Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Hum Reprod ; 39(2): 275-281, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38099857

RESUMEN

Infertility is a complex condition affecting millions of couples worldwide. The current definition of infertility, based on clinical criteria, fails to account for the molecular and cellular changes that may occur during the development of infertility. Recent advancements in sequencing technology and single-cell analysis offer new opportunities to gain a deeper understanding of these changes. The endometrium has a potential role in infertility and has been extensively studied to identify gene expression profiles associated with (impaired) endometrial receptivity. However, limited overlap among studies hampers the identification of relevant downstream pathways that could play a role in the development of endometrial-related infertility. To address these challenges, we propose sequencing the endometrial transcriptome of healthy and infertile women at the single-cell level to consistently identify molecular signatures. Establishing consensus on physiological patterns in endometrial samples can aid in identifying deviations in infertile patients. A similar strategy has been used with great success in cancer research. However, large collaborative initiatives, international uniform protocols of sample collection and processing are crucial to ensure reliability and reproducibility. Overall, the proposed approach holds promise for an objective and accurate classification of endometrial-based infertility and has the potential to improve diagnosis and treatment outcomes.


Asunto(s)
Infertilidad Femenina , Femenino , Humanos , Infertilidad Femenina/diagnóstico , Infertilidad Femenina/genética , Infertilidad Femenina/metabolismo , Reproducibilidad de los Resultados , Endometrio/metabolismo , Transcriptoma , Resultado del Tratamiento , Implantación del Embrión/fisiología
2.
Sci Rep ; 13(1): 6874, 2023 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-37106015

RESUMEN

DNA methylation is important for establishing and maintaining cell identity and for genomic stability. This is achieved by regulating the accessibility of regulatory and transcriptional elements and the compaction of subtelomeric, centromeric, and other inactive genomic regions. Carcinogenesis is accompanied by a global loss in DNA methylation, which facilitates the transformation of cells. Cancer hypomethylation may also cause genomic instability, for example through interference with the protective function of telomeres and centromeres. However, understanding the role(s) of hypomethylation in tumor evolution is incomplete because the precise mutational consequences of global hypomethylation have thus far not been systematically assessed. Here we made genome-wide inventories of all possible genetic variation that accumulates in single cells upon the long-term global hypomethylation by CRISPR interference-mediated conditional knockdown of DNMT1. Depletion of DNMT1 resulted in a genomewide reduction in DNA methylation. The degree of DNA methylation loss was similar to that observed in many cancer types. Hypomethylated cells showed reduced proliferation rates, increased transcription of genes, reactivation of the inactive X-chromosome and abnormal nuclear morphologies. Prolonged hypomethylation was accompanied by increased chromosomal instability. However, there was no increase in mutational burden, enrichment for certain mutational signatures or accumulation of structural variation to the genome. In conclusion, the primary consequence of hypomethylation is genomic instability, which in cancer leads to increased tumor heterogeneity and thereby fuels cancer evolution.


Asunto(s)
Metilación de ADN , Inestabilidad Genómica , Humanos , Mutación , Carcinogénesis , ADN
3.
Nat Commun ; 13(1): 5915, 2022 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-36207433

RESUMEN

Genome-wide mutation analyses have revealed that specific anti-cancer drugs are highly mutagenic to cancer cells, but the mutational impact of anti-cancer therapies on normal cells is not known. Here, we examine genome-wide somatic mutation patterns in 42 healthy adult stem cells (ASCs) of the colon or the liver from 14 cancer patients (mean of 3.2 ASC per donor) that received systemic chemotherapy and/or local radiotherapy. The platinum-based chemo-drug Oxaliplatin induces on average 535 ± 260 mutations in colon ASC, while 5-FU shows a complete mutagenic absence in most, but not all colon ASCs. In contrast with the colon, normal liver ASCs escape mutagenesis from systemic treatment with Oxaliplatin and 5-FU. Thus, while chemotherapies are highly effective at killing cancer cells, their systemic use also increases the mutational burden of long-lived normal stem cells responsible for tissue renewal thereby increasing the risk for developing second cancers.


Asunto(s)
Células Madre Adultas , Células Madre , Adulto , Fluorouracilo , Humanos , Mutación , Oxaliplatino/farmacología
4.
Sci Rep ; 12(1): 10081, 2022 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-35710704

RESUMEN

Bladder cancer has a high recurrence rate and low survival of advanced stage patients. Few genetic drivers of bladder cancer have thus far been identified. We performed in-depth structural variant analysis on whole-genome sequencing data of 206 metastasized urinary tract cancers. In ~ 10% of the patients, we identified recurrent in-frame deletions of exons 8 and 9 in the aryl hydrocarbon receptor gene (AHRΔe8-9), which codes for a ligand-activated transcription factor. Pan-cancer analyses show that AHRΔe8-9 is highly specific to urinary tract cancer and mutually exclusive with other bladder cancer drivers. The ligand-binding domain of the AHRΔe8-9 protein is disrupted and we show that this results in ligand-independent AHR-pathway activation. In bladder organoids, AHRΔe8-9 induces a transformed phenotype that is characterized by upregulation of AHR target genes, downregulation of differentiation markers and upregulation of genes associated with stemness and urothelial cancer. Furthermore, AHRΔe8-9 expression results in anchorage independent growth of bladder organoids, indicating tumorigenic potential. DNA-binding deficient AHRΔe8-9 fails to induce transformation, suggesting a role for AHR target genes in the acquisition of the oncogenic phenotype. In conclusion, we show that AHRΔe8-9 is a novel driver of urinary tract cancer and that the AHR pathway could be an interesting therapeutic target.


Asunto(s)
Receptores de Hidrocarburo de Aril , Neoplasias de la Vejiga Urinaria , Exones/genética , Humanos , Ligandos , Mutación , Receptores de Hidrocarburo de Aril/metabolismo , Neoplasias de la Vejiga Urinaria/genética
5.
Aging Cell ; 21(4): e13562, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35246937

RESUMEN

Organs age differently, causing wide heterogeneity in multimorbidity, but underlying mechanisms are largely elusive. To investigate the basis of organ-specific ageing, we utilized progeroid repair-deficient Ercc1Δ/- mouse mutants and systematically compared at the tissue, stem cell and organoid level two organs representing ageing extremes. Ercc1Δ/- intestine shows hardly any accelerated ageing. Nevertheless, we found apoptosis and reduced numbers of intestinal stem cells (ISCs), but cell loss appears compensated by over-proliferation. ISCs retain their organoid-forming capacity, but organoids perform poorly in culture, compared with WT. Conversely, liver ages dramatically, even causing early death in Ercc1-KO mice. Apoptosis, p21, polyploidization and proliferation of various (stem) cells were prominently elevated in Ercc1Δ/- liver and stem cell populations were either largely unaffected (Sox9+), or expanding (Lgr5+), but were functionally exhausted in organoid formation and development in vitro. Paradoxically, while intestine displays less ageing, repair in WT ISCs appears inferior to liver as shown by enhanced sensitivity to various DNA-damaging agents, and lower lesion removal. Our findings reveal organ-specific anti-ageing strategies. Intestine, with short lifespan limiting time for damage accumulation and repair, favours apoptosis of damaged cells relying on ISC plasticity. Liver with low renewal rates depends more on repair pathways specifically protecting the transcribed compartment of the genome to promote sustained functionality and cell preservation. As shown before, the hematopoietic system with intermediate self-renewal mainly invokes replication-linked mechanisms, apoptosis and senescence. Hence, organs employ different genome maintenance strategies, explaining heterogeneity in organ ageing and the segmental nature of DNA-repair-deficient progerias.


Asunto(s)
Envejecimiento , Daño del ADN , Envejecimiento/genética , Envejecimiento/metabolismo , Animales , Daño del ADN/genética , Reparación del ADN , Ratones , Organoides/metabolismo , Células Madre/metabolismo
6.
iScience ; 25(2): 103736, 2022 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-35118356

RESUMEN

Induced pluripotent stem cells (iPSCs) hold great promise for regenerative medicine, but genetic instability is a major concern. Embryonic pluripotent cells also accumulate mutations during early development, but how this relates to the mutation burden in iPSCs remains unknown. Here, we directly compared the mutation burden of cultured iPSCs with their isogenic embryonic cells during human embryogenesis. We generated developmental lineage trees of human fetuses by phylogenetic inference from somatic mutations in the genomes of multiple stem cells, which were derived from different germ layers. Using this approach, we characterized the mutations acquired pre-gastrulation and found a rate of 1.65 mutations per cell division. When cultured in hypoxic conditions, iPSCs generated from fetal stem cells of the assessed fetuses displayed a similar mutation rate and spectrum. Our results show that iPSCs maintain a genomic integrity during culture at a similar degree as their pluripotent counterparts do in vivo.

7.
Commun Biol ; 4(1): 1301, 2021 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-34795391

RESUMEN

Inflammatory liver disease increases the risk of developing primary liver cancer. The mechanism through which liver disease induces tumorigenesis remains unclear, but is thought to occur via increased mutagenesis. Here, we performed whole-genome sequencing on clonally expanded single liver stem cells cultured as intrahepatic cholangiocyte organoids (ICOs) from patients with alcoholic cirrhosis, non-alcoholic steatohepatitis (NASH), and primary sclerosing cholangitis (PSC). Surprisingly, we find that these precancerous liver disease conditions do not result in a detectable increased accumulation of mutations, nor altered mutation types in individual liver stem cells. This finding contrasts with the mutational load and typical mutational signatures reported for liver tumors, and argues against the hypothesis that liver disease drives tumorigenesis via a direct mechanism of induced mutagenesis. Disease conditions in the liver may thus act through indirect mechanisms to drive the transition from healthy to cancerous cells, such as changes to the microenvironment that favor the outgrowth of precancerous cells.


Asunto(s)
Colangitis Esclerosante/genética , Cirrosis Hepática Alcohólica/genética , Hepatopatías/genética , Mutagénesis , Enfermedad del Hígado Graso no Alcohólico/genética , Lesiones Precancerosas/genética , Células Madre/metabolismo , Humanos , Hígado/fisiología , Organoides/metabolismo
8.
Nat Commun ; 11(1): 3932, 2020 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-32753580

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

9.
Sci Rep ; 10(1): 12991, 2020 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-32737409

RESUMEN

Children show a higher incidence of leukemia compared to young adolescents, yet their cells have less age-related (oncogenic) somatic mutations. Newborns with Down syndrome have an even higher risk of developing leukemia, which is thought to be driven by mutations that accumulate during fetal development. To characterize mutation accumulation in individual stem and progenitor cells of Down syndrome and karyotypically normal fetuses, we clonally expanded single cells and performed whole-genome sequencing. We found a higher mutation rate in haematopoietic stem and progenitor cells during fetal development compared to the post-infant rate. In fetal trisomy 21 cells the number of somatic mutations is even further increased, which was already apparent during the first cell divisions of embryogenesis before gastrulation. The number and types of mutations in fetal trisomy 21 haematopoietic stem and progenitor cells were similar to those in Down syndrome-associated myeloid preleukemia and could be attributed to mutational processes that were active during normal fetal haematopoiesis. Finally, we found that the contribution of early embryonic cells to human fetal tissues can vary considerably between individuals. The increased mutation rates found in this study, may contribute to the increased risk of leukemia early during life and the higher incidence of leukemia in Down syndrome.


Asunto(s)
Linaje de la Célula/genética , Síndrome de Down , Feto/metabolismo , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Acumulación de Mutaciones , Síndrome de Down/embriología , Síndrome de Down/genética , Síndrome de Down/patología , Femenino , Feto/patología , Células Madre Hematopoyéticas/patología , Humanos , Leucemia/embriología , Leucemia/genética , Leucemia/patología , Masculino , Secuenciación Completa del Genoma
10.
Sci Adv ; 6(16): eaaz7602, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32494621

RESUMEN

Genomic instability is common in human embryos, but the underlying causes are largely unknown. Here, we examined the consequences of sperm DNA damage on the embryonic genome by single-cell whole-genome sequencing of individual blastomeres from bovine embryos produced with sperm damaged by γ-radiation. Sperm DNA damage primarily leads to fragmentation of the paternal chromosomes followed by random distribution of the chromosomal fragments over the two sister cells in the first cell division. An unexpected secondary effect of sperm DNA damage is the induction of direct unequal cleavages, which include the poorly understood heterogoneic cell divisions. As a result, chaotic mosaicism is common in embryos derived from fertilizations with damaged sperm. The mosaic aneuploidies, uniparental disomies, and de novo structural variation induced by sperm DNA damage may compromise fertility and lead to rare congenital disorders when embryos escape developmental arrest.


Asunto(s)
Desarrollo Embrionario , Espermatozoides , Animales , Bovinos , Daño del ADN , Desarrollo Embrionario/genética , Femenino , Inestabilidad Genómica , Humanos , Masculino , Mosaicismo , Embarazo
11.
Nat Commun ; 11(1): 2493, 2020 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-32427826

RESUMEN

Genetic changes acquired during in vitro culture pose a risk for the successful application of stem cells in regenerative medicine. To assess the genetic risks induced by culturing, we determined all mutations in individual human stem cells by whole genome sequencing. Individual pluripotent, intestinal, and liver stem cells accumulate 3.5 ± 0.5, 7.2 ± 1.1 and 8.3 ± 3.6 base substitutions per population doubling, respectively. The annual in vitro mutation accumulation rate of adult stem cells is nearly 40-fold higher than the in vivo mutation accumulation rate. Mutational signature analysis reveals that in vitro induced mutations are caused by oxidative stress. Reducing oxygen tension in culture lowers the mutational load. We use the mutation rates, spectra, and genomic distribution to model the accumulation of oncogenic mutations during typical in vitro expansion, manipulation or screening experiments using human stem cells. Our study provides empirically defined parameters to assess the mutational risk of stem cell based therapies.


Asunto(s)
Células Madre Adultas/metabolismo , Análisis Mutacional de ADN/métodos , Células Madre Pluripotentes Inducidas/metabolismo , Mutación , Adulto , Células Madre Adultas/citología , Algoritmos , Células Cultivadas , Humanos , Células Madre Pluripotentes Inducidas/citología , Intestinos/citología , Hígado/citología , Hígado/metabolismo , Modelos Genéticos , Acumulación de Mutaciones , Tasa de Mutación , Medicina Regenerativa/métodos , Secuenciación Completa del Genoma/métodos
12.
Nat Commun ; 10(1): 4571, 2019 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-31594944

RESUMEN

5-Fluorouracil (5-FU) is a chemotherapeutic drug commonly used for the treatment of solid cancers. It is proposed that 5-FU interferes with nucleotide synthesis and incorporates into DNA, which may have a mutational impact on both surviving tumor and healthy cells. Here, we treat intestinal organoids with 5-FU and find a highly characteristic mutational pattern that is dominated by T>G substitutions in a CTT context. Tumor whole genome sequencing data confirms that this signature is also identified in vivo in colorectal and breast cancer patients who have received 5-FU treatment. Taken together, our results demonstrate that 5-FU is mutagenic and may drive tumor evolution and increase the risk of secondary malignancies. Furthermore, the identified signature shows a strong resemblance to COSMIC signature 17, the hallmark signature of treatment-naive esophageal and gastric tumors, which indicates that distinct endogenous and exogenous triggers can converge onto highly similar mutational signatures.


Asunto(s)
Carcinogénesis/efectos de los fármacos , Fluorouracilo/efectos adversos , Neoplasias/genética , Mutación Puntual/efectos de los fármacos , Transcriptoma/efectos de los fármacos , Adulto , Edad de Inicio , Anciano , Biopsia , Carcinogénesis/genética , Técnicas de Cultivo de Célula , Línea Celular , Ensayos Clínicos como Asunto , Estudios de Cohortes , Análisis Mutacional de ADN , Femenino , Humanos , Intestinos/citología , Masculino , Persona de Mediana Edad , Modelos Genéticos , Tasa de Mutación , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Organoides , Polimorfismo de Nucleótido Simple/efectos de los fármacos , Células Madre , Transcriptoma/genética , Secuenciación Completa del Genoma , Adulto Joven
13.
Sci Adv ; 5(5): eaaw1271, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31149636

RESUMEN

A developing human fetus needs to balance rapid cellular expansion with maintaining genomic stability. Here, we accurately quantified and characterized somatic mutation accumulation in fetal tissues by analyzing individual stem cells from human fetal liver and intestine. Fetal mutation rates were about fivefold higher than in tissue-matched adult stem cells. The mutational landscape of fetal intestinal stem cells resembled that of adult intestinal stem cells, while the mutation spectrum of fetal liver stem cells is distinct from stem cells of the fetal intestine and the adult liver. Our analyses indicate that variation in mutational mechanisms, including oxidative stress and spontaneous deamination of methylated cytosines, contributes to the observed divergence in mutation accumulation patterns and drives genetic mosaicism in humans.


Asunto(s)
Feto/fisiología , Mutación , Células Madre Adultas/fisiología , Feto/citología , Humanos , Intestinos/citología , Intestinos/embriología , Hígado/citología , Hígado/embriología , Tasa de Mutación , Especificidad de Órganos , Piel/citología , Piel/embriología
14.
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
15.
Nat Commun ; 9(1): 1873, 2018 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-29760424

RESUMEN

In contrast to mouse, human female germ cells develop asynchronously. Germ cells transition to meiosis, erase genomic imprints, and reactivate the X chromosome. It is unknown if these events all appear asynchronously, and how they relate to each other. Here we combine exome sequencing of human fetal and maternal tissues with single-cell RNA-sequencing of five donors. We reconstruct full parental haplotypes and quantify changes in parental allele-specific expression, genome-wide. First we distinguish primordial germ cells (PGC), pre-meiotic, and meiotic transcriptional stages. Next we demonstrate that germ cells from various stages monoallelically express imprinted genes and confirm this by methylation patterns. Finally, we show that roughly 30% of the PGCs are still reactivating their inactive X chromosome and that this is related to transcriptional stage rather than fetal age. Altogether, we uncover the complexity and cell-to-cell heterogeneity of transcriptional and epigenetic remodeling in female human germ cells.


Asunto(s)
Cromosomas Humanos X/química , Epigénesis Genética , Óvulo/metabolismo , Transcriptoma , Aborto Legal , Adulto , Cromosomas Humanos X/metabolismo , Metilación de ADN , Femenino , Feto , Heterogeneidad Genética , Impresión Genómica , Haplotipos , Humanos , Masculino , Meiosis , Óvulo/crecimiento & desarrollo , Embarazo , Trimestres del Embarazo , Análisis de la Célula Individual/métodos , Secuenciación del Exoma , Inactivación del Cromosoma X
16.
Genome Med ; 9(1): 9, 2017 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-28126037

RESUMEN

BACKGROUND: Germline chromothripsis causes complex genomic rearrangements that are likely to affect multiple genes and their regulatory contexts. The contribution of individual rearrangements and affected genes to the phenotypes of patients with complex germline genomic rearrangements is generally unknown. METHODS: To dissect the impact of germline chromothripsis in a relevant developmental context, we performed trio-based RNA expression analysis on blood cells, induced pluripotent stem cells (iPSCs), and iPSC-derived neuronal cells from a patient with de novo germline chromothripsis and both healthy parents. In addition, Hi-C and 4C-seq experiments were performed to determine the effects of the genomic rearrangements on transcription regulation of genes in the proximity of the breakpoint junctions. RESULTS: Sixty-seven genes are located within 1 Mb of the complex chromothripsis rearrangements involving 17 breakpoints on four chromosomes. We find that three of these genes (FOXP1, DPYD, and TWIST1) are both associated with developmental disorders and differentially expressed in the patient. Interestingly, the effect on TWIST1 expression was exclusively detectable in the patient's iPSC-derived neuronal cells, stressing the need for studying developmental disorders in the biologically relevant context. Chromosome conformation capture analyses show that TWIST1 lost genomic interactions with several enhancers due to the chromothripsis event, which likely led to deregulation of TWIST1 expression and contributed to the patient's craniosynostosis phenotype. CONCLUSIONS: We demonstrate that a combination of patient-derived iPSC differentiation and trio-based molecular profiling is a powerful approach to improve the interpretation of pathogenic complex genomic rearrangements. Here we have applied this approach to identify misexpression of TWIST1, FOXP1, and DPYD as key contributors to the complex congenital phenotype resulting from germline chromothripsis rearrangements.


Asunto(s)
Cromotripsis , Mutación de Línea Germinal , Transcriptoma , Dihidrouracilo Deshidrogenasa (NADP)/genética , Factores de Transcripción Forkhead/genética , Regulación de la Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Leucocitos/metabolismo , Neuronas/metabolismo , Proteínas Nucleares/genética , Proteínas Represoras/genética , Proteína 1 Relacionada con Twist/genética
17.
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
18.
Sci Rep ; 6: 22154, 2016 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-26915950

RESUMEN

The rat is an important model for liver regeneration. However, there is no in vitro culture system that can capture the massive proliferation that can be observed after partial hepatectomy in rats. We here describe the generation of rat liver stem cell lines. Rat liver stem cells, which grow as cystic organoids, were characterized by high expression of the stem cell marker Lgr5, by the expression of liver progenitor and duct markers, and by low expression of hepatocyte markers, oval cell markers, and stellate cell markers. Prolonged cultures of rat liver organoids depended on high levels of WNT-signalling and the inhibition of BMP-signaling. Upon transplantation of clonal lines to a Fah(-/-) Il2rg(-/-) rat model of liver failure, the rat liver stem cells engrafted into the host liver where they differentiated into areas with FAH and Albumin positive hepatocytes. Rat liver stem cell lines hold potential as consistent reliable cell sources for pharmacological, toxicological or metabolic studies. In addition, rat liver stem cell lines may contribute to the development of regenerative medicine in liver disease. To our knowledge, the here described liver stem cell lines represent the first organoid culture system in the rat.


Asunto(s)
Hepatocitos/metabolismo , Hepatocitos/trasplante , Fallo Hepático/terapia , Regeneración Hepática/fisiología , Trasplante de Células Madre , Células Madre/citología , Animales , Biomarcadores/metabolismo , Proteínas Portadoras/farmacología , Técnicas de Cultivo de Célula , Diferenciación Celular/fisiología , Proliferación Celular , Células Cultivadas , Hidrolasas/deficiencia , Hidrolasas/genética , Subunidad gamma Común de Receptores de Interleucina/deficiencia , Subunidad gamma Común de Receptores de Interleucina/genética , Hígado/citología , Fallo Hepático/patología , Trasplante de Hígado , Ratas , Ratas Transgénicas , Proteína Wnt3A/farmacología
19.
EMBO Rep ; 16(7): 791-802, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26077710

RESUMEN

Embryonic stem cell (ESC) cultures display a heterogeneous gene expression profile, ranging from a pristine naïve pluripotent state to a primed epiblast state. Addition of inhibitors of GSK3ß and MEK (so-called 2i conditions) pushes ESC cultures toward a more homogeneous naïve pluripotent state, but the molecular underpinnings of this naïve transition are not completely understood. Here, we demonstrate that DAZL, an RNA-binding protein known to play a key role in germ-cell development, marks a subpopulation of ESCs that is actively transitioning toward naïve pluripotency. Moreover, DAZL plays an essential role in the active reprogramming of cytosine methylation. We demonstrate that DAZL associates with mRNA of Tet1, a catalyst of 5-hydroxylation of methyl-cytosine, and enhances Tet1 mRNA translation. Overexpression of DAZL in heterogeneous ESC cultures results in elevated TET1 protein levels as well as increased global hydroxymethylation. Conversely, null mutation of Dazl severely stunts 2i-mediated TET1 induction and hydroxymethylation. Our results provide insight into the regulation of the acquisition of naïve pluripotency and demonstrate that DAZL enhances TET1-mediated cytosine hydroxymethylation in ESCs that are actively reprogramming to a pluripotent ground state.


Asunto(s)
Proteínas de Unión al ADN/genética , Regulación del Desarrollo de la Expresión Génica , Células Madre Embrionarias de Ratones/fisiología , Células Madre Pluripotentes/fisiología , Proteínas Proto-Oncogénicas/genética , Proteínas de Unión al ARN/metabolismo , Animales , Diferenciación Celular , Reprogramación Celular , Citosina/metabolismo , Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Estratos Germinativos/fisiología , Ratones , Biosíntesis de Proteínas , Proteínas Proto-Oncogénicas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/genética , Transcriptoma
20.
PLoS One ; 10(3): e0122066, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25811670

RESUMEN

Tumorigenesis is often associated with loss of tumor suppressor genes (such as TP53), genomic instability and telomere lengthening. Previously, we generated and characterized a rat p53 knockout model in which the homozygous rats predominantly develop hemangiosarcomas whereas the heterozygous rats mainly develop osteosarcomas. Using genome-wide analyses, we find that the tumors that arise in the heterozygous and homozygous Tp53C273X mutant animals are also different in their genomic instability profiles. While p53 was fully inactivated in both heterozygous and homozygous knockout rats, tumors from homozygous animals show very limited aneuploidy and low degrees of somatic copy number variation as compared to the tumors from heterozygous animals. In addition, complex structural rearrangements such as chromothripsis and breakage-fusion-bridge cycles were never found in tumors from homozygous animals, while these were readily detectable in tumors from heterozygous animals. Finally, we measured telomere length and telomere lengthening pathway activity and found that tumors of homozygous animals have longer telomeres but do not show clear telomerase or alternative lengthening of telomeres (ALT) activity differences as compared to the tumors from heterozygous animals. Taken together, our results demonstrate that host p53 status in this rat p53 knockout model has a large effect on both tumor type and genomic instability characteristics, where full loss of functional p53 is not the main driver of large-scale structural variations. Our results also suggest that chromothripsis primarily occurs under p53 heterozygous rather than p53 null conditions.


Asunto(s)
Inestabilidad Genómica , Neoplasias/genética , Proteína p53 Supresora de Tumor/deficiencia , Animales , Animales Modificados Genéticamente , Hibridación Genómica Comparativa , Variaciones en el Número de Copia de ADN , Modelos Animales de Enfermedad , Femenino , Técnicas de Silenciamiento del Gen , Heterocigoto , Homocigoto , Masculino , Mutación , Neoplasias/patología , Ratas , Telómero/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA