Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 176
Filtrar
1.
Blood ; 137(7): 908-922, 2021 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-33174606

RESUMEN

Epigenetic regulation is essential for the maintenance of the hematopoietic system, and its deregulation is implicated in hematopoietic disorders. In this study, UTX, a demethylase for lysine 27 on histone H3 (H3K27) and a component of COMPASS-like and SWI/SNF complexes, played an essential role in the hematopoietic system by globally regulating aging-associated genes. Utx-deficient (UtxΔ/Δ) mice exhibited myeloid skewing with dysplasia, extramedullary hematopoiesis, impaired hematopoietic reconstituting ability, and increased susceptibility to leukemia, which are the hallmarks of hematopoietic aging. RNA-sequencing (RNA-seq) analysis revealed that Utx deficiency converted the gene expression profiles of young hematopoietic stem-progenitor cells (HSPCs) to those of aged HSPCs. Utx expression in hematopoietic stem cells declined with age, and UtxΔ/Δ HSPCs exhibited increased expression of an aging-associated marker, accumulation of reactive oxygen species, and impaired repair of DNA double-strand breaks. Pathway and chromatin immunoprecipitation analyses coupled with RNA-seq data indicated that UTX contributed to hematopoietic homeostasis mainly by maintaining the expression of genes downregulated with aging via demethylase-dependent and -independent epigenetic programming. Of note, comparison of pathway changes in UtxΔ/Δ HSPCs, aged muscle stem cells, aged fibroblasts, and aged induced neurons showed substantial overlap, strongly suggesting common aging mechanisms among different tissue stem cells.


Asunto(s)
Envejecimiento/genética , Regulación de la Expresión Génica/genética , Hematopoyesis/genética , Sistema Hematopoyético/fisiología , Código de Histonas/genética , Histona Demetilasas/fisiología , Animales , Senescencia Celular/genética , Roturas del ADN de Doble Cadena , Reparación del ADN , Femenino , Predisposición Genética a la Enfermedad , Hematopoyesis Extramedular , Histona Demetilasas/deficiencia , Histona Demetilasas/genética , Reconstitución Inmune , Histona Demetilasas con Dominio de Jumonji/metabolismo , Leucemia Experimental/genética , Leucemia Experimental/virología , Masculino , Ratones , Ratones Noqueados , Virus de la Leucemia Murina de Moloney/fisiología , Células Mieloides/patología , Quimera por Radiación , Especies Reactivas de Oxígeno/metabolismo , Proteínas Recombinantes/metabolismo , Factores de Transcripción/metabolismo , Integración Viral
2.
J Virol ; 94(18)2020 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-32641479

RESUMEN

Apolipoprotein B editing enzyme, catalytic polypeptide 3 (APOBEC3) family members are cytidine deaminases that play important roles in intrinsic responses to retrovirus infection. Complex retroviruses like human immunodeficiency virus type 1 (HIV-1) encode the viral infectivity factor (Vif) protein to counteract APOBEC3 proteins. Vif induces degradation of APOBEC3G and other APOBEC3 proteins and thereby prevents their packaging into virions. It is not known if murine leukemia virus (MLV) encodes a Vif-like protein. Here, we show that the MLV P50 protein, produced from an alternatively spliced gag RNA, interacts with the C terminus of mouse APOBEC3 and prevents its packaging without causing its degradation. By infecting APOBEC3 knockout (KO) and wild-type (WT) mice with Friend or Moloney MLV P50-deficient viruses, we found that APOBEC3 restricts the mutant viruses more than WT viruses in vivo Replication of P50-mutant viruses in an APOBEC3-expressing stable cell line was also much slower than that of WT viruses, and overexpressing P50 in this cell line enhanced mutant virus replication. Thus, MLV encodes a protein, P50, that overcomes APOBEC3 restriction by preventing its packaging into virions.IMPORTANCE MLV has existed in mice for at least a million years, in spite of the existence of host restriction factors that block infection. Although MLV is considered a simple retrovirus compared to lentiviruses, it does encode proteins generated from alternatively spliced RNAs. Here, we show that P50, generated from an alternatively spliced RNA encoded in gag, counteracts APOBEC3 by blocking its packaging. MLV also encodes a protein, glycoGag, that increases capsid stability and limits APOBEC3 access to the reverse transcription complex (RTC). Thus, MLV has evolved multiple means of preventing APOBEC3 from blocking infection, explaining its survival as an infectious pathogen in mice.


Asunto(s)
Citidina Desaminasa/genética , Regulación Viral de la Expresión Génica , Productos del Gen gag/genética , Leucemia Experimental/genética , Virus de la Leucemia Murina de Moloney/genética , Infecciones por Retroviridae/genética , Infecciones Tumorales por Virus/genética , Empalme Alternativo , Animales , Cápside/metabolismo , Citidina Desaminasa/deficiencia , Productos del Gen gag/metabolismo , Células HEK293 , Interacciones Huésped-Patógeno/genética , Humanos , Leucemia Experimental/metabolismo , Leucemia Experimental/virología , Ratones , Ratones Noqueados , Virus de la Leucemia Murina de Moloney/metabolismo , Virus de la Leucemia Murina de Moloney/patogenicidad , Células 3T3 NIH , Infecciones por Retroviridae/metabolismo , Infecciones por Retroviridae/virología , Transducción de Señal , Infecciones Tumorales por Virus/metabolismo , Infecciones Tumorales por Virus/virología , Virión/genética , Virión/metabolismo , Virión/patogenicidad , Replicación Viral
3.
J Virol ; 91(21)2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-28794032

RESUMEN

Ecotropic, xenotropic, and polytropic mouse leukemia viruses (E-, X-, and P-MLVs) exist in mice as infectious viruses and endogenous retroviruses (ERVs) inserted into mouse chromosomes. All three MLV subgroups are linked to leukemogenesis, which involves generation of recombinants with polytropic host range. Although P-MLVs are deemed to be the proximal agents of disease induction, few biologically characterized infectious P-MLVs have been sequenced for comparative analysis. We analyzed the complete genomes of 16 naturally occurring infectious P-MLVs, 12 of which were typed for pathogenic potential. We sought to identify ERV progenitors, recombinational hot spots, and segments that are always replaced, never replaced, or linked to pathogenesis or host range. Each P-MLV has an E-MLV backbone with P- or X-ERV replacements that together cover 100% of the recombinant genomes, with different substitution patterns for X- and P-ERVs. Two segments are always replaced, both coding for envelope (Env) protein segments: the N terminus of the surface subunit and the cytoplasmic tail R peptide. Viral gag gene replacements are influenced by host restriction genes Fv1 and Apobec3 Pathogenic potential maps to the env transmembrane subunit segment encoding the N-heptad repeat (HR1). Molecular dynamics simulations identified three novel interdomain salt bridges in the lymphomagenic virus HR1 that could affect structural stability, entry or sensitivity to host immune responses. The long terminal repeats of lymphomagenic P-MLVs are differentially altered by recombinations, duplications, or mutations. This analysis of the naturally occurring, sometimes pathogenic P-MLV recombinants defines the limits and extent of intersubgroup recombination and identifies specific sequence changes linked to pathogenesis and host interactions.IMPORTANCE During virus-induced leukemogenesis, ecotropic mouse leukemia viruses (MLVs) recombine with nonecotropic endogenous retroviruses (ERVs) to produce polytropic MLVs (P-MLVs). Analysis of 16 P-MLV genomes identified two segments consistently replaced: one at the envelope N terminus that alters receptor choice and one in the R peptide at the envelope C terminus, which is removed during virus assembly. Genome-wide analysis shows that nonecotropic replacements in the progenitor ecotropic MLV genome are more extensive than previously appreciated, covering 100% of the genome; contributions from xenotropic and polytropic ERVs differentially alter the regions responsible for receptor determination or subject to APOBEC3 and Fv1 restriction. All pathogenic viruses had modifications in the regulatory elements in their long terminal repeats and differed in a helical segment of envelope involved in entry and targeted by the host immune system. Virus-induced leukemogenesis thus involves generation of complex recombinants, and specific replacements are linked to pathogenesis and host restrictions.


Asunto(s)
Especificidad del Huésped/genética , Virus de la Leucemia Murina/clasificación , Virus de la Leucemia Murina/patogenicidad , Leucemia Experimental/virología , Infecciones por Retroviridae/virología , Infecciones Tumorales por Virus/virología , Proteínas Virales/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Evolución Molecular , Genoma Viral , Virus de la Leucemia Murina/genética , Ratones , Simulación de Dinámica Molecular , Conformación Proteica , Receptores Virales/genética , Receptores Virales/metabolismo , Homología de Secuencia , Secuencias Repetidas Terminales , Proteínas Virales/química , Proteínas Virales/metabolismo
4.
J Virol ; 90(15): 6896-6905, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27194765

RESUMEN

UNLABELLED: During the early steps of infection, retroviruses must direct the movement of the viral genome into the nucleus to complete their replication cycle. This process is mediated by cellular proteins that interact first with the reverse transcription complex and later with the preintegration complex (PIC), allowing it to reach and enter the nucleus. For simple retroviruses, such as murine leukemia virus (MLV), the identities of the cellular proteins involved in trafficking of the PIC in infection are unknown. To identify cellular proteins that interact with the MLV PIC, we developed a replication-competent MLV in which the integrase protein was tagged with a FLAG epitope. Using a combination of immunoprecipitation and mass spectrometry, we established that the microtubule motor dynein regulator DCTN2/p50/dynamitin interacts with the MLV preintegration complex early in infection, suggesting a direct interaction between the incoming viral particles and the dynein complex regulators. Further experiments showed that RNA interference (RNAi)-mediated silencing of either DCTN2/p50/dynamitin or another dynein regulator, NudEL, profoundly reduced the efficiency of infection by ecotropic, but not amphotropic, MLV reporters. We propose that the cytoplasmic dynein regulators are a critical component of the host machinery needed for infection by the retroviruses entering the cell via the ecotropic envelope pathway. IMPORTANCE: Retroviruses must access the chromatin of host cells to integrate the viral DNA, but before this crucial event, they must reach the nucleus. The movement through the cytoplasm-a crowded environment where diffusion is slow-is thought to utilize retrograde transport along the microtubule network by the dynein complex. Different viruses use different components of this multisubunit complex. We found that the preintegration complex of murine leukemia virus (MLV) interacts with the dynein complex and that regulators of this complex are essential for infection. Our study provides the first insight into the requirements for retrograde transport of the MLV preintegration complex.


Asunto(s)
Dineínas/metabolismo , Virus de la Leucemia Murina/fisiología , Leucemia Experimental/virología , Infecciones por Retroviridae/virología , Infecciones Tumorales por Virus/virología , Animales , Genoma Viral , Leucemia Experimental/metabolismo , Ratones , Células 3T3 NIH , Infecciones por Retroviridae/metabolismo , Infecciones Tumorales por Virus/metabolismo
5.
J Virol ; 90(16): 7118-7130, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27226373

RESUMEN

UNLABELLED: Injection of the LP-BM5 murine leukemia virus into mice causes murine AIDS, a disease characterized by many dysfunctions of immunocompetent cells. To establish whether the disease is characterized by glutathione imbalance, reduced glutathione (GSH) and cysteine were quantified in different organs. A marked redox imbalance, consisting of GSH and/or cysteine depletion, was found in the lymphoid organs, such as the spleen and lymph nodes. Moreover, a significant decrease in cysteine and GSH levels in the pancreas and brain, respectively, was measured at 5 weeks postinfection. The Th2 immune response was predominant at all times investigated, as revealed by the expression of Th1/Th2 cytokines. Furthermore, investigation of the activation status of peritoneal macrophages showed that the expression of genetic markers of alternative activation, namely, Fizz1, Ym1, and Arginase1, was induced. Conversely, expression of inducible nitric oxide synthase, a marker of classical activation of macrophages, was detected only when Th1 cytokines were expressed at high levels. In vitro studies revealed that during the very early phases of infection, GSH depletion and the downregulation of interleukin-12 (IL-12) p40 mRNA were correlated with the dose of LP-BM5 used to infect the macrophages. Treatment of LP-BM5-infected mice with N-(N-acetyl-l-cysteinyl)-S-acetylcysteamine (I-152), an N-acetyl-cysteine supplier, restored GSH/cysteine levels in the organs, reduced the expression of alternatively activated macrophage markers, and increased the level of gamma interferon production, while it decreased the levels of Th2 cytokines, such as IL-4 and IL-5. Our findings thus establish a link between GSH deficiency and Th1/Th2 disequilibrium in LP-BM5 infection and indicate that I-152 can be used to restore the GSH level and a balanced Th1/Th2 response in infected mice. IMPORTANCE: The first report of an association between Th2 polarization and alteration of the redox state in LP-BM5 infection is presented. Moreover, it provides evidence that LP-BM5 infection causes a decrease in the thiol content of peritoneal macrophages, which can influence IL-12 production. The restoration of GSH levels by GSH-replenishing molecules can represent a new therapeutic avenue to fight this retroviral infection, as it reestablishes the Th1/Th2 balance. Immunotherapy based on the use of pro-GSH molecules would permit LP-BM5 infection and probably all those viral infections characterized by GSH deficiency and a Th1/Th2 imbalance to be more effectively combated.


Asunto(s)
Glutatión/deficiencia , Virus de la Leucemia Murina/patogenicidad , Leucemia Experimental/complicaciones , Síndrome de Inmunodeficiencia Adquirida del Murino/etiología , Infecciones por Retroviridae/complicaciones , Células Th2/inmunología , Infecciones Tumorales por Virus/complicaciones , Animales , Células Cultivadas , Citocinas/metabolismo , Femenino , Leucemia Experimental/inmunología , Leucemia Experimental/virología , Activación de Linfocitos , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Macrófagos Peritoneales/virología , Ratones , Ratones Endogámicos C57BL , Síndrome de Inmunodeficiencia Adquirida del Murino/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Murino/patología , Infecciones por Retroviridae/inmunología , Infecciones por Retroviridae/virología , Bazo/inmunología , Bazo/metabolismo , Bazo/virología , Células TH1/inmunología , Células TH1/metabolismo , Células TH1/virología , Células Th2/metabolismo , Células Th2/virología , Infecciones Tumorales por Virus/inmunología , Infecciones Tumorales por Virus/virología
6.
Proc Natl Acad Sci U S A ; 111(23): 8595-600, 2014 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-24912157

RESUMEN

The compound immunodeficiencies in nonobese diabetic (NOD) inbred mice homozygous for the Prkdc(scid) and Il2rg(null) alleles (NSG mice) permit engraftment of a wide-range of primary human cells, enabling sophisticated modeling of human disease. In studies designed to define neoplastic stem cells of primary myelofibrosis (PMF), a myeloproliferative neoplasm characterized by profound disruption of the hematopoietic microenvironment, we observed a high frequency of acute myeloid leukemia (AML) in NSG mice. AML was of mouse origin, confined to PMF-xenografted mice, and contained multiple clonal integrations of ecotropic murine leukemia virus (E-MuLV). Significantly, MuLV replication was not only observed in diseased mice, but also in nontreated NSG controls. Furthermore, in addition to the single ecotropic endogenous retrovirus (eERV) located on chromosome 11 (Emv30) in the NOD genome, multiple de novo germ-line eERV integrations were observed in mice from each of four independent NSG mouse colonies. Analysis confirmed that E-MuLV originated from the Emv30 provirus and that recombination events were not necessary for virus replication or AML induction. Pathogenicity is thus likely attributable to PMF-mediated paracrine stimulation of mouse myeloid cells, which serve as targets for retroviral infection and transformation, as evidenced by integration into the Evi1 locus, a hotspot for retroviral-induced myeloid leukemia. This study thus corroborates a role of paracrine stimulation in PMF disease progression, underlines the importance of target cell type and numbers in MuLV-induced disease, and mandates awareness of replicating MuLV in NOD immunodeficient mice, which can significantly influence experimental results and their interpretation.


Asunto(s)
Retrovirus Endógenos/genética , Leucemia Experimental/genética , Leucemia Mieloide Aguda/genética , Mielofibrosis Primaria/genética , Anciano , Animales , Southern Blotting , Femenino , Humanos , Subunidad gamma Común de Receptores de Interleucina/genética , Subunidad gamma Común de Receptores de Interleucina/metabolismo , Virus de la Leucemia Murina/genética , Leucemia Experimental/patología , Leucemia Experimental/virología , Leucemia Mieloide Aguda/patología , Leucemia Mieloide Aguda/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Ratones Transgénicos , Persona de Mediana Edad , Datos de Secuencia Molecular , Mielofibrosis Primaria/patología , Mielofibrosis Primaria/virología , Provirus/genética , Trasplante Heterólogo , Integración Viral/genética , Replicación Viral/genética , Adulto Joven
7.
J Biol Chem ; 290(45): 27345-27359, 2015 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-26370074

RESUMEN

Siglec-1 (sialoadhesin, CD169) is a surface receptor on human cells that mediates trans-enhancement of HIV-1 infection through recognition of sialic acid moieties in virus membrane gangliosides. Here, we demonstrate that mouse Siglec-1, expressed on the surface of primary macrophages in an interferon-α-responsive manner, captures murine leukemia virus (MLV) particles and mediates their transfer to proliferating lymphocytes. The MLV infection of primary B-cells was markedly more efficient than that of primary T-cells. The major structural protein of MLV particles, Gag, frequently co-localized with Siglec-1, and trans-infection, primarily of surface-bound MLV particles, efficiently occurred. To explore the role of sialic acid for MLV trans-infection at a submolecular level, we analyzed the potential of six sialic acid precursor analogs to modulate the sialylated ganglioside-dependent interaction of MLV particles with Siglec-1. Biosynthetically engineered sialic acids were detected in both the glycolipid and glycoprotein fractions of MLV producer cells. MLV released from cells carrying N-acyl-modified sialic acids displayed strikingly different capacities for Siglec-1-mediated capture and trans-infection; N-butanoyl, N-isobutanoyl, N-glycolyl, or N-pentanoyl side chain modifications resulted in up to 92 and 80% reduction of virus particle capture and trans-infection, respectively, whereas N-propanoyl or N-cyclopropylcarbamyl side chains had no effect. In agreement with these functional analyses, molecular modeling indicated reduced binding affinities for non-functional N-acyl modifications. Thus, Siglec-1 is a key receptor for macrophage/lymphocyte trans-infection of surface-bound virions, and the N-acyl side chain of sialic acid is a critical determinant for the Siglec-1/MLV interaction.


Asunto(s)
Virus de la Leucemia Murina de Moloney/patogenicidad , Lectina 1 Similar a Ig de Unión al Ácido Siálico/química , Lectina 1 Similar a Ig de Unión al Ácido Siálico/fisiología , Animales , Sitios de Unión , Línea Celular , Gangliósidos/química , Gangliósidos/metabolismo , Interacciones Huésped-Patógeno/fisiología , Humanos , Interferón-alfa/fisiología , Leucemia Experimental/fisiopatología , Leucemia Experimental/virología , Linfocitos/fisiología , Linfocitos/virología , Macrófagos/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Moleculares , Virus de la Leucemia Murina de Moloney/genética , Virus de la Leucemia Murina de Moloney/fisiología , Ácido N-Acetilneuramínico/química , Receptores Virales/química , Receptores Virales/fisiología , Infecciones por Retroviridae/fisiopatología , Infecciones por Retroviridae/virología , Lectina 1 Similar a Ig de Unión al Ácido Siálico/genética , Infecciones Tumorales por Virus/fisiopatología , Infecciones Tumorales por Virus/virología
8.
J Virol ; 89(2): 1468-73, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25378499

RESUMEN

Toll-like receptor 7 and Myd88 are required for antiretroviral antibody and germinal center responses, but whether somatic hypermutation and class-switch recombination are required for antiretroviral immunity has not been examined. Mice deficient in activation-induced cytidine deaminase (AID) resisted Friend virus infection, produced virus-neutralizing antibodies, and controlled viremia. Passive transfer demonstrated that immune IgM from AID-deficient mice contributes to Friend virus control in the presence of virus-specific CD4+ T cells.


Asunto(s)
Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Virus de la Leucemia Murina de Friend/inmunología , Leucemia Experimental/inmunología , Infecciones por Retroviridae/inmunología , Hipermutación Somática de Inmunoglobulina , Infecciones Tumorales por Virus/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Citidina Desaminasa/deficiencia , Inmunización Pasiva , Cambio de Clase de Inmunoglobulina , Inmunoglobulina M/inmunología , Leucemia Experimental/virología , Ratones , Infecciones por Retroviridae/virología , Infecciones Tumorales por Virus/virología
9.
J Virol ; 89(1): 155-64, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25320301

RESUMEN

UNLABELLED: Interleukin-1 beta (IL-1ß) is an inflammatory cytokine that is secreted in response to inflammasome activation by innate microbe-sensing pathways. Although some retroviruses can trigger IL-1ß secretion through the DNA-sensing molecule IFI16, the effect of IL-1ß on the course of infection is unknown. To test whether IL-1ß secretion affects retroviral replication in vivo, I constructed a novel murine leukemia virus strain (FMLV-IL-1ß) that encodes the mature form of IL-1ß. This virus replicated with kinetics similar to that of wild-type virus in tissue culture but caused a dramatically more aggressive infection of both C57BL/6 and BALB/c mice. By 7 days postinfection (PI), mice infected with FMLV-IL-1ß exhibited splenomegaly and viral loads 300-fold higher than those in mice infected with wild-type FMLV. Furthermore, the enlarged spleens of FMLV-IL-1ß-infected mice correlated with a large expansion of Gr-1(+) CD11b(+) myeloid-derived suppressor cells, as well as elevated levels of immune activation. Although FMLV-IL-1ß infection was controlled by C57BL/6 mice by 14 days p.i., FMLV-IL-1ß was able to establish a significant persistent infection and immune activation in BALB/c mice. These results demonstrate that IL-1ß secretion is a powerful positive regulator of retroviral infection and that FMLV-IL-1ß represents a new model of proinflammatory retroviral infection. IMPORTANCE: Interleukin-1 beta (IL-1ß) is an inflammatory cytokine released in response to activation of innate pathogen-sensing pathways during microbial infection. To examine the potential impact of IL-1ß on retroviral replication in vivo, I constructed a novel mouse retrovirus strain (FMLV-IL-1ß) that encodes IL-1ß and promotes abundant IL-1ß secretion from infected cells. This virus replicates with normal kinetics in cultured cells but displays a dramatically enhanced ability to replicate in mice and caused persistent infection and immune activation in the BALB/c strain of mice. These results establish IL-1ß as a positive regulator of retroviral replication and suggest that targeting this pathway may have therapeutic benefits in infections with proinflammatory retroviruses. This virus can also be used to further study the impact of inflammatory pathways on retroviral infection.


Asunto(s)
Interleucina-1beta/metabolismo , Virus de la Leucemia Murina/inmunología , Virus de la Leucemia Murina/patogenicidad , Leucemia Experimental/patología , Infecciones por Retroviridae/patología , Infecciones Tumorales por Virus/patología , Animales , Interacciones Huésped-Patógeno , Interleucina-1beta/genética , Virus de la Leucemia Murina/genética , Virus de la Leucemia Murina/fisiología , Leucemia Experimental/virología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Infecciones por Retroviridae/virología , Esplenomegalia/patología , Infecciones Tumorales por Virus/virología , Carga Viral , Virulencia , Replicación Viral
10.
Proc Biol Sci ; 282(1798): 20141568, 2015 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-25392466

RESUMEN

Reduced genetic variation among hosts may favour the emergence of virulent infectious diseases by enhancing pathogen replication and its associated virulence due to adaptation to a limited set of host genotypes. Here, we test this hypothesis using experimental evolution of a mouse-specific retroviral pathogen, Friend virus (FV) complex. We demonstrate rapid fitness (i.e. viral titre) and virulence increases when FV complex serially infects a series of inbred mice representing the same genotype, but not when infecting a diverse array of inbred mouse strains modelling the diversity in natural host populations. Additionally, a single infection of a different host genotype was sufficient to constrain the emergence of a high fitness/high virulence FV complex phenotype in these experiments. The potent inhibition of viral fitness and virulence was associated with an observed loss of the defective retroviral genome (spleen focus-forming virus), whose presence exacerbates infection and drives disease in susceptible mice. Results from our experiments provide an important first step in understanding how genetic variation among vertebrate hosts influences pathogen evolution and suggests that serial exposure to different genotypes within a single host species may act as a constraint on pathogen adaptation that prohibits the emergence of more virulent infections. From a practical perspective, these results have implications for low-diversity host populations such as endangered species and domestic animals.


Asunto(s)
Virus de la Leucemia Murina de Friend/fisiología , Virus de la Leucemia Murina de Friend/patogenicidad , Aptitud Genética , Genotipo , Interacciones Huésped-Patógeno/genética , Leucemia Experimental/genética , Infecciones por Retroviridae/genética , Infecciones Tumorales por Virus/genética , Animales , Evolución Biológica , Femenino , Leucemia Experimental/virología , Ratones , Ratones Endogámicos , Infecciones por Retroviridae/virología , Organismos Libres de Patógenos Específicos , Infecciones Tumorales por Virus/virología , Virulencia/fisiología
11.
Retrovirology ; 11: 126, 2014 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-25539593

RESUMEN

BACKGROUND: Pathogen recognition drives host defense towards viral infections. Specific groups rather than single members of the protein family of pattern recognition receptors (PRRs) such as membrane spanning Toll-like receptors (TLRs) and cytosolic helicases might mediate sensing of replication intermediates of a specific virus species. TLR7 mediates host sensing of retroviruses and could significantly influence retrovirus-specific antibody responses. However, the origin of efficient cell-mediated immunity towards retroviruses is unknown. Double-stranded RNA intermediates produced during retroviral replication are good candidates for immune stimulatory viral products. Thus, we considered TLR3 as primer of cell-mediated immunity against retroviruses in vivo. RESULTS: Infection of mice deficient in TLR3 (TLR3(-/-)) with Friend retrovirus (FV) complex revealed higher viral loads during acute retroviral infection compared to wild type mice. TLR3(-/-) mice exhibited significantly lower expression levels of type I interferons (IFNs) and IFN-stimulated genes like Pkr or Ifi44, as well as reduced numbers of activated myeloid dendritic cells (DCs) (CD86(+) and MHC-II(+)). DCs generated from FV-infected TLR3(-/-) mice were less capable of priming virus-specific CD8(+) T cell proliferation. Moreover, cytotoxicity of natural killer (NK) cells as well as CD8(+) T cells were reduced in vitro and in vivo, respectively, in FV-infected TLR3(-/-) mice. CONCLUSIONS: TLR3 mediates antiretroviral cytotoxic NK cell and CD8(+) T cell activity in vivo. Our findings qualify TLR3 as target of immune therapy against retroviral infections.


Asunto(s)
Virus de la Leucemia Murina de Friend/inmunología , Receptor Toll-Like 3/metabolismo , Animales , Linfocitos T CD8-positivos/inmunología , Femenino , Células Asesinas Naturales/inmunología , Leucemia Experimental/inmunología , Leucemia Experimental/virología , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Retroviridae/inmunología , Infecciones por Retroviridae/virología , Receptor Toll-Like 3/deficiencia , Infecciones Tumorales por Virus/inmunología , Infecciones Tumorales por Virus/virología , Carga Viral
12.
J Immunol ; 189(7): 3724-33, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-22956581

RESUMEN

Distal symmetrical polyneuropathy is the most common form of HIV infection-associated peripheral neuropathy and is often associated with pain. C57BL/6 (B6) mice infected with LP-BM5, a murine retroviral isolate, develop a severe immunodeficiency syndrome similar to that in humans infected with HIV-1, hence the term murine AIDS. We investigated the induction of peripheral neuropathy after LP-BM5 infection in B6 mice. Infected B6 mice, like HIV-infected humans, exhibited behavioral (increased sensitivity to mechanical and heat stimuli) and pathological (transient loss of intraepidermal nerve fibers) signs of peripheral neuropathy. The levels of viral gag RNA were significantly increased in all tissues tested, including spleen, paw skin, lumbar dorsal root ganglia, and lumbar spinal cord, postinfection (p.i.). Correlated with the development of peripheral neuropathy, the tissue levels of several cytokines, including IFN-γ, IL-1ß, IL-6, and IL-12, were significantly elevated p.i. These increases had cytokine-specific and tissue-specific profiles and kinetics. Further, treatment with the antiretroviral agent zidovudine either significantly reduced or completely reversed the aforementioned behavioral, pathologic, and cytokine changes p.i. These data suggest that LP-BM5 infection is a potential mouse model of HIV-associated distal symmetrical polyneuropathy that can be used for investigating the roles of various cytokines in infection-induced neuropathic pain. Further investigation of this model could give a better understanding of, and lead to more effective treatments for, HIV infection-associated painful peripheral neuropathy.


Asunto(s)
Citocinas/biosíntesis , Virus de la Leucemia Murina/inmunología , Síndrome de Inmunodeficiencia Adquirida del Murino/inmunología , Síndrome de Inmunodeficiencia Adquirida del Murino/metabolismo , Enfermedades del Sistema Nervioso Periférico/inmunología , Enfermedades del Sistema Nervioso Periférico/virología , Animales , Citocinas/genética , Modelos Animales de Enfermedad , Hipersensibilidad/inmunología , Hipersensibilidad/metabolismo , Hipersensibilidad/virología , Virus de la Leucemia Murina/aislamiento & purificación , Leucemia Experimental/inmunología , Leucemia Experimental/metabolismo , Leucemia Experimental/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedades del Sistema Nervioso Periférico/metabolismo , ARN Mensajero/biosíntesis , Infecciones por Retroviridae/inmunología , Infecciones por Retroviridae/metabolismo
13.
J Immunol ; 189(5): 2521-9, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22821964

RESUMEN

The immune system is tasked with defending against a myriad of microbial infections, and its response to a given infectious microbe may be strongly influenced by coinfection with another microbe. It was shown that infection of mice with lactate dehydrogenase-elevating virus (LDV) impairs early adaptive immune responses to Friend virus (FV) coinfection. To investigate the mechanism of this impairment, we examined LDV-induced innate immune responses and found LDV-specific induction of IFN-α and IFN-γ. LDV-induced IFN-α had little effect on FV infection or immune responses, but unexpectedly, LDV-induced IFN-γ production dampened Th1 adaptive immune responses and enhanced FV infection. Two distinct effects were identified. First, LDV-induced IFN-γ signaling indirectly modulated FV-specific CD8+ T cell responses. Second, intrinsic IFN-γ signaling in B cells promoted polyclonal B cell activation and enhanced early FV infection, despite promotion of germinal center formation and neutralizing Ab production. Results from this model reveal that IFN-γ production can have detrimental effects on early adaptive immune responses and virus control.


Asunto(s)
Inmunidad Adaptativa , Regulación hacia Abajo/inmunología , Interferón gamma/fisiología , Virus de la Leucemia Murina/inmunología , Infecciones por Retroviridae/inmunología , Inmunidad Adaptativa/genética , Animales , Modelos Animales de Enfermedad , Regulación hacia Abajo/genética , Femenino , Virus de la Leucemia Murina de Friend/inmunología , Virus de la Leucemia Murina de Friend/patogenicidad , Interferón gamma/deficiencia , Interferón gamma/genética , Virus Elevador de Lactato Deshidrogenasa/inmunología , Virus Elevador de Lactato Deshidrogenasa/patogenicidad , Virus de la Leucemia Murina/patogenicidad , Leucemia Experimental/genética , Leucemia Experimental/inmunología , Leucemia Experimental/virología , Ratones , Ratones Congénicos , Ratones Endogámicos A , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Infecciones por Retroviridae/genética , Infecciones por Retroviridae/patología , Virus Formadores de Foco en el Bazo/inmunología , Virus Formadores de Foco en el Bazo/patogenicidad , Infecciones Tumorales por Virus/genética , Infecciones Tumorales por Virus/inmunología , Infecciones Tumorales por Virus/virología
14.
Retrovirology ; 10: 58, 2013 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-23738889

RESUMEN

BACKGROUND: Co-infection of HIV patients with cytomegalovirus (CMV) is associated with enhanced AIDS progression and CMV end-organ diseases. On the other hand, persistent CMV infection has recently been shown to decrease tumor relapse and protect against lethal bacterial infection. The influence of persistent CMV on the outcome of an acute retroviral superinfection is still unknown. RESULTS: Here we show that a persistent murine CMV (mCMV) infection surprisingly confers higher resistance to a primary Friend retrovirus infection (FV) of mice. Decreased FV titers and augmented FV-specific CD8 T-cell responses were found in mCMV infected mice during primary FV superinfection. NK cells produced higher amounts of IFNgamma after FV infection of persistently mCMV infected mice suggesting that these cells were involved in the 'protective' effect. Depletion of NK1.1+ cells or neutralization of IFNgamma during FV superinfection abrogated the mCMV-mediated effect. CONCLUSION: Our data demonstrate for the first time that a persistent CMV infection induces long-lasting NK cell responses that can enhance immunity to primary retroviral infections. To our knowledge, studies investigating primary HIV infection have not analyzed the role of the CMV seropositivity in these patients. Our observations suggest that NK cells in CMV seropositive individuals might contribute to the control of primary HIV infection.


Asunto(s)
Virus de la Leucemia Murina de Friend/inmunología , Infecciones por Herpesviridae/inmunología , Células Asesinas Naturales/inmunología , Leucemia Experimental/inmunología , Muromegalovirus/inmunología , Infecciones por Retroviridae/inmunología , Infecciones Tumorales por Virus/inmunología , Animales , Coinfección/inmunología , Coinfección/virología , Modelos Animales de Enfermedad , Resistencia a la Enfermedad , Infecciones por Herpesviridae/virología , Leucemia Experimental/virología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Infecciones por Retroviridae/virología , Infecciones Tumorales por Virus/virología
15.
Stem Cells ; 30(2): 121-30, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22083997

RESUMEN

Friend virus induces erythroleukemia through a characteristic two-stage progression. The prevailing model proposes that during the initial, polyclonal stage of disease most of the infected cells terminally differentiate, resulting in acute erythrocytosis. In the late stage of disease, a clonal leukemia develops through the acquisition of new mutations--proviral insertional activation of Spi1/Pu.1 and mutation of p53. Previous work from our laboratory demonstrated that Friend virus activates the bone morphogenic protein 4 (BMP4)-dependent stress erythropoiesis pathway, which leads to the rapid expansion of stress erythroid progenitors, which are the targets for Friend virus in the spleen. We recently showed that stress erythroid progenitors have intrinsic self-renewal ability and therefore could function as leukemia stem cells (LSCs) when infected with Friend virus. Here, we show that the two stages of Friend virus-induced disease are caused by infection of distinct stress progenitor populations in the spleen. The development of leukemia relies on the ability of the virus to hijack the intrinsic self-renewal capability of stress erythroid progenitors leading to the generation of LSCs. Two signals are required for the self-renewal of Friend virus LSCs proviral insertional activation of Spi1/Pu.1 and Hedgehog-dependent signaling. Surprisingly, mutation of p53 is not observed in LSCs. These data establish a new model for Friend virus-induced erythroleukemia and demonstrate the utility of Friend virus as a model system to study LSC self-renewal.


Asunto(s)
Virus de la Leucemia Murina de Friend , Proteínas Hedgehog/metabolismo , Leucemia Eritroblástica Aguda/patología , Leucemia Experimental/patología , Células Madre Neoplásicas/fisiología , Proteínas Proto-Oncogénicas/genética , Infecciones por Retroviridae/patología , Transactivadores/genética , Proteína p53 Supresora de Tumor/genética , Infecciones Tumorales por Virus/patología , Integración Viral , Animales , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Virus de la Leucemia Murina de Friend/genética , Leucemia Eritroblástica Aguda/virología , Leucemia Experimental/virología , Ratones , Ratones Endogámicos BALB C , Policitemia , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Provirus , Infecciones por Retroviridae/virología , Transducción de Señal , Bazo/patología , Transactivadores/metabolismo , Infecciones Tumorales por Virus/virología , Proteínas del Envoltorio Viral/metabolismo
16.
Virol J ; 10: 130, 2013 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-23617998

RESUMEN

BACKGROUND: ZASC1 is a zinc finger-containing transcription factor that was previously shown to bind to specific DNA binding sites in the Moloney murine leukemia virus (Mo-MuLV) promoter and is required for efficient viral mRNA transcription (J. Virol. 84:7473-7483, 2010). METHODS: To determine whether this cellular factor influences Mo-MuLV replication and viral disease pathogenesis in vivo, we generated a ZASC1 knockout mouse model and completed both early infection and long term disease pathogenesis studies. RESULTS: Mice lacking ZASC1 were born at the expected Mendelian ratio and showed no obvious physical or behavioral defects. Analysis of bone marrow samples revealed a specific increase in a common myeloid progenitor cell population in ZASC1-deficient mice, a result that is of considerable interest because osteoclasts derived from the myeloid lineage are among the first bone marrow cells infected by Mo-MuLV (J. Virol. 73: 1617-1623, 1999). Indeed, Mo-MuLV infection of neonatal mice revealed that ZASC1 is required for efficient early virus replication in the bone marrow, but not in the thymus or spleen. However, the absence of ZASC1 did not influence the timing of subsequent tumor progression or the types of tumors resulting from virus infection. CONCLUSIONS: These studies have revealed that ZASC1 is important for myeloid cell differentiation in the bone marrow compartment and that this cellular factor is required for efficient Mo-MuLV replication in this tissue at an early time point post-infection.


Asunto(s)
Médula Ósea/virología , Proteínas de Unión al ADN/metabolismo , Leucemia Experimental/virología , Virus de la Leucemia Murina de Moloney/fisiología , Proteínas Nucleares/metabolismo , Infecciones por Retroviridae/virología , Infecciones Tumorales por Virus/virología , Replicación Viral , Animales , Proteínas de Unión al ADN/deficiencia , Interacciones Huésped-Patógeno , Ratones , Ratones Noqueados , Proteínas Nucleares/deficiencia
17.
Int J Cancer ; 131(5): 1083-94, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22034044

RESUMEN

Infection of erythroid cells by Friend spleen focus-forming virus (SFFV) leads to acute erythroid hyperplasia in mice, due to expression of its unique envelope glycoprotein, gp55. Erythroid cells expressing SFFV gp55 proliferate in the absence of their normal regulator, erythropoietin, because of the interaction among the viral envelope protein, the erythropoietin receptor, and a short form of the receptor tyrosine kinase Stk (sf-Stk). This leads to constitutive activation of several signal transduction pathways. Our previous studies showed that sf-Stk interacts with SFFV gp55, forming disulfide-linked complexes. This covalent interaction, along with other noncovalent interactions with SFFV-gp55, results in constitutive tyrosine phosphorylation of sf-Stk and rodent fibroblast transformation. Here, we determined the precise amino acid region within sf-Stk that contributes to fibroblast transformation by the polycythemia-inducing (SFFV-P) and the anemia-inducing (SFFV-A) strains of SFFV. Sf-Stk deletion mutants showed different transforming abilities in fibroblasts infected with SFFV-P and SFFV-A, although the N-terminal extracellular domain of sf-Stk was essential for fibroblast transformation by both viruses. Point mutations of sf-Stk indicated that cysteine 19 was critical for fibroblast transformation by SFFV-P, although all four cysteines (8, 19, 37 and 42) appeared to be important for fibroblast transformation by both SFFV-P and SFFV-A. Mutation of sf-Stk cysteine 19 abolished its ability to form dimers with SFFV-P and SFFV-A gp55. These results suggest that the interaction between sf-Stk and the envelope proteins of the polycythemia- and anemia-inducing variants of SFFV is architecturally different.


Asunto(s)
Anemia/etiología , Transformación Celular Neoplásica/patología , Fibroblastos/patología , Leucemia Experimental/genética , Policitemia/etiología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Infecciones por Retroviridae/genética , Virus Formadores de Foco en el Bazo/genética , Infecciones Tumorales por Virus/genética , Secuencia de Aminoácidos , Anemia/metabolismo , Anemia/patología , Animales , Western Blotting , Transformación Celular Neoplásica/metabolismo , Células Cultivadas , Fibroblastos/metabolismo , Fibroblastos/virología , Humanos , Leucemia Experimental/metabolismo , Leucemia Experimental/virología , Ratones , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Mutación/genética , Fosforilación , Plásmidos/genética , Policitemia/metabolismo , Policitemia/patología , Estructura Terciaria de Proteína , Proteínas Tirosina Quinasas Receptoras/genética , Infecciones por Retroviridae/metabolismo , Infecciones por Retroviridae/virología , Homología de Secuencia de Aminoácido , Transducción de Señal , Infecciones Tumorales por Virus/metabolismo , Infecciones Tumorales por Virus/virología , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo
18.
PLoS Pathog ; 6(11): e1001183, 2010 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-21085616

RESUMEN

The p12 protein is a cleavage product of the Gag precursor of the murine leukemia virus (MLV). Specific mutations in p12 have been described that affect early stages of infection, rendering the virus replication-defective. Such mutants showed normal generation of genomic DNA but no formation of circular forms, which are markers of nuclear entry by the viral DNA. This suggested that p12 may function in early stages of infection but the precise mechanism of p12 action is not known. To address the function and follow the intracellular localization of the wt p12 protein, we generated tagged p12 proteins in the context of a replication-competent virus, which allowed for the detection of p12 at early stages of infection by immunofluorescence. p12 was found to be distributed to discrete puncta, indicative of macromolecular complexes. These complexes were localized to the cytoplasm early after infection, and thereafter accumulated adjacent to mitotic chromosomes. This chromosomal accumulation was impaired for p12 proteins with a mutation that rendered the virus integration-defective. Immunofluorescence demonstrated that intracellular p12 complexes co-localized with capsid, a known constituent of the MLV pre-integration complex (PIC), and immunofluorescence combined with fluorescent in situ hybridization (FISH) revealed co-localization of the p12 proteins with the incoming reverse transcribed viral DNA. Interactions of p12 with the capsid and with the viral DNA were also demonstrated by co-immunoprecipitation. These results imply that p12 proteins are components of the MLV PIC. Furthermore, a large excess of wt PICs did not rescue the defect in integration of PICs derived from mutant p12 particles, demonstrating that p12 exerts its function as part of this complex. Altogether, these results imply that p12 proteins are constituent of the MLV PIC and function in directing the PIC from the cytoplasm towards integration.


Asunto(s)
Productos del Gen gag/química , Productos del Gen gag/metabolismo , Virus de la Leucemia Murina/fisiología , Leucemia Experimental/metabolismo , Infecciones por Retroviridae/metabolismo , Infecciones Tumorales por Virus/metabolismo , Ensamble de Virus , Animales , Western Blotting , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Neoplasias Óseas/virología , Células Cultivadas , Cromosomas de los Mamíferos/genética , ADN Viral/genética , ADN Viral/metabolismo , Fibroblastos/metabolismo , Fibroblastos/virología , Técnica del Anticuerpo Fluorescente , Productos del Gen gag/genética , Humanos , Inmunoprecipitación , Hibridación Fluorescente in Situ , Riñón/citología , Riñón/metabolismo , Riñón/virología , Leucemia Experimental/patología , Leucemia Experimental/virología , Ratones , Mitosis/fisiología , Mutación/genética , Células 3T3 NIH , Osteosarcoma/metabolismo , Osteosarcoma/patología , Osteosarcoma/virología , Filogenia , ARN Mensajero/genética , Infecciones por Retroviridae/patología , Infecciones por Retroviridae/virología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Infecciones Tumorales por Virus/patología , Infecciones Tumorales por Virus/virología , Replicación Viral
19.
J Immunol ; 184(6): 3072-8, 2010 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-20154210

RESUMEN

Chlamydophila pneumoniae infection of the vascular wall as well as activation of the transcription factor IFN regulatory factor (IRF)3 have been linked to development of chronic vascular lesions and atherosclerosis. The innate immune system detects invading pathogens by use of pattern recognition receptors, some of which are able to stimulate IRF3/7 activation and subsequent type I IFN production (e. g., IFN-beta). In this study, we show that infection of human endothelial cells with C. pneumoniae-induced production of IFN-beta, a cytokine that so far has been mainly associated with antiviral immunity. Moreover, C. pneumoniae infection led to IRF3 and IRF7 nuclear translocation in HUVECs and RNA interference experiments showed that IRF3 and IRF7 as well as the mitochondrial antiviral signaling (MAVS) were essential for IFN-beta induction. Finally, C. pneumoniae replication was enhanced in endothelial cells in which IRF3, IRF7, or MAVS expression was inhibited by small interfering RNA and attenuated by IFN-beta treatment. In conclusion, C. pneumoniae infection of endothelial cells activates an MAVS-, IRF3-, and IRF7-dependent signaling, which controls bacterial growth and might modulate development of vascular lesions.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Chlamydophila pneumoniae/crecimiento & desarrollo , Chlamydophila pneumoniae/inmunología , Endotelio Vascular/inmunología , Factor 3 Regulador del Interferón/fisiología , Factor 7 Regulador del Interferón/fisiología , Interferón beta/fisiología , Proteínas Mitocondriales/fisiología , Interferencia de ARN/fisiología , Células Cultivadas , Regulación hacia Abajo/inmunología , Endotelio Vascular/microbiología , Endotelio Vascular/virología , Humanos , Inmunidad Innata , Interferón beta/biosíntesis , Interferón beta/genética , Leucemia Experimental/inmunología , Leucemia Experimental/microbiología , Leucemia Experimental/virología , Virus de la Leucemia Murina de Moloney/inmunología , ARN Viral/antagonistas & inhibidores , Infecciones por Retroviridae/inmunología , Infecciones por Retroviridae/microbiología , Infecciones por Retroviridae/virología , Transducción de Señal/inmunología , Infecciones Tumorales por Virus/inmunología , Infecciones Tumorales por Virus/microbiología , Infecciones Tumorales por Virus/virología
20.
Cell Rep ; 38(3): 110279, 2022 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-35045303

RESUMEN

Murine leukemia virus (MLV)-presenting cells form stable intercellular contacts with target cells during infection of lymphoid tissue, indicating a role of cell-cell contacts in retrovirus dissemination. Whether host cell adhesion proteins are required for retrovirus spread in vivo remains unknown. Here, we demonstrate that the lymphocyte-function-associated-antigen-1 (LFA1) and its ligand intercellular-adhesion-molecule-1 (ICAM1) are important for cell-contact-dependent transmission of MLV between leukocytes. Infection experiments in LFA1- and ICAM1-deficient mice demonstrate a defect in MLV spread within lymph nodes. Co-culture of primary leukocytes reveals a specific requirement for ICAM1 on donor cells and LFA1 on target cells for cell-contact-dependent spread through trans- and cis-infection. Importantly, adoptive transfer experiments combined with a newly established MLV-fusion assay confirm that the directed LFA1-ICAM1 interaction is important for retrovirus fusion and transmission in vivo. Taken together, our data provide insights on how retroviruses exploit host proteins and the biology of cell-cell interactions for dissemination.


Asunto(s)
Molécula 1 de Adhesión Intercelular/metabolismo , Virus de la Leucemia Murina/patogenicidad , Leucemia Experimental/virología , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Infecciones por Retroviridae/virología , Animales , Interacciones Huésped-Patógeno/fisiología , Linfocitos/virología , Macrófagos/virología , Ratones , Ratones Endogámicos C57BL , Infecciones por Retroviridae/transmisión , Infecciones Tumorales por Virus/transmisión , Infecciones Tumorales por Virus/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA