Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 57(7): 1549-1566.e8, 2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-38776917

RESUMEN

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.


Asunto(s)
Linaje de la Célula , Eosinófilos , Interleucina-5 , Ratones Transgénicos , Proteómica , Análisis de la Célula Individual , Transcriptoma , Eosinófilos/inmunología , Eosinófilos/metabolismo , Animales , Interleucina-5/metabolismo , Interleucina-5/genética , Humanos , Ratones , Proteómica/métodos , Análisis de la Célula Individual/métodos , Diferenciación Celular/inmunología , Ratones Endogámicos C57BL , Perfilación de la Expresión Génica/métodos , Subunidad alfa del Receptor de Interleucina-5/metabolismo , Subunidad alfa del Receptor de Interleucina-5/genética , Mielopoyesis/genética , Factores Reguladores del Interferón/metabolismo , Factores Reguladores del Interferón/genética , Ratones Noqueados
2.
Nat Immunol ; 19(9): 1035, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29955109

RESUMEN

In the version of this article initially published, the accession code for the RNA-seq data set deposited in the NCBI public repository Sequence Read Archive was missing from the 'Data availability' subsection of the Methods section. The accession code is SRP125477.

3.
Nat Immunol ; 18(12): 1310-1320, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29035391

RESUMEN

The hygiene hypothesis postulates that the recent increase in allergic diseases such as asthma and hay fever observed in Western countries is linked to reduced exposure to childhood infections. Here we investigated how infection with a gammaherpesvirus affected the subsequent development of allergic asthma. We found that murid herpesvirus 4 (MuHV-4) inhibited the development of house dust mite (HDM)-induced experimental asthma by modulating lung innate immune cells. Specifically, infection with MuHV-4 caused the replacement of resident alveolar macrophages (AMs) by monocytes with regulatory functions. Monocyte-derived AMs blocked the ability of dendritic cells to trigger a HDM-specific response by the TH2 subset of helper T cells. Our results indicate that replacement of embryonic AMs by regulatory monocytes is a major mechanism underlying the long-term training of lung immunity after infection.


Asunto(s)
Asma/terapia , Macrófagos Alveolares/inmunología , Monocitos/inmunología , Pyroglyphidae/inmunología , Rhadinovirus/inmunología , Células Th2/inmunología , Traslado Adoptivo , Animales , Asma/inmunología , Línea Celular , Cricetinae , Células Dendríticas/inmunología , Femenino , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/virología , Macrófagos Alveolares/citología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Células Th2/trasplante
4.
EMBO J ; 41(18): e109353, 2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-35920020

RESUMEN

Macrophage polarization is a process whereby macrophages acquire distinct effector states (M1 or M2) to carry out multiple and sometimes opposite functions. We show here that translational reprogramming occurs during macrophage polarization and that this relies on the Elongator complex subunit Elp3, an enzyme that modifies the wobble uridine base U34 in cytosolic tRNAs. Elp3 expression is downregulated by classical M1-activating signals in myeloid cells, where it limits the production of pro-inflammatory cytokines via FoxO1 phosphorylation, and attenuates experimental colitis in mice. In contrast, alternative M2-activating signals upregulate Elp3 expression through a PI3K- and STAT6-dependent signaling pathway. The metabolic reprogramming linked to M2 macrophage polarization relies on Elp3 and the translation of multiple candidates, including the mitochondrial ribosome large subunit proteins Mrpl3, Mrpl13, and Mrpl47. By promoting translation of its activator Ric8b in a codon-dependent manner, Elp3 also regulates mTORC2 activation. Elp3 expression in myeloid cells further promotes Wnt-driven tumor initiation in the intestine by maintaining a pool of tumor-associated macrophages exhibiting M2 features. Collectively, our data establish a functional link between tRNA modifications, mTORC2 activation, and macrophage polarization.


Asunto(s)
Histona Acetiltransferasas , Activación de Macrófagos , Transducción de Señal , Animales , Codón/metabolismo , Histona Acetiltransferasas/genética , Activación de Macrófagos/genética , Macrófagos/metabolismo , Diana Mecanicista del Complejo 2 de la Rapamicina/genética , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Ratones
5.
PLoS Pathog ; 16(3): e1008405, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32176737

RESUMEN

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.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Gammaherpesvirinae/inmunología , Genes Virales/inmunología , Fiebre Catarral Maligna/inmunología , Proteínas del Envoltorio Viral/inmunología , Animales , Bovinos , Línea Celular , Gammaherpesvirinae/genética , Fiebre Catarral Maligna/genética , Conejos , Proteínas del Envoltorio Viral/genética
6.
EMBO J ; 36(16): 2404-2418, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28716804

RESUMEN

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.


Asunto(s)
Células Dendríticas/inmunología , Interferón Tipo I/metabolismo , Células Th2/inmunología , Alérgenos/inmunología , Animales , Ratones , Ratones Noqueados , Pyroglyphidae/inmunología , Receptor de Interferón alfa y beta/deficiencia , Schistosoma mansoni/inmunología
7.
Eur J Immunol ; 49(7): 1067-1081, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30919955

RESUMEN

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.


Asunto(s)
Granuloma/inmunología , Macrófagos del Hígado/inmunología , Hígado/patología , Macrófagos/inmunología , Receptores de Superficie Celular/metabolismo , Schistosoma mansoni/fisiología , Esquistosomiasis/inmunología , Técnicas de Ablación , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Activación de Macrófagos , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Receptores de Superficie Celular/genética , Transducción de Señal
8.
PLoS Pathog ; 13(10): e1006691, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29059246

RESUMEN

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.


Asunto(s)
Gammaherpesvirinae/inmunología , Evasión Inmune/inmunología , Fiebre Catarral Maligna/inmunología , Proteínas Virales/química , Proteínas Virales/inmunología , Animales , Presentación de Antígeno/inmunología , Bovinos , Ácido Glutámico/inmunología , Glicina/inmunología , Latencia del Virus/inmunología
9.
Parasite Immunol ; 41(3): e12599, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30372527

RESUMEN

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.


Asunto(s)
Feto/inmunología , Helmintiasis/inmunología , Helmintos/inmunología , Inmunidad Materno-Adquirida/inmunología , Animales , Antígenos Helmínticos/inmunología , Femenino , Helmintiasis/parasitología , Humanos , Ratones , Madres
10.
J Environ Manage ; 225: 193-204, 2018 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-30086441

RESUMEN

Urban development may increase the risk of future floods because of local changes in hydrological conditions and an increase in flood exposure that arises from an increasing population and expanding infrastructure within flood-prone zones. Existing urban land use change models generally consider the expansion process and do not consider the densification of existing urban areas. In this paper, we simulate 24 possible urbanization scenarios in Wallonia region (Belgium) until 2100. These scenarios are generated using an agent-based model that considers urban expansion and densification as well as development restrictions in flood-prone zones. The extents of inundation and water depths for each scenario are determined by the WOLF 2D hydraulic model for steady floods corresponding to return periods of 25, 50, and 100 years. Our results show that future flood damages and their spatial distributions vary remarkably from one urbanization scenario to another. A spatial planning policy oriented towards strict development control in flood-prone zones leads to a substantial mitigation of the increased flood damage. By contrast, a spatial planning policy exclusively oriented to infill development with no development restrictions in flood-prone zones would be the most detrimental in terms of exposure to flood risk. Our study enables the identification of the most sensitive locations for flood damage related to urban development, which can help in the design of more resilient spatial planning strategies and localize zones with high levels of flood risk for each scenario.


Asunto(s)
Planificación de Ciudades , Inundaciones , Urbanización , Bélgica , Hidrología , Modelos Teóricos , Riesgo
11.
J Virol ; 89(7): 3630-47, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25589653

RESUMEN

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.


Asunto(s)
Células Dendríticas/inmunología , Gammaherpesvirinae/inmunología , Interacciones Huésped-Patógeno , Linfocitos/fisiología , Fiebre Catarral Maligna/virología , Fagocitosis , Semaforinas/inmunología , Animales , Línea Celular , Movimiento Celular , Proliferación Celular , Citoesqueleto/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Células Dendríticas/virología , Fibroblastos/efectos de los fármacos , Gammaherpesvirinae/genética , Perfilación de la Expresión Génica , Humanos , Evasión Inmune , Fiebre Catarral Maligna/inmunología , Filogenia , Conformación Proteica , Semaforinas/química , Semaforinas/genética , Homología de Secuencia de Aminoácido
12.
J Virol ; 89(22): 11438-56, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26339050

RESUMEN

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.


Asunto(s)
Alphaherpesvirinae/genética , Alphaherpesvirinae/patogenicidad , Encéfalo/virología , Infecciones por Herpesviridae/veterinaria , Tortugas/virología , Vacunas Virales/inmunología , Alphaherpesvirinae/clasificación , Animales , Secuencia de Bases , Línea Celular , Mapeo Cromosómico , ADN Viral/genética , Genoma Viral/genética , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/virología , Interleucina-10/genética , Datos de Secuencia Molecular , Filogenia , Semaforinas/genética , Análisis de Secuencia de ADN , Eliminación de Secuencia/genética
13.
Proc Natl Acad Sci U S A ; 110(21): E1933-42, 2013 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-23630278

RESUMEN

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.


Asunto(s)
Antígenos Nucleares/biosíntesis , Antígenos Virales/biosíntesis , Gammaherpesvirinae/fisiología , Regulación Viral de la Expresión Génica/fisiología , Trastornos Linfoproliferativos/metabolismo , Fiebre Catarral Maligna/metabolismo , Latencia del Virus/fisiología , Enfermedad Aguda , Animales , Antígenos Nucleares/genética , Antígenos Virales/genética , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/patología , Linfocitos T CD8-positivos/virología , Bovinos , Genoma Viral/fisiología , Trastornos Linfoproliferativos/genética , Trastornos Linfoproliferativos/patología , Trastornos Linfoproliferativos/virología , Fiebre Catarral Maligna/patología , Fiebre Catarral Maligna/virología , Plásmidos/genética , Plásmidos/metabolismo , Conejos , Replicación Viral/fisiología
14.
J Gen Virol ; 96(11): 3360-3372, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26329753

RESUMEN

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.


Asunto(s)
Enfermedades de los Bovinos/virología , Gammaherpesvirinae/aislamiento & purificación , Infecciones por Herpesviridae/veterinaria , Fiebre Catarral Maligna/virología , MicroARNs/genética , ARN Viral/genética , Animales , Bovinos , Gammaherpesvirinae/clasificación , Gammaherpesvirinae/genética , Infecciones por Herpesviridae/virología , Secuenciación de Nucleótidos de Alto Rendimiento , Conejos
15.
PLoS Pathog ; 9(10): e1003662, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24204255

RESUMEN

In this study, B cell function in protective T(H)2 immunity against N. brasiliensis infection was investigated. Protection against secondary infection depended on IL-4Rα and IL-13; but not IL-4. Protection did not associate with parasite specific antibody responses. Re-infection of B cell-specific IL-4Rα⁻/⁻ mice resulted in increased worm burdens compared to control mice, despite their equivalent capacity to control primary infection. Impaired protection correlated with reduced lymphocyte IL-13 production and B cell MHC class II and CD86 surface expression. Adoptive transfer of in vivo N. brasiliensis primed IL-4Rα expressing B cells into naïve BALB/c mice, but not IL-4Rα or IL-13 deficient B cells, conferred protection against primary N. brasiliensis infection. This protection required MHC class II compatibility on B cells suggesting cognate interactions by B cells with CD4⁺ T cells were important to co-ordinate immunity. Furthermore, the rapid nature of these protective effects by B cells suggested non-BCR mediated mechanisms, such as via Toll Like Receptors, was involved, and this was supported by transfer experiments using antigen pulsed Myd88⁻/⁻ B cells. These data suggest TLR dependent antigen processing by IL-4Rα-responsive B cells producing IL-13 contribute significantly to CD4⁺ T cell-mediated protective immunity against N. brasiliensis infection.


Asunto(s)
Presentación de Antígeno , Linfocitos B/inmunología , Inmunidad Celular , Nippostrongylus/inmunología , Receptores de Superficie Celular/inmunología , Infecciones por Strongylida/inmunología , Células Th2/inmunología , Animales , Linfocitos B/patología , Antígeno B7-2/genética , Antígeno B7-2/inmunología , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/inmunología , Interleucina-13/genética , Interleucina-13/inmunología , Ratones Endogámicos BALB C , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/inmunología , Receptores de Superficie Celular/genética , Infecciones por Strongylida/genética , Infecciones por Strongylida/patología , Células Th2/patología , Receptores Toll-Like/genética , Receptores Toll-Like/inmunología
16.
Parasite Immunol ; 41(3): e12615, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30805943
17.
Nat Commun ; 15(1): 5960, 2024 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-39013877

RESUMEN

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.


Asunto(s)
Células Dendríticas , Necator americanus , Linfocitos T Reguladores , Humanos , Linfocitos T Reguladores/inmunología , Animales , Células Dendríticas/inmunología , Necator americanus/inmunología , Masculino , Adulto , Necatoriasis/inmunología , Infecciones por Uncinaria/inmunología , Infecciones por Uncinaria/parasitología , Femenino , Enfermedades Endémicas , Adulto Joven , Inmunofenotipificación
19.
Trends Parasitol ; 39(5): 358-372, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36935340

RESUMEN

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.


Asunto(s)
Coinfección , Helmintiasis , Helmintos , Virosis , Virus , Animales , Sistema Inmunológico , Virosis/complicaciones
20.
Animals (Basel) ; 13(11)2023 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-37578748

RESUMEN

Many fluorochromes routinely used in semen quality analysis emit in the green and red channels, limiting their possible combination for multiple parameter analysis. The use of fluorophores emitting in different light channels broadens the possibilities of combination to expand the range of simultaneously evaluated criteria. This is of great interest in cases of small ejaculated volumes, such as those naturally occurring in roosters, small dog breeds and drones (Apis mellifera). The purpose of this experiment is to establish Calcein Violet (CaV), a blue fluorochrome, as a marker of viability and acrosomal integrity in domestic animals in order to free the red and green channels. SYBR®14/Propidium Iodide (PI) was used as reference dye, heat-treated samples as negative controls, serial staining combination for validation and epifluorescence microscopy for observation. Dead spermatozoa marked in red with PI showed no blue fluorescence either from the head or the tail. Live spermatozoa showed a decreasing blue emission from head to tail when single stained with CaV. Unreacted acrosomes showed intense blue fluorescence irrespective of plasma membrane integrity. This needs to be further confirmed for species with small and difficult to observe heads. Establishment of CaV as a marker of membrane integrity by fluorescence microscopy is a decisive first step towards further technical development and use with flow cytometry.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA