Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Res Sq ; 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38765977

RESUMEN

PINK1 loss-of-function mutations and exposure to mitochondrial toxins are causative for Parkinson's disease (PD) and Parkinsonism, respectively. We demonstrate that pathological α-synuclein deposition, the hallmark pathology of idiopathic PD, induces mitochondrial dysfunction, and impairs mitophagy as evidenced by the accumulation of the PINK1 substrate pS65-Ubiquitin (pUb). We discovered MTK458, a brain penetrant small molecule that binds to PINK1 and stabilizes its active complex, resulting in increased rates of mitophagy. Treatment with MTK458 mediates clearance of accumulated pUb and α-synuclein pathology in α-synuclein pathology models in vitro and in vivo. Our findings from preclinical PD models suggest that pharmacological activation of PINK1 warrants further clinical evaluation as a therapeutic strategy for disease modification in PD.

2.
bioRxiv ; 2023 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-36824886

RESUMEN

PINK1 loss-of-function mutations and exposure to mitochondrial toxins are causative for Parkinson's disease (PD) and Parkinsonism, respectively. We demonstrate that pathological α-synuclein deposition, the hallmark pathology of idiopathic PD, induces mitochondrial dysfunction and impairs mitophagy, driving accumulation of the PINK1 substrate pS65-Ubiquitin (pUb) in primary neurons and in vivo. We synthesized MTK458, a brain penetrant small molecule that binds to PINK1 and stabilizes an active heterocomplex, thereby increasing mitophagy. MTK458 mediates clearance of α-synuclein pathology in PFF seeding models in vitro and in vivo and reduces pUb. We developed an ultrasensitive assay to quantify pUb levels in plasma and observed an increase in pUb in PD subjects that correlates with disease progression, paralleling our observations in PD models. Our combined findings from preclinical PD models and patient biofluids suggest that pharmacological activation of PINK1 is worthy of further study as a therapeutic strategy for disease modification in PD. Highlights: Discovery of a plasma Parkinson's Disease biomarker candidate, pS65-Ubiquitin (pUb)Plasma pUb levels correlate with disease status and progression in PD patients.Identification of a potent, brain penetrant PINK1 activator, MTK458MTK458 selectively activates PINK1 by stimulating dimerization and stabilization of the PINK1/TOM complexMTK458 drives clearance of α-synuclein pathology and normalizes pUb in in vivo Parkinson's models.

3.
Nat Med ; 26(1): 118-130, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31873312

RESUMEN

Gene silencing with virally delivered shRNA represents a promising approach for treatment of inherited neurodegenerative disorders. In the present study we develop a subpial technique, which we show in adult animals successfully delivers adeno-associated virus (AAV) throughout the cervical, thoracic and lumbar spinal cord, as well as brain motor centers. One-time injection at cervical and lumbar levels just before disease onset in mice expressing a familial amyotrophic lateral sclerosis (ALS)-causing mutant SOD1 produces long-term suppression of motoneuron disease, including near-complete preservation of spinal α-motoneurons and muscle innervation. Treatment after disease onset potently blocks progression of disease and further α-motoneuron degeneration. A single subpial AAV9 injection in adult pigs or non-human primates using a newly designed device produces homogeneous delivery throughout the cervical spinal cord white and gray matter and brain motor centers. Thus, spinal subpial delivery in adult animals is highly effective for AAV-mediated gene delivery throughout the spinal cord and supraspinal motor centers.


Asunto(s)
Esclerosis Amiotrófica Lateral/terapia , Dependovirus/metabolismo , Silenciador del Gen , Técnicas de Transferencia de Gen , Neuronas Motoras/patología , Degeneración Nerviosa/terapia , Piamadre/patología , Médula Espinal/patología , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/fisiopatología , Animales , Atrofia , Progresión de la Enfermedad , Potenciales Evocados Motores , Femenino , Regulación de la Expresión Génica , Humanos , Inflamación/patología , Interneuronas/patología , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Desarrollo de Músculos , Degeneración Nerviosa/genética , Degeneración Nerviosa/fisiopatología , Piamadre/fisiopatología , Primates , Pliegue de Proteína , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/administración & dosificación , Médula Espinal/diagnóstico por imagen , Médula Espinal/fisiopatología , Superóxido Dismutasa-1/genética , Superóxido Dismutasa-1/metabolismo , Porcinos
4.
Stem Cell Res Ther ; 10(1): 83, 2019 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-30867054

RESUMEN

BACKGROUND: A well-characterized method has not yet been established to reproducibly, efficiently, and safely isolate large numbers of clinical-grade multipotent human neural stem cells (hNSCs) from embryonic stem cells (hESCs). Consequently, the transplantation of neurogenic/gliogenic precursors into the CNS for the purpose of cell replacement or neuroprotection in humans with injury or disease has not achieved widespread testing and implementation. METHODS: Here, we establish an approach for the in vitro isolation of a highly expandable population of hNSCs using the manual selection of neural precursors based on their colony morphology (CoMo-NSC). The purity and NSC properties of established and extensively expanded CoMo-NSC were validated by expression of NSC markers (flow cytometry, mRNA sequencing), lack of pluripotent markers and by their tumorigenic/differentiation profile after in vivo spinal grafting in three different animal models, including (i) immunodeficient rats, (ii) immunosuppressed ALS rats (SOD1G93A), or (iii) spinally injured immunosuppressed minipigs. RESULTS: In vitro analysis of established CoMo-NSCs showed a consistent expression of NSC markers (Sox1, Sox2, Nestin, CD24) with lack of pluripotent markers (Nanog) and stable karyotype for more than 15 passages. Gene profiling and histology revealed that spinally grafted CoMo-NSCs differentiate into neurons, astrocytes, and oligodendrocytes over a 2-6-month period in vivo without forming neoplastic derivatives or abnormal structures. Moreover, transplanted CoMo-NSCs formed neurons with synaptic contacts and glia in a variety of host environments including immunodeficient rats, immunosuppressed ALS rats (SOD1G93A), or spinally injured minipigs, indicating these cells have favorable safety and differentiation characteristics. CONCLUSIONS: These data demonstrate that manually selected CoMo-NSCs represent a safe and expandable NSC population which can effectively be used in prospective human clinical cell replacement trials for the treatment of a variety of neurodegenerative disorders, including ALS, stroke, spinal traumatic, or spinal ischemic injury.


Asunto(s)
Citometría de Flujo , Células Madre Multipotentes/citología , Células-Madre Neurales/citología , Línea Celular , Humanos
5.
Acta Neuropathol ; 133(6): 907-922, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28357566

RESUMEN

Mutations in TDP-43 cause amyotrophic lateral sclerosis (ALS), a fatal paralytic disease characterized by degeneration and premature death of motor neurons. The contribution of mutant TDP-43-mediated damage within motor neurons was evaluated using mice expressing a conditional allele of an ALS-causing TDP-43 mutant (Q331K) whose broad expression throughout the central nervous system mimics endogenous TDP-43. TDP-43Q331K mice develop age- and mutant-dependent motor deficits from degeneration and death of motor neurons. Cre-recombinase-mediated excision of the TDP-43Q331K gene from motor neurons is shown to delay onset of motor symptoms and appearance of TDP-43-mediated aberrant nuclear morphology, and abrogate subsequent death of motor neurons. However, reduction of mutant TDP-43 selectively in motor neurons did not prevent age-dependent degeneration of axons and neuromuscular junction loss, nor did it attenuate astrogliosis or microgliosis. Thus, disease mechanism is non-cell autonomous with mutant TDP-43 expressed in motor neurons determining disease onset but progression defined by mutant acting within other cell types.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Proteínas de Unión al ADN/metabolismo , Neuronas Motoras/metabolismo , Adulto , Anciano de 80 o más Años , Envejecimiento/metabolismo , Envejecimiento/patología , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Animales , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Humanos , Inflamación/metabolismo , Inflamación/patología , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Persona de Mediana Edad , Actividad Motora/fisiología , Neuronas Motoras/patología , Mutación , Unión Neuromuscular/metabolismo , Unión Neuromuscular/patología , Adulto Joven
6.
Proc Natl Acad Sci U S A ; 112(50): E6993-7002, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26621731

RESUMEN

Ubiquitous expression of amyotrophic lateral sclerosis (ALS)-causing mutations in superoxide dismutase 1 (SOD1) provokes noncell autonomous paralytic disease. By combining ribosome affinity purification and high-throughput sequencing, a cascade of mutant SOD1-dependent, cell type-specific changes are now identified. Initial mutant-dependent damage is restricted to motor neurons and includes synapse and metabolic abnormalities, endoplasmic reticulum (ER) stress, and selective activation of the PRKR-like ER kinase (PERK) arm of the unfolded protein response. PERK activation correlates with what we identify as a naturally low level of ER chaperones in motor neurons. Early changes in astrocytes occur in genes that are involved in inflammation and metabolism and are targets of the peroxisome proliferator-activated receptor and liver X receptor transcription factors. Dysregulation of myelination and lipid signaling pathways and activation of ETS transcription factors occur in oligodendrocytes only after disease initiation. Thus, pathogenesis involves a temporal cascade of cell type-selective damage initiating in motor neurons, with subsequent damage within glia driving disease propagation.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Perfilación de la Expresión Génica , Neuronas Motoras/metabolismo , Mutación , Neuroglía/metabolismo , Biosíntesis de Proteínas , Superóxido Dismutasa/genética , Anciano , Esclerosis Amiotrófica Lateral/patología , Animales , Astrocitos/metabolismo , Astrocitos/patología , Humanos , Ratones , Neuronas Motoras/patología , Neuroglía/patología , Superóxido Dismutasa-1
7.
Neuron ; 86(1): 218-32, 2015 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-25801706

RESUMEN

Mutations in superoxide dismutase (SOD1) cause amyotrophic lateral sclerosis (ALS), a neurodegenerative disease characterized by loss of motor neurons and accompanied by accumulation of misfolded SOD1 onto the cytoplasmic faces of intracellular organelles, including mitochondria and the endoplasmic reticulum (ER). Using inhibition of misfolded SOD1 deposition onto mitochondria as an assay, a chaperone activity abundant in nonneuronal tissues is now purified and identified to be the multifunctional macrophage migration inhibitory factor (MIF), whose activities include an ATP-independent protein folding chaperone. Purified MIF is shown to directly inhibit mutant SOD1 misfolding. Elevating MIF in neuronal cells suppresses accumulation of misfolded SOD1 and its association with mitochondria and the ER and extends survival of mutant SOD1-expressing motor neurons. Accumulated MIF protein is identified to be low in motor neurons, implicating correspondingly low chaperone activity as a component of vulnerability to mutant SOD1 misfolding and supporting therapies to enhance intracellular MIF chaperone activity.


Asunto(s)
Factores Inhibidores de la Migración de Macrófagos/metabolismo , Pliegue de Proteína , Superóxido Dismutasa/metabolismo , Fosfatasa Ácida/genética , Animales , Diferenciación Celular/genética , Células Cultivadas , Colina O-Acetiltransferasa/genética , Colina O-Acetiltransferasa/metabolismo , Retículo Endoplásmico/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Células Madre Pluripotentes Inducidas , Isoenzimas/genética , Hígado/metabolismo , Hígado/ultraestructura , Ratones , Ratones Transgénicos , Mitocondrias/metabolismo , Neuronas Motoras/fisiología , Mutación/genética , Proteínas del Tejido Nervioso/metabolismo , Transporte de Proteínas/genética , Ratas , Ratas Transgénicas , Proteínas Ribosómicas/genética , Proteínas Ribosómicas/metabolismo , Médula Espinal/metabolismo , Médula Espinal/ultraestructura , Superóxido Dismutasa/genética , Superóxido Dismutasa-1 , Fosfatasa Ácida Tartratorresistente
8.
Proc Natl Acad Sci U S A ; 111(2): 829-32, 2014 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-24379375

RESUMEN

Amyotrophic lateral sclerosis (ALS) causes motor neuron degeneration, paralysis, and death. Accurate disease modeling, identifying disease mechanisms, and developing therapeutics is urgently needed. We previously reported motor neuron toxicity through postmortem ALS spinal cord-derived astrocytes. However, these cells can only be harvested after death, and their expansion is limited. We now report a rapid, highly reproducible method to convert adult human fibroblasts from living ALS patients to induced neuronal progenitor cells and subsequent differentiation into astrocytes (i-astrocytes). Non-cell autonomous toxicity to motor neurons is found following coculture of i-astrocytes from familial ALS patients with mutation in superoxide dismutase or hexanucleotide expansion in C9orf72 (ORF 72 on chromosome 9) the two most frequent causes of ALS. Remarkably, i-astrocytes from sporadic ALS patients are as toxic as those with causative mutations, suggesting a common mechanism. Easy production and expansion of i-astrocytes now enables rapid disease modeling and high-throughput drug screening to alleviate astrocyte-derived toxicity.


Asunto(s)
Esclerosis Amiotrófica Lateral/fisiopatología , Astrocitos/citología , Desdiferenciación Celular/fisiología , Diferenciación Celular/fisiología , Fibroblastos/citología , Neuronas Motoras/patología , Células-Madre Neurales/citología , Análisis de Varianza , Astrocitos/metabolismo , Comunicación Celular , Técnicas de Cultivo de Célula , Cartilla de ADN/genética , Técnica del Anticuerpo Fluorescente , Humanos , Modelos Biológicos , Neuronas Motoras/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
9.
Mol Ther ; 21(12): 2148-59, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24008656

RESUMEN

Mutations in superoxide dismutase 1 (SOD1) are linked to familial amyotrophic lateral sclerosis (ALS) resulting in progressive motor neuron death through one or more acquired toxicities. Involvement of wild-type SOD1 has been linked to sporadic ALS, as misfolded SOD1 has been reported in affected tissues of sporadic patients and toxicity of astrocytes derived from sporadic ALS patients to motor neurons has been reported to be reduced by lowering the synthesis of SOD1. We now report slowed disease onset and progression in two mouse models following therapeutic delivery using a single peripheral injection of an adeno-associated virus serotype 9 (AAV9) encoding an shRNA to reduce the synthesis of ALS-causing human SOD1 mutants. Delivery to young mice that develop aggressive, fatal paralysis extended survival by delaying both disease onset and slowing progression. In a later-onset model, AAV9 delivery after onset markedly slowed disease progression and significantly extended survival. Moreover, AAV9 delivered intrathecally to nonhuman primates is demonstrated to yield robust SOD1 suppression in motor neurons and glia throughout the spinal cord and therefore, setting the stage for AAV9-mediated therapy in human clinical trials.


Asunto(s)
Esclerosis Amiotrófica Lateral/patología , Esclerosis Amiotrófica Lateral/terapia , Dependovirus/genética , Terapia Genética , Neuronas Motoras/metabolismo , Neuroglía/metabolismo , ARN Interferente Pequeño/genética , Superóxido Dismutasa/genética , Administración Intravenosa , Esclerosis Amiotrófica Lateral/genética , Animales , Células COS , Chlorocebus aethiops , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Vectores Genéticos , Células HEK293 , Humanos , Inyecciones Espinales , Macaca fascicularis , Ratones , Neuronas Motoras/patología , Neuroglía/patología , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa-1
10.
Proc Natl Acad Sci U S A ; 110(8): E736-45, 2013 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-23382207

RESUMEN

Transactivating response region DNA binding protein (TDP-43) is the major protein component of ubiquitinated inclusions found in amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD) with ubiquitinated inclusions. Two ALS-causing mutants (TDP-43(Q331K) and TDP-43(M337V)), but not wild-type human TDP-43, are shown here to provoke age-dependent, mutant-dependent, progressive motor axon degeneration and motor neuron death when expressed in mice at levels and in a cell type-selective pattern similar to endogenous TDP-43. Mutant TDP-43-dependent degeneration of lower motor neurons occurs without: (i) loss of TDP-43 from the corresponding nuclei, (ii) accumulation of TDP-43 aggregates, and (iii) accumulation of insoluble TDP-43. Computational analysis using splicing-sensitive microarrays demonstrates alterations of endogenous TDP-43-dependent alternative splicing events conferred by both human wild-type and mutant TDP-43(Q331K), but with high levels of mutant TDP-43 preferentially enhancing exon exclusion of some target pre-mRNAs affecting genes involved in neurological transmission and function. Comparison with splicing alterations following TDP-43 depletion demonstrates that TDP-43(Q331K) enhances normal TDP-43 splicing function for some RNA targets but loss-of-function for others. Thus, adult-onset motor neuron disease does not require aggregation or loss of nuclear TDP-43, with ALS-linked mutants producing loss and gain of splicing function of selected RNA targets at an early disease stage.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Núcleo Celular/metabolismo , Proteínas de Unión al ADN/genética , Mutación , Empalme del ARN , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/fisiopatología , Animales , Proteínas de Unión al ADN/metabolismo , Ratones , Ratones Transgénicos , Reacción en Cadena en Tiempo Real de la Polimerasa , Ubiquitinación
11.
Oncoimmunology ; 1(1): 94-96, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-22720222

RESUMEN

The mechanism of tumor cell death after treatment with DNA alkylating agents in vivo previously remained largely unknown. We demonstrate that tumor regression after chemotherapy occurs via sporadic necrosis and relies on activation of innate immunity in a manner dependent on high mobility group box 1 protein (HMGB1).

12.
J Immunol ; 186(6): 3517-26, 2011 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-21300822

RESUMEN

Dysregulation of apoptosis is associated with the development of human cancer and resistance to anticancer therapy. We have previously shown in tumor xenografts that DNA alkylating agents induce sporadic cell necrosis and regression of apoptosis-deficient tumors. Sporadic tumor cell necrosis is associated with extracellular release of cellular content such as the high mobility group box 1 (HMGB1) protein and subsequent recruitment of innate immune cells into the tumor tissue. It remained unclear whether HMGB1 and the activation of innate immunity played a role in tumor response to chemotherapy. In this study, we show that whereas DNA alkylating therapy leads to a complete tumor regression in an athymic mouse tumor xenograft model, it fails to do so in tumors deficient in HMGB1. The HMGB1-deficient tumors have an impaired ability to recruit innate immune cells including macrophages, neutrophils, and NK cells into the treated tumor tissue. Cytokine array analysis reveals that whereas DNA alkylating treatment leads to suppression of protumor cytokines such as IL-4, IL-10, and IL-13, loss of HMGB1 leads to elevated levels of these cytokines upon treatment. Suppression of innate immunity and HMGB1 using depleting Abs leads to a failure in tumor regression. Taken together, these results indicate that HMGB1 plays an essential role in activation of innate immunity and tumor clearance in response to DNA alkylating agents.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Resistencia a Antineoplásicos/inmunología , Proteína HMGB1/fisiología , Inmunidad Innata/inmunología , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/prevención & control , Animales , Antineoplásicos Alquilantes/uso terapéutico , Línea Celular Transformada , Línea Celular Tumoral , Células Cultivadas , Células HEK293 , Proteína HMGB1/deficiencia , Proteína HMGB1/metabolismo , Humanos , Masculino , Ratones , Ratones Desnudos , Necrosis , Neoplasias Experimentales/patología , Distribución Aleatoria
13.
Mol Cell ; 36(1): 51-60, 2009 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-19818709

RESUMEN

The activity and specificity of serine/threonine phosphatases are governed largely by their associated proteins. alpha4 is an evolutionarily conserved noncatalytic subunit for PP2A-like phosphatases. Though alpha4 binds to only a minority of PP2A-related catalytic subunits, alpha4 deletion leads to progressive loss of all PP2A, PP4, and PP6 phosphatase complexes. In healthy cells, association with alpha4 renders catalytic (C) subunits enzymatically inactive while protecting them from proteasomal degradation until they are assembled into a functional phosphatase complex. During cellular stress, existing PP2A complexes can become unstable. Under such conditions, alpha4 sequesters released C subunits and is required for the adaptive increase in targeted PP2A activity that can dephosphorylate stress-induced phosphorylated substrates. Consistent with this, overexpression of alpha4 protects cells from a variety of stress stimuli, including DNA damage and nutrient limitation. These findings demonstrate that alpha4 plays a required role in regulating the assembly and maintenance of adaptive PP2A phosphatase complexes.


Asunto(s)
Fosfoproteínas/fisiología , Proteína Fosfatasa 2/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Camptotecina/farmacología , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Supervivencia Celular/fisiología , Daño del ADN/efectos de los fármacos , Daño del ADN/fisiología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Doxorrubicina/farmacología , Estabilidad de Enzimas/fisiología , Femenino , Expresión Génica/efectos de los fármacos , Expresión Génica/genética , Respuesta al Choque Térmico/fisiología , Histonas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular , Hígado/metabolismo , Masculino , Ratones , Ratones Noqueados , Chaperonas Moleculares , Fosfoproteínas Fosfatasas/metabolismo , Fosforilación/efectos de los fármacos , Fosforilación/genética , Multimerización de Proteína/fisiología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Ubiquitinación/genética
14.
Proc Natl Acad Sci U S A ; 106(34): 14391-6, 2009 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-19706526

RESUMEN

Approximately 50% of cancer patients receive radiation treatment, either alone or in combination with other therapies. Tumor hypoxia has long been associated with resistance to radiation therapy. Moreover, the expression of hypoxia inducible factors HIF1alpha and/or HIF2alpha correlates with poor prognosis in many tumors. Recent evidence indicates that HIF1alpha expression can enhance radiation-induced apoptosis in cancer cells. We demonstrate here that HIF2alpha inhibition promotes tumor cell death and, in contrast to HIF1alpha, enhances the response to radiation treatment. Specifically, inhibiting HIF2alpha expression augments p53 activity, increases apoptosis, and reduces clonogenic survival of irradiated and non-irradiated cells. Moreover, HIF2alpha inhibition promotes p53-mediated responses by disrupting cellular redox homeostasis, thereby permitting reactive oxygen species (ROS) accumulation and DNA damage. These results correlate with altered p53 phosphorylation and target gene expression in untreated human tumor samples and show that HIF2alpha likely contributes to tumor cell survival including during radiation therapy.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Neoplasias/metabolismo , Interferencia de ARN , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Caspasa 3/metabolismo , Ciclo Celular/efectos de la radiación , Muerte Celular/efectos de la radiación , Hipoxia de la Célula , Línea Celular Tumoral , Daño del ADN , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Humanos , Immunoblotting , Microscopía Fluorescente , Neoplasias/genética , Neoplasias/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Proto-Oncogénicas/metabolismo , Radiación Ionizante , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de la radiación , Proteína p53 Supresora de Tumor/genética
15.
Cancer Res ; 68(23): 9595-600, 2008 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19047135

RESUMEN

Dysregulation of apoptosis is associated with the development of human cancer and resistance to anticancer therapy. The ultimate goal of cancer treatment is to selectively induce cancer cell death and overcome drug resistance. A deeper understanding of how a given chemotherapy affects tumor cell death is needed to develop strategically designed anticancer agents. Here, we use a xenograft mouse tumor system generated from genetically defined cells deficient in apoptosis to examine the involvement of multiple forms of cell death induced by cyclophosphamide (CP), a DNA alkylating agent commonly used in chemotherapy. We find that although apoptosis facilitates tumor regression, it is dispensable for complete tumor regression as other forms of cell death are activated. Sporadic necrosis is observed in both apoptosis-competent and deficient tumors evident by tumor cell morphology, extracellular release of high mobility group box 1 protein, and activation of innate immune cells in CP-treated tumors. Our findings indicate that in apoptosis-deficient tumors, necrosis may play a fundamental role in tumor clearance by stimulating the innate immune response.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Apoptosis/efectos de los fármacos , Ciclofosfamida/farmacología , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/patología , Proteínas E1A de Adenovirus/genética , Animales , Apoptosis/fisiología , Daño del ADN , ADN de Neoplasias/efectos de los fármacos , ADN de Neoplasias/metabolismo , Resistencia a Antineoplásicos , Genes ras , Humanos , Inmunidad Innata/efectos de los fármacos , Masculino , Ratones , Ratones Desnudos , Necrosis , Neoplasias Experimentales/genética , Neoplasias Experimentales/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína Destructora del Antagonista Homólogo bcl-2/deficiencia , Proteína X Asociada a bcl-2/deficiencia
16.
Curr Opin Genet Dev ; 18(1): 54-61, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18387799

RESUMEN

Tumor cells display increased metabolic autonomy in comparison to non-transformed cells, taking up nutrients and metabolizing them in pathways that support growth and proliferation. Classical work in tumor cell metabolism focused on bioenergetics, particularly enhanced glycolysis and suppressed oxidative phosphorylation (the 'Warburg effect'). But the biosynthetic activities required to create daughter cells are equally important for tumor growth, and recent studies are now bringing these pathways into focus. In this review, we discuss how tumor cells achieve high rates of nucleotide and fatty acid synthesis, how oncogenes and tumor suppressors influence these activities, and how glutamine metabolism enables macromolecular synthesis in proliferating cells.


Asunto(s)
Neoplasias/metabolismo , Proliferación Celular , Ácidos Grasos/biosíntesis , Glutamina/metabolismo , Glucólisis , Humanos , Lípidos/biosíntesis , Modelos Biológicos , Neoplasias/patología , Ribosamonofosfatos/biosíntesis
17.
J Biol Chem ; 282(40): 29712-20, 2007 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-17693407

RESUMEN

Compared with kinases, the role of protein phosphatases in regulating biological functions is less well understood. Here we show that alpha4, a non-catalytic subunit of the protein phosphatase 2A, plays a major role in the control of cell spreading, migration, and cytoskeletal architecture. Fibroblasts lacking alpha4 were impaired in their ability to spread and migrate compared with wild-type cells, whereas enforced expression of alpha4 promoted cell spreading and migration. These effects were not restricted to fibroblasts. Using a T cell-specific alpha4 transgenic mouse model, increased alpha4 expression was found to increase lymphocyte motility and chemotaxis. Elevated alpha4 expression results in an increase in the GTP-bound state of Rac1, and GTP-bound Rac1 was dramatically reduced in alpha4-deficient cells. A constitutively active mutant of Rac1 rescued the defects of cell spreading and migration caused by alpha4 deletion, while inhibition of Rac1 blocked the ability of alpha4 to promote cell migration. Together, these data define a novel role for the protein phosphatase 2A regulatory subunit alpha4 in the regulation of cell spreading and migration.


Asunto(s)
Regulación de la Expresión Génica , Fosfoproteínas/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Línea Celular , Movimiento Celular , Quimiotaxis , Fibroblastos/metabolismo , Eliminación de Gen , Guanosina Trifosfato/metabolismo , Péptidos y Proteínas de Señalización Intercelular , Ratones , Ratones Transgénicos , Modelos Biológicos , Chaperonas Moleculares , Neuropéptidos , Fosfoproteínas/metabolismo , Cicatrización de Heridas , Proteínas de Unión al GTP rac , Proteína de Unión al GTP rac1
18.
J Biol Chem ; 282(24): 17845-54, 2007 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-17430886

RESUMEN

Necrotic cells release inflammatory mediators that activate cytokine production from innate immune cells. One mediator of this activation is high mobility group box 1 protein (HMGB1). HMGB1 is normally a chromatin-associated protein and is sequestered at condensed chromatin during apoptosis. How it is released from chromatin during necrotic cell death is not known. Here we show that after DNA-alkylating damage, the activation of poly(ADP)-ribose polymerase (PARP) regulates the translocation of HMGB1 from the nucleus to the cytosol. This displaced HMGB1 is subject to release if the cell then loses plasma membrane integrity as a result of necrosis. Both full-length HMGB1 and a truncated form of HMGB1 lacking the highly conserved glutamate-rich C-terminal tail can induce macrophage activation and tumor necrosis factor-alpha production. However, displacement of HMGB1 from the nucleus following PARP activation requires the presence of the glutamate-rich C-terminal tail. Although the C-terminal tail is not the sole substrate for PARP modification of HMGB1, it appears to be required to destabilize HMGB1 association with chromatin following PARP-dependent chromatin modifications. These data suggest that PARP-dependent nuclear-to-cytosolic translocation of HMGB1 serves to establish the ability of cells to release this potent inflammatory mediator upon subsequent necrotic death.


Asunto(s)
Núcleo Celular/metabolismo , Proteína HMGB1/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Transporte Activo de Núcleo Celular/fisiología , Secuencia de Aminoácidos , Animales , Muerte Celular/fisiología , Línea Celular , Daño del ADN , Activación Enzimática , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/genética
19.
Cancer Biol Ther ; 3(12): 1214-6, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15611628

RESUMEN

The NF-kappaB family of transcription factors, well-known for regulating immune and inflammatory responses, has recently been identified as a molecular link between chronic inflammation and cancer. Using distinct models of inflammation-associated cancer, three recent reports now show the importance of NF-kappa B in promoting cancer progression during inflammation (Greten et al., Cell 2004, 118:285-96; Luo et al., Cancer Cell 2004, 6:297-305, Pikarsky et al., Nature 2004, 431:461-6). Results from these studies suggest that NF-kappa B exerts its oncogenic effects in both tumor cells and in the tumor microenvironment, promoting the survival of premalignant epithelial cells while also stimulating release of pro-inflammatory mediators from activated macrophages that promote tumor growth.


Asunto(s)
FN-kappa B/metabolismo , Neoplasias/metabolismo , Animales , Regulación Neoplásica de la Expresión Génica , Humanos , Inflamación/metabolismo , Inflamación/patología , Transducción de Señal
20.
Genes Dev ; 18(11): 1272-82, 2004 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-15145826

RESUMEN

Necrosis has been considered a passive form of cell death in which the cell dies as a result of a bioenergetic catastrophe imposed by external conditions. However, in response to alkylating DNA damage, cells undergo necrosis as a self-determined cell fate. This form of death does not require the central apoptotic mediators p53, Bax/Bak, or caspases and actively induces an inflammatory response. Necrosis in response to DNA damage requires activation of the DNA repair protein poly(ADP-ribose) polymerase (PARP), but PARP activation is not sufficient to determine cell fate. Cell death is determined by the effect of PARP-mediated beta-nicotinamide adenine dinucleotide (NAD) consumption on cellular metabolism. Cells using aerobic glycolysis to support their bioenergetics undergo rapid ATP depletion and death in response to PARP activation. In contrast, cells catabolizing nonglucose substrates to maintain oxidative phosphorylation are resistant to ATP depletion and death in response to PARP activation. Because most cancer cells maintain their ATP production through aerobic glycolysis, these data may explain the molecular basis by which DNA-damaging agents can selectively induce tumor cell death independent of p53 or Bcl-2 family proteins.


Asunto(s)
Alquilantes/farmacología , Daño del ADN/fisiología , Necrosis , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2 , Adenosina Trifosfato/metabolismo , Animales , Secuencia de Bases , Células Cultivadas , Activación Enzimática/efectos de los fármacos , Fibroblastos/patología , Proteína HMGB1/metabolismo , Inflamación/metabolismo , Inflamación/patología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/patología , Proteínas de la Membrana/efectos de los fármacos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Mutantes , Datos de Secuencia Molecular , NAD/metabolismo , Poli(ADP-Ribosa) Polimerasas/efectos de los fármacos , Proteínas Proto-Oncogénicas/efectos de los fármacos , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Proteína p53 Supresora de Tumor/efectos de los fármacos , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteína Destructora del Antagonista Homólogo bcl-2 , Proteína X Asociada a bcl-2
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...