Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cells ; 11(14)2022 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-35883685

RESUMEN

Gamma-interferon (γ-IFN) significantly inhibits infection by replication-defective viral vectors derived from the human immunodeficiency virus type 1 (HIV-1) or murine leukemia virus (MLV) but the underlying mechanism remains unclear. Previously we reported that knockdown of γ-IFN-inducible lysosomal thiolreductase (GILT) abrogates the antiviral activity of γ-IFN in TE671 cells but not in HeLa cells, suggesting that other γ-IFN-inducible host factors are involved in its antiviral activity in HeLa cells. We identified cellular factors, the expression of which are induced by γ-IFN in HeLa cells, using a microarray, and analyzed the effects of 11 γ-IFN-induced factors on retroviral vector infection. Our results showed that the exogenous expression of FAT10, IFI6, or IDO1 significantly inhibits both HIV-1- and MLV-based vector infections. The antiviral activity of γ-IFN was decreased in HeLa cells, in which the function of IDO1, IFI6, FAT10, and GILT were simultaneously inhibited. IDO1 is an enzyme that metabolizes an essential amino acid, tryptophan. However, IDO1 did not restrict retroviral vector infection in Atg3-silencing HeLa cells, in which autophagy did not occur. This study found that IDO1, IFI6, FAT10, and GILT are involved in the antiviral activity of γ-IFN, and IDO1 inhibits retroviral infection by inducing autophagy.


Asunto(s)
Infecciones por VIH , VIH-1 , Infecciones por Retroviridae , Antirretrovirales/farmacología , Antivirales/farmacología , Autofagia , Infecciones por VIH/tratamiento farmacológico , VIH-1/metabolismo , Células HeLa , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Interferón gamma/metabolismo , Interferón gamma/farmacología , Virus de la Leucemia Murina , Proteínas Mitocondriales , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro , Ubiquitinas/farmacología
2.
Small GTPases ; 13(1): 162-182, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34180342

RESUMEN

We recently identified a CD63-interacting protein to understand the role of CD63 in virion production of the human immunodeficiency virus type 1, and we have found that Rab3a forms a complex with CD63. In this study, we analysed the effect of Rab3a on virion production of the murine leukaemia virus (MLV), which is another member of the retrovirus family. We found that Rab3a silencing induced lysosomal degradation of the MLV Gag protein, and recovery of the Rab3a expression restored the level of the Gag protein through a complex formation of MLV Gag and Rab3a, indicating that Rab3a is required for MLV Gag protein expression. In contrast, CD63 silencing decreased the infectivity of released virions but had no effect on virion production, indicating that CD63 facilitates the infectivity of released MLV particles. Although Rab3a induced CD63 degradation in uninfected cells, the complex of MLV Gag and Rab3a suppressed the Rab3a-mediated CD63 degradation in MLV-infected cells. Finally, we found that the MLV Gag protein interacts with Rab3a to stabilize its own protein and CD63 that facilitates the infectivity of released MLV particles. Considering the involvement of Rab3a in lysosome trafficking to the plasma membrane, it may also induce cell surface transport of the MLV Gag protein.


Asunto(s)
Productos del Gen gag , Virus de la Leucemia Murina , Ratones , Animales , Humanos , Productos del Gen gag/metabolismo , Virus de la Leucemia Murina/metabolismo , Virión/metabolismo , Membrana Celular/metabolismo , Proteínas de Unión al GTP/metabolismo
3.
Cell Death Differ ; 28(12): 3199-3213, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34663907

RESUMEN

SARS-CoV-2 vaccinations have greatly reduced COVID-19 cases, but we must continue to develop our understanding of the nature of the disease and its effects on human immunity. Previously, we suggested that a dysregulated STAT3 pathway following SARS-Co-2 infection ultimately leads to PAI-1 activation and cascades of pathologies. The major COVID-19-associated metabolic risks (old age, hypertension, cardiovascular diseases, diabetes, and obesity) share high PAI-1 levels and could predispose certain groups to severe COVID-19 complications. In this review article, we describe the common metabolic profile that is shared between all of these high-risk groups and COVID-19. This profile not only involves high levels of PAI-1 and STAT3 as previously described, but also includes low levels of glutamine and NAD+, coupled with overproduction of hyaluronan (HA). SARS-CoV-2 infection exacerbates this metabolic imbalance and predisposes these patients to the severe pathophysiologies of COVID-19, including the involvement of NETs (neutrophil extracellular traps) and HA overproduction in the lung. While hyperinflammation due to proinflammatory cytokine overproduction has been frequently documented, it is recently recognized that the immune response is markedly suppressed in some cases by the expansion and activity of MDSCs (myeloid-derived suppressor cells) and FoxP3+ Tregs (regulatory T cells). The metabolomics profiles of severe COVID-19 patients and patients with advanced cancer are similar, and in high-risk patients, SARS-CoV-2 infection leads to aberrant STAT3 activation, which promotes a cancer-like metabolism. We propose that glutamine deficiency and overproduced HA is the central metabolic characteristic of COVID-19 and its high-risk groups. We suggest the usage of glutamine supplementation and the repurposing of cancer drugs to prevent the development of severe COVID-19 pneumonia.


Asunto(s)
COVID-19/fisiopatología , Glutamina/deficiencia , Animales , COVID-19/sangre , COVID-19/epidemiología , Comorbilidad , Glutamina/sangre , Humanos , Ácido Hialurónico/sangre , Metaboloma , Inhibidor 1 de Activador Plasminogénico/sangre , Factores de Riesgo , Índice de Severidad de la Enfermedad
4.
Sci Rep ; 10(1): 21474, 2020 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-33293588

RESUMEN

Gene editing using CRISPR/Cas9 is a promising method to cure many human genetic diseases. We have developed an efficient system to deliver Cas9 into the adeno-associated virus integration site 1 (AAVS1) locus, known as a safe harbor, using lentivirus and AAV viral vectors, as a step toward future in vivo transduction. First, we introduced Cas9v1 (derived from Streptococcus pyogenes) at random into the genome using a lentiviral vector. Cas9v1 activity was used when the N-terminal 1.9 kb, and C-terminal 2.3 kb fragments of another Cas9v2 (human codon-optimized) were employed sequentially with specific single-guide RNAs (sgRNAs) and homology donors carried by AAV vectors into the AAVS1 locus. Then, Cas9v1 was removed from the genome by another AAV vector containing sgRNA targeting the long terminal repeat of the lentivirus vector. The reconstituted Cas9v2 in the AAVS1 locus was functional and gene editing was efficient.


Asunto(s)
Proteína 9 Asociada a CRISPR/genética , Sistemas CRISPR-Cas , Dependovirus/genética , Lentivirus/genética , Streptococcus pyogenes/genética , Transducción Genética , Edición Génica , Técnicas de Transferencia de Gen , Sitios Genéticos , Vectores Genéticos/genética , Células HEK293 , Humanos , ARN Guía de Kinetoplastida/genética , Transducción Genética/métodos
5.
Cell Death Differ ; 27(12): 3209-3225, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33037393

RESUMEN

COVID-19 is caused by SARS-CoV-2 infection and characterized by diverse clinical symptoms. Type I interferon (IFN-I) production is impaired and severe cases lead to ARDS and widespread coagulopathy. We propose that COVID-19 pathophysiology is initiated by SARS-CoV-2 gene products, the NSP1 and ORF6 proteins, leading to a catastrophic cascade of failures. These viral components induce signal transducer and activator of transcription 1 (STAT1) dysfunction and compensatory hyperactivation of STAT3. In SARS-CoV-2-infected cells, a positive feedback loop established between STAT3 and plasminogen activator inhibitor-1 (PAI-1) may lead to an escalating cycle of activation in common with the interdependent signaling networks affected in COVID-19. Specifically, PAI-1 upregulation leads to coagulopathy characterized by intravascular thrombi. Overproduced PAI-1 binds to TLR4 on macrophages, inducing the secretion of proinflammatory cytokines and chemokines. The recruitment and subsequent activation of innate immune cells within an infected lung drives the destruction of lung architecture, which leads to the infection of regional endothelial cells and produces a hypoxic environment that further stimulates PAI-1 production. Acute lung injury also activates EGFR and leads to the phosphorylation of STAT3. COVID-19 patients' autopsies frequently exhibit diffuse alveolar damage (DAD) and increased hyaluronan (HA) production which also leads to higher levels of PAI-1. COVID-19 risk factors are consistent with this scenario, as PAI-1 levels are increased in hypertension, obesity, diabetes, cardiovascular diseases, and old age. We discuss the possibility of using various approved drugs, or drugs currently in clinical development, to treat COVID-19. This perspective suggests to enhance STAT1 activity and/or inhibit STAT3 functions for COVID-19 treatment. This might derail the escalating STAT3/PAI-1 cycle central to COVID-19.


Asunto(s)
COVID-19/patología , Factores de Transcripción STAT/metabolismo , Transducción de Señal/fisiología , COVID-19/metabolismo , COVID-19/virología , Quimiocinas/metabolismo , Citocinas/metabolismo , Receptores ErbB/metabolismo , Humanos , Interferón Tipo I/metabolismo , SARS-CoV-2/aislamiento & purificación , SARS-CoV-2/metabolismo , Factores de Transcripción STAT/química , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/metabolismo
6.
Artículo en Inglés | MEDLINE | ID: mdl-32411688

RESUMEN

Human immunodeficiency virus type 1 (HIV-1)-based viral vector is widely used as a biomaterial to transfer a gene of interest into target cells in many biological study fields including gene therapy. Vesicular stomatitis virus glycoprotein (VSV-G)-containing HIV-1 vector much more efficiently transduces various mammalian cells than other viral envelope proteins-containing vectors. Understanding the mechanism would contribute to development of a novel method of efficient HIV-1 vector production. HIV-1 vector is generally constructed by transient transfection of human 293T or African green monkey COS7 cells. It was found in this study that HIV-1 Gag protein is constitutively digested in lysosomes of African green monkey cells. Surprisingly, VSV-G elevated HIV-1 Gag protein levels, suggesting that VSV-G protects Gag protein from the lysosomal degradation. Unphosphorylated ezrin, but not phosphorylated ezrin, was detected in COS7 cells, and ezrin silencing elevated Gag protein levels in the presence of VSV-G. Expression of unphosphorylated ezrin reduced Gag protein amounts. These results indicate that unphosphorylated ezrin proteins inhibit the VSV-G-mediated stabilization of HIV-1 Gag protein. Trafficking of HIV-1 Gag-associated intracellular vesicles may be controlled by ezrin. Finally, this study found that ezrin silencing yields higher amount of VSV-G-pseudotyped HIV-1 vector.

7.
Front Microbiol ; 9: 1912, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30210460

RESUMEN

Host-cell expression of the ezrin protein is required for CXCR4 (X4)-tropic HIV-1 infection. Ezrin function is regulated by phosphorylation at threonine-567. This study investigates the role of ezrin phosphorylation in HIV-1 infection and virion release. We analyzed the effects of ezrin mutations involving substitution of threonine-567 by alanine (EZ-TA), a constitutively inactive mutant, or by aspartic acid (EZ-TD), which mimics phosphorylated threonine. We also investigated the effects of ezrin silencing on HIV-1 virion release using a specific siRNA. We observed that X4-tropic HIV-1 vector infection was inhibited by expression of the EZ-TA mutant but increased by expression of the EZ-TD mutant, suggesting that ezrin phosphorylation in target cells is required for efficient HIV-1 entry. Expression of a dominant-negative mutant of ezrin (EZ-N) and ezrin silencing in HIV-1 vector-producing cells significantly reduced the infectivity of released virions without affecting virion production. This result indicates that endogenous ezrin expression is required for virion infectivity. The EZ-TD but not the EZ-TA inhibited virion release from HIV-1 vector-producing cells. Taken together, these findings suggest that ezrin phosphorylation in target cells is required for efficient HIV-1 entry but inhibits virion release from HIV-1 vector-producing cells.

8.
Eur J Immunol ; 48(8): 1319-1328, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29745988

RESUMEN

Interferon regulatory factor 4 (IRF4) has critical roles in immune cell differentiation and function and is indispensable for clonal expansion and effector function in T cells. Here, we demonstrate that the AKT pathway is impaired in murine CD8+ T cells lacking IRF4. The expression of phosphatase and tensin homolog (PTEN), a negative regulator of the AKT pathway, was elevated in Irf4-/- CD8+ T cells. Inhibition of PTEN partially rescued downstream events, suggesting that PTEN constitutes a checkpoint in the IRF4-mediated regulation of cell signaling. Despite the clonal expansion defect, in the absence of IRF4, memory-like CD8+ T cells could be generated and maintained, although unable to expand in recall responses. The homeostatic proliferation of naïve Irf4-/- CD8+ T cells was impaired, whereas their number eventually reached a level similar to that of wild-type CD8+ T cells. Conversely, memory-like Irf4-/- CD8+ T cells underwent homeostatic proliferation in a manner similar to that of wild-type memory CD8+ T cells. These results suggest that IRF4 regulates the clonal expansion of CD8+ T cells at least in part via the AKT signaling pathway. Moreover, IRF4 regulates the homeostatic proliferation of naïve CD8+ T cells, whereas the maintenance of memory CD8+ T cells is IRF4-independent.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica/inmunología , Factores Reguladores del Interferón/inmunología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Antígenos de Linfocitos T/inmunología , Animales , Diferenciación Celular/inmunología , Proliferación Celular , Células Cultivadas , Factores Reguladores del Interferón/genética , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfohidrolasa PTEN/antagonistas & inhibidores , Transducción de Señal/inmunología
9.
Biochem Biophys Res Commun ; 501(4): 833-837, 2018 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-29684346

RESUMEN

Interferon regulatory factor (IRF) 4 and the proto-oncogene c-Rel cooperate in growth and antiviral drug resistance of adult T-cell leukemia/lymphoma (ATLL). To elucidate the target of IRF4 and c-Rel in ATLL, we determined the simultaneous binding sites of IRF4 and c-Rel using ChIP-seq technology. Nine genes were identified within 2 kb of binding sites, including MIR3662. Expression of miR-3662 was regulated by IRF4, and to a lesser extent by c-Rel. Cell proliferation was inhibited by knockdown of miR-3662 and expression of miR-3662 was correlated with antiviral drug resistance in ATLL cell lines. Thus, miR-3662 represents a target for therapies against ATLL.


Asunto(s)
Farmacorresistencia Viral/genética , Regulación Leucémica de la Expresión Génica , Leucemia-Linfoma de Células T del Adulto/genética , Leucemia-Linfoma de Células T del Adulto/virología , MicroARNs/genética , Adulto , Secuencia de Bases , Sitios de Unión/genética , Línea Celular Tumoral , Proliferación Celular/genética , Humanos , Factores Reguladores del Interferón/metabolismo , Leucemia-Linfoma de Células T del Adulto/patología , MicroARNs/metabolismo , Unión Proteica/genética , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-rel/metabolismo
10.
Artículo en Inglés | MEDLINE | ID: mdl-29629340

RESUMEN

Cleavage and activation of hemagglutinin (HA) by trypsin-like proteases in influenza A virus (IAV) are essential prerequisites for its successful infection and spread. In host cells, some transmembrane serine proteases such as TMPRSS2, TMPRSS4 and HAT, along with plasmin in the bloodstream, have been reported to cleave the HA precursor (HA0) molecule into its active forms, HA1 and HA2. Some trypsinogens can also enhance IAV proliferation in some cell types (e.g., rat cardiomyoblasts). However, the precise activation mechanism for this process is unclear, because the expression level of the physiological activator of the trypsinogens, the TMPRSS15 enterokinase, is expected to be very low in such cells, with the exception of duodenal cells. Here, we show that at least two variant enterokinases are expressed in various human cell lines, including A549 lung-derived cells. The exogenous expression of these enterokinases was able to enhance the proliferation of IAV in 293T human kidney cells, but the proliferation was reduced by knocking down the endogenous enterokinase in A549 cells. The enterokinase was able to enhance HA processing in the cells, which activated trypsinogen in vitro and in the IAV-infected cells also. Therefore, we conclude that enterokinase plays a role in IAV infection and proliferation by activating trypsinogen to process viral HA in human cell lines.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/fisiología , Gripe Humana/enzimología , Tripsina/metabolismo , Línea Celular , Activación Enzimática , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Interacciones Huésped-Patógeno , Humanos , Subtipo H1N1 del Virus de la Influenza A/genética , Gripe Humana/genética , Gripe Humana/virología , Procesamiento Proteico-Postraduccional , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Tripsina/genética
12.
Front Microbiol ; 8: 1653, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28900422

RESUMEN

CD63, a member of the tetraspanin family, is involved in virion production by human immunodeficiency virus type 1 (HIV-1), but its mechanism is unknown. In this study, we showed that a small GTP-binding protein, Rab3a, interacts with CD63. When Rab3a was exogenously expressed, the amounts of CD63 decreased in cells. The Rab3a-mediated reduction of CD63 was suppressed by lysosomal and proteasomal inhibitors. The amount of CD63 was increased by reducing the endogenous Rab3a level using a specific shRNA. These results indicate that Rab3a binds to CD63 to induce the degradation of CD63. Rab3a is thought to be involved in exocytosis, but we found that another function of Rab3a affects the fate of CD63 in lysosomes. CD63 interacted with Rab3a and was incorporated into HIV-1 particles. However, Rab3a was not detected in HIV-1 virions, thereby indicating that Rab3a-free CD63, but not Rab3a-bound CD63, is incorporated into HIV-1 particles. Overexpression or silencing of Rab3a moderately reduced HIV-1 virion formation. Overexpression of Rab3a decreased CD63 levels, but did not affect the incorporation of CD63 into HIV-1 particles. This study showed that Rab3a binds to CD63 to induce the degradation of CD63, and only Rab3a-free CD63 is incorporated into HIV-1 particles.

13.
Oncotarget ; 7(44): 71255-71273, 2016 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-27655726

RESUMEN

The mechanism by which type II interferon (IFN) inhibits virus replications remains to be identified. Murine leukemia virus (MLV) replication was significantly restricted by γ-IFN, but not human immunodeficiency virus type 1 (HIV-1) replication. Because MLV enters host cells via endosomes, we speculated that certain cellular factors among γ-IFN-induced, endosome-localized proteins inhibit MLV replication. We found that γ-IFN-inducible lysosomal thiolreductase (GILT) significantly restricts HIV-1 replication as well as MLV replication by its thiolreductase activity. GILT silencing enhanced replication-defective HIV-1 vector infection and virion production in γ-IFN-treated cells, although γ-IFN did not inhibit HIV-1 replication. This result showed that GILT is required for the anti-viral activity of γ-IFN. Interestingly, GILT protein level was increased by γ-IFN in uninfected cells and env-deleted HIV-1-infected cells, but not in full-length HIV-1-infected cells. γ-IFN-induced transcription from the γ-IFN-activation sequence was attenuated by the HIV-1 Env protein. These results suggested that the γ-IFN cannot restrict HIV-1 replication due to the inhibition of γ-IFN signaling by HIV-1 Env. Finally, we found that 4,4'-dithiodipyridine (4-PDS), which inhibits S-S bond formation at acidic pH, significantly suppresses HIV-1 vector infection and virion production, like GILT. In conclusion, this study showed that GILT functions as a host restriction factor against the retroviruses, and a GILT mimic, 4-PDS, is the leading compound for the development of novel concept of anti-viral agents.


Asunto(s)
Antirretrovirales/farmacología , VIH-1/fisiología , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/fisiología , Animales , Células COS , Chlorocebus aethiops , Ácido Ditionitrobenzoico/farmacología , Productos del Gen env/fisiología , VIH-1/efectos de los fármacos , Humanos , Interferón gamma/farmacología , Virus de la Leucemia Murina/efectos de los fármacos , Virus de la Leucemia Murina/fisiología , Ratones , Tetraspanina 30/fisiología , Virión/fisiología , Replicación Viral/efectos de los fármacos
14.
PLoS Pathog ; 12(1): e1005357, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26735137

RESUMEN

Dengue virus (DENV) is one of the most important arthropod-borne pathogens that cause life-threatening diseases in humans. However, no vaccine or specific antiviral is available for dengue. As seen in other RNA viruses, the innate immune system plays a key role in controlling DENV infection and disease outcome. Although the interferon (IFN) response, which is central to host protective immunity, has been reported to limit DENV replication, the molecular details of how DENV infection is modulated by IFN treatment are elusive. In this study, by employing a gain-of-function screen using a type I IFN-treated cell-derived cDNA library, we identified a previously uncharacterized gene, C19orf66, as an IFN-stimulated gene (ISG) that inhibits DENV replication, which we named Repressor of yield of DENV (RyDEN). Overexpression and gene knockdown experiments revealed that expression of RyDEN confers resistance to all serotypes of DENV in human cells. RyDEN expression also limited the replication of hepatitis C virus, Kunjin virus, Chikungunya virus, herpes simplex virus type 1, and human adenovirus. Importantly, RyDEN was considered to be a crucial effector molecule in the IFN-mediated anti-DENV response. When affinity purification-mass spectrometry analysis was performed, RyDEN was revealed to form a complex with cellular mRNA-binding proteins, poly(A)-binding protein cytoplasmic 1 (PABPC1), and La motif-related protein 1 (LARP1). Interestingly, PABPC1 and LARP1 were found to be positive modulators of DENV replication. Since RyDEN influenced intracellular events on DENV replication and, suppression of protein synthesis from DENV-based reporter construct RNA was also observed in RyDEN-expressing cells, our data suggest that RyDEN is likely to interfere with the translation of DENV via interaction with viral RNA and cellular mRNA-binding proteins, resulting in the inhibition of virus replication in infected cells.


Asunto(s)
Virus del Dengue/fisiología , Dengue/inmunología , Interferones/inmunología , Proteínas Virales/genética , Replicación Viral/inmunología , Línea Celular , Virus del Dengue/crecimiento & desarrollo , Técnicas de Silenciamiento del Gen , Humanos , Immunoblotting , Inmunoprecipitación , Espectrometría de Masas , Reacción en Cadena de la Polimerasa , Transfección
15.
Diabetologia ; 58(11): 2606-14, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26271342

RESUMEN

AIMS/HYPOTHESIS: Interferon regulatory factor (IRF)4 plays a critical role in lymphoid development and the regulation of immune responses. Genetic deletion of IRF4 has been shown to suppress autoimmune disease in several mouse models, but its role in autoimmune diabetes in NOD mice remains unknown. METHODS: To address the role of IRF4 in the pathogenesis of autoimmune diabetes in NOD mice, we generated IRF4-knockout NOD mice and investigated the impact of the genetic deletion of IRF4 on diabetes, insulitis and insulin autoantibody; the effector function of T cells in vivo and in vitro; and the proportion of dendritic cell subsets. RESULTS: Heterozygous IRF4-deficient NOD mice maintained the number and phenotype of T cells at levels similar to NOD mice. However, diabetes and autoantibody production were completely suppressed in both heterozygous and homozygous IRF4-deficient NOD mice. The level of insulitis was strongly suppressed in both heterozygous and homozygous IRF4-deficient mice, with minimal insulitis observed in heterozygous mice. An adoptive transfer study revealed that IRF4 deficiency conferred disease resistance in a gene-dose-dependent manner in recipient NOD/severe combined immunodeficiency mice. Furthermore, the proportion of migratory dendritic cells in lymph nodes was reduced in heterozygous and homozygous IRF4-deficient NOD mice in an IRF4 dose-dependent manner. These results suggest that the levels of IRF4 in T cells and dendritic cells are important for the pathogenesis of diabetes in NOD mice. CONCLUSIONS/INTERPRETATION: Haploinsufficiency of IRF4 halted disease development in NOD mice. Our findings suggest that an IRF4-targeted strategy might be useful for modulating autoimmunity in type 1 diabetes.


Asunto(s)
Autoanticuerpos/sangre , Diabetes Mellitus Tipo 1/genética , Haploinsuficiencia , Insulina/inmunología , Factores Reguladores del Interferón/genética , Animales , Células Dendríticas/inmunología , Diabetes Mellitus Tipo 1/sangre , Diabetes Mellitus Tipo 1/inmunología , Inmunoglobulina G/sangre , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Factores Protectores , Linfocitos T/inmunología
16.
Hepatogastroenterology ; 62(139): 624-8, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-26897942

RESUMEN

BACKGROUND/AIMS: Though serum p53 antibody has been widely used, it is mentioned that it is not related to clinical parameters of colorectal cancer (CRC) and has no prognostic significance in long-term follow-up. The aim of this study was to explore the possibility to increase the value of p53 antibody in combination with carcinoembryonic antigen (CEA) and CA19-9 for post operation follow up. METHODOLOGY: One hundred twenty-eight patients with primary CRC who underwent surgery were enrolled in this study. Serum p53 antibodies, CEA and CA19-9 were measured before and after the surgery and their impact on CRC staging patient survival. DNA was extracted from colorectal cancer tissue and KRAS mutation analysis was determined by direct sequencing. RESULTS: Thirty seven point five patients were positive for serum p53 antibodies before the operation. Total 67.2% patients were positive with any tumor markers, but it was only 3.9% that were positive with all markers. There was no association between KRAS mutation and p53 antibody level. CONCLUSIONS: The combined use of tumor markers can be an effective screening method for detection of colorectal cancer. If serum p53 antibody level continues to be positive for long after resection, the case should require subsequent intense follow-up.


Asunto(s)
Autoanticuerpos/sangre , Biomarcadores de Tumor/sangre , Colectomía , Neoplasias Colorrectales/cirugía , Proteína p53 Supresora de Tumor/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/genética , Antígeno CA-19-9/sangre , Antígeno Carcinoembrionario/sangre , Colectomía/efectos adversos , Colectomía/mortalidad , Neoplasias Colorrectales/sangre , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/mortalidad , Neoplasias Colorrectales/patología , Detección Precoz del Cáncer/métodos , Femenino , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Mutación , Estadificación de Neoplasias , Valor Predictivo de las Pruebas , Proteínas Proto-Oncogénicas p21(ras)/genética , Factores de Riesgo , Factores de Tiempo , Resultado del Tratamiento
17.
Biochem Biophys Res Commun ; 447(1): 216-22, 2014 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-24721431

RESUMEN

Xenotropic murine leukemia virus-related virus (XMRV) is a novel gammaretrovirus that was originally isolated from human prostate cancer. It is now believed that XMRV is not the etiologic agent of prostate cancer. An analysis of murine leukemia virus (MLV) infection in various human cell lines revealed that prostate cancer cell lines are preferentially infected by XMRV, and this suggested that XMRV infection may confer some sort of growth advantage to prostate cancer cell lines. To examine this hypothesis, androgen-dependent LNCaP cells were infected with XMRV and tested for changes in certain cell growth properties. We found that XMRV-infected LNCaP cells can proliferate in the absence of the androgen dihydrotestosterone. Moreover, androgen receptor expression is significantly reduced in XMRV-infected LNCaP cells. Such alterations were not observed in uninfected and amphotropic MLV-infected LNCaP cells. This finding explains why prostate cancer cell lines are preferentially infected with XMRV.


Asunto(s)
Andrógenos/farmacología , Proliferación Celular/efectos de los fármacos , Neoplasias de la Próstata/virología , Virus Relacionado con el Virus Xenotrópico de la Leucemia Murina/fisiología , Antagonistas de Receptores Androgénicos/farmacología , Anilidas/farmacología , Animales , Línea Celular Tumoral , Dihidrotestosterona/farmacología , Humanos , Masculino , Ratones , Nitrilos/farmacología , Ratas , Receptores Androgénicos/biosíntesis , Receptores Androgénicos/efectos de los fármacos , Compuestos de Tosilo/farmacología
18.
J Immunol ; 192(5): 2271-9, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24489086

RESUMEN

IRF4 is a transcription factor from the IRF factor family that plays pivotal roles in the differentiation and function of T and B lymphocytes. Although IRF4 is also expressed in dendritic cells (DCs) and macrophages, its roles in these cells in vivo are not clearly understood. In this study, conditional knockout mice that lack IRF4 in DCs or macrophages were generated and infected with Leishmania major. Mice lacking DC expression of IRF4 showed reduced footpad swelling compared with C57BL/6 mice, whereas those lacking IRF4 in macrophages did not. Mice with IRF4-deficient DCs also showed reduced parasite burden, and their CD4(+) T cells produced higher levels of IFN-γ in response to L. major Ag. In the draining lymph nodes, the proportion of activated CD4(+) T cells in these mice was similar to that in the control, but the proportion of IFN-γ-producing cells was increased, suggesting a Th1 bias in the immune response. Moreover, the numbers of migrating Langerhans cells and other migratory DCs in the draining lymph nodes were reduced both before and postinfection in mice with IRF4 defects in DCs, but higher levels of IL-12 were observed in IRF4-deficient DCs. These results imply that IRF4 expression in DCs inhibits their ability to produce IL-12 while promoting their migratory behavior, thus regulating CD4(+) T cell responses against local infection with L. major.


Asunto(s)
Factores Reguladores del Interferón/inmunología , Interleucina-12/inmunología , Células de Langerhans/inmunología , Leishmania major/metabolismo , Leishmaniasis Cutánea/inmunología , Células TH1/inmunología , Animales , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Factores Reguladores del Interferón/genética , Interferón gamma/inmunología , Interleucina-12/genética , Células de Langerhans/patología , Leishmaniasis Cutánea/genética , Leishmaniasis Cutánea/patología , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Noqueados , Células TH1/patología
19.
Virus Genes ; 48(3): 448-56, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24469466

RESUMEN

Ecotropic murine leukemia viruses (Eco-MLVs) infect mouse and rat, but not other mammalian cells, and gain access for infection through binding the cationic amino acid transporter 1 (CAT1). Glycosylation of the rat and hamster CAT1s inhibits Eco-MLV infection, and treatment of rat and hamster cells with a glycosylation inhibitor, tunicamycin, enhances Eco-MLV infection. Although the mouse CAT1 is also glycosylated, it does not inhibit Eco-MLV infection. Comparison of amino acid sequences between the rat and mouse CAT1s shows amino acid insertions in the rat protein near the Eco-MLV-binding motif. In addition to the insertion present in the rat CAT1, the hamster CAT1 has additional amino acid insertions. In contrast, tunicamycin treatment of mink and human cells does not elevate the infection, because their CAT1s do not have the Eco-MLV-binding motif. To define the evolutionary pathway of the Eco-MLV receptor, we analyzed CAT1 sequences and susceptibility to Eco-MLV infection of other several murinae animals, including the southern vole (Microtus rossiaemeridionalis), large Japanese field mouse (Apodemus speciosus), and Eurasian harvest mouse (Micromys minutus). Eco-MLV infection was enhanced by tunicamycin in these cells, and their CAT1 sequences have the insertions like the hamster CAT1. Phylogenetic analysis of mammalian CAT1s suggested that the ancestral CAT1 does not have the Eco-MLV-binding motif, like the human CAT1, and the mouse CAT1 is thought to be generated by the amino acid deletions in the third extracellular loop of CAT1.


Asunto(s)
Transportador de Aminoácidos Catiónicos 1/genética , Evolución Molecular , Virus de la Leucemia Murina/fisiología , Muridae/genética , Receptores Virales/genética , Infecciones por Retroviridae/virología , Enfermedades de los Roedores/virología , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Arvicolinae , Transportador de Aminoácidos Catiónicos 1/química , Transportador de Aminoácidos Catiónicos 1/metabolismo , Línea Celular , Cricetinae , Gerbillinae , Humanos , Ratones , Datos de Secuencia Molecular , Muridae/clasificación , Muridae/virología , Filogenia , Ratas , Receptores Virales/química , Receptores Virales/metabolismo
20.
Influenza Other Respir Viruses ; 8(1): 123-30, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24112132

RESUMEN

BACKGROUND: Influenza A viruses have an envelope made of a lipid bilayer and two surface glycoproteins, the hemagglutinin and the neuraminidase. The structure of the virus is directly dependent on the genetic makeup of the viral genome except the glycosylation moieties and the composition of the lipid bilayer. They both depend on the host cell and are in direct contact with the environment, such as air or water. Virus survival is important for virus transmission from contaminated waters in the case of wild aquatic birds or from contaminated surface or air for humans. OBJECTIVE: The objective of this study was to check whether the origin species of the host cell has an influence on influenza A virus survival. METHOD: The persistence in water at 35°C of viruses grown on either mammalian cells or avian cells and belonging to two different subtypes H1N1 and H5N1 was compared. RESULTS: Both H5N1 and H1N1 viruses remained infectious for periods of time as long as 19-25 days, respectively. However, within the same subtype, viruses grown on mammalian cells were more stable in water at 35°C than their counterparts grown on avian cells, even for viruses sharing the same genetic background. CONCLUSIONS: This difference in virus stability outside the host is probably connected to the nature of the lipid bilayer taken from the cell or to the carbohydrate side chains of the virus surface glycoproteins. Moreover, the long-lasting survival time might have a critical role in the ecology of influenza viruses, especially for avian viruses.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/fisiología , Subtipo H5N1 del Virus de la Influenza A/fisiología , Viabilidad Microbiana , Microbiología del Agua , Animales , Aves , Línea Celular , Perros , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA