Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Cancer Sci ; 113(1): 170-181, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34714577

RESUMEN

The aryl hydrocarbon receptor (AHR) pathway modulates the immune system in response to kynurenine, an endogenous tryptophan metabolite. IDO1 and TDO2 catalyze kynurenine production, which promotes cancer progression by compromising host immunosurveillance. However, it is unclear whether the AHR activation regulates the malignant traits of cancer such as metastatic capability or cancer stemness. Here, we carried out systematic analyses of metabolites in patient-derived colorectal cancer spheroids and identified high levels of kynurenine and TDO2 that were positively associated with liver metastasis. In a mouse colon cancer model, TDO2 expression substantially enhanced liver metastasis, induced AHR-mediated PD-L1 transactivation, and dampened immune responses; these changes were all abolished by PD-L1 knockout. In patient-derived cancer spheroids, TDO2 or AHR activity was required for not only the expression of PD-L1, but also for cancer stem cell (CSC)-related characteristics and Wnt signaling. TDO2 was coexpressed with both PD-L1 and nuclear ß-catenin in colon xenograft tumors, and the coexpression of TDO2 and PD-L1 was observed in clinical colon cancer specimens. Thus, our data indicate that the activation of the TDO2-kynurenine-AHR pathway facilitates liver metastasis of colon cancer via PD-L1-mediated immune evasion and maintenance of stemness.


Asunto(s)
Antígeno B7-H1/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Neoplasias del Colon/patología , Dioxigenasas/metabolismo , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/secundario , Células Madre Neoplásicas/patología , Receptores de Hidrocarburo de Aril/metabolismo , Animales , Línea Celular Tumoral , Neoplasias del Colon/metabolismo , Humanos , Quinurenina , Neoplasias Hepáticas/metabolismo , Ratones , Trasplante de Neoplasias , Células Madre Neoplásicas/metabolismo , Esferoides Celulares/citología , Esferoides Celulares/metabolismo , Escape del Tumor , Regulación hacia Arriba , Vía de Señalización Wnt
2.
Biochem Biophys Res Commun ; 586: 93-99, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34837838

RESUMEN

Dysregulated activation of the WNT/ß-catenin signaling pathway is essential for the initiation and development of various cancers. E7386, a small-molecule compound, attenuates WNT signaling by blocking the interaction between ß-catenin and CREB-binding protein (CBP); hence, it is regarded as a therapeutic candidate for cancers with activated WNT signaling. In the present study, we evaluated the biological characteristics associated with E7386 sensitivity by using a panel of patient-derived colon cancer spheroids. An integrative approach that combined E7386 sensitivity and gene expression profiles revealed that the resistance of the cancer spheroids to E7386 was associated with the activation of the NF-κB pathway. NF-κB pathway inhibitors acted synergistically with E7386 to block proliferation and induce cell cycle arrest in E7386-resistant spheroids. These findings suggest a possibility that a combination of E7386 and NF-κB inhibition may effectively block the proliferation of a subset of colon cancer cells.


Asunto(s)
Proteína de Unión a CREB/genética , FN-kappa B/genética , Fenilendiaminas/farmacología , Pirazinas/farmacología , Esferoides Celulares/efectos de los fármacos , Triazinas/farmacología , beta Catenina/genética , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Proteína de Unión a CREB/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/genética , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Sinergismo Farmacológico , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Cultivo Primario de Células , Esferoides Celulares/metabolismo , Esferoides Celulares/patología , Vía de Señalización Wnt , beta Catenina/metabolismo
3.
BMC Biol ; 19(1): 207, 2021 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-34548081

RESUMEN

BACKGROUND: Intra-tumor heterogeneity (ITH) encompasses cellular differences in tumors and is related to clinical outcomes such as drug resistance. However, little is known about the dynamics of ITH, owing to the lack of time-series analysis at the single-cell level. Mouse models that recapitulate cancer development are useful for controlled serial time sampling. RESULTS: We performed single-cell exome and transcriptome sequencing of 200 cells to investigate how ITH is generated in a mouse colorectal cancer model. In the model, a single normal intestinal cell is grown into organoids that mimic the intestinal crypt structure. Upon RNAi-mediated downregulation of a tumor suppressor gene APC, the transduced organoids were serially transplanted into mice to allow exposure to in vivo microenvironments, which play relevant roles in cancer development. The ITH of the transcriptome increased after the transplantation, while that of the exome decreased. Mutations generated during organoid culture did not greatly change at the bulk-cell level upon the transplantation. The RNA ITH increase was due to the emergence of new transcriptional subpopulations. In contrast to the initial cells expressing mesenchymal-marker genes, new subpopulations repressed these genes after the transplantation. Analyses of colorectal cancer data from The Cancer Genome Atlas revealed a high proportion of metastatic cases in human subjects with expression patterns similar to the new cell subpopulations in mouse. These results suggest that the birth of transcriptional subpopulations may be a key for adaptation to drastic micro-environmental changes when cancer cells have sufficient genetic alterations at later tumor stages. CONCLUSIONS: This study revealed an evolutionary dynamics of single-cell RNA and DNA heterogeneity in tumor progression, giving insights into the mesenchymal-epithelial transformation of tumor cells at metastasis in colorectal cancer.


Asunto(s)
Neoplasias Colorrectales , Animales , Neoplasias Colorrectales/genética , ADN , Exoma/genética , Heterogeneidad Genética , Ratones , ARN , Análisis de Secuencia de ARN , Microambiente Tumoral
4.
Cell Rep Med ; 5(5): 101532, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38670097

RESUMEN

Ovarian clear cell carcinoma (OCCC) is a gynecological cancer with a dismal prognosis; however, the mechanism underlying OCCC chemoresistance is not well understood. To explore the intracellular networks associated with the chemoresistance, we analyze surgical specimens by performing integrative analyses that combine single-cell analyses and spatial transcriptomics. We find that a chemoresistant OCCC subpopulation with elevated HIF activity localizes mainly in areas populated by cancer-associated fibroblasts (CAFs) with a myofibroblastic phenotype, which is corroborated by quantitative immunostaining. CAF-enhanced chemoresistance and HIF-1α induction are recapitulated in co-culture assays, which show that cancer-derived platelet-derived growth factor (PDGF) contributes to the chemoresistance and HIF-1α induction via PDGF receptor signaling in CAFs. Ripretinib is identified as an effective receptor tyrosine kinase inhibitor against CAF survival. In the co-culture system and xenograft tumors, ripretinib prevents CAF survival and suppresses OCCC proliferation in the presence of carboplatin, indicating that combination of conventional chemotherapy and CAF-targeted agents is effective against OCCC.


Asunto(s)
Fibroblastos Asociados al Cáncer , Subunidad alfa del Factor 1 Inducible por Hipoxia , Neoplasias Ováricas , Factor de Crecimiento Derivado de Plaquetas , Transducción de Señal , Femenino , Humanos , Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/patología , Fibroblastos Asociados al Cáncer/efectos de los fármacos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Neoplasias Ováricas/patología , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Ratones , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Progresión de la Enfermedad , Técnicas de Cocultivo , Proliferación Celular/efectos de los fármacos , Ratones Desnudos , Adenocarcinoma de Células Claras/metabolismo , Adenocarcinoma de Células Claras/patología , Adenocarcinoma de Células Claras/tratamiento farmacológico , Adenocarcinoma de Células Claras/genética , Retroalimentación Fisiológica/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Commun Biol ; 6(1): 1183, 2023 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-37985874

RESUMEN

Gastrointestinal tract organs harbor reserve cells, which are endowed with cellular plasticity and regenerate functional units in response to tissue damage. However, whether the reserve cells in gastrointestinal tract exist as long-term quiescent cells remain incompletely understood. In the present study, we systematically examine H2b-GFP label-retaining cells and identify a long-term slow-cycling population in the gastric corpus but not in other gastrointestinal organs. The label-retaining cells, which reside near the basal layers of the corpus, comprise a subpopulation of chief cells. The identified quiescent cells exhibit induction of Atf4 and its target genes including Atf3, a marker of paligenosis, and activation of the unfolded protein response, but do not show elevated expression of Troy, Lgr5, or Mist. External damage to the gastric mucosa induced by indomethacin treatment triggers proliferation of the quiescent Atf4+ population, indicating that the gastric corpus harbors a specific cell population that is primed to facilitate stomach regeneration.


Asunto(s)
Células Principales Gástricas , Células Principales Gástricas/metabolismo , Células Madre/metabolismo , Mucosa Gástrica , Células Epiteliales , Estómago
6.
Cell Rep ; 42(6): 112519, 2023 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-37224811

RESUMEN

Cancer chemoresistance is often attributed to slow-cycling persister populations with cancer stem cell (CSC)-like features. However, how persister populations emerge and prevail in cancer remains obscure. We previously demonstrated that while the NOX1-mTORC1 pathway is responsible for proliferation of a fast-cycling CSC population, PROX1 expression is required for chemoresistant persisters in colon cancer. Here, we show that enhanced autolysosomal activity mediated by mTORC1 inhibition induces PROX1 expression and that PROX1 induction in turn inhibits NOX1-mTORC1 activation. CDX2, identified as a transcriptional activator of NOX1, mediates PROX1-dependent NOX1 inhibition. PROX1-positive and CDX2-positive cells are present in distinct populations, and mTOR inhibition triggers conversion of the CDX2-positive population to the PROX1-positive population. Inhibition of autophagy synergizes with mTOR inhibition to block cancer proliferation. Thus, mTORC1 inhibition-mediated induction of PROX1 stabilizes a persister-like state with high autolysosomal activity via a feedback regulation that involves a key cascade of proliferating CSCs.


Asunto(s)
Neoplasias del Colon , Humanos , Línea Celular Tumoral , Proliferación Celular , Neoplasias del Colon/metabolismo , Retroalimentación , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , NADPH Oxidasa 1
7.
J Clin Invest ; 133(22)2023 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-37966117

RESUMEN

The heterogeneity of cancer stem cells (CSCs) within tumors presents a challenge in therapeutic targeting. To decipher the cellular plasticity that fuels phenotypic heterogeneity, we undertook single-cell transcriptomics analysis in triple-negative breast cancer (TNBC) to identify subpopulations in CSCs. We found a subpopulation of CSCs with ancestral features that is marked by FXYD domain-containing ion transport regulator 3 (FXYD3), a component of the Na+/K+ pump. Accordingly, FXYD3+ CSCs evolve and proliferate, while displaying traits of alveolar progenitors that are normally induced during pregnancy. Clinically, FXYD3+ CSCs were persistent during neoadjuvant chemotherapy, hence linking them to drug-tolerant persisters (DTPs) and identifying them as crucial therapeutic targets. Importantly, FXYD3+ CSCs were sensitive to senolytic Na+/K+ pump inhibitors, such as cardiac glycosides. Together, our data indicate that FXYD3+ CSCs with ancestral features are drivers of plasticity and chemoresistance in TNBC. Targeting the Na+/K+ pump could be an effective strategy to eliminate CSCs with ancestral and DTP features that could improve TNBC prognosis.


Asunto(s)
Neoplasias de la Mama Triple Negativas , Humanos , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología , Células Madre Neoplásicas/patología , Línea Celular Tumoral , Proteínas de la Membrana , Proteínas de Neoplasias/genética
8.
Cancer Lett ; 521: 29-38, 2021 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-34419499

RESUMEN

Patient-derived cells and xenografts retain the biological characteristics of clinical cancers and are instrumental in gaining a better understanding of the chemoresistance of cancer cells. Here, we have established a panel of patient-derived spheroids from clinical materials of ovarian cancer. Systematic evaluation using therapeutic agents indicated that sensitivity to platinum-based compounds significantly varied among the spheroids. To understand the molecular basis of drug sensitivity, we performed integrative analyses combining chemoresistance data and gene expression profiling of the ovarian cancer patient-derived spheroids. Correlation analyses revealed that cisplatin resistance was significantly associated with elevated levels of glucose-6-phosphate dehydrogenase (G6PD) and glutathione-producing redox enzymes. Accordingly, cisplatin-resistant spheroids established in vitro showed elevated levels of G6PD and active glutathione. Moreover, treatment with a G6PD inhibitor in combination with cisplatin suppressed spheroid proliferation in vitro and largely eradicated peritoneal metastasis in mouse xenograft models. Furthermore, G6PD expression was elevated during carcinogenesis and associated with poor prognosis. Thus, the combination of gene expression data and chemosensitivity revealed the essential roles of G6PD-driven redox metabolism in cisplatin resistance, underscoring the significance of an integrative approach using patient-derived cells.

9.
Cancer Res ; 80(20): 4451-4464, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-32816913

RESUMEN

Cancer chemoresistance is often attributed to the presence of cancer stem cell (CSC)-like cells, but whether they are homogeneously chemoresistant remains unclear. We previously showed that in colon tumors, a subpopulation of LGR5+ CSC-like cells driven by TCF1 (TCF7), a Wnt-responsive transcription factor, were responsible for tumorigenicity. Here we demonstrate that the tumorigenic subpopulation of mouse LGR5+ cells exists in a slow-cycling state and identify a unique 22-gene signature that characterizes these slow-cycling CSC. Seven of the signature genes are specifically expressed in slow-cycling LGR5+ cells from xenografted human colon tumors and are upregulated in colon cancer clinical specimens. Among these seven, four genes (APCDD1, NOTUM, PROX1, and SP5) are known to be direct Wnt target genes, and PROX1 was expressed in the invasive fronts of colon tumors. PROX1 was activated by TCF1 to induce CDKN1C and maintain a slow-cycling state in colon cancer organoids. Strikingly, PROX1 was required for recurrent growth after chemotherapeutic treatment, suggesting that inhibition of slow-cycling CSC by targeting the TCF1-PROX1-CDKN1C pathway is an effective strategy to combat refractory colon cancer in combination with conventional chemotherapy. SIGNIFICANCE: These findings illustrate the importance of a slow-cycling CSC subpopulation in colon cancer development and chemoresistance, with potential implications for the identified slow-cycling CSC signatures and the TCF1-PROX1-CDKN1C pathway as therapeutic targets.


Asunto(s)
Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Resistencia a Antineoplásicos/genética , Proteínas de Homeodominio/efectos adversos , Células Madre Neoplásicas/patología , Proteínas Supresoras de Tumor/efectos adversos , Animales , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Ratones Endogámicos C57BL , Receptores Acoplados a Proteínas G/genética , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Esferoides Celulares/patología , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
10.
Cancer Sci ; 100(7): 1291-9, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19432880

RESUMEN

The mdm2 and mdmx oncogenes play essential yet nonredundant roles in synergistic inactivatiosn of p53. However, the biochemical mechanism by which Mdmx synergizes with Mdm2 to inhibit p53 function remains obscure. Here we demonstrate that, using nonphosphorylatable mutants of Mdmx, the cooperative inhibition of p53 by Mdmx and Mdm2 was associated with cytoplasmic localization of p53, and with an increase of the interaction of Mdmx to p53 and Mdm2 in the cytoplasm. In addition, the Mdmx mutant cooperates with Mdm2 to induce ubiquitination of p53 at C-terminal lysine residues, and the integrity of the C-terminal lysines was partly required for the cooperative inhibition. The expression of subcellular localization mutants of Mdmx revealed that subcellular localization of Mdmx dictated p53 localization, and that cytoplasmic Mdmx tethered p53 in the cytoplasm and efficiently inhibited p53 activity. RNAi-mediated inhibition of Mdmx or introduction of the nuclear localization mutant of Mdmx reduced cytoplasmic retention of p53 in neuroblastoma cells, in which cytoplasmic sequestration of p53 is involved in its inactivation. Our data indicate that cytoplasmic tethering of p53 mediated by Mdmx contributes to p53 inactivation in some types of cancer cells.


Asunto(s)
Citoplasma/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Secuencia de Aminoácidos , Línea Celular Tumoral , Humanos , Datos de Secuencia Molecular , Fosforilación , Proteínas Proto-Oncogénicas c-mdm2/genética , Proteína p53 Supresora de Tumor/metabolismo
11.
Cell Rep ; 28(5): 1282-1295.e8, 2019 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-31365870

RESUMEN

Cancer stem cells (CSCs) are associated with the refractory nature of cancer, and elucidating the targetable pathways for CSCs is crucial for devising innovative antitumor therapies. We find that the proliferation of CSC-enriched colon spheroids from clinical specimen is dependent on mTORC1 kinase, which is activated by reactive oxygen species (ROS) produced by NOX1, an NADPH oxidase. In the spheroid-derived xenograft tumors, NOX1 is preferentially expressed in LGR5-positive cells. Dependence on NOX1 expression or mTOR kinase activity is corroborated in the xenograft tumors and mouse colon cancer-derived organoids. NOX1 co-localizes with mTORC1 in VPS41-/VPS39-positive lysosomes, where mTORC1 binds to S100A9, a member of S100 calcium binding proteins, in a NOX1-produced ROS-dependent manner. S100A9 is oxidized by NOX1-produced ROS, which facilitates binding to mTORC1 and its activation. We propose that NOX1-dependent mTORC1 activation via S100A9 oxidation in VPS41-/VPS39-positive lysosomes is crucial for colon CSC proliferation and colon cancer progression.


Asunto(s)
Calgranulina B/metabolismo , Neoplasias del Colon/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , NADPH Oxidasa 1/metabolismo , Proteínas de Neoplasias/metabolismo , Células Madre Neoplásicas/metabolismo , Animales , Calgranulina B/genética , Neoplasias del Colon/patología , Humanos , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Ratones , NADPH Oxidasa 1/genética , Proteínas de Neoplasias/genética , Células Madre Neoplásicas/patología , Oxidación-Reducción
12.
Mol Cell Oncol ; 4(4): e1335271, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28868346

RESUMEN

Leucine-rich repeat-containing G-protein coupled receptor 5 (Lgr5) is considered a representative marker of intestinal stem cells from both non-tumor and tumor tissues. However, it remains unclear whether all or only a fraction of Lgr5-positive cells behave as stem cells. Recently, we reported that Lgr5-positive cells from non-tumor and tumor tissues can be classified into overlapping yet distinct groups and that the tumor-specific groups are associated with tumorigenic capability, suggesting that these cells could represent targets for therapeutic intervention.

13.
Cell Rep ; 19(5): 981-994, 2017 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-28467911

RESUMEN

The generation of tumor-initiating cells during colon carcinogenesis is associated with the dysregulation of Wnt signaling, which is known to act on Lgr5-positive intestinal stem cells. Here, using single-cell qPCR analysis, we identified a subset of Lgr5-positive stem cells that emerged during tumorigenesis in a mouse model of colon cancer. These tumor-specific Lgr5-positive cells expressed low levels of Ceacam1 and increased levels of a specific subset of Wnt targets and showed enhanced tumorigenicity. Among the Wnt targets that were specifically expressed, the long isoform of Tcf1 was required for the proliferation of tumor organoids and drove a unique Wnt target gene expression profile. Tcf1 expression increased at an early stage of colon carcinogenesis and was associated with the nuclear accumulation of ß-catenin, underscoring the importance of the induction of Tcf1 expression in generating tumorigenic colon stem cells.


Asunto(s)
Carcinogénesis/genética , Neoplasias del Colon/patología , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Células Madre Neoplásicas/metabolismo , Proteínas Wnt/genética , Animales , Regulación Neoplásica de la Expresión Génica , Factor Nuclear 1-alfa del Hepatocito/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Wnt/metabolismo
14.
Biochem J ; 392(Pt 3): 511-7, 2005 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-16107205

RESUMEN

DNase X is the first human DNase protein identified as being homologous with DNase I. In the present study we describe the isolation of several mammalian DNase X cDNAs and the molecular characterization of their coding proteins. A sequence comparison reveals some conserved characteristics: all the mammalian DNase X proteins have an N-terminal signal peptide, a potential N-linked glycosylation site and a C-terminal hydrophobic domain. Human DNase X, ectopically expressed in HeLa S3 cells, is located in the ER (endoplasmic reticulum) and is modified by an N-linked glycosylation at Asn-243. Gene expression analyses show that the high expression level in muscular tissues, a known feature of human DNASE X, is also observed in mouse DNase X. Interestingly, the translation of porcine and bovine DNase X proteins occurs in the absence of an in-frame AUG initiation codon. We show that their mRNAs utilize a conserved CUG triplet for translation initiation.


Asunto(s)
Codón Iniciador/genética , Desoxirribonucleasas/química , Desoxirribonucleasas/genética , Iniciación de la Cadena Peptídica Traduccional , ARN Mensajero/metabolismo , Porcinos/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Bovinos , Secuencia de Consenso , Desoxirribonucleasas/biosíntesis , Desoxirribonucleasas/metabolismo , Perfilación de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Células HeLa , Humanos , Ratones , Datos de Secuencia Molecular , Biosíntesis de Proteínas , ARN Mensajero/genética , Homología de Secuencia de Aminoácido
15.
Biochem J ; 376(Pt 2): 377-81, 2003 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-12943533

RESUMEN

Among DNase I family members, only DNase gamma causes DNA fragmentation during apoptosis. However, the molecular basis for this functional feature of DNase gamma is poorly understood. Here we describe the identification of functional NLSs (nuclear localization signals) in DNase gamma and their roles in its apoptotic function. DNase gamma contains two NLSs: a classical bipartite-type NLS (NLS1) located in the N-terminal half, and a short basic domain (NLS2) at the C-terminus. No potential NLSs are found in the primary structures of other DNase I family DNases. Inactivation of either NLS1 or NLS2 causes reduced DNA ladder-producing activity in DNase gamma. Disruption of NLS2 suppresses ladder formation more effectively than disruption of NLS1. DNase gamma doubly mutated in both NLSs is enzymically active, but no longer catalyses apoptotic DNA fragmentation. Although DNase I fails to produce ladder formation during apoptosis, DNase I fused to NLS2 of DNase gamma through its C-terminus is able to catalyse DNA fragmentation in apoptotic cells. These results indicate that the presence of either NLS1 or NLS2 is necessary for the apoptotic function of DNase gamma, and that the most important domain for this function is NLS2. These findings also explain the lack of apoptotic DNase activity in the other DNase I family DNases.


Asunto(s)
Fragmentación del ADN , Endodesoxirribonucleasas/química , Endodesoxirribonucleasas/metabolismo , Animales , Apoptosis , Línea Celular , Desoxirribonucleasa I/genética , Desoxirribonucleasa I/metabolismo , Endodesoxirribonucleasas/fisiología , Humanos , Señales de Localización Nuclear , Proteínas Recombinantes de Fusión/metabolismo
16.
Exp Gerontol ; 39(2): 195-202, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15036412

RESUMEN

Organ-specific endonuclease might play a role in the age-related increase in apoptosis in laboratory rodent tissues. In nuclear extracts from liver tissues of male F344 rats, the DNase activity gel system identified DNase gamma, Ca(2+)/Mg(2+)-dependent endonuclease. The enzyme activity, which was measured at 3, 6, 16, and 24 months (mo) of age, was significantly increased between 16 and 24mo in control rats fed ad libitum (AL). The expression level of DNase gamma-mRNA, estimated by a semi-quantitative reverse transcription-polymerase chain reaction method, was also increased at 24mo in group AL. The proportion of immunohistochemically DNase gamma-positive cells, most of which were light-microscopically confined to apoptotic cells, was also significantly increased between 16 and 24mo. Dietary restriction, a powerful anti-aging intervention, which was achieved by providing 70% of the mean food intake in group AL from 6 weeks of age, inhibited the age-related increase in the enzyme activity and the proportion of immunostained cells; for the mRNA level, statistical significance was not obtained. The present study suggests that DNase gamma is involved in an age-related increase in the apoptosis of rat liver, and that CR inhibits the increase as it minimized the age-related increase in the fraction of DNA-damaged hepatocytes susceptible to apoptosis.


Asunto(s)
Envejecimiento/metabolismo , Apoptosis/fisiología , Endodesoxirribonucleasas/metabolismo , Privación de Alimentos/fisiología , Hígado/enzimología , Envejecimiento/genética , Animales , Núcleo Celular/enzimología , Endodesoxirribonucleasas/genética , Regulación del Desarrollo de la Expresión Génica , Hígado/citología , Masculino , ARN Mensajero/genética , Ratas , Ratas Endogámicas F344 , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
17.
Virchows Arch ; 443(2): 170-4, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12802584

RESUMEN

DNA fragmentation induced by endonucleases is a hallmark of apoptotic process. One of the endonucleases responsible for apoptotic DNA fragmentation has been identified as DNase gamma. It has been suggested that massive nuclear debris observed in histiocytic necrotizing lymphadenitis (HNL, Kikuchi disease) results mainly from apoptosis of T-lymphocytes rather than necrosis. To identify the role of DNase gamma in apoptotic foci of HNL paraffin embedded tissue sections of HNL and lymphadenopathy with hyperplastic germinal centers and paracortex in the neck lymph nodes were immunohistochemically stained for DNase gamma and the results compared with the reactivity of TdT-mediated dUTP nick end labeling (TUNEL). Immunoreactivity for DNase gamma was detected in fragmented and condensed nuclei found abundantly in HNL as well as TUNEL reactivity. Moreover, the ratio of DNase gamma immunoreactivity to TUNEL reactivity, which represents probability of apoptosis relevant to DNase gamma, was higher in foci of nuclear debris in HNL than in the hyperplastic paracortex in lymphadenopathy including T-cell apoptosis. Our findings suggest that apoptosis detected in HNL depends more specifically upon DNase gamma than apoptosis in the hyperplastic paracortex of lymphadenopathy, and that the dysregulation of DNase gamma activation is involved in apoptosis in HNL.


Asunto(s)
Fragmentación del ADN , Endodesoxirribonucleasas/metabolismo , Linfadenitis Necrotizante Histiocítica/enzimología , Adolescente , Adulto , Anciano , Femenino , Centro Germinal/enzimología , Centro Germinal/patología , Linfadenitis Necrotizante Histiocítica/patología , Humanos , Técnicas para Inmunoenzimas , Etiquetado Corte-Fin in Situ , Ganglios Linfáticos/enzimología , Ganglios Linfáticos/patología , Masculino , Persona de Mediana Edad
18.
PLoS One ; 8(12): e80223, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24312463

RESUMEN

Apoptosis and necrosis, two major forms of cell death, can be distinguished morphologically and biochemically. Internucleosomal DNA fragmentation (INDF) is a biochemical hallmark of apoptosis, and caspase-activated DNase (CAD), also known as DNA fragmentation factor 40 kDa (DFF40), is one of the major effector endonucleases. DNase γ, a Mg(2+)/Ca(2+)-dependent endonuclease, is also known to generate INDF but its role among other apoptosis-associated endonucleases in cell death is unclear. Here we show that (i) INDF occurs even during necrosis in cell lines, primary cells, and in tissues of mice in vivo, and (ii) DNase γ, but not CAD, is the effector endonuclease for INDF in cells undergoing necrosis. These results document a previously unappreciated role for INDF in necrosis and define its molecular basis.


Asunto(s)
Fragmentación del ADN , Endodesoxirribonucleasas/metabolismo , Animales , Desoxirribonucleasas/genética , Desoxirribonucleasas/metabolismo , Endodesoxirribonucleasas/genética , Humanos , Ratones , Necrosis , Proteínas de Unión a Poli-ADP-Ribosa , Células U937
19.
Biomed Res ; 30(3): 165-70, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19574717

RESUMEN

DNA fragmentation is a biochemical hallmark of apoptosis. Several endonucleases, including CAD/DFF40 and endonuclease G, are implicated in DNA fragmentation. DNase gamma has also been considered to be one of the enzymes involved, but its role in relation to CAD/DFF40 in apoptosis has not been fully elucidated. Here, we distinguished between DNase gamma-dependent and CAD/DFF40-dependent DNA fragmentations. We found that DNase gamma activities appeared in the late apoptotic phase and accelerated DNA fragmentation. Thus, even if the apoptotic DNA fragmentation is initiated by CAD/DFF40, DNase gamma is required for the more complete digestion of the genomic DNA in dying cells.


Asunto(s)
Apoptosis/fisiología , Linfoma de Burkitt/genética , Línea Celular Tumoral , Fragmentación del ADN , Endodesoxirribonucleasas/metabolismo , Linfoma de Burkitt/patología , Núcleo Celular/metabolismo , Desoxirribonucleasas/genética , Desoxirribonucleasas/metabolismo , Endodesoxirribonucleasas/genética , Inhibidores Enzimáticos/metabolismo , Humanos , Proteínas de Unión a Poli-ADP-Ribosa , Estaurosporina/metabolismo
20.
J Biol Chem ; 282(23): 17132-40, 2007 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-17416904

RESUMEN

DNase X is the first mammalian DNase to be isolated that is homologous to DNase I. In this study, we have examined its function using a novel monoclonal antibody and showed it to be expressed on the cell surface as a glycosylphosphatidylinositolanchored membrane protein. High level expression was observed in human muscular tissues and in myotubes obtained in vitro from RD rhabdomyosarcoma cells. We observed that RD myotubes incorporated a foreign gene, lacZ, by endocytosis but that expression of the encoded coding product, beta-galactosidase, was strongly inhibited. Overexpression of DNase X inhibited endocytosis-mediated gene transfer, whereas knockdown of DNase X with small interfering RNA had the opposite effect. These results reveal that DNase X provides a cell surface barrier to endocytosis-mediated gene transfer.


Asunto(s)
Desoxirribonucleasas/metabolismo , Endocitosis , Técnicas de Transferencia de Gen , Glicosilfosfatidilinositoles/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Diferenciación Celular , Línea Celular , Cartilla de ADN , Técnica del Anticuerpo Fluorescente Indirecta , Humanos , Datos de Secuencia Molecular , Interferencia de ARN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA