Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Dev Cell ; 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38878774

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a rapidly progressing, highly heterogeneous neurodegenerative disease, underscoring the importance of obtaining information to personalize clinical decisions quickly after diagnosis. Here, we investigated whether ALS-relevant signatures can be detected directly from biopsied patient fibroblasts. We profiled familial ALS (fALS) fibroblasts, representing a range of mutations in the fused in sarcoma (FUS) gene and ages of onset. To differentiate FUS fALS and healthy control fibroblasts, machine-learning classifiers were trained separately on high-content imaging and transcriptional profiles. "Molecular ALS phenotype" scores, derived from these classifiers, captured a spectrum from disease to health. Interestingly, these scores negatively correlated with age of onset, identified several pre-symptomatic individuals and sporadic ALS (sALS) patients with FUS-like fibroblasts, and quantified "movement" of FUS fALS and "FUS-like" sALS toward health upon FUS ASO treatment. Taken together, these findings provide evidence that non-neuronal patient fibroblasts can be used for rapid, personalized assessment in ALS.

3.
Nature ; 606(7916): 945-952, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35732742

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a heterogenous neurodegenerative disorder that affects motor neurons and voluntary muscle control1. ALS heterogeneity includes the age of manifestation, the rate of progression and the anatomical sites of symptom onset. Disease-causing mutations in specific genes have been identified and define different subtypes of ALS1. Although several ALS-associated genes have been shown to affect immune functions2, whether specific immune features account for ALS heterogeneity is poorly understood. Amyotrophic lateral sclerosis-4 (ALS4) is characterized by juvenile onset and slow progression3. Patients with ALS4 show motor difficulties by the time that they are in their thirties, and most of them require devices to assist with walking by their fifties. ALS4 is caused by mutations in the senataxin gene (SETX). Here, using Setx knock-in mice that carry the ALS4-causative L389S mutation, we describe an immunological signature that consists of clonally expanded, terminally differentiated effector memory (TEMRA) CD8 T cells in the central nervous system and the blood of knock-in mice. Increased frequencies of antigen-specific CD8 T cells in knock-in mice mirror the progression of motor neuron disease and correlate with anti-glioma immunity. Furthermore, bone marrow transplantation experiments indicate that the immune system has a key role in ALS4 neurodegeneration. In patients with ALS4, clonally expanded TEMRA CD8 T cells circulate in the peripheral blood. Our results provide evidence of an antigen-specific CD8 T cell response in ALS4, which could be used to unravel disease mechanisms and as a potential biomarker of disease state.


Asunto(s)
Esclerosis Amiotrófica Lateral , Linfocitos T CD8-positivos , Células Clonales , Esclerosis Amiotrófica Lateral/inmunología , Esclerosis Amiotrófica Lateral/patología , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Células Clonales/patología , ADN Helicasas/genética , ADN Helicasas/metabolismo , Técnicas de Sustitución del Gen , Ratones , Neuronas Motoras/patología , Enzimas Multifuncionales/genética , Enzimas Multifuncionales/metabolismo , Mutación , ARN Helicasas/genética , ARN Helicasas/metabolismo
4.
Nat Med ; 28(1): 104-116, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35075293

RESUMEN

Fused in sarcoma (FUS) is an RNA-binding protein that is genetically and pathologically associated with rare and aggressive forms of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). To explore the mechanisms by which mutant FUS causes neurodegeneration in ALS-FTD, we generated a series of FUS knock-in mouse lines that express the equivalent of ALS-associated mutant FUSP525L and FUSΔEX14 protein. In FUS mutant mice, we show progressive, age-dependent motor neuron loss as a consequence of a dose-dependent gain of toxic function, associated with the insolubility of FUS and related RNA-binding proteins. In this disease-relevant mouse model of ALS-FUS, we show that ION363, a non-allele-specific FUS antisense oligonucleotide, efficiently silences Fus and reduces postnatal levels of FUS protein in the brain and spinal cord, delaying motor neuron degeneration. In a patient with ALS with a FUSP525L mutation, we provide preliminary evidence that repeated intrathecal infusions of ION363 lower wild-type and mutant FUS levels in the central nervous system, resulting in a marked reduction in the burden of FUS aggregates that are a pathological hallmark of disease. In mouse genetic and human clinical studies, we provide evidence in support of FUS silencing as a therapeutic strategy in FUS-dependent ALS and FTD.


Asunto(s)
Esclerosis Amiotrófica Lateral/terapia , Silenciador del Gen/efectos de los fármacos , Oligonucleótidos Antisentido/farmacología , Proteína FUS de Unión a ARN/genética , Esclerosis Amiotrófica Lateral/genética , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Transgénicos , Mutación
5.
Clin Cancer Res ; 25(13): 4179-4193, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-30867219

RESUMEN

PURPOSE: For many tumors, signaling exchanges between cancer cells and other cells in their microenvironment influence overall tumor signaling. Some of these exchanges depend on expression of the primary cilium on nontransformed cell populations, as extracellular ligands including Sonic Hedgehog (SHH), PDGFRα, and others function through receptors spatially localized to cilia. Cell ciliation is regulated by proteins that are themselves therapeutic targets. We investigated whether kinase inhibitors of clinical interest influence ciliation and signaling by proteins with ciliary receptors in cancer and other cilia-relevant disorders, such as polycystic kidney disease (PKD). EXPERIMENTAL DESIGN: We screened a library of clinical and preclinical kinase inhibitors, identifying drugs that either prevented or induced ciliary disassembly. Specific bioactive protein targets of the drugs were identified by mRNA depletion. Mechanism of action was defined, and activity of select compounds investigated. RESULTS: We identified multiple kinase inhibitors not previously linked to control of ciliation, including sunitinib, erlotinib, and an inhibitor of the innate immune pathway kinase, IRAK4. For all compounds, activity was mediated through regulation of Aurora-A (AURKA) activity. Drugs targeting cilia influenced proximal cellular responses to SHH and PDGFRα. In vivo, sunitinib durably limited ciliation and cilia-related biological activities in renal cells, renal carcinoma cells, and PKD cysts. Extended analysis of IRAK4 defined a subset of innate immune signaling effectors potently affecting ciliation. CONCLUSIONS: These results suggest a paradigm by which targeted drugs may have unexpected off-target effects in heterogeneous cell populations in vivo via control of a physical platform for receipt of extracellular ligands.


Asunto(s)
Cilios/efectos de los fármacos , Cilios/metabolismo , Descubrimiento de Drogas , Animales , Biomarcadores , Línea Celular , Susceptibilidad a Enfermedades , Clorhidrato de Erlotinib/farmacología , Proteínas Hedgehog/metabolismo , Humanos , Enfermedades Renales Quísticas/etiología , Enfermedades Renales Quísticas/metabolismo , Enfermedades Renales Quísticas/patología , Ratones , Modelos Biológicos , Comunicación Paracrina/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Riñón Poliquístico Autosómico Dominante/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , ARN Interferente Pequeño/genética , Transducción de Señal/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas , Sunitinib/farmacología
6.
Cell Rep ; 16(3): 657-71, 2016 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-27396341

RESUMEN

Anti-Müllerian hormone (AMH) and its type II receptor AMHR2, both previously thought to primarily function in gonadal tissue, were unexpectedly identified as potent regulators of transforming growth factor (TGF-ß)/bone morphogenetic protein (BMP) signaling and epithelial-mesenchymal transition (EMT) in lung cancer. AMH is a TGF-ß/BMP superfamily member, and AMHR2 heterodimerizes with type I receptors (ALK2, ALK3) also used by the type II receptor for BMP (BMPR2). AMH signaling regulates expression of BMPR2, ALK2, and ALK3, supports protein kinase B-nuclear factor κB (AKT-NF-κB) and SMAD survival signaling, and influences BMP-dependent signaling in non-small cell lung cancer (NSCLC). AMH and AMHR2 are selectively expressed in epithelial versus mesenchymal cells, and loss of AMH/AMHR2 induces EMT. Independent induction of EMT reduces expression of AMH and AMHR2. Importantly, EMT associated with depletion of AMH or AMHR2 results in chemoresistance but sensitizes cells to the heat shock protein 90 (HSP90) inhibitor ganetespib. Recognition of this AMH/AMHR2 axis helps to further elucidate TGF-ß/BMP resistance-associated signaling and suggests new strategies for therapeutic targeting of EMT.


Asunto(s)
Hormona Antimülleriana/metabolismo , Plasticidad de la Célula/fisiología , Resistencia a Antineoplásicos/fisiología , Células Epiteliales/metabolismo , Células Epiteliales/fisiología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Animales , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Transición Epitelial-Mesenquimal/fisiología , Regulación de la Expresión Génica/fisiología , Proteínas de Choque Térmico/metabolismo , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Ratones , Ratones SCID , FN-kappa B/metabolismo , Receptores de Péptidos/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/metabolismo
7.
Colloids Surf B Biointerfaces ; 140: 269-277, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26764110

RESUMEN

Gemini surfactants with hexadecyl tails and hydroxyethylated head groups bridged with tetramethylene (G4), hexamethylene (G6) and dodecamethylene (G12) spacers were shown to self-assemble at the lower critical micelle concentration compared to their conventional m-s-m analogs. The lipoplex formation and the plasmid DNA transfer into different kinds of host cells were studied. In the case of eukaryotic cells, high transfection efficacy has been demonstrated for DNA-gemini complexes, which increased as follows: G6G4>G12 has been obtained in the case of transformation of bacterial cells with plasmid DNA-gemini complexes, mediated by electroporation technique. Solely G6 shows transformation efficacy exceeding the control result (uncomplexed DNA), while the inhibitory effect occurs for G4 and G12. Analysis of physico-chemical features of single surfactants and lipoplexes shows that compaction and condensation effects change as follows: G6

Asunto(s)
Técnicas de Transferencia de Gen , Compuestos de Amonio Cuaternario/química , Tensoactivos/química , Transfección/métodos , ADN/química , ADN/genética , Electroporación , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Microscopía de Fuerza Atómica , Estructura Molecular , Conformación de Ácido Nucleico , Plásmidos/química , Plásmidos/genética , Reproducibilidad de los Resultados , Relación Estructura-Actividad , Termodinámica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA