Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.869
Filtrar
1.
J Virol ; 95(22): e0124421, 2021 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-34468176

RESUMEN

Apolipoprotein B mRNA editing enzyme catalytic subunit 3 (APOBEC3) proteins are critical for the control of infection by retroviruses. These proteins deaminate cytidines in negative-strand DNA during reverse transcription, leading to G-to-A changes in coding strands. Uracil DNA glycosylase (UNG) is a host enzyme that excises uracils in genomic DNA, which the base excision repair machinery then repairs. Whether UNG removes uracils found in retroviral DNA after APOBEC3-mediated mutation is not clear, and whether this occurs in vivo has not been demonstrated. To determine if UNG plays a role in the repair of retroviral DNA, we used APOBEC3G (A3G) transgenic mice which we showed previously had extensive deamination of murine leukemia virus (MLV) proviruses. The A3G transgene was crossed onto an Ung and mouse Apobec3 knockout background (UNG-/-APO-/-), and the mice were infected with MLV. We found that virus infection levels were decreased in A3G UNG-/-APO-/- compared with A3G APO-/- mice. Deep sequencing of the proviruses showed that there were significantly higher levels of G-to-A mutations in proviral DNA from A3G transgenic UNG-/-APO-/- than A3G transgenic APO-/- mice, suggesting that UNG plays a role in the repair of uracil-containing proviruses. In in vitro studies, we found that cytoplasmic viral DNA deaminated by APOBEC3G was uracilated. In the absence of UNG, the uracil-containing proviruses integrated at higher levels into the genome than those made in the presence of UNG. Thus, UNG also functions in the nucleus prior to integration by nicking uracil-containing viral DNA, thereby blocking integration. These data show that UNG plays a critical role in the repair of the damage inflicted by APOBEC3 deamination of reverse-transcribed DNA. IMPORTANCE While APOBEC3-mediated mutation of retroviruses is well-established, what role the host base excision repair enzymes play in correcting these mutations is not clear. This question is especially difficult to address in vivo. Here, we use a transgenic mouse developed by our lab that expresses human APOBEC3G and also lacks the endogenous uracil DNA glycosylase (Ung) gene and show that UNG removes uracils introduced by this cytidine deaminase in MLV reverse transcripts, thereby reducing G-to-A mutations in proviruses. Furthermore, our data suggest that UNG removes uracils at two stages in infection-first, in unintegrated nuclear viral reverse-transcribed DNA, resulting in its degradation; and second, in integrated proviruses, resulting in their repair. These data suggest that retroviruses damaged by host cytidine deaminases take advantage of the host DNA repair system to overcome this damage.


Asunto(s)
Desaminasa APOBEC-3G/inmunología , ADN Viral/inmunología , Infecciones por Retroviridae , Retroviridae , Uracil-ADN Glicosidasa/inmunología , Animales , Reparación del ADN , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Células 3T3 NIH , Retroviridae/genética , Retroviridae/inmunología , Infecciones por Retroviridae/inmunología , Infecciones por Retroviridae/virología
2.
Curr Issues Mol Biol ; 43(1): 52-64, 2021 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-33946297

RESUMEN

Koala populations are currently declining and under threat from koala retrovirus (KoRV) infection both in the wild and in captivity. KoRV is assumed to cause immunosuppression and neoplastic diseases, favoring chlamydiosis in koalas. Currently, 10 KoRV subtypes have been identified, including an endogenous subtype (KoRV-A) and nine exogenous subtypes (KoRV-B to KoRV-J). The host's immune response acts as a safeguard against pathogens. Therefore, a proper understanding of the immune response mechanisms against infection is of great importance for the host's survival, as well as for the development of therapeutic and prophylactic interventions. A vaccine is an important protective as well as being a therapeutic tool against infectious disease, and several studies have shown promise for the development of an effective vaccine against KoRV. Moreover, CRISPR/Cas9-based genome editing has opened a new window for gene therapy, and it appears to be a potential therapeutic tool in many viral infections, which could also be investigated for the treatment of KoRV infection. Here, we discuss the recent advances made in the understanding of the immune response in KoRV infection, as well as the progress towards vaccine development against KoRV infection in koalas.


Asunto(s)
Citocinas/inmunología , Phascolarctidae/virología , Infecciones por Retroviridae/prevención & control , Retroviridae/inmunología , Receptores Toll-Like/inmunología , Vacunación/métodos , Animales , Citocinas/metabolismo , Phascolarctidae/inmunología , Infecciones por Retroviridae/inmunología , Infecciones por Retroviridae/virología , Receptores Toll-Like/metabolismo
3.
Viruses ; 13(3)2021 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-33801276

RESUMEN

The SAM and HD domain-containing protein 1 (SAMHD1) is a dNTP triphosphohydrolase that plays a crucial role for a variety of different cellular functions. Besides balancing intracellular dNTP concentrations, facilitating DNA damage repair, and dampening excessive immune responses, SAMHD1 has been shown to act as a major restriction factor against various virus species. In addition to its well-described activity against retroviruses such as HIV-1, SAMHD1 has been identified to reduce the infectivity of different DNA viruses such as the herpesviruses CMV and EBV, the poxvirus VACV, or the hepadnavirus HBV. While some viruses are efficiently restricted by SAMHD1, others have developed evasion mechanisms that antagonize the antiviral activity of SAMHD1. Within this review, we summarize the different cellular functions of SAMHD1 and highlight the countermeasures viruses have evolved to neutralize the restriction factor SAMHD1.


Asunto(s)
Infecciones por Virus ADN/inmunología , Interacciones Huésped-Patógeno/inmunología , Infecciones por Retroviridae/inmunología , Proteína 1 que Contiene Dominios SAM y HD/inmunología , Virus ADN/inmunología , Humanos , Retroviridae/inmunología , Interferencia Viral
4.
Viruses ; 13(3)2021 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-33801908

RESUMEN

An evolutionary arms race has been ongoing between retroviruses and their primate hosts for millions of years. Within the last century, a zoonotic transmission introduced the Human Immunodeficiency Virus (HIV-1), a retrovirus, to the human population that has claimed the lives of millions of individuals and is still infecting over a million people every year. To counteract retroviruses such as this, primates including humans have evolved an innate immune sensor for the retroviral capsid lattice known as TRIM5α. Although the molecular basis for its ability to restrict retroviruses is debated, it is currently accepted that TRIM5α forms higher-order assemblies around the incoming retroviral capsid that are not only disruptive for the virus lifecycle, but also trigger the activation of an antiviral state. More recently, it was discovered that TRIM5α restriction is broader than previously thought because it restricts not only the human retroelement LINE-1, but also the tick-borne flaviviruses, an emergent group of RNA viruses that have vastly different strategies for replication compared to retroviruses. This review focuses on the underlying mechanisms of TRIM5α-mediated restriction of retroelements and flaviviruses and how they differ from the more widely known ability of TRIM5α to restrict retroviruses.


Asunto(s)
Cápside/inmunología , Inmunidad Innata , Virus ARN/inmunología , Virus ARN/metabolismo , Proteínas de Motivos Tripartitos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Factores de Restricción Antivirales , Cápside/metabolismo , Proteínas Portadoras/genética , Flavivirus/inmunología , Flavivirus/metabolismo , Humanos , Virus ARN/clasificación , Virus ARN/genética , Retroviridae/inmunología , Retroviridae/metabolismo , Infecciones por Retroviridae/inmunología , Infecciones por Retroviridae/prevención & control , Proteínas de Motivos Tripartitos/genética , Proteínas de Motivos Tripartitos/inmunología , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/inmunología
5.
Commun Biol ; 4(1): 318, 2021 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-33750893

RESUMEN

Bone morphogenetic protein (BMP) is a kind of classical multi-functional growth factor that plays a vital role in the formation and maintenance of bone, cartilage, muscle, blood vessels, and the regulation of adipogenesis and thermogenesis. However, understanding of the role of BMPs in antiviral immunity is still limited. Here we demonstrate that Bmp8a is a newly-identified positive regulator for antiviral immune responses. The bmp8a-/- zebrafish, when infected with viruses, show reduced antiviral immunity and increased viral load and mortality. We also show for the first time that Bmp8a interacts with Alk6a, which promotes the phosphorylation of Tbk1 and Irf3 through p38 MAPK pathway, and induces the production of type I interferons (IFNs) in response to viral infection. Our study uncovers a previously unrecognized role of Bmp8a in regulation of antiviral immune responses and provides a target for controlling viral infection.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Interferón Tipo I/metabolismo , Infecciones por Retroviridae/virología , Retroviridae/patogenicidad , Proteínas de Pez Cebra/metabolismo , Pez Cebra/virología , Animales , Animales Modificados Genéticamente , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Proteínas Morfogenéticas Óseas/genética , Técnicas de Inactivación de Genes , Interacciones Huésped-Patógeno , Factor 3 Regulador del Interferón/metabolismo , Interferón Tipo I/inmunología , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Retroviridae/crecimiento & desarrollo , Retroviridae/inmunología , Infecciones por Retroviridae/genética , Infecciones por Retroviridae/inmunología , Infecciones por Retroviridae/metabolismo , Transducción de Señal , Carga Viral , Replicación Viral , Pez Cebra/genética , Pez Cebra/inmunología , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
6.
J Vet Diagn Invest ; 33(1): 87-94, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33225861

RESUMEN

CD71 is an immunohistochemical marker used in diagnosing acute myeloid leukemia (AML) M6-Er in humans; however, to our knowledge, it has not been reportedly used for immunohistochemistry in veterinary medicine. We evaluated the pathologic features of AML M6-Er in a retrovirus-negative cat and used CD71 to support the diagnosis. A 4-y-old spayed female Scottish Fold cat was presented with lethargy, anorexia, and fever. Whole-blood PCR assay results for pro feline leukemia virus/pro feline immunodeficiency virus and feline vector-borne diseases were negative. Early erythroid precursors were observed in the peripheral blood smear. Fine-needle aspiration of the enlarged spleen and splenic lymph node showed many early erythroid precursors. Bone marrow aspirate smears revealed erythroid hyperplasia with 68.4% erythroid lineage and 3.6% rubriblasts. Dysplastic cells infiltrated other organs. The patient was diagnosed with myelodysplastic syndrome, progressing to the early phase of AML M6-Er. The patient died on day 121 despite multidrug treatments. Postmortem examination revealed neoplastic erythroblasts infiltrating the bone marrow and other organs. Neoplastic cells were immunopositive for CD71 but immunonegative for CD3, CD20, granzyme B, von Willebrand factor, CD61, myeloperoxidase, and Iba-1. Although further studies are necessary for the application of CD71, our results supported the morphologic diagnosis of AML M6-Er.


Asunto(s)
Antígenos CD/sangre , Enfermedades de los Gatos/diagnóstico , Virus de la Leucemia Felina/aislamiento & purificación , Leucemia Eritroblástica Aguda/veterinaria , Receptores de Transferrina/sangre , Animales , Biomarcadores de Tumor/sangre , Médula Ósea/patología , Enfermedades de los Gatos/sangre , Gatos , Diagnóstico Diferencial , Femenino , Inmunohistoquímica/veterinaria , Leucemia Eritroblástica Aguda/diagnóstico , Retroviridae/inmunología
7.
Virol J ; 17(1): 168, 2020 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-33129323

RESUMEN

Koala retrovirus (KoRV) is believed to be in an active state of endogenization into the koala genome. KoRV is present as both an endogenous and exogenous infection in all koalas in northern Australia. KoRV has been linked to koala pathologies including neoplasia and increased susceptibility to Chlamydia. A KoRV vaccine recently trialled in 10 northern koalas improved antibody response and reduced viral load. This communication reports the expression of key immune genes underlining the innate and adaptive immune response to vaccination in these northern koalas. The results showed that prior to vaccination, IL-8 was expressed at the highest levels, with at least 200-fold greater expression compared to other cytokines, while CD8 mRNA expression was significantly higher than CD4 mRNA expression level. Interferon-γ was up-regulated at both 4- and 8-weeks post-vaccination while IL-8 was down-regulated at 8-weeks post-vaccination.


Asunto(s)
Citocinas/genética , Interferón gamma/genética , Phascolarctidae/virología , Infecciones por Retroviridae/inmunología , Infecciones por Retroviridae/veterinaria , Retroviridae/inmunología , Vacunas Virales/inmunología , Animales , Formación de Anticuerpos , Australia , Estudios de Cohortes , Citocinas/inmunología , Retrovirus Endógenos/genética , Retrovirus Endógenos/inmunología , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Phascolarctidae/inmunología , Retroviridae/genética , Infecciones por Retroviridae/prevención & control , Regulación hacia Arriba , Vacunas Virales/administración & dosificación
8.
J Virol ; 94(24)2020 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-32999021

RESUMEN

Viruses, including retroviruses, can be passed from mothers to their progeny during birth and breastfeeding. It is assumed that newborns may develop immune tolerance to milk-transmitted pathogens similarly to food antigens. I/LnJ mice are uniquely resistant to retroviruses acquired as newborns or as adults as they produce virus-neutralizing antibodies (Abs). A loss-of-function allele of H2-Ob (Ob), originally mapped within the virus infectivity controller 1 (vic1) locus, is responsible for production of antiretrovirus Abs in I/LnJ mice. Importantly, Ob-deficient and vic1 I/LnJ congenic mice on other genetic backgrounds produce antivirus Abs when infected as adults, but not as newborns. We report here that I/LnJ mice carry an additional genetic locus, virus infectivity controller 2 (vic2), that abrogates neonatal immune tolerance to retroviruses. Further genetic analysis mapped the vic2 locus to the telomeric end of chromosome 15. Identification of the vic2 gene and understanding of the related signaling pathways would make blocking of neonatal immune tolerance to retroviruses an achievable goal.IMPORTANCE This work describes a previously unknown genetic mechanism that allows neonates to respond to infections as efficiently as adults.


Asunto(s)
Tolerancia Inmunológica/genética , Infecciones por Retroviridae/inmunología , Retroviridae/inmunología , Animales , Anticuerpos Neutralizantes , Mapeo Cromosómico , Femenino , Ratones , Ratones Congénicos , Ratones Endogámicos BALB C , Ratones Endogámicos , Infecciones por Retroviridae/virología
9.
Elife ; 92020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32930662

RESUMEN

Host antiviral proteins engage in evolutionary arms races with viruses, in which both sides rapidly evolve at interaction interfaces to gain or evade immune defense. For example, primate TRIM5α uses its rapidly evolving 'v1' loop to bind retroviral capsids, and single mutations in this loop can dramatically improve retroviral restriction. However, it is unknown whether such gains of viral restriction are rare, or if they incur loss of pre-existing function against other viruses. Using deep mutational scanning, we comprehensively measured how single mutations in the TRIM5α v1 loop affect restriction of divergent retroviruses. Unexpectedly, we found that the majority of mutations increase weak antiviral function. Moreover, most random mutations do not disrupt potent viral restriction, even when it is newly acquired via a single adaptive substitution. Our results indicate that TRIM5α's adaptive landscape is remarkably broad and mutationally resilient, maximizing its chances of success in evolutionary arms races with retroviruses.


The evolutionary battle between viruses and the immune system is essentially a high-stakes arms race. The immune system makes antiviral proteins, called restriction factors, which can stop the virus from replicating. In response, viruses evolve to evade the effects of restriction factors. To counter this, restriction factors evolve too, and the cycle continues. The challenge for the immune system is that mammals do not evolve as fast as viruses. How then, in the face of this disadvantage, can the immune system hope to keep pace with viral evolution? One human antiviral protein that seems to have struggled to keep up is TRIM5α. In rhesus macaques, it is very effective at stopping the replication of HIV-1 and related viruses. But in humans, it is not effective at all. But why? Protein evolution happens due to small genetic mutations, but not every mutation makes a protein better. If a protein is resilient, it can tolerate lots of neutral or negative mutations without breaking, until it mutates in a way that makes it better. But, if a protein is fragile, even small changes can render it completely unable to do its job. It is possible that restriction factors, like TRIM5α, are evolutionarily 'fragile', and therefore easy to break. But it is difficult to test whether this is the case, because existing mutations have already passed the test of natural selection. This means that either the mutation is somehow useful for the protein, or that it is not harmful enough to be removed. Tenthorey et al. devised a way to introduce all possible changes to the part of TRIM5α that binds to viruses. This revealed that TRIM5α is not fragile; most random mutations increased, rather than decreased, the protein's ability to prevent viral infection. In fact, it appears it would only take a single mutation to make TRIM5α better at blocking HIV-1 in humans, and there are many possible single mutations that would work. Thus, it would appear that human TRIM5α can easily gain the ability to block HIV-1. The next step was to find out whether these gains in antiviral activity are just as easily lost. To do this, Tenthorey et al. performed the same tests on TRIM5α from rhesus macaques and an HIV-blocking mutant version of human TRIM5α. This showed that the majority of random mutations do not break TRIM5α's virus-blocking ability. Thus, TRIM5α can readily gain antiviral activity and, once gained, does not lose it easily during subsequent mutation. Antiviral proteins like TRIM5α engage in uneven evolutionary battles with fast-evolving viruses. But, although they are resilient and able to evolve, they are not always able to find the right mutations on their own. Experiments like these suggest that it might be possible to give them a helping hand. Identifying mutations that help human TRIM5α to strongly block HIV-1 could pave the way for future gene therapy. This step would demand significant advances in gene therapy efficacy and safety, but it could offer a new way to block virus infection in the future.


Asunto(s)
Catarrinos/genética , Interacciones Huésped-Patógeno , Mutación/genética , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas , Animales , Antivirales , Factores de Restricción Antivirales , Células Cultivadas , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Retroviridae/inmunología , Proteínas de Motivos Tripartitos/química , Proteínas de Motivos Tripartitos/genética , Proteínas de Motivos Tripartitos/inmunología , Proteínas de Motivos Tripartitos/metabolismo , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/inmunología , Ubiquitina-Proteína Ligasas/metabolismo , Virosis/inmunología
10.
Viruses ; 12(8)2020 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-32751803

RESUMEN

Retroviruses are major causes of disease in animals and human. Better understanding of the initial host immune response to these viruses could provide insight into how to limit infection. Mouse retroviruses that are endemic in their hosts provide an important genetic tool to dissect the different arms of the innate immune system that recognize retroviruses as foreign. Here, we review what is known about the major branches of the innate immune system that respond to mouse retrovirus infection, Toll-like receptors and nucleic acid sensors, and discuss the importance of these responses in activating adaptive immunity and controlling infection.


Asunto(s)
Inmunidad Innata , Ratones/virología , Infecciones por Retroviridae/inmunología , Retroviridae/inmunología , Animales , Interacciones Microbiota-Huesped/inmunología , Retroviridae/genética , Infecciones por Retroviridae/virología , Replicación Viral
11.
Virus Res ; 290: 198117, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32800804

RESUMEN

OBJECTIVES: Many studies have investigated the utility of hepatitis B virus (HBV) serological markers in HBV-infected patients. However, only a few studies have examined HBV serological markers in HBV-human immunodeficiency virus (HIV) co-infected patients. Here, we conducted a cross-sectional study to evaluate correlations of HBV serological markers in treatment-naïve HBV mono-infected patients and HBV-HIV co-infected patients. METHODS: HBsAg, HBV DNA, HBV RNA, and HBcrAg were quantified in 51 HBV mono-infected patients and 33 HBV-HIV co-infected patients recruited at Tianjin Second People's Hospital from 2016 to 2019. RESULTS: There was no significant difference in serum levels of HBV DNA (P = 0.056), HBV RNA (P = 0.387), HBcrAg (P = 0.714) and HBsAg (P = 0.165) between the patient groups. In HBV mono-infected patients, strong positive correlations were confirmed between HBV RNA and HBV DNA (r=0.620, P < 0.01), HBcrAg and HBV DNA (r=0.802, P < 0.001), and HBcrAg and HBV RNA (r=0.727, P < 0.01). In HBV-HIV co-infected patients, serum HBsAg was very strongly correlated with HBcrAg (r=0.838, P < 0.001). In HBeAg-positive HBV mono-infected patients, all HBV serological markers correlated with each other, whereas only HBV RNA correlated with HBcrAg in HBeAg-negative HBV mono-infected patients (r=0.688, P = 0.007). In HBeAg-positive HBV-HIV co-infected patients, only HBsAg correlated with HBcrAg (r=0.725, P<0.001), whereas HBcrAg and HBV RNA correlated with each other in HBeAg-negative patients (r = 0.683, P=0.010). Moreover, CD4 T-cell counts were not significantly associated with HBsAg, HBV DNA, HBV RNA, and HBcrAg levels. CONCLUSION: Compared with HBsAg and HBV DNA, which are widely used in clinical settings, our study confirmed that new HBV serological markers, such as HBV RNA and HBcrAg, have some utility in HBV mono-infected patients and HBV-HIV co-infected patients for monitoring the progression of liver disease.


Asunto(s)
ADN Viral/sangre , Infecciones por VIH/sangre , Infecciones por VIH/virología , Hepatitis B Crónica/sangre , Hepatitis B Crónica/virología , Adulto , Biomarcadores/sangre , Estudios Transversales , Femenino , Infecciones por VIH/diagnóstico , Antígenos de Superficie de la Hepatitis B/sangre , Antígenos de Superficie de la Hepatitis B/genética , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/inmunología , Hepatitis B Crónica/diagnóstico , Humanos , Masculino , Persona de Mediana Edad , ARN Viral/sangre , ARN Viral/genética , Retroviridae/genética , Retroviridae/inmunología
12.
Methods Enzymol ; 632: 503-519, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32000912

RESUMEN

The interaction strength between CD8+ T cells' TCR and cognate peptide-MHC (pMHC) impacts on the CD8+ T cell response against pathogens and tumors (Martinez-Usatorre, Donda, Zehn, & Romero, 2018; Zehn, Lee, & Bevan, 2009). CD8+ T cell responses against tumors are characterized by the presence of low affinity CD8+ T cells specific for nonmutated tumor associated self-antigens (TAA) and potentially high affinity tumor specific CD8+ T cells recognizing mutated self-antigens (Gros et al., 2016; Kvistborg et al., 2012; McMahan & Slansky, 2007). High affinity T cells display enhanced survival, expansion capacity and tumor control (Martinez-Usatorre et al., 2018; Schmid et al., 2010). In fact, recent clinical trials using neoantigen tumor vaccines showed prolonged progression free survival in melanoma patients (Ott et al., 2017; Sahin et al., 2017), while only modest clinical efficacy was obtained with TAA vaccines (Romero et al., 2016). However, the highly individual nature of neoantigens constitutes a major technical and economical hurdle for routine clinical application. Thus, the characterization of TAA-specific CD8+ T cell responses may reveal new strategies to enhance their anti-tumor properties. In parallel, the identification of high affinity antigens and CD8+ T cells may be essential to design effective tumor vaccines and adoptive cell transfer therapies. Therefore, in this chapter, we describe how to generate tumor cell lines with stable expression of affinity-ranged antigens and methods to assess T-cell affinity.


Asunto(s)
Antígenos de Neoplasias/inmunología , Línea Celular Tumoral/inmunología , Antígenos de Neoplasias/genética , Autoantígenos/genética , Autoantígenos/inmunología , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral/metabolismo , Proliferación Celular , Células Clonales/inmunología , Células Clonales/metabolismo , Células HEK293 , Humanos , Neoplasias/genética , Neoplasias/inmunología , Retroviridae/genética , Retroviridae/inmunología , Transducción Genética
13.
Front Immunol ; 11: 584148, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33488585

RESUMEN

Adoptive T cell therapy (ACT) holds great promise for cancer treatment. One approach, which has regained wide interest in recent years, employs antitumor T cells isolated from tumor lesions ("tumor-infiltrating lymphocytes" or TIL). It is now appreciated that a considerable proportion of anti-melanoma TIL recognize new HLA-binding peptides resulting from somatic mutations, which occurred during tumor progression. The clinical efficacy of TIL can potentially be improved via their genetic modification, designed to enhance their survival, homing capacity, resistance to suppression, tumor killing ability and additional properties of clinical relevance. Successful implementation of such gene-based strategies critically depends on efficient and reproducible protocols for gene delivery into clinical TIL preparations. Here we describe an optimized protocol for the retroviral transduction of TIL. As the experimental system we employed anti-melanoma TIL cultures prepared from four patients, recombinant retrovirus encoding an anti-CD19 chimeric antigen receptor (CAR) as a model gene of interest and CD19+ and CD19- human cell lines serving as target cells. Transduction on day 7 of the rapid expansion protocol (REP) resulted in 69 ± 8% CAR positive TIL. Transduced, but not untransduced TIL, from the four patients responded robustly to CD19+, but not CD19- cell lines, as judged by substantial secretion of IFN-γ following co-culture. In light of the rekindled interest in antitumor TIL, this protocol can be incorporated into a broad range of gene-based approaches for improving the in-vivo survival and functionality of TIL in the clinical setting.


Asunto(s)
Linfocitos Infiltrantes de Tumor/inmunología , Retroviridae/inmunología , Antígenos CD19/inmunología , Línea Celular Tumoral , Humanos , Inmunoterapia Adoptiva/métodos , Interferón gamma/inmunología , Células K562 , Activación de Linfocitos/inmunología , Melanoma/inmunología , Receptores Quiméricos de Antígenos/inmunología
14.
J Med Virol ; 92(3): 394-398, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31670405

RESUMEN

The mechanisms that contribute to retinal tissue destruction during the onset and progression of AIDS-related human cytomegalovirus (HCMV) retinitis remain unclear. Evidence for the stimulation of multiple cell death pathways including apoptosis, necroptosis, and pyroptosis during the pathogenesis of experimental murine cytomegalovirus (MCMV) retinitis in mice with retrovirus-induced immunosuppression (MAIDS) has been reported. Parthanatos is a caspase-independent cell death pathway mediated by rapid overactivation of poly (ADP-ribose) polymerase-1 (PARP-1) and distinct from other cell death pathways. Using the MAIDS model of MCMV retinitis, studies were performed to test the hypothesis that intraocular MCMV infection of mice with MAIDS stimulates parthanatos-associated messenger RNAs (mRNAs) and proteins within the eye during the development of retinal necrosis that takes place by 10 days after MCMV infection. MCMV-infected eyes of MAIDS mice exhibited significant stimulation of PARP-1 mRNA and proteins at 3 days after infection but declined thereafter at 6 and 10 days after infection. Additional studies showed the intraocular stimulation of mRNAs or proteins before MCMV retinitis development for two additional participants in parthanatos, polymer of ADP-ribose and poly (ADP-ribose) glycohydrolase. These results provide new evidence for a role for parthanatos during MAIDS-related MCMV retinitis that may also extend to AIDS-related HCMV retinitis.


Asunto(s)
Retinitis por Citomegalovirus/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Murino/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Murino/virología , Parthanatos , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Animales , Muerte Celular , Retinitis por Citomegalovirus/complicaciones , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Glicósido Hidrolasas/genética , Glicósido Hidrolasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Síndrome de Inmunodeficiencia Adquirida del Murino/complicaciones , Muromegalovirus , Poli(ADP-Ribosa) Polimerasa-1/genética , Poli Adenosina Difosfato Ribosa/genética , Poli Adenosina Difosfato Ribosa/metabolismo , ARN Mensajero/metabolismo , Retina/patología , Retina/virología , Retroviridae/inmunología
15.
J Virol ; 93(21)2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31375593

RESUMEN

Adenovirus (AdV)-based vectors are popular experimental vaccine vectors, but despite their ability to induce strong immune responses, their application is impeded by widespread preexisting immunity against many AdV types that can impair or even abrogate the induction of transgene-specific immune responses. Therefore, the development of vectors based on AdV types with a low seroprevalence is important for effective AdV-based immunization in humans. We investigated the immunization efficacy of vectors based on AdV type 48 (Ad48) and Ad50 in the ovalbumin (ova) model as well as the Friend retrovirus (FV) model, which allows testing of the protective effect of vaccine-induced immunity. Using ova-encoding vectors, we found a significantly lower induction of ova-specific CD8+ T cells and antibody responses by Ad48- and Ad50-based vectors than by Ad5-based vectors. Similarly, we found a reduced induction of FV-specific CD8+ T cell responses in Ad48- and Ad50.Leader-Gag-immunized mice compared with that in Ad5-immunized mice; however, some of those mice were able to control the FV infection, and protection correlated with the level of neutralizing antibodies 10 days after FV challenge. Analyses of the AdV-specific antibodies and CD8+ T cells induced by the individual AdV types revealed a high level of cross-reactivity, and the efficacy of Ad48-based immunization was impaired in Ad5-preimmune mice. Our results show that the immunity induced by Ad48- and Ad50-based vectors is reduced compared to that induced by Ad5 and is sufficient to control FV infection in only some of the immunized mice. A high level of cross-reactivity suggests that AdV preimmunity must be considered even when applying rare AdV-based vectors.IMPORTANCE AdV-based vectors are important tools for the development of vaccines against a wide range of pathogens. While AdV vectors are generally considered safe and highly effective, their application can be severely impaired by preexisting immunity due to the widespread seroprevalence of some AdV types. The characterization of different AdV types with regard to immunogenicity and efficacy in challenge models is of great importance for the development of improved AdV-based vectors that allow for efficient immunization despite anti-AdV immunity. We show that the immunity induced by an Ad48-based vector is inferior to that induced by an Ad5-based vector but can still mediate the control of an FV infection in highly FV-susceptible mice. However, the efficacy of Ad48-based immunization was impaired in Ad5-preimmune mice. Importantly, we found cross-reactivity of both the humoral and cellular immune responses raised by the individual AdV types, suggesting that switching to a different AdV type may not be sufficient to circumvent preexisting anti-AdV immunity.


Asunto(s)
Infecciones por Adenoviridae/inmunología , Adenoviridae/clasificación , Adenoviridae/inmunología , Vacunas contra el Adenovirus/administración & dosificación , Anticuerpos Antivirales/inmunología , Inmunidad Celular/inmunología , Infecciones por Retroviridae/inmunología , Infecciones por Adenoviridae/prevención & control , Infecciones por Adenoviridae/virología , Vacunas contra el Adenovirus/inmunología , Animales , Antígenos Virales/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Femenino , Vectores Genéticos/administración & dosificación , Humanos , Inmunización , Ratones , Ratones Endogámicos BALB C , Ovalbúmina/inmunología , Retroviridae/inmunología , Infecciones por Retroviridae/prevención & control , Infecciones por Retroviridae/virología
16.
Br J Haematol ; 185(4): 679-690, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30828801

RESUMEN

Acute myeloid leukaemia (AML) is a lethal haematological malignancy characterized by an immunosuppressive milieu in the tumour microenvironment (TME) that fosters disease growth and therapeutic resistance. Hypomethylating agents (HMAs) demonstrate clinical efficacy in AML patients and exert immunomodulatory activities. In the present study, we show that guadecitabine augments both antigen processing and presentation, resulting in increased AML susceptibility to T cell-mediated killing. Exposure to HMA results in the activation of the endogenous retroviral pathway with concomitant downstream amplification of critical mediators of inflammation. In an immunocompetent murine leukaemia model, guadecitabine negatively regulates inhibitory accessory cells in the TME by decreasing PD-1 (also termed PDCD1) expressing T cells and reducing AML-mediated expansion of myeloid-derived suppressor cells. Therapy with guadecitabine results in enhanced leukaemia-specific immunity, as manifested by increased CD4 and CD8 cells targeting syngeneic leukaemia cells. We have previously reported that vaccination with AML/dendritic cell fusions elicits the expansion of leukaemia-specific T cells and protects against disease relapse. In the present study, we demonstrate that vaccination in conjunction with HMA therapy results in enhanced anti-leukaemia immunity and survival. The combination of a novel personalized dendritic cell/AML fusion vaccine and an HMA has therapeutic potential, and a clinical trial investigating this combination is planned.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Azacitidina/análogos & derivados , Vacunas contra el Cáncer/inmunología , Leucemia Mieloide Aguda/tratamiento farmacológico , Microambiente Tumoral/inmunología , Animales , Antineoplásicos Inmunológicos/inmunología , Azacitidina/inmunología , Azacitidina/farmacología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Metilación de ADN/efectos de los fármacos , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/inmunología , Humanos , Inmunidad Celular/efectos de los fármacos , Leucemia Mieloide Aguda/inmunología , Ratones Endogámicos C57BL , Trasplante de Neoplasias , Receptor de Muerte Celular Programada 1/metabolismo , Retroviridae/inmunología , Activación Viral/inmunología
17.
Front Immunol ; 10: 54, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30804928

RESUMEN

Cytotoxic CD8+ T lymphocytes (CTL) efficiently control acute virus infections but can become exhausted when a chronic infection develops. The checkpoint receptor PD-1 suppresses the functionality of virus-specific CD8+ T cells during chronic infection. However, the role of the PD-L1/PD-1 pathway during the acute phase of infections has not been well characterized. In the current study the effects of PD-1 or PD-L1 deficiency on the CD8+ T cell response against Friend retroviral (FV) infection of knockout mice was analyzed during acute infection. We observed an enhanced proliferation, functional maturation, and reduced apoptosis of effector CD8+ T cells in the absence of PD-1 or PD-L1. The knockout of PD-L1 had a stronger effect on the functionality of CD8+ T cells than that of PD-1. Augmented CTL responses were associated with an improved control of FV replication. The strong phenotype of FV-infected PD-L1 knockout mice was independent of the interaction with CD80 as an additional receptor for PD-L1. Furthermore, we performed a detailed analysis of the production of different granzymes in virus-specific CD8+ T cells and observed that especially the simultaneous production of multiple granzymes in individual T cells (multifunctionality) was under the control of the PD-1/PD-L1 pathway. The findings from this study allow for a better understanding of the development of antiviral cytotoxic immunity during acute viral infections.


Asunto(s)
Antígeno B7-H1/metabolismo , Receptor de Muerte Celular Programada 1/metabolismo , Infecciones por Retroviridae/inmunología , Infecciones por Retroviridae/metabolismo , Retroviridae/inmunología , Transducción de Señal , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Animales , Apoptosis , Caspasas/metabolismo , Citotoxicidad Inmunológica , Modelos Animales de Enfermedad , Ratones , Ratones Transgénicos , Proteoma , Proteómica/métodos , Infecciones por Retroviridae/virología
18.
Front Immunol ; 9: 2755, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30538707

RESUMEN

Costimulatory and coinhibitory receptors play a key role in regulating immune responses to infection and cancer. Coinhibitory receptors include programmed cell death 1 (PD-1), cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), and T cell immunoglobulin and ITIM domain (TIGIT), which suppress immune responses. Coinhibitory receptors are highly expressed on exhausted virus-specific T cells, indicating that viruses evade host immune responses through enhanced expression of these molecules. Human retroviruses, human immunodeficiency virus (HIV) and human T-cell leukemia virus type 1 (HTLV-1), infect T cells, macrophages and dendritic cells. Therefore, one needs to consider the effects of coinhibitory receptors on both uninfected effector T cells and infected target cells. Coinhibitory receptors are implicated not only in the suppression of immune responses to viruses by inhibition of effector T cells, but also in the persistence of infected cells in vivo. Here we review recent studies on coinhibitory receptors and their roles in retroviral infections such as HIV and HTLV-1.


Asunto(s)
Receptores de Superficie Celular/inmunología , Infecciones por Retroviridae/inmunología , Retroviridae/inmunología , Animales , Humanos , Linfocitos T/inmunología
19.
Front Immunol ; 9: 1260, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29951052

RESUMEN

CD4+ T cell differentiation is influenced by a plethora of intrinsic and extrinsic factors, providing the immune system with the ability to tailor its response according to specific stimuli. Indeed, different classes of pathogens may induce a distinct balance of CD4+ T cell differentiation programmes. Here, we report an uncommonly strong bias toward follicular helper (Tfh) differentiation of CD4+ T cells reactive with a retroviral envelope glycoprotein model antigen, presented in its natural context during retroviral infection. Conversely, the response to the same antigen, presented in different immunization regimens, elicited a response typically balanced between Tfh and T helper 1 cells. Comprehensive quantitation of variables known to influence Tfh differentiation revealed the closest correlation with the strength of T cell receptor (TCR) signaling, leading to PD-1 expression, but not with surface TCR downregulation, irrespective of TCR clonotypic avidity. In contrast, strong TCR signaling leading to TCR downregulation and induction of LAG3 expression in high TCR avidity clonotypes restrained CD4+ T cell commitment and further differentiation. Finally, stunted Th1 differentiation, correlating with limited IL-2 availability in retroviral infection, provided permissive conditions for Tfh development, suggesting that Tfh differentiation is the default program of envelope-reactive CD4+ T cells.


Asunto(s)
Antígenos Virales/inmunología , Linfocitos T CD4-Positivos/inmunología , Infecciones por Retroviridae/inmunología , Retroviridae/inmunología , Animales , Linfocitos T CD4-Positivos/metabolismo , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Citocinas/metabolismo , Perfilación de la Expresión Génica , Ratones , Ratones Noqueados , Ratones Transgénicos , Infecciones por Retroviridae/genética , Infecciones por Retroviridae/virología , Transducción de Señal , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo , Transcriptoma
20.
PLoS Pathog ; 14(2): e1006776, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29447279

RESUMEN

Tight regulation of immune responses is not only critical for preventing autoimmune diseases but also for preventing immunopathological damage during infections in which overactive immune responses may be more harmful for the host than the pathogen itself. Regulatory T cells (Tregs) play a critical role in this regulation, which was discovered using the Friend retrovirus (FV) mouse model. Subsequent FV studies revealed basic biological information about Tregs, including their suppressive activity on effector cells as well as the molecular mechanisms of virus-induced Treg expansion. Treg suppression not only limits immunopathology but also prevents complete elimination of pathogens contributing to chronic infections. Therefore, Tregs play a complex role in the pathogenesis of persistent retroviral infections. New therapeutic concepts to reactivate effector T-cell responses in chronic viral infections by manipulating Tregs also came from work with the FV model. This knowledge initiated many studies to characterize the role of Tregs in HIV pathogenesis in humans, where a complex picture is emerging. On one hand, Tregs suppress HIV-specific effector T-cell responses and are themselves targets of infection, but on the other hand, Tregs suppress HIV-induced immune hyperactivation and thus slow the infection of conventional CD4+ T cells and limit immunopathology. In this review, the basic findings from the FV mouse model are put into perspective with clinical and basic research from HIV studies. In addition, the few Treg studies performed in the simian immunodeficiency virus (SIV) monkey model will also be discussed. The review provides a comprehensive picture of the diverse role of Tregs in different retroviral infections and possible therapeutic approaches to treat retroviral chronicity and pathogenesis by manipulating Treg responses.


Asunto(s)
Interacciones Huésped-Patógeno , Modelos Inmunológicos , Infecciones por Retroviridae/inmunología , Retroviridae/inmunología , Linfocitos T Reguladores/inmunología , Animales , Humanos , Tolerancia Inmunológica , Terapia de Inmunosupresión , Linfopoyesis , Retroviridae/fisiología , Infecciones por Retroviridae/patología , Infecciones por Retroviridae/terapia , Infecciones por Retroviridae/virología , Linfocitos T Reguladores/patología , Linfocitos T Reguladores/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...