immune responses, primarily characterized by IFN-γ secretion. Furthermore, the preparation of this novel quadrivalent NoV vaccine requires only a single recombinant adenovirus to provide broad preventive immunity against the major GI/GII epidemic strains, making it a promising vaccine candidate for further development.


Assuntos
Adenoviridae , Anticorpos Antivirais , Infecções por Caliciviridae , Vetores Genéticos , Camundongos Endogâmicos BALB C , Norovirus , Pan troglodytes , Vacinas Virais , Animais , Norovirus/imunologia , Norovirus/genética , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Camundongos , Infecções por Caliciviridae/prevenção & controle , Infecções por Caliciviridae/imunologia , Vacinas Virais/imunologia , Vacinas Virais/genética , Vacinas Virais/administração & dosagem , Vetores Genéticos/genética , Vetores Genéticos/imunologia , Adenoviridae/genética , Adenoviridae/imunologia , Feminino , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/imunologia , Imunoglobulina G/sangue , Gastroenterite/prevenção & controle , Gastroenterite/virologia , Gastroenterite/imunologia , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Imunoglobulina A/sangue , Genótipo , Saliva/imunologia , Saliva/virologia , Líquido da Lavagem Broncoalveolar/imunologia , Líquido da Lavagem Broncoalveolar/virologia
3.
Virulence ; 15(1): 2368080, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38899573

RESUMO

Dendritic cells (DCs) present an ideal target for delivering immunogenic cargo due to their potent antigen-presenting capabilities. This targeting approach holds promise in vaccine development by enhancing the efficiency of antigen recognition and capture by DCs. To identify a high-affinity targeting peptide binding to rabbit DCs, rabbit monocyte-derived DCs (raMoDCs) were isolated and cultured, and a novel peptide, HS (HSLRHDYGYPGH), was identified using a phage-displayed peptide library. Alongside HS, two other DC-targeting peptides, KC1 and MY, previously validated in our laboratory, were employed to construct recombinant Lactgobacillus reuteri fusion-expressed rabbit hemorrhagic disease virus (RHDV) capsid protein VP60. These recombinant Lactobacillus strains were named HS-VP60/L. reuteri, KC1-VP60/L. reuteri, and MY-VP60/L. reuteri. The ability of these recombinant Lactobacillus to bind rabbit DCs was evaluated both in vivo and in vitro. Results demonstrated that the DC-targeting peptide KC1 significantly enhanced the capture efficiency of recombinant Lactobacillus by raMoDCs, promoted DC maturation, and increased cytokine secretion. Furthermore, oral administration of KC1-VP60/L. reuteri effectively induced SIgA and IgG production in rabbits, prolonged rabbit survival post-challenge, and reduced RHDV copies in organs. In summary, the DC-targeting peptide KC1 exhibited robust binding to raMoDCs, and recombinant Lactobacillus expressing KC1-VP60 protein antigens efficiently induced systemic and mucosal immune responses in rabbits, conferring protective efficacy against RHDV. This study offers valuable insights for the development of novel RHDV vaccines.


Assuntos
Células Dendríticas , Vírus da Doença Hemorrágica de Coelhos , Limosilactobacillus reuteri , Peptídeos , Animais , Células Dendríticas/imunologia , Coelhos , Vírus da Doença Hemorrágica de Coelhos/imunologia , Vírus da Doença Hemorrágica de Coelhos/genética , Limosilactobacillus reuteri/genética , Limosilactobacillus reuteri/imunologia , Peptídeos/imunologia , Peptídeos/genética , Infecções por Caliciviridae/prevenção & controle , Infecções por Caliciviridae/imunologia , Infecções por Reoviridae/prevenção & controle , Infecções por Reoviridae/imunologia , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/imunologia , Vacinas Virais/imunologia , Vacinas Virais/genética , Lactobacillus/genética , Lactobacillus/imunologia
4.
Glycobiology ; 34(6)2024 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-38590172

RESUMO

Human noroviruses, globally the main cause of viral gastroenteritis, show strain specific affinity for histo-blood group antigens (HBGA) and can successfully be propagated ex vivo in human intestinal enteroids (HIEs). HIEs established from jejunal stem cells of individuals with different ABO, Lewis and secretor geno- and phenotypes, show varying susceptibility to such infections. Using bottom-up glycoproteomic approaches we have defined and compared the N-linked glycans of glycoproteins of seven jejunal HIEs. Membrane proteins were extracted, trypsin digested, and glycopeptides enriched by hydrophilic interaction liquid chromatography and analyzed by nanoLC-MS/MS. The Byonic software was used for glycopeptide identification followed by hands-on verifications and interpretations. Glycan structures and attachment sites were identified from MS2 spectra obtained by higher-energy collision dissociation through analysis of diagnostic saccharide oxonium ions (B-ions), stepwise glycosidic fragmentation of the glycans (Y-ions), and peptide sequence ions (b- and y-ions). Altogether 694 unique glycopeptides from 93 glycoproteins were identified. The N-glycans encompassed pauci- and oligomannose, hybrid- and complex-type structures. Notably, polyfucosylated HBGA-containing glycopeptides of the four glycoproteins tetraspanin-8, carcinoembryonic antigen-related cell adhesion molecule 5, sucrose-isomaltase and aminopeptidase N were especially prominent and were characterized in detail and related to donor ABO, Lewis and secretor types of each HIE. Virtually no sialylated N-glycans were identified for these glycoproteins suggesting that terminal sialylation was infrequent compared to fucosylation and HBGA biosynthesis. This approach gives unique site-specific information on the structural complexity of N-linked glycans of glycoproteins of human HIEs and provides a platform for future studies on the role of host glycoproteins in gastrointestinal infectious diseases.


Assuntos
Antígenos de Grupos Sanguíneos , Infecções por Caliciviridae , Fucose , Glicoproteínas , Antígenos de Histocompatibilidade , Jejuno , Organoides , Glicômica , Proteômica , Genótipo , Fenótipo , Glicoproteínas/química , Glicoproteínas/genética , Glicoproteínas/metabolismo , Fucose/metabolismo , Glicosilação , Antígenos de Grupos Sanguíneos/química , Antígenos de Grupos Sanguíneos/genética , Antígenos de Grupos Sanguíneos/metabolismo , Antígenos de Histocompatibilidade/química , Antígenos de Histocompatibilidade/genética , Antígenos de Histocompatibilidade/metabolismo , Humanos , Glicopeptídeos/química , Infecções por Caliciviridae/sangue , Infecções por Caliciviridae/imunologia , Infecções por Caliciviridae/metabolismo , Organoides/metabolismo , Jejuno/metabolismo , Jejuno/virologia
5.
Gut Microbes ; 15(2): 2249960, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37655966

RESUMO

Over 90% of epidemic non-bacterial gastroenteritis are caused by human noroviruses (NoVs), which persist in a substantial subset of people allowing their spread worldwide. This has led to a significant number of endemic cases and up to 70,000 children deaths in developing countries. NoVs are primarily transmitted through the fecal-oral route. To date, studies have focused on the influence of the gut microbiota on enteric viral clearance by mucosal immunity. In this study, the use of mouse norovirus S99 (MNoV_S99) and CR6 (MNoV_CR6), two persistent strains, allowed us to provide evidence that the norovirus-induced exacerbation of colitis severity relied on bacterial sensing by nucleotide-binding oligomerization domain 2 (Nod2). Consequently, Nod2-deficient mice showed reduced levels of gravity of Dextran sodium sulfate (DSS)-induced colitis with both viral strains. And MNoV_CR6 viremia was heightened in Nod2-/- mice in comparison with animals hypomorphic for Atg16l1, which are prone to aggravated inflammation under DSS. Accordingly, the infection of macrophages derived from WT mice promoted the phosphorylation of Signal Transducer and Activator of Transcription 1 (STAT1) and NOD2's expression levels. Higher secretion of Tumor Necrosis Factor alpha (TNFα) following NOD2 activation and better viral clearance were measured in these cells. By contrast, reduced levels of pSTAT1 and blunted downstream secretion of TNFα were found in Nod2-deficient macrophages infected by MNoV_S99. Hence, our results uncover a previously unidentified virus-host-bacterial interplay that may represent a novel therapeutic target for treating noroviral origin gastroenteritis that may be linked with susceptibility to several common illnesses such as Crohn's disease.


Assuntos
Infecções por Caliciviridae , Colite , Gastroenterite , Microbioma Gastrointestinal , Proteína Adaptadora de Sinalização NOD2 , Animais , Camundongos , Infecções por Caliciviridae/imunologia , Colite/induzido quimicamente , Colite/virologia , Gastroenterite/imunologia , Gastroenterite/virologia , Proteína Adaptadora de Sinalização NOD2/metabolismo
6.
J Virol ; 96(3): e0192321, 2022 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-34787460

RESUMO

Akt (protein kinase B) is a key signaling protein in eukaryotic cells that controls many cellular processes, such as glucose metabolism and cell proliferation, for survival. As obligate intracellular pathogens, viruses modulate host cellular processes, including Akt signaling, for optimal replication. The mechanisms by which viruses modulate Akt and the resulting effects on the infectious cycle differ widely depending on the virus. In this study, we explored the effect of Akt serine 473 phosphorylation (p-Akt) during murine norovirus (MNV) infection. p-Akt increased during infection of murine macrophages with acute MNV-1 and persistent CR3 and CR6 strains. Inhibition of Akt with MK2206, an inhibitor of all three isoforms of Akt (Akt1/2/3), reduced infectious virus progeny of all three virus strains. This reduction was due to decreased viral genome replication (CR3), defective virus assembly (MNV-1), or altered cellular egress (CR3 and CR6) in a virus strain-dependent manner. Collectively, our data demonstrate that Akt activation increases in macrophages during the later stages of the MNV infectious cycle, which may enhance viral infection in unique ways for different virus strains. The data, for the first time, indicate a role for Akt signaling in viral assembly and highlight additional phenotypic differences between closely related MNV strains. IMPORTANCE Human noroviruses (HNoV) are a leading cause of viral gastroenteritis, resulting in high annual economic burden and morbidity, yet there are no small-animal models supporting productive HNoV infection or robust culture systems producing cell culture-derived virus stocks. As a result, research on drug discovery and vaccine development against norovirus infection has been challenging, and no targeted antivirals or vaccines against HNoV are approved. On the other hand, murine norovirus (MNV) replicates to high titers in cell culture and is a convenient and widespread model in norovirus research. Our data demonstrate the importance of Akt signaling during the late stage of the MNV life cycle. Notably, the effect of Akt signaling on genome replication, virus assembly, and cellular egress is virus strain specific, highlighting the diversity of biological phenotypes despite small genetic variability among norovirus strains. This study is the first to demonstrate a role for Akt in viral assembly.


Assuntos
Infecções por Caliciviridae/metabolismo , Infecções por Caliciviridae/virologia , Macrófagos/metabolismo , Macrófagos/virologia , Norovirus/fisiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Replicação Viral , Animais , Infecções por Caliciviridae/imunologia , Suscetibilidade a Doenças , Interações Hospedeiro-Patógeno , Ativação de Macrófagos , Macrófagos/imunologia , Camundongos , Fosforilação , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Especificidade da Espécie
7.
Front Immunol ; 12: 781718, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34868056

RESUMO

Norovirus (NoV) is a zoonotic virus that causes diarrhea in humans and animals. Outbreaks in nosocomial settings occur annually worldwide, endangering public health and causing serious social and economic burdens. The latter quarter of 2016 witnessed the emergence of the GII.P16-GII.2 recombinant norovirus throughout Asia. This genotype exhibits strong infectivity and replication characteristics, proposing its potential to initiate a pandemic. There is no vaccine against GII.P16-GII.2 recombinant norovirus, so it is necessary to design a preventive vaccine. In this study, GII.P16-GII.2 type norovirus virus-like particles (VLPs) were constructed using the baculovirus expression system and used to conduct immunizations in mice. After immunization of mice, mice were induced to produce memory T cells and specific antibodies, indicating that the VLPs induced specific cellular and humoral immune responses. Further experiments were then initiated to understand the underlying mechanisms involved in antigen presentation. Towards this, we established co-cultures between dendritic cells (DCs) or macrophages (Mø) and naïve CD4+T cells and simulated the antigen presentation process by incubation with VLPs. Thereafter, we detected changes in cell surface molecules, cytokines and related proteins. The results indicated that VLPs effectively promoted the phenotypic maturation of Mø but not DCs, as indicated by significant changes in the expression of MHC-II, costimulatory factors and related cytokines in Mø. Moreover, we found VLPs caused Mø to polarize to the M1 type and release inflammatory cytokines, thereby inducing naïve CD4+ T cells to perform Th1 immune responses. Therefore, this study reveals the mechanism of antigen presentation involving GII.P16-GII.2 recombinant norovirus VLPs, providing a theoretical basis for both understanding responses to norovirus infection as well as opportunities for vaccine development.


Assuntos
Infecções por Caliciviridae/imunologia , Interações Hospedeiro-Patógeno/imunologia , Ativação de Macrófagos/imunologia , Macrófagos/imunologia , Norovirus/imunologia , Células Th1/imunologia , Vacinas de Partículas Semelhantes a Vírus/imunologia , Animais , Anticorpos Neutralizantes , Anticorpos Antivirais/imunologia , Especificidade de Anticorpos/imunologia , Apresentação de Antígeno , Antígenos Virais/genética , Antígenos Virais/imunologia , Infecções por Caliciviridae/prevenção & controle , Infecções por Caliciviridae/virologia , Modelos Animais de Doenças , Feminino , Humanos , Imunidade Celular , Macrófagos/metabolismo , Camundongos , Norovirus/classificação , Norovirus/genética , Proteínas Recombinantes , Células Th1/metabolismo , Vacinas de Partículas Semelhantes a Vírus/isolamento & purificação , Vacinas de Partículas Semelhantes a Vírus/ultraestrutura
8.
J Virol ; 95(20): e0113421, 2021 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-34346771

RESUMO

Murine norovirus (MNV) infection results in a late translation shutoff that is proposed to contribute to the attenuated and delayed innate immune response observed both in vitro and in vivo. Recently, we further demonstrated the activation of the α subunit of eukaryotic initiation factor 2 (eIF2α) kinase GCN2 during MNV infection, which has been previously linked to immunomodulation and resistance to inflammatory signaling during metabolic stress. While viral infection is usually associated with activation of double-stranded RNA (dsRNA) binding pattern recognition receptor PKR, we hypothesized that the establishment of a metabolic stress in infected cells is a proviral event, exploited by MNV to promote replication through weakening the activation of the innate immune response. In this study, we used multi-omics approaches to characterize cellular responses during MNV replication. We demonstrate the activation of pathways related to the integrated stress response, a known driver of anti-inflammatory phenotypes in macrophages. In particular, MNV infection causes an amino acid imbalance that is associated with GCN2 and ATF2 signaling. Importantly, this reprogramming lacks the features of a typical innate immune response, with the ATF/CHOP target GDF15 contributing to the lack of antiviral responses. We propose that MNV-induced metabolic stress supports the establishment of host tolerance to viral replication and propagation. IMPORTANCE During viral infection, host defenses are typically characterized by the secretion of proinflammatory autocrine and paracrine cytokines, potentiation of the interferon (IFN) response, and induction of the antiviral response via activation of JAK and Stat signaling. To avoid these and propagate, viruses have evolved strategies to evade or counteract host sensing. In this study, we demonstrate that murine norovirus controls the antiviral response by activating a metabolic stress response that activates the amino acid response and impairs inflammatory signaling. This highlights novel tools in the viral countermeasures arsenal and demonstrates the importance of the currently poorly understood metabolic reprogramming occurring during viral infections.


Assuntos
Infecções por Caliciviridae/imunologia , Macrófagos/virologia , Fator 2 Ativador da Transcrição/metabolismo , Animais , Antivirais , Infecções por Caliciviridae/metabolismo , Linhagem Celular , Fator de Iniciação 2 em Eucariotos/metabolismo , Imunidade Inata/imunologia , Inflamação/imunologia , Interferons , Macrófagos/imunologia , Camundongos , Norovirus/patogenicidade , Proteínas Serina-Treonina Quinases/metabolismo , Células RAW 264.7 , RNA de Cadeia Dupla/genética , Transdução de Sinais/imunologia , Proteínas não Estruturais Virais/metabolismo , Replicação Viral/genética
9.
Viruses ; 13(2)2021 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-33494515

RESUMO

Norovirus is the leading cause of acute gastroenteritis worldwide. The pathogenesis of norovirus and the induced immune response remain poorly understood due to the lack of a robust virus culture system. The monolayers of two secretor-positive Chinese human intestinal enteroid (HIE) lines were challenged with two norovirus pandemic GII.4 Sydney strains. Norovirus RNA replication in supernatants and cell lysates were quantified by RT-qPCR. RNA expression levels of immune-related genes were profiled using PCR arrays. The secreted protein levels of shortlisted upregulated genes were measured in supernatants using analyte-specific enzyme-linked immunosorbent assay (ELISA). Productive norovirus replications were achieved in three (75%) out of four inoculations. The two most upregulated immune-related genes were CXCL10 (93-folds) and IFI44L (580-folds). Gene expressions of CXCL10 and IFI44L were positively correlated with the level of norovirus RNA replication (CXCL10: Spearman's r = 0.779, p < 0.05; IFI44L: r = 0.881, p < 0.01). The higher level of secreted CXCL10 and IFI44L proteins confirmed their elevated gene expression. The two genes have been reported to be upregulated in norovirus volunteer challenges and natural human infections by other viruses. Our data suggested that HIE could mimic the innate immune response elicited in natural norovirus infection and, therefore, could serve as an experimental model for future virus-host interaction and antiviral studies.


Assuntos
Infecções por Caliciviridae/imunologia , Quimiocina CXCL10/metabolismo , Intestinos/virologia , Proteínas Supressoras de Tumor/metabolismo , Idoso , Idoso de 80 Anos ou mais , Linhagem Celular , Quimiocina CXCL10/genética , Feminino , Interações entre Hospedeiro e Microrganismos , Humanos , Imunidade Inata , Interferons/genética , Interferons/metabolismo , Intestinos/imunologia , Masculino , Pessoa de Meia-Idade , Modelos Biológicos , Norovirus/patogenicidade , Norovirus/fisiologia , Organoides/imunologia , Organoides/virologia , Análise de Sequência de RNA , Proteínas Supressoras de Tumor/genética , Replicação Viral
10.
J Virol ; 95(3)2021 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-33177207

RESUMO

Noroviruses are a leading cause of gastrointestinal infection in humans and mice. Understanding human norovirus (HuNoV) cell tropism has important implications for our understanding of viral pathogenesis. Murine norovirus (MNoV) is extensively used as a surrogate model for HuNoV. We previously identified CD300lf as the receptor for MNoV. Here, we generated a Cd300lf conditional knockout (CD300lfF/F ) mouse to elucidate the cell tropism of persistent and nonpersistent strains of murine norovirus. Using this mouse model, we demonstrated that CD300lf expression on intestinal epithelial cells (IECs), and on tuft cells in particular, is essential for transmission of the persistent MNoV strain CR6 (MNoVCR6) in vivo In contrast, the nonpersistent MNoV strain CW3 (MNoVCW3) does not require CD300lf expression on IECs for infection. However, deletion of CD300lf in myelomonocytic cells (LysM Cre+) partially reduces CW3 viral load in lymphoid and intestinal tissues. Disruption of CD300lf expression on B cells (CD19 Cre), neutrophils (Mrp8 Cre), and dendritic cells (CD11c Cre) did not affect MNoVCW3 viral RNA levels. Finally, we show that the transcription factor STAT1, which is critical for the innate immune response, partially restricts the cell tropism of MNoVCW3 to LysM+ cells. Taken together, these data demonstrate that CD300lf expression on tuft cells is essential for MNoVCR6; that myelomonocytic cells are a major, but not exclusive, target cell of MNoVCW3; and that STAT1 signaling restricts the cellular tropism of MNoVCW3 This study provides the first genetic system for studying the cell type-specific role of CD300lf in norovirus pathogenesis.IMPORTANCE Human noroviruses (HuNoVs) are a leading cause of gastroenteritis resulting in up to 200,000 deaths each year. The receptor and cell tropism of HuNoV in immunocompetent humans are unclear. We use murine norovirus (MNoV) as a model for HuNoV. We recently identified CD300lf as the sole physiologic receptor for MNoV. Here, we leverage this finding to generate a Cd300lf conditional knockout mouse to decipher the contributions of specific cell types to MNoV infection. We demonstrate that persistent MNoVCR6 requires CD300lf expression on tuft cells. In contrast, multiple CD300lf+ cell types, dominated by myelomonocytic cells, are sufficient for nonpersistent MNoVCW3 infection. CD300lf expression on epithelial cells, B cells, neutrophils, and dendritic cells is not critical for MNoVCW3 infection. Mortality associated with the MNoVCW3 strain in Stat1-/- mice does not require CD300lf expression on LysM+ cells, highlighting that both CD300lf receptor expression and innate immunity regulate MNoV cell tropism in vivo.


Assuntos
Células Epiteliais/imunologia , Interações Hospedeiro-Patógeno , Imunidade Inata/imunologia , Intestinos/imunologia , Norovirus/fisiologia , Receptores Imunológicos/fisiologia , Tropismo Viral , Animais , Infecções por Caliciviridae/imunologia , Infecções por Caliciviridae/metabolismo , Infecções por Caliciviridae/virologia , Células Epiteliais/virologia , Feminino , Intestinos/virologia , Masculino , Camundongos , Camundongos Knockout
11.
Sheng Wu Gong Cheng Xue Bao ; 36(8): 1536-1545, 2020 Aug 25.
Artigo em Chinês | MEDLINE | ID: mdl-32924352

RESUMO

Moschus chrysogaster (sifanicus) viral hemorrhagic disease (McVHD) is an acute and highly lethal infectious disease caused by Moschus chrysogaster hemorrhagic disease virus (McHDV) whose genome sequence is highly homologous with rabbit hemorrhagic disease virus. To screen the protective antigen of McHDV and set the basis for study of McVHD vaccine, the antigen epitope of major structural protein VP60 of McHDV was analyzed, and the specific primers were designed to obtain three amplified DNA sequences encoding the main antigen epitope of VP60 from McHDV by using RT-PCR. Then the three DNA fragments were sequenced and cloned to prokaryotic expression vector with pET-28a(+) by using overlap extension PCR, and finally the prokaryotic expression plasmid pET-truncated-VP60 was constructed. Subsequently, the pET-truncated-VP60 was transformed into Escherichia coli BL21(DE3), and the recombinant proteins were expressed by IPTG induction. Finally, the expressed protein was purified and applied to immunize that without immunizing with RHD vaccine, then the antiserum titers were evaluated by the hemagglutination inhibition test, and the immune-protective efficacy of the recombinant proteins was observed and analyzed through animal challenge test. The results showed that the multi-epitope DNA fragments of VP60 of McHDV was successfully expressed in the form of inclusion bodies in E. coli, and the relative molecular weight of recombinant proteins is about 45 kDa. After immunized with the recombinant proteins, 100% of New Zealand white rabbits were resistant to attack of McHDV, which indicates efficient immune-protective efficacy of chosen epitope recombinant protein. The study laid a foundation for the development of the new subunit vaccines of McVHD.


Assuntos
Infecções por Caliciviridae , Expressão Gênica , Vírus da Doença Hemorrágica de Coelhos , Proteínas Estruturais Virais , Animais , Infecções por Caliciviridae/imunologia , Infecções por Caliciviridae/virologia , Epitopos/genética , Escherichia coli/genética , Coelhos , Proteínas Estruturais Virais/genética , Proteínas Estruturais Virais/metabolismo
12.
Virology ; 546: 109-121, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32452409

RESUMO

The inflammasome machinery has recently been recognized as an emerging pillar of innate immunity. However, little is known regarding the interaction between the classical interferon (IFN) response and inflammasome activation in response to norovirus infection. We found that murine norovirus (MNV-1) infection induces the transcription of IL-1ß, a hallmark of inflammasome activation, which is further increased by inhibition of IFN response, but fails to trigger the release of mature IL-1ß. Interestingly, pharmacological inflammasome inhibitors do not affect viral replication, but slightly reverse the inflammasome activator lipopolysaccharide (LPS)-mediated inhibition of MNV replication. LPS efficiently stimulates the transcription of IFN-ß through NF-ĸB, which requires the transcription factors IRF3 and IRF7. This activates downstream antiviral IFN-stimulated genes (ISGs) via the JAK-STAT pathway. Moreover, inhibition of NF-ĸB and JAK-STAT signaling partially reverse LPS-mediated anti-MNV activity, suggesting additional antiviral mechanisms activated by NF-ĸB. This study reveals additional insight in host defense against MNV infection.


Assuntos
Antivirais/farmacologia , Infecções por Caliciviridae/imunologia , Janus Quinases/imunologia , Lipopolissacarídeos/farmacologia , Macrófagos/imunologia , NF-kappa B/imunologia , Norovirus/efeitos dos fármacos , Fatores de Transcrição STAT/imunologia , Animais , Infecções por Caliciviridae/genética , Infecções por Caliciviridae/virologia , Humanos , Inflamassomos/genética , Inflamassomos/imunologia , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/imunologia , Janus Quinases/genética , Macrófagos/virologia , Camundongos , NF-kappa B/genética , Norovirus/genética , Norovirus/fisiologia , Fatores de Transcrição STAT/genética , Transdução de Sinais/efeitos dos fármacos
13.
J Immunol Res ; 2020: 3194704, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32411793

RESUMO

We have previously shown that rotavirus (RV) inner capsid protein VP6 has an adjuvant effect on norovirus (NoV) virus-like particle- (VLP-) induced immune responses and studied the adjuvant mechanism in immortalized cell lines used as antigen-presenting cells (APCs). Here, we investigated the uptake and presentation of RV VP6 and NoV GII.4 VLPs by primary bone marrow-derived dendritic cells (BMDCs). The adjuvant effect of VP6 on GII.4 VLP presentation and NoV-specific immune response induction by BMDC in vivo was also studied. Intracellular staining demonstrated that BMDCs internalized both antigens, but VP6 more efficiently than NoV VLPs. Both antigens were processed and presented to antigen-primed T cells, which responded by robust interferon γ secretion. When GII.4 VLPs and VP6 were mixed in the same pulsing reaction, a subpopulation of the cells had uptaken both antigens. Furthermore, VP6 copulsing increased GII.4 VLP uptake by 37% and activated BMDCs to secrete 2-5-fold increased levels of interleukin 6 and tumor necrosis factor α compared to VLP pulsing alone. When in vitro-pulsed BMDCs were transferred to syngeneic BALB/c mice, VP6 improved NoV-specific antibody responses. The results of this study support the earlier findings of VP6 adjuvant effect in vitro and in vivo.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Antígenos Virais/administração & dosagem , Infecções por Caliciviridae/prevenção & controle , Proteínas do Capsídeo/administração & dosagem , Células Dendríticas/imunologia , Norovirus/imunologia , Vacinas Virais/administração & dosagem , Animais , Antígenos Virais/imunologia , Infecções por Caliciviridae/imunologia , Infecções por Caliciviridae/virologia , Proteínas do Capsídeo/imunologia , Células Cultivadas , Reações Cruzadas , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Imunogenicidade da Vacina , Interleucina-6/metabolismo , Camundongos , Cultura Primária de Células , Fator de Necrose Tumoral alfa/metabolismo , Vacinas Virais/imunologia
14.
Cell Mol Gastroenterol Hepatol ; 10(2): 245-267, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32289501

RESUMO

BACKGROUND & AIMS: Human norovirus infection is the leading cause of acute gastroenteritis. Genetic polymorphisms, mediated by the FUT2 gene (secretor enzyme), define strain susceptibility. Secretors express a diverse set of fucosylated histoblood group antigen carbohydrates (HBGA) on mucosal cells; nonsecretors (FUT2-/-) express a limited array of HBGAs. Thus, nonsecretors have less diverse norovirus strain infections, including resistance to the epidemiologically dominant GII.4 strains. Because future human norovirus vaccines will comprise GII.4 antigen and because secretor phenotype impacts GII.4 infection and immunity, nonsecretors may mimic young children immunologically in response to GII.4 vaccination, providing a needed model to study cross-protection in the context of limited pre-exposure. METHODS: By using specimens collected from the first characterized nonsecretor cohort naturally infected with GII.2 human norovirus, we evaluated the breadth of serologic immunity by surrogate neutralization assays, and cellular activation and cytokine production by flow cytometry. RESULTS: GII.2 infection resulted in broad antibody and cellular immunity activation that persisted for at least 30 days for T cells, monocytes, and dendritic cells, and for 180 days for blocking antibody. Multiple cellular lineages expressing interferon-γ and tumor necrosis factor-α dominated the response. Both T-cell and B-cell responses were cross-reactive with other GII strains, but not GI strains. To promote entry mechanisms, inclusion of bile acids was essential for GII.2 binding to nonsecretor HBGAs. CONCLUSIONS: These data support development of within-genogroup, cross-reactive antibody and T-cell immunity, key outcomes that may provide the foundation for eliciting broad immune responses after GII.4 vaccination in individuals with limited GII.4 immunity, including young children.


Assuntos
Infecções por Caliciviridae/imunologia , Resistência à Doença/genética , Gastroenterite/imunologia , Interações entre Hospedeiro e Microrganismos/genética , Norovirus/imunologia , Adulto , Antígenos Virais/imunologia , Antígenos Virais/metabolismo , Antígenos de Grupos Sanguíneos/metabolismo , Infecções por Caliciviridae/sangue , Infecções por Caliciviridae/genética , Infecções por Caliciviridae/virologia , Estudos de Coortes , Reações Cruzadas , Feminino , Fucosiltransferases/genética , Fucosiltransferases/metabolismo , Gastroenterite/sangue , Gastroenterite/genética , Gastroenterite/virologia , Interações entre Hospedeiro e Microrganismos/imunologia , Humanos , Imunidade Celular/genética , Masculino , Pessoa de Meia-Idade , Norovirus/genética , Norovirus/isolamento & purificação , Norovirus/patogenicidade , Linfócitos T/imunologia , Adulto Jovem , Galactosídeo 2-alfa-L-Fucosiltransferase
15.
J Virol ; 94(10)2020 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-32161178

RESUMO

The rabbit hemorrhagic disease virus (RHDV), which belongs to the family Caliciviridae and the genus Lagovirus, causes lethal fulminant hepatitis in rabbits. RHDV decreases the activity of antioxidant enzymes regulated by Nrf2 in the liver. Antioxidants are important for the maintenance of cellular integrity and cytoprotection. However, the mechanism underlying the regulation of the Nrf2-antioxidant response element (ARE) signaling pathway by RHDV remains unclear. Using isobaric tags for relative and absolute quantification (iTRAQ) technology, the current study demonstrated that RHDV inhibits the induction of ARE-regulated genes and increases the expression of the p50 subunit of the NF-κB transcription factor. We showed that RHDV replication causes a remarkable increase in reactive oxygen species (ROS), which is simultaneously accompanied by a significant decrease in Nrf2. It was found that nuclear translocation of Keap1 plays a key role in the nuclear export of Nrf2, leading to the inhibition of Nrf2 transcriptional activity. The p50 protein partners with Keap1 to form the Keap1-p50/p65 complex, which is involved in the nuclear translocation of Keap1. Moreover, upregulation of Nrf2 protein levels in liver cell nuclei by tert-butylhydroquinone (tBHQ) delayed rabbit deaths due to RHDV infection. Considered together, our findings suggest that RHDV inhibits the Nrf2-dependent antioxidant response via nuclear translocation of Keap1-NF-κB complex and nuclear export of Nrf2 and provide new insight into the importance of oxidative stress during RHDV infection.IMPORTANCE Recent studies have reported that rabbit hemorrhagic disease virus (RHDV) infection reduced Nrf2-related antioxidant function. However, the regulatory mechanisms underlying this process remain unclear. The current study showed that the NF-κB p50 subunit partners with Keap1 to form the Keap1-NF-κB complex, which plays a key role in the inhibition of Nrf2 transcriptional activity. More importantly, upregulated Nrf2 activity delayed the death of RHDV-infected rabbits, strongly indicating the importance of oxidative damage during RHDV infection. These findings may provide novel insights into the pathogenesis of RHDV.


Assuntos
Antioxidantes/metabolismo , Infecções por Caliciviridae/metabolismo , Vírus da Doença Hemorrágica de Coelhos/imunologia , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , NF-kappa B/metabolismo , Animais , Elementos de Resposta Antioxidante , Antioxidantes/farmacologia , Infecções por Caliciviridae/imunologia , Infecções por Caliciviridae/patologia , Núcleo Celular/metabolismo , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica , Células HEK293 , Vírus da Doença Hemorrágica de Coelhos/patogenicidade , Humanos , Hidroquinonas , Proteína 1 Associada a ECH Semelhante a Kelch/genética , Fígado/lesões , Fígado/metabolismo , Fígado/patologia , Fator 2 Relacionado a NF-E2/genética , Estresse Oxidativo , Proteômica , Coelhos , Transdução de Sinais/efeitos dos fármacos , Fator de Transcrição RelA , Replicação Viral
16.
Biotechnol Lett ; 42(7): 1211-1218, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32088791

RESUMO

OBJECTIVES: Noroviruses (NoVs) are major cause of acute viral gastroenteritis in worldwide, and the lack of a cell culture system that must be considered the virus like particles (VLPs) are used as an effective vaccine development. MATERIALS AND METHODS: In the present study, we investigated the expression of the major capsid protein (VP1) of the Genogroup II, genotype 17 (GII.17) NoV, using recombinant baculovirus system in insect cells, as well as a saliva binding blockade assay to detect their protective potency. RESULTS: Our results showed that GII.17 VLPs could be successfully generated in sf9 insect cells, and electron microscopic revealed that GII.17 VLPs appeared as spherical particles with a - 35 nm diameter. Immunized mice with purified VLPs produced GII.17 specific sera and could efficiently block GII.17 VLPs binding to the saliva histo-blood group antigens (HBGAs). CONCLUSIONS: Together, these results suggested that GII.17 VLPs represent a promising vaccine candidate against NoV GII.17 infection and strongly support further preclinical and clinical studies.


Assuntos
Anticorpos Antivirais/imunologia , Infecções por Caliciviridae/imunologia , Norovirus/imunologia , Proteínas Recombinantes/imunologia , Vacinas de Partículas Semelhantes a Vírus/imunologia , Animais , Anticorpos Antivirais/sangue , Anticorpos Antivirais/metabolismo , Antígenos de Grupos Sanguíneos/metabolismo , Infecções por Caliciviridae/prevenção & controle , Modelos Animais de Doenças , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Norovirus/genética , Coelhos , Proteínas Recombinantes/genética , Saliva/química , Células Sf9 , Vacinas de Partículas Semelhantes a Vírus/genética , Proteínas do Core Viral/genética , Proteínas do Core Viral/imunologia
17.
J Feline Med Surg ; 22(6): 602-607, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31411533

RESUMO

OBJECTIVES: Feline calicivirus (FCV) is a highly variable and globally important feline pathogen for which vaccination has been the mainstay of control. Here, we test whether the continued use of FCV-F9, one of the most frequently used vaccine strains globally, is driving the emergence of vaccine-resistant viruses in the field. METHODS: This study made use of two representative panels of field isolates previously collected from cats visiting randomly selected veterinary practices across the UK as part of separate cross-sectional studies from 2001 and 2013/2014. Phylogenetic analysis and in vitro virus neutralisation tests were used to compare the genetic and antigenic relationships between these populations and FCV-F9. RESULTS: Phylogenetic analysis showed a typically radial distribution dominated by 52 distinct strains, with strains from both 2001 and 2013/2014 intermingled. The sequence for FCV-F9 appeared to be integral to this phylogeny and there were no significant differences in the genetic distances within each studied population (intra-population distances), or between them (inter-population distances), or between each population and FCV-F9. A 1 in 8 dilution neutralised 97% and 100% of the 2001 and 2013/14 isolates, respectively, and a 1 in 16 dilution neutralised 87% and 75% of isolates, respectively. There was no significant difference either in variance between the FCV-F9 neutralising titres for the two populations, or in the distribution of neutralisation titres across the two populations. CONCLUSIONS AND RELEVANCE: Although FCV is a highly variable virus, we found no evidence for a progressive divergence of field virus from vaccine strain FCV-F9, either phylogenetically or antigenically, with FCV-F9 antisera remaining broadly and equally cross-reactive to two geographically representative and temporally separated FCV populations. We suggest this may be because the immunodominant region of the FCV capsid responsible for neutralisation may have structural constraints preventing its longer term progressive antigenic evolution.


Assuntos
Infecções por Caliciviridae/veterinária , Calicivirus Felino/classificação , Doenças do Gato/imunologia , Doenças do Gato/prevenção & controle , Soros Imunes/imunologia , Vacinação/veterinária , Vacinas Virais/imunologia , Animais , Infecções por Caliciviridae/imunologia , Infecções por Caliciviridae/prevenção & controle , Infecções por Caliciviridae/virologia , Calicivirus Felino/imunologia , Doenças do Gato/virologia , Gatos , Reino Unido
18.
Nat Immunol ; 20(12): 1681-1691, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31636462

RESUMO

Much attention has focused on commensal bacteria in health and disease, but the role of commensal viruses is understudied. Although metagenomic analysis shows that the intestine of healthy humans and animals harbors various commensal viruses and the dysbiosis of these viruses can be associated with inflammatory diseases, there is still a lack of causal data and underlying mechanisms to understand the physiological role of commensal viruses in intestinal homeostasis. In the present study, we show that commensal viruses are essential for the homeostasis of intestinal intraepithelial lymphocytes (IELs). Mechanistically, the cytosolic viral RNA-sensing receptor RIG-I in antigen-presenting cells can recognize commensal viruses and maintain IELs via a type I interferon-independent, but MAVS-IRF1-IL-15 axis-dependent, manner. The recovery of IELs by interleukin-15 administration reverses the susceptibility of commensal virus-depleted mice to dextran sulfate sodium-induced colitis. Collectively, our results indicate that commensal viruses maintain the IELs and consequently sustain intestinal homeostasis via noncanonical RIG-I signaling.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Infecções por Caliciviridae/imunologia , Colite/imunologia , Proteína DEAD-box 58/metabolismo , Intestinos/imunologia , Linfócitos Intraepiteliais/imunologia , Norovirus/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Infecções por Caliciviridae/virologia , Células Cultivadas , Colite/induzido quimicamente , Colite/virologia , Proteína DEAD-box 58/genética , Sulfato de Dextrana , Suscetibilidade a Doenças , Homeostase , Fator Regulador 1 de Interferon/genética , Fator Regulador 1 de Interferon/metabolismo , Interleucina-15/metabolismo , Intestinos/virologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais , Simbiose/imunologia
19.
Vet Microbiol ; 238: 108429, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31648721

RESUMO

Viral haemorrhagic disease (VHD) and colibacillosis are common diseases in rabbits that cause economic losses worldwide. The effect of colibacillosis on the immune response of vaccinated rabbits against rabbit haemorrhagic disease virus (RHDV) was studied. Four groups (G1-G4) were included. G1 was the negative control group; G2 was the RHDV vaccine group; G3 was the E. coli-infected group; and G4 was the E. coli-infected + RHDV vaccine group. The E. coli infection and RHDV vaccination were simultaneously performed, with another previous infection, 3 days before vaccination. At 28 days post-vaccination (PV), the rabbits (G2-G4) were challenged intramuscularly with 0.5 ml of RHDV at a dose of 103 50% median lethal dose (LD50)/rabbit. The rabbits were observed for clinical signs, body weight gain and mortality rates. Tissue, blood, serum, and faecal samples and rectal swabs were collected at 3, 5, 7, 14, 21 and 28 days PV. Significant clinical signs and mortality and a decrease in BW were observed in the infected + RHDV vaccine group. On the 3rd day post-infection (PI), compared with all the other groups, the vaccinated group (G2) had significantly upregulated hepatic tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) levels; however, the infected + RHDV vaccine group had significantly higher intestinal levels of TNF-α and IL-6 than the other groups. Furthermore, E. coli infection in vaccinated rabbits led to immunosuppression, as shown by significant decreases (P < 0.05) in heterophil phagocytic activity, the CD4+/CD8+ ratio, and HI antibody responses to RHDV and a significant increase in the heterophil to lymphocyte (H/L) ratio. In conclusion, colibacillosis leads to immunosuppression involving a shift in the equilibrium of cytokines and reduced weight gain and mortality in vaccinated rabbits and could be a contributing factor in RHDV vaccination failure in rabbit farming.


Assuntos
Infecções por Caliciviridae/veterinária , Infecções por Escherichia coli/veterinária , Coelhos/imunologia , Vacinação/veterinária , Vacinas Virais/imunologia , Animais , Infecções por Caliciviridae/imunologia , Infecções por Caliciviridae/mortalidade , Citocinas/genética , Infecções por Escherichia coli/imunologia , Infecções por Escherichia coli/mortalidade , Infecções por Escherichia coli/fisiopatologia , Regulação da Expressão Gênica/imunologia , Vírus da Doença Hemorrágica de Coelhos/imunologia , Coelhos/microbiologia , Coelhos/virologia , Vacinação/normas
20.
Curr Opin Virol ; 37: 16-25, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31163292

RESUMO

The co-evolution of the microbiota and immune system has forged a mutually beneficial relationship. This relationship allows the host to maintain the balance between active immunity to pathogens and vaccines and tolerance to self-antigens and food antigens. In children living in low-income and middle-income countries, undernourishment and repetitive gastrointestinal infections are associated with the failure of oral vaccines. Intestinal dysbiosis associated with these environmental influences, as well as some host-related factors, compromises immune responses and negatively impacts vaccine efficacy. To understand how immune responses to viral vaccines can be optimally modulated, mechanistic studies of the relationship between the microbiome, host genetics, viral infections and the development and function of the immune system are needed. We discuss the potential role of the microbiome in modulating vaccine responses in the context of a growing understanding of the relationship between the gastrointestinal microbiota, host related factors (including histo-blood group antigens) and resident immune cell populations.


Assuntos
Imunidade Adaptativa , Disbiose , Microbioma Gastrointestinal/imunologia , Interações Microbianas , Vacinas Virais/imunologia , Sistema ABO de Grupos Sanguíneos , Animais , Infecções por Caliciviridae/sangue , Infecções por Caliciviridae/imunologia , Infecções por Caliciviridae/prevenção & controle , Dieta , Disbiose/imunologia , Disbiose/microbiologia , Disbiose/virologia , Interações entre Hospedeiro e Microrganismos/genética , Humanos , Sistema Imunitário/microbiologia , Sistema Imunitário/virologia , Imunidade Inata , Antígenos do Grupo Sanguíneo de Lewis , Probióticos , Infecções por Rotavirus/sangue , Infecções por Rotavirus/imunologia , Infecções por Rotavirus/prevenção & controle , Viroses/sangue , Viroses/imunologia , Viroses/prevenção & controle
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA