Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Hepatol Commun ; 3(8): 1085-1097, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31388629

RESUMEN

Farnesoid X receptor (FXR) agonism is emerging as an important potential therapeutic mechanism of action for multiple chronic liver diseases. The bile acid-derived FXR agonist obeticholic acid (OCA) has shown promise in a phase 2 study in patients with nonalcoholic steatohepatitis (NASH). Here, we report efficacy of the novel nonbile acid FXR agonist tropifexor (LJN452) in two distinct preclinical models of NASH. The efficacy of tropifexor at <1 mg/kg doses was superior to that of OCA at 25 mg/kg in the liver in both NASH models. In a chemical and dietary model of NASH (Stelic animal model [STAM]), tropifexor reversed established fibrosis and reduced the nonalcoholic fatty liver disease activity score and hepatic triglycerides. In an insulin-resistant obese NASH model (amylin liver NASH model [AMLN]), tropifexor markedly reduced steatohepatitis, fibrosis, and profibrogenic gene expression. Transcriptome analysis of livers from AMLN mice revealed 461 differentially expressed genes following tropifexor treatment that included a combination of signatures associated with reduction of oxidative stress, fibrogenesis, and inflammation. Conclusion: Based on preclinical validation in animal models, tropifexor is a promising investigational therapy that is currently under phase 2 development for NASH.

2.
J Med Chem ; 61(24): 11021-11036, 2018 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-30407821

RESUMEN

Spinal muscular atrophy (SMA), a rare neuromuscular disorder, is the leading genetic cause of death in infants and toddlers. SMA is caused by the deletion or a loss of function mutation of the survival motor neuron 1 (SMN1) gene. In humans, a second closely related gene SMN2 exists; however it codes for a less stable SMN protein. In recent years, significant progress has been made toward disease modifying treatments for SMA by modulating SMN2 pre-mRNA splicing. Herein, we describe the discovery of LMI070/branaplam, a small molecule that stabilizes the interaction between the spliceosome and SMN2 pre-mRNA. Branaplam (1) originated from a high-throughput phenotypic screening hit, pyridazine 2, and evolved via multiparameter lead optimization. In a severe mouse SMA model, branaplam treatment increased full-length SMN RNA and protein levels, and extended survival. Currently, branaplam is in clinical studies for SMA.


Asunto(s)
Encéfalo/efectos de los fármacos , Canal de Potasio ERG1/metabolismo , Atrofia Muscular Espinal/tratamiento farmacológico , Piridazinas/química , Administración Oral , Animales , Encéfalo/metabolismo , Línea Celular , Cristalografía por Rayos X , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos/métodos , Canal de Potasio ERG1/antagonistas & inhibidores , Humanos , Ratones Endogámicos C57BL , Neuronas Motoras/efectos de los fármacos , Atrofia Muscular Espinal/genética , Piridazinas/farmacología , Relación Estructura-Actividad Cuantitativa , Empalme del ARN , Ratas Sprague-Dawley , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Proteína 1 para la Supervivencia de la Neurona Motora/metabolismo , Proteína 2 para la Supervivencia de la Neurona Motora/genética
5.
Nat Chem Biol ; 11(7): 511-7, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26030728

RESUMEN

Spinal muscular atrophy (SMA), which results from the loss of expression of the survival of motor neuron-1 (SMN1) gene, represents the most common genetic cause of pediatric mortality. A duplicate copy (SMN2) is inefficiently spliced, producing a truncated and unstable protein. We describe herein a potent, orally active, small-molecule enhancer of SMN2 splicing that elevates full-length SMN protein and extends survival in a severe SMA mouse model. We demonstrate that the molecular mechanism of action is via stabilization of the transient double-strand RNA structure formed by the SMN2 pre-mRNA and U1 small nuclear ribonucleic protein (snRNP) complex. The binding affinity of U1 snRNP to the 5' splice site is increased in a sequence-selective manner, discrete from constitutive recognition. This new mechanism demonstrates the feasibility of small molecule-mediated, sequence-selective splice modulation and the potential for leveraging this strategy in other splicing diseases.


Asunto(s)
Empalme Alternativo , Atrofia Muscular Espinal/tratamiento farmacológico , ARN Bicatenario/agonistas , Ribonucleoproteína Nuclear Pequeña U1/agonistas , Bibliotecas de Moléculas Pequeñas/farmacología , Proteína 2 para la Supervivencia de la Neurona Motora/metabolismo , Animales , Sitios de Unión , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Humanos , Ratones , Ratones Transgénicos , Modelos Moleculares , Atrofia Muscular Espinal/metabolismo , Atrofia Muscular Espinal/mortalidad , Atrofia Muscular Espinal/patología , Unión Proteica/efectos de los fármacos , Estabilidad Proteica/efectos de los fármacos , Proteolisis , Precursores del ARN/agonistas , Precursores del ARN/química , Precursores del ARN/metabolismo , ARN Bicatenario/química , ARN Bicatenario/metabolismo , Ribonucleoproteína Nuclear Pequeña U1/química , Ribonucleoproteína Nuclear Pequeña U1/metabolismo , Bibliotecas de Moléculas Pequeñas/síntesis química , Bibliotecas de Moléculas Pequeñas/metabolismo , Análisis de Supervivencia , Proteína 2 para la Supervivencia de la Neurona Motora/química , Proteína 2 para la Supervivencia de la Neurona Motora/genética
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.
Nat Immunol ; 9(10): 1171-8, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18724372

RESUMEN

Inflammasomes are cytosolic multiprotein complexes that sense microbial infection and trigger cytokine production and cell death. However, the molecular components of inflammasomes and what they sense remain poorly defined. Here we demonstrate that 35 amino acids of the carboxyl terminus of flagellin triggered inflammasome activation in the absence of bacterial contaminants or secretion systems. To further elucidate the host flagellin-sensing pathway, we generated mice deficient in the intracellular sensor Naip5. These mice failed to activate the inflammasome in response to the 35 amino acids of flagellin or in response to Legionella pneumophila infection. Our data clarify the molecular basis for the cytosolic response to flagellin.


Asunto(s)
Flagelina/inmunología , Macrófagos/inmunología , Complejos Multiproteicos/inmunología , Proteína Inhibidora de la Apoptosis Neuronal/inmunología , Secuencias de Aminoácidos/inmunología , Animales , Proteínas Reguladoras de la Apoptosis/inmunología , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas de Unión al Calcio/inmunología , Proteínas de Unión al Calcio/metabolismo , Citosol , Ensayo de Inmunoadsorción Enzimática , Flagelina/química , Immunoblotting , Legionella pneumophila/inmunología , Enfermedad de los Legionarios/inmunología , Macrófagos/microbiología , Ratones , Proteína Inhibidora de la Apoptosis Neuronal/genética , Receptor Toll-Like 5/inmunología , Receptor Toll-Like 5/metabolismo , Transducción Genética
8.
J Immunol ; 180(9): 6237-45, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18424746

RESUMEN

Chlamydiae are obligate intracellular bacterial pathogens that exhibit a broad range of host tropism. Differences in host tropism between Chlamydia species have been linked to host variations in IFN-gamma-mediated immune responses. In mouse cells, IFN-gamma can effectively restrict growth of the human pathogen Chlamydia trachomatis but fails to control growth of the closely related mouse pathogen Chlamydia muridarum. The ability of mouse cells to resist C. trachomatis replication is largely dependent on the induction of a family of IFN-gamma-inducible GTPases called immunity-related GTPases or IRGs. In this study we demonstrate that C. muridarum can specifically evade IRG-mediated host resistance. It has previously been suggested that C. muridarum inactivates the IRG protein Irga6 (Iigp1) to dampen the murine immune response. However, we show that Irga6 is dispensable for the control of C. trachomatis replication. Instead, an effective IFN-gamma response to C. trachomatis requires the IRG proteins Irgm1 (Lrg47), Irgm3 (Igtp), and Irgb10. Ectopic expression of Irgb10 in the absence of IFN-gamma is sufficient to reduce intracellular growth of C. trachomatis but fails to restrict growth of C. muridarum, indicating that C. muridarum can specifically evade Irgb10-driven host responses. Importantly, we find that Irgb10 protein intimately associates with inclusions harboring C. trachomatis but is absent from inclusions formed by C. muridarum. These data suggest that C. muridarum has evolved a mechanism to escape the murine IFN-gamma response by restricting access of Irgb10 and possibly other IRG proteins to the inclusion.


Asunto(s)
Infecciones por Chlamydia/inmunología , Chlamydia muridarum/inmunología , GTP Fosfohidrolasas/inmunología , Inmunidad Innata , Cuerpos de Inclusión/inmunología , Interferón gamma/inmunología , Animales , Evolución Biológica , Células Cultivadas , Infecciones por Chlamydia/genética , Chlamydia muridarum/crecimiento & desarrollo , Chlamydia trachomatis/crecimiento & desarrollo , Chlamydia trachomatis/inmunología , GTP Fosfohidrolasas/genética , Inmunidad Innata/genética , Cuerpos de Inclusión/genética , Cuerpos de Inclusión/microbiología , Interferón gamma/genética , Ratones , Ratones Noqueados
9.
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
10.
Cell Microbiol ; 9(10): 2344-57, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17506816

RESUMEN

Macrophages from the C57BL/6 (B6) mouse strain restrict intracellular growth of Legionella pneumophila, whereas A/J macrophages are highly permissive. The mechanism by which B6 macrophages restrict Legionella growth remains poorly understood, but is known to require the cytosolic microbe sensors Naip5 (Birc1e) and Ipaf. We hypothesized that Naip5 and Ipaf may act in partnership with other antimicrobial signalling pathways in macrophages. Indeed, we found that macrophages lacking either tumour necrosis factor (TNF)-alpha or type I interferon (IFN) signalling are permissive for growth of L. pneumophila, even in the presence of functional Naip5 and Ipaf alleles. Similarly, macrophages lacking Naip5 and/or Ipaf signalling were permissive even though we found that Naip5 or Ipaf were not required for induction of TNF-alpha and type I IFN. Therefore, our data suggest that the mechanism by which B6 macrophages restrict intracellular replication of L. pneumophila is more complex than previously appreciated, and involves the concerted action of cytokine and intracellular microbe sensor signalling pathways.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/fisiología , Proteínas de Unión al Calcio/fisiología , Interferón Tipo I/fisiología , Legionella pneumophila/fisiología , Macrófagos/microbiología , Proteína Inhibidora de la Apoptosis Neuronal/fisiología , Factor de Necrosis Tumoral alfa/fisiología , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas de Unión al Calcio/genética , Células Cultivadas , Interferón Tipo I/biosíntesis , Mediciones Luminiscentes , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Inhibidora de la Apoptosis Neuronal/genética , Transducción de Señal , Factor de Necrosis Tumoral alfa/biosíntesis
11.
Proc Natl Acad Sci U S A ; 103(38): 14092-7, 2006 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-16959883

RESUMEN

Infections caused by the bacteria Chlamydia trachomatis contribute to diverse pathologies in a variety of human populations. We previously used a systemic model of C. trachomatis infection in mice to map three quantitative trait loci that influence in vivo susceptibility differences between the C57BL/6J and C3H/HeJ inbred strains of mouse. One of these quantitative trait loci, Ctrq-3, influences an IFN-gamma-dependent susceptibility difference in primary embryonic fibroblasts isolated from these strains. Here we use fine structure mapping in congenic fibroblasts carrying DNA from the susceptible parent to localize the effect of Ctrq-3 to a 1.2-megabase interval of genomic DNA that contains Irgb10 and Igtp, two members of the IFN-gamma-inducible p47 family of GTPases. This class of proteins has been widely implicated in resistance to intracellular pathogens in mice. We analyzed expression of Irgb10 and Igtp in parental and congenic embryonic fibroblasts treated with IFN-gamma and found that relatively resistant fibroblasts express more Irgb10 than relatively susceptible fibroblasts. However, we also found that abolishing the expression of either Irgb10 or Igtp increases susceptibility of embryonic fibroblasts to C. trachomatis. Thus, we conclude that, although a difference in Irgb10 expression is likely responsible for the effect of Ctrq-3 on susceptibility to C. trachomatis, both genes play a role in intracellular resistance to C. trachomatis.


Asunto(s)
Infecciones por Chlamydia/genética , Chlamydia trachomatis , Mapeo Cromosómico , Susceptibilidad a Enfermedades , GTP Fosfohidrolasas , Secuencia de Aminoácidos , Animales , Células Cultivadas , Infecciones por Chlamydia/inmunología , Chlamydia trachomatis/inmunología , Cromosomas de los Mamíferos , Fibroblastos/citología , Fibroblastos/fisiología , GTP Fosfohidrolasas/genética , GTP Fosfohidrolasas/metabolismo , Humanos , Interferón gamma/metabolismo , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Modelos Genéticos , Datos de Secuencia Molecular
12.
PLoS Pathog ; 2(3): e18, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16552444

RESUMEN

Macrophages from C57BL/6J (B6) mice restrict growth of the intracellular bacterial pathogen Legionella pneumophila. Restriction of bacterial growth requires caspase-1 and the leucine-rich repeat-containing protein Naip5 (Birc1e). We identified mutants of L. pneumophila that evade macrophage innate immunity. All mutants were deficient in expression of flagellin, the primary flagellar subunit, and failed to induce caspase-1-mediated macrophage death. Interestingly, a previously isolated flagellar mutant (fliI) that expresses, but does not assemble, flagellin did not replicate in macrophages, and induced macrophage death. Thus, flagellin itself, not flagella or motility, is required to initiate macrophage innate immunity. Immunity to Legionella did not require MyD88, an essential adaptor for toll-like receptor 5 (TLR5) signaling. Moreover, flagellin of Legionella and Salmonella induced cytotoxicity when delivered to the macrophage cytosol using Escherichia coli as a heterologous host. It thus appears that macrophages sense cytosolic flagellin via a TLR5-independent pathway that leads to rapid caspase-1-dependent cell death and provides defense against intracellular bacterial pathogens.


Asunto(s)
Caspasa 1/fisiología , Flagelina/genética , Flagelina/metabolismo , Inmunidad/fisiología , Legionella/genética , Macrófagos/inmunología , Mutación , Proteína Inhibidora de la Apoptosis Neuronal/fisiología , Animales , Muerte Celular/fisiología , Citotoxicidad Inmunológica/fisiología , Genotipo , Legionella/crecimiento & desarrollo , Legionella/ultraestructura , Macrófagos/fisiología , Ratones , Ratones Noqueados , Salmonella/metabolismo , Receptor Toll-Like 5/fisiología
13.
Nat Genet ; 38(2): 240-4, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16429160

RESUMEN

The pathogenesis of Bacillus anthracis, the bacterium that causes anthrax, depends on secretion of three factors that combine to form two bipartite toxins. Edema toxin, consisting of protective antigen (PA) and edema factor (EF), causes the edema associated with cutaneous anthrax infections, whereas lethal toxin (LeTx), consisting of PA and lethal factor (LF), is believed to be responsible for causing death in systemic anthrax infections. EF and LF can be transported by PA into the cytosol of many cell types. In mouse macrophages, LF can cause rapid necrosis that may be related to the pathology of systemic infections. Inbred mouse strains display variable sensitivity to LeTx-induced macrophage necrosis. This trait difference has been mapped to a locus on chromosome 11 named Ltxs1 (refs. 7,8). Here we show that an extremely polymorphic gene in this locus, Nalp1b, is the primary mediator of mouse macrophage susceptibility to LeTx. We also show that LeTx-induced macrophage death requires caspase-1, which is activated in susceptible, but not resistant, macrophages after intoxication, suggesting that Nalp1b directly or indirectly activates caspase-1 in response to LeTx.


Asunto(s)
Antígenos Bacterianos/toxicidad , Proteínas Reguladoras de la Apoptosis/metabolismo , Toxinas Bacterianas/toxicidad , Macrófagos/efectos de los fármacos , Alelos , Animales , Proteínas Reguladoras de la Apoptosis/antagonistas & inhibidores , Secuencia de Bases , Caspasa 1/metabolismo , Supervivencia Celular , Susceptibilidad a Enfermedades , Exones/genética , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Polimorfismo Genético , ARN Mensajero/genética , ARN Mensajero/metabolismo
14.
Nat Immunol ; 7(3): 318-25, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16444259

RESUMEN

Baculovirus inhibitor of apoptosis repeat-containing 1 (Birc1) proteins have homology to several germline-encoded receptors of the innate immune system. However, their function in immune surveillance is not clear. Here we describe a Birc1e-dependent signaling pathway that restricted replication of the intracellular pathogen Legionella pneumophila in mouse macrophages. Translocation of bacterial products into host-cell cytosol was essential for Birc1e-mediated control of bacterial replication. Caspase-1 was required for Birc1e-dependent antibacterial responses ex vivo in macrophages and in a mouse model of Legionnaires' disease. The interleukin 1beta converting enzyme-protease-activating factor was necessary for L. pneumophila growth restriction, but interleukin 1beta was not required. These results establish Birc1e as a nucleotide-binding oligomerization-leucine-rich repeat protein involved in the detection and control of intracellular L. pneumophila.


Asunto(s)
Enfermedad de los Legionarios/inmunología , Proteína Inhibidora de la Apoptosis Neuronal/inmunología , Transducción de Señal/inmunología , Animales , Traslocación Bacteriana , Caspasa 1/inmunología , Caspasa 1/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Immunoblotting , Legionella pneumophila/fisiología , Macrófagos/inmunología , Macrófagos/microbiología , Ratones , Proteína Inhibidora de la Apoptosis Neuronal/metabolismo , Transfección
15.
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
16.
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
17.
Curr Biol ; 13(1): 27-36, 2003 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-12526741

RESUMEN

BACKGROUND: Legionella pneumophila is a gram-negative bacterial pathogen that is the cause of Legionnaires' Disease. Legionella produces disease because it can replicate inside a specialized compartment of host macrophages. Macrophages isolated from various inbred mice exhibit large differences in permissiveness for intracellular replication of Legionella. A locus affecting this host-resistance phenotype, Lgn1, has been mapped to chromosome 13, but the responsible gene has not been identified. RESULTS: Here, we report that Naip5 (also known as Birc1e) influences susceptibility to Legionella. Naip5 encodes a protein that is homologous to plant innate immunity (so-called "resistance") proteins and has been implicated in signaling pathways related to apoptosis regulation. Detailed recombination mapping and analysis of expression implicates Naip5 in the Legionella permissiveness differences among mouse strains. A bacterial artificial chromosome (BAC) transgenic line expressing a nonpermissive allele of Naip5 exhibits a reduction in macrophage Legionella permissiveness. In addition, morpholino-based antisense inhibition of Naip5 causes an increase in the Legionella permissiveness of macrophages. CONCLUSIONS: We conclude that polymorphisms in Naip5 are involved in the permissiveness differences of mouse macrophages for intracellular Legionella replication. We speculate that Naip5 is a functional mammalian homolog of plant "resistance" proteins that monitor for, and initiate host response to, the presence of secreted bacterial virulence proteins.


Asunto(s)
Predisposición Genética a la Enfermedad , Legionella pneumophila/patogenicidad , Enfermedad de los Legionarios/genética , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Animales , Elementos sin Sentido (Genética)/farmacología , Células Cultivadas , Mapeo Cromosómico , Cromosomas Artificiales Bacterianos , Regulación de la Expresión Génica , Enfermedad de los Legionarios/microbiología , Macrófagos/efectos de los fármacos , Macrófagos/microbiología , Ratones , Ratones Endogámicos , Ratones Transgénicos , Proteína Inhibidora de la Apoptosis Neuronal , Polimorfismo Genético , Transgenes
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...