Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 50(2): 403-417.e4, 2019 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-30709740

RESUMEN

The tolerogenic microenvironment of the liver is associated with impaired hepatic T cell function. Here, we examined the contribution of liver-resident natural killer (LrNK) cells, a prominent hepatic NK cell compartment, to T cell antiviral responses in the liver. The number of virus-specific T cells increased in LrNK-cell-deficient mice during both acute and chronic lymphocytic choriomeningitis virus infection. Upon infection with adenovirus, hepatic T cells from these mice produced more cytokines, which was accompanied by reduced viral loads. Transfer of LrNK cells into LrNK-cell-deficient or wild-type mice inhibited hepatic T cell function, resulting in impaired viral clearance, whereas transfer of conventional NK cells promoted T cell antiviral responses. LrNK-cell-mediated inhibition of T cell function was dependent on the PD-1-PD-L1 axis. Our findings reveal a role for LrNK cells in the regulation of T cell immunity and provide insight into the mechanisms of immune tolerance in the liver.


Asunto(s)
Antígeno B7-H1/inmunología , Células Asesinas Naturales/inmunología , Hígado/inmunología , Receptor de Muerte Celular Programada 1/inmunología , Linfocitos T/inmunología , Animales , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Hepatocitos/inmunología , Hepatocitos/metabolismo , Hepatocitos/virología , Células Asesinas Naturales/metabolismo , Hígado/metabolismo , Hígado/virología , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/metabolismo , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/fisiología , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/metabolismo , Transducción de Señal/inmunología , Linfocitos T/metabolismo , Linfocitos T/virología , Transcriptoma/genética , Transcriptoma/inmunología
2.
Proc Natl Acad Sci U S A ; 120(23): e2220005120, 2023 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-37252973

RESUMEN

Dengue virus (DENV) is the most important human virus transmitted by mosquitos. Dengue pathogenesis is characterized by a large induction of proinflammatory cytokines. This cytokine induction varies among the four DENV serotypes (DENV1 to 4) and poses a challenge for live DENV vaccine design. Here, we identify a viral mechanism to limit NF-κB activation and cytokine secretion by the DENV protein NS5. Using proteomics, we found that NS5 binds and degrades the host protein ERC1 to antagonize NF-κB activation, limit proinflammatory cytokine secretion, and reduce cell migration. We found that ERC1 degradation involves unique properties of the methyltransferase domain of NS5 that are not conserved among the four DENV serotypes. By obtaining chimeric DENV2 and DENV4 viruses, we map the residues in NS5 for ERC1 degradation, and generate recombinant DENVs exchanging serotype properties by single amino acid substitutions. This work uncovers a function of the viral protein NS5 to limit cytokine production, critical to dengue pathogenesis. Importantly, the information provided about the serotype-specific mechanism for counteracting the antiviral response can be applied to improve live attenuated vaccines.


Asunto(s)
Virus del Dengue , Dengue , Proteínas no Estructurales Virales , Humanos , Citocinas , FN-kappa B/metabolismo , Serogrupo , Proteínas no Estructurales Virales/metabolismo
3.
Trends Biochem Sci ; 46(9): 758-771, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33736931

RESUMEN

Modified bases act as marks on cellular RNAs so that they can be distinguished from foreign RNAs, reducing innate immune responses to endogenous RNA. In humans, mutations giving reduced levels of one base modification, adenosine-to-inosine deamination, cause a viral infection mimic syndrome, a congenital encephalitis with aberrant interferon induction. These Aicardi-Goutières syndrome 6 mutations affect adenosine deaminase acting on RNA 1 (ADAR1), which generates inosines in endogenous double-stranded (ds)RNA. The inosine base alters dsRNA structure to prevent aberrant activation of antiviral cytosolic helicase RIG-I-like receptors. We review how effects of inosines, ADARs, and other modified bases have been shown to be important in innate immunity and cancer.


Asunto(s)
Inmunidad Innata , Edición de ARN , Proteínas de Unión al ARN , Adenosina Desaminasa/genética , Adenosina Desaminasa/metabolismo , Humanos , ARN Bicatenario , Proteínas de Unión al ARN/metabolismo , Transcriptoma
4.
J Virol ; 97(3): e0175822, 2023 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-36916936

RESUMEN

Recent studies have begun to reveal the complex and multifunctional roles of N6-methyladenosine (m6A) modifications and their associated writer, reader, and eraser proteins in infection by diverse RNA and DNA viruses. However, little is known about their regulation and functions during infection by several viruses, including poxviruses. Here, we show that members of the YTH Domain Family (YTHDF), in particular YTHDF2, are downregulated as the prototypical poxvirus, vaccinia virus (VacV) enters later stages of replication in a variety of natural target cell types, but not in commonly used transformed cell lines wherein the control of YTHDF2 expression appears to be dysregulated. YTHDF proteins also decreased at late stages of infection by herpes simplex virus 1 (HSV-1) but not human cytomegalovirus, suggesting that YTHDF2 is downregulated in response to infections that induce host shutoff. In line with this idea, YTHDF2 was potently downregulated upon infection with a VacV mutant expressing catalytically inactive forms of the decapping enzymes, D9 and D10, which fails to degrade dsRNA and induces a protein kinase R response that itself inhibits protein synthesis. Overexpression and RNAi-mediated depletion approaches further demonstrate that YTHDF2 does not directly affect VacV replication. Instead, experimental downregulation of YTHDF2 or the related family member, YTHDF1, induces a potent increase in interferon-stimulated gene expression and establishes an antiviral state that suppresses infection by either VacV or HSV-1. Combined, our data suggest that YTHDF2 is destabilized in response to infection-induced host shutoff and serves to augment host antiviral responses. IMPORTANCE There is increasing recognition of the importance of N6-methyladenosine (m6A) modifications to both viral and host mRNAs and the complex roles this modification plays in determining the fate of infection by diverse RNA and DNA viruses. However, in many instances, the functional contributions and importance of specific m6A writer, reader, and eraser proteins remains unknown. Here, we show that natural target cells but not transformed cell lines downregulate the YTH Domain Family (YTHDF) of m6A reader proteins, in particular YTHDF2, in response to shutoff of protein synthesis upon infection with the large DNA viruses, vaccinia virus (VacV), or herpes simplex virus type 1. We further reveal that YTHDF2 downregulation also occurs as part of the host protein kinase R response to a VacV shutoff mutant and that this downregulation of YTHDF family members functions to enhance interferon-stimulated gene expression to create an antiviral state.


Asunto(s)
Poxviridae , Proteínas de Unión al ARN , Virus Vaccinia , Vaccinia , Humanos , Expresión Génica , Interferones/metabolismo , Poxviridae/genética , Proteínas Quinasas/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción/metabolismo , Vaccinia/virología , Virus Vaccinia/metabolismo , Replicación Viral , Infecciones por Poxviridae/virología , Interacciones Huésped-Patógeno
5.
Fish Shellfish Immunol ; 151: 109646, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38810712

RESUMEN

To achieve insights in antiviral immune defense of the central nervous system (CNS), we investigated T cells and CD45 cells in the marine fish model Dicentrarchus labrax infected with the CNS-tropic virus betanodavirus. By employing markers for pan-T cells (mAb DLT15) and CD45-cells (mAb DLT22) in immunofluorescence (IIF) of leukocytes from brain, we obtained 3,7 ± 2.3 % of T cells and 7.3 ± 3.2 % of CD45+ cells. Both IIF and immunoelectron microscopy confirmed a leukocyte/glial morphology for the immunoreactive cells. Quantitative immunohistochemistry (qIHC) of brain/eye sections showed 1.9 ± 0.8 % of T+ cells and 2 ± 0.9 % of CD45+ cells in the brain, and 3.6 ± 1.9 % and 4.1 ± 2.2 % in the eye, respectively. After in vivo RGNNV infection the number of T cells/CD45+ leukocytes in the brain increased to 8.3 ± 2.1 % and 11.6 ± 4.4 % (by IIF), and 26.1 ± 3.4 % and 45.6 ± 5.9 % (by qIHC), respectively. In the eye we counted after infection 8.5 ± 4.4 % of T cells and 10.2 ± 5.8 % of CD45 cells. Gene transcription analysis of brain mRNA revealed a strong increase of gene transcripts coding for: antiviral proteins Mx and ISG-12; T-cell related CD3ε/δ, TcRß, CD4, CD8α, CD45; and for immuno-modulatory cytokines TNFα, IL-2, IL-10. A RAG-1 gene product was also present and upregulated, suggesting somatic recombination in the fish brain. Similar transcription data were obtained in the eye, albeit with differences. Our findings provide first evidence for a recruitment and involvement of T cells and CD45+ leukocytes in the fish eye-brain axis during antiviral responses and suggest similarities in the CNS immune defense across evolutionary distant vertebrates.


Asunto(s)
Lubina , Enfermedades de los Peces , Antígenos Comunes de Leucocito , Nodaviridae , Infecciones por Virus ARN , Linfocitos T , Animales , Nodaviridae/fisiología , Enfermedades de los Peces/inmunología , Enfermedades de los Peces/virología , Lubina/inmunología , Infecciones por Virus ARN/veterinaria , Infecciones por Virus ARN/inmunología , Infecciones por Virus ARN/virología , Antígenos Comunes de Leucocito/metabolismo , Antígenos Comunes de Leucocito/genética , Antígenos Comunes de Leucocito/inmunología , Linfocitos T/inmunología , Proteínas de Peces/genética , Proteínas de Peces/inmunología , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/virología , Encéfalo/virología , Encéfalo/inmunología
6.
J Virol ; 96(11): e0063422, 2022 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-35575553

RESUMEN

The global amphibian declines are compounded by infections with members of the Ranavirus genus such as Frog Virus 3 (FV3). Premetamorphic anuran amphibians are believed to be significantly more susceptible to FV3 while this pathogen targets the kidneys of both pre- and postmetamorphic animals. Paradoxically, FV3-challenged Xenopus laevis tadpoles exhibit lower kidney viral loads than adult frogs. Presently, we demonstrate that X. laevis tadpoles are intrinsically more resistant to FV3 kidney infections than cohort-matched metamorphic and postmetamorphic froglets and that this resistance appears to be epigenetically conferred by endogenous retroviruses (ERVs). Using a X. laevis kidney-derived cell line, we show that enhancing ERV gene expression activates cellular double-stranded RNA-sensing pathways, resulting in elevated mRNA levels of antiviral interferon (IFN) cytokines and thus greater anti-FV3 protection. Finally, our results indicate that large esterase-positive myeloid-lineage cells, rather than renal cells, are responsible for the elevated ERV/IFN axis seen in the tadpole kidneys. This conclusion is supported by our observation that CRISPR-Cas9 ablation of colony-stimulating factor-3 results in abolished homing of these myeloid cells to tadpole kidneys, concurrent with significantly abolished tadpole kidney expression of both ERVs and IFNs. We believe that the manuscript marks an important step forward in understanding the mechanisms controlling amphibian antiviral defenses and thus susceptibility and resistance to pathogens like FV3. IMPORTANCE Global amphibian biodiversity is being challenged by pathogens like the Frog Virus 3 (FV3) ranavirus, underlining the need to gain a greater understanding of amphibian antiviral defenses. While it was previously believed that anuran (frog/toad) amphibian tadpoles are more susceptible to FV3, we demonstrated that tadpoles are in fact more resistant to this virus than metamorphic and postmetamorphic froglets. We showed that this resistance is conferred by large myeloid cells within the tadpole kidneys (central FV3 target), which possess an elevated expression of endogenous retroviruses (ERVs). In turn, these ERVs activate cellular double-stranded RNA-sensing pathways, resulting in a greater expression of antiviral interferon cytokines, thereby offering the observed anti-FV3 protection.


Asunto(s)
Infecciones por Virus ADN , Retrovirus Endógenos , Ranavirus , Xenopus laevis , Animales , Línea Celular , Infecciones por Virus ADN/inmunología , Infecciones por Virus ADN/veterinaria , Resistencia a la Enfermedad , Retrovirus Endógenos/inmunología , Interferones/inmunología , Riñón/virología , Larva/inmunología , Larva/virología , ARN Bicatenario , Ranavirus/patogenicidad , Xenopus laevis/virología
7.
EMBO Rep ; 22(11): e52101, 2021 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-34467630

RESUMEN

N6 -methyladenosine (m6 A) is a chemical modification present in multiple RNA species and is most abundant in mRNAs. Studies on m6 A reveal its comprehensive roles in almost every aspect of mRNA metabolism, as well as in a variety of physiological processes. Although some recent discoveries indicate that m6 A can affect the life cycles of numerous viruses as well as the cellular antiviral immune response, the roles of m6 A modification in type I interferon (IFN-I) signaling are still largely unknown. Here, we reveal that WT1-associated protein (WTAP), one of the m6 A "writers", is degraded via the ubiquitination-proteasome pathway upon activation of IFN-I signaling. With the degradation of WTAP, the m6 A levels of IFN-regulatory factor 3 (IRF3) and interferon alpha/beta receptor subunit 1 (IFNAR1) mRNAs are reduced, leading to translational suppression of IRF3 and instability of IFNAR1 mRNA. Thus, the WTAP-IRF3/IFNAR1 axis may serve as negative feedback pathway to fine-tune the activation of IFN-I signaling, which highlights the roles of m6 A in the antiviral response by dictating the fate of mRNAs associated with IFN-I signaling.


Asunto(s)
Antivirales , Factor 3 Regulador del Interferón , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/metabolismo , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ubiquitinación
8.
Fish Shellfish Immunol ; 123: 182-193, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35227882

RESUMEN

Spring viraemia of carp virus (SVCV) is a fierce pathogen causing high mortality in the common carp. At present, the treatment of spring viraemia of carp (SVC) is limited. Innate immunity is the host's first line of defense against microbial pathogens. Retinoic acid-inducible gene I (RIG-I) activation plays an essential role in the antiviral immune response. Virus infection can activate the RIG-I signaling and induce the production of interferon (IFN) and the expression of IFN-stimulated genes (ISGs). STUB1 (STIP1 homology and U-box containing protein 1) is a highly conserved cytoplasmic protein. This protein is known to exist widely in many biological systems and plays an important role in the process of immune regulation, but little is known in fish. To explore the immune function of STUB1 in fish, STUB1 gene was cloned from zebrafish and analyzed in this study. Zebrafish STUB1 showed 77% and 79% amino acid sequence homology with those from human and mouse, respectively. The amino acid sequence of zebrafish STUB1 contains three TPR domains and one U-box domain. Subcellular localization study revealed that STUB1 is located in the cytoplasm. And overexpression of zebrafish STUB1 resulted in the activation of the transcription of IFN1 and ISGs. Functional analysis showed that STUB1 was able to activate RIG-I signaling, and promote the expression of RIG-I, but STUB1 can degrade RIG-I in mammals. The proliferation of SVCV was significantly inhibited after the overexpression of STUB1 and N-terminal TPR domain of STUB1 in EPC cells. And through secondary structure analysis, overexpression of the mutant of STUB1 110 amino acid resulted in weakened antiviral ability. The expression of STUB1 was attenuated by poly(I:C) treatment and SVCV infection. In summary, this study demonstrated for the first time that STUB1 can induce the production of IFN, enhance the expression of ISGs by promoting the expression of RIG-I and inhibiting viral replication in fish. These findings may form the essential basis for the development of antiviral targets and drugs.


Asunto(s)
Carpas , Rhabdoviridae , Animales , Antivirales/metabolismo , Antivirales/farmacología , Carpas/metabolismo , Inmunidad Innata/genética , Mamíferos/metabolismo , Ratones , Rhabdoviridae/fisiología , Ubiquitina-Proteína Ligasas/metabolismo , Pez Cebra , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
9.
Cell Mol Life Sci ; 78(4): 1423-1444, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33084946

RESUMEN

Antiviral responses of interferons (IFNs) are crucial in the host immune response, playing a relevant role in controlling viralw infections. Three types of IFNs, type I (IFN-α, IFN-ß), II (IFN-γ) and III (IFN-λ), are classified according to their receptor usage, mode of induction, biological activity and amino acid sequence. Here, we provide a comprehensive review of type I IFN responses and different mechanisms that viruses employ to circumvent this response. In the first part, we will give an overview of the different induction and signaling cascades induced in the cell by IFN-I after virus encounter. Next, highlights of some of the mechanisms used by viruses to counteract the IFN induction will be described. And finally, we will address different mechanism used by viruses to interference with the IFN signaling cascade and the blockade of IFN induced antiviral activities.


Asunto(s)
Interacciones Huésped-Patógeno/genética , Inmunidad Innata/genética , Interferones/genética , Virosis/genética , Humanos , Evasión Inmune/genética , Interferón-alfa/genética , Interferón-alfa/inmunología , Interferones/clasificación , Interferones/inmunología , Transducción de Señal , Virosis/inmunología
10.
Cell Microbiol ; 22(11): e13242, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32596986

RESUMEN

Influenza A virus (IAV) infection regulates the expression of numerous host genes. However, the precise mechanism underlying implication of these genes in IAV pathogenesis remains largely unknown. Here, we employed isobaric tags for relative and absolute quantification (iTRAQ) to identify host proteins regulated by IAV infection. iTRAQ analysis of mouse lungs infected or uninfected with IAV showed a total of 167 differentially upregulated proteins in response to the viral infection. Interestingly, we observed that p27Kip1, a potent cyclin-dependent kinase inhibitor, was markedly induced by IAV both at mRNA and protein levels through in vitro and in vivo studies. Furthermore, it was shown that innate immune signalling positively regulated p27Kip1 expression in response to IAV infection. Ectopic expression of p27Kip1 in A549 cells dramatically inhibited IAV replication, whereas, p27Kip1 knockdown significantly enhanced the virus replication. in vivo experiments demonstrated that p27Kip1 knockout (KO) mice were more susceptible to IAV than wild-type (WT) mice: exhibiting higher viral load in lung tissue, faster body-weight loss, reduced survival rate and more severe organ damage. Moreover, we found that p27Kip1 overexpression facilitated the degradation of viral NS1 protein, caused a dramatic STAT1 activation and promoted the expression of IFN-ß and several critical antiviral interferon-stimulated genes (ISGs). Increased p27Kip1 expression also restricted infections of several other viruses. Conversely, IAV-infected p27Kip1 KO mice exhibited a sharp increase in NS1 protein accumulation, reduced level of STAT1 activation and decreased expression of IFN-ß and the ISGs in the lung compared to WT animals. These findings reveal a key role of p27Kip1 in enhancing antiviral innate immunity.


Asunto(s)
Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Inmunidad Innata , Subtipo H1N1 del Virus de la Influenza A/inmunología , Infecciones por Orthomyxoviridae/inmunología , Animales , Línea Celular , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/biosíntesis , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Interacciones Huésped-Patógeno , Humanos , Subtipo H1N1 del Virus de la Influenza A/metabolismo , Subtipo H1N1 del Virus de la Influenza A/fisiología , Gripe Humana/genética , Gripe Humana/inmunología , Gripe Humana/metabolismo , Pulmón/metabolismo , Pulmón/virología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/virología , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Transducción de Señal , Regulación hacia Arriba , Proteínas no Estructurales Virales/metabolismo , Virosis/inmunología , Virosis/metabolismo , Virosis/virología , Replicación Viral
11.
Int J Med Sci ; 18(12): 2561-2569, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34104087

RESUMEN

SARS-CoV-2 infection poses a global challenge to human health. Upon viral infection, host cells initiate the innate antiviral response, which primarily involves type I interferons (I-IFNs), to enable rapid elimination of the invading virus. Previous studies revealed that SARS-CoV-2 infection limits the expression of I-IFNs in vitro and in vivo, but the underlying mechanism remains incompletely elucidated. In the present study, we performed data mining and longitudinal data analysis using SARS-CoV-2-infected normal human bronchial epithelial (NHBE) cells and ferrets, and the results confirmed the strong inhibitory effect of SARS-CoV-2 on the induction of I-IFNs. Moreover, we identified genes that are negatively correlated with IFNB1 expression in vitro and in vivo based on Pearson correlation analysis. We found that SARS-CoV-2 activates numerous intrinsic pathways, such as the circadian rhythm, phosphatidylinositol signaling system, peroxisome, and TNF signaling pathways, to inhibit I-IFNs. These intrinsic inhibitory pathways jointly facilitate the successful immune evasion of SARS-CoV-2. Our study elucidates the underlying mechanism by which SARS-CoV-2 evades the host innate antiviral response in vitro and in vivo, providing theoretical evidence for targeting these immune evasion-associated pathways to combat SARS-CoV-2 infection.


Asunto(s)
COVID-19/inmunología , Interacciones Huésped-Patógeno/inmunología , Interferón gamma/metabolismo , SARS-CoV-2/inmunología , Animales , Bronquios/citología , COVID-19/virología , Línea Celular , Conjuntos de Datos como Asunto , Modelos Animales de Enfermedad , Células Epiteliales , Hurones , Regulación de la Expresión Génica/inmunología , Interacciones Huésped-Patógeno/genética , Humanos , Inmunidad Innata , Interferón gamma/inmunología , RNA-Seq , Mucosa Respiratoria/citología , Transducción de Señal/genética , Transducción de Señal/inmunología
12.
J Invertebr Pathol ; 180: 107545, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33571511

RESUMEN

Outbreaks of white spot syndrome virus (WSSV) have caused serious damage to penaeid shrimp aquaculture worldwide. Despite great efforts to characterize the virus, the conditions that lead to infection and the infection mechanisms, there is still a lack of understanding regarding these complex virus-host interactions, which is needed to develop consistent and effective treatment methods for WSSV. In this study, we used a gas chromatography - mass spectrometry (GC-MS)-based metabolomics approach to compare the metabolite profiles of gills, haemolymph and hepatopancreas from whiteleg shrimp (Penaeus vannamei) exposed to WSSV and corresponding controls. The results revealed clear discriminations between metabolite profiles of WSSV-challenged shrimp and controlled shrimp in each tissue. The responses of shrimp gills to WSSV infection were characterized by increases of many fatty acids and amino acids in WSSV-challenged shrimp compared to the controls. Changes in haemolymph metabolite profiles include the increased levels of itaconic acid, energy-related metabolites, metabolites in glutathione cycle and decrease of amino acids. The WSSV challenge led to the decreases of several fatty acids and amino acids and increases of other amino acids, lactic acid and other organic compounds (levulinic acid, malonic acid and putrescine) in hepatopancreas. These alterations of shrimp metabolites suggest several immune responses of shrimp to WSSV in a tissue-specific manner, including upregulation of osmoregulation, antimicrobial activity, metabolic rate, gluconeogenesis, glutathione pathway in control of oxidative stress and shift from aerobic to anaerobic metabolism in shrimp which indicates the Warburg effect. The findings from this study provide a better understanding of molecular process of shrimp response against WSSV invasion which may be useful for development of disease management strategies.


Asunto(s)
Penaeidae/metabolismo , Penaeidae/virología , Virus del Síndrome de la Mancha Blanca 1/fisiología , Animales , Acuicultura , Cromatografía de Gases y Espectrometría de Masas , Branquias/virología , Hemolinfa/virología , Hepatopáncreas/virología
13.
Int J Mol Sci ; 22(9)2021 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-33946935

RESUMEN

Interferons (IFNs) are induced by viruses and are the main regulators of the host antiviral response. They balance tissue tolerance and immune resistance against viral challenges. Like all cells in the human body, neutrophils possess the receptors for IFNs and contribute to antiviral host defense. To combat viruses, neutrophils utilize various mechanisms, such as viral sensing, neutrophil extracellular trap formation, and antigen presentation. These mechanisms have also been linked to tissue damage during viral infection and inflammation. In this review, we presented evidence that a complex cross-regulatory talk between IFNs and neutrophils initiates appropriate antiviral immune responses and regulates them to minimize tissue damage. We also explored recent exciting research elucidating the interactions between IFNs, neutrophils, and severe acute respiratory syndrome-coronavirus-2, as an example of neutrophil and IFN cross-regulatory talk. Dissecting the IFN-neutrophil paradigm is needed for well-balanced antiviral therapeutics and development of novel treatments against many major epidemic or pandemic viral infections, including the ongoing pandemic of the coronavirus disease that emerged in 2019.


Asunto(s)
COVID-19/inmunología , Interferón Tipo I/inmunología , Neutrófilos/inmunología , Virosis/inmunología , Animales , Antivirales/inmunología , Trampas Extracelulares/inmunología , Humanos , SARS-CoV-2/inmunología , Transducción de Señal , Virus/inmunología
14.
Biochem Biophys Res Commun ; 524(1): 89-95, 2020 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-31980167

RESUMEN

Glycogen storage disease type Ib (GSD-Ib), caused by a deficiency in glucose-6-phosphate transporter (G6PT), is characterized by disrupted glucose homeostasis, inflammatory bowel disease, neutropenia, and neutrophil dysfunction. The purpose of this study was to investigate the role of G6PT on macrophage functions and metabolism. Peritoneal macrophages of G6pt-/- mice were lower in number and their effector functions including migration, superoxide production, and phagocytosis were impaired. To investigate the underlying mechanisms of macrophage dysfunction, the G6PT gene was mutated in porcine alveolar macrophage 3D4/31 cells using the CRISPR/Cas9 technology. The G6PT-deficient macrophages exhibited significant decline in cell growth, bactericidal activity, and antiviral response. These phenotypes are associated with the impaired glycolysis and mitochondrial oxidative phosphorylation. We therefore propose that the G6PT-mediated metabolism is essential for effector functions of macrophage, the immune deficiencies observed in GSD-Ib extend beyond neutropenia and neutrophil dysfunction, and future therapeutic targets aimed both the neutrophils and macrophages may be necessary.


Asunto(s)
Antiportadores/genética , Antiportadores/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo I/metabolismo , Macrófagos/metabolismo , Proteínas de Transporte de Monosacáridos/genética , Proteínas de Transporte de Monosacáridos/metabolismo , Animales , Sistemas CRISPR-Cas/genética , Línea Celular , Proliferación Celular , Glucosa/metabolismo , Glucólisis , Humanos , Macrófagos/citología , Ratones , Mitocondrias/metabolismo , Modelos Animales , Mutación , Neutrófilos/metabolismo , Oxidación-Reducción , Fenotipo , Fosforilación , Porcinos
15.
J Virol ; 93(21)2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31434731

RESUMEN

We describe a novel function for the interferon (IFN)-induced protein 44-like (IFI44L) gene in negatively modulating innate immune responses induced after virus infections. Furthermore, we show that decreasing IFI44L expression impairs virus production and that IFI44L expression negatively modulates the antiviral state induced by an analog of double-stranded RNA (dsRNA) or by IFN treatment. The mechanism likely involves the interaction of IFI44L with cellular FK506-binding protein 5 (FKBP5), which in turn interacts with kinases essential for type I and III IFN responses, such as inhibitor of nuclear factor kappa B (IκB) kinase alpha (IKKα), IKKß, and IKKε. Consequently, binding of IFI44L to FKBP5 decreased interferon regulatory factor 3 (IRF-3)-mediated and nuclear factor kappa-B (NF-κB) inhibitor (IκBα)-mediated phosphorylation by IKKε and IKKß, respectively. According to these results, IFI44L is a good target for treatment of diseases associated with excessive IFN levels and/or proinflammatory responses and for reduction of viral replication.IMPORTANCE Excessive innate immune responses can be deleterious for the host, and therefore, negative feedback is needed. Here, we describe a completely novel function for IFI44L in negatively modulating innate immune responses induced after virus infections. In addition, we show that decreasing IFI44L expression impairs virus production and that IFI44L expression negatively modulates the antiviral state induced by an analog of dsRNA or by IFN treatment. IFI44L binds to the cellular protein FKBP5, which in turn interacts with kinases essential for type I and III IFN induction and signaling, such as the kinases IKKα, IKKß, and IKKε. IFI44L binding to FKBP5 decreased the phosphorylation of IRF-3 and IκBα mediated by IKKε and IKKß, respectively, providing an explanation for the function of IFI44L in negatively modulating IFN responses. Therefore, IFI44L is a candidate target for reducing virus replication.


Asunto(s)
Quinasa I-kappa B/metabolismo , Inmunidad Innata/inmunología , Interferones/farmacología , Proteínas de Unión a Tacrolimus/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Replicación Viral , Secuencia de Aminoácidos , Antivirales/farmacología , Retroalimentación Fisiológica , Humanos , Quinasa I-kappa B/genética , Inmunidad Innata/efectos de los fármacos , Gripe Humana/inmunología , Gripe Humana/patología , Gripe Humana/virología , FN-kappa B , Neoplasias/inmunología , Neoplasias/patología , Neoplasias/virología , Orthomyxoviridae/efectos de los fármacos , Orthomyxoviridae/inmunología , Fosforilación , Homología de Secuencia , Transducción de Señal , Proteínas de Unión a Tacrolimus/genética , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor/genética
16.
Cytokine ; 127: 154961, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31901597

RESUMEN

Some of the respiratory viral infections in chickens pose a significant threat to the poultry industry and public health. In response to viral infections, host innate responses provide the first line of defense against viruses, which often act even before the establishment of the infection. Host cells sense the presence of viral components through germinal encoded pattern recognition receptors (PRRs). The engagement of PRRs with pathogen-associated molecular patterns leads to the induction of pro-inflammatory and interferon productions. Induced antiviral responses play a critical role in the outcome of the infections. In order to improve current strategies for control of viral infections or to advance new strategies aimed against viral infections, a deep understanding of host-virus interaction and induction of antiviral responses is required. In this review, we summarized recent progress in understanding innate antiviral responses in chickens with a focus on the avian influenza virus and infectious bronchitis virus.


Asunto(s)
Antivirales/farmacología , Pollos/virología , Infecciones por Coronavirus/tratamiento farmacológico , Virus de la Bronquitis Infecciosa/efectos de los fármacos , Virus de la Influenza A/efectos de los fármacos , Gripe Aviar/tratamiento farmacológico , Infecciones del Sistema Respiratorio/tratamiento farmacológico , Animales , Infecciones por Coronavirus/virología , Humanos , Gripe Aviar/virología , Infecciones del Sistema Respiratorio/virología
17.
Fish Shellfish Immunol ; 106: 920-929, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32931945

RESUMEN

Galectins are well-known ß-galactoside-binding proteins, which play vital roles in innate immune responses of both vertebrates and invertebrates. However, knowledge regarding invertebrate galectins is still in its infancy. With the intention of filling the knowledge gap, here we identified a quadruple domain-containing galectin from marine invertebrate disk abalone, Haliotis discus discus (AbGalec), and characterized it. AbGalec consisted of four distinct carbohydrate-recognition domains (CRDs) and lacked a signal peptide. Expression analysis revealed AbGalec to be ubiquitously expressed in all the examined early embryonic stages of abalone, with highest expression in the 16-cell stage, suggesting the importance of AbGalec in early developmental processes. Tissue distribution analysis revealed the highest expression of AbGalec in abalone mantle, followed by that in gills and hemocytes. Immune challenge experiments revealed significant upregulation of AbGalec at 24 h and 48 h post injection (p.i.) with bacterial and viral components. These results suggested the possible involvement of AbGalec in host defense mechanisms. Polyinosinic: polycytidylic acid (Poly I:C) and viral hemorrhagic septicemia virus (VHSV) injections were capable of inducing AbGalec transcript expression more prominently than bacterial stimulants, thus providing evidence for its role in viral infections. We determined the virus-neutralizing ability of a quadruple domain-containing galectin for the first time, by analyzing the downregulation of VHSV transcripts during the overexpression of AbGalec. Significant downregulation of VHSV transcripts was observed after 24 h and 48 h of post infection. Collectively, our findings reveal the potent antiviral responses of molluscan quadruple domain-containing galectin, AbGalec, along with its involvement in innate immunity.


Asunto(s)
Galectinas/inmunología , Gastrópodos/inmunología , Novirhabdovirus , Infecciones por Rhabdoviridae/inmunología , Animales , Galectinas/genética , Gastrópodos/embriología , Gastrópodos/genética , Gastrópodos/virología , Novirhabdovirus/genética , Poli I-C/farmacología , Dominios Proteicos , Infecciones por Rhabdoviridae/veterinaria , Proteínas Virales/genética
18.
BMC Evol Biol ; 19(1): 149, 2019 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-31337330

RESUMEN

BACKGROUND: Adenosine deaminase enzymes of the ADAR family are conserved in metazoans. They convert adenine into inosine in dsRNAs and thus alter both structural properties and the coding potential of their substrates. Acting on exogenous dsRNAs, ADAR1 exerts a pro- or anti-viral role in vertebrates and Drosophila. RESULTS: We traced 4 ADAR homologs in 14 lophotrochozoan genomes and we classified them into ADAD, ADAR1 or ADAR2, based on phylogenetic and structural analyses of the enzymatic domain. Using RNA-seq and quantitative real time PCR we demonstrated the upregulation of one ADAR1 homolog in the bivalve Crassostrea gigas and in the gastropod Haliotis diversicolor supertexta during Ostreid herpesvirus-1 or Haliotid herpesvirus-1 infection. Accordingly, we demonstrated an extensive ADAR-mediated editing of viral RNAs. Single nucleotide variation (SNV) profiles obtained by pairing RNA- and DNA-seq data from the viral infected individuals resulted to be mostly compatible with ADAR-mediated A-to-I editing (up to 97%). SNVs occurred at low frequency in genomic hotspots, denoted by the overlapping of viral genes encoded on opposite DNA strands. The SNV sites and their upstream neighbor nucleotide indicated the targeting of selected adenosines. The analysis of viral sequences suggested that, under the pressure of the ADAR editing, the two Malacoherpesviridae genomes have evolved to reduce the number of deamination targets. CONCLUSIONS: We report, for the first time, evidence of an extensive editing of Malacoherpesviridae RNAs attributable to host ADAR1 enzymes. The analysis of base neighbor preferences, structural features and expression profiles of molluscan ADAR1 supports the conservation of the enzyme function among metazoans and further suggested that ADAR1 exerts an antiviral role in mollusks.


Asunto(s)
Antivirales/metabolismo , Virus ADN/genética , Moluscos/virología , Edición de ARN/genética , ARN Viral/genética , Proteínas de Unión al ARN/metabolismo , Animales , Teorema de Bayes , Virus ADN/fisiología , Regulación de la Expresión Génica , Genoma Viral , Modelos Moleculares , Moluscos/genética , Filogenia , Polimorfismo de Nucleótido Simple/genética , Dominios Proteicos , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética , Transcriptoma/genética
19.
Fish Shellfish Immunol ; 85: 99-105, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29969707

RESUMEN

The highly effective DNA vaccines against diseases caused by fish rhabdoviruses in farmed fish consist of a DNA plasmid vector encoding the viral glycoprotein under the control of a constitutive cytomegalovirus promoter (CMV). Among others, attempts to improve efficacy and safety of these DNA vaccines have focused on regulatory elements of plasmid vectors, which play a major role in controlling expression levels of vaccine antigens. Depending on the context, use of a fish-derived promoter with minimal activity in mammalian cells could be preferable. Another aspect related to the CMV promoter is that constitutive expression of the vaccine antigen may lead to rapid elimination of antigen expressing cells in the fish and thereby potentially reduce the long-term effects of the vaccine. In this study, we compared DNA vaccines with the interferon-inducible Mx promoter from rainbow trout and the CMV promoter, respectively. Plasmid constructs encoding the enhanced green fluorescent protein (EGFP) were used for the in vitro analysis, whereas DNA vaccines encoding the glycoprotein (G) of the viral haemorrhagic septicaemia virus (VHSV) were applied for the in vivo examination. The in vitro analysis showed that while the DNA vaccine with the CMV promoter constitutively drove the expression of EGFP in both fish and human cell lines, the DNA vaccine with the Mx promoter inducibly enhanced the expression of EGFP in the fish cell line. To address the impact on protection, a time-course model was followed as suggested by Kurath et al. (2006), where vaccinated fish were challenged with VHSV at 2, 8 and 78 weeks post-vaccination (wpv). The DNA vaccine with the CMV promoter protected at all times, while vaccination with the DNA vaccine containing the Mx promoter only protected the fish at 8 wpv. However, following induction with Poly (I:C) one week before the challenge, high protection was also evident at 2 wpv. In conclusion, the results revealed a more fish host dependent activity of the trout Mx promoter compared to the traditionally used cross species-active CMV promoter, but improvements will be needed for its application in DNA vaccines to ensure long term protection.


Asunto(s)
Enfermedades de los Peces/prevención & control , Septicemia Hemorrágica Viral/prevención & control , Novirhabdovirus/inmunología , Oncorhynchus mykiss , Vacunas de ADN/farmacología , Vacunas Virales/farmacología , Animales , Línea Celular , Cyprinidae , Femenino , Enfermedades de los Peces/inmunología , Enfermedades de los Peces/virología , Células HeLa , Septicemia Hemorrágica Viral/inmunología , Septicemia Hemorrágica Viral/virología , Humanos , Interferones/inmunología , Perciformes , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacología , Vacunas de ADN/administración & dosificación , Proteínas Virales de Fusión/administración & dosificación , Proteínas Virales de Fusión/farmacología , Vacunas Virales/administración & dosificación
20.
Fish Shellfish Immunol ; 77: 304-311, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29621634

RESUMEN

L-type lectins (LTLs) play important roles in the secretory pathway of animals, including selective protein trafficking, sorting, and targeting. They have a leguminous lectin domain and can bind to high-mannose-type oligosaccharides. In this study, a novel LTL, designated as MrVIP36, was identified from Macrobrachium rosenbergii. The full-length cDNA of MrVIP36 was 1687 bp with a 972 bp open reading frame encoding a putative protein of 323 deduced amino acids. The deduced MrVIP36 protein contained an LTL-like domain (LTLD) and a transmembrane domain. Phylogenetic tree analysis indicated that MrVIP36 was a member of invertebrate LTLs. It has a closer evolutionary distance with invertebrate LTLs than vertebrate LTLs. Quantitative real time polymerase chain reaction showed that MrVIP36 is expressed widely in all tested tissues, especially in the hepatopancreas and intestine. MrVIP36 was significantly up-regulated in hemocytes of prawns at different time points after Staphylococcus aureus, Vibrio parahaemolyticus, and White spot syndrome virus (WSSV) infections. The recombinant protein MrLTLD (rMrLTLD) could bind and agglutinate all tested bacteria. Sugar binding assay revealed that rMrLTLD could also bind to the glycoconjugates of the bacterial surface, such as lipopolysaccharide and peptidoglycan. Moreover, rMrLTLD could inhibit the growth activities of microorganisms in vitro and accelerate the bacterial clearance in vivo. rMrLTLD could also inhibit WSSV replication in vivo. Survival rate analysis showed that rMrLTLD could protect prawns against WSSV infection. Taken together, our results suggested that MrVIP36 functioned as a pattern recognition receptor involved in the antibacterial and antiviral immune responses of M. rosenbergii.


Asunto(s)
Regulación de la Expresión Génica/inmunología , Inmunidad Innata/genética , Lectinas/genética , Lectinas/inmunología , Palaemonidae/genética , Palaemonidae/inmunología , Animales , Proteínas de Artrópodos/química , Proteínas de Artrópodos/genética , Proteínas de Artrópodos/inmunología , Perfilación de la Expresión Génica , Lectinas/química , Análisis de Secuencia de Proteína , Staphylococcus aureus/fisiología , Vibrio parahaemolyticus/fisiología , Virus del Síndrome de la Mancha Blanca 1/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA