Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
J Neurosci ; 42(12): 2598-2612, 2022 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-35121635

RESUMEN

Tuberous sclerosis complex (TSC) is caused by mutations in Tsc1 or Tsc2, whose gene products inhibit the small G-protein Rheb1. Rheb1 activates mTORC1, which may cause refractory epilepsy, intellectual disability, and autism. The mTORC1 inhibitors have been used for TSC patients with intractable epilepsy. However, its effectiveness for cognitive symptoms remains unclear. We found a new signaling pathway for synapse formation through Rheb1 activation, but not mTORC1. Here, we show that treatment with the farnesyltransferase inhibitor lonafarnib increased unfarnesylated (inactive) Rheb1 levels and restored synaptic abnormalities in cultured Tsc2+/- neurons, whereas rapamycin did not enhance spine synapse formation. Lonafarnib treatment also restored the plasticity-related Arc (activity-regulated cytoskeleton-associated protein) expression in cultured Tsc2+/- neurons. Lonafarnib action was partly dependent on the Rheb1 reduction with syntenin. Oral administration of lonafarnib increased unfarnesylated protein levels without affecting mTORC1 and MAP (mitogen-activated protein (MAP)) kinase signaling, and restored dendritic spine morphology in the hippocampi of male Tsc2+/- mice. In addition, lonafarnib treatment ameliorated contextual memory impairments and restored memory-related Arc expression in male Tsc2+/- mice in vivo Heterozygous Rheb1 knockout in male Tsc2+/- mice reproduced the results observed with pharmacological treatment. These results suggest that the Rheb1 activation may be responsible for synaptic abnormalities and memory impairments in Tsc2+/- mice, and its inhibition by lonafarnib could provide insight into potential treatment options for TSC-associated neuropsychiatric disorders.SIGNIFICANCE STATEMENT Tuberous sclerosis complex (TSC) is an autosomal-dominant disease that causes neuropsychiatric symptoms, including intractable epilepsy, intellectual disability (ID) and autism. No pharmacological treatment for ID has been reported so far. To develop a pharmacological treatment for ID, we investigated the mechanism of TSC and found that Rheb1 activation is responsible for synaptic abnormalities in TSC neurons. To inhibit Rheb1 function, we used the farnesyltransferase inhibitor lonafarnib, because farnesylation of Rheb1 is required for its activation. Lonafarnib treatment increased inactive Rheb1 and recovered proper synapse formation and plasticity-related Arc (activity-regulated cytoskeleton-associated protein) expression in TSC neurons. Furthermore, in vivo lonafarnib treatment restored contextual memory and Arc induction in TSC mice. Together, Rheb1 inhibition by lonafarnib could provide insight into potential treatments for TSC-associated ID.


Asunto(s)
Epilepsia Refractaria , Discapacidad Intelectual , Esclerosis Tuberosa , Animales , Cognición , Farnesiltransferasa , Humanos , Discapacidad Intelectual/tratamiento farmacológico , Discapacidad Intelectual/genética , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Esclerosis Tuberosa/genética
2.
Immunity ; 32(2): 200-13, 2010 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-20153219

RESUMEN

Neutrophils rapidly undergo polarization and directional movement to infiltrate the sites of infection and inflammation. Here, we show that an inhibitory MHC I receptor, Ly49Q, was crucial for the swift polarization of and tissue infiltration by neutrophils. During the steady state, Ly49Q inhibited neutrophil adhesion by preventing focal-complex formation, likely by inhibiting Src and PI3 kinases. However, in the presence of inflammatory stimuli, Ly49Q mediated rapid neutrophil polarization and tissue infiltration in an ITIM-domain-dependent manner. These opposite functions appeared to be mediated by distinct use of effector phosphatase SHP-1 and SHP-2. Ly49Q-dependent polarization and migration were affected by Ly49Q regulation of membrane raft functions. We propose that Ly49Q is pivotal in switching neutrophils to their polarized morphology and rapid migration upon inflammation, through its spatiotemporal regulation of membrane rafts and raft-associated signaling molecules.


Asunto(s)
Microdominios de Membrana/metabolismo , Subfamilia A de Receptores Similares a Lectina de Células NK/metabolismo , Neutrófilos/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Animales , Movimiento Celular/genética , Movimiento Celular/inmunología , Polaridad Celular/genética , Polaridad Celular/inmunología , Células Cultivadas , Adhesiones Focales/genética , Adhesiones Focales/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Subfamilia A de Receptores Similares a Lectina de Células NK/genética , Subfamilia A de Receptores Similares a Lectina de Células NK/inmunología , Activación Neutrófila , Neutrófilos/inmunología , Neutrófilos/patología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Transporte de Proteínas , Proteína Tirosina Fosfatasa no Receptora Tipo 11/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 6/inmunología , Familia-src Quinasas/antagonistas & inhibidores
3.
Hum Mol Genet ; 25(10): 2045-2059, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-26936824

RESUMEN

Most clinical reports have suggested that patients with congenital profound hearing loss have recessive mutations in deafness genes, whereas dominant alleles are associated with progressive hearing loss (PHL). Jackson shaker (Ush1gjs) is a mouse model of recessive deafness that exhibits congenital profound deafness caused by the homozygous mutation of Ush1g/Sans on chromosome 11. We found that C57BL/6J-Ush1gjs/+ heterozygous mice exhibited early-onset PHL (ePHL) accompanied by progressive degeneration of stereocilia in the cochlear outer hair cells. Interestingly, ePHL did not develop in mutant mice with the C3H/HeN background, thus suggesting that other genetic factors are required for ePHL development. Therefore, we performed classical genetic analyses and found that the occurrence of ePHL in Ush1gjs/+ mice was associated with an interval in chromosome 10 that contains the cadherin 23 gene (Cdh23), which is also responsible for human deafness. To confirm this mutation effect, we generated C57BL/6J-Ush1gjs/+, Cdh23c.753A/G double-heterozygous mice by using the CRISPR/Cas9-mediated Cdh23c.753A>G knock-in method. The Cdh23c.753A/G mice harbored a one-base substitution (A for G), and the homozygous A allele caused moderate hearing loss with aging. Analyses revealed the complete recovery of ePHL and stereocilia degeneration in C57BL/6J-Ush1gjs/+ mice. These results clearly show that the development of ePHL requires at least two mutant alleles of the Ush1g and Cdh23 genes. Our results also suggest that because the SANS and CDH23 proteins form a complex in the stereocilia, the interaction between these proteins may play key roles in the maintenance of stereocilia and the prevention of ePHL.


Asunto(s)
Cadherinas/genética , Pérdida Auditiva/genética , Mutación/genética , Proteínas del Tejido Nervioso/genética , Alelos , Secuencia de Aminoácidos/genética , Animales , Cromosomas Humanos Par 10/genética , Modelos Animales de Enfermedad , Células Ciliadas Auditivas Externas/patología , Pérdida Auditiva/patología , Heterocigoto , Homocigoto , Humanos , Ratones , Estereocilios/patología
4.
J Biol Chem ; 291(30): 15588-601, 2016 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-27226632

RESUMEN

Phospholipase A2 enzymes have long been implicated in the promotion of inflammation by mobilizing pro-inflammatory lipid mediators, yet recent evidence suggests that they also contribute to anti-inflammatory or pro-resolving programs. Group IID-secreted phospholipase A2 (sPLA2-IID) is abundantly expressed in dendritic cells in lymphoid tissues and resolves the Th1 immune response by controlling the steady-state levels of anti-inflammatory lipids such as docosahexaenoic acid and its metabolites. Here, we show that psoriasis and contact dermatitis were exacerbated in Pla2g2d-null mice, whereas they were ameliorated in Pla2g2d-overexpressing transgenic mice, relative to littermate wild-type mice. These phenotypes were associated with concomitant alterations in the tissue levels of ω3 polyunsaturated fatty acid (PUFA) metabolites, which had the capacity to reduce the expression of pro-inflammatory and Th1/Th17-type cytokines in dendritic cells or lymph node cells. In the context of cancer, however, Pla2g2d deficiency resulted in marked attenuation of skin carcinogenesis, likely because of the augmented anti-tumor immunity. Altogether, these results underscore a general role of sPLA2-IID as an immunosuppressive sPLA2 that allows the microenvironmental lipid balance toward an anti-inflammatory state, exerting beneficial or detrimental impact depending upon distinct pathophysiological contexts in inflammation and cancer.


Asunto(s)
Fosfolipasas A2 Grupo II/inmunología , Inmunidad Celular , Proteínas de Neoplasias/inmunología , Neoplasias Cutáneas/inmunología , Células TH1/inmunología , Células Th17/inmunología , Animales , Ácidos Grasos Omega-3/genética , Ácidos Grasos Omega-3/inmunología , Fosfolipasas A2 Grupo II/genética , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Ratones , Ratones Noqueados , Proteínas de Neoplasias/genética , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Células TH1/patología , Células Th17/patología
5.
Transgenic Res ; 26(4): 559-565, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28220277

RESUMEN

Mitochondrial DNA segregation is one of the characteristic modes of mitochondrial inheritance in which the heteroplasmic state of mitochondrial DNA is transmitted to the next generation in variable proportions. To analyze mitochondrial DNA segregation, we produced a heteroplasmic mouse strain with interspecific mitochondrial DNA haplotypes, which contains two types of mitochondrial DNA derived from C57BL/6J and Mus spretus strains. The strain was produced on a C57BL/6J nuclear genomic background by microinjection of donor cytoplasm into fertilized eggs. The PCR-RFLP semi-quantitative analysis method, which was improved to reduce the effect of heteroduplex formation, was used to measure the proportion of heteroplasmic mitochondrial DNA in tissues. Founder mice contained up to approximately 14% of exogenous Mus spretus mitochondrial DNA molecules in their tails, and the detected proportions differed among tissues of the same individual. Heteroplasmic mitochondrial DNA is transmitted to the next generation in varying proportions under the maternal inheritance mode. This mitochondrial heteroplasmic mouse strain and the improved PCR-RFLP measurement system enable analysis of the transmission of heteroplasmic mitochondrial DNA variants between tissues and generations.


Asunto(s)
ADN Mitocondrial/genética , Haplotipos/genética , Polimorfismo de Longitud del Fragmento de Restricción/genética , Animales , Femenino , Ratones , Microinyecciones , Reacción en Cadena de la Polimerasa , Cigoto/crecimiento & desarrollo
6.
J Immunol ; 195(3): 982-93, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26136427

RESUMEN

Many members of the BTB-ZF family have been shown to play important roles in lymphocyte development and function. The role of zinc finger Znf131 (also known as Zbtb35) in T cell lineage was elucidated through the production of mice with floxed allele to disrupt at different stages of development. In this article, we present that Znf131 is critical for T cell development during double-negative to double-positive stage, with which significant cell expansion triggered by the pre-TCR signal is coupled. In mature T cells, Znf131 is required for the activation of effector genes, as well as robust proliferation induced upon TCR signal. One of the cyclin-dependent kinase inhibitors, p21(Cip1) encoded by cdkn1a gene, is one of the targets of Znf131. The regulation of T cell proliferation by Znf131 is in part attributed to its suppression on the expression of p21(Cip1).


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Proteínas de Unión al ADN/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Linfocitos T/inmunología , Factores de Transcripción/inmunología , Células 3T3 , Animales , Diferenciación Celular/inmunología , Línea Celular , Proliferación Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/biosíntesis , Proteínas de Unión al ADN/genética , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Nucleares/inmunología , Regiones Promotoras Genéticas/genética , Proteínas Inhibidoras de STAT Activados/inmunología , Proteínas Proto-Oncogénicas c-myb/genética , Proteínas Proto-Oncogénicas c-myb/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Linfocitos T/citología , Factores de Transcripción/genética , Ubiquitina-Proteína Ligasas
7.
Proc Natl Acad Sci U S A ; 110(36): 14753-8, 2013 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-23959904

RESUMEN

Enterovirus 71 (EV71) typically causes mild hand-foot-and-mouth disease in children, but it can also cause severe neurological disease. Recently, epidemic outbreaks of EV71 with significant mortality have been reported in the Asia-Pacific region, and EV71 infection has become a serious public health concern worldwide. However, there is little information available concerning EV71 neuropathogenesis, and no vaccines or anti-EV71 drugs have been developed. Previous studies of this disease have used monkeys and neonatal mice that are susceptible to some EV71 strains as models. The monkey model is problematic for ethical and economical reasons, and mice that are more than a few weeks old lose their susceptibility to EV71. Thus, the development of an appropriate small animal model would greatly contribute to the study of this disease. Mice lack EV71 susceptibility due to the absence of a receptor for this virus. Previously, we identified the human scavenger receptor class B, member 2 (hSCARB2) as a cellular receptor for EV71. In the current study, we generated a transgenic (Tg) mouse expressing hSCARB2 with an expression profile similar to that in humans. Tg mice infected with EV71 exhibited ataxia, paralysis, and death. The most severely affected cells were neurons in the spinal cord, brainstem, cerebellum, hypothalamus, thalamus, and cerebrum. The pathological features in these Tg mice were generally similar to those of EV71 encephalomyelitis in humans and experimentally infected monkeys. These results suggest that this Tg mouse could represent a useful animal model for the study of EV71 infection.


Asunto(s)
Enfermedades del Sistema Nervioso Central/genética , Modelos Animales de Enfermedad , Infecciones por Enterovirus/genética , Proteínas de Membrana de los Lisosomas/genética , Receptores Depuradores/genética , Animales , Western Blotting , Encéfalo/metabolismo , Encéfalo/patología , Encéfalo/virología , Línea Celular Tumoral , Enfermedades del Sistema Nervioso Central/metabolismo , Enfermedades del Sistema Nervioso Central/virología , Chlorocebus aethiops , Enterovirus Humano A/fisiología , Infecciones por Enterovirus/metabolismo , Infecciones por Enterovirus/virología , Interacciones Huésped-Patógeno , Humanos , Inmunohistoquímica , Proteínas de Membrana de los Lisosomas/metabolismo , Ratones , Ratones Transgénicos , Receptores Depuradores/metabolismo , Médula Espinal/metabolismo , Médula Espinal/patología , Médula Espinal/virología , Factores de Tiempo , Células Vero
8.
Nat Cell Biol ; 9(1): 36-44, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17173042

RESUMEN

Proper neutrophil migration into inflammatory sites ensures host defense without tissue damage. Phosphoinositide 3-kinase (PI(3)K) and its lipid product phosphatidylinositol 3,4,5-trisphosphate (PtdIns(3,4,5)P(3)) regulate cell migration, but the role of PtdIns(3,4,5)P(3)-degrading enzymes in this process is poorly understood. Here, we show that Src homology 2 (SH2) domain-containing inositol-5-phosphatase 1 (SHIP1), a PtdIns(3,4,5)P(3) phosphatase, is a key regulator of neutrophil migration. Genetic inactivation of SHIP1 led to severe defects in neutrophil polarization and motility. In contrast, loss of the PtdIns(3,4,5)P(3) phosphatase PTEN had no impact on neutrophil chemotaxis. To study PtdIns(3,4,5)P(3) metabolism in living primary cells, we generated a novel transgenic mouse (AktPH-GFP Tg) expressing a bioprobe for PtdIns(3,4,5)P(3.) Time-lapse footage showed rapid, localized binding of AktPH-GFP to the leading edge membrane of chemotaxing ship1(+/+)AktPH-GFP Tg neutrophils, but only diffuse localization in ship1(-/-)AktPH-GFP Tg neutrophils. By directing where PtdIns(3,4,5)P(3) accumulates, SHIP1 governs the formation of the leading edge and polarization required for chemotaxis.


Asunto(s)
Movimiento Celular , Polaridad Celular , Quimiotaxis , Fosfatos de Fosfatidilinositol/metabolismo , Monoéster Fosfórico Hidrolasas/fisiología , Animales , Células Cultivadas , Fosfatidilinositol 3-Quinasa Clase Ib , Humanos , Inositol Polifosfato 5-Fosfatasas , Isoenzimas/metabolismo , Isoenzimas/fisiología , Macrófagos/fisiología , Ratones , Ratones Transgénicos , Modelos Biológicos , Neutrófilos/fisiología , Proteína Oncogénica v-akt/genética , Proteína Oncogénica v-akt/metabolismo , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatidilinositol 3-Quinasas/fisiología , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas , Monoéster Fosfórico Hidrolasas/genética , Monoéster Fosfórico Hidrolasas/metabolismo
9.
Biochem Biophys Res Commun ; 436(3): 400-5, 2013 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-23747725

RESUMEN

By using the toxin receptor-mediated cell knockout (TRECK) method, we have generated two transgenic (Tg) murine lines that model type 1 (insulin-dependent) diabetes. The first strain, C.B-17/Icr-Prkdc(scid)/Prkdc(scid)-INS-TRECK-Tg, carries the diphtheria toxin receptor (hDTR) driven by the human insulin gene promoter, while the other strain, C57BL/6-ins2(BAC)-TRECK-Tg, expresses hDTR cDNA under the control of the mouse insulin II gene promoter. With regard to the C.B-17/Icr-Prkdc(scid)/Prkdc(scid)-INS-TRECK-Tg strain, only one of three Tg strains exhibited proper expression of hDTR in pancreatic ß cells. By contrast, hDTR was expressed in the pancreatic ß cells of all four of the generated C57BL/6-ins2(BAC)-TRECK-Tg strains. Hyperglycemia, severe ablation of pancreatic ß cells and depletion of serum insulin were observed within 3days after the administration of diphtheria toxin (DT) in these Tg mice. Subcutaneous injection of a suitable dosage of insulin was sufficient for recovery from hyperglycemia in all of the examined strains. Using the C.B-17/Icr-Prkdc(scid)/Prkdc(scid)-INS-TRECK-Tg model, we tried to perform regenerative therapeutic approaches: allogeneic transplantation of pancreatic islet cells from C57BL/6 and xenogeneic transplantation of CD34(+) human umbilical cord blood cells. Both approaches successfully rescued C.B-17/Icr-Prkdc(scid)/Prkdc(scid)-INS-TRECK-Tg mice from hyperglycemia caused by DT administration. The high specificity with which DT causes depletion in pancreatic ß cells of these Tg mice is highly useful for diabetogenic research.


Asunto(s)
Diabetes Mellitus Tipo 1/patología , Toxina Diftérica/efectos adversos , Células Secretoras de Insulina/metabolismo , Receptores de Superficie Celular/antagonistas & inhibidores , Animales , Antígenos CD34/metabolismo , Trasplante de Células Madre de Sangre del Cordón Umbilical , Diabetes Mellitus Experimental/patología , Toxina Diftérica/metabolismo , Técnicas de Inactivación de Genes , Glucosa/farmacología , Humanos , Hiperglucemia/patología , Hiperglucemia/terapia , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/patología , Insulinas/administración & dosificación , Insulinas/sangre , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Ratones Transgénicos , Receptores de Superficie Celular/metabolismo , Trasplante Heterólogo , Trasplante Homólogo
10.
Proc Natl Acad Sci U S A ; 107(16): 7586-91, 2010 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-20368433

RESUMEN

Atypical Rho-guanine nucleotide exchange factors (Rho-GEFs) that contain Dock homology regions (DHR-1 and DHR-2) are expressed in a variety of tissues; however, their functions and mechanisms of action remain unclear. We identify key conserved amino acids in the DHR-2 domain that are critical for the catalytic activity of Dock-GEFs (Dock1-4). We further demonstrate that Dock-GEFs directly associate with WASP family verprolin-homologous (WAVE) proteins through the DHR-1 domain. Brain-derived neurotrophic factor (BDNF)-TrkB signaling recruits the Dock3/WAVE1 complex to the plasma membrane, whereupon Dock3 activates Rac and dissociates from the WAVE complex in a phosphorylation-dependent manner. BDNF induces axonal sprouting through Dock-dependent Rac activation, and adult transgenic mice overexpressing Dock3 exhibit enhanced optic nerve regeneration after injury without affecting WAVE expression levels. Our results highlight a unique mechanism through which Dock-GEFs achieve spatial and temporal restriction of WAVE signaling, and identify Dock-GEF activity as a potential therapeutic target for axonal regeneration.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/metabolismo , Factores de Intercambio de Guanina Nucleótido/fisiología , Proteínas del Tejido Nervioso/fisiología , Familia de Proteínas del Síndrome de Wiskott-Aldrich/metabolismo , Secuencia de Aminoácidos , Animales , Axones/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Células COS , Chlorocebus aethiops , Hipocampo/metabolismo , Humanos , Ratones , Datos de Secuencia Molecular , Regeneración Nerviosa , Neuronas/metabolismo , Nervio Óptico/metabolismo , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho , Homología de Secuencia de Aminoácido , Transducción de Señal
11.
Transgenic Res ; 21(1): 51-62, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21431867

RESUMEN

The proximal straight tubule (S3 segment) of the kidney is highly susceptible to ischemia and toxic insults but has a remarkable capacity to repair its structure and function. In response to such injuries, complex processes take place to regenerate the epithelial cells of the S3 segment; however, the precise molecular mechanisms of this regeneration are still being investigated. By applying the "toxin receptor mediated cell knockout" method under the control of the S3 segment-specific promoter/enhancer, Gsl5, which drives core 2 ß-1,6-N-acetylglucosaminyltransferase gene expression, we established a transgenic mouse line expressing the human diphtheria toxin (DT) receptor only in the S3 segment. The administration of DT to these transgenic mice caused the selective ablation of S3 segment cells in a dose-dependent manner, and transgenic mice exhibited polyuria containing serum albumin and subsequently developed oliguria. An increase in the concentration of blood urea nitrogen was also observed, and the peak BUN levels occurred 3-7 days after DT administration. Histological analysis revealed that the most severe injury occurred in the S3 segments of the proximal tubule, in which tubular cells were exfoliated into the tubular lumen. In addition, aquaporin 7, which is localized exclusively to the S3 segment, was diminished. These results indicate that this transgenic mouse can suffer acute kidney injury (AKI) caused by S3 segment-specific damage after DT administration. This transgenic line offers an excellent model to uncover the mechanisms of AKI and its rapid recovery.


Asunto(s)
Lesión Renal Aguda/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Túbulos Renales Proximales/patología , Lesión Renal Aguda/patología , Secuencia de Aminoácidos , Animales , Toxina Diftérica/toxicidad , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Células Epiteliales/patología , Factor de Crecimiento Similar a EGF de Unión a Heparina , Humanos , Túbulos Renales Proximales/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Datos de Secuencia Molecular , N-Acetilglucosaminiltransferasas/genética , Regiones Promotoras Genéticas , Secuencias Reguladoras de Ácidos Nucleicos
12.
J Exp Med ; 201(6): 859-70, 2005 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-15767368

RESUMEN

The membrane phospholipid phosphatidylinositol 4, 5-bisphosphate [PI(4,5)P(2)] is a critical signal transducer in eukaryotic cells. However, the physiological roles of the type I phosphatidylinositol phosphate kinases (PIPKIs) that synthesize PI(4,5)P(2) are largely unknown. Here, we show that the alpha isozyme of PIPKI (PIPKIalpha) negatively regulates mast cell functions and anaphylactic responses. In vitro, PIPKIalpha-deficient mast cells exhibited increased degranulation and cytokine production after Fcepsilon receptor-I cross-linking. In vivo, PIPKIalpha(-/-) mice displayed enhanced passive cutaneous and systemic anaphylaxis. Filamentous actin was diminished in PIPKIalpha(-/-) mast cells, and enhanced degranulation observed in the absence of PIPKIalpha was also seen in wild-type mast cells treated with latrunculin, a pharmacological inhibitor of actin polymerization. Moreover, the association of FcepsilonRI with lipid rafts and FcepsilonRI-mediated activation of signaling proteins was augmented in PIPKIalpha(-/-) mast cells. Thus, PIPKIalpha is a negative regulator of FcepsilonRI-mediated cellular responses and anaphylaxis, which functions by controlling the actin cytoskeleton and dynamics of FcepsilonRI signaling. Our results indicate that the different PIPKI isoforms might be functionally specialized.


Asunto(s)
Anafilaxia/metabolismo , Señalización del Calcio/fisiología , Degranulación de la Célula/fisiología , Mastocitos/fisiología , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Actinas/metabolismo , Anafilaxia/genética , Animales , Señalización del Calcio/genética , Degranulación de la Célula/genética , Células Cultivadas , Isoenzimas/genética , Isoenzimas/metabolismo , Microdominios de Membrana/metabolismo , Ratones , Ratones Noqueados , Antígenos de Histocompatibilidad Menor , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Receptores de IgE/metabolismo , Tiazoles/farmacología
13.
Nature ; 434(7036): 1035-40, 2005 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-15815647

RESUMEN

Robust type-I interferon (IFN-alpha/beta) induction in plasmacytoid dendritic cells, through the activation of Toll-like receptor 9 (TLR9), constitutes a critical aspect of immunity. It is absolutely dependent on the transcription factor IRF-7, which interacts with and is activated by the adaptor MyD88. How plasmacytoid dendritic cells, but not other cell types (such as conventional dendritic cells), are able to activate the MyD88-IRF-7-dependent IFN induction pathway remains unknown. Here we show that the spatiotemporal regulation of MyD88-IRF-7 signalling is critical for a high-level IFN induction in response to TLR9 activation. The IFN-inducing TLR9 ligand, A/D-type CpG oligodeoxynucleotide (CpG-A), is retained for long periods in the endosomal vesicles of plasmacytoid dendritic cells, together with the MyD88-IRF-7 complex. However, in conventional dendritic cells, CpG-A is rapidly transferred to lysosomal vesicles. We further show that conventional dendritic cells can also mount a robust IFN induction if CpG-A is manipulated for endosomal retention using a cationic lipid. This strategy also allows us to demonstrate endosomal activation of the IFN pathway by the otherwise inactive TLR9 ligand B/K-type oligodeoxynucleotide (CpG-B). Thus, our study offers insights into the regulation of TLR9 signalling in space, potentially suggesting a new avenue for therapeutic intervention.


Asunto(s)
Antígenos de Diferenciación/metabolismo , Proteínas de Unión al ADN/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Interferón Tipo I/inmunología , Receptores Inmunológicos/metabolismo , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales , Animales , Antígenos de Diferenciación/genética , Transporte Biológico , Células de la Médula Ósea/citología , Línea Celular , Islas de CpG/genética , Proteínas de Unión al ADN/genética , Células Dendríticas/clasificación , Células Dendríticas/citología , Endosomas/metabolismo , Factor 7 Regulador del Interferón , Interferón Tipo I/metabolismo , Macrófagos/citología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Factor 88 de Diferenciación Mieloide , Receptores de Superficie Celular/metabolismo , Receptores Inmunológicos/genética , Factores de Tiempo , Receptor Toll-Like 9
14.
Proc Natl Acad Sci U S A ; 105(51): 20446-51, 2008 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-19074283

RESUMEN

A conundrum of innate antiviral immunity is how nucleic acid-sensing Toll-like receptors (TLRs) and RIG-I/MDA5 receptors cooperate during virus infection. The conventional wisdom has been that the activation of these receptor pathways evokes type I IFN (IFN) responses. Here, we provide evidence for a critical role of a Toll-like receptor 3 (TLR3)-dependent type II IFN signaling pathway in antiviral innate immune response against Coxsackievirus group B serotype 3 (CVB3), a member of the positive-stranded RNA virus family picornaviridae and most prevalent virus associated with chronic dilated cardiomyopathy. TLR3-deficient mice show a vulnerability to CVB3, accompanied by acute myocarditis, whereas transgenic expression of TLR3 endows even type I IFN signal-deficient mice resistance to CVB3 and other types of viruses, provided that type II IFN signaling remains intact. Taken together, our results indicate a critical cooperation of the RIG-I/MDA5-type I IFN and the TLR3-type II IFN signaling axes for efficient innate antiviral immune responses.


Asunto(s)
Inmunidad Innata , Interferón gamma/inmunología , Transducción de Señal/inmunología , Receptor Toll-Like 3/inmunología , Virosis/inmunología , Animales , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/metabolismo , Enterovirus/inmunología , Interferón Tipo I/inmunología , Interferón Tipo I/metabolismo , Helicasa Inducida por Interferón IFIH1 , Interferón gamma/metabolismo , Ratones , Ratones Noqueados , Miocarditis/inmunología , Miocarditis/virología , Receptor Toll-Like 3/deficiencia , Receptor Toll-Like 3/metabolismo
15.
Cells ; 10(7)2021 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-34359862

RESUMEN

Tissue-resident mast cells (MCs) have important roles in IgE-associated and -independent allergic reactions. Although microenvironmental alterations in MC phenotypes affect the susceptibility to allergy, understanding of the regulation of MC maturation is still incomplete. We previously reported that group III secreted phospholipase A2 (sPLA2-III) released from immature MCs is functionally coupled with lipocalin-type prostaglandin D2 (PGD2) synthase in neighboring fibroblasts to supply a microenvironmental pool of PGD2, which in turn acts on the PGD2 receptor DP1 on MCs to promote their proper maturation. In the present study, we reevaluated the role of sPLA2-III in MCs using a newly generated MC-specific Pla2g3-deficient mouse strain. Mice lacking sPLA2-III specifically in MCs, like those lacking the enzyme in all tissues, had immature MCs and displayed reduced local and systemic anaphylactic responses. Furthermore, MC-specific Pla2g3-deficient mice, as well as MC-deficient KitW-sh mice reconstituted with MCs prepared from global Pla2g3-null mice, displayed a significant reduction in irritant contact dermatitis (ICD) and an aggravation of contact hypersensitivity (CHS). The increased CHS response by Pla2g3 deficiency depended at least partly on the reduced expression of hematopoietic PGD2 synthase and thereby reduced production of PGD2 due to immaturity of MCs. Overall, our present study has confirmed that MC-secreted sPLA2-III promotes MC maturation, thereby facilitating acute anaphylactic and ICD reactions and limiting delayed CHS response.


Asunto(s)
Diferenciación Celular , Eliminación de Gen , Mastocitos/enzimología , Mastocitos/patología , Fosfolipasas A2 Secretoras/metabolismo , Anafilaxia/patología , Animales , Dermatitis/patología , Dermatitis por Contacto/patología , Fibroblastos/patología , Ratones Endogámicos C57BL , Fosfolipasas A2 Secretoras/deficiencia
16.
Dev Biol ; 330(2): 427-39, 2009 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-19371732

RESUMEN

During mouse gastrulation, primordial germ cells (PGCs) become clustered at the base of the allantois and move caudally into the hindgut endoderm before entering the genital ridges. The precise roles of endoderm tissues in PGC migration, however, remain unclear. By using Sox17 mutants with a specific endoderm deficiency, we provide direct evidence for the crucial role of hindgut expansion in directing proper PGC migration. In Sox17-null embryos, PGCs normally colonize in the allantois and then a small front-row population of PGCs moves properly into the most posterior gut endoderm. Defective hindgut expansion, however, causes the failure of further lateral PGC movement, resulting in the immobilization of PGCs in the hindgut entrance at the later stages. In contrast, the majority of the remaining PGCs moves into the visceral endoderm layer, but relocate outside of the embryonic gut domain. This leads to a scattering of PGCs in the extraembryonic yolk sac endoderm. This aberrant migration of Sox17-null PGCs can be rescued by the supply of wildtype hindgut cells in chimeric embryos. Therefore, these data indicate that hindgut morphogenic movement is crucial for directing PGC movement toward the embryonic gut side, but not for their relocation from the mesoderm into the endoderm.


Asunto(s)
Movimiento Celular , Embrión de Mamíferos/citología , Desarrollo Embrionario , Células Germinativas/citología , Animales , Metilación de ADN , Epigénesis Genética , Proteínas HMGB/genética , Inmunohistoquímica , Hibridación in Situ , Ratones , Ratones Endogámicos ICR , Microscopía Electrónica de Transmisión , Factores de Transcripción SOXF/genética
17.
Biochem Biophys Res Commun ; 391(1): 357-63, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19913509

RESUMEN

In early-organogenesis-stage mouse embryos, the posteroventral foregut endoderm adjacent to the heart tube gives rise to liver, ventral pancreas and gallbladder. Hepatic and pancreatic primordia become specified in the posterior segment of the ventral foregut endoderm at early somite stages. The mechanisms for demarcating gallbladder and bile duct primordium, however, are poorly understood. Here, we demonstrate that the gallbladder and bile duct progenitors are specified in the paired lateral endoderm domains outside the heart field at almost the same timing as hepatic and pancreatic induction. In the anterior definitive endoderm, Sox17 reactivation occurs in a certain population within the most lateral domains posterolateral to the anterior intestinal portal (AIP) lip on both the left and right sides. During foregut formation, the paired Sox17-positive domains expand ventromedially to merge in the midline of the AIP lip and become localized between the liver and pancreatic primordia. In Sox17-null embryos, these lateral domains are missing, resulting in a complete loss of the gallbladder/bile-duct structure. Chimera analyses revealed that Sox17-null endoderm cells in the posteroventral foregut do not display any gallbladder/bile-duct molecular characters. Our findings show that Sox17 functions cell-autonomously to specify gallbladder/bile-duct in the mouse embryo.


Asunto(s)
Conductos Biliares/embriología , Vesícula Biliar/embriología , Proteínas HMGB/fisiología , Intestinos/embriología , Morfogénesis , Factores de Transcripción SOXF/fisiología , Animales , Conductos Biliares/anomalías , Conductos Biliares/metabolismo , Tipificación del Cuerpo , Embrión de Mamíferos/metabolismo , Endodermo/metabolismo , Femenino , Vesícula Biliar/anomalías , Vesícula Biliar/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas HMGB/genética , Mucosa Intestinal/metabolismo , Ratones , Ratones Endogámicos C57BL , Factores de Transcripción SOXF/genética
18.
Mol Cell Biol ; 26(16): 6149-56, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16880525

RESUMEN

The mammalian small GTPase ADP-ribosylation factor 6 (ARF6) plays important roles in a wide variety of cellular events, including endocytosis, actin cytoskeletal reorganization, and phosphoinositide metabolism. However, physiological functions for ARF6 have not previously been examined. Here, we described the consequence of ARF6 ablation in mice, which manifests most obviously in the context of liver development. Livers from ARF6-/- embryos are smaller and exhibit hypocellularity, due to the onset of midgestational liver cell apoptosis. Preceding the apoptosis, however, defective hepatic cord formation is observed; the liver cells migrate abnormally upon exiting the primordial hepatic epithelial sheet and clump rather than becoming dispersed. Consistent with this observation, the ability of hepatocyte growth factor/scatter factor (HGF) to induce hepatic cord-like structures from ARF6-/- fetal hepatocytes cultured in vitro in collagen gel matrix is impaired. Finally, we show that endogenous ARF6 in wild-type fetal hepatocytes is activated in response to HGF stimulation. These results provide evidence that ARF6 is an essential component in the signaling pathway coupling HGF signaling to hepatic cord formation.


Asunto(s)
Factores de Ribosilacion-ADP/metabolismo , Hígado/embriología , Proteínas de Unión al GTP Monoméricas/metabolismo , Factor 6 de Ribosilación del ADP , Factores de Ribosilacion-ADP/deficiencia , Animales , Apoptosis , Células Cultivadas , Colágeno/metabolismo , Embrión de Mamíferos/anomalías , Embrión de Mamíferos/citología , Embrión de Mamíferos/embriología , Exones/genética , Geles , Marcación de Gen , Genotipo , Factor de Crecimiento de Hepatocito/farmacología , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Hepatocitos/patología , Hígado/anomalías , Hígado/citología , Hígado/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
19.
Sci Rep ; 9(1): 5285, 2019 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-30918314

RESUMEN

Outer hair cells (OHCs) are responsible for the amplification of sound, and the death of these cells leads to hearing loss. Although the mechanisms for sound amplification and OHC death have been well investigated, the effects on the cochlea after OHC death are poorly understood. To study the consequences of OHC death, we established an OHC knockout system using a novel mouse model, Prestin-hDTR, which uses the prestin promoter to express the human diphtheria toxin (DT) receptor gene (hDTR). Administration of DT to adult Prestin-hDTR mice results in the depletion of almost all OHCs without significant damage to other cochlear and vestibular cells, suggesting that this system is an effective tool for the analysis of how other cells in the cochlea and vestibula are affected after OHC death. To evaluate the changes in the cochlea after OHC death, we performed differential gene expression analysis between the untreated and DT-treated groups of wild-type and Prestin-hDTR mice. This analysis revealed that genes associated with inflammatory/immune responses were significantly upregulated. Moreover, we found that several genes linked to hearing loss were strongly downregulated by OHC death. Together, these results suggest that this OHC knockout system is a useful tool to identify biomarkers associated with OHC death.


Asunto(s)
Cóclea/metabolismo , Células Ciliadas Auditivas Externas/metabolismo , Pérdida Auditiva/metabolismo , Animales , Toxina Diftérica/metabolismo , Modelos Animales de Enfermedad , Inmunohistoquímica , Ratones Endogámicos C57BL , Proteínas Motoras Moleculares/metabolismo
20.
Sci Rep ; 7(1): 11208, 2017 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-28894263

RESUMEN

Using a forward genetics approach to map loci in a mouse skin cancer model, we previously identified a genetic locus, Skin tumour modifier of MSM 1 (Stmm1) on chromosome 7, conferring strong tumour resistance. Sub-congenic mapping localized Parathyroid hormone (Pth) in Stmm1b. Here, we report that serum intact-PTH (iPTH) and a genetic polymorphism in Pth are important for skin tumour resistance. We identified higher iPTH levels in sera from cancer-resistant MSM/Ms mice compared with susceptible FVB/NJ mice. Therefore, we performed skin carcinogenesis experiments with MSM-BAC transgenic mice (Pth MSM-Tg) and Pth knockout heterozygous mice (Pth +/-). As a result, the higher amounts of iPTH in sera conferred stronger resistance to skin tumours. Furthermore, we found that the coding SNP (rs51104087, Val28Met) localizes in the mouse Pro-PTH encoding region, which is linked to processing efficacy and increased PTH secretion. Finally, we report that PTH increases intracellular calcium in keratinocytes and promotes their terminal differentiation. Taken together, our data suggest that Pth is one of the genes responsible for Stmm1, and serum iPTH could serve as a prevention marker of skin cancer and a target for new therapies.


Asunto(s)
Hormonas y Agentes Reguladores de Calcio/genética , Hormonas y Agentes Reguladores de Calcio/metabolismo , Predisposición Genética a la Enfermedad , Hormona Paratiroidea/genética , Hormona Paratiroidea/metabolismo , Neoplasias Cutáneas/epidemiología , Neoplasias Cutáneas/genética , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Noqueados , Ratones Transgénicos , Polimorfismo de Nucleótido Simple
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA