Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 456
Filtrar
1.
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
2.
J Virol ; 95(18): e0063421, 2021 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-34190600

RESUMEN

The host transmembrane protein SERINC5 is incorporated into viral particles and restricts infection by certain retroviruses. However, what motif of SERINC5 mediates this process remains elusive. By conducting mutagenesis analyses, we found that the substitution of phenylalanine with alanine at position 412 (F412A) resulted in a >75-fold reduction in SERINC5's restriction function. The F412A substitution also resulted in the loss of SERINC5's function to sensitize HIV-1 neutralization by antibodies recognizing the envelope's membrane proximal region. A series of biochemical analyses revealed that F412A showed steady-state protein expression, localization at the cellular membrane, and incorporation into secreted virus particles to a greater extent than in the wild type. Furthermore, introduction of several amino acid mutations at this position revealed that the aromatic side chains, including phenylalanine, tyrosine, and tryptophan, were required to maintain SERINC5 functions to impair the virus-cell fusion process and virion infectivity. Moreover, the wild-type SERINC5 restricted infection of lentiviruses pseudotyped with envelopes of murine leukemia viruses, simian immunodeficiency virus, and HIV-2, and F412A abrogated this function. Taken together, our results highlight the importance of the aromatic side chain at SERINC5 position 412 to maintain its restriction function against diverse retrovirus envelopes. IMPORTANCE The host protein SERINC5 is incorporated into progeny virions of certain retroviruses and restricts the infectivity of these viruses or sensitizes the envelope glycoprotein to a class of neutralizing antibodies. However, how and which part of SERINC5 engages with the diverse array of retroviral envelopes and exerts its antiretroviral functions remain elusive. During mutagenesis analyses, we eventually found that the single substitution of phenylalanine with alanine, but not with tyrosine or tryptophan, at position 412 (F412A) resulted in the loss of SERINC5's functions toward diverse retroviruses, whereas F412A showed steady-state protein expression, localization at the cellular membrane, and incorporation into progeny virions to a greater extent than the wild type. Results suggest that the aromatic side chain at position 412 of SERINC5 plays a critical role in mediating antiviral functions toward various retroviruses, thus providing additional important information regarding host and retrovirus interaction.


Asunto(s)
Aminoácidos Aromáticos/genética , Membrana Celular/metabolismo , Infecciones por VIH/virología , VIH-1/patogenicidad , Virus de la Leucemia Murina/patogenicidad , Proteínas de la Membrana/metabolismo , Mutación , Células HEK293 , Infecciones por VIH/genética , VIH-1/genética , Interacciones Huésped-Patógeno , Humanos , Virus de la Leucemia Murina/genética , Proteínas de la Membrana/genética , Virulencia
3.
J Ethnopharmacol ; 267: 113519, 2021 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-33137432

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Astragalus glycyphyllos L. has been extensively used in Bulgarian folk medicine as an antihypertensive, diuretic, anti-inflammatory, anti-tumour, in cases of cardiac insufficiency, renal inflammation, calculosis, etc. AIM OF THE STUDY: To evaluate the possible in vitro/in vivo anti-proliferative/anti-tumour activity of a purified saponins' mixture (PSM) obtained from the plant. MATERIALS AND METHODS: Viability and proliferative activity of the Graffi myeloid tumour cells was assessed by MTT test. The morphological alterations were visualized and analysed by fluorescent microscopy after intravital double staining. An in vivo model of Graffi tumour bearing hamsters was used to examine the influence of PSM on transplantability, tumour growth, survival and mortality as well as to observe pathomorphological changes. RESULTS: Graffi tumour cells were sensitive to purified saponins' mixture after 24 and 48 h treatment. The treatment induced a statistically significant decrease of the viability/proliferation of the Graffi tumour cells. These effects were concentration- and time-dependent. Fluorescent microscopy studies showed that these antiproliferative effects were connected to the induction of apoptosis. The in vivo study showed the presence of a stromal component, single mononuclear cells in the stroma. Multiple incorrect mitotic figures were observed in the tumour tissue from the control group. Well-formed stroma with accumulation of mononuclear cells and mitotic cells were found in the group, treated with PSM. The tumour weight was decreased in the group, treated with PMS. CONCLUSION: The results indicate that PSM exhibited in vitro/in vivo antiproliferative/anti-tumour effects.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Planta del Astrágalo , Proliferación Celular/efectos de los fármacos , Leucemia Experimental/tratamiento farmacológico , Extractos Vegetales/farmacología , Saponinas/farmacología , Animales , Antineoplásicos Fitogénicos/aislamiento & purificación , Apoptosis/efectos de los fármacos , Planta del Astrágalo/química , Cricetinae , Femenino , Virus de la Leucemia Murina/patogenicidad , Leucemia Experimental/patología , Leucemia Experimental/virología , Masculino , Mesocricetus , Extractos Vegetales/aislamiento & purificación , Cultivo Primario de Células , Infecciones por Retroviridae/virología , Saponinas/aislamiento & purificación , Carga Tumoral/efectos de los fármacos , Células Tumorales Cultivadas , Infecciones Tumorales por Virus/virología
4.
Viruses ; 12(11)2020 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-33121095

RESUMEN

Apolipoprotein B mRNA editing enzyme, catalytic peptide 3 (APOBEC3) proteins are critical host proteins that counteract and prevent the replication of retroviruses. Unlike the genome of humans and other species, the mouse genome encodes a single Apobec3 gene, which has undergone positive selection, as reflected by the allelic variants found in different inbred mouse strains. This positive selection was likely due to infection by various mouse retroviruses, which have persisted in their hosts for millions of years. While mouse retroviruses are inhibited by APOBEC3, they nonetheless still remain infectious, likely due to the actions of different viral proteins that counteract this host factor. The study of viruses in their natural hosts provides important insight into their co-evolution.


Asunto(s)
Citidina Desaminasa/genética , Interacciones Huésped-Patógeno/genética , Infecciones por Retroviridae/virología , Retroviridae/patogenicidad , Animales , Virus de la Leucemia Murina/patogenicidad , Virus del Tumor Mamario del Ratón/patogenicidad , Ratones , Infecciones Tumorales por Virus/virología , Replicación Viral
5.
mBio ; 11(4)2020 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-32665269

RESUMEN

The serine incorporator (SERINC) proteins are multipass transmembrane proteins that affect sphingolipid and phosphatidylserine synthesis. Human SERINC5 and SERINC3 were recently shown to possess antiretroviral activity for a number of retroviruses, including human immunodeficiency virus (HIV), murine leukemia virus (MLV), and equine infectious anemia virus (EIAV). In the case of MLV, the glycosylated Gag (glyco-Gag) protein was shown to counteract SERINC5-mediated restriction in in vitro experiments and the viral envelope was found to determine virion sensitivity or resistance to SERINC5. However, nothing is known about the in vivo function of SERINC5. Antiretroviral function of a host factor in vitro is not always associated with antiretroviral function in vivo Using SERINC5-/- mice that we had generated, we showed that mouse SERINC5 (mSERINC5) restriction of MLV infection in vivo is influenced not only by glyco-Gag but also by the retroviral envelope. Finally, we also examined the in vivo function of the other SERINC gene with known antiretroviral functions, SERINC3. By using SERINC3-/- mice, we found that the murine homologue, mSERINC3, had no antiretroviral role either in vivo or in vitro To our knowledge, this report provides the first data showing that SERINC5 restricts retrovirus infection in vivo and that restriction of retrovirus infectivity in vivo is dependent on the presence of both glyco-Gag and the viral envelope.IMPORTANCE This study examined for the first time the in vivo function of the serine incorporator (SERINC) proteins during retrovirus infection. SERINC3 and SERINC5 (SERINC3/5) restrict a number of retroviruses, including human immunodeficiency virus 1 (HIV-1) and murine leukemia virus (MLV), by blocking their entry into cells. Nevertheless, HIV-1 and MLV encode factors, Nef and glycosylated Gag, respectively, that counteract SERINC3/5 in vitro We recently developed SERINC3 and SERINC5 knockout mice to examine the in vivo function of these genes. We found that SERINC5 restriction is dependent on the absence of glycosylated Gag and the expression of a specific viral envelope glycoprotein. On the other hand, SERINC3 had no antiviral function. Our findings have implications for the development of therapeutics that target SERINC5 during retrovirus infection.


Asunto(s)
Interacciones Huésped-Patógeno , Leucemia Experimental/virología , Proteínas de la Membrana/genética , Infecciones por Retroviridae/virología , Infecciones Tumorales por Virus/virología , Animales , Femenino , Glicosilación , Virus de la Leucemia Murina/patogenicidad , Masculino , Glicoproteínas de Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
6.
J Virol ; 94(13)2020 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-32321818

RESUMEN

RNA modifications play diverse roles in regulating RNA function, and viruses co-opt these pathways for their own benefit. While recent studies have highlighted the importance of N6-methyladenosine (m6A)-the most abundant mRNA modification-in regulating retrovirus replication, the identification and function of other RNA modifications in viral biology have been largely unexplored. Here, we characterized the RNA modifications present in a model retrovirus, murine leukemia virus (MLV), using mass spectrometry and sequencing. We found that 5-methylcytosine (m5C) is highly enriched in viral genomic RNA relative to uninfected cellular mRNAs, and we mapped at single-nucleotide resolution the m5C sites, which are located in multiple clusters throughout the MLV genome. Further, we showed that the m5C reader protein ALYREF plays an important role in regulating MLV replication. Together, our results provide a complete m5C profile in a virus and its function in a eukaryotic mRNA.IMPORTANCE Over 130 modifications have been identified in cellular RNAs, which play critical roles in many cellular processes, from modulating RNA stability to altering translation efficiency. One such modification, 5-methylcytosine, is relatively abundant in mammalian mRNAs, but its precise location and function are not well understood. In this study, we identified unexpectedly high levels of m5C in the murine leukemia virus RNA, precisely mapped its location, and showed that ALYREF, a reader protein that specifically recognizes m5C, regulates viral production. Together, our findings provide a high-resolution atlas of m5C in murine leukemia virus and reveal a functional role of m5C in viral replication.


Asunto(s)
5-Metilcitosina/metabolismo , Virus de la Leucemia Murina/genética , 5-Metilcitosina/fisiología , Animales , Metilación de ADN/genética , Genoma Viral/genética , Células HEK293 , Humanos , Virus de la Leucemia Murina/metabolismo , Virus de la Leucemia Murina/patogenicidad , Metiltransferasas/metabolismo , Ratones , Células 3T3 NIH , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Estabilidad del ARN , ARN Mensajero/genética , ARN Viral/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Retroviridae/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcriptoma/genética , Replicación Viral/genética
7.
Biotechnol Prog ; 36(3): e2953, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31846227

RESUMEN

Triton X-100 has long been used either alone or in combination with solvent to inactivate enveloped viruses in biopharmaceutical manufacturing. However, European Chemicals Agency (ECHA) officially placed Triton X-100 on the Annex XIV authorization list in 2017 because 4-(1,1,3,3-tetramethylbutyl) phenol, a degradation product of Triton X-100, is of harmful endocrine disrupting activities. As a result, any use of Triton X-100 in the European Economic Area would require an ECHA issued authorization after the sunset date of January 4, 2021. In search of possible replacements for Triton X-100, we discovered that polysorbate 80 (PS80) in absence of any solvents was able to effectively inactive enveloped viruses such as xenotropic murine leukemia virus and pseudorabies virus with comparable efficacy as measured by log reduction factors. Interestingly, PS80 did not show any virucidal activities in phosphate buffered saline (PBS) while achieving robust virus inactivation in cell-free Chinese hamster ovary (CHO) bioreactor harvests. This intriguing observation led us to speculate that virus inactivation by PS80 involved components in the cell-free CHO bioreactor harvests that were absent in PBS. Specifically, we hypothesized that esterase and/or lipases in the cell-free bioreactor harvests hydrolyzed PS80 to yield oleic acid, a known potent virucidal agent, which in turn inactivated viruses. This theory was confirmed using purified recombinant lysosomal phospholipase A2 isomer (rLPLA2) in PBS. Subsequent characterization work has indicated that virus inactivation by PS80 is effective and robust within temperature and concentration ranges comparable to those of Triton X-100. Similar to Triton X-100, virus inactivation by PS80 is dually dependent on treatment time and temperature. Unlike Triton X-100, PS80 inactivation does not correlate with concentrations in a simple manner. Additionally, we have demonstrated that PS20 exhibits similar virus inactivation activities as PS80. Based on the findings described in the current work, we believe that PS80 is potentially a viable replacement for Triton X-100 and can be used in manufacturing processes for wide spectrum of biopharmaceuticals to achieve desirable virus clearance. Finally, the advantages and disadvantages of using PS80 for virus inactivation are discussed in the contexts of GMP manufacturing.


Asunto(s)
Sistema Libre de Células , Virus de la Leucemia Murina/efectos de los fármacos , Polisorbatos/farmacología , Inactivación de Virus/efectos de los fármacos , Animales , Células CHO , Cricetinae , Cricetulus , Detergentes/química , Detergentes/farmacología , Hidrólisis/efectos de los fármacos , Cinética , Virus de la Leucemia Murina/patogenicidad , Ratones , Octoxinol , Solventes/química
8.
J Neuroimmune Pharmacol ; 14(3): 391-400, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31209775

RESUMEN

HIV-associated neurocognitive disorders (HAND) have been linked to dysregulation of glutamate metabolism in the central nervous system (CNS) culminating in elevated extracellular glutamate and disrupted glutamatergic neurotransmission. Increased glutamate synthesis via upregulation of glutaminase (GLS) activity in brain immune cells has been identified as one potential source of excess glutamate in HAND. However, direct evidence for this hypothesis in an animal model is lacking, and the viability of GLS as a drug target has not been explored. In this brief report, we demonstrate that GLS inhibition with the glutamine analogue 6-diazo-5-oxo-L-norleucine (DON) can reverse cognitive impairment in the EcoHIV-infected mouse model of HAND. However, due to peripheral toxicity DON is not amenable to clinical use in a chronic disease such as HAND. We thus tested JHU083, a novel, brain penetrant DON prodrug predicted to exhibit improved tolerability. Systemic administration of JHU083 reversed cognitive impairment in EcoHIV-infected mice similarly to DON, and simultaneously normalized EcoHIV-induced increases in cerebrospinal fluid (CSF) glutamate and GLS activity in microglia-enriched brain CD11b + cells without observed toxicity. These studies support the mechanistic involvement of elevated microglial GLS activity in HAND pathogenesis, and identify JHU083 as a potential treatment option. Graphical Abstract Please provide Graphical Abstract caption.Glutamine Antagonist JHU083 Normalizes Aberrant Glutamate Production and Cognitive Deficits in the EcoHIV Murine Model of HIV-Associated Neurocognitive Disorders .


Asunto(s)
Complejo SIDA Demencia , Compuestos Azo/uso terapéutico , Caproatos/uso terapéutico , Trastornos del Conocimiento/tratamiento farmacológico , Glutamatos/biosíntesis , Glutamina/antagonistas & inhibidores , Profármacos/uso terapéutico , Animales , Compuestos Azo/farmacocinética , Antígeno CD11b/análisis , Caproatos/farmacocinética , Trastornos del Conocimiento/líquido cefalorraquídeo , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/virología , Condicionamiento Clásico/efectos de los fármacos , Miedo , Glutamatos/líquido cefalorraquídeo , VIH-1/genética , VIH-1/patogenicidad , Virus de la Leucemia Murina/genética , Virus de la Leucemia Murina/patogenicidad , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Memoria a Corto Plazo/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microglía/metabolismo , Norleucina/análogos & derivados , Norleucina/uso terapéutico , Profármacos/farmacocinética , Virus Reordenados/genética , Virus Reordenados/patogenicidad , Aprendizaje Espacial/efectos de los fármacos
9.
Biotechnol J ; 14(8): e1800646, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30810288

RESUMEN

Continuous virus inactivation (VI) remains one of the missing pieces while the biopharma industry moves toward continuous manufacturing. The challenges of adapting VI to the continuous operation are two-fold: 1) achieving fluid homogeneity and 2) a narrow residence time distribution (RTD) for fluid incubation. To address these challenges, a dynamic active in-line mixer and a packed-bed continuous virus inactivation reactor (CVIR) are implemented, which act as a narrow RTD incubation chamber. The developed concept is applied using solvent/detergent (S/D) treatment for inactivation of two commonly used model viruses. The in-line mixer is characterized and enables mixing of the viscous S/D chemicals to ±1.0% of the target concentration in a small dead volume. The reactor's RTD is characterized and additional control experiments confirm that the VI is due to the S/D action and not induced by system components. The CVIR setup achieves steady state rapidly before two reactor volumes and the logarithmic reduction values of the continuous inactivation process are identical to those obtained by the traditional batch operation. The packed-bed reactor for continuous VI unites fully continuous processing with very low-pressure drop and scalability.


Asunto(s)
Biotecnología/instrumentación , Biotecnología/métodos , Solventes , Inactivación de Virus , Animales , Virus de la Diarrea Viral Bovina/patogenicidad , Diseño de Equipo , Cinética , Virus de la Leucemia Murina/patogenicidad
10.
Cancer Immunol Res ; 6(11): 1292-1300, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30143537

RESUMEN

Mouse models have been instrumental in establishing fundamental principles of cancer initiation and progression and continue to be invaluable in the discovery and further development of cancer therapies. Nevertheless, important aspects of human disease are imperfectly approximated in mouse models, notably the involvement of endogenous retroviruses (ERVs). Replication-defective ERVs, present in both humans and mice, may affect tumor development and antitumor immunity through mechanisms not involving infection. Here, we revealed an adverse effect of murine ERVs with restored infectivity on the behavior of mouse cancer models. In contrast to human cancer, where infectious ERVs have never been detected, we found that ERV infectivity was frequently restored in transplantable, as well as genetic, mouse cancer models. Such replication-competent, ERV-derived retroviruses were responsible for unusually high expression of retroviral nucleic acids and proteins in mouse cancers. Infectious ERV-derived retroviruses produced by mouse cancer cells could directly infect tumor-infiltrating host immune cells and fundamentally modified the host's immune defenses to cancer, as well as the outcome of immunotherapy. Therefore, infectious retroviruses, variably arising in mouse cancer models, but not in human cancer, have the potential to confound many immunologic studies and should be considered as a variable, if not altogether avoided. Cancer Immunol Res; 6(11); 1292-300. ©2018 AACR.


Asunto(s)
Retrovirus Endógenos/patogenicidad , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/virología , Animales , Línea Celular Tumoral , Femenino , Virus de la Leucemia Murina/genética , Virus de la Leucemia Murina/patogenicidad , Linfocitos Infiltrantes de Tumor/patología , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Transgénicos , Neoplasias Experimentales/patología , Factor 1 de Unión al Dominio 1 de Regulación Positiva/genética , Proteínas Proto-Oncogénicas B-raf/genética , Infecciones por Retroviridae/virología , Tropismo Viral/fisiología
11.
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
12.
J Mol Biol ; 429(8): 1171-1191, 2017 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-28315663

RESUMEN

The retroviral restriction factors of the APOBEC3 (A3) cytidine deaminase family catalyze the deamination of cytidines in single-stranded viral DNA. APOBEC3C (A3C) is a strong antiviral factor against viral infectivity factor (vif)-deficient simian immunodeficiency virus Δvif, which is, however, a weak inhibitor against human immunodeficiency virus (HIV)-1 for reasons unknown. The precise link between the antiretroviral effect of A3C and its catalytic activity is incompletely understood. Here, we show that the S61P mutation in human A3C (A3C.S61P) boosted hypermutation in the viral genomes of simian immunodeficiency virus Δvif and murine leukemia virus but not in human immunodeficiency virus HIV-1Δvif. The enhanced antiviral activity of A3C.S61P correlated with enhanced in vitro cytidine deamination. Furthermore, the S61P mutation did not change the substrate specificity of A3C, ribonucleoprotein complex formation, self-association, Zinc coordination, or viral incorporation features. We propose that local structural changes induced by the serine-to-proline substitution are responsible for the gain of catalytic activity of A3C.S61P. Our results are a first step toward an understanding of A3C's DNA binding capacity, deamination-dependent editing, and antiviral functions at the molecular level. We conclude that the enhanced enzymatic activity of A3C is insufficient to restrict HIV-1, indicating an unknown escape mechanism of HIV-1.


Asunto(s)
Citidina Desaminasa/química , Citidina Desaminasa/metabolismo , VIH-1/patogenicidad , Sustitución de Aminoácidos , Animales , Citidina Desaminasa/genética , Citosina/metabolismo , ADN de Cadena Simple/química , ADN de Cadena Simple/metabolismo , ADN Viral/metabolismo , Células HEK293/virología , VIH-1/genética , Interacciones Huésped-Patógeno , Humanos , Virus de la Leucemia Murina/metabolismo , Virus de la Leucemia Murina/patogenicidad , Pan troglodytes , Conformación Proteica , Virus de la Inmunodeficiencia de los Simios/metabolismo , Virus de la Inmunodeficiencia de los Simios/patogenicidad , Zinc/metabolismo , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/metabolismo
13.
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
14.
J Virol ; 90(7): 3385-99, 2016 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-26764005

RESUMEN

UNLABELLED: Certain murine leukemia viruses (MLVs) are capable of inducing fatal progressive spongiform motor neuron disease in mice that is largely mediated by viral Env glycoprotein expression within central nervous system (CNS) glia. While the etiologic mechanisms and the glial subtypes involved remain unresolved, infection of NG2 glia was recently observed to correlate spatially and temporally with altered neuronal physiology and spongiogenesis. Since one role of NG2 cells is to serve as oligodendrocyte (OL) progenitor cells (OPCs), we examined here whether their infection by neurovirulent (FrCasE) or nonneurovirulent (Fr57E) ecotropic MLVs influenced their viability and/or differentiation. Here, we demonstrate that OPCs, but not OLs, are major CNS targets of both FrCasE and Fr57E. We also show that MLV infection of neural progenitor cells (NPCs) in culture did not affect survival, proliferation, or OPC progenitor marker expression but suppressed certain glial differentiation markers. Assessment of glial differentiation in vivo using transplanted transgenic NPCs showed that, while MLVs did not affect cellular engraftment or survival, they did inhibit OL differentiation, irrespective of MLV neurovirulence. In addition, in chimeric brains, where FrCasE-infected NPC transplants caused neurodegeneration, the transplanted NPCs proliferated. These results suggest that MLV infection is not directly cytotoxic to OPCs but rather acts to interfere with OL differentiation. Since both FrCasE and Fr57E viruses restrict OL differentiation but only FrCasE induces overt neurodegeneration, restriction of OL maturation alone cannot account for neuropathogenesis. Instead neurodegeneration may involve a two-hit scenario where interference with OPC differentiation combined with glial Env-induced neuronal hyperexcitability precipitates disease. IMPORTANCE: A variety of human and animal retroviruses are capable of causing central nervous system (CNS) neurodegeneration manifested as motor and cognitive deficits. These retroviruses infect a variety of CNS cell types; however, the specific role each cell type plays in neuropathogenesis remains to be established. The NG2 glia, whose CNS functions are only now emerging, are a newly appreciated viral target in murine leukemia virus (MLV)-induced neurodegeneration. Since one role of NG2 glia is that of oligodendrocyte progenitor cells (OPCs), we investigated here whether their infection by the neurovirulent MLV FrCasE contributed to neurodegeneration by affecting OPC viability and/or development. Our results show that both neurovirulent and nonneurovirulent MLVs interfere with oligodendrocyte differentiation. Thus, NG2 glial infection could contribute to neurodegeneration by preventing myelin formation and/or repair and by suspending OPCs in a state of persistent susceptibility to excitotoxic insult mediated by neurovirulent virus effects on other glial subtypes.


Asunto(s)
Virus de la Leucemia Murina/patogenicidad , Enfermedad de la Neurona Motora/virología , Células-Madre Neurales/virología , Neurogénesis/fisiología , Neuroglía/virología , Infecciones por Retroviridae/complicaciones , Células 3T3 , Animales , Línea Celular , Proliferación Celular , Supervivencia Celular , Femenino , Productos del Gen env/biosíntesis , Masculino , Ratones , Ratones Transgénicos , Oligodendroglía/citología , Oligodendroglía/virología
15.
Biotechnol Prog ; 32(1): 89-97, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26488618

RESUMEN

To ensure the viral safety of protein therapeutics made in mammalian cells, purification processes include dedicated viral clearance steps to remove or inactivate adventitious and endogenous viruses. One such dedicated step is low pH treatment, a robust and effective method commonly used in monoclonal antibody production to inactivate enveloped viruses. To characterize the operating space for low pH viral inactivation, we performed a statistically designed experiment evaluating the effect of pH, temperature, hold duration, acid type, and buffer concentration on inactivation of the retrovirus model, XMuLV. An additional single factor experiment was performed to study the effect of protein concentration. These data were used to generate predictive models of inactivation at each time point studied, which can be used to identify conditions for robust and effective XMuLV inactivation. At pH 3.6, XMuLV inactivation was rapid, robust, and relatively unaffected by the other factors studied, providing support for this as a generic viral inactivation condition for products that can tolerate this low pH. At pH 3.7 and 3.8, other factors besides pH affected XMuLV inactivation. By understanding the impact of each factor on inactivation, the factors can be manipulated within the operating space to ensure effective inactivation while achieving desired product quality goals.


Asunto(s)
Virus de la Leucemia Murina/crecimiento & desarrollo , Proteínas del Envoltorio Viral/química , Inactivación de Virus , Animales , Anticuerpos Monoclonales/inmunología , Biotecnología , Concentración de Iones de Hidrógeno , Cinética , Virus de la Leucemia Murina/patogenicidad , Retroviridae/efectos de los fármacos , Temperatura , Proteínas del Envoltorio Viral/inmunología
16.
Virology ; 487: 273-84, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26580513

RESUMEN

Human SAMHD1 (hSAMHD1) restricts HIV-1 infection in non-dividing cells by depleting intracellular dNTPs to limit viral reverse transcription. Phosphorylation of hSAMHD1 at threonine (T) 592 by cyclin-dependent kinase (CDK) 1 and CDK2 negatively regulates HIV-1 restriction. Mouse SAMHD1 (mSAMHD1) restricts HIV-1 infection in non-dividing cells, but whether its phosphorylation regulates retroviral restriction is unknown. Here we identified six phospho-sites of mSAMHD1, including T634 that is homologous to T592 of hSAMHD1 and phosphorylated by CDK1 and CDK2. We found that wild-type (WT) mSAMHD1 and a phospho-ablative mutant, but not a phospho-mimetic mutant, restricted HIV-1 infection in differentiated U937 cells. Murine leukemia virus (MLV) infection of dividing NIH3T3 cells was modestly restricted by mSAMHD1 WT and phospho-mutants, but not by a dNTPase-defective mutant. Our results suggest that phosphorylation of mSAMHD1 at T634 by CDK1/2 negatively regulates its HIV-1 restriction in differentiated cells, but does not affect its MLV restriction in dividing cells.


Asunto(s)
Quinasa 2 Dependiente de la Ciclina/genética , Quinasas Ciclina-Dependientes/genética , VIH-1/patogenicidad , Virus de la Leucemia Murina/patogenicidad , Proteínas de Unión al GTP Monoméricas/metabolismo , Células 3T3 , Animales , Proteína Quinasa CDC2 , Línea Celular , Quinasa 2 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Células HEK293 , Infecciones por VIH/patología , VIH-1/genética , Humanos , Virus de la Leucemia Murina/genética , Leucemia Experimental/patología , Ratones , Fosforilación , Interferencia de ARN , ARN Interferente Pequeño , Infecciones por Retroviridae/patología , Proteína 1 que Contiene Dominios SAM y HD , Transfección , Infecciones Tumorales por Virus/patología , Células U937 , Replicación Viral/genética
17.
J Med Food ; 18(8): 882-9, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26076116

RESUMEN

The immunomodulatory effects of a dietary supplement of purple sweet potato extract (PSPE) in LP-BM5 murine leukemia virus (MuLV)-induced immune-deficient mice were investigated. Mice were divided into six groups: normal control, infected control (LP-BM5 MuLV infection), positive control (LP-BM5 MuLV infection+dietary supplement of red ginseng 300 mg/kg), purple sweet potato water extract (PSPWE) (LP-BM5 MuLV infection+dietary supplement of PSPE 300 mg/kg), PSP10EE (LP-BM5 MuLV infection+dietary supplement of 10% ethanol PSPE 300 mg/kg), and PSP80EE (LP-BM5 MuLV infection+dietary supplement of 80% ethanol PSPE 300 mg/kg). Dietary supplementation began on the day of LP-BM5 MuLV infection and continued for 12 weeks. Dietary supplementation of PSPE inhibited LP-BM5 MuLV-induced splenomegaly and lymphadenopathy and attenuated the suppression of T- and B-cell proliferation and T helper 1/T helper 2 cytokine imbalance in LP-BM5 MuLV-infected mice. Dietary supplement of PSPE increased the activity of the antioxidant enzymes, superoxide dismutase and glutathione peroxidase. The data suggest that PSPE may ameliorate immune dysfunction due to LP-BM5 MuLV infection by modulating antioxidant defense systems.


Asunto(s)
Antioxidantes/farmacología , Inmunomodulación/efectos de los fármacos , Ipomoea batatas/química , Ipomoea batatas/inmunología , Virus de la Leucemia Murina , Síndrome de Inmunodeficiencia Adquirida del Murino/dietoterapia , Animales , Antioxidantes/química , Linfocitos B/citología , Linfocitos B/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Citocinas/análisis , Modelos Animales de Enfermedad , Glutatión Peroxidasa/metabolismo , Virus de la Leucemia Murina/patogenicidad , Enfermedades Linfáticas/dietoterapia , Ratones , Ratones Endogámicos C57BL , Síndrome de Inmunodeficiencia Adquirida del Murino/inmunología , Síndrome de Inmunodeficiencia Adquirida del Murino/patología , Síndrome de Inmunodeficiencia Adquirida del Murino/virología , Extractos Vegetales/farmacología , Preparaciones de Plantas/química , Esplenomegalia/dietoterapia , Superóxido Dismutasa/metabolismo , Linfocitos T/citología , Linfocitos T/efectos de los fármacos
18.
Biologicals ; 43(4): 256-65, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25997567

RESUMEN

Infectivity and reverse transcriptase quantitative real-time polymerase chain reaction (qRT-PCR) assays have been optimized and validated for xenotropic murine leukemia virus (X-MuLV) detection. We have evaluated the assays systematically with regard to specificity, linearity, lower limit of detection (LLOD), lower limit of quantification (LLOQ), and precision. Both assays are specific for X-MuLV detection, with a linear detection range of 0.6-5.6 log(10) TCID(50)/mL for the infectivity assay, and 1.4-6.5 log(10) particles/mL for the qRT-PCR assay. The LLOD and LLOQ of the infectivity and the qRT-PCR assays are determined as 0.5 and 1.0 log(10)/mL, and 1.4 and 2.2 log(10)/mL. The inter-assay repeatability of qRT-PCR assay (4.2% coefficient of variation [CV]) is higher than the infectivity assay (7.9% CV). We have shown that both assays are closely correlated (r = 0.85, P < 0.05, n = 22). The particle/infectivity ratio is determined as 66. Both assays were applied to evaluate virus removal using virus clearance samples of chromatographic and filtration processes. Here, we have demonstrated that the qRT-PCR assay is much faster in testing and is approximately 8-fold more sensitive than the infectivity assay. Therefore, the qRT-PCR assay can replace the infectivity assay in many cases, but both assays are complementary in elucidating the mechanism of virus inactivation and removal in virus clearance validation.


Asunto(s)
Virus de la Leucemia Murina/patogenicidad , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Virulencia , Animales , Gatos , Línea Celular , Virus de la Leucemia Murina/aislamiento & purificación , Límite de Detección , Reproducibilidad de los Resultados
19.
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
20.
Transgenic Res ; 23(3): 539-56, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24474164

RESUMEN

Draft genome sequences for the human schistosomes, Schistosoma japonicum, S. mansoni and S. haematobium are now available. The schistosome genome contains ~11,000 protein encoding genes for which the functions of few are well understood. Nonetheless, the newly described gene products and novel non-coding RNAs represent potential intervention targets, and molecular tools are being developed to determine their importance. Over the past decade, noteworthy advances has been reported towards development of tools for gene manipulation of schistosomes, including gene expression perturbation by RNAi, and transient and stable transfection including transgenesis mediated by genome integration competent vectors. Retrovirus-mediated transgenesis is an established functional genomic approach for model species. It offers the means to establish gain- or loss-of-function phenotypes, supports vector-based RNA interference, and represents a powerful forward genetics tool for insertional mutagenesis. Murine leukemia virus (MLV) pseudotyped with vesicular stomatitis virus glycoprotein mediates somatic transgenesis in S. mansoni, and vertical transmission of integrated transgenes in S. mansoni has been demonstrated, leading the establishment of transgenic lines. In addition, MLV transgenes encoding antibiotic resistance allow the selection of MLV-transduced parasites with the appropriate antibiotics. Here we describe detailed methods to produce and quantify pseudotyped MLV particles for use in transducing developmental stages of schistosomes. Approaches to analyze MLV-transduced schistosomes, including qPCR and high throughput approaches to verify and map genome integration of transgenes are also presented. We anticipate these tools should find utility in genetic investigations in other laboratories and for other helminth pathogens of important neglected tropical diseases.


Asunto(s)
Técnicas de Transferencia de Gen , Genoma , Virus de la Leucemia Murina/genética , Schistosoma mansoni/genética , Animales , Células Germinativas/virología , Humanos , Virus de la Leucemia Murina/patogenicidad , Ratones , Ratones Transgénicos/genética , Mutagénesis Insercional , Transducción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA