Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 14: 1173605, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37435074

RESUMEN

Human metapneumovirus (HMPV) is a pneumovirus that may cause severe respiratory disease in humans. HMPV infection has been found to increase susceptibility to bacterial superinfections leading to increased morbidity and mortality. The molecular mechanisms underlying HMPV-mediated increase in bacterial susceptibility are poorly understood and largely understudied. Type I interferons (IFNs), while critical for antiviral defenses, may often have detrimental effects by skewing the host immune response and cytokine output of immune cells. It is currently unknown if HMPV skews the inflammatory response in human macrophages triggered by bacterial stimuli. Here we report that HMPV pre-infection impacts production of specific cytokines. HMPV strongly suppresses IL-1ß transcription in response to LPS or heat-killed Pseudomonas aeruginosa and Streptococcus pneumonia, while enhancing mRNA levels of IL-6, TNF-α and IFN-ß. We demonstrate that in human macrophages the HMPV-mediated suppression of IL-1ß transcription requires TANK-binding kinase 1 (TBK1) and signaling via the IFN-ß-IFNAR axis. Interestingly, our results show that HMPV pre-infection did not impair the LPS-stimulated activation of NF-κB and HIF-1α, transcription factors that stimulate IL-1ß mRNA synthesis in human cells. Furthermore, we determined that sequential HMPV-LPS treatment resulted in accumulation of the repressive epigenetic mark H3K27me3 at the IL1B promoter. Thus, for the first time we present data revealing the molecular mechanisms by which HMPV shapes the cytokine output of human macrophages exposed to bacterial pathogens/LPS, which appears to be dependent on epigenetic reprogramming at the IL1B promoter leading to reduced synthesis of IL-1ß. These results may improve current understanding of the role of type I IFNs in respiratory disease mediated not only by HMPV, but also by other respiratory viruses that are associated with superinfections.


Asunto(s)
Infecciones Bacterianas , Interferón beta , Interleucina-1beta , Infecciones por Paramyxoviridae , Sobreinfección , Humanos , Citocinas , Metapneumovirus , Transcripción Genética , Infecciones Bacterianas/inmunología , Infecciones por Paramyxoviridae/inmunología
2.
Front Immunol ; 12: 563336, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34248923

RESUMEN

The innate immune and host-protective responses to viruses, such as the airway pathogen human metapneumovirus (HMPV), depend on interferons (IFNs) that is induced through TANK-binding kinase 1 (TBK1) and IFN regulatory factors (IRFs). The transcription factor IRF1 is important for host resistance against several viruses and has a key role in induction of IFN-λ at mucosal surfaces. In most cell types IRF1 is expressed at very low levels, but its mRNA is rapidly induced when the demand for IRF1 activity arises. Despite general recognition of the importance of IRF1 to antiviral responses, the molecular mechanisms by which IRF1 is regulated during viral infections are not well understood. Here we identify the serine/threonine kinase TBK1 and IFN-ß as critical regulators of IRF1 mRNA and protein levels in human monocyte-derived macrophages. We find that inhibition of TBK1 activity either by the semi-selective TBK1/IKKε inhibitor BX795 or by siRNA-mediated knockdown abrogates HMPV-induced expression of IRF1. Moreover, we show that canonical NF-κB signaling is involved in IRF1 induction and that the TBK1/IKKε inhibitor BX795, but not siTBK1 treatment, impairs HMPV-induced phosphorylation of the NF-κB subunit p65. At later time-points of the infection, IRF1 expression depended heavily on IFN-ß-mediated signaling via the IFNAR-STAT1 pathway. Hence, our results suggest that TBK1 activation and TBK1/IKKε-mediated phosphorylation of the NF-κB subunit p65 control transcription of IRF1. Our study identifies a novel mechanism for IRF1 induction in response to viral infection of human macrophages that could be relevant not only to defense against HMPV, but also to other viral, bacterial and fungal pathogens.


Asunto(s)
Inmunidad Innata , Factor 1 Regulador del Interferón/genética , Factor 1 Regulador del Interferón/inmunología , Interferón Tipo I/inmunología , Metapneumovirus/inmunología , Proteínas Serina-Treonina Quinasas/inmunología , Línea Celular , Células Cultivadas , Humanos , Interferón Tipo I/genética , Metapneumovirus/genética , Monocitos/inmunología , Monocitos/virología , Proteínas Serina-Treonina Quinasas/genética , Transducción de Señal
3.
PLoS One ; 13(11): e0207374, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30485323

RESUMEN

ROR family of nuclear receptor transcription factors forms nodes connecting metabolic and inflammatory signaling pathways. The RORα members of the family have intrinsic transcriptional activity and they are involved in both activation and repression of a wide range of genes. The role of RORα in control of inflammation has been extensively studied using animal models but its function in human cells is not as well understood. To address this shortcoming, we analyzed how RORα is shaping the inflammatory state of human macrophages. Using CRISPR-Cas9 system, we deleted RORA in THP-1 human monocytic cell line. In mutant cells we observed a dramatic increase in basal expression of a subset of NF-κB regulated genes, including TNF, IL-1ß and IL-6, at both transcriptional and translational levels. Furthermore, RORA-deletion cells produced notable amounts of pro-IL-1ß even in the absence of LPS stimulation. Subsequent LPS stimulation induced cleavage of pro-IL-1ß to mature form. Our RNAseq analysis further confirmed the key role of RORA in setting the inflammatory state of macrophages and defined the set of differentially regulated genes. Overall, our data provides evidence supporting the anti-inflammatory function of RORα in human macrophages.


Asunto(s)
Citocinas/inmunología , Regulación de la Expresión Génica/inmunología , Macrófagos/inmunología , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/inmunología , Biosíntesis de Proteínas/inmunología , Transcripción Genética/inmunología , Citocinas/genética , Eliminación de Gen , Regulación de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Inflamación/inducido químicamente , Inflamación/genética , Inflamación/patología , Lipopolisacáridos/toxicidad , Macrófagos/patología , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Biosíntesis de Proteínas/efectos de los fármacos , Células THP-1 , Transcripción Genética/efectos de los fármacos
4.
J Immunol ; 192(5): 2291-304, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24477914

RESUMEN

The transcriptional repressor B lymphocyte-induced maturation protein 1 (BLIMP1) is a master regulator of B and T cell differentiation. To examine the role of BLIMP1 in innate immunity, we used a conditional knockout (CKO) of Blimp1 in myeloid cells and found that Blimp1 CKO mice were protected from lethal infection induced by Listeria monocytogenes. Transcriptome analysis of Blimp1 CKO macrophages identified the murine chemokine (C-C motif) ligand 8, CCL8, as a direct target of Blimp1-mediated transcriptional repression in these cells. BLIMP1-deficient macrophages expressed elevated levels of Ccl8, and consequently Blimp1 CKO mice had higher levels of circulating CCL8, resulting in increased neutrophils in the peripheral blood, promoting a more aggressive antibacterial response. Mice lacking the Ccl8 gene were more susceptible to L. monocytogenes infection than were wild-type mice. Although CCL8 failed to recruit neutrophils directly, it was chemotactic for γ/δ T cells, and CCL8-responsive γ/δ T cells were enriched for IL-17F. Finally, CCL8-mediated enhanced clearance of L. monocytogenes was dependent on γ/δ T cells. Collectively, these data reveal an important role for BLIMP1 in modulating host defenses by suppressing expression of the chemokine CCL8.


Asunto(s)
Quimiocina CCL8/inmunología , Regulación de la Expresión Génica/inmunología , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Macrófagos/inmunología , Factores de Transcripción/inmunología , Animales , Quimiocina CCL8/genética , Regulación de la Expresión Génica/genética , Listeriosis/genética , Macrófagos/patología , Ratones , Ratones Noqueados , Neutrófilos/inmunología , Neutrófilos/patología , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Linfocitos T/inmunología , Linfocitos T/patología , Factores de Transcripción/genética , Transcripción Genética/genética , Transcripción Genética/inmunología
5.
PLoS Genet ; 8(6): e1002761, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22719268

RESUMEN

Hepatic gluconeogenesis is required for maintaining blood glucose homeostasis; yet, in diabetes mellitus, this process is unrestrained and is a major contributor to fasting hyperglycemia. To date, the impacts of chromatin modifying enzymes and chromatin landscape on gluconeogenesis are poorly understood. Through catalyzing the removal of methyl groups from specific lysine residues in the histone tail, histone demethylases modulate chromatin structure and, hence, gene expression. Here we perform an RNA interference screen against the known histone demethylases and identify a histone H3 lysine 36 (H3K36) demethylase, Jhdm1a, as a key negative regulator of gluconeogenic gene expression. In vivo, silencing of Jhdm1a promotes liver glucose synthesis, while its exogenous expression reduces blood glucose level. Importantly, the regulation of gluconeogenesis by Jhdm1a requires its demethylation activity. Mechanistically, we find that Jhdm1a regulates the expression of a major gluconeogenic regulator, C/EBPα. This is achieved, at least in part, by its USF1-dependent association with the C/EBPα promoter and its subsequent demethylation of dimethylated H3K36 on the C/EBPα locus. Our work provides compelling evidence that links histone demethylation to transcriptional regulation of gluconeogenesis and has important implications for the treatment of diabetes.


Asunto(s)
Glucemia , Gluconeogénesis , Histona Demetilasas con Dominio de Jumonji , Hígado/metabolismo , Animales , Glucemia/metabolismo , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Células Cultivadas , Regulación de la Expresión Génica , Gluconeogénesis/genética , Glucosa-6-Fosfatasa/metabolismo , Hepatocitos/metabolismo , Humanos , Histona Demetilasas con Dominio de Jumonji/genética , Histona Demetilasas con Dominio de Jumonji/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Interferente Pequeño/genética , Ratas , Factores Estimuladores hacia 5'/metabolismo
6.
PLoS One ; 7(5): e35237, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22615734

RESUMEN

Angiogenesis is the process by which new blood vessels are formed from existing vessels. Mammalian populations, including humans and mice, harbor genetic variations that alter angiogenesis. Angiogenesis-regulating gene variants can result in increased susceptibility to multiple angiogenesis-dependent diseases in humans. Our efforts to dissect the complexity of the genetic diversity that regulates angiogenesis have used laboratory animals due to the availability of genome sequence for many species and the ability to perform high volume controlled breeding. Using the murine corneal micropocket assay, we have observed more than ten-fold difference in angiogenic responsiveness among various mouse strains. This degree of difference is observed with either bFGF or VEGF induced corneal neovascularization. Ongoing mapping studies have identified multiple loci that affect angiogenic responsiveness in several mouse models. In this study, we used F2 intercrosses between C57BL/6J and the 129 substrains 129P1/ReJ and 129P3/J, as well as the SJL/J strain, where we have identified new QTLs that affect angiogenic responsiveness. In the case of AngFq5, on chromosome 7, congenic animals were used to confirm the existence of this locus and subcongenic animals, combined with a haplotype-based mapping approach that identified the pink-eyed dilution mutation as a candidate polymorphism to explain AngFq5. The ability of mutations in the pink-eyed dilution gene to affect angiogenic response was demonstrated using the p-J allele at the same locus. Using this allele, we demonstrate that pink-eyed dilution mutations in Oca2 can affect both bFGF and VEGF-induced corneal angiogenesis.


Asunto(s)
Color del Ojo/genética , Mutación , Neovascularización Fisiológica/genética , Animales , Haplotipos , Ratones , Ratones Endogámicos C57BL , Polimorfismo Genético
7.
Infect Immun ; 79(4): 1526-35, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21263022

RESUMEN

Host cells use metabolic signaling through the LXRα nuclear receptor to defend against Listeria monocytogenes infection. 25-Hydroxycholesterol is a natural ligand of LXRs that is produced by the enzyme cholesterol 25-hydroxylase (CH25H). We found that expression of Ch25h is upregulated following L. monocytogenes infection in a beta interferon (IFN-ß)-dependent fashion. Moreover, increased Ch25h expression promotes survival of L. monocytogenes-infected cells and increases sensitivity of the host to infection. We determined that expression of Cd5l, a prosurvival gene, is controlled by CH25H. In addition, we found that CD5L inhibits activation of caspase-1, promoting survival of infected macrophages. Our results reveal a mechanism by which an intracellular pathogen can prolong survival of infected cells, thus providing itself with a protected environment in which to replicate.


Asunto(s)
Listeria monocytogenes/fisiología , Listeriosis/metabolismo , Macrófagos/metabolismo , Macrófagos/microbiología , Transducción de Señal/fisiología , Esteroide Hidroxilasas/biosíntesis , Animales , Proteínas Reguladoras de la Apoptosis/biosíntesis , Western Blotting , Femenino , Expresión Génica , Regulación de la Expresión Génica/genética , Etiquetado Corte-Fin in Situ , Listeriosis/inmunología , Ratones , Ratones Endogámicos C57BL , Receptores Inmunológicos/biosíntesis , Receptores Depuradores , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Regulación hacia Arriba
8.
Proc Natl Acad Sci U S A ; 107(4): 1594-9, 2010 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-20080646

RESUMEN

Candida albicans is a ubiquitous opportunistic pathogen that is the most prevalent cause of hospital-acquired fungal infections. In mammalian hosts, C. albicans is engulfed by phagocytes that attack the pathogen with DNA-damaging reactive oxygen species (ROS). Acetylation of histone H3 lysine 56 (H3K56) by the fungal-specific histone acetyltransferase Rtt109 is important for yeast model organisms to survive DNA damage and maintain genome integrity. To assess the importance of Rtt109 for C. albicans pathogenicity, we deleted the predicted homolog of Rtt109 in the clinical C. albicans isolate, SC5314. C. albicans rtt109(-/-) mutant cells lack acetylated H3K56 (H3K56ac) and are hypersensitive to genotoxic agents. Additionally, rtt109(-/-) mutant cells constitutively display increased H2A S129 phosphorylation and elevated DNA repair gene expression, consistent with endogenous DNA damage. Importantly, C. albicans rtt109(-/-) cells are significantly less pathogenic in mice and more susceptible to killing by macrophages in vitro than are wild-type cells. Via pharmacological inhibition of the host NADPH oxidase enzyme, we show that the increased sensitivity of rtt109(-/-) cells to macrophages depends on the host's ability to generate ROS, providing a mechanistic link between the drug sensitivity, gene expression, and pathogenesis phenotypes. We conclude that Rtt109 is particularly important for fungal pathogenicity, suggesting a unique target for therapeutic antifungal compounds.


Asunto(s)
Candida albicans/enzimología , Candidiasis/microbiología , Histona Acetiltransferasas/metabolismo , Animales , Candida albicans/genética , Candida albicans/crecimiento & desarrollo , Candida albicans/patogenicidad , Supervivencia Celular , Reparación del ADN , ADN de Hongos/genética , ADN de Hongos/metabolismo , Femenino , Regulación Fúngica de la Expresión Génica , Histona Acetiltransferasas/genética , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno/metabolismo
9.
Infect Immun ; 78(2): 586-94, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19995898

RESUMEN

Host susceptibility to infection is controlled in large measure by the genetic makeup of the host. Spirochetes of the genus Borrelia include nearly 40 species of vector-borne spirochetes that are capable of infecting a wide range of mammalian hosts, causing Lyme disease and relapsing fever. Relapsing fever is associated with high-level bacteremia, as well as hematologic manifestations, such as thrombocytopenia (i.e., low platelet numbers) and anemia. To facilitate studies of genetic control of susceptibility to Borrelia hermsii infection, we performed a systematic analysis of the course of infection using immunocompetent and immunocompromised inbred strains of mice. Our analysis revealed that sensitivity to B. hermsii infections is genetically controlled. In addition, whereas the role of adaptive immunity to relapsing fever-causing spirochetes is well documented, we found that innate immunity contributes significantly to the reduction of bacterial burden. Similar to human infection, the progression of the disease in mice was associated with thrombocytopenia and anemia. Histological and fluorescence in situ hybridization (FISH) analysis of infected tissues indicated that red blood cells (RBCs) were removed by tissue-resident macrophages, a process that could lead to anemia. Spirochetes in the spleen and liver were often visualized associated with RBCs, lending support to the hypothesis that direct interaction of B. hermsii spirochetes with RBCs leads to clearance of bacteria from the bloodstream by tissue phagocytes.


Asunto(s)
Predisposición Genética a la Enfermedad , Inmunidad Innata/genética , Fiebre Recurrente/genética , Fiebre Recurrente/inmunología , Anemia/genética , Anemia/microbiología , Animales , Progresión de la Enfermedad , Femenino , Citometría de Flujo , Hibridación Fluorescente in Situ , Masculino , Ratones , Ratones Endogámicos , Fiebre Recurrente/patología , Factores Sexuales , Trombocitopenia/genética , Trombocitopenia/microbiología
10.
Neoplasia ; 11(9): 835-45, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19724677

RESUMEN

Intrahepatic and extrahepatic metastases are common findings in hepatocellular carcinoma (HCC). Insulin-like growth factor 2 (IGF2) expression is frequently induced in HCC, and serum IGF2 levels correlate with the presence of extrahepatic metastases. Yet, the role of IGF-induced signaling in the dissemination of HCC remains unclear. We have previously observed elevated IGF2 levels in tumors with metastatic potential in an HCC mouse model. Here, we demonstrate that inhibition of IGF2, or its receptor IGF1R, impairs the migration and invasion activities of murine HCC cells. Furthermore, inhibition of IGF1R also impairs the ability of HCC cells to colonize the lungs after introduction into the circulation through the tail vein but does not impair subcutaneous tumor growth. Collectively, these findings suggest that IGF1R-mediated signaling plays a causative role in tumor dissemination but is not required for tumor growth per se. Although previous studies indicate that IGF ligands can signal through IGF1R/insulin receptor (IR) heterodimers, and IR-A homodimers, we demonstrate that the IR is not required for invasion and metastasis by HCC cells. Finally, we identify matrix metalloproteinase 2 as a mediator of the invasive phenotype downstream of IGF1R-induced signaling. Thus, our studies demonstrate the importance of IGF2-induced signaling in the dissemination of HCC cells.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Receptor IGF Tipo 1/fisiología , Receptor de Insulina/fisiología , Transducción de Señal , Animales , Western Blotting , Carcinoma Hepatocelular/patología , Movimiento Celular , Proliferación Celular , Immunoblotting , Proteínas Sustrato del Receptor de Insulina/metabolismo , Factor II del Crecimiento Similar a la Insulina/metabolismo , Neoplasias Hepáticas/patología , Masculino , Metaloproteinasa 3 de la Matriz/genética , Metaloproteinasa 3 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Desnudos , Invasividad Neoplásica , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
11.
Infect Immun ; 77(5): 1945-58, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19289513

RESUMEN

Haemophilus influenzae efficiently colonizes and persists at the human nasopharyngeal mucosa, causing disease when it spreads to other sites. Nitric oxide (NO) represents a major antimicrobial defense deployed by host cells in locations colonized by H. influenzae during pathogenesis that are likely to vary in oxygen levels. Formate-dependent nitrite reductase regulator (FNR) is an oxygen-sensitive regulator in several bacterial pathogens. We report that fnr of H. influenzae is required for anaerobic defense against exposure to NO donors and to resist NO-dependent effects of gamma interferon (IFN-gamma)-activated murine bone marrow-derived macrophages. To understand the mechanism of resistance, we investigated the role of FNR-regulated genes in defense against NO sources. Expression analysis revealed FNR-dependent activation of nrfA, dmsA, napA, and ytfE. Nonpolar deletion mutants of nrfA and ytfE exhibited sensitivity to NO donors, and the ytfE gene was more critical for survival. Compared to the wild-type strain, the ytfE mutant exhibited decreased survival when exposed to macrophages, a defect that was more pronounced after prior stimulation of macrophages with IFN-gamma or lipopolysaccharide. Complementation restored survival of the mutant to the level in the parental strain. Increased sensitivity of the ytfE mutant relative to that of the parent was abrogated by treatment of macrophages with a NO synthase inhibitor, implicating YtfE in resistance to a NO-dependent pathway. These results identify a requirement for FNR in positive control of ytfE and indicate a critical role for ytfE in resistance of H. influenzae to reactive nitrogen species and the antibacterial effects of macrophages.


Asunto(s)
Proteínas Bacterianas/fisiología , Regulación de la Expresión Génica , Haemophilus influenzae/fisiología , Macrófagos/microbiología , Óxido Nítrico/toxicidad , Factores de Transcripción/fisiología , Animales , Proteínas Bacterianas/genética , Eliminación de Gen , Perfilación de la Expresión Génica , Prueba de Complementación Genética , Haemophilus influenzae/efectos de los fármacos , Haemophilus influenzae/genética , Haemophilus influenzae/inmunología , Humanos , Ratones , Viabilidad Microbiana , Factores de Transcripción/genética , Factores de Virulencia/genética , Factores de Virulencia/fisiología
12.
PLoS Genet ; 3(9): 1587-97, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17845078

RESUMEN

Genetic makeup of the host plays a significant role in the course and outcome of infection. Inbred strains of mice display a wide range of sensitivities to Listeria monocytogenes infection and thus serve as a good model for analysis of the effect of genetic polymorphism. The outcome of L. monocytogenes infection in mice is influenced by the ability of this bacterium to induce expression of interferon beta mRNA, encoded in mouse by the Ifnb1 (interferon beta 1, fibroblast) gene. Mouse strains that lack components of the IFN beta signaling pathway are substantially more resistant to infection. We found that macrophages from the ByJ substrain of the common C57BL/6 inbred strain of mice are impaired in their ability to induce Ifnb1 expression in response to bacterial and viral infections. We mapped the locus that controls differential expression of Ifnb1 to a region on Chromosome 7 that includes interferon regulatory factor 3 (Irf3), which encodes a transcription factor responsible for early induction of Ifnb1 expression. In C57BL/6ByJ mice, Irf3 mRNA was inefficiently spliced, with a significant proportion of the transcripts retaining intron 5. Analysis of the Irf3 locus identified a single base-pair polymorphism and revealed that intron 5 of Irf3 is spliced by the atypical U12-type spliceosome. We found that the polymorphism disrupts a U12-type branchpoint and has a profound effect on the efficiency of splicing of Irf3. We demonstrate that a naturally occurring change in the splicing control element has a dramatic effect on the resistance to L. monocytogenes infection. Thus, the C57BL/6ByJ mouse strain serves as an example of how a mammalian host can counter bacterial virulence strategies by introducing subtle alteration of noncoding sequences.


Asunto(s)
Factor 3 Regulador del Interferón/genética , Interferón beta/biosíntesis , Listeriosis/metabolismo , Polimorfismo Genético , Animales , Línea Celular , Interferón beta/genética , Listeriosis/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
13.
Cancer Res ; 67(16): 7589-96, 2007 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-17699762

RESUMEN

Hepatocellular carcinoma (HCC) is a leading cause of cancer-related death worldwide. HCC patients frequently present with disease that has metastasized to other regions of the liver, the portal vein, lymph nodes, or lungs, leading to poor prognoses. Therefore, model systems that allow exploration of the molecular mechanisms underlying metastasis in this disease are greatly needed. We describe here a metastatic HCC model generated after the somatic introduction of the mouse polyoma virus middle T antigen to mice with liver-specific deletion of the Trp53 tumor suppressor locus and show the cell autonomous effect of p53 loss of function on HCC metastasis. We additionally find that cholangiocarcinoma also develops in these mice, and some tumors display features of both HCC and cholangiocarcinoma, suggestive of origin from liver progenitor cells. Concomitant loss of the Ink4a/Arf tumor suppressor locus accelerates tumor formation and metastasis, suggesting potential roles for the p16 and p19 tumor suppressors in this process. Significantly, tumor cell lines isolated from tumors lacking both Trp53 and Ink4a/Arf display enhanced invasion activity in vitro relative to those lacking Trp53 alone. Thus, our data illustrate a new model system amenable for the analysis of HCC metastasis.


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina/deficiencia , Neoplasias Hepáticas Experimentales/patología , Neoplasias Pulmonares/secundario , Proteína p53 Supresora de Tumor/deficiencia , Animales , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Eliminación de Gen , Neoplasias Hepáticas Experimentales/genética , Ratones , Ratones Transgénicos , Proteína p53 Supresora de Tumor/genética
14.
Nat Genet ; 39(8): 1025-32, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17632513

RESUMEN

We undertook a quantitative trait locus (QTL) analysis in mice to identify modifier genes that might influence the severity of human iron disorders. We identified a strong QTL on mouse chromosome 9 that differentially affected macrophage iron burden in C57BL/10J and SWR/J mice. A C57BL/10J missense allele of an evolutionarily conserved gene, Mon1a, cosegregated with the QTL in congenic mouse lines. We present evidence that Mon1a is involved in trafficking of ferroportin, the major mammalian iron exporter, to the surface of iron-recycling macrophages. Differences in amounts of surface ferroportin correlate with differences in cellular iron content. Mon1a is also important for trafficking of cell-surface and secreted molecules unrelated to iron metabolism, suggesting that it has a fundamental role in the mammalian secretory apparatus.


Asunto(s)
Proteínas Portadoras/genética , Hierro/metabolismo , Macrófagos/metabolismo , Sitios de Carácter Cuantitativo , Animales , Proteínas Portadoras/metabolismo , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Cromosomas de los Mamíferos , Cruzamientos Genéticos , Femenino , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos , Transporte de Proteínas , ARN Interferente Pequeño
15.
Brief Funct Genomic Proteomic ; 4(3): 258-69, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16420751

RESUMEN

For almost half a century, the mouse model of Listeria monocytogenes infection has been used to analyse both innate and adaptive components of immunity and to discover key immune genes. Vast accumulated knowledge about the disease in mice provides a unique framework for identifying and characterising immune molecules using a variety of experimental approaches. To illustrate the range of questions that can be addressed using modern genetics and genomics tools, the authors provide an overview of the analysis of components of immune signalling networks using the mouse model of L. monocytogenes infection.


Asunto(s)
Listeria monocytogenes/metabolismo , Animales , Complemento C5/genética , Variación Genética , Genética , Genoma , Genómica , Humanos , Sistema Inmunológico/patología , Inmunidad Activa , Inmunidad Innata , Listeriosis/genética , Ratones , Mutación , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas , Transducción de Señal , Transgenes
16.
J Immunol ; 173(8): 5112-20, 2004 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-15470055

RESUMEN

Epidemiological, clinical, and experimental approaches have convincingly demonstrated that host resistance to infection with intracellular pathogens is significantly influenced by genetic polymorphisms. Using a mouse model of infection with virulent Mycobacterium tuberculosis (MTB), we have previously identified the sst1 locus as a genetic determinant of host resistance to tuberculosis. In this study we demonstrate that susceptibility to another intracellular pathogen, Listeria monocytogenes, is also influenced by the sst1 locus. The contribution of sst1 to anti-listerial immunity is much greater in immunodeficient scid mice, indicating that this locus controls innate immunity and becomes particularly important when adaptive immunity is significantly depressed. Similar to our previous observations using infection with MTB, the resistant allele of sst1 prevents formation of necrotic infectious lesions in vivo. We have shown that macrophages obtained from sst1-resistant congenic mice possess superior ability to kill L. monocytogenes in vitro. The bactericidal effect of sst1 is dependent on IFN-gamma activation and reactive oxygen radical production by activated macrophages after infection, but is independent of NO production. It is possible that there is a single gene that controls common IFN-dependent macrophage function, which is important in the pathogenesis of infections caused by both MTB and L. monocytogenes. However, host resistance to the two pathogens may be controlled by two different polymorphic genes encoded within the sst1 locus. The polymorphic gene(s) encoded within the sst1 locus that controls macrophage interactions with the two intracellular pathogens remains to be elucidated.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Inmunidad Innata/genética , Listeriosis/inmunología , Animales , Mapeo Cromosómico , Interferón gamma/farmacología , Células Asesinas Naturales/inmunología , Listeria monocytogenes/inmunología , Listeriosis/genética , Activación de Macrófagos , Macrófagos/microbiología , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones SCID , Óxido Nítrico/fisiología , Fagocitos/fisiología , Polimorfismo Genético , Especies Reactivas de Oxígeno , Linfocitos T/inmunología
17.
Proc Natl Acad Sci U S A ; 100(20): 11594-9, 2003 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-14500783

RESUMEN

The B10.Q/J strain of mice was serendipitously discovered to be highly susceptible to infection by the intracellular protozoan parasite, Toxoplasma gondii but markedly resistant to induction of autoimmune arthritis. We have previously shown that the B10.Q/J phenotype is controlled by a single recessive locus and is associated with lymphocyte hyporesponsiveness to IL-12. Using genetic approaches, we have now localized the B10.Q/J locus to chromosome 9 and established its identity as Tyk2, a Janus kinase essential for IL-12 and IFN-alpha/beta cytokine signaling. The B10.Q/J Tyk2 gene contained a single missense mutation resulting in a nonconservative amino acid substitution (E775K) in an invariant motif of the pseudokinase (Janus kinase homology 2) domain. This mutation appeared to result in the absence of the B10.Q/J-encoded Tyk2 protein, despite presence of Tyk2-specific transcripts. Phenotypically, B10.Q/J cells were indistinguishable from Tyk2-deficient cells, showing impaired signaling and biologic responses to IL-12, IL-23, and type I IFNs. The analogous E782K mutant of human Tyk2 failed to restore IFN-alpha responsiveness in Tyk2 null 11,1 cells. Our results indicate a crucial role for Tyk2 in T helper 1-mediated protective and pathogenic immune responses. An additional implication of our findings is that naturally occurring mutations in the Tyk2 gene may underlie altered susceptibilities to infectious or autoimmune diseases in human and animal populations.


Asunto(s)
Autoinmunidad/genética , Predisposición Genética a la Enfermedad , Infecciones/genética , Mutación , Proteínas Tirosina Quinasas , Proteínas/inmunología , Animales , Regulación Enzimológica de la Expresión Génica , Prueba de Complementación Genética , Ratones , Ratones Endogámicos BALB C , Mutagénesis , Mutación Puntual , Proteínas/genética , TYK2 Quinasa
18.
Genetics ; 161(2): 509-19, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12072450

RESUMEN

Spt3 of Saccharomyces cerevisiae is required for the normal transcription of many genes in vivo. Past studies have shown that Spt3 is required for both mating and sporulation, two events that initiate when cells are at G(1)/START. We now show that Spt3 is needed for two other events that begin at G(1)/START, diploid filamentous growth and haploid invasive growth. In addition, Spt3 is required for normal expression of FLO11, a gene required for filamentous growth, although this defect is not the sole cause of the spt3Delta/spt3Delta filamentous growth defect. To extend our studies of Spt3's role in filamentous growth to the pathogenic yeast Candida albicans, we have identified the C. albicans SPT3 gene and have studied its role in C. albicans filamentous growth and virulence. Surprisingly, C. albicans spt3Delta/spt3Delta mutants are hyperfilamentous, the opposite phenotype observed for S. cerevisiae spt3Delta/spt3Delta mutants. Furthermore, C. albicans spt3Delta/spt3Delta mutants are avirulent in mice. These experiments demonstrate that Spt3 plays important but opposite roles in filamentous growth in S. cerevisiae and C. albicans.


Asunto(s)
Candida albicans/crecimiento & desarrollo , Proteínas Fúngicas/fisiología , Saccharomyces cerevisiae/crecimiento & desarrollo , Animales , Candida albicans/genética , Candida albicans/patogenicidad , Proteínas Fúngicas/genética , Glicoproteínas de Membrana , Proteínas de la Membrana/genética , Ratones , Datos de Secuencia Molecular , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/fisiología , Factores de Transcripción , Virulencia
19.
Curr Opin Microbiol ; 5(1): 111-7, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11834379

RESUMEN

Although the role of host heredity in susceptibility to infectious diseases is significant, the genetic control of immunity to infection remains poorly understood. Advances in experimental and epidemiological analyses of complex genetic traits have led to the discoveries of novel genetic determinants of host resistance. New loci that control susceptibility to a number of intracellular pathogens have been identified using mouse models of infectious diseases. The contributions of individual loci, however, vary in quantitative and qualitative manner, depending on mechanisms of pathogen virulence and genetic background of the host. In this review, we discuss how genetic analysis of host resistance contributes to further understanding of host immunity and pathogenesis of intracellular infections.


Asunto(s)
Predisposición Genética a la Enfermedad , Inmunidad/genética , Infecciones/genética , Infecciones/inmunología , Carácter Cuantitativo Heredable , Animales , Humanos , Ratones , Mutación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...