Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(13): e2313652121, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38498709

RESUMEN

Huntington's disease (HD) is an inherited neurodegenerative disorder caused by an expanded CAG repeat in the huntingtin (HTT) gene. The repeat-expanded HTT encodes a mutated HTT (mHTT), which is known to induce DNA double-strand breaks (DSBs), activation of the cGAS-STING pathway, and apoptosis in HD. However, the mechanism by which mHTT triggers these events is unknown. Here, we show that HTT interacts with both exonuclease 1 (Exo1) and MutLα (MLH1-PMS2), a negative regulator of Exo1. While the HTT-Exo1 interaction suppresses the Exo1-catalyzed DNA end resection during DSB repair, the HTT-MutLα interaction functions to stabilize MLH1. However, mHTT displays a significantly reduced interaction with Exo1 or MutLα, thereby losing the ability to regulate Exo1. Thus, cells expressing mHTT exhibit rapid MLH1 degradation and hyperactive DNA excision, which causes severe DNA damage and cytosolic DNA accumulation. This activates the cGAS-STING pathway to mediate apoptosis. Therefore, we have identified unique functions for both HTT and mHTT in modulating DNA repair and the cGAS-STING pathway-mediated apoptosis by interacting with MLH1. Our work elucidates the mechanism by which mHTT causes HD.


Asunto(s)
Enfermedad de Huntington , Humanos , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Proteínas Mutantes/genética , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Nucleotidiltransferasas/genética , ADN , Apoptosis/genética , Homólogo 1 de la Proteína MutL/genética
2.
Gastroenterology ; 164(7): 1232-1247, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36842710

RESUMEN

BACKGROUND & AIMS: Although small patient subsets benefit from current targeted strategies or immunotherapy, gemcitabine remains the first-line drug for pancreatic cancer (PC) treatment. However, gemcitabine resistance is widespread and compromises long-term survival. Here, we identified ubiquitin-conjugating enzyme E2T (UBE2T) as a potential therapeutic target to combat gemcitabine resistance in PC. METHODS: Proteomics and metabolomics were combined to examine the effect of UBE2T on pyrimidine metabolism remodeling. Spontaneous PC mice (LSL-KrasG12D/+, LSL-Trp53R172H/+, Pdx1-Cre; KPC) with Ube2t-conditional knockout, organoids, and large-scale clinical samples were used to determine the effect of UBE2T on gemcitabine efficacy. Organoids, patient-derived xenografts (PDX), and KPC mice were used to examine the efficacy of the combination of a UBE2T inhibitor and gemcitabine. RESULTS: Spontaneous PC mice with Ube2t deletion had a marked survival advantage after gemcitabine treatment, and UBE2T levels were positively correlated with gemcitabine resistance in clinical patients. Mechanistically, UBE2T catalyzes ring finger protein 1 (RING1)-mediated ubiquitination of p53 and relieves the transcriptional repression of ribonucleotide reductase subunits M1 and M2, resulting in unrestrained pyrimidine biosynthesis and alleviation of replication stress. Additionally, high-throughput compound library screening using organoids identified pentagalloylglucose (PGG) as a potent UBE2T inhibitor and gemcitabine sensitizer. The combination of gemcitabine and PGG diminished tumor growth in PDX models and prolonged long-term survival in spontaneous PC mice. CONCLUSIONS: Collectively, UBE2T-mediated p53 degradation confers PC gemcitabine resistance by promoting pyrimidine biosynthesis and alleviating replication stress. This study offers an opportunity to improve PC survival by targeting UBE2T and develop a promising gemcitabine sensitizer in clinical translation setting.


Asunto(s)
Gemcitabina , Neoplasias Pancreáticas , Humanos , Ratones , Animales , Enzimas Ubiquitina-Conjugadoras/genética , Enzimas Ubiquitina-Conjugadoras/metabolismo , Proteína p53 Supresora de Tumor/genética , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Modelos Animales de Enfermedad , Línea Celular Tumoral , Neoplasias Pancreáticas
3.
Nucleic Acids Res ; 50(10): 5635-5651, 2022 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-35580045

RESUMEN

Non-homologous end joining (NHEJ) is the major pathway that mediates the repair of DNA double-strand breaks (DSBs) generated by ionizing radiation (IR). Previously, the DNA helicase RECQL4 was implicated in promoting NHEJ, but its role in the pathway remains unresolved. In this study, we report that RECQL4 stabilizes the NHEJ machinery at DSBs to promote repair. Specifically, we find that RECQL4 interacts with the NHEJ core factor DNA-PKcs and the interaction is increased following IR. RECQL4 promotes DNA end bridging mediated by DNA-PKcs and Ku70/80 in vitro and the accumulation/retention of NHEJ factors at DSBs in vivo. Moreover, interaction between DNA-PKcs and the other core NHEJ proteins following IR treatment is attenuated in the absence of RECQL4. These data indicate that RECQL4 promotes the stabilization of the NHEJ factors at DSBs to support formation of the NHEJ long-range synaptic complex. In addition, we observed that the kinase activity of DNA-PKcs is required for accumulation of RECQL4 to DSBs and that DNA-PKcs phosphorylates RECQL4 at six serine/threonine residues. Blocking phosphorylation at these sites reduced the recruitment of RECQL4 to DSBs, attenuated the interaction between RECQL4 and NHEJ factors, destabilized interactions between the NHEJ machinery, and resulted in decreased NHEJ. Collectively, these data illustrate reciprocal regulation between RECQL4 and DNA-PKcs in NHEJ.


Asunto(s)
Roturas del ADN de Doble Cadena , Proteínas de Unión al ADN , ADN/genética , ADN/metabolismo , Reparación del ADN por Unión de Extremidades , Reparación del ADN , Proteína Quinasa Activada por ADN/genética , Proteína Quinasa Activada por ADN/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Fosforilación , RecQ Helicasas/genética , RecQ Helicasas/metabolismo
4.
Biochem Biophys Res Commun ; 508(4): 1240-1244, 2019 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-30563767

RESUMEN

Genome integrity is important for cell growth, development and proliferation. The E3 ligase RAD18 plays a vital role in the DNA damage response (DDR) to maintain genome integrity. Recent studies reveal that RAD18 has non-ubiquitinated and mono-ubiquitinated form in normal cells. However, whether RAD18 undergoes other post-translational modification remains to be investigated. Here we show that RAD18 is a target of NEDD8, an ubiquitin-like protein. In response to hydrogen peroxide (H2O2)-induced oxidative stress, RAD18 NEDDylation increases significantly, while its ubiquitination decreases. Moreover, NEDD8 overexpression or deNEDDylase NEDP1 deletion further antagonizes RAD18 ubiquitination. In addition, treatment with MLN4924, an inhibitor of NEDD8-activating Enzyme, reduces the interaction between PCNA and RAD18, which blocks the localization of RAD18 to form foci, and thus inhibiting polymerase η recruitment after oxidative stress. Together, our study demonstrates that RAD18 NEDDylation regulates its localization and involves in the DDR pathway by modulating RAD18 ubiquitination.


Asunto(s)
Daño del ADN , Proteínas de Unión al ADN/metabolismo , Proteína NEDD8/metabolismo , Estrés Oxidativo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , ADN Polimerasa Dirigida por ADN/metabolismo , Células HEK293 , Células HeLa , Humanos , Peróxido de Hidrógeno/toxicidad , Transporte de Proteínas , Especificidad por Sustrato
6.
J Biol Chem ; 292(20): 8472-8483, 2017 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-28348081

RESUMEN

Herpes simplex virus 1 (HSV-1) infection manipulates distinct host DNA-damage responses to facilitate virus proliferation, but the molecular mechanisms remain to be elucidated. One possible HSV-1 target might be DNA damage-tolerance mechanisms, such as the translesion synthesis (TLS) pathway. In TLS, proliferating cell nuclear antigen (PCNA) is monoubiquitinated in response to DNA damage-caused replication fork stalling. Ubiquitinated PCNA then facilitates the error-prone DNA polymerase η (polη)-mediated TLS, allowing the fork to bypass damaged sites. Because of the involvement of PCNA ubiquitination in DNA-damage repair, we hypothesized that the function of PCNA might be altered by HSV-1. Here we show that PCNA is a substrate of the HSV-1 deubiquitinase UL36USP, which has previously been shown to be involved mainly in virus uptake and maturation. In HSV-1-infected cells, viral infection-associated UL36USP consistently reduced PCNA ubiquitination. The deubiquitination of PCNA inhibited the formation of polη foci and also increased cell sensitivity to DNA-damage agents. Moreover, the catalytically inactive mutant UL36C40A failed to deubiquitinate PCNA. Of note, the levels of virus marker genes increased strikingly in cells infected with wild-type HSV-1, but only moderately in UL36C40A mutant virus-infected cells, indicating that the UL36USP deubiquitinating activity supports HSV-1 virus replication during infection. These findings suggest a role of UL36USP in the DNA damage-response pathway.


Asunto(s)
Reparación del ADN , Enzimas Desubicuitinizantes/metabolismo , Herpesvirus Humano 1/fisiología , Antígeno Nuclear de Célula en Proliferación/metabolismo , Proteínas Virales/metabolismo , Replicación Viral/fisiología , Sustitución de Aminoácidos , Animales , Chlorocebus aethiops , Daño del ADN , Enzimas Desubicuitinizantes/genética , Células HeLa , Humanos , Mutación Missense , Antígeno Nuclear de Célula en Proliferación/genética , Células Vero , Proteínas Virales/genética
7.
Biochem Cell Biol ; 95(4): 459-467, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28257582

RESUMEN

Stroke is a common cerebrovascular disease in aging populations, and constitutes the second highest principle cause of mortality and the principle cause of permanent disability, and ischemic stroke is the primary form. Osthole is a coumarin derivative extracted from the fruits of Cnidium monnieri (L.) Cusson. In this study, we established a rat model of middle cerebral artery occlusion/reperfusion (MCAO/R) in vivo and found that MCAO/R caused cerebral infarction, hippocampus neuronal injury and apoptosis, and also activated the Notch 1 signaling pathway. However, treatment with osthole further enhanced the activity of Notch 1 signaling and reduced the cerebral infarction as well as the hippocampus neuronal injury and apoptosis induced by MCAO/R in a dose-dependent manner. The same results were observed in a primary neuronal oxygen glucose deficiency/reperfusion (OGD/R) model in vitro, and the effect of osthole could be blocked by an inhibitor of Notch 1 signaling, N-[N-(3,5-difluorophenacetyl)-l-alanyl]-S-phenylglycine tert-butyl ester (DAPT). Therefore, we demonstrated that osthole injection prevented rat ischemia-reperfusion injury via activating the Notch 1 signaling pathway in vivo and in vitro in a dose-dependent manner, which may be significant for clinical treatment of ischemic stroke.


Asunto(s)
Isquemia Encefálica/prevención & control , Cumarinas/farmacología , Receptores Notch/metabolismo , Daño por Reperfusión/metabolismo , Daño por Reperfusión/prevención & control , Transducción de Señal/efectos de los fármacos , Animales , Isquemia Encefálica/metabolismo , Cumarinas/administración & dosificación , Masculino , Ratas , Ratas Sprague-Dawley
8.
Biochem Biophys Res Commun ; 482(4): 632-637, 2017 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-27864145

RESUMEN

Myeloid differentiation factor 88 (MyD88) plays a central role in innate immunity response, however, how its activity is tightly regulated remains largely unknown. In this study, we identify MyD88 as a novel substrate of NEDD8, and demonstrate that MyD88 NEDDylation antagonizes its ubiquitination. Interestingly, in response to the stimulation of IL-1ß, MyD88 NEDDylation is downregulated while its ubiquitination is upregulated. We also show that deNEDDylase NEDP1 serves as a regulator of this process. Furthermore, we demonstrate that NEDD8 negatively regulates the dimerization of MyD88 and suppresses MyD88-dependent NF-κB signaling. Taken together, this study reveals that NEDDylation of MyD88 regulates NF-κB activity through antagonizing its ubiquitination, suggesting a novel mechanism of modulating NF-κB signaling pathway.


Asunto(s)
Factor 88 de Diferenciación Mieloide/inmunología , FN-kappa B/inmunología , Ubiquitinación , Ubiquitinas/inmunología , Endopeptidasas/inmunología , Células HEK293 , Humanos , Inmunidad Innata , Interleucina-1beta/inmunología , Proteína NEDD8 , Transducción de Señal
9.
J Cell Sci ; 127(Pt 10): 2238-48, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24634510

RESUMEN

NEDD8 is an important regulatory factor in many biological processes. However, the substrates for neddylation, and the relationship between the ubiquitin and NEDD8 pathways remain largely unknown. Here, we show that NEDD8 is covalently conjugated to histone 2A (H2A), and that neddylation of H2A antagonizes its ubiquitylation. NEDD8 suppresses ubiquitylation of H2A, and a decreased level of free NEDD8 promotes H2A ubiquitylation. Furthermore, we found that the E3 ligase RNF168 promotes both H2A ubiquitylation and neddylation. Interestingly, RNF168 is itself a substrate for NEDD8, and neddylation of RNF168 is necessary for its E3 ubiquitin activity. Inhibition of RNF168 neddylation impairs the interaction between RNF168 and its E2 enzyme Ubc13 (also known as UBE2N). Moreover, in response to DNA damage, the level of H2A neddylation decreased with an increase in the ubiquitylation of H2A, which facilitates DNA damage repair. During the later stages of damage repair, H2A neddylation increased gradually, whereas ubiquitylation decreased to basal levels. Mechanistically, NEDD8 negatively regulates the DNA damage repair process through suppression of the ubiquitylation of H2A and γH2AX, which further blocks the recruitment of the damage response protein BRCA1. Our findings elucidate the relationship of H2A ubiquitylation and neddylation, and suggest a novel modulatory approach to DNA damage repair through the neddylation pathway.


Asunto(s)
Daño del ADN , Reparación del ADN , Histonas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinas/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Células HEK293 , Células HeLa , Histonas/genética , Humanos , Proteína NEDD8 , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación , Ubiquitinas/genética
10.
Clin Exp Pharmacol Physiol ; 43(1): 125-34, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26385023

RESUMEN

Bone morphogenetic protein (BMP)-7 mediated neuroprotective effect of cerebral ischemic preconditioning (IPC) has been studied in an ischemic animal model, but the underlying cellular mechanisms have not been clearly clarified. In this study, primary cortical neurons and the SH-SY5Y cell line were used to investigate the role of BMP-7 and its downstream signals in the neuroprotective effects of oxygen-glucose deprivation preconditioning (OGDPC). Immunocytochemistry was used to detect the expression of neurofilament in neurons. MTT and lactate dehydrogenase activity assays were used to measure the cytotoxicity. Western blot was used to detect the protein expression of BMP-7 and downstream signals. BMP inhibitor, mitogen-activated protein kinase inhibitors, Smad inhibitor and siRNA of Smad 1 were used to investigate the role of corresponding signalling pathways in the OGDPC. Results showed that OGDPC-induced overexpression of BMP-7 in primary cortical neurons and SH-SY5Y cells. Both of endogenous and exogenous BMP-7 could replicate the neuroprotective effects seen in OGDPC pretreatment. In addition, extracellular regulated protein kinases, p38 and Smad signalling pathway were found to be involved in the neuroprotective effects mediated by OGDPC via BMP-7. This study primarily reveals the cellular mechanisms of the neuroprotection mediated by OGDPC, and provides evidence for better understanding of this intrinsic factor against ischemia.


Asunto(s)
Proteína Morfogenética Ósea 7/metabolismo , Glucosa/deficiencia , Precondicionamiento Isquémico , Sistema de Señalización de MAP Quinasas , Daño por Reperfusión Miocárdica/patología , Neuronas/patología , Oxígeno/metabolismo , Animales , Proteína Morfogenética Ósea 7/genética , Proteína Morfogenética Ósea 7/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Daño por Reperfusión Miocárdica/metabolismo , Neuronas/metabolismo , Neuroprotección/efectos de los fármacos , Ratas , Proteínas Smad/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
11.
Traffic ; 13(6): 790-9, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22348310

RESUMEN

HSCARG is a newly identified nuclear factor-κB (NF-κB) inhibitor that plays important roles in cell growth. Our previous study found that HSCARG could shuttle between the nucleus and cytoplasm by sensing the change in cellular redox states. To further investigate the mechanism of HSCARG translocation and its effect on the regulation of NF-κB activity, we identified a previously uncharacterized nuclear export signal (NES) at residues 272-278 of HSCARG that is required for its cytoplasmic translocation. This leucine-rich NES was found to be mediated by chromosome region maintenance 1. More importantly, accumulation of HSCARG in the nucleus occurred following a mutation in the NES or oxidative stress, which attenuated the inhibition of NF-κB by HSCARG. These results indicate that nucleocytoplasmic translocation of HSCARG plays an important role in fine-tuning NF-κB signaling.


Asunto(s)
Citoplasma/metabolismo , Regulación Neoplásica de la Expresión Génica , Carioferinas/fisiología , FN-kappa B/metabolismo , Receptores Citoplasmáticos y Nucleares/fisiología , Factores de Transcripción/metabolismo , Transporte Activo de Núcleo Celular , Núcleo Celular/metabolismo , Análisis Mutacional de ADN , Células HEK293 , Células HeLa , Humanos , Carioferinas/metabolismo , Leucina/metabolismo , Microscopía Fluorescente/métodos , Modelos Biológicos , Señales de Exportación Nuclear , Oxidación-Reducción , Estrés Oxidativo , Receptores Citoplasmáticos y Nucleares/metabolismo , Transducción de Señal , Proteína Exportina 1
12.
Biochem Biophys Res Commun ; 441(3): 560-6, 2013 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-24184479

RESUMEN

A mild cerebral ischemic insult, also known as ischemic preconditioning (IPC), confers transient tolerance to a subsequent ischemic challenge in the brain. This study was conducted to investigate whether bone morphogenetic protein-7 (BMP-7) is involved in neuroprotection elicited by IPC in a rat model of ischemia. Ischemic tolerance was induced in rats by IPC (15 min middle cerebral artery occlusion, MCAO) at 48 h before lethal ischemia (2h MCAO). The present data showed that IPC increased BMP-7 mRNA and protein expression after 24h reperfusion following ischemia in the brain. In rats of ischemia, IPC-induced reduction of cerebral infarct volume and improvement of neuronal morphology were attenuated when BMP-7 was inhibited either by antagonist noggin or short interfering RNA (siRNA) pre-treatment. Besides, cerebral IPC-induced up-regulation of B-cell lymphoma 2 (Bcl-2) and down-regulation of cleaved caspase-3 at 24h after ischemia/reperfusion (I/R) injury were reversed via inhibition of BMP-7. These findings indicate that BMP-7 mediates IPC-induced tolerance to cerebral I/R, probably through inhibition of apoptosis.


Asunto(s)
Apoptosis , Proteína Morfogenética Ósea 7/fisiología , Encéfalo/irrigación sanguínea , Precondicionamiento Isquémico , Accidente Cerebrovascular/patología , Animales , Proteína Morfogenética Ósea 7/biosíntesis , Proteína Morfogenética Ósea 7/genética , Modelos Animales de Enfermedad , Infarto de la Arteria Cerebral Media , Masculino , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Ratas , Ratas Wistar , Accidente Cerebrovascular/metabolismo
13.
Int J Mol Sci ; 14(2): 3314-24, 2013 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-23385236

RESUMEN

The excision repair cross-complementing rodent repair deficiency complementation group 1 (ERCC1), and X-ray repair cross-complementing group 1 (XRCC1) genes appear to protect mammalian cells from the harmful effects of ionizing radiation. We conducted a large case-control study to investigate the association of polymorphisms in ERCC1 C118T, ERCC1 C8092A, XRCC1 A194T, XRCC1 A194T, and XRCC3 C241T, with glioma risk in a Chinese population. Five single nucleotide polymorphisms (SNPs) were genotyped, using the MassARRAY IPLEX platform, in 443 glioma cases and 443 controls. Association analyses based on an χ2 test and binary logistic regression were performed to determine the odds ratio (OR) and a 95% confidence interval (95% CI) for each SNP. For XRCC1 Arg194Trp, the variant genotype T/T was strongly associated with a lower risk of glioma cancer when compared with the wild type C/C (OR = 2.45, 95% CI = 1.43-4.45). Individuals carrying the XRCC1 399A allele had an increased risk of glioma (OR = 1.33, 95% CI = 1.02-1.64). The XRCC3 241T/T genotype was associated with a strong increased glioma risk (OR = 3.78, 95% CI = 1.86-9.06). Further analysis of the interactions of two susceptibility-associated SNPs, XRCC1 Arg194Trp and XRCC3 Thr241Met, showed that the combination of the XRCC1 194T and XRCC3 241T alleles brought a large increase in glioma risk (OR = 2.75, 95% CI = 1.54-4.04). XRCC1 Arg194Trp, XRCC1 Arg399Gln, and XRCC3 C241T, appear to be associated with susceptibility to glioma in a Chinese population.

14.
NAR Cancer ; 5(3): zcad031, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37325548

RESUMEN

Tumors defective in DNA mismatch repair (dMMR) exhibit microsatellite instability (MSI). Currently, patients with dMMR tumors are benefitted from anti-PD-1/PDL1-based immune checkpoint inhibitor (ICI) therapy. Over the past several years, great progress has been made in understanding the mechanisms by which dMMR tumors respond to ICI, including the identification of mutator phenotype-generated neoantigens, cytosolic DNA-mediated activation of the cGAS-STING pathway, type-I interferon signaling and high tumor-infiltration of lymphocytes in dMMR tumors. Although ICI therapy shows great clinical benefits, ∼50% of dMMR tumors are eventually not responsive. Here we review the discovery, development and molecular basis of dMMR-mediated immunotherapy, as well as tumor resistant problems and potential therapeutic interventions to overcome the resistance.

15.
Cancer Res ; 83(22): 3767-3782, 2023 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-37646571

RESUMEN

The chemotherapeutic agent 5-fluorouracil (5-FU) remains the backbone of postoperative adjuvant treatment for gastric cancer. However, fewer than half of patients with gastric cancer benefit from 5-FU-based chemotherapies owing to chemoresistance and limited clinical biomarkers. Here, we identified the SNF2 protein Polo-like kinase 1-interacting checkpoint helicase (PICH) as a predictor of 5-FU chemosensitivity and characterized a transcriptional function of PICH distinct from its role in chromosome separation. PICH formed a transcriptional complex with RNA polymerase II (Pol II) and ATF4 at the CCNA1 promoter in an ATPase-dependent manner. Binding of the PICH complex promoted cyclin A1 transcription and accelerated S-phase progression. Overexpressed PICH impaired 5-FU chemosensitivity in human organoids and patient-derived xenografts. Furthermore, elevated PICH expression was negatively correlated with survival in postoperative patients receiving 5-FU chemotherapy. Together, these findings reveal an ATPase-dependent transcriptional function of PICH that promotes cyclin A1 transcription to drive 5-FU chemoresistance, providing a potential predictive biomarker of 5-FU chemosensitivity for postoperative patients with gastric cancer and prompting further investigation into the transcriptional activity of PICH. SIGNIFICANCE: PICH binds Pol II and ATF4 in an ATPase-dependent manner to form a transcriptional complex that promotes cyclin A1 expression, accelerates S-phase progression, and impairs 5-FU chemosensitivity in gastric cancer.


Asunto(s)
Neoplasias Gástricas , Humanos , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Resistencia a Antineoplásicos/genética , Ciclina A1 , ADN Helicasas/metabolismo , Fluorouracilo/farmacología , Adenosina Trifosfatasas/uso terapéutico , Quinasa Tipo Polo 1
16.
Acta Neurochir (Wien) ; 154(8): 1469-76; discussion 1476, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22661329

RESUMEN

BACKGROUND: The c-Jun N-terminal kinase (JNK) proteins are encoded by three genes (JNK1, JNK2, and JNK3), giving rise to multiple isoforms via alternative splicing. JNK inhibition using a chemical inhibitor SP600125 confers neuroprotection in an animal model of subarachnoid hemorrhage (SAH). The aim of this study is to investigate whether the protective effects of SP600125 were associated with modulation of tight junction proteins including claudin-5 and ZO-1 and to define which JNK isoforms were involved in the early brain injury after SAH. METHODS: Seventy-five male Sprague Dawley rats (weighing 300-350 g) were randomly assigned to five groups (n = 15): (1) sham, (2) SAH, (3) SAH + DMSO (dimethyl sulfoxide), (4) SAH + 10 mg/kg SP600125, and (5) SAH + 30 mg/kg SP600125. SP600125 or DMSO was injected intraperitoneally 1 h before and 6 h after the induction of SAH. Animals from all the groups were killed 24 h after SAH, and brain tissues were dissected and subjected to electron microscopic examination, Western blot analysis, and histological evaluation. RESULTS: SP600125 pretreatment restored tight junctions and attenuated blood-brain barrier (BBB) disruption and cerebral edema after SAH, coupled with reduced apoptosis in the cerebral cortex. SP600125 exposure restored the reduced expression of both claudin-5 and ZO-1 following SAH and normalized the levels of JNK1 and JNK3. CONCLUSION: Our data demonstrate that the JNK signaling plays an important role in the regulation of tight junction proteins and BBB integrity, and thus represents a promising target against brain injuries after SAH.


Asunto(s)
Antracenos/uso terapéutico , Barrera Hematoencefálica/efectos de los fármacos , Claudina-5/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Hemorragia Subaracnoidea/tratamiento farmacológico , Proteína de la Zonula Occludens-1/metabolismo , Animales , Apoptosis/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Masculino , Microscopía Electrónica de Transmisión , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Hemorragia Subaracnoidea/complicaciones , Hemorragia Subaracnoidea/metabolismo , Hemorragia Subaracnoidea/patología , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/patología , Uniones Estrechas/ultraestructura
17.
Aging (Albany NY) ; 14(1): 462-476, 2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-35017318

RESUMEN

Intracerebral hemorrhage (ICH) is a common neurological condition that causes severe disability and even death. Even though the mechanism is not clear, increasing evidence shows the efficacy of atorvastatin on treating ICH. In this study, we examined the impact of atorvastatin on the NOD-like receptor protein 3 (NLRP3) inflammasome and inflammatory pathways following ICH. Mouse models of ICH were established by collagenase injection in adult C57BL/6 mice. IHC mice received atorvastatin treatment 2 h after hematoma establishment. First, the changes of glial cells and neurons in the brains of ICH patients and mice were detected by immunohistochemistry and western blotting. Second, the molecular mechanisms underlying the microglial activation and neuronal loss were evaluated after the application of atorvastatin. Finally, the behavioral deficits of ICH mice without or with the treatment of atorvastatin were determined by neurological defect scores. The results demonstrated that atorvastatin significantly deactivated glial cells by reducing the expression of glial fibrillary acidic protein (GFAP), Ionized calcium binding adapter molecule 1 (Iba1), tumor necrosis factor (TNF)-α, and interleukin (IL)-6 in ICH model mice. For inflammasomes, atorvastatin also showed its efficacy by decreasing the expression of NLRP3, cleaved caspase-1, and IL-1ß in ICH mice. Moreover, atorvastatin markedly inhibited the upregulation of toll-like receptor 4 (TLR4) and myeloid differentiation factor 88 (MyD88), which indicated deactivation of NLRP3 inflammasomes. By inhibiting the activities of inflammasomes in glial cells, neuronal loss was partially prevented by suppressing the apoptosis in the brains of ICH mice, protecting them from neurological defects.


Asunto(s)
Atorvastatina/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Factor 88 de Diferenciación Mieloide/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Hemorragia Cerebral , Citocinas/genética , Citocinas/metabolismo , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Factor 88 de Diferenciación Mieloide/genética , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Neuronas/efectos de los fármacos , Transducción de Señal , Receptor Toll-Like 4/genética
18.
Cancer Cell ; 39(1): 109-121.e5, 2021 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-33338427

RESUMEN

Tumors with defective mismatch repair (dMMR) are responsive to immunotherapy because of dMMR-induced neoantigens and activation of the cGAS-STING pathway. While neoantigens result from the hypermutable nature of dMMR, it is unknown how dMMR activates the cGAS-STING pathway. We show here that loss of the MutLα subunit MLH1, whose defect is responsible for ~50% of dMMR cancers, results in loss of MutLα-specific regulation of exonuclease 1 (Exo1) during DNA repair. This leads to unrestrained DNA excision by Exo1, which causes increased single-strand DNA formation, RPA exhaustion, DNA breaks, and aberrant DNA repair intermediates. Ultimately, this generates chromosomal abnormalities and the release of nuclear DNA into the cytoplasm, activating the cGAS-STING pathway. In this study, we discovered a hitherto unknown MMR mechanism that modulates genome stability and has implications for cancer therapy.


Asunto(s)
Aberraciones Cromosómicas , Enzimas Reparadoras del ADN/metabolismo , Exodesoxirribonucleasas/metabolismo , Homólogo 1 de la Proteína MutL/deficiencia , Neoplasias/genética , Transducción de Señal , Animales , Línea Celular Tumoral , Roturas del ADN de Cadena Simple , Reparación de la Incompatibilidad de ADN , Reparación del ADN , ADN de Cadena Simple/metabolismo , Células HeLa , Humanos , Proteínas de la Membrana/metabolismo , Ratones , Homólogo 1 de la Proteína MutL/metabolismo , Neoplasias/metabolismo , Nucleotidiltransferasas/metabolismo , Proteína de Replicación A/metabolismo
19.
Cancer Cell ; 39(1): 96-108.e6, 2021 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-33338425

RESUMEN

Increased neoantigens in hypermutated cancers with DNA mismatch repair deficiency (dMMR) are proposed as the major contributor to the high objective response rate in anti-PD-1 therapy. However, the mechanism of drug resistance is not fully understood. Using tumor models defective in the MMR gene Mlh1 (dMLH1), we show that dMLH1 tumor cells accumulate cytosolic DNA and produce IFN-ß in a cGAS-STING-dependent manner, which renders dMLH1 tumors slowly progressive and highly sensitive to checkpoint blockade. In neoantigen-fixed models, dMLH1 tumors potently induce T cell priming and lose resistance to checkpoint therapy independent of tumor mutational burden. Accordingly, loss of STING or cGAS in tumor cells decreases tumor infiltration of T cells and endows resistance to checkpoint blockade. Clinically, downregulation of cGAS/STING in human dMMR cancers correlates with poor prognosis. We conclude that DNA sensing within tumor cells is essential for dMMR-triggered anti-tumor immunity. This study provides new mechanisms and biomarkers for anti-dMMR-cancer immunotherapy.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Proteínas de la Membrana/genética , Homólogo 1 de la Proteína MutL/deficiencia , Neoplasias/genética , Nucleotidiltransferasas/genética , Animales , Línea Celular Tumoral , Reparación de la Incompatibilidad de ADN , Regulación hacia Abajo , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Interferón beta/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Trasplante de Neoplasias , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología , Nucleotidiltransferasas/metabolismo , Pronóstico , Transducción de Señal/efectos de los fármacos
20.
Acta Neurochir (Wien) ; 151(9): 1127-34, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19444374

RESUMEN

BACKGROUND: Although there are still some unresolved aspects, current research has revealed that vascular cell proliferation probably plays an important part in the pathological formation process of cerebral vasospasm. Using a "two-hemorrhage" model of subarachnoid hemorrhage (SAH), this study investigated the function of ERK1/2 and vascular wall cell proliferation in pathological development of cerebral vasospasm. METHODS: Fifty rabbits were randomly divided into five groups: (1) SAH day 1, (2) SAH day 3, (3) SAH day 7, (4) SAH + DMSO (dimethyl sufoxide) solution, (5) SAH + PD98059 (a mitogen-activated protein kinase inhibitor) dissolved in DMSO solution. In the SAH + PD98059/DMSO group and SAH + DMSO control group, PD98059 in DMSO (2 mmol/l) or an equal quantity of DMSO, respectively, was injected into the cisterna magna, once a day from SAH day 1 to day 3. Western protein blotting was used to detect the expression of proliferating cell nuclear antigen (PCNA) and extracellular signal-regulated protein kinases 1 and 2 (ERK1/2) in each group's basilar arteries. Light microscopy and electron microscopy were used for dynamic histological detection at each observation point of the SAH vascular wall under the effects of SAH and the mitogen-activated protein kinase inhibitor. Another 18 rabbits were randomly divided into three groups: SAH, SAH + DMSO and SAH + PD98059/DMSO; cerebral angiograpathy was conducted on SAH days 1 and 7, and the progression of angiographic vasospasm evaluated. RESULTS: Compared with the control group, the extent of vasospasm after SAH increased with time. PD98059 significantly reduced angiographic and morphological vasospasm. In cerebral vasospasm, the expression of T-ERK1/2 showed no significant change. However, expression of p-ERK1/2 and PCNA began to increase significantly on day 3, and achieved a peak on day 7. PD98059 significantly inhibited the expression of p-ERK1/2 and PCNA (p < 0.05). CONCLUSIONS: Cell proliferation on the vascular wall plays an important part in the pathological formation process of cerebral vasospasm. ERK1/2 phosphorylation, as an important signaling pathway, taking part in the process of vascular-wall pathological proliferation of cerebral vasospasm.


Asunto(s)
Proliferación Celular , Hipertrofia/enzimología , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Músculo Liso Vascular/enzimología , Hemorragia Subaracnoidea/complicaciones , Vasoespasmo Intracraneal/enzimología , Animales , Arteria Basilar/citología , Arteria Basilar/enzimología , Modelos Animales de Enfermedad , Activación Enzimática/fisiología , Inhibidores Enzimáticos/farmacología , Flavonoides/farmacología , Hipertrofia/tratamiento farmacológico , Hipertrofia/fisiopatología , Microscopía Electrónica , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/enzimología , Antígeno Nuclear de Célula en Proliferación , Conejos , Regulación hacia Arriba/fisiología , Vasoespasmo Intracraneal/tratamiento farmacológico , Vasoespasmo Intracraneal/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA