Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-38464227

RESUMEN

Selective and controlled expansion of endogenous ß-cells has been pursued as a potential therapy for diabetes. Ideally, such therapies would preserve feedback control of ß-cell proliferation to avoid excessive ß-cell expansion and an increased risk of hypoglycemia. Here, we identified a regulator of ß-cell proliferation whose inactivation results in controlled ß-cell expansion: the protein deacetylase Sirtuin 2 (SIRT2). Sirt2 deletion in ß-cells of mice increased ß-cell proliferation during hyperglycemia with little effect in homeostatic conditions, indicating preservation of feedback control of ß-cell mass. SIRT2 restrains proliferation of human islet ß-cells cultured in glucose concentrations above the glycemic set point, demonstrating conserved SIRT2 function. Analysis of acetylated proteins in islets treated with a SIRT2 inhibitor revealed that SIRT2 deacetylates enzymes involved in oxidative phosphorylation, dampening the adaptive increase in oxygen consumption during hyperglycemia. At the transcriptomic level, Sirt2 inactivation has context-dependent effects on ß-cells, with Sirt2 controlling how ß-cells interpret hyperglycemia as a stress. Finally, we provide proof-of-principle that systemic administration of a GLP1-coupled Sirt2-targeting antisense oligonucleotide achieves ß-cell selective Sirt2 inactivation and stimulates ß-cell proliferation under hyperglycemic conditions. Overall, these studies identify a therapeutic strategy for increasing ß-cell mass in diabetes without circumventing feedback control of ß-cell proliferation.

2.
Front Mol Biosci ; 10: 1096286, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36814640

RESUMEN

Insulin resistance is the major risk factor for Type 2 diabetes (T2D). In vulnerable individuals, insulin resistance induces a progressive loss of insulin secretion with islet pathology revealing a partial deficit of beta cells and islet amyloid derived from islet amyloid polypeptide (IAPP). IAPP is co-expressed and secreted with insulin by beta cells, expression of both proteins being upregulated in response to insulin resistance. If IAPP expression exceeds the threshold for clearance of misfolded proteins, beta cell failure occurs exacerbated by the action of IAPP toxicity to compromise the autophagy lysosomal pathway. We postulated that suppression of IAPP expression by an IAPP antisense oligonucleotide delivered to beta cells by the GLP-1 agonist exenatide (eGLP1-IAPP-ASO) is a potential disease modifying therapy for T2D. While eGLP1-IAPP-ASO suppressed mouse IAPP and transgenic human IAPP expression in mouse islets, it had no discernable effects on IAPP expression in human islets under the conditions studied. Suppression of transgenic human IAPP expression in mouse islets attenuated disruption of the autophagy lysosomal pathway in beta cells, supporting the potential of this strategy.

3.
Proc Natl Acad Sci U S A ; 118(11)2021 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-33692125

RESUMEN

Rare genetic mutations result in aggregation and spreading of cognate proteins in neurodegenerative disorders; however, in the absence of mutation (i.e., in the vast majority of "sporadic" cases), mechanisms for protein misfolding/aggregation remain largely unknown. Here, we show environmentally induced nitrosative stress triggers protein aggregation and cell-to-cell spread. In patient brains with amyotrophic lateral sclerosis (ALS)/frontotemporal dementia (FTD), aggregation of the RNA-binding protein TDP-43 constitutes a major component of aberrant cytoplasmic inclusions. We identify a pathological signaling cascade whereby reactive nitrogen species cause S-nitrosylation of TDP-43 (forming SNO-TDP-43) to facilitate disulfide linkage and consequent TDP-43 aggregation. Similar pathological SNO-TDP-43 levels occur in postmortem human FTD/ALS brains and in cell-based models, including human-induced pluripotent stem cell (hiPSC)-derived neurons. Aggregated TDP-43 triggers additional nitrosative stress, representing positive feed forward leading to further SNO-TDP-43 formation and disulfide-linked oligomerization/aggregation. Critically, we show that these redox reactions facilitate cell spreading in vivo and interfere with the TDP-43 RNA-binding activity, affecting SNMT1 and phospho-(p)CREB levels, thus contributing to neuronal damage in ALS/FTD disorders.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Proteínas de Unión al ADN/metabolismo , Demencia Frontotemporal/metabolismo , S-Nitrosotioles/metabolismo , Esclerosis Amiotrófica Lateral/patología , Encéfalo/metabolismo , Encéfalo/patología , Cisteína/metabolismo , Proteínas de Unión al ADN/química , Demencia Frontotemporal/patología , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Neuronas Motoras/metabolismo , Óxido Nítrico/metabolismo , Agregación Patológica de Proteínas , Procesamiento Postranscripcional del ARN , Especies de Nitrógeno Reactivo/metabolismo , S-Nitrosotioles/química , Estrés Fisiológico
4.
J Am Chem Soc ; 143(9): 3416-3429, 2021 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-33626278

RESUMEN

The extra hepatic delivery of antisense oligonucleotides (ASOs) remains a challenge and hampers the widespread application of this powerful class of therapeutic agents. In that regard, pancreatic beta cells are a particularly attractive but challenging cell type because of their pivotal role in diabetes and the fact that they are refractory to uptake of unconjugated ASOs. To circumvent this, we have expanded our understanding of the structure activity relationship of ASOs conjugated to Glucagon Like Peptide 1 Receptor (GLP1R) agonist peptide ligands. We demonstrate the key role of the linker chemistry and its optimization to design maleimide based conjugates with improved in vivo efficacy. In addition, truncation studies and scoping of a diverse set of GLP1R agonists proved fruitful to identify additional targeting ligands efficacious in vivo including native hGLP1(7-36)NH2. Variation of the carrier peptide also shed some light on the dramatic impact of subtle sequence differences on the corresponding ASO conjugate performance in vivo, an area which clearly warrant further investigations. We have confirmed the remarkable potential of GLP1R agonist conjugation for the delivery of ASOs to pancreatic beta cell by effectively knocking down islet amyloid polypeptide (IAPP) mRNA, a potential proapoptotic target, in mice.


Asunto(s)
Portadores de Fármacos/química , Receptor del Péptido 1 Similar al Glucagón/química , Células Secretoras de Insulina/efectos de los fármacos , Oligonucleótidos Antisentido/farmacología , Péptidos/química , Secuencia de Aminoácidos , Animales , Receptor del Péptido 1 Similar al Glucagón/agonistas , Células HEK293 , Humanos , Polipéptido Amiloide de los Islotes Pancreáticos/genética , Ratones Endogámicos C57BL , Estructura Molecular , ARN Mensajero/metabolismo , Relación Estructura-Actividad
5.
Nucleic Acid Ther ; 29(5): 266-277, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31368839

RESUMEN

Although antisense oligonucleotides (ASOs) are well tolerated preclinically and in the clinic, some sequences of ASOs can trigger an inflammatory response leading to B cell and macrophage activation in rodents. This prompted our investigation into the contribution of genetic architecture to the ASO-mediated inflammatory response. Genome-wide association (GWA) and transcriptomic analysis in a hybrid mouse diversity panel (HMDP) were used to identify and validate novel genes involved in the acute and delayed inflammatory response to a single 75 mg/kg dose of an inflammatory 2'-O-methoxyethyl (2'MOE) modified ASO. The acute response was measured 6 h after ASO administration, via evaluation for increased plasma production of interleukin 6 (IL6), IL10, monocyte chemoattractant protein 1 (MCP-1) and macrophage inflammatory protein-1ß (MIP-1ß). Delayed inflammation was evaluated by spleen weight increases after 96 h. We identified single nucleotide polymorphisms (SNPs) on chromosomes 16 and 17 associated with plasma MIP-1ß, IL6, and MCP-1 levels, and one on chromosome 8 associated with increases in spleen weight. Systems genetic analysis utilizing transcriptomic data from HMDP strain macrophages determined that the acute inflammatory SNPs were expression quantitative trait locis (eQTLs) for CCAAT/enhancer-binding protein beta (Cebpb) and salt inducible kinase 1 (Sik1). The delayed inflammatory SNP was an eQTL for Rho guanine nucleotide exchange factor 10 (Arhgef10). In vitro assays in mouse primary cells and human cell lines have confirmed the HMDP finding that lower Sik1 expression increases the acute inflammatory response. Our results demonstrate the utility of using mouse GWA study (GWAS) and the HMDP for detecting genes modulating the inflammatory response to pro-inflammatory ASOs in a pharmacological setting.


Asunto(s)
Predisposición Genética a la Enfermedad , Inflamación/terapia , Oligonucleótidos Antisentido/farmacología , Transcriptoma/genética , Animales , Proteína beta Potenciadora de Unión a CCAAT/genética , Línea Celular , Quimiocina CCL2/genética , Quimiocina CCL4/genética , Perfilación de la Expresión Génica/métodos , Estudio de Asociación del Genoma Completo/métodos , Humanos , Inflamación/genética , Inflamación/patología , Ratones , Polimorfismo de Nucleótido Simple/genética , Proteínas Serina-Treonina Quinasas/genética
6.
PLoS Genet ; 14(10): e1007732, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30372444

RESUMEN

Antisense oligonucleotides (ASOs) have demonstrated variation of efficacy in patient populations. This has prompted our investigation into the contribution of genetic architecture to ASO pharmacokinetics (PK) and pharmacodynamics (PD). Genome wide association (GWA) and transcriptomic analysis in a hybrid mouse diversity panel (HMDP) were used to identify and validate novel genes involved in the uptake and efficacy of a single dose of a Malat1 constrained ethyl (cEt) modified ASO. The GWA of the HMDP identified two significant associations on chromosomes 4 and 10 with hepatic Malat1 ASO concentrations. Stabilin 2 (Stab2) and vesicle associated membrane protein 3 (Vamp3) were identified by cis-eQTL analysis. HMDP strains with lower Stab2 expression and Stab2 KO mice displayed significantly lower PK than strains with higher Stab2 expression and the wild type (WT) animals respectively, confirming the role of Stab2 in regulating hepatic Malat1 ASO uptake. GWA examining ASO efficacy uncovered three loci associated with Malat1 potency: Small Subunit Processome Component (Utp11l) on chromosome 4, Rho associated coiled-coil containing protein kinase 2 (Rock2) and Aci-reductone dioxygenase (Adi1) on chromosome 12. Our results demonstrate the utility of mouse GWAS using the HMDP in detecting genes capable of impacting the uptake of ASOs, and identifies genes critical for the activity of ASOs in vivo.


Asunto(s)
Oligonucleótidos Antisentido/farmacocinética , ARN Largo no Codificante/genética , ARN Largo no Codificante/fisiología , Animales , Moléculas de Adhesión Celular Neuronal/genética , Moléculas de Adhesión Celular Neuronal/metabolismo , Perfilación de la Expresión Génica/métodos , Variación Genética , Estudio de Asociación del Genoma Completo , Hígado/metabolismo , Ratones , Ratones Noqueados , Oligonucleótidos Antisentido/genética , ARN Mensajero/metabolismo , Proteína 3 de Membrana Asociada a Vesículas/genética , Proteína 3 de Membrana Asociada a Vesículas/metabolismo
7.
Acta Neuropathol ; 136(3): 405-423, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29881994

RESUMEN

Sporadic amyotrophic lateral sclerosis (sALS) is the most common form of ALS, however, the molecular mechanisms underlying cellular damage and motor neuron degeneration remain elusive. To identify molecular signatures of sALS we performed genome-wide expression profiling in laser capture microdissection-enriched surviving motor neurons (MNs) from lumbar spinal cords of sALS patients with rostral onset and caudal progression. After correcting for immunological background, we discover a highly specific gene expression signature for sALS that is associated with phosphorylated TDP-43 (pTDP-43) pathology. Transcriptome-pathology correlation identified casein kinase 1ε (CSNK1E) mRNA as tightly correlated to levels of pTDP-43 in sALS patients. Enhanced crosslinking and immunoprecipitation in human sALS patient- and healthy control-derived frontal cortex, revealed that TDP-43 binds directly to and regulates the expression of CSNK1E mRNA. Additionally, we were able to show that pTDP-43 itself binds RNA. CK1E, the protein product of CSNK1E, in turn interacts with TDP-43 and promotes cytoplasmic accumulation of pTDP-43 in human stem-cell-derived MNs. Pathological TDP-43 phosphorylation is therefore, reciprocally regulated by CK1E activity and TDP-43 RNA binding. Our framework of transcriptome-pathology correlations identifies candidate genes with relevance to novel mechanisms of neurodegeneration.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Quinasa de la Caseína I/metabolismo , Proteínas de Unión al ADN/metabolismo , Neuronas Motoras/metabolismo , Médula Espinal/metabolismo , Transcriptoma , Anciano , Anciano de 80 o más Años , Esclerosis Amiotrófica Lateral/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neuronas Motoras/patología , Fosforilación , Médula Espinal/patología
8.
Cell ; 170(5): 899-912.e10, 2017 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-28803727

RESUMEN

Microsatellite repeat expansions in DNA produce pathogenic RNA species that cause dominantly inherited diseases such as myotonic dystrophy type 1 and 2 (DM1/2), Huntington's disease, and C9orf72-linked amyotrophic lateral sclerosis (C9-ALS). Means to target these repetitive RNAs are required for diagnostic and therapeutic purposes. Here, we describe the development of a programmable CRISPR system capable of specifically visualizing and eliminating these toxic RNAs. We observe specific targeting and efficient elimination of microsatellite repeat expansion RNAs both when exogenously expressed and in patient cells. Importantly, RNA-targeting Cas9 (RCas9) reverses hallmark features of disease including elimination of RNA foci among all conditions studied (DM1, DM2, C9-ALS, polyglutamine diseases), reduction of polyglutamine protein products, relocalization of repeat-bound proteins to resemble healthy controls, and efficient reversal of DM1-associated splicing abnormalities in patient myotubes. Finally, we report a truncated RCas9 system compatible with adeno-associated viral packaging. This effort highlights the potential of RCas9 for human therapeutics.


Asunto(s)
Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Terapia Genética/métodos , Oligonucleótidos Antisentido/farmacología , Animales , Células COS , Línea Celular , Células Cultivadas , Chlorocebus aethiops , Repeticiones de Microsatélite , Empalme del ARN , Expansión de Repetición de Trinucleótido
9.
Neurobiol Dis ; 84: 99-108, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25796565

RESUMEN

Nitric oxide (NO) is a gasotransmitter that impacts fundamental aspects of neuronal function in large measure through S-nitrosylation, a redox reaction that occurs on regulatory cysteine thiol groups. For instance, S-nitrosylation regulates enzymatic activity of target proteins via inhibition of active site cysteine residues or via allosteric regulation of protein structure. During normal brain function, protein S-nitrosylation serves as an important cellular mechanism that modulates a diverse array of physiological processes, including transcriptional activity, synaptic plasticity, and neuronal survival. In contrast, emerging evidence suggests that aging and disease-linked environmental risk factors exacerbate nitrosative stress via excessive production of NO. Consequently, aberrant S-nitrosylation occurs and represents a common pathological feature that contributes to the onset and progression of multiple neurodegenerative disorders, including Alzheimer's, Parkinson's, and Huntington's diseases. In the current review, we highlight recent key findings on aberrant protein S-nitrosylation showing that this reaction triggers protein misfolding, mitochondrial dysfunction, transcriptional dysregulation, synaptic damage, and neuronal injury. Specifically, we discuss the pathological consequences of S-nitrosylated parkin, myocyte enhancer factor 2 (MEF2), dynamin-related protein 1 (Drp1), protein disulfide isomerase (PDI), X-linked inhibitor of apoptosis protein (XIAP), and glyceraldehyde-3-phosphate dehydrogenase (GAPDH) under neurodegenerative conditions. We also speculate that intervention to prevent these aberrant S-nitrosylation events may produce novel therapeutic agents to combat neurodegenerative diseases.


Asunto(s)
Enfermedades Neurodegenerativas/metabolismo , Proteína S/metabolismo , Animales , Humanos
10.
Anesth Analg ; 116(1): 224-31, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23223118

RESUMEN

BACKGROUND: Painful neuropathy is a dose-limiting side effect in cancer chemotherapy. To characterize this phenomenon, we examined pain behavior and analgesic actions in a mouse model of cisplatin polyneuropathy. METHODS: Male C57BL/6 mice received intraperitoneal cisplatin or saline (2.3 mg/kg/d) every other day 6 times over 2 weeks for a total dose of 13.8 mg/kg. Thermal escape latencies, mechanical allodynia using von Frey hairs, and observation of behavior/morbidity and body weights were assessed. After onset of allodynia, we examined the actions of intraperitoneal gabapentin (100 mg/kg), etanercept (20 and 40 mg/kg), ketorolac (15 mg/kg), and morphine (1, 3, and 10 mg/kg). Additionally, using the conditioned place preference (CPP) paradigm, we examined the effects of gabapentin and ketorolac on the presumed pain state initiated by cisplatin. Additionally, we examined the spinal cord and dorsal root ganglia (DRG) of cisplatin-treated mice. RESULTS: Cisplatin, but not saline treatment, produced persistent hindpaw tactile allodynia, which persisted 46 days with no effect on thermal escape. Gabapentin and morphine, but neither etanercept nor ketorolac, produced a complete but transient (2-hour) reversal of the allodynia. Etanercept (40 mg/kg) pretreatment resulted in a delay in onset of mechanical allodynia. Using CPP, gabapentin, but not ketorolac, in cisplatin animals resulted in a significant preference for the drug-associated treatment compartment. There was no place preference in non-cisplatin-treated (nonallodynic) mice after gabapentin injection. Immunohistochemistry in cisplatin-treated mice showed no change in glial fibrillary acidic protein (astrocyte) or Iba1 (ionized calcium binding adaptor molecule 1) (microglia) activation states, but a significant increase in activated transcription factor 3 was observed in the DRG. CONCLUSIONS: Cisplatintreated mice display allodynia and an activation of DRG activated transcription factor 3, which is paralleled by its effects on behavior in the CPP system, wherein gabapentin, but not ketorolac, in the presence of the cisplatin polyneuropathy, is positively rewarding, confirming that this neuropathy is an aversive (painful) state that is ameliorated by gabapentin.


Asunto(s)
Aminas/farmacología , Analgésicos/farmacología , Antiinflamatorios no Esteroideos/farmacología , Antineoplásicos , Cisplatino , Condicionamiento Operante/efectos de los fármacos , Ácidos Ciclohexanocarboxílicos/farmacología , Ganglios Espinales/efectos de los fármacos , Hiperalgesia/inducido químicamente , Hiperalgesia/psicología , Inmunoglobulina G/farmacología , Ketorolaco Trometamina/farmacología , Factor de Transcripción 3/metabolismo , Ácido gamma-Aminobutírico/farmacología , Analgésicos Opioides/farmacología , Animales , Conducta Animal/efectos de los fármacos , Etanercept , Gabapentina , Hiperalgesia/patología , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Morfina/farmacología , Umbral del Dolor/efectos de los fármacos , Receptores del Factor de Necrosis Tumoral
11.
Proc Natl Acad Sci U S A ; 109(31): 12286-93, 2012 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-22761313

RESUMEN

Using chemical germ-line mutagenesis, we screened mice for defects in the humoral immune response to a type II T-independent immunogen and an experimental alphavirus vector. A total of 26 mutations that impair humoral immunity were recovered, and 19 of these mutations have been positionally cloned. Among the phenovariants were bumble, cellophane, and Worker ascribed to mutations in Nfkbid, Zeb1, and Ruvbl2, respectively. We show that IκBNS, the nuclear IκB-like protein encoded by Nfkbid, is required for the development of marginal zone and peritoneal B-1 B cells and additionally required for extrafollicular antibody responses to T-independent and -dependent immunogens. Zeb1 is also required for marginal zone and peritoneal B-1 B-cell development as well as T-cell development, germinal center formation, and memory B-cell responses. Finally, Ruvbl2 is required for T-cell development and maximal T-dependent antibody responses. Collectively, the mutations that we identified give us insight into the points at which disruption of an antibody response can occur. All of the mutations identified to date directly affect lymphocyte development or function; none have an exclusive effect on cells of the innate immune system.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , ADN Helicasas/inmunología , Proteínas de Homeodominio/inmunología , Inmunidad Humoral/fisiología , Factores de Transcripción de Tipo Kruppel/inmunología , Proteínas/inmunología , ATPasas Asociadas con Actividades Celulares Diversas , Animales , Células Cultivadas , ADN Helicasas/genética , Proteínas de Homeodominio/genética , Inmunidad Innata/fisiología , Péptidos y Proteínas de Señalización Intracelular , Factores de Transcripción de Tipo Kruppel/genética , Ratones , Ratones Noqueados , Mutación , Proteínas/genética , Linfocitos T/inmunología , Homeobox 1 de Unión a la E-Box con Dedos de Zinc
12.
Proc Natl Acad Sci U S A ; 109(31): 12650-5, 2012 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-22802641

RESUMEN

Using an environmentally sensitized genetic screen we identified mutations that cause inflammatory colitis in mice. The X-linked Klein-Zschocher (KLZ) mutation created a null allele of Yipf6, a member of a gene family believed to regulate vesicular transport in yeast, but without known functions in mammals. Yipf6 is a five transmembrane-spanning protein associated with Golgi compartments. Klein-Zschocher mutants were extremely sensitive to colitis induced by dextran sodium sulfate (DSS) and developed spontaneous ileitis and colitis after 16 mo of age in specific pathogen-free housing conditions. Electron microscopy, gene expression, and immunocytochemistry analyses provided evidence that impaired intestinal homeostasis stemmed from defective formation and secretion of large secretory granules from Paneth and goblet cells. These studies support a tissue- and organ-specific function for Yipf6 in the maintenance of intestinal homeostasis and implicate the orthologous human gene as a disease susceptibility locus.


Asunto(s)
Colitis/metabolismo , Células Caliciformes/metabolismo , Proteínas de la Membrana/metabolismo , Mutación , Células de Paneth/metabolismo , Animales , Colitis/inducido químicamente , Colitis/genética , Colitis/patología , Sulfato de Dextran/toxicidad , Femenino , Regulación de la Expresión Génica , Sitios Genéticos , Predisposición Genética a la Enfermedad , Células Caliciformes/ultraestructura , Aparato de Golgi/genética , Aparato de Golgi/metabolismo , Aparato de Golgi/ultraestructura , Humanos , Ileítis/inducido químicamente , Ileítis/genética , Ileítis/metabolismo , Ileítis/patología , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Transgénicos , Células de Paneth/ultraestructura
13.
Nat Immunol ; 12(5): 434-40, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21423172

RESUMEN

B lymphopoiesis begins in the fetal liver, switching after birth to the bone marrow, where it persists for life. The unique developmental outcomes of each phase are well documented, yet their molecular requirements are not. Here we describe two allelic X-linked mutations in mice that caused cell-intrinsic arrest of adult B lymphopoiesis. Mutant fetal liver progenitors generated B cells in situ but not in irradiated adult bone marrow, which emphasizes a necessity for the affected pathway only in the context of adult bone marrow. The causative mutations were ascribed to Atp11c, which encodes a P4-type ATPase with no previously described function. Our data establish an essential, cell-autonomous and context-sensitive function for ATP11C, a putative aminophospholipid flippase, in B cell development.


Asunto(s)
Adenosina Trifosfatasas/inmunología , Linfocitos B/inmunología , Médula Ósea/inmunología , Linfopoyesis/inmunología , Adenosina Trifosfatasas/genética , Animales , Linfocitos B/enzimología , Femenino , Regulación del Desarrollo de la Expresión Génica , Genotipo , Inmunoglobulina M/genética , Inmunoglobulina M/inmunología , Inmunofenotipificación , Interleucina-7/inmunología , Linfopoyesis/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Transgénicos , ARN/química , ARN/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transactivadores/genética , Transactivadores/inmunología
14.
Proc Natl Acad Sci U S A ; 107(7): 3046-51, 2010 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-20133626

RESUMEN

Null alleles of the gene encoding NEMO (NF-kappaB essential modulator) are lethal in hemizygous mice and men, whereas hypomorphic alleles typically cause a syndrome of immune deficiency and ectodermal dysplasia. Here we describe an allele of Ikbkg in mice that impaired Toll-like receptor signaling, lymph node formation, development of memory and regulatory T cells, and Ig production, but did not cause ectodermal dysplasia. Degradation of IkappaB alpha, which is considered a primary requirement for NEMO-mediated immune signaling, occurred normally in response to Toll-like receptor stimulation, yet ERK phosphorylation and NF-kappaB p65 nuclear translocation were severely impaired. This selective loss of function highlights the immunological importance of NEMO-regulated pathways beyond IkappaB alpha degradation, and offers a biochemical explanation for rare immune deficiencies in man.


Asunto(s)
Síndromes de Inmunodeficiencia/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Mutación/genética , Transducción de Señal/genética , Animales , Western Blotting , Citocinas/metabolismo , Etilnitrosourea , Citometría de Flujo , Quinasa I-kappa B/metabolismo , Péptidos y Proteínas de Señalización Intracelular/inmunología , Ganglios Linfáticos/crecimiento & desarrollo , Masculino , Ratones , Ratones Transgénicos , Mutagénesis , Óxido Nítrico/metabolismo , Transducción de Señal/inmunología , Linfocitos T Reguladores , Receptores Toll-Like/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...