Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 101
Filtrar
1.
Acta Neuropathol ; 147(1): 9, 2024 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-38175301

RESUMEN

Nuclear clearance and cytoplasmic accumulations of the RNA-binding protein TDP-43 are pathological hallmarks in almost all patients with amyotrophic lateral sclerosis (ALS) and up to 50% of patients with frontotemporal dementia (FTD) and Alzheimer's disease. In Alzheimer's disease, TDP-43 pathology is predominantly observed in the limbic system and correlates with cognitive decline and reduced hippocampal volume. Disruption of nuclear TDP-43 function leads to abnormal RNA splicing and incorporation of erroneous cryptic exons in numerous transcripts including Stathmin-2 (STMN2, also known as SCG10) and UNC13A, recently reported in tissues from patients with ALS and FTD. Here, we identify both STMN2 and UNC13A cryptic exons in Alzheimer's disease patients, that correlate with TDP-43 pathology burden, but not with amyloid-ß or tau deposits. We also demonstrate that processing of the STMN2 pre-mRNA is more sensitive to TDP-43 loss of function than UNC13A. In addition, full-length RNAs encoding STMN2 and UNC13A are suppressed in large RNA-seq datasets generated from Alzheimer's disease post-mortem brain tissue. Collectively, these results open exciting new avenues to use STMN2 and UNC13A as potential therapeutic targets in a broad range of neurodegenerative conditions with TDP-43 proteinopathy including Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer , Esclerosis Amiotrófica Lateral , Demencia Frontotemporal , Enfermedad de Pick , Humanos , Enfermedad de Alzheimer/genética , Proteínas de Unión al ADN/genética , Empalme del ARN , ARN Mensajero/genética , Estatmina/genética
2.
Nat Neurosci ; 27(1): 34-47, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37996528

RESUMEN

The mRNA transcript of the human STMN2 gene, encoding for stathmin-2 protein (also called SCG10), is profoundly impacted by TAR DNA-binding protein 43 (TDP-43) loss of function. The latter is a hallmark of several neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS). Using a combination of approaches, including transient antisense oligonucleotide-mediated suppression, sustained shRNA-induced depletion in aging mice, and germline deletion, we show that stathmin-2 has an important role in the establishment and maintenance of neurofilament-dependent axoplasmic organization that is critical for preserving the caliber and conduction velocity of myelinated large-diameter axons. Persistent stathmin-2 loss in adult mice results in pathologies found in ALS, including reduced interneurofilament spacing, axonal caliber collapse that drives tearing within outer myelin layers, diminished conduction velocity, progressive motor and sensory deficits, and muscle denervation. These findings reinforce restoration of stathmin-2 as an attractive therapeutic approach for ALS and other TDP-43-dependent neurodegenerative diseases.


Asunto(s)
Esclerosis Amiotrófica Lateral , Animales , Ratones , Esclerosis Amiotrófica Lateral/metabolismo , Axones/fisiología , Desnervación , Proteínas de Unión al ADN/genética , Filamentos Intermedios/metabolismo , Filamentos Intermedios/patología , Neuronas Motoras/metabolismo , Estatmina/genética , Estatmina/metabolismo
3.
Nat Rev Genet ; 25(3): 196-210, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37938738

RESUMEN

Complex chromosome rearrangements, known as chromoanagenesis, are widespread in cancer. Based on large-scale DNA sequencing of human tumours, the most frequent type of complex chromosome rearrangement is chromothripsis, a massive, localized and clustered rearrangement of one (or a few) chromosomes seemingly acquired in a single event. Chromothripsis can be initiated by mitotic errors that produce a micronucleus encapsulating a single chromosome or chromosomal fragment. Rupture of the unstable micronuclear envelope exposes its chromatin to cytosolic nucleases and induces chromothriptic shattering. Found in up to half of tumours included in pan-cancer genomic analyses, chromothriptic rearrangements can contribute to tumorigenesis through inactivation of tumour suppressor genes, activation of proto-oncogenes, or gene amplification through the production of self-propagating extrachromosomal circular DNAs encoding oncogenes or genes conferring anticancer drug resistance. Here, we discuss what has been learned about the mechanisms that enable these complex genomic rearrangements and their consequences in cancer.


Asunto(s)
Cromotripsis , Neoplasias , Humanos , Cromatina , ADN/genética , Núcleo Celular , Neoplasias/genética , Reordenamiento Génico , Aberraciones Cromosómicas
4.
J Med Chem ; 66(22): 15084-15093, 2023 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-37937963

RESUMEN

Biomolecular condensates are proposed to cause diseases, such as cancer and neurodegeneration, by concentrating proteins at abnormal subcellular loci. Imaging-based compound screens have been used to identify small molecules that reverse or promote biomolecular condensates. However, limitations of conventional imaging-based methods restrict the screening scale. Here, we used a graph convolutional network (GCN)-based computational approach and identified small molecule candidates that reduce the nuclear liquid-liquid phase separation of TAR DNA-binding protein 43 (TDP-43), an essential protein that undergoes phase transition in neurodegenerative diseases. We demonstrated that the GCN-based deep learning algorithm is suitable for spatial information extraction from the molecular graph. Thus, this is a promising method to identify small molecule candidates with novel scaffolds. Furthermore, we validated that these candidates do not affect the normal splicing function of TDP-43. Taken together, a combination of an imaging-based screen and a GCN-based deep learning method dramatically improves the speed and accuracy of the compound screen for biomolecular condensates.


Asunto(s)
Condensados Biomoleculares , Aprendizaje Profundo , Algoritmos , Proteínas de Unión al ADN , Diagnóstico por Imagen
5.
Nature ; 618(7967): 1049-1056, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37316668

RESUMEN

Chromothripsis, the shattering and imperfect reassembly of one (or a few) chromosome(s)1, is an ubiquitous2 mutational process generating localized and complex chromosomal rearrangements that drive genome evolution in cancer. Chromothripsis can be initiated by mis-segregation errors in mitosis3,4 or DNA metabolism5-7 that lead to entrapment of chromosomes within micronuclei and their subsequent fragmentation in the next interphase or following mitotic entry6,8-10. Here we use inducible degrons to demonstrate that chromothriptically produced pieces of a micronucleated chromosome are tethered together in mitosis by a protein complex consisting of mediator of DNA damage checkpoint 1 (MDC1), DNA topoisomerase II-binding protein 1 (TOPBP1) and cellular inhibitor of PP2A (CIP2A), thereby enabling en masse segregation to the same daughter cell. Such tethering is shown to be crucial for the viability of cells undergoing chromosome mis-segregation and shattering after transient inactivation of the spindle assembly checkpoint. Transient, degron-induced reduction in CIP2A following chromosome micronucleation-dependent chromosome shattering is shown to drive acquisition of segmental deletions and inversions. Analyses of pancancer tumour genomes showed that expression of CIP2A and TOPBP1 was increased overall in cancers with genomic rearrangements, including copy number-neutral chromothripsis with minimal deletions, but comparatively reduced in cancers with canonical chromothripsis in which deletions were frequent. Thus, chromatin-bound tethers maintain the proximity of fragments of a shattered chromosome enabling their re-encapsulation into, and religation within, a daughter cell nucleus to form heritable, chromothriptically rearranged chromosomes found in the majority of human cancers.


Asunto(s)
Núcleo Celular , Segregación Cromosómica , Cromosomas Humanos , Cromotripsis , Mitosis , Humanos , Núcleo Celular/genética , Núcleo Celular/metabolismo , Neoplasias/genética , Cromatina/genética
6.
Nature ; 616(7957): 504-509, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37046091

RESUMEN

Epstein-Barr virus (EBV) is an oncogenic herpesvirus associated with several cancers of lymphocytic and epithelial origin1-3. EBV encodes EBNA1, which binds to a cluster of 20 copies of an 18-base-pair palindromic sequence in the EBV genome4-6. EBNA1 also associates with host chromosomes at non-sequence-specific sites7, thereby enabling viral persistence. Here we show that the sequence-specific DNA-binding domain of EBNA1 binds to a cluster of tandemly repeated copies of an EBV-like, 18-base-pair imperfect palindromic sequence encompassing a region of about 21 kilobases at human chromosome 11q23. In situ visualization of the repetitive EBNA1-binding site reveals aberrant structures on mitotic chromosomes characteristic of inherently fragile DNA. We demonstrate that increasing levels of EBNA1 binding trigger dose-dependent breakage at 11q23, producing a fusogenic centromere-containing fragment and an acentric distal fragment, with both mis-segregated into micronuclei in the next cell cycles. In cells latently infected with EBV, elevating EBNA1 abundance by as little as twofold was sufficient to trigger breakage at 11q23. Examination of whole-genome sequencing of EBV-associated nasopharyngeal carcinomas revealed that structural variants are highly enriched on chromosome 11. Presence of EBV is also shown to be associated with an enrichment of chromosome 11 rearrangements across 2,439 tumours from 38 cancer types. Our results identify a previously unappreciated link between EBV and genomic instability, wherein EBNA1-induced breakage at 11q23 triggers acquisition of structural variations in chromosome 11.


Asunto(s)
Rotura Cromosómica , ADN , Herpesvirus Humano 4 , Proteínas Virales , Humanos , Sitios de Unión , ADN/química , ADN/metabolismo , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/metabolismo , Herpesvirus Humano 4/patogenicidad , Proteínas Virales/genética , Proteínas Virales/metabolismo , Roturas del ADN de Doble Cadena , Cromosomas Humanos Par 11/química , Cromosomas Humanos Par 11/genética , Cromosomas Humanos Par 11/metabolismo , Inestabilidad Genómica , Mitosis
7.
Science ; 379(6637): 1140-1149, 2023 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-36927019

RESUMEN

Loss of nuclear TDP-43 is a hallmark of neurodegeneration in TDP-43 proteinopathies, including amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). TDP-43 mislocalization results in cryptic splicing and polyadenylation of pre-messenger RNAs (pre-mRNAs) encoding stathmin-2 (also known as SCG10), a protein that is required for axonal regeneration. We found that TDP-43 binding to a GU-rich region sterically blocked recognition of the cryptic 3' splice site in STMN2 pre-mRNA. Targeting dCasRx or antisense oligonucleotides (ASOs) suppressed cryptic splicing, which restored axonal regeneration and stathmin-2-dependent lysosome trafficking in TDP-43-deficient human motor neurons. In mice that were gene-edited to contain human STMN2 cryptic splice-polyadenylation sequences, ASO injection into cerebral spinal fluid successfully corrected Stmn2 pre-mRNA misprocessing and restored stathmin-2 expression levels independently of TDP-43 binding.


Asunto(s)
Proteínas de Unión al ADN , Edición Génica , Poliadenilación , Empalme del ARN , Estatmina , Proteinopatías TDP-43 , Animales , Humanos , Ratones , Proteínas de Unión al ADN/metabolismo , Precursores del ARN/genética , Precursores del ARN/metabolismo , Estatmina/genética , Estatmina/metabolismo , Proteinopatías TDP-43/genética , Proteinopatías TDP-43/terapia , Sitios de Empalme de ARN , Oligonucleótidos Antisentido/genética , Proyección Neuronal
8.
Nat Cell Biol ; 24(9): 1378-1393, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36075972

RESUMEN

While acetylated, RNA-binding-deficient TDP-43 reversibly phase separates within nuclei into complex droplets (anisosomes) comprised of TDP-43-containing liquid outer shells and liquid centres of HSP70-family chaperones, cytoplasmic aggregates of TDP-43 are hallmarks of multiple neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS). Here we show that transient oxidative stress, proteasome inhibition or inhibition of the ATP-dependent chaperone activity of HSP70 provokes reversible cytoplasmic TDP-43 de-mixing and transition from liquid to gel/solid, independently of RNA binding or stress granules. Isotope labelling mass spectrometry was used to identify that phase-separated cytoplasmic TDP-43 is bound by the small heat-shock protein HSPB1. Binding is direct, mediated through TDP-43's RNA binding and low-complexity domains. HSPB1 partitions into TDP-43 droplets, inhibits TDP-43 assembly into fibrils, and is essential for disassembly of stress-induced TDP-43 droplets. A decrease in HSPB1 promotes cytoplasmic TDP-43 de-mixing and mislocalization. HSPB1 depletion was identified in spinal motor neurons of patients with ALS containing aggregated TDP-43. These findings identify HSPB1 to be a regulator of cytoplasmic TDP-43 phase separation and aggregation.


Asunto(s)
Proteínas de Unión al ADN , Proteínas de Choque Térmico Pequeñas , Proteínas de Choque Térmico , Transición de Fase , Adenosina Trifosfato , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/metabolismo , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas de Choque Térmico/química , Proteínas de Choque Térmico/metabolismo , Humanos , Chaperonas Moleculares/genética , Complejo de la Endopetidasa Proteasomal , ARN/metabolismo
9.
Mol Neurodegener ; 16(1): 61, 2021 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-34488813

RESUMEN

Mutations in FUS, an RNA-binding protein involved in multiple steps of RNA metabolism, are associated with the most severe forms of amyotrophic lateral sclerosis (ALS). Accumulation of cytoplasmic FUS is likely to be a major culprit in the toxicity of FUS mutations. Thus, preventing cytoplasmic mislocalization of the FUS protein may represent a valuable therapeutic strategy. FUS binds to its own pre-mRNA creating an autoregulatory loop efficiently buffering FUS excess through multiple proposed mechanisms including retention of introns 6 and/or 7. Here, we introduced a wild-type FUS gene allele, retaining all intronic sequences, in mice whose heterozygous or homozygous expression of a cytoplasmically retained FUS protein (Fus∆NLS) was previously shown to provoke ALS-like disease or postnatal lethality, respectively. Wild-type FUS completely rescued the early lethality caused by the two Fus∆NLS alleles, and improved the age-dependent motor deficits and reduced lifespan caused by heterozygous expression of mutant FUS∆NLS. Mechanistically, wild-type FUS decreased the load of cytoplasmic FUS, increased retention of introns 6 and 7 in the endogenous mouse Fus mRNA, and decreased expression of the mutant mRNA. Thus, the wild-type FUS allele activates the homeostatic autoregulatory loop, maintaining constant FUS levels and decreasing the mutant protein in the cytoplasm. These results provide proof of concept that an autoregulatory competent wild-type FUS expression could protect against this devastating, currently intractable, neurodegenerative disease.


Asunto(s)
Esclerosis Amiotrófica Lateral/terapia , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/genética , Proteína FUS de Unión a ARN/fisiología , Alelos , Esclerosis Amiotrófica Lateral/genética , Animales , Citoplasma/metabolismo , Demencia Frontotemporal/genética , Genes Letales , Prueba de Complementación Genética , Humanos , Intrones/genética , Ratones , Ratones Transgénicos , Mutación , Unión Proteica , Precursores del ARN/metabolismo , Proteína FUS de Unión a ARN/deficiencia , Proteína FUS de Unión a ARN/genética , Proteínas Recombinantes/metabolismo , Transgenes
10.
Genes Dev ; 35(15-16): 1093-1108, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34266887

RESUMEN

Abnormal numerical and structural chromosome content is frequently found in human cancer. To test the role of aneuploidy in tumor initiation and progression, we generated mice with random aneuploidies by transient induction of polo-like kinase 4 (Plk4), a master regulator of centrosome number. Short-term chromosome instability (CIN) from transient Plk4 induction resulted in formation of aggressive T-cell lymphomas in mice with heterozygous inactivation of one p53 allele and accelerated tumor development in the absence of p53. Transient CIN increased the frequency of lymphoma-initiating cells with a specific karyotype profile, including trisomy of chromosomes 4, 5, 14, and 15 occurring early in tumorigenesis. Tumor development in mice with chronic CIN induced by an independent mechanism (through inactivation of the spindle assembly checkpoint) gradually trended toward a similar karyotypic profile, as determined by single-cell whole-genome DNA sequencing. Overall, we show how transient CIN generates cells with random aneuploidies from which ones that acquire a karyotype with specific chromosome gains are sufficient to drive cancer formation, and that distinct CIN mechanisms can lead to similar karyotypic cancer-causing outcomes.


Asunto(s)
Aneuploidia , Inestabilidad Cromosómica , Animales , Transformación Celular Neoplásica/genética , Centrosoma , Inestabilidad Cromosómica/genética , Evolución Clonal , Inestabilidad Genómica/genética , Ratones
11.
Nat Neurosci ; 24(8): 1089-1099, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34083786

RESUMEN

Methods to enhance adult neurogenesis by reprogramming glial cells into neurons enable production of new neurons in the adult nervous system. Development of therapeutically viable approaches to induce new neurons is now required to bring this concept to clinical application. Here, we successfully generate new neurons in the cortex and dentate gyrus of the aged adult mouse brain by transiently suppressing polypyrimidine tract binding protein 1 using an antisense oligonucleotide delivered by a single injection into cerebral spinal fluid. Radial glial-like cells and other GFAP-expressing cells convert into new neurons that, over a 2-month period, acquire mature neuronal character in a process mimicking normal neuronal maturation. The new neurons functionally integrate into endogenous circuits and modify mouse behavior. Thus, generation of new neurons in the dentate gyrus of the aging brain can be achieved with a therapeutically feasible approach, thereby opening prospects for production of neurons to replace those lost to neurodegenerative disease.


Asunto(s)
Giro Dentado , Células Ependimogliales , Neurogénesis/fisiología , Neuronas , Proteína de Unión al Tracto de Polipirimidina/antagonistas & inhibidores , Animales , Reprogramación Celular/fisiología , Giro Dentado/citología , Giro Dentado/fisiología , Células Ependimogliales/citología , Células Ependimogliales/fisiología , Ratones , Neuronas/citología , Neuronas/fisiología , Oligonucleótidos Antisentido
12.
Proc Natl Acad Sci U S A ; 118(19)2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33952700

RESUMEN

An aneuploid-immune paradox encompasses somatic copy-number alterations (SCNAs), unleashing a cytotoxic response in experimental precancer systems, while conversely being associated with immune suppression and cytotoxic-cell depletion in human tumors, especially head and neck cancer (HNSC). We present evidence from patient samples and cell lines that alterations in chromosome dosage contribute to an immune hot-to-cold switch during human papillomavirus-negative (HPV-) head and neck tumorigenesis. Overall SCNA (aneuploidy) level was associated with increased CD3+ and CD8+ T cell microenvironments in precancer (mostly CD3+, linked to trisomy and aneuploidy), but with T cell-deficient tumors. Early lesions with 9p21.3 loss were associated with depletion of cytotoxic T cell infiltration in TP53 mutant tumors; and with aneuploidy were associated with increased NK-cell infiltration. The strongest driver of cytotoxic T cell and Immune Score depletion in oral cancer was 9p-arm level loss, promoting profound decreases of pivotal IFN-γ-related chemokines (e.g., CXCL9) and pathway genes. Chromosome 9p21.3 deletion contributed mainly to cell-intrinsic senescence suppression, but deletion of the entire arm was necessary to diminish levels of cytokine, JAK-STAT, and Hallmark NF-κB pathways. Finally, 9p arm-level loss and JAK2-PD-L1 codeletion (at 9p24) were predictive markers of poor survival in recurrent HPV- HNSC after anti-PD-1 therapy; likely amplified by independent aneuploidy-induced immune-cold microenvironments observed here. We hypothesize that 9p21.3 arm-loss expansion and epistatic interactions allow oral precancer cells to acquire properties to overcome a proimmunogenic aneuploid checkpoint, transform and invade. These findings enable distinct HNSC interception and precision-therapeutic approaches, concepts that may apply to other CN-driven neoplastic, immune or aneuploid diseases, and immunotherapies.


Asunto(s)
Aneuploidia , Deleción Cromosómica , Neoplasias de Cabeza y Cuello/genética , Evasión Inmune , Infecciones por Papillomavirus , Adulto , Anciano , Anciano de 80 o más Años , Antígeno B7-H1 , Complejo CD3 , Linfocitos T CD8-positivos , Línea Celular Tumoral , Cromosomas , Citocinas , Variaciones en el Número de Copia de ADN , Regulación Neoplásica de la Expresión Génica , Genes p53/genética , Humanos , Evasión Inmune/genética , Inmunoterapia , Janus Quinasa 2 , Persona de Mediana Edad , Infecciones por Papillomavirus/genética , Linfocitos T Citotóxicos , Microambiente Tumoral , Adulto Joven
14.
Curr Opin Cell Biol ; 70: 91-99, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33610905

RESUMEN

Micronuclei are small membrane-bounded compartments with a DNA content encapsulated by a nuclear envelope and spatially separated from the primary nucleus. Micronuclei have long been linked to chromosome instability, genome rearrangements, and mutagenesis. They are frequently found in cancers, during senescence, and after genotoxic stress. Compromised integrity of the micronuclear envelope delays or disrupts DNA replication, inhibits DNA repair, and exposes micronuclear DNA directly to cytoplasm. Micronuclei play a central role in tumorigenesis, with micronuclear DNA being a source of complex genome rearrangements (including chromothripsis) and promoting a cyclic GMP-AMP synthase (cGAS)-mediated cellular immune response that may contribute to cancer metastasis. Here, we discuss recent findings on how micronuclei are generated, what the consequences are, and what cellular mechanisms can be applied to protect against micronucleation.


Asunto(s)
Cromotripsis , Micronúcleos con Defecto Cromosómico , Daño del ADN , Inestabilidad Genómica , Humanos , Membrana Nuclear
15.
Science ; 371(6529)2021 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-33335017

RESUMEN

The RNA binding protein TDP-43 forms intranuclear or cytoplasmic aggregates in age-related neurodegenerative diseases. In this study, we found that RNA binding-deficient TDP-43 (produced by neurodegeneration-causing mutations or posttranslational acetylation in its RNA recognition motifs) drove TDP-43 demixing into intranuclear liquid spherical shells with liquid cores. These droplets, which we named "anisosomes", have shells that exhibit birefringence, thus indicating liquid crystal formation. Guided by mathematical modeling, we identified the primary components of the liquid core to be HSP70 family chaperones, whose adenosine triphosphate (ATP)-dependent activity maintained the liquidity of shells and cores. In vivo proteasome inhibition within neurons, to mimic aging-related reduction of proteasome activity, induced TDP-43-containing anisosomes. These structures converted to aggregates when ATP levels were reduced. Thus, acetylation, HSP70, and proteasome activities regulate TDP-43 phase separation and conversion into a gel or solid phase.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Agregado de Proteínas , Proteínas de Unión al ARN/metabolismo , Envejecimiento/metabolismo , Animales , Anisotropía , Microscopía por Crioelectrón , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Células HEK293 , Histona Desacetilasas/metabolismo , Humanos , Cristales Líquidos/química , Ratones , Ratones Endogámicos C57BL , Mutación , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , Neuronas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/farmacología , Dominios Proteicos , Proteínas de Unión al ARN/genética , Ratas , Ratas Sprague-Dawley
16.
Nature ; 591(7848): 137-141, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33361815

RESUMEN

Focal chromosomal amplification contributes to the initiation of cancer by mediating overexpression of oncogenes1-3, and to the development of cancer therapy resistance by increasing the expression of genes whose action diminishes the efficacy of anti-cancer drugs. Here we used whole-genome sequencing of clonal cell isolates that developed chemotherapeutic resistance to show that chromothripsis is a major driver of circular extrachromosomal DNA (ecDNA) amplification (also known as double minutes) through mechanisms that depend on poly(ADP-ribose) polymerases (PARP) and the catalytic subunit of DNA-dependent protein kinase (DNA-PKcs). Longitudinal analyses revealed that a further increase in drug tolerance is achieved by structural evolution of ecDNAs through additional rounds of chromothripsis. In situ Hi-C sequencing showed that ecDNAs preferentially tether near chromosome ends, where they re-integrate when DNA damage is present. Intrachromosomal amplifications that formed initially under low-level drug selection underwent continuing breakage-fusion-bridge cycles, generating amplicons more than 100 megabases in length that became trapped within interphase bridges and then shattered, thereby producing micronuclei whose encapsulated ecDNAs are substrates for chromothripsis. We identified similar genome rearrangement profiles linked to localized gene amplification in human cancers with acquired drug resistance or oncogene amplifications. We propose that chromothripsis is a primary mechanism that accelerates genomic DNA rearrangement and amplification into ecDNA and enables rapid acquisition of tolerance to altered growth conditions.


Asunto(s)
Cromotripsis , Evolución Molecular , Amplificación de Genes/genética , Neoplasias/genética , Oncogenes/genética , Daño del ADN , Reparación del ADN por Unión de Extremidades , ADN Circular/química , ADN Circular/metabolismo , ADN de Neoplasias/química , ADN de Neoplasias/metabolismo , Proteína Quinasa Activada por ADN , Resistencia a Antineoplásicos , Células HEK293 , Células HeLa , Humanos , Micronúcleos con Defecto Cromosómico , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Neoplasias/patología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Selección Genética , Secuenciación Completa del Genoma
17.
Nature ; 582(7813): 550-556, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32581380

RESUMEN

Parkinson's disease is characterized by loss of dopamine neurons in the substantia nigra1. Similar to other major neurodegenerative disorders, there are no disease-modifying treatments for Parkinson's disease. While most treatment strategies aim to prevent neuronal loss or protect vulnerable neuronal circuits, a potential alternative is to replace lost neurons to reconstruct disrupted circuits2. Here we report an efficient one-step conversion of isolated mouse and human astrocytes to functional neurons by depleting the RNA-binding protein PTB (also known as PTBP1). Applying this approach to the mouse brain, we demonstrate progressive conversion of astrocytes to new neurons that innervate into and repopulate endogenous neural circuits. Astrocytes from different brain regions are converted to different neuronal subtypes. Using a chemically induced model of Parkinson's disease in mouse, we show conversion of midbrain astrocytes to dopaminergic neurons, which provide axons to reconstruct the nigrostriatal circuit. Notably, re-innervation of striatum is accompanied by restoration of dopamine levels and rescue of motor deficits. A similar reversal of disease phenotype is also accomplished by converting astrocytes to neurons using antisense oligonucleotides to transiently suppress PTB. These findings identify a potentially powerful and clinically feasible approach to treating neurodegeneration by replacing lost neurons.


Asunto(s)
Astrocitos/citología , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/citología , Enfermedad de Parkinson/patología , Enfermedad de Parkinson/terapia , Sustancia Negra/citología , Sustancia Negra/fisiología , Animales , Axones/fisiología , Dopamina/biosíntesis , Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Femenino , Ribonucleoproteínas Nucleares Heterogéneas/deficiencia , Ribonucleoproteínas Nucleares Heterogéneas/genética , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Humanos , Técnicas In Vitro , Masculino , Ratones , Neostriado/citología , Neostriado/fisiología , Vías Nerviosas , Neurogénesis , Enfermedad de Parkinson/metabolismo , Fenotipo , Proteína de Unión al Tracto de Polipirimidina/deficiencia , Proteína de Unión al Tracto de Polipirimidina/genética , Proteína de Unión al Tracto de Polipirimidina/metabolismo , Sustancia Negra/metabolismo
18.
Nat Med ; 26(1): 118-130, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31873312

RESUMEN

Gene silencing with virally delivered shRNA represents a promising approach for treatment of inherited neurodegenerative disorders. In the present study we develop a subpial technique, which we show in adult animals successfully delivers adeno-associated virus (AAV) throughout the cervical, thoracic and lumbar spinal cord, as well as brain motor centers. One-time injection at cervical and lumbar levels just before disease onset in mice expressing a familial amyotrophic lateral sclerosis (ALS)-causing mutant SOD1 produces long-term suppression of motoneuron disease, including near-complete preservation of spinal α-motoneurons and muscle innervation. Treatment after disease onset potently blocks progression of disease and further α-motoneuron degeneration. A single subpial AAV9 injection in adult pigs or non-human primates using a newly designed device produces homogeneous delivery throughout the cervical spinal cord white and gray matter and brain motor centers. Thus, spinal subpial delivery in adult animals is highly effective for AAV-mediated gene delivery throughout the spinal cord and supraspinal motor centers.


Asunto(s)
Esclerosis Amiotrófica Lateral/terapia , Dependovirus/metabolismo , Silenciador del Gen , Técnicas de Transferencia de Gen , Neuronas Motoras/patología , Degeneración Nerviosa/terapia , Piamadre/patología , Médula Espinal/patología , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/fisiopatología , Animales , Atrofia , Progresión de la Enfermedad , Potenciales Evocados Motores , Femenino , Regulación de la Expresión Génica , Humanos , Inflamación/patología , Interneuronas/patología , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Desarrollo de Músculos , Degeneración Nerviosa/genética , Degeneración Nerviosa/fisiopatología , Piamadre/fisiopatología , Primates , Pliegue de Proteína , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/administración & dosificación , Médula Espinal/diagnóstico por imagen , Médula Espinal/fisiopatología , Superóxido Dismutasa-1/genética , Superóxido Dismutasa-1/metabolismo , Porcinos
19.
J Clin Invest ; 129(8): 3103-3120, 2019 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-31112137

RESUMEN

Mechanisms underlying motor neuron degeneration in amyotrophic lateral sclerosis (ALS) are yet unclear. Specific deletion of the ER-component membralin in astrocytes manifested postnatal motor defects and lethality in mice, causing the accumulation of extracellular glutamate through reducing the glutamate transporter EAAT2. Restoring EAAT2 levels in membralin KO astrocytes limited astrocyte-dependent excitotoxicity in motor neurons. Transcriptomic profiles from mouse astrocytic membralin KO motor cortex indicated significant perturbation in KEGG pathway components related to ALS, including downregulation of Eaat2 and upregulation of Tnfrsf1a. Changes in gene expression with membralin deletion also overlapped with mouse ALS models and reactive astrocytes. Our results shown that activation of TNF receptor (TNFR1)-NFκB pathway known to suppress Eaat2 transcription was upregulated with membralin deletion. Further, reduced membralin and EAAT2 levels correlated with disease progression in spinal cord from SOD1-mutant mouse models, and reductions in membralin/EAAT2 were observed in human ALS spinal cord. Importantly, overexpression of membralin in SOD1G93A astrocytes decreased TNFR1 levels and increased EAAT2 expression, and improved motor neuron survival. Importantly, upregulation of membralin in SOD1G93A mice significantly prolonged mouse survival. Together, our study provided a mechanism for ALS pathogenesis where membralin limited glutamatergic neurotoxicity, suggesting that modulating membralin had potentials in ALS therapy.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Astrocitos/metabolismo , Ácido Glutámico/metabolismo , Corteza Motora/metabolismo , Proteínas del Tejido Nervioso/deficiencia , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Animales , Astrocitos/patología , Regulación hacia Abajo , Transportador 2 de Aminoácidos Excitadores/biosíntesis , Transportador 2 de Aminoácidos Excitadores/genética , Ácido Glutámico/genética , Humanos , Ratones , Ratones Noqueados , Corteza Motora/patología , Proteínas del Tejido Nervioso/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/biosíntesis , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Superóxido Dismutasa/biosíntesis , Superóxido Dismutasa/genética , Transcripción Genética , Regulación hacia Arriba
20.
Nat Genet ; 51(4): 705-715, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30833795

RESUMEN

Cancer genomes are frequently characterized by numerical and structural chromosomal abnormalities. Here we integrated a centromere-specific inactivation approach with selection for a conditionally essential gene, a strategy termed CEN-SELECT, to systematically interrogate the structural landscape of mis-segregated chromosomes. We show that single-chromosome mis-segregation into a micronucleus can directly trigger a broad spectrum of genomic rearrangement types. Cytogenetic profiling revealed that mis-segregated chromosomes exhibit 120-fold-higher susceptibility to developing seven major categories of structural aberrations, including translocations, insertions, deletions, and complex reassembly through chromothripsis coupled to classical non-homologous end joining. Whole-genome sequencing of clonally propagated rearrangements identified random patterns of clustered breakpoints with copy-number alterations resulting in interspersed gene deletions and extrachromosomal DNA amplification events. We conclude that individual chromosome segregation errors during mitotic cell division are sufficient to drive extensive structural variations that recapitulate genomic features commonly associated with human disease.


Asunto(s)
Segregación Cromosómica/genética , Reordenamiento Génico/genética , Animales , Células Cultivadas , Aberraciones Cromosómicas , Variaciones en el Número de Copia de ADN/genética , Genoma Humano/genética , Genómica/métodos , Células HEK293 , Humanos , Neoplasias/genética , Translocación Genética/genética , Secuenciación Completa del Genoma/métodos , Xenopus laevis/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA