Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Proc Natl Acad Sci U S A ; 117(23): 12806-12816, 2020 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-32444488

RESUMEN

The most prevalent human carcinogen is sunlight-associated ultraviolet (UV), a physiologic dose of which generates thousands of DNA lesions per cell, mostly of two types: cyclobutane pyrimidine dimers (CPDs) and 6-4 photoproducts (6-4PPs). It has not been possible, in living cells, to precisely characterize the respective contributions of these two lesion types to the signals that regulate cell cycle progression, DNA replication, and cell survival. Here we coupled multiparameter flow cytometry with lesion-specific photolyases that eliminate either CPDs or 6-4PPs and determined their respective contributions to DNA damage responses. Strikingly, only 6-4PP lesions activated the ATR-Chk1 DNA damage response pathway. Mechanistically, 6-4PPs, but not CPDs, impeded DNA replication across the genome as revealed by microfluidic-assisted replication track analysis. Furthermore, single-stranded DNA accumulated preferentially at 6-4PPs during DNA replication, indicating selective and prolonged replication blockage at 6-4PPs. These findings suggest that 6-4PPs, although eightfold fewer in number than CPDs, are the trigger for UV-induced DNA damage responses.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Daño del ADN , Replicación del ADN , Dímeros de Pirimidina/genética , Rayos Ultravioleta , Animales , Células Cultivadas , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Reparación del ADN , Células HCT116 , Humanos
2.
Int J Mol Sci ; 22(1)2020 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-33396303

RESUMEN

Endoplasmic reticulum (ER) stress response is an adaptive program to cope with cellular stress that disturbs the function and homeostasis of ER, which commonly occurs during cancer progression to late stage. Late-stage cancers, mostly requiring chemotherapy, often develop treatment resistance. Chemoresistance has been linked to ER stress response; however, most of the evidence has come from studies that correlate the expression of stress markers with poor prognosis or demonstrate proapoptosis by the knockdown of stress-responsive genes. Since ER stress in cancers usually persists and is essentially not induced by genetic manipulations, we used low doses of ER stress inducers at levels that allowed cell adaptation to occur in order to investigate the effect of stress response on chemoresistance. We found that prolonged tolerable ER stress promotes mesenchymal-epithelial transition, slows cell-cycle progression, and delays the S-phase exit. Consequently, cisplatin-induced apoptosis was significantly decreased in stress-adapted cells, implying their acquisition of cisplatin resistance. Molecularly, we found that proliferating cell nuclear antigen (PCNA) ubiquitination and the expression of polymerase η, the main polymerase responsible for translesion synthesis across cisplatin-DNA damage, were up-regulated in ER stress-adaptive cells, and their enhanced cisplatin resistance was abrogated by the knockout of polymerase η. We also found that a fraction of p53 in stress-adapted cells was translocated to the nucleus, and that these cells exhibited a significant decline in the level of cisplatin-DNA damage. Consistently, we showed that the nuclear p53 coincided with strong positivity of glucose-related protein 78 (GRP78) on immunostaining of clinical biopsies, and the cisplatin-based chemotherapy was less effective for patients with high levels of ER stress. Taken together, this study uncovers that adaptation to ER stress enhances DNA repair and damage tolerance, with which stressed cells gain resistance to chemotherapeutics.


Asunto(s)
Adaptación Fisiológica , Cisplatino/farmacología , Reparación del ADN , ADN Polimerasa Dirigida por ADN/metabolismo , Resistencia a Antineoplásicos , Estrés del Retículo Endoplásmico , Neoplasias de la Boca/tratamiento farmacológico , Antineoplásicos/farmacología , Apoptosis , Proliferación Celular , Daño del ADN , Replicación del ADN , ADN Polimerasa Dirigida por ADN/genética , Chaperón BiP del Retículo Endoplásmico , Humanos , Neoplasias de la Boca/metabolismo , Neoplasias de la Boca/patología , Células Tumorales Cultivadas
3.
Cancer ; 123(8): 1464-1474, 2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-27925665

RESUMEN

BACKGROUND: Merkel cell carcinoma (MCC) is an aggressive skin cancer with a recurrence rate of >40%. Of the 2000 MCC cases per year in the United States, most are caused by the Merkel cell polyomavirus (MCPyV). Antibodies to MCPyV oncoprotein (T-antigens) have been correlated with MCC tumor burden. The present study assesses the clinical utility of MCPyV-oncoprotein antibody titers for MCC prognostication and surveillance. METHODS: MCPyV-oncoprotein antibody detection was optimized in a clinical laboratory. A cohort of 219 patients with newly diagnosed MCC were followed prospectively (median follow-up, 1.9 years). Among the seropositive patients, antibody titer and disease status were serially tracked. RESULTS: Antibodies to MCPyV oncoproteins were rare among healthy individuals (1%) but were present in most patients with MCC (114 of 219 patients [52%]; P < .01). Seropositivity at diagnosis independently predicted decreased recurrence risk (hazard ratio, 0.58; P = .04) in multivariate analyses adjusted for age, sex, stage, and immunosuppression. After initial treatment, seropositive patients whose disease did not recur had rapidly falling titers that became negative by a median of 8.4 months. Among seropositive patients who underwent serial evaluation (71 patients; 282 time points), an increasing oncoprotein titer had a positive predictive value of 66% for clinically evident recurrence, whereas a decreasing titer had a negative predictive value of 97%. CONCLUSIONS: Determination of oncoprotein antibody titer assists in the clinical management of patients with newly diagnosed MCC by stratifying them into a higher risk seronegative cohort, in which radiologic imaging may play a more prominent role, and into a lower risk seropositive cohort, in which disease status can be tracked in part by oncoprotein antibody titer. Cancer 2017;123:1464-1474. © 2016 American Cancer Society.


Asunto(s)
Anticuerpos Antivirales/inmunología , Carcinoma de Células de Merkel/diagnóstico , Carcinoma de Células de Merkel/etiología , Proteínas Oncogénicas Virales/inmunología , Anciano , Anciano de 80 o más Años , Anticuerpos Antivirales/sangre , Biomarcadores , Carcinoma de Células de Merkel/epidemiología , Carcinoma de Células de Merkel/mortalidad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia , Estadificación de Neoplasias , Vigilancia de la Población , Estudios Prospectivos , Reproducibilidad de los Resultados , Estudios Seroepidemiológicos , Infecciones Tumorales por Virus/complicaciones , Infecciones Tumorales por Virus/inmunología , Infecciones Tumorales por Virus/virología
4.
Proc Natl Acad Sci U S A ; 108(17): 6757-62, 2011 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-21502524

RESUMEN

Unique chemical methodology enables the synthesis of innovative and diverse scaffolds and chemotypes and allows access to previously unexplored "chemical space." Compound collections based on such new synthetic methods can provide small-molecule probes of proteins and/or pathways whose functions are not fully understood. We describe the identification, characterization, and evolution of two such probes. In one example, a pathway-based screen for DNA damage checkpoint inhibitors identified a compound, MARPIN (ATM and ATR pathway inhibitor) that sensitizes p53-deficient cells to DNA-damaging agents. Modification of the small molecule and generation of an immobilized probe were used to selectively bind putative protein target(s) responsible for the observed activity. The second example describes a focused library approach that relied on tandem multicomponent reaction methodologies to afford a series of modulators of the heat shock protein 70 (Hsp70) molecular chaperone. The synthesis of libraries based on the structure of MAL3-101 generated a collection of chemotypes, each modulating Hsp70 function, but exhibiting divergent pharmacological activities. For example, probes that compromise the replication of a disease-associated polyomavirus were identified. These projects highlight the importance of chemical methodology development as a source of small-molecule probes and as a drug discovery starting point.


Asunto(s)
Diseño de Fármacos , Proteínas HSP70 de Choque Térmico/antagonistas & inhibidores , Sondas Moleculares , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Proteínas de Unión al ADN/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Humanos , Sondas Moleculares/síntesis química , Sondas Moleculares/química , Sondas Moleculares/farmacología , Poliomavirus/fisiología , Infecciones por Polyomavirus/tratamiento farmacológico , Infecciones por Polyomavirus/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas Supresoras de Tumor/metabolismo , Replicación Viral/efectos de los fármacos
5.
Proc Natl Acad Sci U S A ; 108(33): 13716-21, 2011 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-21844338

RESUMEN

Multiple human epidemiologic studies link caffeinated (but not decaffeinated) beverage intake with significant decreases in several types of cancer, including highly prevalent UV-associated skin carcinomas. The mechanism by which caffeine protects against skin cancer is unknown. Ataxia telangiectasia and Rad3-related (ATR) is a replication checkpoint kinase activated by DNA stresses and is one of several targets of caffeine. Suppression of ATR, or its downstream target checkpoint kinase 1 (Chk1), selectively sensitizes DNA-damaged and malignant cells to apoptosis. Agents that target this pathway are currently in clinical trials. Conversely, inhibition of other DNA damage response pathways, such as ataxia telangiectasia mutated (ATM) and BRCA1, promotes cancer. To determine the effect of replication checkpoint inhibition on carcinogenesis, we generated transgenic mice with diminished ATR function in skin and crossed them into a UV-sensitive background, Xpc(-/-). Unlike caffeine, this genetic approach was selective and had no effect on ATM activation. These transgenic mice were viable and showed no histological abnormalities in skin. Primary keratinocytes from these mice had diminished UV-induced Chk1 phosphorylation and twofold augmentation of apoptosis after UV exposure (P = 0.006). With chronic UV treatment, transgenic mice remained tumor-free for significantly longer (P = 0.003) and had 69% fewer tumors at the end of observation of the full cohort (P = 0.019), compared with littermate controls with the same genetic background. This study suggests that inhibition of replication checkpoint function can suppress skin carcinogenesis and supports ATR inhibition as the relevant mechanism for the protective effect of caffeinated beverage intake in human epidemiologic studies.


Asunto(s)
Proteínas de Ciclo Celular/antagonistas & inhibidores , Queratinocitos/efectos de la radiación , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Neoplasias Cutáneas/prevención & control , Rayos Ultravioleta/efectos adversos , Animales , Apoptosis , Proteínas de la Ataxia Telangiectasia Mutada , Cafeína/farmacología , Proteínas de Ciclo Celular/genética , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Queratinocitos/citología , Ratones , Ratones Transgénicos , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Neoplasias Cutáneas/etiología , Neoplasias Cutáneas/patología
6.
J Chin Med Assoc ; 84(5): 459-464, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33871392

RESUMEN

Dry eye disease (DED), also called the keratoconjunctivitis sicca, is one of the most common diseases in the ophthalmology clinics. While DED is not a life-threatening disease, life quality may be substantially affected by the discomfort and the complications of poor vision. As such, a large number of studies have made contributions to the investigation of the DED pathogenesis and novel treatments. DED is a multifactorial disease featured with various phenotypic consequences; therefore, animal models are valuable tools suitable for the related studies. Accordingly, selection of the animal model to recapitulate the clinical presentation of interest is important for appropriately addressing the research objective. To this end, we systemically reviewed different murine and rabbit models of DED, which are categorized into the quantitative (aqueous-deficient) type and the qualitative (evaporative) type, based on the schemes to establish. The clinical manifestations of dry eye on animal models can be induced by mechanical or surgical approaches, iatrogenic immune response, topical eye drops, blockage of neural pathway, or others. Although these models have shown promising results, each has its own limitation and cannot fully reproduce the pathophysiological mechanisms that occur in patients. Nonetheless, the animal models remain the best approximation of human DED and represent the valuable tool for the DED studies.


Asunto(s)
Modelos Animales de Enfermedad , Síndromes de Ojo Seco , Animales , Síndromes de Ojo Seco/tratamiento farmacológico , Síndromes de Ojo Seco/etiología , Síndromes de Ojo Seco/fisiopatología , Ratones , Conejos
7.
Photochem Photobiol ; 96(3): 478-499, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32119110

RESUMEN

Ultraviolet (UV) radiation is among the most prevalent environmental factors that influence human health and disease. Even 1 h of UV irradiation extensively damages the genome. To cope with resulting deleterious DNA lesions, cells activate a multitude of DNA damage response pathways, including DNA repair. Strikingly, UV-induced DNA damage formation and repair are affected by chromatin state. When cells enter S phase with these lesions, a distinct mutation signature is created via error-prone translesion synthesis. Chronic UV exposure leads to high mutation burden in skin and consequently the development of skin cancer, the most common cancer in the United States. Intriguingly, UV-induced oxidative stress has opposing effects on carcinogenesis. Elucidating the molecular mechanisms of UV-induced DNA damage responses will be useful for preventing and treating skin cancer with greater precision. Excitingly, recent studies have uncovered substantial depth of novel findings regarding the molecular and cellular consequences of UV irradiation. In this review, we will discuss updated mechanisms of UV-induced DNA damage responses including the ATR pathway, which maintains genome integrity following UV irradiation. We will also present current strategies for preventing and treating nonmelanoma skin cancer, including ATR pathway inhibition for prevention and photodynamic therapy for treatment.


Asunto(s)
Daño del ADN , Neoplasias Cutáneas/prevención & control , Neoplasias Cutáneas/terapia , Piel/efectos de la radiación , Rayos Ultravioleta , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Reparación del ADN , Replicación del ADN , Humanos , Dímeros de Pirimidina/metabolismo , Piel/metabolismo , Transcripción Genética/efectos de la radiación
8.
Sci Rep ; 10(1): 7908, 2020 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-32404939

RESUMEN

Most systemic lupus erythematosus (SLE) patients are photosensitive and ultraviolet B light (UVB) exposure worsens cutaneous disease and precipitates systemic flares of disease. The pathogenic link between skin disease and systemic exacerbations in SLE remains elusive. In an acute model of UVB-triggered inflammation, we observed that a single UV exposure triggered a striking IFN-I signature not only in the skin, but also in the blood and kidneys. The early IFN-I signature was significantly higher in female compared to male mice. The early IFN-I response in the skin was almost entirely, and in the blood partly, dependent on the presence of cGAS, as was skin inflammatory cell infiltration. Inhibition of cGAMP hydrolysis augmented the UVB-triggered IFN-I response. UVB skin exposure leads to cGAS-activation and both local and systemic IFN-I signature and could contribute to acute flares of disease in susceptible subjects such as patients with SLE.


Asunto(s)
Exposición a Riesgos Ambientales , Interferón Tipo I/biosíntesis , Nucleotidiltransferasas/metabolismo , Rayos Ultravioleta/efectos adversos , Animales , Citocinas/metabolismo , Dermatitis/etiología , Dermatitis/metabolismo , Dermatitis/patología , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Femenino , Humanos , Mediadores de Inflamación/metabolismo , Masculino , Ratones , Ratones Noqueados , Nucleotidiltransferasas/genética , Factores Sexuales , Piel/metabolismo , Piel/patología , Piel/efectos de la radiación
9.
Int J Biol Sci ; 15(5): 1080-1090, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31182927

RESUMEN

Up-regulation of ASB6 has been previously associated with late-stage and poor prognosis of oral squamous cell carcinoma (OSCC) patients. To explore the cellular and molecular basis of how ASB6 enhances the malignancy of OSCC, we employed the clonogenicity and migration assays, murine pulmonary metastasis model, Western blot, and immunofluorescence microscopy to characterize the phenotypes of OSCC cells with lentiviral-based stable overexpression or knockdown of ASB6. We found that ASB6 overexpression increases, whereas ASB6 knockdown decreases, the potential of tumor-sphere formation, colony formation, and expression of Oct-4 and Nanog. While knockdown of ASB6 decreases cell migration in vitro and lung metastasis in mice, the migratory potential was however not promoted by ASB6 overexpression. ASB6 knockdown down-regulates the level of vimentin, and the loss of filopodia formation became more prominent following CRISPR/Cas9-directed knockout of ASB6. Moreover, ASB6 was up-regulated when cells were grown in selective condition featured with a collateral effect of enhancing intracellular stress, and the level of endoplasmic reticulum (ER) stress was further increased by knockdown of ASB6. Thus, ASB6 may attenuate ER stress that would otherwise accumulate and subsequently impede the potential of cells to acquire or sustain the stemness properties and metastatic capacity, thereby enhancing the malignancy of OSCC by increasing the population of cancer stem or stem-like cells.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Regulación Neoplásica de la Expresión Génica/fisiología , Animales , Biomarcadores de Tumor/genética , Western Blotting , Carcinoma de Células Escamosas , Línea Celular Tumoral , Movimiento Celular/genética , Movimiento Celular/fisiología , Estrés del Retículo Endoplásmico/genética , Estrés del Retículo Endoplásmico/fisiología , Técnica del Anticuerpo Fluorescente , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Ratones , Microscopía Confocal , Proteínas Supresoras de la Señalización de Citocinas/genética , Proteínas Supresoras de la Señalización de Citocinas/metabolismo
10.
Cell Rep ; 19(1): 162-174, 2017 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-28380355

RESUMEN

Ultraviolet (UV) radiation is a carcinogen that generates DNA lesions. Here, we demonstrate an unexpected role for DGCR8, an RNA binding protein that canonically functions with Drosha to mediate microRNA processing, in the repair of UV-induced DNA lesions. Treatment with UV induced phosphorylation on serine 153 (S153) of DGCR8 in both human and murine cells. S153 phosphorylation was critical for cellular resistance to UV, the removal of UV-induced DNA lesions, and the recovery of RNA synthesis after UV exposure but not for microRNA expression. The RNA-binding and Drosha-binding activities of DGCR8 were not critical for UV resistance. DGCR8 depletion was epistatic to defects in XPA, CSA, and CSB for UV sensitivity. DGCR8 physically interacted with CSB and RNA polymerase II. JNKs were involved in the UV-induced S153 phosphorylation. These findings suggest that UV-induced S153 phosphorylation mediates transcription-coupled nucleotide excision repair of UV-induced DNA lesions in a manner independent of microRNA processing.


Asunto(s)
Daño del ADN , MicroARNs/metabolismo , Proteínas de Unión al ARN/metabolismo , Ribonucleasa III/metabolismo , Animales , Anisomicina/metabolismo , Antracenos/metabolismo , ADN/metabolismo , ADN/efectos de la radiación , Reparación del ADN , Células HCT116 , Células HeLa , Humanos , MAP Quinasa Quinasa 4/antagonistas & inhibidores , MAP Quinasa Quinasa 4/metabolismo , Ratones , Fosforilación , ARN Polimerasa II/metabolismo , Proteínas de Unión al ARN/genética , Ribonucleasa III/genética , Rayos Ultravioleta
11.
Mol Neurobiol ; 30(2): 185-200, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15475626

RESUMEN

Alzheimer's disease is characterized pathologically by senile plaques in the brain. The major component of senile plaques is amyloid-beta (Abeta), which is cleaved from Alzheimer's Abeta protein precursor (AbetaPP). Recently, information regarding the cytoplasmic tail of AbetaPP has started to emerge, opening up various insights into the physiological roles of AbetaPP and its pathological role in Alzheimer's disease. The cytoplasmic domain of AbetaPP shares the evolutionarily conserved GYENPTY motif, which binds to a number of adaptor proteins containing the phosphotyrosine interaction domain (PID). Among the PID-containing proteins, this article focuses on four groups of adaptor proteins of AbetaPP: Fe65, X11, mDab1, and c-Jun N-terminal kinase-interacting protein 1b/islet-brain 1.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Citoplasma/metabolismo , Enfermedad de Alzheimer/genética , Secuencia de Aminoácidos , Precursor de Proteína beta-Amiloide/genética , Animales , Citoplasma/genética , Humanos , Datos de Secuencia Molecular
12.
Cancer Res ; 74(24): 7534-45, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25336189

RESUMEN

Resistance to DNA-damaging chemotherapy is a barrier to effective treatment that appears to be augmented by p53 functional deficiency in many cancers. In p53-deficient cells in which the G1-S checkpoint is compromised, cell viability after DNA damage relies upon intact intra-S and G2-M checkpoints mediated by the ATR (ataxia telangiectasia and Rad3 related) and Chk1 kinases. Thus, a logical rationale to sensitize p53-deficient cancers to DNA-damaging chemotherapy is through the use of ATP-competitive inhibitors of ATR or Chk1. To discover small molecules that may act on uncharacterized components of the ATR pathway, we performed a phenotype-based screen of 9,195 compounds for their ability to inhibit hydroxyurea-induced phosphorylation of Ser345 on Chk1, known to be a critical ATR substrate. This effort led to the identification of four small-molecule compounds, three of which were derived from known bioactive library (anthothecol, dihydrocelastryl, and erysolin) and one of which was a novel synthetic compound termed MARPIN. These compounds all inhibited ATR-selective phosphorylation and sensitized p53-deficient cancer cells to DNA-damaging agents in vitro and in vivo. Notably, these compounds did not inhibit ATR catalytic activity in vitro, unlike typical ATP-competitive inhibitors, but acted in a mechanistically distinct manner to disable ATR-Chk1 function. Our results highlight a set of novel molecular probes to further elucidate druggable mechanisms to improve cancer therapeutic responses produced by DNA-damaging drugs.


Asunto(s)
Neoplasias/genética , Proteínas Quinasas/biosíntesis , Proteína p53 Supresora de Tumor/genética , Proteínas de la Ataxia Telangiectasia Mutada/antagonistas & inhibidores , Proteínas de la Ataxia Telangiectasia Mutada/biosíntesis , Catálisis/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Daño del ADN/efectos de los fármacos , Puntos de Control de la Fase G2 del Ciclo Celular , Células HeLa , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Proteínas Quinasas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/administración & dosificación
13.
Front Oncol ; 3: 144, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23785666

RESUMEN

Sunlight-induced non-melanoma skin cancer is the most prevalent cancer in the United States with more than two million cases per year. Several studies have shown an inhibitory effect of caffeine administration on UVB-induced skin cancer in mice, and these studies are paralleled by epidemiology studies that indicate an inhibitory effect of coffee drinking on non-melanoma skin cancer in humans. Strikingly, decaffeinated coffee consumption had no such inhibitory effect. Mechanism studies indicate that caffeine has a sunscreen effect that inhibits UVB-induced formation of thymine dimers and sunburn lesions in the epidermis of mice. In addition, caffeine administration has a biological effect that enhances UVB-induced apoptosis thereby enhancing the elimination of damaged precancerous cells, and caffeine administration also enhances apoptosis in tumors. Caffeine administration enhances UVB-induced apoptosis by p53-dependent and p53-independent mechanisms. Exploration of the p53-independent effect indicated that caffeine administration enhanced UVB-induced apoptosis by inhibiting the UVB-induced increase in ATR-mediated formation of phospho-Chk1 (Ser345) and abolishing the UVB-induced decrease in cyclin B1 which resulted in caffeine-induced premature and lethal mitosis in mouse skin. In studies with cultured primary human keratinocytes, inhibition of ATR with siRNA against ATR inhibited Chk1 phosphorylation and enhanced UVB-induced apoptosis. Transgenic mice with decreased epidermal ATR function that were irradiated chronically with UVB had 69% fewer tumors at the end of the study compared with irradiated littermate controls with normal ATR function. These results, which indicate that genetic inhibition of ATR (like pharmacologic inhibition of ATR via caffeine) inhibits UVB-induced carcinogenesis support the concept that ATR-mediated phosphorylation of Chk1 is an important target for caffeine's inhibitory effect on UVB-induced carcinogenesis.

14.
J Invest Dermatol ; 129(7): 1805-15, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19242509

RESUMEN

New approaches to prevent and reverse UV damage are needed to combat rising sunlight-induced skin cancer rates. Mouse studies have shown that oral or topical caffeine promotes elimination of UV-damaged keratinocytes through apoptosis and markedly inhibits subsequent skin cancer development. This potentially important therapeutic effect has not been studied in human skin cells. Here, we use primary human keratinocytes to examine which of several caffeine effects mediates this process. In these cells, caffeine more than doubled apoptosis after 75 mJ cm(-2) of ultraviolet light B (UVB). Selectively targeting two of caffeine's known effects did not alter UVB-induced apoptosis: inhibition of ataxia-telangiectasia mutated and augmentation of cyclic AMP levels. In contrast, siRNA against ataxia-telangiectasia and Rad3-related (ATR) doubled apoptosis after UV through a p53-independent mechanism. Caffeine did not further augment apoptosis after UVB in cells in which ATR had been specifically depleted, suggesting that a key target of caffeine in this effect is ATR. Inhibition of a central ATR target, checkpoint kinase 1 (Chk1), through siRNA or a new and highly specific inhibitor (PF610666) also augmented UVB-induced apoptosis. These data suggest that a relevant target of caffeine is the ATR-Chk1 pathway and that inhibiting ATR or Chk1 might have promise in preventing or reversing UV damage.


Asunto(s)
Apoptosis , Cafeína/farmacología , Proteínas de Ciclo Celular/metabolismo , Queratinocitos/metabolismo , Queratinocitos/efectos de la radiación , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Rayos Ultravioleta/efectos adversos , Adulto , Anciano , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Apoptosis/efectos de la radiación , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/genética , Células Cultivadas , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Células Epidérmicas , Femenino , Humanos , Queratinocitos/citología , Inhibidores de Fosfodiesterasa/farmacología , Fosforilación/efectos de los fármacos , Fosforilación/efectos de la radiación , Proteínas Quinasas/genética , Proteínas Serina-Treonina Quinasas/genética , ARN Interferente Pequeño , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
15.
J Invest Dermatol ; 127(7): 1577-84, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17568801

RESUMEN

Chemical genetics employs diverse small-molecule compounds to elucidate biological processes in a manner analogous to the mutagenesis strategies at the core of classical genetics. Screening small-molecule libraries for compounds that induce a phenotype of interest represents the forward chemical genetic approach, whereas the reverse approach involves small molecules targeting a single protein. Here, we review key differences between the goals for small-molecule screening in industry versus academia, recent developments in high-throughput screening, and publicly available resources of compound collections, screening facilities, and databases. A particularly exciting outcome of a chemical genetic screen is the discovery of a previously unknown role for a protein in a pathway together with compounds that affect the function of that protein. In illustrative cases, such discoveries have led to progress toward therapeutic development and more commonly have increased the size of the small molecule "toolbox" available to the research community for the study of biological processes.


Asunto(s)
Pruebas Genéticas/métodos , Terapia Genética/métodos , Análisis por Micromatrices , Biología Molecular/métodos , Animales , Animales Modificados Genéticamente , Bases de Datos Genéticas , Diagnóstico por Imagen , Pruebas Genéticas/tendencias , Terapia Genética/tendencias , Genotipo , Humanos , Biología Molecular/tendencias , Mutagénesis , Fenotipo
16.
J Neurosci Res ; 81(5): 720-9, 2005 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16035108

RESUMEN

Amyotrophic lateral sclerosis (ALS) is the most common fatal motor neuron disease. It has been generally accepted that the proapoptotic property of the familial ALS (FALS)-linked mutant SOD1 genes plays an important role in the pathogenesis of some FALS cases. We found here that expression of N19S-SOD1, a novel SOD1 mutant originally found in a sporadic ALS patient, induces lower grade death in NSC34 cells than FALS-linked mutant SOD1. In agreement, intracytoplasmic aggregate formation and SOD1 polymerization are less prominently induced by ectopic expression of N19S-SOD1 than FALS-linked mutant SOD1. We further found that additional cell stresses, such as inhibition of proteasomal activity or up-regulation of intracellular oxidative stress, enhance N19S-SOD1-induced aggregate formation and polymerization of N19S-SOD1. Such analysis of the intracellular polymerization and the ubiquitination of N19S-SOD1 have further suggested that it is recognized as a misfolded protein, like FALS-linked mutant SOD1, whereas wild-type SOD1 is not. Altogether, it is speculated that the N19S mutation of SOD1 in cooperation with associated cell stresses contributes to the onset of ALS as a risk factor.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Neuronas Motoras/patología , Superóxido Dismutasa/metabolismo , Esclerosis Amiotrófica Lateral/patología , Animales , Células CHO , Muerte Celular/fisiología , Cricetinae , Humanos , Immunoblotting , Ratones , Mutación , Estrés Oxidativo , Superóxido Dismutasa/genética , Superóxido Dismutasa-1 , Transfección
17.
J Neurosci Res ; 79(5): 714-23, 2005 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-15678515

RESUMEN

Humanin (HN) is a 24-amino acid peptide that protects neuronal cells from death caused by Alzheimer's disease (AD)-related genes and amyloid-beta (Abeta). Multiple studies have revealed its biochemical and neuroprotective characteristics in vitro; however, little has been known regarding whether HN is effective in vivo in AD model systems. We examined the effect of S14G-HN, a 1,000-fold more potent derivative of HN in vitro, on amnesia induced by Abeta25-35 in mice. The Y-maze test revealed that at least 50 pmol of S14G-HN by intracerebroventricular injection prevented Abeta-induced impairment of short-term/spatial working memory; however, 5 nmol of S14A-HN, a neuroprotection-defective mutant in vitro, did not prevent Abeta-induced amnesia. These results are in agreement with the structure-function correlation shown previously in vitro. In the water-finding task, S14G-HN prevented prolongation of finding latency (the time to find water) observed in Abeta-amnesic mice, indicating that S14G-HN also blocked Abeta-induced impairment of latent learning. In accordance with these observations, immunohistochemical analysis showed that S14G-HN sustained the number of cholinergic neurons in the basal forebrain and the striata nearly to the normal level. Furthermore, genistein, a specific inhibitor of tyrosine kinases, blocked recovery from scopolamine-induced amnesia by S14G-HN, suggesting that certain tyrosine kinase(s) are involved in the inhibitory function of S14G-HN in vivo. Taking these findings together, we conclude that S14G-HN has rescue activity against memory impairment caused by AD-related insults in vivo by activating the same intracellular neuroprotective machinery as elucidated previously in vitro.


Asunto(s)
Trastornos de la Memoria/prevención & control , Fármacos Neuroprotectores/uso terapéutico , Proteínas/uso terapéutico , Amnesia/inducido químicamente , Amnesia/tratamiento farmacológico , Péptidos beta-Amiloides/toxicidad , Análisis de Varianza , Animales , Conducta Animal , Encéfalo/anatomía & histología , Encéfalo/metabolismo , Recuento de Células/métodos , Colina O-Acetiltransferasa , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Inhibidores Enzimáticos/farmacología , Conducta Exploratoria/efectos de los fármacos , Genisteína/farmacología , Humanos , Inmunohistoquímica/métodos , Inyecciones Intraventriculares/métodos , Péptidos y Proteínas de Señalización Intracelular , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Trastornos de la Memoria/inducido químicamente , Memoria a Corto Plazo/efectos de los fármacos , Ratones , Fármacos Neuroprotectores/química , Proteínas/química , Tiempo de Reacción/efectos de los fármacos , Escopolamina , Percepción Espacial/efectos de los fármacos
18.
Neurosignals ; 11(5): 236-50, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12566925

RESUMEN

To develop a therapeutic intervention for Alzheimer's disease (AD), it is necessary to clarify the mechanisms underlying the pathogenesis of AD, in which senile plaques, neurofibrillary tangles and neuronal loss in the cerebrum are the central abnormalities. A number of studies have focused on the major component of the senile plaques, which is amyloid-beta (Abeta) and its precursor protein APP, and have investigated the roles of these molecules in the onset, progression and inhibition of AD. For multiple reasons, however, their roles in AD, especially in neuronal death, remain elusive and a unified concept for their roles has not yet been established. Recently, it has been found that APP functions normally as a neuronal surface transmembrane protein. In this article, we review the molecular mechanisms of neuronal cell death by these APP-relevant insults and discuss the functions of APP in regard to intracellular signal transducers, including c-Jun N-terminal kinase. We also revise the roles of Abeta in neuronal death and survival.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Precursor de Proteína beta-Amiloide/fisiología , Neuronas/patología , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/toxicidad , Animales , Muerte Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Noqueados , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Neurotoxinas
19.
Eur J Neurosci ; 19(10): 2826-38, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15147316

RESUMEN

While the exact aetiology of Alzheimer's disease (AD) is unknown, distinct genetic mutations have been identified for the rare cases of familial AD (FAD). V642I mutation in amyloid precursor protein (APP) co-segregates with FAD with perfect penetration, and the clinicopathological characteristics of patients with this mutation resemble that of sporadic AD. To examine the pathogenic process of this FAD-linked trait in vivo, we produced a mouse with the corresponding point mutation in the APP gene using homologous recombination and Cre-loxP site-specific recombination ('knock-in' technique). Mice with the heterozygous V642I-APP allele most precisely reflected the genotype of humans bearing this mutation. For the observation period of 2.5 years the mutants stayed apparently indistinguishable from the wild-type littermates. However, behavioural analysis revealed significantly deteriorated long-term memory in mutants when examined for the retention of spatial attention. Interestingly, acquisition of spatial memory was slightly affected but short-term working memory was not deteriorated at all. Histological examination was negative for formation of neuritic plaques or neurofibrillary tangles, whereas the relative amount of longer form of beta-amyloid species A beta 42(43) was significantly increased against that of the shorter form (A beta 40) in the mutant brain homogenates. We conclude that a V642I-APP mutant allele in aged mice confers functional components, but not organic components, of the AD-related phenotype that are observed in the early stage of AD. This V642I-APP knock-in mutant line may serve as a model to study the early pathogenic processes of AD in vivo and to develop therapeutics for this stage.


Asunto(s)
Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Mutación , Factores de Edad , Alelos , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/fisiología , Animales , Conducta Animal , Southern Blotting/métodos , Química Encefálica , Conducta de Elección/fisiología , Modelos Animales de Enfermedad , Femenino , Genotipo , Humanos , Inmunohistoquímica/métodos , Isoleucina/genética , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Actividad Motora/genética , Ovillos Neurofibrilares , Fragmentos de Péptidos/metabolismo , ARN Mensajero/biosíntesis , Tiempo de Reacción/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Factores Sexuales , Tasa de Supervivencia , Valina/genética , Proteínas tau/metabolismo
20.
Eur J Neurosci ; 17(6): 1150-8, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12670303

RESUMEN

Humanin (HN) is a newly identified neuroprotective peptide that specifically suppresses Alzheimer's disease (AD)-related neurotoxicity. HN peptide has been detected in the human AD brain as well as in mouse testis and colon by immunoblot and immunohistochemical analyses. By means of yeast two-hybrid screening, we identified TRIM11 as a novel HN-interacting protein. TRIM11, which is a member of protein family containing a tripartite motif (TRIM), is composed of a RING finger domain, which is a putative E3 ubiquitin ligase, a B-box domain, a coiled-coil domain and a B30.2 domain. Deletion of the B30.2 domain in TRIM11 abolished the interaction with HN, whereas the B30.2 domain alone did not interact with HN. For their interaction, at least the coiled-coil domain was indispensable together with the B30.2 domain. The intracellular level of glutathione S-transferase-fused or EGFP-fused HN peptides or plain HN was drastically reduced by the coexpression of TRIM11. Disruption of the RING finger domain by deleting the first consensus cysteine or proteasome inhibitor treatment significantly diminished the effect of TRIM11 on the intracellular level of HN. These results suggest that TRIM11 plays a role in the regulation of intracellular HN level through ubiquitin-mediated protein degradation pathways.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Enfermedad de Alzheimer/metabolismo , Proteínas Portadoras/metabolismo , Proteínas de la Membrana/metabolismo , Fármacos Neuroprotectores/metabolismo , Proteínas/metabolismo , Animales , Immunoblotting , Péptidos y Proteínas de Señalización Intracelular , Ratones , Reacción en Cadena de la Polimerasa , Técnicas del Sistema de Dos Híbridos , Ubiquitina/metabolismo , Levaduras
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA