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










Base de datos
Intervalo de año de publicación
3.
Autophagy ; 18(8): 2003-2005, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35343362

RESUMEN

Neurons depend on macroautophagy/autophagy to maintain cellular homeostasis, and loss of autophagy leads to neurodegeneration. To better understand the role of basal autophagy in neurons, we enriched autophagic vesicles from healthy adult mouse brain and performed mass spectrometry to identify cargos cleared by autophagy. We found that synaptic and mitochondrial proteins comprise nearly half of the unique AV cargos identified in brain. Similarly, synaptic and mitochondrial proteins are major cargos for basal autophagy in neurons. Strikingly, we noted a specific enrichment of mitochondrial nucleoids within neuronal autophagosomes, which occurs through a mechanism distinct from damage-associated mitophagy. Here, we discuss the implications of these findings for our understanding of homeostatic mechanisms in neurons and how the age-dependent decline of autophagy in neurons may contribute to the onset or progression of neurodegenerative disease.


Asunto(s)
Autofagia , Enfermedades Neurodegenerativas , Animales , Autofagia/fisiología , Homeostasis , Ratones , Proteínas Mitocondriales/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Neuronas/metabolismo , Proteómica
4.
Neuron ; 110(6): 967-976.e8, 2022 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-35051374

RESUMEN

Neurons depend on autophagy to maintain cellular homeostasis, and defects in autophagy are pathological hallmarks of neurodegenerative disease. To probe the role of basal autophagy in the maintenance of neuronal health, we isolated autophagic vesicles from mouse brain tissue and used proteomics to identify the major cargos engulfed within autophagosomes, validating our findings in rodent primary and human iPSC-derived neurons. Mitochondrial proteins were identified as a major cargo in the absence of mitophagy adaptors such as OPTN. We found that nucleoid-associated proteins are enriched compared with other mitochondrial components. In the axon, autophagic engulfment of nucleoid-enriched mitochondrial fragments requires the mitochondrial fission machinery Drp1. We proposed that localized Drp1-dependent fission of nucleoid-enriched fragments in proximity to the sites of autophagosome biogenesis enhances their capture. The resulting efficient autophagic turnover of nucleoids may prevent accumulation of mitochondrial DNA in the neuron, thus mitigating activation of proinflammatory pathways that contribute to neurodegeneration.


Asunto(s)
Autofagosomas , Enfermedades Neurodegenerativas , Animales , Autofagosomas/metabolismo , Autofagia/fisiología , Encéfalo , Ratones , Enfermedades Neurodegenerativas/metabolismo , Neuronas/metabolismo
5.
Genes Dev ; 35(13-14): 963-975, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34168038

RESUMEN

Autophagy inhibitors are currently being evaluated in clinical trials for the treatment of diverse cancers, largely due to their ability to impede tumor cell survival and metabolic adaptation. More recently, there is growing interest in whether and how modulating autophagy in the host stroma influences tumorigenesis. Fibroblasts play prominent roles in cancer initiation and progression, including depositing type 1 collagen and other extracellular matrix (ECM) components, thereby stiffening the surrounding tissue to enhance tumor cell proliferation and survival, as well as secreting cytokines that modulate angiogenesis and the immune microenvironment. This constellation of phenotypes, pathologically termed desmoplasia, heralds poor prognosis and reduces patient survival. Using mouse mammary cancer models and syngeneic transplantation assays, we demonstrate that genetic ablation of stromal fibroblast autophagy significantly impedes fundamental elements of the stromal desmoplastic response, including collagen and proinflammatory cytokine secretion, extracellular matrix stiffening, and neoangiogenesis. As a result, autophagy in stromal fibroblasts is required for mammary tumor growth in vivo, even when the cancer cells themselves remain autophagy-competent . We propose the efficacy of autophagy inhibition is shaped by this ability of host stromal fibroblast autophagy to support tumor desmoplasia.


Asunto(s)
Células del Estroma , Microambiente Tumoral , Animales , Autofagia/genética , Línea Celular Tumoral , Transformación Celular Neoplásica/patología , Fibroblastos/metabolismo , Humanos , Ratones , Microambiente Tumoral/genética
6.
J Cell Sci ; 134(11)2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-34096604

RESUMEN

In Saccharomyces cerevisiae, the selective autophagic degradation of mitochondria, termed mitophagy, is critically regulated by the adapter protein Atg32. Despite our knowledge about the molecular mechanisms by which Atg32 controls mitophagy, its physiological roles in yeast survival and fitness remains less clear. Here, we demonstrate a requirement for Atg32 in promoting spermidine production during respiratory growth and heat-induced mitochondrial stress. During respiratory growth, mitophagy-deficient yeast exhibit profound heat-stress induced defects in growth and viability due to impaired biosynthesis of spermidine and its biosynthetic precursor S-adenosyl methionine. Moreover, spermidine production is crucial for the induction of cytoprotective nitric oxide (NO) during heat stress. Hence, the re-addition of spermidine to Atg32 mutant yeast is sufficient to both enhance NO production and restore respiratory growth during heat stress. Our findings uncover a previously unrecognized physiological role for yeast mitophagy in spermidine metabolism and illuminate new interconnections between mitophagy, polyamine biosynthesis and NO signaling.


Asunto(s)
Mitofagia , Proteínas de Saccharomyces cerevisiae , Autofagia/genética , Proteínas Relacionadas con la Autofagia/genética , Respuesta al Choque Térmico/genética , Óxido Nítrico , Receptores Citoplasmáticos y Nucleares , Proteínas de Saccharomyces cerevisiae/genética , Espermidina
7.
J Cell Biol ; 220(7)2021 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-34014261

RESUMEN

Autophagy is a degradative pathway required to maintain homeostasis. Neuronal autophagosomes form constitutively at the axon terminal and mature via lysosomal fusion during dynein-mediated transport to the soma. How the dynein-autophagosome interaction is regulated is unknown. Here, we identify multiple dynein effectors on autophagosomes as they transit along the axons of primary neurons. In the distal axon, JIP1 initiates autophagosomal transport. Autophagosomes in the mid-axon require HAP1 and Huntingtin. We find that HAP1 is a dynein activator, binding the dynein-dynactin complex via canonical and noncanonical interactions. JIP3 is on most axonal autophagosomes, but specifically regulates the transport of mature autolysosomes. Inhibiting autophagosomal transport disrupts maturation, and inhibiting autophagosomal maturation perturbs the association and function of dynein effectors; thus, maturation and transport are tightly linked. These results reveal a novel maturation-based dynein effector handoff on neuronal autophagosomes that is key to motility, cargo degradation, and the maintenance of axonal health.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Autofagosomas/genética , Axones/metabolismo , Proteína Huntingtina/genética , Proteínas del Tejido Nervioso/genética , Autofagia/genética , Transporte Axonal/genética , Complejo Dinactina/genética , Dineínas/genética , Homeostasis , Humanos , Lisosomas/genética , Proteínas Asociadas a Microtúbulos/genética , Neuronas/metabolismo , Neuronas/patología , Fagosomas/genética
8.
Curr Biol ; 31(10): 2140-2154.e6, 2021 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-33765413

RESUMEN

Parkinson's disease-causing mutations in the leucine-rich repeat kinase 2 (LRRK2) gene hyperactivate LRRK2 kinase activity and cause increased phosphorylation of Rab GTPases, important regulators of intracellular trafficking. We found that the most common LRRK2 mutation, LRRK2-G2019S, dramatically reduces the processivity of autophagosome transport in neurons in a kinase-dependent manner. This effect was consistent across an overexpression model, neurons from a G2019S knockin mouse, and human induced pluripotent stem cell (iPSC)-derived neurons gene edited to express the G2019S mutation, and the effect was reversed by genetic or pharmacological inhibition of LRRK2. Furthermore, LRRK2 hyperactivation induced by overexpression of Rab29, a known activator of LRRK2 kinase, disrupted autophagosome transport to a similar extent. Mechanistically, we found that hyperactive LRRK2 recruits the motor adaptor JNK-interacting protein 4 (JIP4) to the autophagosomal membrane, inducing abnormal activation of kinesin that we propose leads to an unproductive tug of war between anterograde and retrograde motors. Disruption of autophagosome transport correlated with a significant defect in autophagosome acidification, suggesting that the observed transport deficit impairs effective degradation of autophagosomal cargo in neurons. Our results robustly link increased LRRK2 kinase activity to defects in autophagosome transport and maturation, further implicating defective autophagy in the pathogenesis of Parkinson's disease.


Asunto(s)
Autofagosomas , Autofagia , Transporte Axonal , Células Madre Pluripotentes Inducidas , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Animales , Autofagosomas/metabolismo , Autofagia/genética , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Ratones , Mutación , Enfermedad de Parkinson
9.
Neuron ; 107(1): 6-8, 2020 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-32645308

RESUMEN

In this issue of Neuron, Orr et al. (2020) identify an evolutionarily conserved mechanism of presynaptic homeostatic plasticity induced by ALS-like motor neuron degeneration, which maintains excitatory potentials until a threshold of synaptic loss is reached. Past this tipping point, disease onset progresses rapidly.


Asunto(s)
Plasticidad Neuronal , Neuroprotección , Esclerosis Amiotrófica Lateral , Animales , Progresión de la Enfermedad , Homeostasis , Ratones
10.
Commun Biol ; 3(1): 388, 2020 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-32681145

RESUMEN

Autophagy promotes protein degradation, and therefore has been proposed to maintain amino acid pools to sustain protein synthesis during metabolic stress. To date, how autophagy influences the protein synthesis landscape in mammalian cells remains unclear. Here, we utilize ribosome profiling to delineate the effects of genetic ablation of the autophagy regulator, ATG12, on translational control. In mammalian cells, genetic loss of autophagy does not impact global rates of cap dependent translation, even under starvation conditions. Instead, autophagy supports the translation of a subset of mRNAs enriched for cell cycle control and DNA damage repair. In particular, we demonstrate that autophagy enables the translation of the DNA damage repair protein BRCA2, which is functionally required to attenuate DNA damage and promote cell survival in response to PARP inhibition. Overall, our findings illuminate that autophagy impacts protein translation and shapes the protein landscape.


Asunto(s)
Autofagia , Regulación de la Expresión Génica , Biosíntesis de Proteínas , ARN Mensajero/metabolismo , Ribosomas/metabolismo , Animales , Autofagia/fisiología , Proteína 12 Relacionada con la Autofagia/metabolismo , Proteína BRCA2/metabolismo , Daño del ADN , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , ARN Mensajero/fisiología , Ribosomas/fisiología
11.
Nat Cell Biol ; 22(2): 187-199, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31932738

RESUMEN

Traditionally viewed as an autodigestive pathway, autophagy also facilitates cellular secretion; however, the mechanisms underlying these processes remain unclear. Here, we demonstrate that components of the autophagy machinery specify secretion within extracellular vesicles (EVs). Using a proximity-dependent biotinylation proteomics strategy, we identify 200 putative targets of LC3-dependent secretion. This secretome consists of a highly interconnected network enriched in RNA-binding proteins (RBPs) and EV cargoes. Proteomic and RNA profiling of EVs identifies diverse RBPs and small non-coding RNAs requiring the LC3-conjugation machinery for packaging and secretion. Focusing on two RBPs, heterogeneous nuclear ribonucleoprotein K (HNRNPK) and scaffold-attachment factor B (SAFB), we demonstrate that these proteins interact with LC3 and are secreted within EVs enriched with lipidated LC3. Furthermore, their secretion requires the LC3-conjugation machinery, neutral sphingomyelinase 2 (nSMase2) and LC3-dependent recruitment of factor associated with nSMase2 activity (FAN). Hence, the LC3-conjugation pathway controls EV cargo loading and secretion.


Asunto(s)
Autofagosomas/metabolismo , Autofagia/genética , Vesículas Extracelulares/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Proteínas de Unión al ARN/genética , Proteínas Adaptadoras del Transporte Vesicular/deficiencia , Proteínas Adaptadoras del Transporte Vesicular/genética , Animales , Autofagosomas/química , Proteína 7 Relacionada con la Autofagia/deficiencia , Proteína 7 Relacionada con la Autofagia/genética , Proteínas Relacionadas con la Autofagia/deficiencia , Proteínas Relacionadas con la Autofagia/genética , Transporte Biológico , Biotinilación , Vesículas Extracelulares/química , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Células HEK293 , Ribonucleoproteína Heterogénea-Nuclear Grupo K/genética , Ribonucleoproteína Heterogénea-Nuclear Grupo K/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lisosomas/química , Lisosomas/metabolismo , Proteínas de Unión a la Región de Fijación a la Matriz/genética , Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Asociadas a Matriz Nuclear/genética , Proteínas Asociadas a Matriz Nuclear/metabolismo , Proteómica/métodos , Células RAW 264.7 , ARN Pequeño no Traducido/genética , ARN Pequeño no Traducido/metabolismo , Proteínas de Unión al ARN/clasificación , Proteínas de Unión al ARN/metabolismo , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Esfingomielina Fosfodiesterasa/genética , Esfingomielina Fosfodiesterasa/metabolismo
12.
Nat Commun ; 9(1): 476, 2018 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-29396402

RESUMEN

Though used widely in cancer therapy, paclitaxel only elicits a response in a fraction of patients. A strong determinant of paclitaxel tumor response is the state of microtubule dynamic instability. However, whether the manipulation of this physiological process can be controlled to enhance paclitaxel response has not been tested. Here, we show a previously unrecognized role of the microtubule-associated protein CRMP2 in inducing microtubule bundling through its carboxy terminus. This activity is significantly decreased when the FER tyrosine kinase phosphorylates CRMP2 at Y479 and Y499. The crystal structures of wild-type CRMP2 and CRMP2-Y479E reveal how mimicking phosphorylation prevents tetramerization of CRMP2. Depletion of FER or reducing its catalytic activity using sub-therapeutic doses of inhibitors increases paclitaxel-induced microtubule stability and cytotoxicity in ovarian cancer cells and in vivo. This work provides a rationale for inhibiting FER-mediated CRMP2 phosphorylation to enhance paclitaxel on-target activity for cancer therapy.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/metabolismo , Microtúbulos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neoplasias Ováricas/tratamiento farmacológico , Paclitaxel/farmacología , Proteínas Tirosina Quinasas/genética , Tratamiento con ARN de Interferencia , Moduladores de Tubulina/farmacología , Animales , Línea Celular Tumoral , Femenino , Humanos , Ratones , Ratones Desnudos , Microscopía Confocal , Microscopía Fluorescente , Microtúbulos/efectos de los fármacos , Microtúbulos/ultraestructura , Simulación de Dinámica Molecular , Terapia Molecular Dirigida , Trasplante de Neoplasias , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/ultraestructura , Fosforilación/efectos de los fármacos , Fosforilación/genética , Multimerización de Proteína/efectos de los fármacos , Multimerización de Proteína/genética , Proteínas Tirosina Quinasas/metabolismo , ARN Interferente Pequeño
13.
J Clin Invest ; 126(12): 4417-4429, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27775547

RESUMEN

The rising success of cancer immunotherapy has produced immense interest in defining the clinical contexts that may benefit from this therapeutic approach. To this end, there is a need to ascertain how the therapeutic modulation of intrinsic cancer cell programs influences the anticancer immune response. For example, the role of autophagy as a tumor cell survival and metabolic fitness pathway is being therapeutically targeted in ongoing clinical trials that combine cancer therapies with antimalarial drugs for the treatment of a broad spectrum of cancers, many of which will likely benefit from immunotherapy. However, our current understanding of the interplay between autophagy and the immune response remains incomplete. Here, we have evaluated how autophagy inhibition impacts the antitumor immune response in immune-competent mouse models of melanoma and mammary cancer. We observed equivalent levels of T cell infiltration and function within autophagy-competent and -deficient tumors, even upon treatment with the anthracycline chemotherapeutic doxorubicin. Similarly, we found equivalent T cell responses upon systemic treatment of tumor-bearing mice with antimalarial drugs. Our findings demonstrate that antitumor adaptive immunity is not adversely impaired by autophagy inhibition in these models, allowing for the future possibility of combining autophagy inhibitors with immunotherapy in certain clinical contexts.


Asunto(s)
Antimaláricos/farmacología , Autofagia/efectos de los fármacos , Inmunidad Celular/efectos de los fármacos , Neoplasias Mamarias Experimentales , Melanoma , Linfocitos T/inmunología , Animales , Autofagia/inmunología , Línea Celular Tumoral , Femenino , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/patología , Melanoma/tratamiento farmacológico , Melanoma/inmunología , Melanoma/patología , Ratones , Ratones Transgénicos , Linfocitos T/patología
14.
J Cell Biol ; 212(5): 577-90, 2016 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-26903539

RESUMEN

Autophagy is a catabolic pathway involving the sequestration of cellular contents into a double-membrane vesicle, the autophagosome. Although recent studies have demonstrated that autophagy supports cell migration, the underlying mechanisms remain unknown. Using live-cell imaging, we uncover that autophagy promotes optimal migratory rate and facilitates the dynamic assembly and disassembly of cell-matrix focal adhesions (FAs), which is essential for efficient motility. Additionally, our studies reveal that autophagosomes associate with FAs primarily during disassembly, suggesting autophagy locally facilitates the destabilization of cell-matrix contact sites. Furthermore, we identify the selective autophagy cargo receptor neighbor of BRCA1 (NBR1) as a key mediator of autophagy-dependent FA remodeling. NBR1 depletion impairs FA turnover and decreases targeting of autophagosomes to FAs, whereas ectopic expression of autophagy-competent, but not autophagy-defective, NBR1 enhances FA disassembly and reduces FA lifetime during migration. Our findings provide mechanistic insight into how autophagy promotes migration by revealing a requirement for NBR1-mediated selective autophagy in enabling FA disassembly in motile cells.


Asunto(s)
Autofagia , Adhesiones Focales , Proteínas/metabolismo , Animales , Células Cultivadas , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Endogámicos C57BL
15.
Methods Enzymol ; 542: 25-57, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24862259

RESUMEN

The metabolism of malignant cells is profoundly altered in order to maintain their survival and proliferation in adverse microenvironmental conditions. Autophagy is an intracellular recycling process that maintains basal levels of metabolites and biosynthetic intermediates under starvation or other forms of stress, hence serving as an important mechanism for metabolic adaptation in cancer cells. Although it is widely acknowledged that autophagy sustains metabolism in neoplastic cells under duress, many questions remain with regard to the mutual relationship between autophagy and metabolism in cancer. Importantly, autophagy has often been described as a "double-edged sword" that can either impede or promote cancer initiation and progression. Here, we overview such a dual function of autophagy in tumorigenesis and our current understanding of the coordinated regulation of autophagy and cancer cell metabolism in the control of tumor growth, progression, and resistance to therapy.


Asunto(s)
Autofagia/fisiología , Neoplasias/metabolismo , Neoplasias/patología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Aminoácidos/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Relacionadas con la Autofagia , Beclina-1 , Hipoxia de la Célula , Supervivencia Celular , Fosfatidilinositol 3-Quinasas Clase III/metabolismo , Glucosa/metabolismo , Glutamina/metabolismo , Glucólisis , Humanos , Proteínas de la Membrana/metabolismo , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteína Sequestosoma-1 , Células del Estroma/metabolismo , Células del Estroma/patología
16.
Cancer Res ; 71(17): 5806-17, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21775522

RESUMEN

The extracellular matrix protein TGFBI enhances the cytotoxic response of cancer cells to paclitaxel by affecting integrin signals that stabilize microtubules. Extending the implications of this knowledge, we tested the more general hypothesis that cancer cell signals which increase microtubule stability before exposure to paclitaxel may increase its ability to stabilize microtubules and thereby enhance its cytotoxicity. Toward this end, we carried out an siRNA screen to evaluate how genetic depletion affected microtubule stabilization, cell viability, and apoptosis. High content microscopic analysis was carried out in the absence or presence of paclitaxel. Kinase knockdowns that stabilized microtubules strongly enhanced the effects of paclitaxel treatment. Conversely, kinase knockdowns that enhanced paclitaxel-mediated cytotoxicity sensitized cells to microtubule stabilization by paclitaxel. The siRNA screen identified several genes that have not been linked previously to microtubule regulation or paclitaxel response. Gene shaving and Bayesian resampling used to classify these genes suggested three pathways of paclitaxel-induced cell death related to apoptosis and microtubule stability, apoptosis alone, or neither process. Our results offer a functional classification of the genetic basis for paclitaxel sensitivity and they support the hypothesis that stabilizing microtubules prior to therapy could enhance antitumor responses to paclitaxel treatment.


Asunto(s)
Citotoxinas/farmacología , Resistencia a Antineoplásicos , Microtúbulos/metabolismo , Neoplasias/metabolismo , Paclitaxel/farmacología , Moduladores de Tubulina/farmacología , Línea Celular Tumoral , Humanos , Microtúbulos/genética , ARN Interferente Pequeño/genética
17.
Cancer Chemother Pharmacol ; 68(4): 1033-44, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21340606

RESUMEN

PURPOSE: To investigate the mechanistic basis of the anti-tumor effect of the compound ITB-301. METHODS: Chemical modifications of genistein have been introduced to improve its solubility and efficacy. The anti-tumor effects were tested in ovarian cancer cells using proliferation assays, cell cycle analysis, immunofluorescence, and microscopy. RESULTS: In this work, we show that a unique glycoside of genistein, ITB-301, inhibits the proliferation of SKOv3 ovarian cancer cells. We found that the 50% growth inhibitory concentration of ITB-301 in SKOv3 cells was 0.5 µM. Similar results were obtained in breast cancer, ovarian cancer, and acute myelogenous leukemia cell lines. ITB-301 induced significant time- and dose-dependent microtubule depolymerization. This depolymerization resulted in mitotic arrest and inhibited proliferation in all ovarian cancer cell lines examined including SKOv3, ES2, HeyA8, and HeyA8-MDR cells. The cytotoxic effect of ITB-301 was dependent on its induction of mitotic arrest as siRNA-mediated depletion of BUBR1 significantly reduced the cytotoxic effects of ITB-301, even at a concentration of 10 µM. Importantly, efflux-mediated drug resistance did not alter the cytotoxic effect of ITB-301 in two independent cancer cell models of drug resistance. CONCLUSION: These results identify ITB-301 as a novel anti-tubulin agent that could be used in cancers that are multidrug resistant. We propose a structural model for the binding of ITB-301 to α- and ß-tubulin dimers on the basis of molecular docking simulations. This model provides a rationale for future work aimed at designing of more potent analogs.


Asunto(s)
Antineoplásicos/farmacología , Genisteína/análogos & derivados , Glicósidos/farmacología , Microtúbulos/efectos de los fármacos , Mitosis/efectos de los fármacos , Neoplasias Ováricas/tratamiento farmacológico , Antineoplásicos/administración & dosificación , Antineoplásicos/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos , Femenino , Genisteína/administración & dosificación , Genisteína/metabolismo , Genisteína/farmacología , Glicósidos/administración & dosificación , Glicósidos/metabolismo , Humanos , Concentración 50 Inhibidora , Modelos Moleculares , Simulación de Dinámica Molecular , Neoplasias Ováricas/patología , Unión Proteica , Tubulina (Proteína)/metabolismo
18.
BMC Med Genomics ; 1: 50, 2008 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-18925931

RESUMEN

BACKGROUND: It has previously been shown that specific microdeletions and microduplications, many of which also associated with cognitive impairment (CI), can present with autism spectrum disorders (ASDs). Multiplex ligation-dependent probe amplification (MLPA) represents an efficient method to screen for such recurrent microdeletions and microduplications. METHODS: In the current study, a total of 279 unrelated subjects ascertained for ASDs were screened for genomic disorders associated with CI using MLPA. Fluorescence in situ hybridization (FISH), quantitative polymerase chain reaction (Q-PCR) and/or direct DNA sequencing were used to validate potential microdeletions and microduplications. Methylation-sensitive MLPA was used to characterize individuals with duplications in the Prader-Willi/Angelman (PWA) region. RESULTS: MLPA showed two subjects with typical ASD-associated interstitial duplications of the 15q11-q13 PWA region of maternal origin. Two additional subjects showed smaller, de novo duplications of the PWA region that had not been previously characterized. Genes in these two novel duplications include GABRB3 and ATP10A in one case, and MKRN3, MAGEL2 and NDN in the other. In addition, two subjects showed duplications of the 22q11/DiGeorge syndrome region. One individual was found to carry a 12 kb deletion in one copy of the ASPA gene on 17p13, which when mutated in both alleles leads to Canavan disease. Two subjects showed partial duplication of the TM4SF2 gene on Xp11.4, previously implicated in X-linked non-specific mental retardation, but in our subsequent analyses such variants were also found in controls. A partial duplication in the ASMT gene, located in the pseudoautosomal region 1 (PAR1) of the sex chromosomes and previously suggested to be involved in ASD susceptibility, was observed in 6-7% of the cases but in only 2% of controls (P = 0.003). CONCLUSION: MLPA proves to be an efficient method to screen for chromosomal abnormalities. We identified duplications in 15q11-q13 and in 22q11, including new de novo small duplications, as likely contributing to ASD in the current sample by increasing liability and/or exacerbating symptoms. Our data indicate that duplications in TM4SF2 are not associated with the phenotype given their presence in controls. The results in PAR1/PAR2 are the first large-scale studies of gene dosage in these regions, and the findings at the ASMT locus indicate that further studies of the duplication of the ASMT gene are needed in order to gain insight into its potential involvement in ASD. Our studies also identify some limitations of MLPA, where single base changes in probe binding sequences alter results. In summary, our studies indicate that MLPA, with a focus on accepted medical genetic conditions, may be an inexpensive method for detection of microdeletions and microduplications in ASD patients for purposes of genetic counselling if MLPA-identified deletions are validated by additional methods.

19.
BMC Med Genet ; 8: 68, 2007 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-18001468

RESUMEN

BACKGROUND: Sotos syndrome is an overgrowth syndrome characterized by macrocephaly, advanced bone age, characteristic facial features, and learning disabilities, caused by mutations or deletions of the NSD1 gene, located at 5q35. Sotos syndrome has been described in a number of patients with autism spectrum disorders, suggesting that NSD1 could be involved in other cases of autism and macrocephaly. METHODS: We screened the NSD1 gene for mutations and deletions in 88 patients with autism spectrum disorders and macrocephaly (head circumference 2 standard deviations or more above the mean). Mutation analysis was performed by direct sequencing of all exons and flanking regions. Dosage analysis of NSD1 was carried out using multiplex ligation-dependent probe amplification. RESULTS: We identified three missense variants (R604L, S822C and E1499G) in one patient each, but none is within a functional domain. In addition, segregation analysis showed that all variants were inherited from healthy parents and in two cases were also present in unaffected siblings, indicating that they are probably nonpathogenic. No partial or whole gene deletions/duplications were observed. CONCLUSION: Our findings suggest that Sotos syndrome is a rare cause of autism spectrum disorders and that screening for NSD1 mutations and deletions in patients with autism and macrocephaly is not warranted in the absence of other features of Sotos syndrome.


Asunto(s)
Sustitución de Aminoácidos/genética , Trastorno Autístico/genética , Anomalías Craneofaciales/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas Nucleares/genética , Adolescente , Adulto , Niño , Preescolar , Análisis Mutacional de ADN , Femenino , Pruebas Genéticas , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina , Humanos , Masculino , Síndrome
20.
Am J Med Genet B Neuropsychiatr Genet ; 144B(4): 484-91, 2007 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-17427195

RESUMEN

Mutations in the PTEN gene are associated with a broad spectrum of disorders, including Cowden syndrome (CS), Bannayan-Riley-Ruvalcaba syndrome, Proteus syndrome, and Lhermitte-Duclos disease. In addition, PTEN mutations have been described in a few patients with autism spectrum disorders (ASDs) and macrocephaly. In this study, we screened the PTEN gene for mutations and deletions in 88 patients with ASDs and macrocephaly (defined as >or=2 SD above the mean). Mutation analysis was performed by direct sequencing of all exons and flanking regions, as well as the promoter region. Dosage analysis of PTEN was carried out using multiplex ligation-dependent probe amplification (MLPA). No partial or whole gene deletions were observed. We identified a de novo missense mutation (D326N) in a highly conserved amino acid in a 5-year-old boy with autism, mental retardation, language delay, extreme macrocephaly (+9.6 SD) and polydactyly of both feet. Polydactyly has previously been described in two patients with Lhermitte-Duclos disease and CS and is thus likely to be a rare sign of PTEN mutations. Our findings suggest that PTEN mutations are a relatively infrequent cause of ASDs with macrocephaly. Screening of PTEN mutations is warranted in patients with autism and pronounced macrocephaly, even in the absence of other features of PTEN-related tumor syndromes.


Asunto(s)
Trastorno Autístico/complicaciones , Trastorno Autístico/genética , Anomalías Craneofaciales/complicaciones , Anomalías Craneofaciales/genética , Pruebas Genéticas , Mutación/genética , Fosfohidrolasa PTEN/genética , Anomalías Múltiples/genética , Adolescente , Adulto , Secuencia de Aminoácidos , Asparagina/genética , Ácido Aspártico/genética , Niño , Preescolar , Análisis Mutacional de ADN , Exones/genética , Femenino , Humanos , Intrones/genética , Masculino , Datos de Secuencia Molecular , Fosfohidrolasa PTEN/química , Síndrome
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...