Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Int J Mol Sci ; 22(15)2021 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-34360808

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a progressive disease leading to the degeneration of motor neurons (MNs). Neuroinflammation is involved in the pathogenesis of ALS; however, interactions of specific immune cell types and MNs are not well studied. We recently found a shift toward T helper (Th)1/Th17 cell-mediated, pro-inflammatory immune responses in the peripheral immune system of ALS patients, which positively correlated with disease severity and progression. Whether Th17 cells or their central mediator, Interleukin-17 (IL-17), directly affects human motor neuron survival is currently unknown. Here, we evaluated the contribution of Th17 cells and IL-17 on MN degeneration using the co-culture of iPSC-derived MNs of fused in sarcoma (FUS)-ALS patients and isogenic controls with Th17 lymphocytes derived from ALS patients, healthy controls, and multiple sclerosis (MS) patients (positive control). Only Th17 cells from MS patients induced severe MN degeneration in FUS-ALS as well as in wildtype MNs. Their main effector, IL-17A, yielded in a dose-dependent decline of the viability and neurite length of MNs. Surprisingly, IL-17F did not influence MNs. Importantly, neutralizing IL-17A and anti-IL-17 receptor A treatment reverted all effects of IL-17A. Our results offer compelling evidence that Th17 cells and IL-17A do directly contribute to MN degeneration.


Asunto(s)
Esclerosis Amiotrófica Lateral/inmunología , Células Madre Pluripotentes Inducidas/inmunología , Interleucina-17/inmunología , Neuronas Motoras/inmunología , Proteína FUS de Unión a ARN/inmunología , Células Th17/inmunología , Esclerosis Amiotrófica Lateral/patología , Supervivencia Celular/inmunología , Humanos , Células Madre Pluripotentes Inducidas/patología , Neuronas Motoras/patología , Células Th17/patología
2.
Cell Rep ; 29(13): 4496-4508.e4, 2019 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-31875556

RESUMEN

Mutations in the FUS gene cause familial amyotrophic lateral sclerosis (ALS-FUS). In ALS-FUS, FUS-positive inclusions are detected in the cytoplasm of neurons and glia, a condition known as FUS proteinopathy. Mutant FUS incorporates into stress granules (SGs) and can spontaneously form cytoplasmic RNA granules in cultured cells. However, it is unclear what can trigger the persistence of mutant FUS assemblies and lead to inclusion formation. Using CRISPR/Cas9 cell lines and patient fibroblasts, we find that the viral mimic dsRNA poly(I:C) or a SG-inducing virus causes the sustained presence of mutant FUS assemblies. These assemblies sequester the autophagy receptor optineurin and nucleocytoplasmic transport factors. Furthermore, an integral component of the antiviral immune response, type I interferon, promotes FUS protein accumulation by increasing FUS mRNA stability. Finally, mutant FUS-expressing cells are hypersensitive to dsRNA toxicity. Our data suggest that the antiviral immune response is a plausible second hit for FUS proteinopathy.


Asunto(s)
Esclerosis Amiotrófica Lateral/inmunología , Interacciones Huésped-Patógeno/inmunología , Neuronas Motoras/inmunología , Proteína FUS de Unión a ARN/inmunología , Virus Sincitiales Respiratorios/inmunología , Médula Espinal/inmunología , Transporte Activo de Núcleo Celular/genética , Transporte Activo de Núcleo Celular/inmunología , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Esclerosis Amiotrófica Lateral/virología , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/inmunología , Línea Celular , Gránulos Citoplasmáticos/genética , Gránulos Citoplasmáticos/inmunología , Gránulos Citoplasmáticos/virología , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/inmunología , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno/efectos de los fármacos , Interacciones Huésped-Patógeno/genética , Humanos , Cuerpos de Inclusión/genética , Cuerpos de Inclusión/inmunología , Cuerpos de Inclusión/virología , Interferón Tipo I/genética , Interferón Tipo I/inmunología , Masculino , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/inmunología , Neuronas Motoras/metabolismo , Neuronas Motoras/virología , Neuroglía/inmunología , Neuroglía/metabolismo , Neuroglía/virología , Proteínas de Transporte Nucleocitoplasmático/genética , Proteínas de Transporte Nucleocitoplasmático/inmunología , Poli I-C/farmacología , Cultivo Primario de Células , Agregado de Proteínas/genética , Agregado de Proteínas/inmunología , Estabilidad del ARN , ARN Mensajero/genética , ARN Mensajero/inmunología , Proteína FUS de Unión a ARN/genética , Virus Sincitiales Respiratorios/patogenicidad , Médula Espinal/metabolismo , Médula Espinal/patología , Médula Espinal/virología
3.
J Mol Biol ; 431(9): 1818-1829, 2019 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-30763568

RESUMEN

Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are neurodegenerative disorders with clear similarities regarding their clinical, genetic and pathological features. Both are progressive, lethal disorders, with no current curative treatment available. Several genes that correlated with ALS and FTD are implicated in the same molecular pathways. Strikingly, many of these genes are not exclusively expressed in neurons, but also in glial cells, suggesting a multicellular pathogenesis. Moreover, chronic inflammation is a common feature observed in ALS and FTD, indicating an essential role of microglia, the resident immune cells of the central nervous system, in disease development and progression. In this review, we will provide a comprehensive overview of the implications of microglia in ALS and FTD. Specifically, we will focus on the role of impaired phagocytosis and increased inflammatory responses and their impact on microglial function. Several genes associated with the disorders can directly be linked to microglial activation, phagocytosis and neuroinflammation. Other genes associated with the disorders are implicated in biological pathways involved in protein degradation and autophagy. In general such mutations have been shown to cause abnormal protein accumulation and impaired autophagy. These impairments have previously been linked to affect the innate immune system in the central nervous system through inappropriate activation of microglia and neuroinflammation, highlighted in this review. Although it has been well established that microglia play essential roles in neurodegenerative disorders, the precise underlying mechanisms remain to be elucidated.


Asunto(s)
Esclerosis Amiotrófica Lateral/inmunología , Autofagia/inmunología , Demencia Frontotemporal/inmunología , Regulación de la Expresión Génica/inmunología , Microglía/inmunología , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Animales , Autofagia/genética , Proteína C9orf72/genética , Proteína C9orf72/inmunología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/inmunología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Demencia Frontotemporal/genética , Demencia Frontotemporal/patología , Humanos , Inmunidad Innata , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Ratones , Microglía/patología , Proteína FUS de Unión a ARN/genética , Proteína FUS de Unión a ARN/inmunología , Receptores Inmunológicos/genética , Receptores Inmunológicos/inmunología , Superóxido Dismutasa-1/genética , Superóxido Dismutasa-1/inmunología , Proteínas tau/genética , Proteínas tau/inmunología
4.
J Neurophysiol ; 116(2): 671-85, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27146988

RESUMEN

Voltage-dependent potassium channels (Kv) go beyond the stabilization of the resting potential and regulate biochemical pathways, regulate intracellular signaling, and detect energy homeostasis. Because targeted deletion and pharmacological block of the Kv1.3 channel protein produce marked changes in metabolism, resistance to diet-induced obesity, and changes in olfactory structure and function, this investigation explored Nedd4-2-mediated ubiquitination and degradation to regulate Kv1.3 channel density. Heterologous coexpression of Nedd4-2 ligase and Kv1.3 in HEK 293 cells reduced Kv1.3 current density without modulation of kinetic properties as measured by patch-clamp electrophysiology. Modulation of current density was dependent on ligase activity and was lost through point mutation of cysteine 938 in the catalytic site of the ligase (Nedd4-2CS). Incorporation of adaptor protein Grb10 relieved Nedd4-2-induced current suppression as did application of the proteasome inhibitor Mg-132. SDS-PAGE and immunoprecipitation strategies demonstrated a channel/adaptor/ligase signalplex. Pixel immunodensity was reduced for Kv1.3 in the presence of Nedd4-2, which was eliminated upon additional incorporation of Grb10. We confirmed Nedd4-2/Grb10 coimmunoprecipitation and observed an increased immunodensity for Nedd4-2 in the presence of Kv1.3 plus Grb10, regardless of whether the catalytic site was active. Kv1.3/Nedd4-2 were reciprocally coimmunoprecipated, whereby mutation of the COOH-terminal, SH3-recognition (493-498), or ubiquitination sites on Kv1.3 (lysines 467, 476, 498) retained coimmunoprecipitation, while the latter prevented the reduction in channel density. A model is presented for which an atypical interaction outside the canonical PY motif may permit channel/ligase interaction to lead to protein degradation and reduced current density, which can involve Nedd4-2/Grb10 interactions to disrupt Kv1.3 loss of current density.


Asunto(s)
Regulación hacia Abajo/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Regulación de la Expresión Génica/genética , Canal de Potasio Kv1.3/metabolismo , Potenciales de la Membrana/genética , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Anticuerpos/farmacología , Línea Celular Transformada , Cisteína/genética , Inhibidores de Cisteína Proteinasa/farmacología , Estimulación Eléctrica , Proteína Adaptadora GRB10/farmacología , Células HEK293 , Humanos , Canal de Potasio Kv1.3/efectos de los fármacos , Leupeptinas/farmacología , Lisina/metabolismo , Potenciales de la Membrana/efectos de los fármacos , Modelos Biológicos , Mutación/genética , Ubiquitina-Proteína Ligasas Nedd4 , Técnicas de Placa-Clamp , Proteína FUS de Unión a ARN/inmunología , Ubiquitinación/efectos de los fármacos , Ubiquitinación/genética
5.
Acta Neuropathol ; 131(4): 587-604, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26895297

RESUMEN

Deposition of the nuclear DNA/RNA-binding protein Fused in sarcoma (FUS) in cytosolic inclusions is a common hallmark of some cases of frontotemporal lobar degeneration (FTLD-FUS) and amyotrophic lateral sclerosis (ALS-FUS). Whether both diseases also share common pathological mechanisms is currently unclear. Based on our previous finding that FUS deposits are hypomethylated in FTLD-FUS but not in ALS-FUS, we have now investigated whether genetic or pharmacological inactivation of Protein arginine methyltransferase 1 (PRMT1) activity results in unmethylated FUS or in alternatively methylated forms of FUS. To do so, we generated FUS-specific monoclonal antibodies that specifically recognize unmethylated arginine (UMA), monomethylated arginine (MMA) or asymmetrically dimethylated arginine (ADMA). Loss of PRMT1 indeed not only results in an increase of UMA FUS and a decrease of ADMA FUS, but also in a significant increase of MMA FUS. Compared to ADMA FUS, UMA and MMA FUS exhibit much higher binding affinities to Transportin-1, the nuclear import receptor of FUS, as measured by pull-down assays and isothermal titration calorimetry. Moreover, we show that MMA FUS occurs exclusively in FTLD-FUS, but not in ALS-FUS. Our findings therefore provide additional evidence that FTLD-FUS and ALS-FUS are caused by distinct disease mechanisms although both share FUS deposits as a common denominator.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Degeneración Lobar Frontotemporal/metabolismo , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteína FUS de Unión a ARN/metabolismo , beta Carioferinas/metabolismo , Esclerosis Amiotrófica Lateral/genética , Animales , Anticuerpos/farmacología , Arginina/metabolismo , Células Cultivadas , Corteza Cerebral/citología , Embrión de Mamíferos , Células Madre Embrionarias , Inhibidores Enzimáticos/farmacología , Femenino , Degeneración Lobar Frontotemporal/genética , Humanos , Cuerpos de Inclusión/efectos de los fármacos , Cuerpos de Inclusión/metabolismo , Masculino , Metilación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Unión Proteica/efectos de los fármacos , Unión Proteica/genética , Proteína-Arginina N-Metiltransferasas/genética , Proteína FUS de Unión a ARN/inmunología , Ratas , beta Carioferinas/inmunología
6.
J Proteome Res ; 14(5): 2121-42, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25815715

RESUMEN

Viruses employ numerous host cell metabolic functions to propagate and manage to evade the host immune system. For herpes simplex virus type 1 (HSV1), a virus that has evolved to efficiently infect humans without seriously harming the host in most cases, the virus-host interaction is specifically interesting. This interaction can be best characterized by studying the proteomic changes that occur in the host during infection. Previous studies have been successful at identifying numerous host proteins that play important roles in HSV infection; however, there is still much that we do not know. This study identifies host metabolic functions and proteins that play roles in HSV infection, using global quantitative stable isotope labeling by amino acids in cell culture (SILAC) proteomic profiling of the host cell combined with LC-MS/MS. We showed differential proteins during early, mid and late infection, using both cytosolic and nuclear fractions. We identified hundreds of differentially regulated proteins involved in fundamental cellular functions, including gene expression, DNA replication, inflammatory response, cell movement, cell death, and RNA post-transcriptional modification. Novel differentially regulated proteins in HSV infections include some previously identified in other virus systems, as well as fusion protein, involved in malignant liposarcoma (FUS) and hypoxia up-regulated 1 protein precursor (HYOU1), which have not been identified previously in any virus infection.


Asunto(s)
Herpesvirus Humano 1/fisiología , Redes y Vías Metabólicas/genética , Mapas de Interacción de Proteínas/genética , Proteoma/genética , Animales , Isótopos de Carbono , Chlorocebus aethiops , Cromatografía Liquida , Regulación de la Expresión Génica , Células HEK293 , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/inmunología , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Marcaje Isotópico , Redes y Vías Metabólicas/inmunología , Mapas de Interacción de Proteínas/inmunología , Proteoma/inmunología , Proteína FUS de Unión a ARN/genética , Proteína FUS de Unión a ARN/inmunología , Espectrometría de Masas en Tándem , Células Vero
7.
J Allergy Clin Immunol ; 135(6): 1569-77, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25678086

RESUMEN

BACKGROUND: Common variable immunodeficiency (CVID) is characterized clinically by inadequate quantity and quality of serum immunoglobulins with increased susceptibility to infections, resulting in significant morbidity and mortality. Only a few genes have been uncovered, and the genetic background of CVID remains elusive to date for the majority of patients. OBJECTIVE: We sought to seek novel associations of genes and genetic variants with CVID. METHODS: We performed association analyses in a discovery cohort of 164 patients with CVID and 19,542 healthy control subjects genotyped on the Immuno BeadChip from Illumina platform; replication of findings was examined in an independent cohort of 135 patients with CVID and 2,066 healthy control subjects, followed by meta-analysis. RESULTS: We identified 11 single nucleotide polymorphisms (SNPs) at the 16p11.2 locus associated with CVID at a genome-wide significant level in the discovery cohort. The most significant SNP, rs929867 (P = 6.21 × 10(-9)), is in the gene fused-in-sarcoma (FUS), with 4 other SNPs mapping to integrin CD11b (ITGAM). Results were confirmed in our replication cohort. Conditional association analysis suggests a single association signal at the 16p11.2 locus. A strong trend of association was also seen for 38 SNPs (P < 5 × 10(-5)) in the MHC region, supporting that this is a genuine CVID locus. Interestingly, we found that 80% of patients with the rare ITGAM variants have reduced switched memory B-cell counts. CONCLUSION: We report a novel association of CVID with rare variants at the FUS/ITGAM (CD11b) locus on 16p11.2. The association signal is enriched for promoter/enhancer markers in the ITGAM gene. ITGAM encodes the integrin CD11b, a part of complement receptor 3, a novel candidate gene implicated here for the first time in the pathogenesis of CVID.


Asunto(s)
Antígeno CD11b/genética , Cromosomas Humanos Par 16 , Inmunodeficiencia Variable Común/genética , Predisposición Genética a la Enfermedad , Polimorfismo de Nucleótido Simple , Proteína FUS de Unión a ARN/genética , Adulto , Linfocitos B/inmunología , Linfocitos B/metabolismo , Linfocitos B/patología , Antígeno CD11b/inmunología , Estudios de Casos y Controles , Preescolar , Estudios de Cohortes , Inmunodeficiencia Variable Común/diagnóstico , Inmunodeficiencia Variable Común/inmunología , Inmunodeficiencia Variable Común/patología , Elementos de Facilitación Genéticos , Femenino , Sitios Genéticos , Humanos , Memoria Inmunológica , Desequilibrio de Ligamiento , Masculino , Regiones Promotoras Genéticas , Proteína FUS de Unión a ARN/inmunología
8.
PLoS One ; 7(6): e39483, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22724023

RESUMEN

Mutations in the fused in sarcoma/translated in liposarcoma gene (FUS/TLS, FUS) have been identified in sporadic and familial forms of amyotrophic lateral sclerosis (ALS). FUS is an RNA-binding protein that is normally localized in the nucleus, but is mislocalized to the cytoplasm in ALS, and comprises cytoplasmic inclusions in ALS-affected areas. However, it is still unknown whether the neurodegeneration that occurs in ALS is caused by the loss of FUS nuclear function, or by the gain of toxic function due to cytoplasmic FUS aggregation. Cabeza (Caz) is a Drosophila orthologue of human FUS. Here, we generated Drosophila models with Caz knockdown, and investigated their phenotypes. In wild-type Drosophila, Caz was strongly expressed in the central nervous system of larvae and adults. Caz did not colocalize with a presynaptic marker, suggesting that Caz physiologically functions in neuronal cell bodies and/or their axons. Fly models with neuron-specific Caz knockdown exhibited reduced climbing ability in adulthood and anatomical defects in presynaptic terminals of motoneurons in third instar larvae. Our results demonstrated that decreased expression of Drosophila Caz is sufficient to cause degeneration of motoneurons and locomotive disability in the absence of abnormal cytoplasmic Caz aggregates, suggesting that the pathogenic mechanism underlying FUS-related ALS should be ascribed more to the loss of physiological FUS functions in the nucleus than to the toxicity of cytoplasmic FUS aggregates. Since the Caz-knockdown Drosophila model we presented recapitulates key features of human ALS, it would be a suitable animal model for the screening of genes and chemicals that might modify the pathogenic processes that lead to the degeneration of motoneurons in ALS.


Asunto(s)
Drosophila/genética , Drosophila/metabolismo , Actividad Motora/genética , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Proteína FUS de Unión a ARN/genética , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Animales Modificados Genéticamente , Anticuerpos/inmunología , Especificidad de Anticuerpos , Encéfalo/metabolismo , Técnicas de Silenciamiento del Gen , Genotipo , Humanos , Longevidad/genética , Masculino , Datos de Secuencia Molecular , Terminales Presinápticos/metabolismo , Dominios y Motivos de Interacción de Proteínas , Transporte de Proteínas , Proteína FUS de Unión a ARN/química , Proteína FUS de Unión a ARN/inmunología
9.
Neuropathol Appl Neurobiol ; 38(4): 322-8, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21883376

RESUMEN

AIMS: Recent studies have shown that fused-in-sarcoma (FUS) protein is a component of 'neuronal' intranuclear inclusion bodies (INIBs) in the brains of patients with intranuclear inclusion body disease (INIBD). However, the extent and frequency of FUS-immunoreactive structures in INIBD are uncertain. METHODS: We immunohistochemically examined the brain, spinal cord and peripheral ganglia from five patients with INIBD and five control subjects, using anti-FUS antibodies. RESULTS: In controls, the nuclei of both neurones and glial cells were intensely immunolabelled with anti-FUS and neuronal cytoplasm was weakly positive for FUS. In INIBD, neuronal and glial INIBs in the brain and spinal cord were positive for FUS. FUS-positive INIBs were also found in the peripheral ganglia. The proportion of FUS-positive neuronal INIBs relative to the total number of inclusion-bearing neurones ranged from 55.6% to 83.3% (average 73.2%) and that of FUS-positive glial INIBs ranged from 45.9% to 85.7% (average 62.7%). The nucleus and cytoplasm of inclusion-bearing neurones and glial cells showed no FUS immunoreactivity. CONCLUSIONS: These findings suggest that FUS is incorporated into INIBs in both neurones and glial cells and that loss of normal FUS immunoreactivity may result from reduced protein expression and/or sequestration within inclusions.


Asunto(s)
Cuerpos de Inclusión Intranucleares/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Neuroglía/metabolismo , Neuronas/metabolismo , Proteína FUS de Unión a ARN/metabolismo , Anciano , Encéfalo/inmunología , Encéfalo/metabolismo , Encéfalo/patología , Femenino , Humanos , Inmunohistoquímica , Cuerpos de Inclusión Intranucleares/inmunología , Cuerpos de Inclusión Intranucleares/patología , Persona de Mediana Edad , Enfermedades Neurodegenerativas/inmunología , Enfermedades Neurodegenerativas/patología , Neuroglía/inmunología , Neuroglía/patología , Neuronas/inmunología , Neuronas/patología , Proteína FUS de Unión a ARN/inmunología , Médula Espinal/inmunología , Médula Espinal/metabolismo , Médula Espinal/patología
10.
Am J Surg Pathol ; 30(3): 351-6, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16538055

RESUMEN

The fusion oncoproteins, TLS-CHOP and EWS-CHOP, are characteristic markers for myxoid and round cell liposarcomas (MLS/RCLS). Especially, the peptide sequence of 26 amino acids corresponding to the normally untranslated CHOP exon 2 and parts of exon 3 (5'-UTR) is a unique structure for these chimeric proteins. In this report, we have generated monoclonal antibodies against the unique peptide sequence of TLS/EWS-CHOP oncoproteins. These antibodies reacted with TLS-CHOP fusion protein, but not reacted with normal TLS and CHOP proteins by Western blot analysis. In addition, one of the antibodies also recognized the chimeric oncoprotein in archival paraffin-embedded tissue samples of MLS/RCLS. The oncoprotein was detectable by the antibody even in the paraffin-embedded tissue samples whose mRNAs were too degraded to be detected by a nested reverse transcription-polymerase chain reaction-based assay. Thus, the molecular assay using the novel antibody is expected to be one of the most sensitive diagnostic assays for MLS/RCLS.


Asunto(s)
Anticuerpos Monoclonales , Liposarcoma/diagnóstico , Proteínas de Fusión Oncogénica/inmunología , Proteína EWS de Unión a ARN/inmunología , Proteína FUS de Unión a ARN/inmunología , Factor de Transcripción CHOP/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Monoclonales/inmunología , Western Blotting , Niño , Femenino , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Adhesión en Parafina , Proteína EWS de Unión a ARN/genética , Proteína EWS de Unión a ARN/metabolismo , Proteína FUS de Unión a ARN/genética , Proteína FUS de Unión a ARN/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sensibilidad y Especificidad , Factor de Transcripción CHOP/genética , Factor de Transcripción CHOP/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA