Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
1.
Cell ; 172(1-2): 373-386.e10, 2018 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-29224780

RESUMEN

Breast cancer (BC) comprises multiple distinct subtypes that differ genetically, pathologically, and clinically. Here, we describe a robust protocol for long-term culturing of human mammary epithelial organoids. Using this protocol, >100 primary and metastatic BC organoid lines were generated, broadly recapitulating the diversity of the disease. BC organoid morphologies typically matched the histopathology, hormone receptor status, and HER2 status of the original tumor. DNA copy number variations as well as sequence changes were consistent within tumor-organoid pairs and largely retained even after extended passaging. BC organoids furthermore populated all major gene-expression-based classification groups and allowed in vitro drug screens that were consistent with in vivo xeno-transplantations and patient response. This study describes a representative collection of well-characterized BC organoids available for cancer research and drug development, as well as a strategy to assess in vitro drug response in a personalized fashion.


Asunto(s)
Neoplasias de la Mama/patología , Heterogeneidad Genética , Organoides/patología , Bancos de Tejidos , Animales , Antineoplásicos/farmacología , Neoplasias de la Mama/genética , Células Cultivadas , Ensayos de Selección de Medicamentos Antitumorales/métodos , Femenino , Humanos , Ratones , Ratones Desnudos , Organoides/efectos de los fármacos , Medicina de Precisión/métodos
2.
Cell ; 160(1-2): 299-312, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25533785

RESUMEN

Despite the enormous replication potential of the human liver, there are currently no culture systems available that sustain hepatocyte replication and/or function in vitro. We have shown previously that single mouse Lgr5+ liver stem cells can be expanded as epithelial organoids in vitro and can be differentiated into functional hepatocytes in vitro and in vivo. We now describe conditions allowing long-term expansion of adult bile duct-derived bipotent progenitor cells from human liver. The expanded cells are highly stable at the chromosome and structural level, while single base changes occur at very low rates. The cells can readily be converted into functional hepatocytes in vitro and upon transplantation in vivo. Organoids from α1-antitrypsin deficiency and Alagille syndrome patients mirror the in vivo pathology. Clonal long-term expansion of primary adult liver stem cells opens up experimental avenues for disease modeling, toxicology studies, regenerative medicine, and gene therapy.


Asunto(s)
Hígado/citología , Técnicas de Cultivo de Órganos , Animales , Inestabilidad Genómica , Hepatocitos/citología , Humanos , Ratones , Organoides/citología
3.
Cell ; 160(1-2): 324-38, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25557080

RESUMEN

Pancreatic cancer is one of the most lethal malignancies due to its late diagnosis and limited response to treatment. Tractable methods to identify and interrogate pathways involved in pancreatic tumorigenesis are urgently needed. We established organoid models from normal and neoplastic murine and human pancreas tissues. Pancreatic organoids can be rapidly generated from resected tumors and biopsies, survive cryopreservation, and exhibit ductal- and disease-stage-specific characteristics. Orthotopically transplanted neoplastic organoids recapitulate the full spectrum of tumor development by forming early-grade neoplasms that progress to locally invasive and metastatic carcinomas. Due to their ability to be genetically manipulated, organoids are a platform to probe genetic cooperation. Comprehensive transcriptional and proteomic analyses of murine pancreatic organoids revealed genes and pathways altered during disease progression. The confirmation of many of these protein changes in human tissues demonstrates that organoids are a facile model system to discover characteristics of this deadly malignancy.


Asunto(s)
Carcinoma Ductal Pancreático/patología , Modelos Biológicos , Técnicas de Cultivo de Órganos , Organoides/patología , Neoplasias Pancreáticas/patología , Animales , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Páncreas/metabolismo , Páncreas/patología
4.
Cell ; 159(1): 163-175, 2014 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-25201529

RESUMEN

The prostate gland consists of basal and luminal cells arranged as pseudostratified epithelium. In tissue recombination models, only basal cells reconstitute a complete prostate gland, yet murine lineage-tracing experiments show that luminal cells generate basal cells. It has remained challenging to address the molecular details of these transitions and whether they apply to humans, due to the lack of culture conditions that recapitulate prostate gland architecture. Here, we describe a 3D culture system that supports long-term expansion of primary mouse and human prostate organoids, composed of fully differentiated CK5+ basal and CK8+ luminal cells. Organoids are genetically stable, reconstitute prostate glands in recombination assays, and can be experimentally manipulated. Single human luminal and basal cells give rise to organoids, yet luminal-cell-derived organoids more closely resemble prostate glands. These data support a luminal multilineage progenitor cell model for prostate tissue and establish a robust, scalable system for mechanistic studies.


Asunto(s)
Técnicas de Cultivo de Órganos , Organoides , Próstata/citología , Andrógenos/metabolismo , Humanos , Masculino , Células Madre/citología , Células Madre/metabolismo
5.
Nature ; 597(7876): 381-386, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34433962

RESUMEN

Over the course of an individual's lifetime, normal human cells accumulate mutations1. Here we compare the mutational landscape in 29 cell types from the soma and germline using multiple samples from the same individuals. Two ubiquitous mutational signatures, SBS1 and SBS5/40, accounted for the majority of acquired mutations in most cell types, but their absolute and relative contributions varied substantially. SBS18, which potentially reflects oxidative damage2, and several additional signatures attributed to exogenous and endogenous exposures contributed mutations to subsets of cell types. The rate of mutation was lowest in spermatogonia, the stem cells from which sperm are generated and from which most genetic variation in the human population is thought to originate. This was due to low rates of ubiquitous mutational processes and may be partially attributable to a low rate of cell division in basal spermatogonia. These results highlight similarities and differences in the maintenance of the germline and soma.


Asunto(s)
Células Germinativas/metabolismo , Mutación de Línea Germinal , Tasa de Mutación , Especificidad de Órganos/genética , Anciano , Células Clonales/metabolismo , Femenino , Salud , Humanos , Masculino , Microdisección , Persona de Mediana Edad , Estrés Oxidativo , Espermatogonias/metabolismo
6.
Nature ; 580(7802): 269-273, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32106218

RESUMEN

Various species of the intestinal microbiota have been associated with the development of colorectal cancer1,2, but it has not been demonstrated that bacteria have a direct role in the occurrence of oncogenic mutations. Escherichia coli can carry the pathogenicity island pks, which encodes a set of enzymes that synthesize colibactin3. This compound is believed to alkylate DNA on adenine residues4,5 and induces double-strand breaks in cultured cells3. Here we expose human intestinal organoids to genotoxic pks+ E. coli by repeated luminal injection over five months. Whole-genome sequencing of clonal organoids before and after this exposure revealed a distinct mutational signature that was absent from organoids injected with isogenic pks-mutant bacteria. The same mutational signature was detected in a subset of 5,876 human cancer genomes from two independent cohorts, predominantly in colorectal cancer. Our study describes a distinct mutational signature in colorectal cancer and implies that the underlying mutational process results directly from past exposure to bacteria carrying the colibactin-producing pks pathogenicity island.


Asunto(s)
Neoplasias Colorrectales/genética , Neoplasias Colorrectales/microbiología , Escherichia coli/genética , Escherichia coli/patogenicidad , Islas Genómicas/genética , Mutagénesis , Mutación , Técnicas de Cocultivo , Estudios de Cohortes , Secuencia de Consenso , Daño del ADN , Microbioma Gastrointestinal , Humanos , Organoides/citología , Organoides/metabolismo , Organoides/microbiología , Péptidos/genética , Policétidos
7.
PLoS Biol ; 20(1): e3001527, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35089911

RESUMEN

CRISPR-associated nucleases are powerful tools for precise genome editing of model systems, including human organoids. Current methods describing fluorescent gene tagging in organoids rely on the generation of DNA double-strand breaks (DSBs) to stimulate homology-directed repair (HDR) or non-homologous end joining (NHEJ)-mediated integration of the desired knock-in. A major downside associated with DSB-mediated genome editing is the required clonal selection and expansion of candidate organoids to verify the genomic integrity of the targeted locus and to confirm the absence of off-target indels. By contrast, concurrent nicking of the genomic locus and targeting vector, known as in-trans paired nicking (ITPN), stimulates efficient HDR-mediated genome editing to generate large knock-ins without introducing DSBs. Here, we show that ITPN allows for fast, highly efficient, and indel-free fluorescent gene tagging in human normal and cancer organoids. Highlighting the ease and efficiency of ITPN, we generate triple fluorescent knock-in organoids where 3 genomic loci were simultaneously modified in a single round of targeting. In addition, we generated model systems with allele-specific readouts by differentially modifying maternal and paternal alleles in one step. ITPN using our palette of targeting vectors, publicly available from Addgene, is ideally suited for generating error-free heterozygous knock-ins in human organoids.


Asunto(s)
ADN/genética , Desoxirribonucleasa I/metabolismo , Sitios Genéticos , Organoides/metabolismo , Reparación del ADN por Recombinación , Coloración y Etiquetado/métodos , Alelos , Secuencia de Bases , Proteína 9 Asociada a CRISPR/genética , Proteína 9 Asociada a CRISPR/metabolismo , Colon/citología , Colon/metabolismo , ADN/metabolismo , Reparación del ADN por Unión de Extremidades , Desoxirribonucleasa I/genética , Electroporación/métodos , Células Epiteliales/citología , Células Epiteliales/metabolismo , Colorantes Fluorescentes/química , Colorantes Fluorescentes/metabolismo , Técnicas de Sustitución del Gen , Vectores Genéticos , Genoma Humano , Heterocigoto , Humanos , Organoides/citología
8.
J Pathol ; 263(2): 217-225, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38551073

RESUMEN

Environmental factors like the pathogenicity island polyketide synthase positive (pks+) Escherichia coli (E. coli) could have potential for risk stratification in colorectal cancer (CRC) screening. The association between pks+ E. coli measured in fecal immunochemical test (FIT) samples and the detection of advanced neoplasia (AN) at colonoscopy was investigated. Biobanked FIT samples were analyzed for both presence of E. coli and pks+ E. coli and correlated with colonoscopy findings; 5020 CRC screening participants were included. Controls were participants in which no relevant lesion was detected because of FIT-negative results (cut-off ≥15 µg Hb/g feces), a negative colonoscopy, or a colonoscopy during which only a nonadvanced polyp was detected. Cases were participants with AN [CRC, advanced adenoma (AA), or advanced serrated polyp (ASP)]. Existing DNA isolation and quantitative polymerase chain reaction (qPCR) procedures were used for the detection of E. coli and pks+ E. coli in stool. A total of 4542 (90.2%) individuals were E. coli positive, and 1322 (26.2%) were pks+ E. coli positive. The prevalence of E. coli in FIT samples from individuals with AN was 92.9% compared to 89.7% in FIT samples of controls (p = 0.010). The prevalence of pks+ E. coli in FIT samples from individuals with AN (28.6%) and controls (25.9%) was not significantly different (p = 0.13). The prevalences of pks+ E. coli in FIT samples from individuals with CRC, AA, or ASP were 29.6%, 28.3%, and 32.1%, respectively. In conclusion, the prevalence of pks+ E. coli in a screening population was 26.2% and did not differ significantly between individuals with AN and controls. These findings disqualify the straightforward option of using a snapshot measurement of pks+ E. coli in FIT samples as a stratification biomarker for CRC risk. © 2024 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Asunto(s)
Neoplasias Colorrectales , Detección Precoz del Cáncer , Escherichia coli , Heces , Sintasas Poliquetidas , Humanos , Neoplasias Colorrectales/microbiología , Neoplasias Colorrectales/diagnóstico , Heces/microbiología , Heces/enzimología , Escherichia coli/aislamiento & purificación , Escherichia coli/enzimología , Escherichia coli/genética , Masculino , Detección Precoz del Cáncer/métodos , Femenino , Persona de Mediana Edad , Anciano , Sintasas Poliquetidas/genética , Colonoscopía , Factores de Riesgo , Adenoma/microbiología , Adenoma/diagnóstico , Medición de Riesgo , Biomarcadores de Tumor , Estudios de Casos y Controles
9.
Proc Natl Acad Sci U S A ; 118(47)2021 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-34795059

RESUMEN

Barrett's esophagus (BE) is categorized, based on morphological appearance, into different stages, which correlate with the risk of developing esophageal adenocarcinoma. More advanced stages are more likely to acquire chromosomal instabilities, but stage-specific markers remain elusive. Here, we performed single-cell DNA-sequencing experiments (scDNAseq) with fresh BE biopsies. Dysplastic BE cells frequently contained chromosomal instability (CIN) regions, and these CIN cells carried mutations corresponding to the COSMIC mutational signature SBS17, which were not present in biopsy-matched chromosomally stable (CS) cells or patient-matched nondiseased control cells. CS cells were predominantly found in nondysplastic BE biopsies. The single-base substitution (SBS) signatures of all CS BE cells analyzed were indistinguishable from those of nondiseased esophageal or gastric cells. Single-cell RNA-sequencing (scRNAseq) experiments with BE biopsies identified two sets of marker genes which facilitate the distinction between columnar BE epithelium and nondysplastic/dysplastic stages. Moreover, histological validation confirmed a correlation between increased CLDN2 expression and the presence of dysplastic BE stages. Our scDNAseq and scRNAseq datasets, which are a useful resource for the community, provide insight into the mutational landscape and gene expression pattern at different stages of BE development.


Asunto(s)
Esófago de Barrett/genética , Esófago de Barrett/patología , Análisis de la Célula Individual/métodos , Adenocarcinoma/genética , Esófago de Barrett/diagnóstico , Biomarcadores , Biopsia , Inestabilidad Cromosómica , Epitelio , Esófago , Expresión Génica , Humanos , Hiperplasia/patología , Mutación , Análisis de Secuencia de ADN , Secuenciación del Exoma
10.
Proc Natl Acad Sci U S A ; 118(36)2021 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-34479993

RESUMEN

Neuroblastomas are childhood tumors with frequent fatal relapses after induction treatment, which is related to tumor evolution with additional genomic events. Our whole-genome sequencing data analysis revealed a high frequency of somatic cytosine > adenine (C > A) substitutions in primary neuroblastoma tumors, which was associated with poor survival. We showed that increased levels of C > A substitutions correlate with copy number loss (CNL) of OGG1 or MUTYH Both genes encode DNA glycosylases that recognize 8-oxo-guanine (8-oxoG) lesions as a first step of 8-oxoG repair. Tumor organoid models with CNL of OGG1 or MUTYH show increased 8-oxoG levels compared to wild-type cells. We used CRISPR-Cas9 genome editing to create knockout clones of MUTYH and OGG1 in neuroblastoma cells. Whole-genome sequencing of single-cell OGG1 and MUTYH knockout clones identified an increased accumulation of C > A substitutions. Mutational signature analysis of these OGG1 and MUTYH knockout clones revealed enrichment for C > A signatures 18 and 36, respectively. Clustering analysis showed that the knockout clones group together with tumors containing OGG1 or MUTYH CNL. In conclusion, we demonstrate that defects in 8-oxoG repair cause accumulation of C > A substitutions in neuroblastoma, which contributes to mutagenesis and tumor evolution.


Asunto(s)
Reparación del ADN/genética , Guanosina/análogos & derivados , Neuroblastoma/genética , Adenina/metabolismo , Niño , Citosina/metabolismo , Daño del ADN , ADN Glicosilasas/genética , ADN Glicosilasas/metabolismo , Femenino , Guanina/metabolismo , Guanosina/genética , Guanosina/metabolismo , Humanos , Masculino , Mutagénesis , Recurrencia Local de Neoplasia/genética , Neuroblastoma/metabolismo , Estrés Oxidativo , Polimorfismo de Nucleótido Simple/genética
11.
BMC Genomics ; 23(1): 134, 2022 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-35168570

RESUMEN

BACKGROUND: The collective of somatic mutations in a genome represents a record of mutational processes that have been operative in a cell. These processes can be investigated by extracting relevant mutational patterns from sequencing data. RESULTS: Here, we present the next version of MutationalPatterns, an R/Bioconductor package, which allows in-depth mutational analysis of catalogues of single and double base substitutions as well as small insertions and deletions. Major features of the package include the possibility to perform regional mutation spectra analyses and the possibility to detect strand asymmetry phenomena, such as lesion segregation. On top of this, the package also contains functions to determine how likely it is that a signature can cause damaging mutations (i.e., mutations that affect protein function). This updated package supports stricter signature refitting on known signatures in order to prevent overfitting. Using simulated mutation matrices containing varied signature contributions, we showed that reliable refitting can be achieved even when only 50 mutations are present per signature. Additionally, we incorporated bootstrapped signature refitting to assess the robustness of the signature analyses. Finally, we applied the package on genome mutation data of cell lines in which we deleted specific DNA repair processes and on large cancer datasets, to show how the package can be used to generate novel biological insights. CONCLUSIONS: This novel version of MutationalPatterns allows for more comprehensive analyses and visualization of mutational patterns in order to study the underlying processes. Ultimately, in-depth mutational analyses may contribute to improved biological insights in mechanisms of mutation accumulation as well as aid cancer diagnostics. MutationalPatterns is freely available at http://bioconductor.org/packages/MutationalPatterns .


Asunto(s)
Genoma Humano , Neoplasias , Análisis Mutacional de ADN , Reparación del ADN , Humanos , Mutación , Acumulación de Mutaciones , Neoplasias/genética
12.
Genome Res ; 29(7): 1067-1077, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31221724

RESUMEN

Nucleotide excision repair (NER) is one of the main DNA repair pathways that protect cells against genomic damage. Disruption of this pathway can contribute to the development of cancer and accelerate aging. Mutational characteristics of NER-deficiency may reveal important diagnostic opportunities, as tumors deficient in NER are more sensitive to certain treatments. Here, we analyzed the genome-wide somatic mutational profiles of adult stem cells (ASCs) from NER-deficient Ercc1 -/Δ mice. Our results indicate that NER-deficiency increases the base substitution load twofold in liver but not in small intestinal ASCs, which coincides with the tissue-specific aging pathology observed in these mice. Moreover, NER-deficient ASCs of both tissues show an increased contribution of Signature 8 mutations, which is a mutational pattern with unknown etiology that is recurrently observed in various cancer types. The scattered genomic distribution of the base substitutions indicates that deficiency of global-genome NER (GG-NER) underlies the observed mutational consequences. In line with this, we observe increased Signature 8 mutations in a GG-NER-deficient human organoid culture, in which XPC was deleted using CRISPR-Cas9 gene-editing. Furthermore, genomes of NER-deficient breast tumors show an increased contribution of Signature 8 mutations compared with NER-proficient tumors. Elevated levels of Signature 8 mutations could therefore contribute to a predictor of NER-deficiency based on a patient's mutational profile.


Asunto(s)
Reparación del ADN/genética , Mutación , Neoplasias/genética , Células Madre Adultas , Animales , Neoplasias de la Mama/genética , Estudios de Cohortes , Análisis Mutacional de ADN , ADN de Neoplasias , Proteínas de Unión al ADN/genética , Endonucleasas/genética , Femenino , Humanos , Ratones , Organoides , Técnicas de Cultivo de Tejidos , Secuenciación Completa del Genoma
13.
Immunity ; 39(2): 298-310, 2013 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-23954131

RESUMEN

Foxp3 is crucial for both the development and function of regulatory T (Treg) cells; however, the posttranslational mechanisms regulating Foxp3 transcriptional output remain poorly defined. Here, we demonstrate that T cell factor 1 (TCF1) and Foxp3 associates in Treg cells and that active Wnt signaling disrupts Foxp3 transcriptional activity. A global chromatin immunoprecipitation sequencing comparison in Treg cells revealed considerable overlap between Foxp3 and Wnt target genes. The activation of Wnt signaling reduced Treg-mediated suppression both in vitro and in vivo, whereas disruption of Wnt signaling in Treg cells enhanced their suppressive capacity. The activation of effector T cells increased Wnt3a production, and Wnt3a levels were found to be greatly increased in mononuclear cells isolated from synovial fluid versus peripheral blood of arthritis patients. We propose a model in which Wnt produced under inflammatory conditions represses Treg cell function, allowing a productive immune response, but, if uncontrolled, could lead to the development of autoimmunity.


Asunto(s)
Artritis/inmunología , Colitis/inmunología , Factores de Transcripción Forkhead/metabolismo , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Linfocitos T Reguladores/inmunología , Animales , Proliferación Celular , Células Cultivadas , Factores de Transcripción Forkhead/genética , Células HEK293 , Factor Nuclear 1-alfa del Hepatocito/genética , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Líquido Sinovial/citología , Linfocitos T Reguladores/metabolismo , Vía de Señalización Wnt , Proteína Wnt3A/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
14.
Nature ; 538(7624): 260-264, 2016 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-27698416

RESUMEN

The gradual accumulation of genetic mutations in human adult stem cells (ASCs) during life is associated with various age-related diseases, including cancer. Extreme variation in cancer risk across tissues was recently proposed to depend on the lifetime number of ASC divisions, owing to unavoidable random mutations that arise during DNA replication. However, the rates and patterns of mutations in normal ASCs remain unknown. Here we determine genome-wide mutation patterns in ASCs of the small intestine, colon and liver of human donors with ages ranging from 3 to 87 years by sequencing clonal organoid cultures derived from primary multipotent cells. Our results show that mutations accumulate steadily over time in all of the assessed tissue types, at a rate of approximately 40 novel mutations per year, despite the large variation in cancer incidence among these tissues. Liver ASCs, however, have different mutation spectra compared to those of the colon and small intestine. Mutational signature analysis reveals that this difference can be attributed to spontaneous deamination of methylated cytosine residues in the colon and small intestine, probably reflecting their high ASC division rate. In liver, a signature with an as-yet-unknown underlying mechanism is predominant. Mutation spectra of driver genes in cancer show high similarity to the tissue-specific ASC mutation spectra, suggesting that intrinsic mutational processes in ASCs can initiate tumorigenesis. Notably, the inter-individual variation in mutation rate and spectra are low, suggesting tissue-specific activity of common mutational processes throughout life.


Asunto(s)
Células Madre Adultas/metabolismo , Envejecimiento/genética , Acumulación de Mutaciones , Tasa de Mutación , Especificidad de Órganos , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Niño , Preescolar , Colon/metabolismo , Análisis Mutacional de ADN , Femenino , Genes Relacionados con las Neoplasias/genética , Humanos , Incidencia , Intestino Delgado/metabolismo , Hígado/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Células Madre Multipotentes/metabolismo , Neoplasias/epidemiología , Neoplasias/genética , Organoides/metabolismo , Mutación Puntual/genética , Adulto Joven
15.
Nature ; 521(7550): 43-7, 2015 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-25924068

RESUMEN

Crypt stem cells represent the cells of origin for intestinal neoplasia. Both mouse and human intestinal stem cells can be cultured in medium containing the stem-cell-niche factors WNT, R-spondin, epidermal growth factor (EGF) and noggin over long time periods as epithelial organoids that remain genetically and phenotypically stable. Here we utilize CRISPR/Cas9 technology for targeted gene modification of four of the most commonly mutated colorectal cancer genes (APC, P53 (also known as TP53), KRAS and SMAD4) in cultured human intestinal stem cells. Mutant organoids can be selected by removing individual growth factors from the culture medium. Quadruple mutants grow independently of all stem-cell-niche factors and tolerate the presence of the P53 stabilizer nutlin-3. Upon xenotransplantation into mice, quadruple mutants grow as tumours with features of invasive carcinoma. Finally, combined loss of APC and P53 is sufficient for the appearance of extensive aneuploidy, a hallmark of tumour progression.


Asunto(s)
Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Intestinos/patología , Mutación/genética , Organoides/metabolismo , Organoides/patología , Células Madre/patología , Aneuploidia , Animales , Sistemas CRISPR-Cas , Niño , Preescolar , Neoplasias Colorrectales/metabolismo , Femenino , Genes APC , Genes p53/genética , Xenoinjertos , Humanos , Imidazoles , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Mucosa Intestinal/metabolismo , Ratones , Persona de Mediana Edad , Mutagénesis Sitio-Dirigida , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Trasplante de Neoplasias , Piperazinas , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Proteína Smad4/deficiencia , Nicho de Células Madre/fisiología , Células Madre/metabolismo
16.
Nature ; 513(7518): 422-425, 2014 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-25043003

RESUMEN

The somatic mutations present in the genome of a cell accumulate over the lifetime of a multicellular organism. These mutations can provide insights into the developmental lineage tree, the number of divisions that each cell has undergone and the mutational processes that have been operative. Here we describe whole genomes of clonal lines derived from multiple tissues of healthy mice. Using somatic base substitutions, we reconstructed the early cell divisions of each animal, demonstrating the contributions of embryonic cells to adult tissues. Differences were observed between tissues in the numbers and types of mutations accumulated by each cell, which likely reflect differences in the number of cell divisions they have undergone and varying contributions of different mutational processes. If somatic mutation rates are similar to those in mice, the results indicate that precise insights into development and mutagenesis of normal human cells will be possible.


Asunto(s)
Linaje de la Célula/genética , Células Clonales/citología , Células Clonales/metabolismo , Genoma/genética , Mutagénesis/genética , Mutación/genética , Animales , Relojes Biológicos/genética , División Celular , Células Cultivadas , Embrión de Mamíferos/citología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Tasa de Mutación , Organoides/citología , Organoides/metabolismo , Filogenia , Análisis de Secuencia de ADN , Cola (estructura animal)/citología
17.
Proc Natl Acad Sci U S A ; 114(52): E11190-E11198, 2017 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-29237753

RESUMEN

During kidney development, progressively committed progenitor cells give rise to the distinct segments of the nephron, the functional unit of the kidney. Similar segment-committed progenitor cells are thought to be involved in the homeostasis of adult kidney. However, markers for most segment-committed progenitor cells remain to be identified. Here, we evaluate Troy/TNFRSF19 as a segment-committed nephron progenitor cell marker. Troy is expressed in the ureteric bud during embryonic development. During postnatal nephrogenesis, Troy+ cells are present in the cortex and papilla and display an immature tubular phenotype. Tracing of Troy+ cells during nephrogenesis demonstrates that Troy+ cells clonally give rise to tubular structures that persist for up to 2 y after induction. Troy+ cells have a 40-fold higher capacity than Troy- cells to form organoids, which is considered a stem cell property in vitro. In the adult kidney, Troy+ cells are present in the papilla and these cells continue to contribute to collecting duct formation during homeostasis. The number of Troy-derived cells increases after folic acid-induced injury. Our data show that Troy marks a renal stem/progenitor cell population in the developing kidney that in adult kidney contributes to homeostasis, predominantly of the collecting duct, and regeneration.


Asunto(s)
Células Epiteliales/metabolismo , Nefronas/embriología , Organogénesis/fisiología , Receptores del Factor de Necrosis Tumoral/metabolismo , Células Madre/metabolismo , Animales , Ratones , Ratones Transgénicos , Receptores del Factor de Necrosis Tumoral/genética
18.
Proc Natl Acad Sci U S A ; 114(12): E2357-E2364, 2017 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-28270604

RESUMEN

In the adenoma-carcinoma sequence, it is proposed that intestinal polyps evolve through a set of defined mutations toward metastatic colorectal cancer (CRC). Here, we dissect this adenoma-carcinoma sequence in vivo by using an orthotopic organoid transplantation model of human colon organoids engineered to harbor different CRC mutation combinations. We demonstrate that sequential accumulation of oncogenic mutations in Wnt, EGFR, P53, and TGF-ß signaling pathways facilitates efficient tumor growth, migration, and metastatic colonization. We show that reconstitution of specific niche signals can restore metastatic growth potential of tumor cells lacking one of the oncogenic mutations. Our findings imply that the ability to metastasize-i.e., to colonize distant sites-is the direct consequence of the loss of dependency on specific niche signals.


Asunto(s)
Neoplasias Colorrectales/genética , Organoides/trasplante , Adulto , Animales , Movimiento Celular , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/fisiopatología , Modelos Animales de Enfermedad , Receptores ErbB/genética , Receptores ErbB/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Ingeniería Genética , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Persona de Mediana Edad , Mutación , Metástasis de la Neoplasia/genética , Procesos Neoplásicos , Organoides/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo
19.
BMC Cancer ; 19(1): 457, 2019 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-31092228

RESUMEN

BACKGROUND: In the past decade, systematic and comprehensive analyses of cancer genomes have identified cancer driver genes and revealed unprecedented insight into the molecular mechanisms underlying the initiation and progression of cancer. These studies illustrate that although every cancer has a unique genetic make-up, there are only a limited number of mechanisms that shape the mutational landscapes of cancer genomes, as reflected by characteristic computationally-derived mutational signatures. Importantly, the molecular mechanisms underlying specific signatures can now be dissected and coupled to treatment strategies. Systematic characterization of mutational signatures in a cancer patient's genome may thus be a promising new tool for molecular tumor diagnosis and classification. RESULTS: In this review, we describe the status of mutational signature analysis in cancer genomes and discuss the opportunities and relevance, as well as future challenges, for further implementation of mutational signatures in clinical tumor diagnostics and therapy guidance. CONCLUSIONS: Scientific studies have illustrated the potential of mutational signature analysis in cancer research. As such, we believe that the implementation of mutational signature analysis within the diagnostic workflow will improve cancer diagnosis in the future.


Asunto(s)
Análisis Mutacional de ADN/métodos , Neoplasias/diagnóstico , Humanos , Técnicas de Diagnóstico Molecular , Neoplasias/genética , Secuenciación Completa del Genoma
20.
Blood ; 126(18): 2098-109, 2015 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-26289642

RESUMEN

Expression of the forkhead transcription factor FOXP1 is essential for early B-cell development, whereas downregulation of FOXP1 at the germinal center (GC) stage is required for GC B-cell function. Aberrantly high FOXP1 expression is frequently observed in diffuse large B-cell lymphoma and mucosa-associated lymphoid tissue lymphoma, being associated with poor prognosis. Here, by gene expression analysis upon ectopic overexpression of FOXP1 in primary human memory B cells (MBCs) and B-cell lines, combined with chromatin immunoprecipitation and sequencing, we established that FOXP1 directly represses expression of PRDM1, IRF4, and XBP1, transcriptional master regulators of plasma cell (PC) differentiation. In accordance, FOXP1 is prominently expressed in primary human naive and MBCs, but expression strongly decreases during PC differentiation. Moreover, as compared with immunoglobulin (Ig) M(+) MBCs, IgG(+) MBCs combine lower expression of FOXP1 with an enhanced intrinsic PC differentiation propensity, and constitutive (over)expression of FOXP1 in B-cell lines and primary human MBCs represses their ability to differentiate into PCs. Taken together, our data indicate that proper control of FOXP1 expression plays a critical role in PC differentiation, whereas aberrant expression of FOXP1 might contribute to lymphomagenesis by blocking this terminal B-cell differentiation.


Asunto(s)
Linfocitos B/citología , Factores de Transcripción Forkhead/metabolismo , Células Plasmáticas/citología , Proteínas Represoras/metabolismo , Linfocitos B/metabolismo , Diferenciación Celular , Línea Celular , Células Cultivadas , Factores de Transcripción Forkhead/genética , Humanos , Inmunoglobulina G/análisis , Inmunoglobulina M/análisis , Células Plasmáticas/metabolismo , Proteínas Represoras/genética , Activación Transcripcional , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda