Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 14(1): 7295, 2023 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-37957154

RESUMEN

Mutations in SNCA, the gene encoding α-synuclein (αSyn), cause familial Parkinson's disease (PD) and aberrant αSyn is a key pathological hallmark of idiopathic PD. This α-synucleinopathy leads to mitochondrial dysfunction, which may drive dopaminergic neurodegeneration. PARKIN and PINK1, mutated in autosomal recessive PD, regulate the preferential autophagic clearance of dysfunctional mitochondria ("mitophagy") by inducing ubiquitylation of mitochondrial proteins, a process counteracted by deubiquitylation via USP30. Here we show that loss of USP30 in Usp30 knockout mice protects against behavioral deficits and leads to increased mitophagy, decreased phospho-S129 αSyn, and attenuation of SN dopaminergic neuronal loss induced by αSyn. These observations were recapitulated with a potent, selective, brain-penetrant USP30 inhibitor, MTX115325, with good drug-like properties. These data strongly support further study of USP30 inhibition as a potential disease-modifying therapy for PD.


Asunto(s)
Enfermedad de Parkinson , Tioléster Hidrolasas , Animales , Ratones , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Neuronas Dopaminérgicas/metabolismo , Ratones Noqueados , Mitocondrias/metabolismo , Enfermedad de Parkinson/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Tioléster Hidrolasas/genética
2.
Nat Commun ; 13(1): 3707, 2022 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-35764636

RESUMEN

SHLD1 is part of the Shieldin (SHLD) complex, which acts downstream of 53BP1 to counteract DNA double-strand break (DSB) end resection and promote DNA repair via non-homologous end-joining (NHEJ). While 53BP1 is essential for immunoglobulin heavy chain class switch recombination (CSR), long-range V(D)J recombination and repair of RAG-induced DSBs in XLF-deficient cells, the function of SHLD during these processes remains elusive. Here we report that SHLD1 is dispensable for lymphocyte development and RAG-mediated V(D)J recombination, even in the absence of XLF. By contrast, SHLD1 is essential for restricting resection at AID-induced DSB ends in both NHEJ-proficient and NHEJ-deficient B cells, providing an end-protection mechanism that permits productive CSR by NHEJ and alternative end-joining. Finally, we show that this SHLD1 function is required for orientation-specific joining of AID-initiated DSBs. Our data thus suggest that 53BP1 promotes V(D)J recombination and CSR through two distinct mechanisms: SHLD-independent synapsis of V(D)J segments and switch regions within chromatin, and SHLD-dependent protection of AID-DSB ends against resection.


Asunto(s)
Roturas del ADN de Doble Cadena , Recombinación V(D)J , Reparación del ADN por Unión de Extremidades , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Cambio de Clase de Inmunoglobulina/genética , Recombinación V(D)J/genética
3.
Nat Commun ; 13(1): 2692, 2022 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-35577786

RESUMEN

Soluble aggregates of the microtubule-associated protein tau have been challenging to assemble and characterize, despite their important role in the development of tauopathies. We found that sequential hyperphosphorylation by protein kinase A in conjugation with either glycogen synthase kinase 3ß or stress activated protein kinase 4 enabled recombinant wild-type tau of isoform 0N4R to spontaneously polymerize into small amorphous aggregates in vitro. We employed tandem mass spectrometry to determine the phosphorylation sites, high-resolution native mass spectrometry to measure the degree of phosphorylation, and super-resolution microscopy and electron microscopy to characterize the morphology of aggregates formed. Functionally, compared with the unmodified aggregates, which require heparin induction to assemble, these self-assembled hyperphosphorylated tau aggregates more efficiently disrupt membrane bilayers and induce Toll-like receptor 4-dependent responses in human macrophages. Together, our results demonstrate that hyperphosphorylated tau aggregates are potentially damaging to cells, suggesting a mechanism for how hyperphosphorylation could drive neuroinflammation in tauopathies.


Asunto(s)
Tauopatías , Receptor Toll-Like 4 , Proteínas tau , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Heparina , Humanos , Fosforilación , Agregación Patológica de Proteínas/metabolismo , Isoformas de Proteínas/metabolismo , Tauopatías/metabolismo , Receptor Toll-Like 4/metabolismo , Proteínas tau/metabolismo , Proteínas tau/ultraestructura
4.
Nat Neurosci ; 24(11): 1542-1554, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34675437

RESUMEN

Amyotrophic lateral sclerosis overlapping with frontotemporal dementia (ALS/FTD) is a fatal and currently untreatable disease characterized by rapid cognitive decline and paralysis. Elucidating initial cellular pathologies is central to therapeutic target development, but obtaining samples from presymptomatic patients is not feasible. Here, we report the development of a cerebral organoid slice model derived from human induced pluripotent stem cells (iPSCs) that recapitulates mature cortical architecture and displays early molecular pathology of C9ORF72 ALS/FTD. Using a combination of single-cell RNA sequencing and biological assays, we reveal distinct transcriptional, proteostasis and DNA repair disturbances in astroglia and neurons. We show that astroglia display increased levels of the autophagy signaling protein P62 and that deep layer neurons accumulate dipeptide repeat protein poly(GA), DNA damage and undergo nuclear pyknosis that could be pharmacologically rescued by GSK2606414. Thus, patient-specific iPSC-derived cortical organoid slice cultures are a reproducible translational platform to investigate preclinical ALS/FTD mechanisms as well as novel therapeutic approaches.


Asunto(s)
Esclerosis Amiotrófica Lateral/patología , Astrocitos/patología , Demencia Frontotemporal/patología , Neuronas/patología , Organoides/patología , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/metabolismo , Astrocitos/metabolismo , Proteína C9orf72/genética , Proteína C9orf72/metabolismo , Demencia Frontotemporal/genética , Demencia Frontotemporal/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Neuronas/metabolismo , Técnicas de Cultivo de Órganos/métodos , Organoides/metabolismo
5.
Cell Rep ; 36(9): 109649, 2021 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-34469738

RESUMEN

CAG repeat expansion in the HTT gene drives Huntington's disease (HD) pathogenesis and is modulated by DNA damage repair pathways. In this context, the interaction between FAN1, a DNA-structure-specific nuclease, and MLH1, member of the DNA mismatch repair pathway (MMR), is not defined. Here, we identify a highly conserved SPYF motif at the N terminus of FAN1 that binds to MLH1. Our data support a model where FAN1 has two distinct functions to stabilize CAG repeats. On one hand, it binds MLH1 to restrict its recruitment by MSH3, thus inhibiting the assembly of a functional MMR complex that would otherwise promote CAG repeat expansion. On the other hand, it promotes accurate repair via its nuclease activity. These data highlight a potential avenue for HD therapeutics in attenuating somatic expansion.


Asunto(s)
Encéfalo/enzimología , Daño del ADN , Reparación de la Incompatibilidad de ADN , Endodesoxirribonucleasas/metabolismo , Exodesoxirribonucleasas/metabolismo , Proteína Huntingtina/genética , Enfermedad de Huntington/enzimología , Enzimas Multifuncionales/metabolismo , Homólogo 1 de la Proteína MutL/metabolismo , Expansión de Repetición de Trinucleótido , Animales , Unión Competitiva , Encéfalo/patología , Línea Celular Tumoral , Endodesoxirribonucleasas/genética , Exodesoxirribonucleasas/genética , Células HEK293 , Humanos , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Ratones , Enzimas Multifuncionales/genética , Homólogo 1 de la Proteína MutL/genética , Proteína 3 Homóloga de MutS/genética , Proteína 3 Homóloga de MutS/metabolismo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas
6.
Essays Biochem ; 64(5): 847-861, 2020 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-33078197

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a rapidly disabling and fatal neurodegenerative disease. Due to insufficient disease-modifying treatments, there is an unmet and urgent need for elucidating disease mechanisms that occur early and represent common triggers in both familial and sporadic ALS. Emerging evidence suggests that impaired DNA damage response contributes to age-related somatic accumulation of genomic instability and can trigger or accelerate ALS pathological manifestations. In this review, we summarize and discuss recent studies indicating a direct link between DNA damage response and ALS. Further mechanistic understanding of the role genomic instability is playing in ALS disease pathophysiology will be critical for discovering new therapeutic avenues.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Daño del ADN , Humanos
9.
Nat Protoc ; 14(7): 1991-2014, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31160788

RESUMEN

Ploidy represents the number of chromosome sets in a cell. Although gametes have a haploid genome (n), most mammalian cells have diploid genomes (2n). The diploid status of most cells correlates with the number of probable alleles for each autosomal gene and makes it difficult to target these genes via mutagenesis techniques. Here, we describe a 7-week protocol for the derivation of mouse haploid embryonic stem cells (hESCs) from female gametes that also outlines how to maintain the cells once derived. We detail additional procedures that can be used with cell lines obtained from the mouse Haplobank, a biobank of >100,000 individual mouse hESC lines with targeted mutations in 16,970 genes. hESCs can spontaneously diploidize and can be maintained in both haploid and diploid states. Mouse hESCs are genomically and karyotypically stable, are innately immortal and isogenic, and can be derived in an array of differentiated cell types; they are thus highly amenable to genetic screens and to defining molecular connectivity pathways.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Haploidia , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/fisiología , Animales , Blastocisto/citología , Línea Celular , Separación Celular/métodos , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Flujo de Trabajo
10.
Nat Commun ; 10(1): 87, 2019 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-30622252

RESUMEN

Mutations in the ATM tumor suppressor gene confer hypersensitivity to DNA-damaging chemotherapeutic agents. To explore genetic resistance mechanisms, we performed genome-wide CRISPR-Cas9 screens in cells treated with the DNA topoisomerase I inhibitor topotecan. Thus, we here establish that inactivating terminal components of the non-homologous end-joining (NHEJ) machinery or of the BRCA1-A complex specifically confer topotecan resistance to ATM-deficient cells. We show that hypersensitivity of ATM-mutant cells to topotecan or the poly-(ADP-ribose) polymerase (PARP) inhibitor olaparib reflects delayed engagement of homologous recombination at DNA-replication-fork associated single-ended double-strand breaks (DSBs), allowing some to be subject to toxic NHEJ. Preventing DSB ligation by NHEJ, or enhancing homologous recombination by BRCA1-A complex disruption, suppresses this toxicity, highlighting a crucial role for ATM in preventing toxic LIG4-mediated chromosome fusions. Notably, suppressor mutations in ATM-mutant backgrounds are different to those in BRCA1-mutant scenarios, suggesting new opportunities for patient stratification and additional therapeutic vulnerabilities for clinical exploitation.


Asunto(s)
Antineoplásicos/farmacología , Proteínas de la Ataxia Telangiectasia Mutada/genética , Reparación del ADN por Unión de Extremidades/genética , Resistencia a Antineoplásicos/genética , Animales , Antineoplásicos/uso terapéutico , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteína BRCA1/metabolismo , Sistemas CRISPR-Cas/genética , Línea Celular Tumoral , Supervivencia Celular/genética , Roturas del ADN de Doble Cadena/efectos de los fármacos , ADN Ligasa (ATP)/metabolismo , Replicación del ADN/efectos de los fármacos , Replicación del ADN/genética , Femenino , Humanos , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Células Madre Embrionarias de Ratones , Mutación , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Ftalazinas/farmacología , Ftalazinas/uso terapéutico , Piperazinas/farmacología , Piperazinas/uso terapéutico , Topotecan/farmacología , Topotecan/uso terapéutico
11.
Nat Cell Biol ; 20(8): 954-965, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30022119

RESUMEN

BRCA1 deficiencies cause breast, ovarian, prostate and other cancers, and render tumours hypersensitive to poly(ADP-ribose) polymerase (PARP) inhibitors. To understand the resistance mechanisms, we conducted whole-genome CRISPR-Cas9 synthetic-viability/resistance screens in BRCA1-deficient breast cancer cells treated with PARP inhibitors. We identified two previously uncharacterized proteins, C20orf196 and FAM35A, whose inactivation confers strong PARP-inhibitor resistance. Mechanistically, we show that C20orf196 and FAM35A form a complex, 'Shieldin' (SHLD1/2), with FAM35A interacting with single-stranded DNA through its C-terminal oligonucleotide/oligosaccharide-binding fold region. We establish that Shieldin acts as the downstream effector of 53BP1/RIF1/MAD2L2 to promote DNA double-strand break (DSB) end-joining by restricting DSB resection and to counteract homologous recombination by antagonizing BRCA2/RAD51 loading in BRCA1-deficient cells. Notably, Shieldin inactivation further sensitizes BRCA1-deficient cells to cisplatin, suggesting how defining the SHLD1/2 status of BRCA1-deficient tumours might aid patient stratification and yield new treatment opportunities. Highlighting this potential, we document reduced SHLD1/2 expression in human breast cancers displaying intrinsic or acquired PARP-inhibitor resistance.


Asunto(s)
Proteína BRCA1/genética , Neoplasias Óseas/tratamiento farmacológico , Neoplasias de la Mama/tratamiento farmacológico , Reparación del ADN por Unión de Extremidades , Resistencia a Antineoplásicos , Osteosarcoma/tratamiento farmacológico , Neoplasias Ováricas/tratamiento farmacológico , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Proteínas/metabolismo , Reparación del ADN por Recombinación , Animales , Proteína BRCA1/deficiencia , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proteínas de Ciclo Celular , Línea Celular Tumoral , Cisplatino/farmacología , Roturas del ADN de Doble Cadena , Proteínas de Unión al ADN , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos/genética , Femenino , Células HEK293 , Humanos , Proteínas Mad2/genética , Proteínas Mad2/metabolismo , Ratones , Complejos Multiproteicos , Osteosarcoma/genética , Osteosarcoma/metabolismo , Osteosarcoma/patología , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Proteínas/genética , Proteínas de Unión a Telómeros/genética , Proteínas de Unión a Telómeros/metabolismo , Proteína 1 de Unión al Supresor Tumoral P53/genética , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Nat Commun ; 9(1): 1700, 2018 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-29703891

RESUMEN

Hutchinson-Gilford Progeria Syndrome (HGPS) is a rare, but devastating genetic disease characterized by segmental premature aging, with cardiovascular disease being the main cause of death. Cells from HGPS patients accumulate progerin, a permanently farnesylated, toxic form of Lamin A, disrupting the nuclear shape and chromatin organization, leading to DNA-damage accumulation and senescence. Therapeutic approaches targeting farnesylation or aiming to reduce progerin levels have provided only partial health improvements. Recently, we identified Remodelin, a small-molecule agent that leads to amelioration of HGPS cellular defects through inhibition of the enzyme N-acetyltransferase 10 (NAT10). Here, we show the preclinical data demonstrating that targeting NAT10 in vivo, either via chemical inhibition or genetic depletion, significantly enhances the healthspan in a Lmna G609G HGPS mouse model. Collectively, the data provided here highlights NAT10 as a potential therapeutic target for HGPS.


Asunto(s)
Envejecimiento Prematuro/tratamiento farmacológico , Inestabilidad Genómica/efectos de los fármacos , Hidrazonas/farmacología , Acetiltransferasa A N-Terminal/antagonistas & inhibidores , Progeria/tratamiento farmacológico , Tiazoles/farmacología , Envejecimiento Prematuro/genética , Envejecimiento Prematuro/mortalidad , Envejecimiento Prematuro/patología , Animales , Daño del ADN/efectos de los fármacos , Daño del ADN/genética , Modelos Animales de Enfermedad , Femenino , Inestabilidad Genómica/genética , Humanos , Hidrazonas/uso terapéutico , Estimación de Kaplan-Meier , Lamina Tipo A/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Acetiltransferasa A N-Terminal/genética , Acetiltransferasa A N-Terminal/metabolismo , Acetiltransferasas N-Terminal , Progeria/genética , Progeria/mortalidad , Progeria/patología , Tiazoles/uso terapéutico
13.
Viruses ; 9(11)2017 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-29144403

RESUMEN

Herpes simplex virus 1 (HSV-1) has extensive interactions with the host DNA damage response (DDR) machinery that can be either detrimental or beneficial to the virus. Proteins in the homologous recombination pathway are known to be required for efficient replication of the viral genome, while different members of the classical non-homologous end-joining (c-NHEJ) pathway have opposing effects on HSV-1 infection. Here, we have investigated the role of the recently-discovered c-NHEJ component, PAXX (Paralogue of XRCC4 and XLF), which we found to be excluded from the nucleus during HSV-1 infection. We have established that cells lacking PAXX have an intact innate immune response to HSV-1 but show a defect in viral genome replication efficiency. Counterintuitively, PAXX-/- cells were able to produce greater numbers of infectious virions, indicating that PAXX acts to restrict HSV-1 infection in a manner that is different from other c-NHEJ factors.


Asunto(s)
Reparación del ADN por Unión de Extremidades , Proteínas de Unión al ADN/metabolismo , Herpes Simple/virología , Herpesvirus Humano 1/fisiología , Animales , Línea Celular , Genes Virales/genética , Genoma Viral , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/crecimiento & desarrollo , Humanos , Interferones/análisis , Interferones/biosíntesis , Ratones , Proteínas Virales/biosíntesis , Virión/aislamiento & purificación , Replicación Viral
14.
Genes Dev ; 30(19): 2152-2157, 2016 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-27798842

RESUMEN

PAXX was identified recently as a novel nonhomologous end-joining DNA repair factor in human cells. To characterize its physiological roles, we generated Paxx-deficient mice. Like Xlf-/- mice, Paxx-/- mice are viable, grow normally, and are fertile but show mild radiosensitivity. Strikingly, while Paxx loss is epistatic with Ku80, Lig4, and Atm deficiency, Paxx/Xlf double-knockout mice display embryonic lethality associated with genomic instability, cell death in the central nervous system, and an almost complete block in lymphogenesis, phenotypes that closely resemble those of Xrcc4-/- and Lig4-/- mice. Thus, combined loss of Paxx and Xlf is synthetic-lethal in mammals.


Asunto(s)
Proteínas de Unión al ADN/genética , Desarrollo Embrionario/genética , Mutaciones Letales Sintéticas/genética , Trisacáridos/genética , Animales , Apoptosis/genética , Proteínas de Unión al ADN/metabolismo , Epistasis Genética , Inestabilidad Genómica/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Tolerancia a Radiación/genética , Trisacáridos/metabolismo
15.
Cell Rep ; 16(11): 2967-2979, 2016 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-27601299

RESUMEN

Paralog of XRCC4 and XLF (PAXX) is a member of the XRCC4 superfamily and plays a role in nonhomologous end-joining (NHEJ), a DNA repair pathway critical for lymphocyte antigen receptor gene assembly. Here, we find that the functions of PAXX and XLF in V(D)J recombination are masked by redundant joining activities. Thus, combined PAXX and XLF deficiency leads to an inability to join RAG-cleaved DNA ends. Additionally, we demonstrate that PAXX function in V(D)J recombination depends on its interaction with Ku. Importantly, we show that, unlike XLF, the role of PAXX during the repair of DNA breaks does not overlap with ATM and the RAG complex. Our findings illuminate the role of PAXX in V(D)J recombination and support a model in which PAXX and XLF function during NHEJ repair of DNA breaks, whereas XLF, the RAG complex, and the ATM-dependent DNA damage response promote end joining by stabilizing DNA ends.


Asunto(s)
Linfocitos B/metabolismo , Enzimas Reparadoras del ADN/metabolismo , Proteínas de Unión al ADN/química , Homología de Secuencia de Aminoácido , Recombinación V(D)J/genética , Animales , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Sistemas CRISPR-Cas/genética , Daño del ADN , Reparación del ADN , Proteínas de Unión al ADN/metabolismo , Eliminación de Gen , Edición Génica , Reordenamiento Génico de Linfocito B , Inmunoglobulinas/genética , Autoantígeno Ku/metabolismo , Modelos Biológicos , Proteínas Oncogénicas v-abl/metabolismo
16.
J Biol Chem ; 291(31): 16011-23, 2016 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-27467582

RESUMEN

Li-Fraumeni syndrome (LFS) patients harbor germ line mutations in the TP53 gene and are at increased risk of hormone receptor-positive breast cancers. Recently, elevated levels of aromatase, the rate-limiting enzyme for estrogen biosynthesis, were found in the breast tissue of LFS patients. Although p53 down-regulates aromatase expression, the underlying mechanisms are incompletely understood. In the present study, we found that LFS stromal cells expressed higher levels of Hsp90 ATPase activity and aromatase compared with wild-type stromal cells. Inhibition of Hsp90 ATPase suppressed aromatase expression. Silencing Aha1 (activator of Hsp90 ATPase 1), a co-chaperone of Hsp90 required for its ATPase activity, led to both inhibition of Hsp90 ATPase activity and reduced aromatase expression. In comparison with wild-type stromal cells, increased levels of the Hsp90 client proteins, HIF-1α, and PKM2 were found in LFS stromal cells. A complex comprised of HIF-1α and PKM2 was recruited to the aromatase promoter II in LFS stromal cells. Silencing either HIF-1α or PKM2 suppressed aromatase expression in LFS stromal cells. CP-31398, a p53 rescue compound, suppressed levels of Aha1, Hsp90 ATPase activity, levels of PKM2 and HIF-1α, and aromatase expression in LFS stromal cells. Consistent with these in vitro findings, levels of Hsp90 ATPase activity, Aha1, HIF-1α, PKM2, and aromatase were increased in the mammary glands of p53 null versus wild-type mice. PKM2 and HIF-1α were shown to co-localize in the nucleus of stromal cells of LFS breast tissue. Taken together, our results show that the Aha1-Hsp90-PKM2/HIF-1α axis mediates the induction of aromatase in LFS.


Asunto(s)
Tejido Adiposo/metabolismo , Aromatasa/biosíntesis , Mama/metabolismo , Proteínas Portadoras/metabolismo , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Proteínas HSP90 de Choque Térmico/metabolismo , Síndrome de Li-Fraumeni/metabolismo , Glándulas Mamarias Animales/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Hormonas Tiroideas/metabolismo , Tejido Adiposo/patología , Animales , Aromatasa/genética , Mama/patología , Proteínas Portadoras/genética , Línea Celular , Femenino , Proteínas HSP90 de Choque Térmico/genética , Humanos , Síndrome de Li-Fraumeni/genética , Síndrome de Li-Fraumeni/patología , Glándulas Mamarias Animales/patología , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Proteínas de Neoplasias/genética , Células del Estroma/metabolismo , Células del Estroma/patología , Hormonas Tiroideas/genética , Proteínas de Unión a Hormona Tiroide
17.
G3 (Bethesda) ; 6(8): 2343-54, 2016 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-27233670

RESUMEN

In mammals the regulation of genomic instability plays a key role in tumor suppression and also controls genome plasticity, which is important for recombination during the processes of immunity and meiosis. Most studies to identify regulators of genomic instability have been performed in cells in culture or in systems that report on gross rearrangements of the genome, yet subtle differences in the level of genomic instability can contribute to whole organism phenotypes such as tumor predisposition. Here we performed a genome-wide association study in a population of 1379 outbred Crl:CFW(SW)-US_P08 mice to dissect the genetic landscape of micronucleus formation, a biomarker of chromosomal breaks, whole chromosome loss, and extranuclear DNA. Variation in micronucleus levels is a complex trait with a genome-wide heritability of 53.1%. We identify seven loci influencing micronucleus formation (false discovery rate <5%), and define candidate genes at each locus. Intriguingly at several loci we find evidence for sexual dimorphism in micronucleus formation, with a locus on chromosome 11 being specific to males.


Asunto(s)
Rotura Cromosómica , Estudio de Asociación del Genoma Completo , Micronúcleos con Defecto Cromosómico , Sitios de Carácter Cuantitativo/genética , Animales , Mapeo Cromosómico , Femenino , Inestabilidad Genómica , Genotipo , Masculino , Ratones , Fenotipo , Polimorfismo de Nucleótido Simple , Caracteres Sexuales
18.
Nat Protoc ; 10(1): 205-15, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25551665

RESUMEN

We describe a sensitive, robust, high-throughput method for quantifying the formation of micronuclei, markers of genome instability, in mouse erythrocytes. Micronuclei are whole chromosomes or chromosome segments that have been separated from the nucleus. Other methods of detection rely on labor-intensive, microscopy-based techniques. Here we describe a 2-d, 96-well plate-based flow cytometric method of micronucleus scoring that is simple enough for a research technician experienced in flow cytometry to perform. The assay detects low levels of genome instability that cannot be readily identified by classic phenotyping, using 25 µl of blood. By using this assay, we have screened >10,000 blood samples and discovered novel genes that contribute to vertebrate genome maintenance, as well as novel disease models and mechanisms of genome instability disorders. We discuss experimental design considerations, including statistical power calculation, we provide troubleshooting tips and we discuss factors that contribute to a false-positive increase in the number of micronucleated red blood cells and to experimental variability.


Asunto(s)
Eritropoyesis/fisiología , Inestabilidad Genómica/genética , Ensayos Analíticos de Alto Rendimiento/métodos , Pruebas de Micronúcleos/métodos , Animales , Eritrocitos/metabolismo , Eritropoyesis/genética , Citometría de Flujo/métodos , Ratones
19.
Curr Opin Genet Dev ; 24: 1-7, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24657530

RESUMEN

Genome instability is a feature of nearly all cancers and can be exploited for therapy. In addition, a growing number of genome maintenance genes have been associated with developmental disorders. Efforts to understand the role of genome instability in these processes will be greatly facilitated by a more comprehensive understanding of their genetic network. We highlight recent genetic screens in model organisms that have assisted in the discovery of novel regulators of genome stability and focus on the contribution of mice as a model organism to understanding the role of genome instability during embryonic development, tumour formation and cancer therapy.


Asunto(s)
Genoma , Neoplasias/genética , Animales , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Pruebas Genéticas , Inestabilidad Genómica , Humanos , Ratones
20.
J Biol Chem ; 289(10): 6513-6525, 2014 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-24451373

RESUMEN

The p53 tumor suppressor gene encodes a homotetrameric transcription factor which is activated in response to a variety of cellular stressors, including DNA damage and oncogene activation. p53 mutations occur in >50% of human cancers. Although p53 has been shown to regulate Wnt signaling, the underlying mechanisms are not well understood. Here we show that silencing p53 in colon cancer cells led to increased expression of Aha1, a co-chaperone of Hsp90. Heat shock factor-1 was important for mediating the changes in Aha1 levels. Increased Aha1 levels were associated with enhanced interactions with Hsp90, resulting in increased Hsp90 ATPase activity. Moreover, increased Hsp90 ATPase activity resulted in increased phosphorylation of Akt and glycogen synthase kinase-3ß (GSK3ß), leading to enhanced expression of Wnt target genes. Significantly, levels of Aha1, Hsp90 ATPase activity, Akt, and GSK3ß phosphorylation and expression of Wnt target genes were increased in the colons of p53-null as compared with p53 wild type mice. Using p53 heterozygous mutant epithelial cells from Li-Fraumeni syndrome patients, we show that a monoallelic mutation of p53 was sufficient to activate the Aha1/Hsp90 ATPase axis leading to stimulation of Wnt signaling and increased expression of Wnt target genes. Pharmacologic intervention with CP-31398, a p53 rescue agent, inhibited recruitment of Aha1 to Hsp90 and suppressed Wnt-mediated gene expression in colon cancer cells. Taken together, this study provides new insights into the mechanism by which p53 regulates Wnt signaling and raises the intriguing possibility that p53 status may affect the efficacy of anticancer therapies targeting Hsp90 ATPase.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Neoplasias del Colon/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas HSP90 de Choque Térmico/metabolismo , Chaperonas Moleculares/genética , Proteína p53 Supresora de Tumor/metabolismo , Vía de Señalización Wnt/genética , Animales , Neoplasias del Colon/genética , Modelos Animales de Enfermedad , Humanos , Síndrome de Li-Fraumeni , Ratones , Ratones Transgénicos , Pirimidinas/farmacología , Proteína p53 Supresora de Tumor/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...