Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Immunity ; 57(7): 1549-1566.e8, 2024 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-38776917

RESUMO

The activities, ontogeny, and mechanisms of lineage expansion of eosinophils are less well resolved than those of other immune cells, despite the use of biological therapies targeting the eosinophilia-promoting cytokine interleukin (IL)-5 or its receptor, IL-5Rα. We combined single-cell proteomics and transcriptomics and generated transgenic IL-5Rα reporter mice to revisit eosinophilopoiesis. We reconciled human and murine eosinophilopoiesis and provided extensive cell-surface immunophenotyping and transcriptomes at different stages along the continuum of eosinophil maturation. We used these resources to show that IL-5 promoted eosinophil-lineage expansion via transit amplification, while its deletion or neutralization did not compromise eosinophil maturation. Informed from our resources, we also showed that interferon response factor-8, considered an essential promoter of myelopoiesis, was not intrinsically required for eosinophilopoiesis. This work hence provides resources, methods, and insights for understanding eosinophil ontogeny, the effects of current precision therapeutics, and the regulation of eosinophil development and numbers in health and disease.


Assuntos
Linhagem da Célula , Eosinófilos , Interleucina-5 , Camundongos Transgênicos , Proteômica , Análise de Célula Única , Transcriptoma , Eosinófilos/imunologia , Eosinófilos/metabolismo , Animais , Interleucina-5/metabolismo , Interleucina-5/genética , Humanos , Camundongos , Proteômica/métodos , Análise de Célula Única/métodos , Diferenciação Celular/imunologia , Camundongos Endogâmicos C57BL , Perfilação da Expressão Gênica/métodos , Subunidade alfa de Receptor de Interleucina-5/metabolismo , Subunidade alfa de Receptor de Interleucina-5/genética , Mielopoese/genética , Fatores Reguladores de Interferon/metabolismo , Fatores Reguladores de Interferon/genética , Camundongos Knockout
2.
EMBO J ; 43(18): 3916-3947, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39085648

RESUMO

Intestinal tuft cells are critical for anti-helminth parasite immunity because they produce IL-25, which triggers IL-13 secretion by activated group 2 innate lymphoid cells (ILC2s) to expand both goblet and tuft cells. We show that epithelial Elp3, a tRNA-modifying enzyme, promotes tuft cell differentiation and is consequently critical for IL-25 production, ILC2 activation, goblet cell expansion and control of Nippostrongylus brasiliensis helminth infection in mice. Elp3 is essential for the generation of intestinal immature tuft cells and for the IL-13-dependent induction of glycolytic enzymes such as Hexokinase 1 and Aldolase A. Importantly, loss of epithelial Elp3 in the intestine blocks the codon-dependent translation of the Gator1 subunit Nprl2, an mTORC1 inhibitor, which consequently enhances mTORC1 activation and stabilizes Atf4 in progenitor cells. Likewise, Atf4 overexpression in mouse intestinal epithelium blocks tuft cell differentiation in response to intestinal helminth infection. Collectively, our data define Atf4 as a negative regulator of tuft cells and provide insights into promotion of intestinal type 2 immune response to parasites through tRNA modifications.


Assuntos
Fator 4 Ativador da Transcrição , Diferenciação Celular , Mucosa Intestinal , Alvo Mecanístico do Complexo 1 de Rapamicina , Animais , Camundongos , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Fator 4 Ativador da Transcrição/metabolismo , Fator 4 Ativador da Transcrição/genética , Mucosa Intestinal/metabolismo , Mucosa Intestinal/parasitologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/citologia , Nippostrongylus/imunologia , Células Caliciformes/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Interleucina-13/metabolismo , Interleucina-13/genética
3.
Proc Natl Acad Sci U S A ; 121(32): e2404536121, 2024 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-39088396

RESUMO

Alcelaphine gammaherpesvirus 1 (AlHV-1) asymptomatically persists in its natural host, the wildebeest. However, cross-species transmission to cattle results in the induction of an acute and lethal peripheral T cell lymphoma-like disease (PTCL), named malignant catarrhal fever (MCF). Our previous findings demonstrated an essential role for viral genome maintenance in infected CD8+ T lymphocytes but the exact mechanism(s) leading to lymphoproliferation and MCF remained unknown. To decipher how AlHV-1 dysregulates T lymphocytes, we first examined the global phenotypic changes in circulating CD8+ T cells after experimental infection of calves. T cell receptor repertoire together with transcriptomics and epigenomics analyses demonstrated an oligoclonal expansion of infected CD8+ T cells displaying effector and exhaustion gene signatures, including GZMA, GNLY, PD-1, and TOX2 expression. Then, among viral genes expressed in infected CD8+ T cells, we uncovered A10 that encodes a transmembrane signaling protein displaying multiple tyrosine residues, with predicted ITAM and SH3 motifs. Impaired A10 expression did not affect AlHV-1 replication in vitro but rendered AlHV-1 unable to induce MCF. Furthermore, A10 was phosphorylated in T lymphocytes in vitro and affected T cell signaling. Finally, while AlHV-1 mutants expressing mutated forms of A10 devoid of ITAM or SH3 motifs (or both) were able to induce MCF, a recombinant virus expressing a mutated form of A10 unable to phosphorylate its tyrosine residues resulted in the lack of MCF and protected against a wild-type virus challenge. Thus, we could characterize the nature of this γ-herpesvirus-induced PTCL-like disease and identify an essential mechanism explaining its development.


Assuntos
Linfócitos T CD8-Positivos , Gammaherpesvirinae , Animais , Linfócitos T CD8-Positivos/imunologia , Gammaherpesvirinae/genética , Gammaherpesvirinae/imunologia , Bovinos , Febre Catarral Maligna/virologia , Febre Catarral Maligna/imunologia , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/virologia
4.
PLoS Pathog ; 16(3): e1008405, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32176737

RESUMO

Alcelaphine herpesvirus 1 (AlHV-1) is a gammaherpesvirus that is carried asymptomatically by wildebeest. Upon cross-species transmission to other ruminants, including domestic cattle, AlHV-1 induces malignant catarrhal fever (MCF), which is a fatal lymphoproliferative disease resulting from proliferation and uncontrolled activation of latently infected CD8+ T cells. Two laboratory strains of AlHV-1 are used commonly in research: C500, which is pathogenic, and WC11, which has been attenuated by long-term maintenance in cell culture. The published genome sequence of a WC11 seed stock from a German laboratory revealed the deletion of two major regions. The sequence of a WC11 seed stock used in our laboratory also bears these deletions and, in addition, the duplication of an internal sequence in the terminal region. The larger of the two deletions has resulted in the absence of gene A7 and a large portion of gene A8. These genes are positional orthologs of the Epstein-Barr virus genes encoding envelope glycoproteins gp42 and gp350, respectively, which are involved in viral propagation and switching of cell tropism. To investigate the degree to which the absence of A7 and A8 participates in WC11 attenuation, recombinant viruses lacking these individual functions were generated in C500. Using bovine nasal turbinate and embryonic lung cell lines, increased cell-free viral propagation and impaired syncytia formation were observed in the absence of A7, whereas cell-free viral spread was inhibited in the absence of A8. Therefore, A7 appears to be involved in cell-to-cell viral spread, and A8 in viral cell-free propagation. Finally, infection of rabbits with either mutant did not induce the signs of MCF or the expansion of infected CD8+ T cells. These results demonstrate that A7 and A8 are both essential for regulating viral spread and suggest that AlHV-1 requires both genes to efficiently spread in vivo and reach CD8+ T lymphocytes and induce MCF.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Gammaherpesvirinae/imunologia , Genes Virais/imunologia , Febre Catarral Maligna/imunologia , Proteínas do Envelope Viral/imunologia , Animais , Bovinos , Linhagem Celular , Gammaherpesvirinae/genética , Febre Catarral Maligna/genética , Coelhos , Proteínas do Envelope Viral/genética
5.
EMBO J ; 36(16): 2404-2418, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28716804

RESUMO

Type 2 inflammation is a defining feature of infection with parasitic worms (helminths), as well as being responsible for widespread suffering in allergies. However, the precise mechanisms involved in T helper (Th) 2 polarization by dendritic cells (DCs) are currently unclear. We have identified a previously unrecognized role for type I IFN (IFN-I) in enabling this process. An IFN-I signature was evident in DCs responding to the helminth Schistosoma mansoni or the allergen house dust mite (HDM). Further, IFN-I signaling was required for optimal DC phenotypic activation in response to helminth antigen (Ag), and efficient migration to, and localization with, T cells in the draining lymph node (dLN). Importantly, DCs generated from Ifnar1-/- mice were incapable of initiating Th2 responses in vivo These data demonstrate for the first time that the influence of IFN-I is not limited to antiviral or bacterial settings but also has a central role to play in DC initiation of Th2 responses.


Assuntos
Células Dendríticas/imunologia , Interferon Tipo I/metabolismo , Células Th2/imunologia , Alérgenos/imunologia , Animais , Camundongos , Camundongos Knockout , Pyroglyphidae/imunologia , Receptor de Interferon alfa e beta/deficiência , Schistosoma mansoni/imunologia
6.
Eur J Immunol ; 49(7): 1067-1081, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30919955

RESUMO

Alternatively activated Mφs (AAMφ) accumulate in hepatic granulomas during schistosomiasis and have been suggested to originate in the bone marrow. What is less understood is how these Mφ responses are regulated after S. mansoni infection. Here, we investigated the role of IL-4 receptor α-chain (IL-4Rα)-signalling in the dynamics of liver Mφ responses. We observed that IL-4Rα signalling was dispensable for the recruitment of Ly6Chi monocytes and for their conversion into F4/80hi CD64hi CD11bhi Mφ. Moreover, while IL-4Rα provided an AAMφ phenotype to liver F4/80hi CD64hi CD11bhi Mφ that was associated with regulation of granuloma formation, it was dispensable for host survival. Resident F4/80hi CD64hi CD11blo Mφ did not upregulate the AAMφ signature gene Ym1. Rather, resident Mφ nearly disappeared by week 8 after infection and artificial ablation of resident Mφ in CD169DTR mice did not affect the response to S. mansoni infection. Interestingly, ablation of CD169+ cells in naive mice resulted in the accumulation of F4/80hi CD64hi CD11bhi Mφ, which was amplified when ablation occurred during schistosomiasis. Altogether, our results suggest the ablation of resident KCs after S. mansoni infection to be associated with the recruitment and accumulation of F4/80hi CD64hi CD11bhi Mφ with lyz2-dependent IL-4Rα contributing to the regulation of granuloma inflammation but being dispensable for host survival.


Assuntos
Granuloma/imunologia , Células de Kupffer/imunologia , Fígado/patologia , Macrófagos/imunologia , Receptores de Superfície Celular/metabolismo , Schistosoma mansoni/fisiologia , Esquistossomose/imunologia , Técnicas de Ablação , Animais , Modelos Animais de Doenças , Feminino , Humanos , Ativação de Macrófagos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Receptores de Superfície Celular/genética , Transdução de Sinais
7.
PLoS Pathog ; 13(10): e1006691, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29059246

RESUMO

Alcelaphine herpesvirus 1 (AlHV-1) is a γ-herpesvirus (γ-HV) belonging to the macavirus genus that persistently infects its natural host, the wildebeest, without inducing any clinical sign. However, cross-transmission to other ruminant species causes a deadly lymphoproliferative disease named malignant catarrhal fever (MCF). AlHV-1 ORF73 encodes the latency-associated nuclear antigen (LANA)-homolog protein (aLANA). Recently, aLANA has been shown to be essential for viral persistence in vivo and induction of MCF, suggesting that aLANA shares key properties of other γ-HV genome maintenance proteins. Here we have investigated the evasion of the immune response by aLANA. We found that a glycin/glutamate (GE)-rich repeat domain was sufficient to inhibit in cis the presentation of an epitope linked to aLANA. Although antigen presentation in absence of GE was dependent upon proteasomal degradation of aLANA, a lack of GE did not affect protein turnover. However, protein self-synthesis de novo was downregulated by aLANA GE, a mechanism directly associated with reduced antigen presentation in vitro. Importantly, codon-modification of aLANA GE resulted in increased antigen presentation in vitro and enhanced induction of antigen-specific CD8+ T cell responses in vivo, indicating that mRNA constraints in GE rather than peptidic sequence are responsible for cis-limitation of antigen presentation. Nonetheless, GE-mediated limitation of antigen presentation in cis of aLANA was dispensable during MCF as rabbits developed the disease after virus infection irrespective of the expression of full-length or GE-deficient aLANA. Altogether, we provide evidence that inhibition in cis of protein synthesis through GE is likely involved in long-term immune evasion of AlHV-1 latent persistence in the wildebeest natural host, but dispensable in MCF pathogenesis.


Assuntos
Gammaherpesvirinae/imunologia , Evasão da Resposta Imune/imunologia , Febre Catarral Maligna/imunologia , Proteínas Virais/química , Proteínas Virais/imunologia , Animais , Apresentação de Antígeno/imunologia , Bovinos , Ácido Glutâmico/imunologia , Glicina/imunologia , Latência Viral/imunologia
8.
Parasite Immunol ; 41(3): e12599, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30372527

RESUMO

Helminth infections leave a long-lasting immunological footprint on their hosts. Clinical studies have provided first evidence that maternal helminth infections can result in an altered immune profile in their offspring which can potentially shape how they respond to conditions throughout life. This can relate to changes in offspring induction of immune responses against other diseases. However, whether these changes result in actual changes in offspring ability to control disease is unclear. Our understanding of which immune mechanisms are altered and how they are changed is limited. In this review, we highlight what we know from human and mouse studies about this important context of helminth exposure. Moreover, we discuss how mechanisms such as antibody transfer, antigen exposure, maternal cell uptake, chimerism and epigenetics are all likely to be functional contributors to the striking changes that are seen in offspring born or nursed by helminth exposed mothers.


Assuntos
Feto/imunologia , Helmintíase/imunologia , Helmintos/imunologia , Imunidade Materno-Adquirida/imunologia , Animais , Antígenos de Helmintos/imunologia , Feminino , Helmintíase/parasitologia , Humanos , Camundongos , Mães
9.
J Virol ; 89(7): 3630-47, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25589653

RESUMO

UNLABELLED: Viral semaphorins are semaphorin 7A (sema7A) mimics found in pox- and herpesviruses. Among herpesviruses, semaphorins are encoded by gammaherpesviruses of the Macavirus genus only. Alcelaphine herpesvirus 1 (AlHV-1) is a macavirus that persistently infects wildebeest asymptomatically but induces malignant catarrhal fever (MCF) when transmitted to several species of susceptible ruminants and the rabbit model. MCF is caused by the activation/proliferation of latently infected T lymphocytes. Viral semaphorins have been suggested to mediate immune evasion mechanisms and/or directly alter host T cell function. We studied AlHV-sema, the viral semaphorin encoded by the A3 gene of AlHV-1. Phylogenetic analyses revealed independent acquisition of pox- and herpesvirus semaphorins, suggesting that these proteins might have distinct functions. AlHV-sema showed a predicted three-dimensional structure very similar to sema7A and conserved key residues in sema7A-plexinC1 interaction. Expression analyses revealed that AlHV-sema is a secreted 93-kDa glycoprotein expressed during the early phase of virus replication. Purified AlHV-sema was able to bind to fibroblasts and dendritic cells and induce F-actin condensation and cell retraction through a plexinC1 and Rho/cofilin-dependent mechanism. Cytoskeleton rearrangement was further associated with inhibition of phagocytosis by dendritic cells and migration to the draining lymph node. Next, we generated recombinant viruses and demonstrated that the lack of A3 did not significantly affect virus growth in vitro and did not impair MCF induction and associated lymphoproliferative lesions. In conclusion, AlHV-sema has immune evasion functions through mechanisms similar to poxvirus semaphorin but is not directly involved in host T cell activation during MCF. IMPORTANCE: Whereas most poxviruses encode viral semaphorins, semaphorin-like genes have only been identified in few gammaherpesviruses belonging to the Macavirus genus. Alcelaphine herpesvirus 1 (AlHV-1) is a macavirus carried asymptomatically by wildebeest but induces a latency-associated lymphoproliferative disease of T lymphocytes in various ruminant species, namely, malignant catarrhal fever (MCF). Viral semaphorins have been hypothesized to have immune evasion functions and/or be involved in activating latently infected T cells. We present evidence that the viral semaphorin AlHV-sema inhibits dendritic cell phagocytosis and migration to the draining lymph node, both being indispensable mechanisms for protective antiviral responses. Next, we engineered recombinant viruses unable to express AlHV-sema and demonstrated that this protein is dispensable for the induction of MCF. In conclusion, this study suggests that herpesvirus and poxvirus semaphorins have independently evolved similar functions to thwart the immune system of the host while AlHV-sema is not directly involved in MCF-associated T-cell activation.


Assuntos
Células Dendríticas/imunologia , Gammaherpesvirinae/imunologia , Interações Hospedeiro-Patógeno , Linfócitos/fisiologia , Febre Catarral Maligna/virologia , Fagocitose , Semaforinas/imunologia , Animais , Linhagem Celular , Movimento Celular , Proliferação de Células , Citoesqueleto/efeitos dos fármacos , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/virologia , Fibroblastos/efeitos dos fármacos , Gammaherpesvirinae/genética , Perfilação da Expressão Gênica , Humanos , Evasão da Resposta Imune , Febre Catarral Maligna/imunologia , Filogenia , Conformação Proteica , Semaforinas/química , Semaforinas/genética , Homologia de Sequência de Aminoácidos
10.
J Virol ; 89(22): 11438-56, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26339050

RESUMO

UNLABELLED: Testudinid herpesvirus 3 (TeHV-3) is the causative agent of a lethal disease affecting several tortoise species. The threat that this virus poses to endangered animals is focusing efforts on characterizing its properties, in order to enable the development of prophylactic methods. We have sequenced the genomes of the two most studied TeHV-3 strains (1976 and 4295). TeHV-3 strain 1976 has a novel genome structure and is most closely related to a turtle herpesvirus, thus supporting its classification into genus Scutavirus, subfamily Alphaherpesvirinae, family Herpesviridae. The sequence of strain 1976 also revealed viral counterparts of cellular interleukin-10 and semaphorin, which have not been described previously in members of subfamily Alphaherpesvirinae. TeHV-3 strain 4295 is a mixture of three forms (m1, m2, and M), in which, in comparison to strain 1976, the genomes exhibit large, partially overlapping deletions of 12.5 to 22.4 kb. Viral subclones representing these forms were isolated by limiting dilution assays, and each replicated in cell culture comparably to strain 1976. With the goal of testing the potential of the three forms as attenuated vaccine candidates, strain 4295 was inoculated intranasally into Hermann's tortoises (Testudo hermanni). All inoculated subjects died, and PCR analyses demonstrated the ability of the m2 and M forms to spread and invade the brain. In contrast, the m1 form was detected in none of the organs tested, suggesting its potential as the basis of an attenuated vaccine candidate. Our findings represent a major step toward characterizing TeHV-3 and developing prophylactic methods against it. IMPORTANCE: Testudinid herpesvirus 3 (TeHV-3) causes a lethal disease in tortoises, several species of which are endangered. We have characterized the viral genome and used this information to take steps toward developing an attenuated vaccine. We have sequenced the genomes of two strains (1976 and 4295), compared their growth in vitro, and investigated the pathogenesis of strain 4295, which consists of three deletion mutants. The major findings are that (i) TeHV-3 has a novel genome structure, (ii) its closest relative is a turtle herpesvirus, (iii) it contains interleukin-10 and semaphorin genes (the first time these have been reported in an alphaherpesvirus), (iv) a sizeable region of the genome is not required for viral replication in vitro or virulence in vivo, and (v) one of the components of strain 4295, which has a deletion of 22.4 kb, exhibits properties indicating that it may serve as the starting point for an attenuated vaccine.


Assuntos
Alphaherpesvirinae/genética , Alphaherpesvirinae/patogenicidade , Encéfalo/virologia , Infecções por Herpesviridae/veterinária , Tartarugas/virologia , Vacinas Virais/imunologia , Alphaherpesvirinae/classificação , Animais , Sequência de Bases , Linhagem Celular , Mapeamento Cromossômico , DNA Viral/genética , Genoma Viral/genética , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/virologia , Interleucina-10/genética , Dados de Sequência Molecular , Filogenia , Semaforinas/genética , Análise de Sequência de DNA , Deleção de Sequência/genética
11.
Proc Natl Acad Sci U S A ; 110(21): E1933-42, 2013 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-23630278

RESUMO

Wildebeests carry asymptomatically alcelaphine herpesvirus 1 (AlHV-1), a γ-herpesvirus inducing malignant catarrhal fever (MCF) to several ruminant species (including cattle). This acute and lethal lymphoproliferative disease occurs after a prolonged asymptomatic incubation period after transmission. Our recent findings with the rabbit model indicated that AlHV-1 infection is not productive during MCF. Here, we investigated whether latency establishment could explain this apparent absence of productive infection and sought to determine its role in MCF pathogenesis. First, whole-genome cellular and viral gene expression analyses were performed in lymph nodes of MCF-developing calves. Whereas a severe disruption in cellular genes was observed, only 10% of the entire AlHV-1 genome was expressed, contrasting with the 45% observed during productive infection in vitro. In vivo, the expressed viral genes included the latency-associated nuclear antigen homolog ORF73 but none of the regions known to be essential for productive infection. Next, genomic conformation analyses revealed that AlHV-1 was essentially episomal, further suggesting that MCF might be the consequence of a latent infection rather than abortive lytic infection. This hypothesis was further supported by the high frequencies of infected CD8(+) T cells during MCF using immunodetection of ORF73 protein and single-cell RT-PCR approaches. Finally, the role of latency-associated ORF73 was addressed. A lack of ORF73 did not impair initial virus replication in vivo, but it rendered AlHV-1 unable to induce MCF and persist in vivo and conferred protection against a lethal challenge with a WT virus. Together, these findings suggest that a latent infection is essential for MCF induction.


Assuntos
Antígenos Nucleares/biossíntese , Antígenos Virais/biossíntese , Gammaherpesvirinae/fisiologia , Regulação Viral da Expressão Gênica/fisiologia , Transtornos Linfoproliferativos/metabolismo , Febre Catarral Maligna/metabolismo , Latência Viral/fisiologia , Doença Aguda , Animais , Antígenos Nucleares/genética , Antígenos Virais/genética , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/patologia , Linfócitos T CD8-Positivos/virologia , Bovinos , Genoma Viral/fisiologia , Transtornos Linfoproliferativos/genética , Transtornos Linfoproliferativos/patologia , Transtornos Linfoproliferativos/virologia , Febre Catarral Maligna/patologia , Febre Catarral Maligna/virologia , Plasmídeos/genética , Plasmídeos/metabolismo , Coelhos , Replicação Viral/fisiologia
12.
J Gen Virol ; 96(11): 3360-3372, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26329753

RESUMO

Alcelaphine herpesvirus 1 (AlHV-1) is a c-herpesvirus (c-HV) carried asymptomatically by wildebeest. Upon cross-species transmission, AlHV-1 induces a fatal lymphoproliferative disease named malignant catarrhal fever (MCF) in many ruminants, including cattle, and the rabbit model. Latency has been shown to be essential for MCF induction. However, the mechanisms causing the activation and proliferation of infected CD8+T cells are unknown. Many c-HVs express microRNAs (miRNAs). These small non-coding RNAs can regulate expression of host or viral target genes involved in various pathways and are thought to facilitate viral infection and/or mediate activation and proliferation of infected lymphocytes. The AlHV-1 genome has been predicted to encode a large number of miRNAs. However, their precise contribution in viral infection and pathogenesis in vivo remains unknown. Here, using cloning and sequencing of small RNAs we identified 36 potential miRNAs expressed in a lymphoblastoid cell line propagated from a calf infected with AlHV-1 and developing MCF. Among the sequenced candidate miRNAs, 32 were expressed on the reverse strand of the genome in two main clusters. The expression of these 32 viral miRNAs was further validated using Northern blot and quantitative reverse transcription PCR in lymphoid organs of MCF developing calves or rabbits. To determine the concerted contribution in MCF of 28 viralmiRNAs clustered in the non-protein-coding region of the AlHV-1 genome, a recombinant virus was produced. The absence of these 28 miRNAs did not affect viral growth in vitro or MCF induction in rabbits, indicating that the AlHV-1 miRNAs clustered in this non-protein-coding genomic region are dispensable for MCF induction.


Assuntos
Doenças dos Bovinos/virologia , Gammaherpesvirinae/isolamento & purificação , Infecções por Herpesviridae/veterinária , Febre Catarral Maligna/virologia , MicroRNAs/genética , RNA Viral/genética , Animais , Bovinos , Gammaherpesvirinae/classificação , Gammaherpesvirinae/genética , Infecções por Herpesviridae/virologia , Sequenciamento de Nucleotídeos em Larga Escala , Coelhos
13.
Nat Commun ; 15(1): 5960, 2024 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-39013877

RESUMO

Hookworm infection remains a significant public health concern, particularly in low- and middle-income countries, where mass drug administration has not stopped reinfection. Developing a vaccine is crucial to complement current control measures, which necessitates a thorough understanding of host immune responses. By leveraging controlled human infection models and high-dimensional immunophenotyping, here we investigated the immune remodeling following infection with 50 Necator americanus L3 hookworm larvae in four naïve volunteers over two years of follow-up and compared the profiles with naturally infected populations in endemic areas. Increased plasmacytoid dendritic cell frequency and diminished responsiveness to Toll-like receptor 7/8 ligand were observed in both controlled and natural infection settings. Despite the increased CD45RA+ regulatory T cell (Tregs) frequencies in both settings, markers of Tregs function, including inducible T-cell costimulatory (ICOS), tumor necrosis factor receptor 2 (TNFR2), and latency-associated peptide (LAP), as well as in vitro Tregs suppressive capacity were higher in natural infections. Taken together, this study provides unique insights into the immunological trajectories following a first-in-life hookworm infection compared to natural infections.


Assuntos
Células Dendríticas , Necator americanus , Linfócitos T Reguladores , Humanos , Linfócitos T Reguladores/imunologia , Animais , Células Dendríticas/imunologia , Necator americanus/imunologia , Masculino , Adulto , Necatoríase/imunologia , Infecções por Uncinaria/imunologia , Infecções por Uncinaria/parasitologia , Feminino , Doenças Endêmicas , Adulto Jovem , Imunofenotipagem
14.
Sci Immunol ; 9(98): eado1227, 2024 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-39093958

RESUMO

The lung is constantly exposed to airborne pathogens and particles that can cause alveolar damage. Hence, appropriate repair responses are essential for gas exchange and life. Here, we deciphered the spatiotemporal trajectory and function of an atypical population of macrophages after lung injury. Post-influenza A virus (IAV) infection, short-lived monocyte-derived Ly6G-expressing macrophages (Ly6G+ Macs) were recruited to the alveoli of lung perilesional areas. Ly6G+ Macs engulfed immune cells, exhibited a high metabolic potential, and clustered with alveolar type 2 epithelial cells (AT2s) in zones of active epithelial regeneration. Ly6G+ Macs were partially dependent on granulocyte-macrophage colony-stimulating factor and interleukin-4 receptor signaling and were essential for AT2-dependent alveolar regeneration. Similar macrophages were recruited in other models of injury and in the airspaces of lungs from patients with suspected pneumonia. This study identifies perilesional alveolar Ly6G+ Macs as a spatially restricted, short-lived macrophage subset promoting epithelial regeneration postinjury, thus representing an attractive therapeutic target for treating lung damage.


Assuntos
Antígenos Ly , Lesão Pulmonar , Macrófagos Alveolares , Camundongos Endogâmicos C57BL , Regeneração , Animais , Antígenos Ly/metabolismo , Antígenos Ly/imunologia , Camundongos , Regeneração/imunologia , Lesão Pulmonar/imunologia , Macrófagos Alveolares/imunologia , Masculino , Humanos , Feminino , Infecções por Orthomyxoviridae/imunologia , Alvéolos Pulmonares/imunologia , Vírus da Influenza A/imunologia , Vírus da Influenza A/fisiologia
16.
Trends Parasitol ; 39(5): 358-372, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36935340

RESUMO

Helminths are parasitic worms that coevolve with their host, usually resulting in long-term persistence through modulating host immunity. The multifarious mechanisms altering the immune system induced by helminths have significant implications on the control of coinfecting pathogens such as viruses. Here, we explore the recent literature to highlight the main immune alterations and mechanisms that affect the control of viral coinfection. Insights from these mechanisms are valuable in the understanding of clinical observations in helminth-prevalent areas and in the design of new therapeutic and vaccination strategies to control viral diseases.


Assuntos
Coinfecção , Helmintíase , Helmintos , Viroses , Vírus , Animais , Sistema Imunitário , Viroses/complicações
17.
Viruses ; 15(2)2023 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-36851740

RESUMO

Gammaherpesviruses (γHVs) include viruses that can induce lymphoproliferative diseases and tumors. These viruses can persist in the long term in the absence of any pathological manifestation in their natural host. Alcelaphine gammaherpesvirus 1 (AlHV-1) belongs to the genus Macavirus and asymptomatically infects its natural host, the wildebeest (Connochaetes spp.). However, when transmitted to several susceptible species belonging to the order Artiodactyla, AlHV-1 is responsible for the induction of a lethal lymphoproliferative disease, named wildebeest-derived malignant catarrhal fever (WD-MCF). Understanding the pathogenic mechanisms responsible for the induction of WD-MCF is important to better control the risks of transmission and disease development in susceptible species. The aim of this review is to synthesize the current knowledge on WD-MCF with a particular focus on the mechanisms by which AlHV-1 induces the disease. We discuss the potential mechanisms of pathogenesis from viral entry into the host to the maintenance of viral genomes in infected CD8+ T lymphocytes, and we present current hypotheses to explain how AlHV-1 infection induces a peripheral T cell lymphoma-like disease.


Assuntos
Antílopes , Gammaherpesvirinae , Linfoma de Células T Periférico , Febre Catarral Maligna , Bovinos , Animais
18.
Virologie (Montrouge) ; 16(5): 299-314, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-33065914

RESUMO

Gammaherpesviruses include viruses able to induce lymphoproliferative diseases and tumors. These viruses can also establish a long and persistent asymptomatic infection in their natural host. Among gammaherpesviruses, AlHV-1 belongs to the Macavirus genus and infects asymptomatically its natural host, the wildebeest (Connochaetes spp.). However, this virus induces a lethal lymphoproliferative disease named the African form of malignant catarrhal fever (MCF) when transmitted to a large number of susceptible species belonging to the Artiodactyla order. Control of MCF in regions where AlHV-1 is endemic is of great importance and directly depends on the understanding of the pathogenic mechanisms responsible for the induction of MCF. The goal of the present review is to summarize current knowledge on AlHV-1 and MCF with a particular interest on the mechanisms used by AlHV-1 to induce the disease. Amongst different models of pathogenesis, the particular role of viral latency will be addressed based on current data. Finally, the evolutionary relationship between the wildebeest, AlHV-1 and the susceptible species to MCF will be discussed.

19.
Vaccines (Basel) ; 10(12)2022 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-36560568

RESUMO

Malignant catarrhal fever (MCF) is a complex and often fatal disease of ungulates. Effective vaccines are needed to avoid MCF outbreaks and mitigate losses. This study aimed to evaluate a sheep-associated MCF (SA-MCF) vaccine candidate targeting ovine herpesvirus 2 (OvHV-2) glycoprotein B (gB). Rabbits were used as a laboratory animal model to test the safety, immunogenicity, and protective efficacy of a chimeric virus consisting of a recombinant, non-pathogenic strain of alcelaphine herpesvirus-1 encoding OvHV-2 ORF8 to express gB (AlHV-1∆ORF73/OvHV-2-ORF8). Viral-vectored immunizations were performed by using the AlHV-1∆ORF73/OvHV-2-ORF8 chimera alone or as a DNA prime (OvHV-2-ORF8)-virus boost regimen. The viral vector was inoculated by intravenous or intramuscular routes and the DNA was delivered by intradermal shots using a gene gun. The vaccine candidates were deemed safe as no clinical signs were observed following any of the immunizations. Anti-OvHV-2 gB antibodies with neutralizing activity were induced by all immunogens. At three weeks post-final immunization, all animals were challenged intranasally with a lethal dose of OvHV-2. MCF protection rates ranging from 66.7% to 71.4% were observed in vaccinated rabbits, while all mock-vaccinated animals developed the disease. The significant protective efficacy obtained with the vaccine platforms tested in this study encourages further trials in relevant livestock species, such as cattle and bison.

20.
Vet Res ; 42: 95, 2011 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-21859474

RESUMO

Alcelaphine herpesvirus 1 (AlHV-1) is carried by wildebeest asymptomatically. It causes a fatal lymphoproliferative disease named wildebeest-derived malignant catarrhal fever (WD-MCF) when cross-species transmitted to a variety of susceptible species of the Artiodactyla order. WD-MCF can be reproduced experimentally in rabbits. In a previous report, we demonstrated that WD-MCF induced by AlHV-1 is associated with a severe proliferation of CD8(+) T cells in the lymphoid tissues. Here, we further studied the mononuclear leukocytic populations in both the lymphoid (throughout the infection and at time of euthanasia) and non-lymphoid (at time of euthanasia) organs during WD-MCF induced experimentally in rabbits. To reach that goal, we performed multi-colour flow cytometry stainings. The results obtained demonstrate that the development of WD-MCF correlates in peripheral blood with a severe increase of CD8(+) cell percentages; and that CD3(+)CD8(+)CD4(-) T cells were the predominant cell type in both lymphoid and non-lymphoid organs at time of euthanasia. Further characterization of the mononuclear leukocytes isolated from both lymphoid and non-lymphoid tissues revealed that the CD8(+) T cells express high levels of the activation markers CD25 and CD44, produce high amount of gamma-interferon (IFN-γ) and perforin, and showed a reduction of interleukin-2 (IL-2) gene expression. These data demonstrate that the development of WD-MCF is associated with the expansion and infiltration of activated and cytotoxic CD3(+)CD8(+)CD4(-) T cells secreting high amount of IFN-γ but low IL-2.


Assuntos
Gammaherpesvirinae/fisiologia , Infecções por Herpesviridae/veterinária , Tecido Linfoide/imunologia , Febre Catarral Maligna/imunologia , Linfócitos T/imunologia , Animais , Complexo CD3/metabolismo , Citometria de Fluxo/veterinária , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/virologia , Tecido Linfoide/virologia , Febre Catarral Maligna/virologia , Perforina/metabolismo , Coelhos , Reação em Cadeia da Polimerase em Tempo Real/veterinária , Organismos Livres de Patógenos Específicos , Linfócitos T/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA