Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell Commun Signal ; 17(1): 74, 2019 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-31291965

RESUMEN

BACKGROUND: We have found that erbB receptor tyrosine kinases drive Ras hyperactivation and growth in NF1-null malignant peripheral nerve sheath tumors (MPNSTs). However, MPNSTs variably express multiple erbB receptors with distinct functional characteristics and it is not clear which of these receptors drive MPNST pathogenesis. Here, we test the hypothesis that altered erbB4 expression promotes MPNST pathogenesis by uniquely activating key cytoplasmic signaling cascades. METHODS: ErbB4 expression was assessed using immunohistochemistry, immunocytochemistry, immunoblotting and real-time PCR. To define erbB4 functions, we generated mice that develop MPNSTs with floxed Erbb4 alleles (P0-GGFß3;Trp53+/-;Erbb4flox/flox mice) and ablated Erbb4 in these tumors. MPNST cell proliferation and survival was assessed using 3H-thymidine incorporation, MTT assays, Real-Time Glo and cell count assays. Control and Erbb4-null MPNST cells were orthotopically xenografted in immunodeficient mice and the growth, proliferation (Ki67 labeling), apoptosis (TUNEL labeling) and angiogenesis of these grafts was analyzed. Antibody arrays querying cytoplasmic kinases were used to identify erbB4-responsive kinases. Pharmacologic or genetic inhibition was used to identify erbB4-responsive kinases that drive proliferation. RESULTS: Aberrant erbB4 expression was evident in 25/30 surgically resected human MPNSTs and in MPNSTs from genetically engineered mouse models (P0-GGFß3 and P0-GGFß3;Trp53+/- mice); multiple erbB4 splice variants that differ in their ability to activate PI3 kinase and nuclear signaling were present in MPNST-derived cell lines. Erbb4-null MPNST cells demonstrated decreased proliferation and survival and altered morphology relative to non-ablated controls. Orthotopic allografts of Erbb4-null cells were significantly smaller than controls, with reduced proliferation, survival and vascularization. ERBB4 knockdown in human MPNST cells similarly inhibited DNA synthesis and viability. Although we have previously shown that broad-spectrum erbB inhibitors inhibit Ras activation, Erbb4 ablation did not affect Ras activation, suggesting that erbB4 drives neoplasia via non-Ras dependent pathways. An analysis of 43 candidate kinases identified multiple NRG1ß-responsive and erbB4-dependent signaling cascades including the PI3K, WNK1, STAT3, STAT5 and phospholipase-Cγ pathways. Although WNK1 inhibition did not alter proliferation, inhibition of STAT3, STAT5 and phospholipase-Cγ markedly reduced proliferation. CONCLUSIONS: ErbB4 promotes MPNST growth by activating key non-Ras dependent signaling cascades including the STAT3, STAT5 and phospholipase-Cγ pathways. ErbB4 and its effector pathways are thus potentially useful therapeutic targets in MPNSTs.


Asunto(s)
Neoplasias de la Vaina del Nervio/patología , Receptor ErbB-4/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Fosfolipasa C gamma/metabolismo , Fosforilación , Receptor ErbB-4/genética , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT5/metabolismo , Transducción de Señal , Proteínas ras/metabolismo
2.
Am J Pathol ; 187(1): 2-3, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27993238

RESUMEN

This Editorial highlights the value of The American Journal of Pathology beyond normal metrics.


Asunto(s)
Bibliometría , Publicaciones Periódicas como Asunto , Edición/economía
4.
Am J Pathol ; 186(10): 2518, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27560710

RESUMEN

This Editorial introduces the Lung Ontogeny and Injury Theme Issue, which provides critical insights into lung development, injury, and repair as well as key pulmonary diseases.


Asunto(s)
Lesión Pulmonar/terapia , Pulmón/embriología , Fibroblastos/fisiología , Hernias Diafragmáticas Congénitas/patología , Humanos , Organogénesis , Pericitos/fisiología , Células Madre/fisiología , Cicatrización de Heridas
5.
Am J Pathol ; 185(1): 2-3, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25529794

RESUMEN

This Editorial introduces readers to the US National Institutes of Health's Proposed Principles and Guidelines for Reporting Preclinical Research.


Asunto(s)
Guías como Asunto , Ciencia/métodos , Ciencia/tendencias , Políticas Editoriales , Humanos , National Institutes of Health (U.S.) , Patología/normas , Publicaciones , Reproducibilidad de los Resultados , Estados Unidos
6.
Am J Pathol ; 185(3): 600-1, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25701882

RESUMEN

This editorial discusses the rise of computational pathology as a major driver of experimental pathology research.


Asunto(s)
Patología , Biología Computacional , Humanos , Investigación
7.
Am J Pathol ; 182(3): 646-67, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23321323

RESUMEN

Patients with neurofibromatosis type 1 (NF1) develop benign plexiform neurofibromas that frequently progress to become malignant peripheral nerve sheath tumors (MPNSTs). A genetically engineered mouse model that accurately models plexiform neurofibroma-MPNST progression in humans would facilitate identification of somatic mutations driving this process. We previously reported that transgenic mice overexpressing the growth factor neuregulin-1 in Schwann cells (P(0)-GGFß3 mice) develop MPNSTs. To determine whether P(0)-GGFß3 mice accurately model human neurofibroma-MPNST progression, cohorts of these animals were monitored through death and were necropsied; 94% developed multiple neurofibromas, with 70% carrying smaller numbers of MPNSTs. Nascent MPNSTs were identified within neurofibromas, suggesting that these sarcomas arise from neurofibromas. Although neurofibromin expression was maintained, P(0)-GGFß3 MPNSTs exhibited Ras hyperactivation, as in human NF1-associated MPNSTs. P(0)-GGFß3 MPNSTs also exhibited abnormalities in the p16(INK4A)-cyclin D/CDK4-Rb and p19(ARF)-Mdm-p53 pathways, analogous to their human counterparts. Array comparative genomic hybridization (CGH) demonstrated reproducible chromosomal alterations in P(0)-GGFß3 MPNST cells (including universal chromosome 11 gains) and focal gains and losses affecting 39 neoplasia-associated genes (including Pten, Tpd52, Myc, Gli1, Xiap, and Bbc3/PUMA). Array comparative genomic hybridization also identified recurrent focal copy number variations affecting genes not previously linked to neurofibroma or MPNST pathogenesis. We conclude that P(0)-GGFß3 mice represent a robust model of neurofibroma-MPNST progression useful for identifying novel genes driving neurofibroma and MPNST pathogenesis.


Asunto(s)
Transformación Celular Neoplásica/genética , Cromosomas de los Mamíferos/genética , Variaciones en el Número de Copia de ADN/genética , Progresión de la Enfermedad , Neoplasias de la Vaina del Nervio/patología , Neurregulina-1/metabolismo , Neurofibroma/patología , Animales , Emparejamiento Base/genética , Biomarcadores de Tumor/metabolismo , Ciclo Celular , Transformación Celular Neoplásica/patología , Hibridación Genómica Comparativa , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neoplasias de la Vaina del Nervio/genética , Neurofibroma/genética , Neurofibromina 1/metabolismo , Sistema Nervioso Periférico/metabolismo , Sistema Nervioso Periférico/patología , Transducción de Señal/genética , Proteínas ras/metabolismo
8.
Acta Neuropathol ; 127(4): 573-91, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24232507

RESUMEN

Malignant peripheral nerve sheath tumors (MPNSTs) are Schwann cell-derived malignancies that arise from plexiform neurofibromas in patients with mutation of the neurofibromin 1 (NF1) gene. We have shown that the growth factor neuregulin-1 (NRG1) also contributes to human neurofibroma and MPNST pathogenesis and that outbred C57BL/6J × SJL/J transgenic mice overexpressing NRG1 in Schwann cells (P0-GGFß3 mice) recapitulate the process of neurofibroma-MPNST progression. However, it is unclear whether NRG1 acts predominantly within NF1-regulated signaling cascades or instead activates other essential cascades that cooperate with NF1 loss to promote tumorigenesis. We now report that tumorigenesis is suppressed in inbred P0-GGFß3 mice on a C57BL/6J background. To determine whether NRG1 overexpression interacts with reduced Nf1 or Trp53 gene dosage to "unmask" tumorigenesis in these animals, we followed cohorts of inbred P0-GGFß3;Nf1+/−, P0-GGFß3;Trp53+/− and control (P0-GGFß3, Nf1+/− and Trp53+/−) mice for 1 year. We found no reduction in survival or tumors in control and P0-GGFß3;Nf1+/− mice. In contrast, P0-GGFß3;Trp53+/− mice died on average at 226 days, with MPNSTs present in 95 % of these mice. MPNSTs in inbred P0-GGFß3;Trp53+/− mice arose de novo from micro-MPNSTs that uniformly develop intraganglionically. These micro-MPNSTs are of lower grade (WHO grade II-III) than the major MPNSTs (WHO grade III-IV); array comparative genomic hybridization showed that lower grade MPNSTs also had fewer genomic abnormalities. Thus, P0-GGFß3;Trp53+/− mice represent a novel model of low- to high-grade MPNST progression. We further conclude that NRG1 promotes peripheral nervous system neoplasia predominantly via its effects on the signaling cascades affected by Nf1 loss.


Asunto(s)
Expresión Génica , Haploinsuficiencia/genética , Neurregulina-1/metabolismo , Neoplasias del Nervio Óptico/genética , Neoplasias del Nervio Óptico/patología , Proteína p53 Supresora de Tumor/genética , Animales , Desmina/metabolismo , Humanos , Etiquetado Corte-Fin in Situ , Antígeno Ki-67/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación/genética , Proteína P0 de la Mielina/genética , Proteína P0 de la Mielina/metabolismo , Neurofibroma/genética , Subunidad beta de la Proteína de Unión al Calcio S100/metabolismo , Médula Espinal/metabolismo , Médula Espinal/patología , Raíces Nerviosas Espinales/metabolismo , Raíces Nerviosas Espinales/patología
9.
J Histochem Cytochem ; 71(5): 289-290, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37199234

RESUMEN

This commentary reflects on the significance and impact of the highly cited companion article that was published in the Journal of Histochemistry and Cytochemistry in 1997 (Gijlswijk RPM et al. Fluorochrome-labeled Tyramides: Use in Immunocytochemistry and Fluorescence In Situ Hybridization. Journal of Histochemistry & Cytochemistry. 1997;45(3):375-382).


Asunto(s)
Colorantes Fluorescentes , Tiramina , Inmunohistoquímica , Hibridación Fluorescente in Situ , Histocitoquímica
10.
Nat Med ; 11(7): 797-803, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15965483

RESUMEN

We describe the generation and characterization of the first inducible 'fatless' model system, the FAT-ATTAC mouse (fat apoptosis through targeted activation of caspase 8). This transgenic mouse develops identically to wild-type littermates. Apoptosis of adipocytes can be induced at any developmental stage by administration of a FK1012 analog leading to the dimerization of a membrane-bound, adipocyte-specific caspase 8-FKBP fusion protein. Within 2 weeks of dimerizer administration, FAT-ATTAC mice show near-knockout levels of circulating adipokines and markedly reduced levels of adipose tissue. FAT-ATTAC mice are glucose intolerant, have diminished basal and endotoxin-stimulated systemic inflammation, are less responsive to glucose-stimulated insulin secretion and show increased food intake independent of the effects of leptin. Most importantly, we show that functional adipocytes can be recovered upon cessation of treatment, allowing the study of adipogenesis in vivo, as well as a detailed examination of the importance of the adipocyte in the regulation of multiple physiological functions and pathological states.


Asunto(s)
Tejido Adiposo/patología , Apoptosis/fisiología , Caspasas/metabolismo , Lipodistrofia/metabolismo , Tacrolimus/análogos & derivados , Adipocitos/efectos de los fármacos , Adipocitos/patología , Tejido Adiposo/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Caspasa 8 , Caspasas/genética , Dimerización , Ingestión de Alimentos , Activación Enzimática , Intolerancia a la Glucosa/genética , Inflamación/genética , Insulina/metabolismo , Secreción de Insulina , Leptina/deficiencia , Leptina/genética , Leptina/metabolismo , Lipopolisacáridos , Ratones , Ratones Transgénicos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Tacrolimus/farmacología , Proteínas de Unión a Tacrolimus/genética , Proteínas de Unión a Tacrolimus/metabolismo
11.
J Biol Chem ; 285(14): 10497-507, 2010 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-20123985

RESUMEN

Macroautophagy (autophagy) is a process wherein bulk cytosolic proteins and damaged organelles are sequestered and degraded via the lysosome. Alterations in autophagy-associated proteins have been shown to cause neural tube closure defects, neurodegeneration, and tumor formation. Normal lysosome function is critical for autophagy completion and when altered may lead to an accumulation of autophagic vacuoles (AVs) and caspase activation. The tumor suppressor p53 is highly expressed in neural precursor cells (NPCs) and has an important role in the regulation of both autophagy and apoptosis. We hypothesized that altered lysosome function would lead to NPC death via an interaction between autophagy- and apoptosis-associated proteins. To test our hypothesis, we utilized FGF2-expanded NPCs and the neural stem cell line, C17.2, in combination with the lysosomotropic agent chloroquine (CQ) and the vacuolar ATPase inhibitor bafilomycin A1 (Baf A1). Both CQ and Baf A1 caused concentration- and time-dependent AV accumulation, p53 phosphorylation, increased damage regulator autophagy modulator levels, caspase-3 activation, and cell death. Short hairpin RNA knockdown of Atg7, but not Beclin1, expression significantly inhibited CQ- and Baf A1-induced cell death, indicating that Atg7 is an upstream mediator of lysosome dysfunction-induced cell death. Cell death and/or caspase-3 activation was also attenuated by protein synthesis inhibition, p53 deficiency, or Bax deficiency, indicating involvement of the intrinsic apoptotic death pathway. In contrast to lysosome dysfunction, starvation-induced AV accumulation was inhibited by either Atg7 or Beclin1 knockdown, and Atg7 knockdown had no effect on starvation-induced death. These findings indicate that Atg7- and Beclin1-induced autophagy plays a cytoprotective role during starvation but that Atg7 has a unique pro-apoptotic function in response to lysosome dysfunction.


Asunto(s)
Apoptosis , Cerebelo/metabolismo , Lisosomas/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Neuronas/metabolismo , Células Madre/metabolismo , Animales , Antifúngicos/farmacología , Antimaláricos/farmacología , Autofagia , Proteína 7 Relacionada con la Autofagia , Western Blotting , Caspasas/metabolismo , Cerebelo/citología , Cloroquina/farmacología , Factor 2 de Crecimiento de Fibroblastos , Técnica del Anticuerpo Fluorescente , Lisosomas/patología , Macrólidos/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/antagonistas & inhibidores , Proteínas Asociadas a Microtúbulos/genética , Neuronas/citología , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/citología , Proteína p53 Supresora de Tumor/fisiología , Proteína X Asociada a bcl-2/fisiología
12.
Am J Pathol ; 176(3): 1065-71, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20150434

RESUMEN

Cell survival depends on a complex interplay of processes including homeostatic pathways and cytoprotective mechanisms. Autophagy is a physiological process involved in the basal turnover of long-lived proteins and organelles and also comprises an integral part of the cellular stress response. The significance of autophagy in regulating neural cell fate has become increasingly recognized and agents targeting autophagy are of increasing therapeutic interest. This review summarizes the recent expansion of our understanding of the scope of this physiological process.


Asunto(s)
Autofagia , Encéfalo/patología , Envejecimiento/patología , Animales , Humanos , Neoplasias/patología , Enfermedades Neurodegenerativas/patología , Estrés Fisiológico
13.
Am J Pathol ; 176(6): 2695-706, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20472884

RESUMEN

Several different deletions within the N-terminal tail of the prion protein (PrP) induce massive neuronal death when expressed in transgenic mice. This toxicity is dose-dependently suppressed by coexpression of full-length PrP, suggesting that it results from subversion of a normal physiological activity of cellular PrP. We performed a combined biochemical and morphological analysis of Tg(DeltaCR) mice, which express PrP carrying a 21-aa deletion (residues 105-125) within a highly conserved region of the protein. Death of cerebellar granule neurons in Tg(DeltaCR) mice is not accompanied by activation of either caspase-3 or caspase-8 or by increased levels of the autophagy marker, LC3-II. In electron micrographs, degenerating granule neurons displayed a unique morphology characterized by heterogeneous condensation of the nuclear matrix without formation of discrete chromatin masses typical of neuronal apoptosis. Our data demonstrate that perturbations in PrP functional activity induce a novel, nonapoptotic, nonautophagic form of neuronal death whose morphological features are reminiscent of those associated with excitotoxic stress.


Asunto(s)
Muerte Celular/fisiología , Cerebelo/citología , Neuronas/fisiología , Proteínas PrPC/toxicidad , Animales , Apoptosis/fisiología , Autofagia/fisiología , Biomarcadores/metabolismo , Caspasa 3/metabolismo , Caspasa 8/metabolismo , Forma de la Célula , Activación Enzimática , Ratones , Ratones Endogámicos CBA , Ratones Mutantes Neurológicos , Ratones Transgénicos , Neuronas/patología , Neuronas/ultraestructura , Proteínas PrPC/genética , Priones/genética , Priones/metabolismo
14.
Cells ; 10(7)2021 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-34209035

RESUMEN

Glioblastoma is a high-grade glial neoplasm with a patient survival of 12-18 months. Therefore, the identification of novel therapeutic targets is an urgent need. RAB38 is a GTPase protein implicated in regulating cell proliferation and survival in tumors. The role of RAB38 in glioblastoma is relatively unexplored. Here, we test the hypothesis that RAB38 regulates glioblastoma growth using human glioblastoma cell lines. We found that genetic interference of RAB38 resulted in a decrease in glioblastoma growth through inhibition of proliferation and cell death induction. Transcriptome analysis showed that RAB38 silencing leads to changes in genes related to mitochondrial metabolism and intrinsic apoptosis (e.g., Bcl-xL). Consistently, rescue experiments demonstrated that loss of RAB38 causes a reduction in glioblastoma viability through downregulation of Bcl-xL. Moreover, RAB38 knockdown inhibited both glycolysis and oxidative phosphorylation. Interference with RAB38 enhanced cell death induced by BH3-mimetics. RAB38 antagonists are under development, but not yet clinically available. We found that FDA-approved statins caused a rapid reduction in RAB38 protein levels, increased cell death, and phenocopied some of the molecular changes elicited by loss of RAB38. In summary, our findings suggest that RAB38 is a potential therapeutic target for glioblastoma treatment.


Asunto(s)
Metabolismo Energético , Glioblastoma/metabolismo , Glioblastoma/patología , Proteínas de Unión al GTP rab/metabolismo , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Biomarcadores de Tumor/metabolismo , Ciclo Celular/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Humanos , Mitocondrias/metabolismo , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Pronóstico , Proteínas Proto-Oncogénicas c-myc/metabolismo , Simvastatina/farmacología , Proteína bcl-X/metabolismo
15.
Sci Rep ; 11(1): 5690, 2021 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-33707600

RESUMEN

Malignant peripheral nerve sheath tumors (MPNSTs) are aggressive Schwann cell-derived neoplasms that occur sporadically or in patients with neurofibromatosis type 1 (NF1). Preclinical research on sporadic MPNSTs has been limited as few cell lines exist. We generated and characterized a new sporadic MPNST cell line, 2XSB, which shares the molecular and genomic features of the parent tumor. These cells have a highly complex karyotype with extensive chromothripsis. 2XSB cells show robust invasive 3-dimensional and clonogenic culture capability and form solid tumors when xenografted into immunodeficient mice. High-density single nucleotide polymorphism array and whole exome sequencing analyses indicate that, unlike NF1-associated MPNSTs, 2XSB cells have intact, functional NF1 alleles with no evidence of mutations in genes encoding components of Polycomb Repressor Complex 2. However, mutations in other genes implicated in MPNST pathogenesis were identified in 2XSB cells including homozygous deletion of CDKN2A and mutations in TP53 and PTEN. We also identified mutations in genes not previously associated with MPNSTs but associated with the pathogenesis of other human cancers. These include DNMT1, NUMA1, NTRK1, PDE11A, CSMD3, LRP5 and ACTL9. This sporadic MPNST-derived cell line provides a useful tool for investigating the biology and potential treatment regimens for sporadic MPNSTs.


Asunto(s)
Genoma Humano , Neoplasias de la Vaina del Nervio/genética , Neoplasias de la Vaina del Nervio/patología , Secuencias Repetitivas de Ácidos Nucleicos , Línea Celular Tumoral , Proliferación Celular , Dosificación de Gen , Genes Relacionados con las Neoplasias , Humanos , Cariotipificación , Mutación/genética , Polimorfismo de Nucleótido Simple/genética , Secuencias Repetitivas de Ácidos Nucleicos/genética , Secuenciación del Exoma
16.
Acad Pathol ; 8: 23742895211006818, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34013020

RESUMEN

The COVID-19 pandemic, caused by severe acute respiratory syndrome coronavirus 2, created an unprecedented need for comprehensive laboratory testing of populations, in order to meet the needs of medical practice and to guide the management and functioning of our society. With the greater New York metropolitan area as an epicenter of this pandemic beginning in March 2020, a consortium of laboratory leaders from the assembled New York academic medical institutions was formed to help identify and solve the challenges of deploying testing. This report brings forward the experience of this consortium, based on the real-world challenges which we encountered in testing patients and in supporting the recovery effort to reestablish the health care workplace. In coordination with the Greater New York Hospital Association and with the public health laboratory of New York State, this consortium communicated with state leadership to help inform public decision-making addressing the crisis. Through the length of the pandemic, the consortium has been a critical mechanism for sharing experience and best practices in dealing with issues including the following: instrument platforms, sample sources, test performance, pre- and post-analytical issues, supply chain, institutional testing capacity, pooled testing, biospecimen science, and research. The consortium also has been a mechanism for staying abreast of state and municipal policies and initiatives, and their impact on institutional and laboratory operations. The experience of this consortium may be of value to current and future laboratory professionals and policy-makers alike, in dealing with major events that impact regional laboratory services.

17.
J Neurochem ; 114(4): 1193-204, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20534000

RESUMEN

We have shown previously that the plecomacrolide antibiotics bafilomycin A1 and B1 significantly attenuate cerebellar granule neuron death resulting from agents that disrupt lysosome function. To further characterize bafilomycin-mediated cytoprotection, we examined its ability to attenuate the death of naïve and differentiated neuronal SH-SY5Y human neuroblastoma cells from agents that induce lysosome dysfunction in vitro, and from in vivo dopaminergic neuron death in C. elegans. Low-dose bafilomycin significantly attenuated SH-SY5Y cell death resulting from treatment with chloroquine, hydroxychloroquine amodiaquine and staurosporine. Bafilomycin also attenuated the chloroquine-induced reduction in processing of cathepsin D, the principal lysosomal aspartic acid protease, to its mature 'active' form. Chloroquine induced autophagic vacuole accumulation and inhibited autophagic flux, effects that were attenuated upon treatment with bafilomycin and were associated with a significant decrease in chloroquine-induced accumulation of detergent-insoluble alpha-synuclein oligomers. In addition, bafilomycin significantly and dose-dependently attenuated dopaminergic neuron death in C. elegans resulting from in vivo over-expression of human wild-type alpha-synuclein. Together, our findings suggest that low-dose bafilomycin is cytoprotective in part through its maintenance of the autophagy-lysosome pathway, and underscores its therapeutic potential for treating Parkinson's disease and other neurodegenerative diseases that exhibit disruption of protein degradation pathways and accumulation of toxic protein species.


Asunto(s)
Autofagia/efectos de los fármacos , Citoprotección/efectos de los fármacos , Lisosomas/efectos de los fármacos , Macrólidos/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Autofagia/fisiología , Caenorhabditis elegans/efectos de los fármacos , Línea Celular Tumoral , Citoprotección/fisiología , Progresión de la Enfermedad , Humanos , Lisosomas/fisiología , Degeneración Nerviosa/tratamiento farmacológico , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Neuronas/efectos de los fármacos , Neuronas/metabolismo
18.
JCO Precis Oncol ; 4: 1038-1048, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35050771

RESUMEN

PURPOSE: The routine use of large next-generation sequencing (NGS) pan-cancer panels is required to identify the increasing number of, but often uncommon, actionable alterations to guide therapy. Inconsistent coverage and variable payment is hindering NGS adoption into clinical practice. A review of test utilization, clinical utility, coverage, and reimbursement was conducted in a cohort of patients diagnosed with high-risk cancer who received pan-cancer panel testing as part of their clinical care. MATERIALS AND METHODS: The Columbia Combined Cancer Panel (CCCP), a 467-gene panel designed to detect DNA variations in solid and liquid tumors, was performed in the Laboratory of Personalized Genomic Medicine at Columbia University Irving Medical Center. Utilization was characterized at test order. Results were reviewed by a molecular pathologist, followed by a multidisciplinary molecular tumor board where clinical utility was classified by consensus. Reimbursement was reviewed after payers provided final coverage decisions. RESULTS: NGS was performed on 359 high-risk tumors from 349 patients. Reimbursement data were available for 246 cases. The most common reason providers ordered CCCP testing was for patients diagnosed with a treatment-resistant or recurrent tumor (n = 214; 61%). Findings were clinically impactful for 229 cases (64%). Molecular alterations that may inform future therapy in the event of progression or relapse were found in 42% of cases, and a targeted therapy was initiated in 23 cases (6.6%). The majority of tests were denied coverage by payers (n = 190; 77%). On average, insurers reimbursed 10.75% of the total NGS service charge. CONCLUSION: CCCP testing identified clinically impactful alterations in 64% of cases. Limited coverage and low reimbursement remain barriers, and broader reimbursement policies are needed to adopt pan-cancer NGS testing that benefits patients into clinical practice.

19.
Artículo en Inglés | MEDLINE | ID: mdl-32839179

RESUMEN

We report a case of a slow-growing, diffuse, infiltrating glioma in the right brainstem of a 9-yr-old boy. The tumor was negative by immunohistochemical staining for histone H3 K27M, BRAF V600E, and IDH1 R132H mutations. Fluorescence in situ hybridization did not reveal a BRAF duplication. Genomic profiling of the tumor, by DNA methylation array and cancer whole-exome and transcriptome sequencing, was performed. This analysis showed copy-number alterations, including gains of several chromosomes. In addition, a novel fusion involving the first 17 exons of FGFR2 fused to exon 2 of VPS35 was identified. This novel fusion is predicted to result in activation of fibroblast growth factor receptor (FGFR) signaling and is potentially targetable using FGFR inhibitors. This tumor expands the spectrum of pediatric diffuse gliomas.


Asunto(s)
Glioma/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Proteínas de Transporte Vesicular/genética , Neoplasias Encefálicas/genética , Niño , Humanos , Hibridación Fluorescente in Situ/métodos , Masculino , Mutación/genética , Secuenciación del Exoma/métodos
20.
J Neurosci ; 28(6): 1490-7, 2008 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-18256270

RESUMEN

Previous studies have shown that caspases and Apaf-1 are required for the normal programmed cell death (PCD) in vivo of immature postmitotic neurons and mitotically active neuronal precursor cells. In contrast, caspase activity is not necessary for the normal PCD of more mature postmitotic neurons that are establishing synaptic connections. Although normally these cells use caspases for PCD, in the absence of caspase activity these neurons undergo a distinct nonapoptotic type of degeneration. We examined the survival of these more mature postmitotic neuronal populations in mice in which Apaf-1 has been genetically deleted and find that they exhibit quantitatively normal PCD of developing postmitotic neurons. We next characterized the morphological mode of PCD in these mice and show that the neurons degenerate by a caspase-independent, nonapoptotic pathway that involves autophagy. However, autophagy does not appear to be involved in the normal PCD of postmitotic neurons in which caspases and Apaf-1 are present and functional because quantitatively normal neuronal PCD occurred in the absence of a key gene required for autophagy (ATG7). Finally, we examined the possible role of another caspase-independent type of neuronal PCD involving the apoptosis-inducing factor (AIF). Mice deficient in AIF also exhibit quantitatively normal PCD of postmitotic neurons after caspase inhibition. Together, these data indicate that, when key components of the type 1 apoptotic pathway (i.e., caspases and Apaf-1) are perturbed in vivo, developing postmitotic neurons nonetheless undergo quantitatively normal PCD by a caspase-independent pathway involving autophagy and not requiring AIF.


Asunto(s)
Apoptosis , Factor Apoptótico 1 Activador de Proteasas/deficiencia , Factor Apoptótico 1 Activador de Proteasas/genética , Autofagia , Caspasas/fisiología , Mitosis , Neuronas/fisiología , Animales , Animales Recién Nacidos , Apoptosis/genética , Factor Apoptótico 1 Activador de Proteasas/fisiología , Autofagia/genética , Muerte Celular/genética , Supervivencia Celular/genética , Femenino , Masculino , Ratones , Ratones Noqueados , Mitosis/genética , Neuronas/enzimología , Transducción de Señal/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA