Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Nucleic Acids Res ; 50(8): 4500-4514, 2022 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-35451487

RESUMEN

Histone H3.3 is an H3 variant which differs from the canonical H3.1/2 at four residues, including a serine residue at position 31 which is evolutionarily conserved. The H3.3 S31 residue is phosphorylated (H3.3 S31Ph) at heterochromatin regions including telomeres and pericentric repeats. However, the role of H3.3 S31Ph in these regions remains unknown. In this study, we find that H3.3 S31Ph regulates heterochromatin accessibility at telomeres during replication through regulation of H3K9/K36 histone demethylase KDM4B. In mouse embryonic stem (ES) cells, substitution of S31 with an alanine residue (H3.3 A31 -phosphorylation null mutant) results in increased KDM4B activity that removes H3K9me3 from telomeres. In contrast, substitution with a glutamic acid (H3.3 E31, mimics S31 phosphorylation) inhibits KDM4B, leading to increased H3K9me3 and DNA damage at telomeres. H3.3 E31 expression also increases damage at other heterochromatin regions including the pericentric heterochromatin and Y chromosome-specific satellite DNA repeats. We propose that H3.3 S31Ph regulation of KDM4B is required to control heterochromatin accessibility of repetitive DNA and preserve chromatin integrity.


Asunto(s)
Heterocromatina , Histonas , Animales , Ratones , Histonas/genética , Histonas/metabolismo , Heterocromatina/genética , Histona Demetilasas/metabolismo , Fosforilación , Ensamble y Desensamble de Cromatina
2.
Proc Natl Acad Sci U S A ; 115(18): 4737-4742, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29669917

RESUMEN

ATRX (alpha thalassemia/mental retardation X-linked) complexes with DAXX to deposit histone variant H3.3 into repetitive heterochromatin. Recent genome sequencing studies in cancers have revealed mutations in ATRX and their association with ALT (alternative lengthening of telomeres) activation. Here we report depletion of ATRX in mouse ES cells leads to selective loss in ribosomal RNA gene (rDNA) copy number. Supporting this, ATRX-mutated human ALT-positive tumors also show a substantially lower rDNA copy than ALT-negative tumors. Further investigation shows that the rDNA copy loss and repeat instability are caused by a disruption in H3.3 deposition and thus a failure in heterochromatin formation at rDNA repeats in the absence of ATRX. We also find that ATRX-depleted cells are reduced in ribosomal RNA transcription output and show increased sensitivity to RNA polymerase I (Pol I) transcription inhibitor CX5461. In addition, human ALT-positive cancer cell lines are also more sensitive to CX5461 treatment. Our study provides insights into the contribution of ATRX loss of function to tumorigenesis through the loss of rDNA stability and suggests the therapeutic potential of targeting Pol I transcription in ALT cancers.


Asunto(s)
ADN de Neoplasias/metabolismo , ADN Ribosómico/metabolismo , Dosificación de Gen , Mutación , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Proteína Nuclear Ligada al Cromosoma X/metabolismo , Benzotiazoles/farmacología , Línea Celular Tumoral , ADN de Neoplasias/genética , ADN Ribosómico/genética , Inestabilidad Genómica , Humanos , Naftiridinas/farmacología , Proteínas de Neoplasias/genética , Neoplasias/genética , Neoplasias/patología , ARN Polimerasa I/antagonistas & inhibidores , ARN Polimerasa I/genética , ARN Polimerasa I/metabolismo , Transcripción Genética/efectos de los fármacos , Transcripción Genética/genética , Proteína Nuclear Ligada al Cromosoma X/genética
3.
Nucleic Acids Res ; 45(21): 12340-12353, 2017 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-29040668

RESUMEN

AURKB (Aurora Kinase B) is a serine/threonine kinase better known for its role at the mitotic kinetochore during chromosome segregation. Here, we demonstrate that AURKB localizes to the telomeres in mouse embryonic stem cells, where it interacts with the essential telomere protein TERF1. Loss of AURKB function affects TERF1 telomere binding and results in aberrant telomere structure. In vitro kinase experiments successfully identified Serine 404 on TERF1 as a putative AURKB target site. Importantly, in vivo overexpression of S404-TERF1 mutants results in fragile telomere formation. These findings demonstrate that AURKB is an important regulator of telomere structural integrity.


Asunto(s)
Aurora Quinasa B/metabolismo , Telómero/enzimología , Proteína 1 de Unión a Repeticiones Teloméricas/metabolismo , Animales , Aurora Quinasa B/fisiología , Línea Celular , Células Madre Embrionarias/enzimología , Humanos , Interfase/genética , Ratones , Mitosis/genética , Mutación , Unión Proteica , Telómero/ultraestructura , Proteína 1 de Unión a Repeticiones Teloméricas/química , Proteína 1 de Unión a Repeticiones Teloméricas/genética
4.
PLoS Biol ; 8(4): e1000361, 2010 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-20436908

RESUMEN

Autocrine priming of cells by small quantities of constitutively produced type I interferon (IFN) is a well-known phenomenon. In the absence of type I IFN priming, cells display attenuated responses to other cytokines, such as anti-viral protection in response to IFNgamma. This phenomenon was proposed to be because IFNalpha/beta receptor1 (IFNAR1) is a component of the IFNgamma receptor (IFNGR), but our new data are more consistent with a previously proposed model indicating that regulated expression of STAT1 may also play a critical role in the priming process. Initially, we noticed that DNA binding activity of STAT1 was attenuated in c-Jun(-/-) fibroblasts because they expressed lower levels of STAT1 than wild-type cells. However, expression of STAT1 was rescued by culturing c-Jun(-/-) fibroblasts in media conditioned by wild-type fibroblasts suggesting they secreted a STAT1-inducing factor. The STAT1-inducing factor in fibroblast-conditioned media was IFNbeta, as it was inhibited by antibodies to IFNAR1, or when IFNbeta expression was knocked down in wild-type cells. IFNAR1(-/-) fibroblasts, which cannot respond to this priming, also expressed reduced levels of STAT1, which correlated with their poor responses to IFNgamma. The lack of priming in IFNAR1(-/-) fibroblasts was compensated by over-expression of STAT1, which rescued molecular responses to IFNgamma and restored the ability of IFNgamma to induce protective anti-viral immunity. This study provides a comprehensive description of the molecular events involved in priming by type I IFN. Adding to the previous working model that proposed an interaction between type I and II IFN receptors, our work and that of others demonstrates that type I IFN primes IFNgamma-mediated immune responses by regulating expression of STAT1. This may also explain how type I IFN can additionally prime cells to respond to a range of other cytokines that use STAT1 (e.g., IL-6, M-CSF, IL-10) and suggests a potential mechanism for the changing levels of STAT1 expression observed during viral infection.


Asunto(s)
Interferón Tipo I/metabolismo , Interferón gamma/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Factor de Transcripción STAT1/metabolismo , Transducción de Señal/fisiología , Animales , Células Cultivadas , Fibroblastos/citología , Fibroblastos/fisiología , Interferón Tipo I/genética , Interferón gamma/genética , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Ratones , Ratones Noqueados , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/metabolismo , Factor de Transcripción STAT1/genética , Bazo/citología
5.
Cancers (Basel) ; 15(18)2023 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-37760615

RESUMEN

Tumour progression relies on interactions with untransformed cells in the tumour microenvironment (TME), including cancer-associated fibroblasts (CAFs), which promote blood supply, tumour progression, and immune evasion. Eph receptor tyrosine kinases are cell guidance receptors that are most active during development but re-emerge in cancer and are recognised drug targets. EphA3 is overexpressed in a wide range of tumour types, and we previously found expression particularly in stromal and vascular tissues of the TME. To investigate its role in the TME, we generated transgenic mice with inducible shRNA-mediated knockdown of EphA3 expression. EphA3 knockdown was confirmed in aortic mesenchymal stem cells (MSCs), which displayed reduced angiogenic capacity. In mice with syngeneic lung tumours, EphA3 knockdown reduced vasculature and CAF/MSC-like cells in tumours, and inhibited tumour growth, which was confirmed also in a melanoma model. Single cell RNA sequencing analysis of multiple human tumour types confirmed EphA3 expression in CAFs, including in breast cancer, where EphA3 was particularly prominent in perivascular- and myofibroblast-like CAFs. Our results thus indicate expression of the cell guidance receptor EphA3 in distinct CAF subpopulations is important in supporting tumour angiogenesis and tumour growth, highlighting its potential as a therapeutic target.

6.
Genome Biol ; 24(1): 284, 2023 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-38066546

RESUMEN

BACKGROUND: Point mutations in histone variant H3.3 (H3.3K27M, H3.3G34R) and the H3.3-specific ATRX/DAXX chaperone complex are frequent events in pediatric gliomas. These H3.3 point mutations affect many chromatin modifications but the exact oncogenic mechanisms are currently unclear. Histone H3.3 is known to localize to nuclear compartments known as promyelocytic leukemia (PML) nuclear bodies, which are frequently mutated and confirmed as oncogenic drivers in acute promyelocytic leukemia. RESULTS: We find that the pediatric glioma-associated H3.3 point mutations disrupt the formation of PML nuclear bodies and this prevents differentiation down glial lineages. Similar to leukemias driven by PML mutations, H3.3-mutated glioma cells are sensitive to drugs that target PML bodies. We also find that point mutations in IDH1/2-which are common events in adult gliomas and myeloid leukemias-also disrupt the formation of PML bodies. CONCLUSIONS: We identify PML as a contributor to oncogenesis in a subset of gliomas and show that targeting PML bodies is effective in treating these H3.3-mutated pediatric gliomas.


Asunto(s)
Neoplasias Encefálicas , Glioma , Histonas , Adulto , Niño , Humanos , Neoplasias Encefálicas/genética , Glioma/genética , Histonas/genética , Mutación , Cuerpos Nucleares de la Leucemia Promielocítica/genética , Cuerpos Nucleares de la Leucemia Promielocítica/patología
7.
Cancers (Basel) ; 14(13)2022 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-35804938

RESUMEN

ADAM10 is a transmembrane metalloprotease that sheds a variety of cell surface proteins, including receptors and ligands that regulate a range of developmental processes which re-emerge during tumour development. While ADAM10 is ubiquitously expressed, its activity is normally tightly regulated, but becomes deregulated in tumours. We previously reported the generation of a monoclonal antibody, 8C7, which preferentially recognises an active form of ADAM10 in human and mouse tumours. We now report our investigation of the mechanism of this specificity, and the preferential targeting of 8C7 to human tumour cell xenografts in mice. We also report the development of novel 8C7 antibody-drug conjugates that preferentially kill cells displaying the 8C7 epitope, and that can inhibit tumour growth in mice. This study provides the first demonstration that antibody-drug conjugates targeting an active conformer of ADAM10, a widely expressed transmembrane metalloprotease, enable tumour-selective targeting and inhibition.

8.
J Cell Biochem ; 109(1): 103-12, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19885868

RESUMEN

Expression of the human HIN-200 family member IFI 16 has been reported to suppress cell growth and contribute to the onset of cellular senescence. However the molecular events involved in this process have not been fully characterised. We fused IFI 16 to the estrogen receptor ligand-binding domain to establish an inducible model for studying the molecular events that cause these phenomena. In cells induced to express the ER-IFI 16 within the nucleus there was a decrease in cellular proliferation and concomitant growth arrest in the G1 phase of the cell cycle. Unlike previous reports, this did not appear to involve the p53-p21(WAF1/CIP1)-cdk2-pRb pathway. Following nuclear expression of ER-IFI 16 we noted senescence-like morphological changes and expression of senescence-associated beta-galactosidase in growth arrested cells. Importantly, we also found a marked reduction in telomerase activity in arrested cells compared to controls. Moreover, IFI 16 and hTERT co-localised within the nucleus and these two proteins physically interacted in vivo and in vitro. Together, these data suggest that IFI 16 may act as an endogenous regulator of telomerase activity and, through its interaction with hTERT, contributes to the inhibition of proliferation and induces a senescence-like state.


Asunto(s)
Proliferación Celular , Senescencia Celular/fisiología , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Telomerasa/metabolismo , Western Blotting , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Técnica del Anticuerpo Fluorescente , Humanos , Inmunoprecipitación
9.
J Cell Biochem ; 103(4): 1270-82, 2008 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-17786933

RESUMEN

HIN-200 proteins are interferon-inducible proteins capable of regulating cell growth, senescence, differentiation and death. Using a combination of in silico analysis of NCBI EST databases and screening of murine C57BL/6 cDNA libraries we isolated novel murine HIN-200 cDNAs designated Ifi206S and Ifi206L encoding two putative mRNA splice variants. The p206S and p206L protein isoforms have a modular domain structure consisting of an N-terminal PAAD/DAPIN/Pyrin domain, a region rich in serine, threonine and proline residues and a C-terminal 200 B domain characteristic of other HIN-200 proteins. Ifi206 mRNA was detected only in the spleen and lung of BALB/c and C57BL/6 mice and expression was up-regulated by both types I and II IFN subtypes. p206 protein was predominantly expressed in the cytoplasm and addition of LMB, a CRM1 dependent nuclear export inhibitor, caused p206 to accumulate in the nucleus. Unlike other human and mouse HIN-200 proteins that contain only a single 200 amino acid domain, overexpression of p206 impaired the clonogenic growth of tumour cell lines. Thus, p206 represents the newest HIN-200 family member discovered. It has distinct and restricted pattern of expression however maintains many of the hallmarks of HIN-200 proteins including the presence of a characteristic 200 X domain, induction by interferon and an ability to suppress tumour cell growth.


Asunto(s)
Proteínas Nucleares/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Células 3T3 , Empalme Alternativo , Animales , Línea Celular Tumoral , Proliferación Celular , Clonación Molecular , Citoplasma/metabolismo , Biblioteca de Genes , Interferón Tipo I/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Proteínas Nucleares/genética , Especificidad de Órganos , Conformación Proteica , ARN Mensajero/metabolismo , Proteínas Recombinantes , Transfección , Proteínas Supresoras de Tumor/genética
10.
J Interferon Cytokine Res ; 28(1): 5-11, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18370867

RESUMEN

HIN-200 proteins are interferon (IFN)-inducible proteins that can regulate cell proliferation and differentiation in vitro. Characterization of the lineage and cell type-dependent expression of Ifi202 revealed little or no expression of Ifi202 in the Lin(-)/c-Kit+ fraction enriched for immature hematopoietic progenitor cells (HPCs) but higher levels in more differentiated Lin(-)/c-Kit(-) and Lin+ populations. The highest levels of Ifi202 expression were observed in CD11b+/Gr-1dim immature granulocytes in the bone marrow. In the peripheral blood, Ifi202 was expressed only in the myeloid lineage, with the highest level of expression seen in CD11b+/Gr-1dim immature granulocytes. Constitutive expression of p202 in primary HPCs delayed proliferation of these cells in vitro, caused a reduction in the number and size of myeloid colonies growing on methylcellulose, and affected the ability of the cells to reconstitute irradiated mice but did not significantly affect cell differentiation. Thus, p202 plays a role in regulating the proliferative capacity of hematopoietic cells.


Asunto(s)
Diferenciación Celular , Sistema Hematopoyético/citología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Fosfoproteínas/metabolismo , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Proliferación Celular , Ensayo de Unidades Formadoras de Colonias , Metilcelulosa , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Bazo/citología , Bazo/metabolismo , Células Madre/citología , Células Madre/metabolismo , Transducción Genética
11.
Nat Commun ; 9(1): 3142, 2018 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-30087349

RESUMEN

An array of oncogenic histone point mutations have been identified across a number of different cancer studies. It has been suggested that some of these mutant histones can exert their effects by inhibiting epigenetic writers. Here, we report that the H3.3 G34R (glycine to arginine) substitution mutation, found in paediatric gliomas, causes widespread changes in H3K9me3 and H3K36me3 by interfering with the KDM4 family of K9/K36 demethylases. Expression of a targeted single-copy of H3.3 G34R at endogenous levels induced chromatin alterations that were comparable to a KDM4 A/B/C triple-knockout. We find that H3.3 G34R preferentially binds KDM4 while simultaneously inhibiting its enzymatic activity, demonstrating that histone mutations can act through inhibition of epigenetic erasers. These results suggest that histone point mutations can exert their effects through interactions with a range of epigenetic readers, writers and erasers.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Cromatina/química , Glioblastoma/metabolismo , Histonas/metabolismo , Mutación , Mutación Puntual , Animales , Arginina/química , Biotinilación , Neoplasias Encefálicas/genética , Niño , Modelos Animales de Enfermedad , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Glicina/química , Histonas/genética , Humanos , Ratones , Unión Proteica , Análisis de Secuencia de ARN , Transgenes
12.
J Exp Med ; 213(9): 1741-57, 2016 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-27503072

RESUMEN

The transmembrane metalloprotease ADAM10 sheds a range of cell surface proteins, including ligands and receptors of the Notch, Eph, and erbB families, thereby activating signaling pathways critical for tumor initiation and maintenance. ADAM10 is thus a promising therapeutic target. Although widely expressed, its activity is normally tightly regulated. We now report prevalence of an active form of ADAM10 in tumors compared with normal tissues, in mouse models and humans, identified by our conformation-specific antibody mAb 8C7. Structure/function experiments indicate mAb 8C7 binds an active conformation dependent on disulfide isomerization and oxidative conditions, common in tumors. Moreover, this active ADAM10 form marks cancer stem-like cells with active Notch signaling, known to mediate chemoresistance. Importantly, specific targeting of active ADAM10 with 8C7 inhibits Notch activity and tumor growth in mouse models, particularly regrowth after chemotherapy. Our results indicate targeted inhibition of active ADAM10 as a potential therapy for ADAM10-dependent tumor development and drug resistance.


Asunto(s)
Proteína ADAM10/fisiología , Neoplasias Experimentales/patología , Células Madre Neoplásicas/patología , Proteína ADAM10/antagonistas & inhibidores , Proteína ADAM10/química , Proteína ADAM17/fisiología , Secuencias de Aminoácidos , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Receptores Notch/fisiología
13.
Cancer Res ; 74(16): 4470-81, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-25125683

RESUMEN

Eph receptor tyrosine kinases are critical for cell-cell communication during normal and oncogenic tissue patterning and tumor growth. Somatic mutation profiles of several cancer genomes suggest EphA3 as a tumor suppressor, but its oncogenic expression pattern and role in tumorigenesis remain largely undefined. Here, we report unexpected EphA3 overexpression within the microenvironment of a range of human cancers and mouse tumor xenografts where its activation inhibits tumor growth. EphA3 is found on mouse bone marrow-derived cells with mesenchymal and myeloid phenotypes, and activation of EphA3(+)/CD90(+)/Sca1(+) mesenchymal/stromal cells with an EphA3 agonist leads to cell contraction, cell-cell segregation, and apoptosis. Treatment of mice with an agonistic α-EphA3 antibody inhibits tumor growth by severely disrupting the integrity and function of newly formed tumor stroma and microvasculature. Our data define EphA3 as a novel target for selective ablation of the tumor microenvironment and demonstrate the potential of EphA3 agonists for anticancer therapy.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Proteínas Tirosina Quinasas Receptoras/agonistas , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Receptor EphA3/agonistas , Receptor EphA3/biosíntesis , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Transformación Celular Neoplásica , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Ratones , Ratones Desnudos , Terapia Molecular Dirigida , Proteínas Tirosina Quinasas Receptoras/inmunología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptor EphA3/inmunología , Receptor EphA3/metabolismo , Transducción de Señal , Células del Estroma/efectos de los fármacos , Células del Estroma/patología , Microambiente Tumoral/efectos de los fármacos
14.
J Cell Biol ; 195(6): 1033-45, 2011 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-22144690

RESUMEN

Eph receptors interact with ephrin ligands on adjacent cells to facilitate tissue patterning during normal and oncogenic development, in which unscheduled expression and somatic mutations contribute to tumor progression. EphA and B subtypes preferentially bind A- and B-type ephrins, respectively, resulting in receptor complexes that propagate via homotypic Eph-Eph interactions. We now show that EphA and B receptors cocluster, such that specific ligation of one receptor promotes recruitment and cross-activation of the other. Remarkably, coexpression of a kinase-inactive mutant EphA3 with wild-type EphB2 can cause either cross-activation or cross-inhibition, depending on relative expression. Our findings indicate that cellular responses to ephrin contact are determined by the EphA/EphB receptor profile on a given cell rather than the individual Eph subclass. Importantly, they imply that in tumor cells coexpressing different Ephs, functional mutations in one subtype may cause phenotypes that are a result of altered signaling from heterotypic rather from homotypic Eph clusters.


Asunto(s)
Polimerizacion , Receptores de la Familia Eph/metabolismo , Animales , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Glioma/enzimología , Células HEK293 , Humanos , Masculino , Neoplasias de la Próstata/enzimología , Receptores de la Familia Eph/agonistas , Receptores de la Familia Eph/química , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA