Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 180(2): 233-247.e21, 2020 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-31978343

RESUMEN

Wnt dependency and Lgr5 expression define multiple mammalian epithelial stem cell types. Under defined growth factor conditions, such adult stem cells (ASCs) grow as 3D organoids that recapitulate essential features of the pertinent epithelium. Here, we establish long-term expanding venom gland organoids from several snake species. The newly assembled transcriptome of the Cape coral snake reveals that organoids express high levels of toxin transcripts. Single-cell RNA sequencing of both organoids and primary tissue identifies distinct venom-expressing cell types as well as proliferative cells expressing homologs of known mammalian stem cell markers. A hard-wired regional heterogeneity in the expression of individual venom components is maintained in organoid cultures. Harvested venom peptides reflect crude venom composition and display biological activity. This study extends organoid technology to reptilian tissues and describes an experimentally tractable model system representing the snake venom gland.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Organoides/crecimiento & desarrollo , Venenos de Serpiente/metabolismo , Células Madre Adultas/metabolismo , Animales , Serpientes de Coral/metabolismo , Perfilación de la Expresión Génica/métodos , Organoides/metabolismo , Glándulas Salivales/metabolismo , Venenos de Serpiente/genética , Serpientes/genética , Serpientes/crecimiento & desarrollo , Células Madre/metabolismo , Toxinas Biológicas/genética , Transcriptoma/genética
2.
Cell ; 165(7): 1564-1566, 2016 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-27315471

RESUMEN

The notion that the colon's deep crypt pockets provide a protected location that shields stem cells from potentially toxic substances is widely accepted. In this issue of Cell, Kaiko et al. reveal how a metabolite abundantly produced by the gut microbiota can inhibit stem cell proliferation but is blocked from doing so by crypt architecture.


Asunto(s)
Colon/metabolismo , Células Madre , Proliferación Celular , Mucosa Intestinal
3.
Nature ; 607(7919): 548-554, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35831497

RESUMEN

The morphology and functionality of the epithelial lining differ along the intestinal tract, but tissue renewal at all sites is driven by stem cells at the base of crypts1-3. Whether stem cell numbers and behaviour vary at different sites is unknown. Here we show using intravital microscopy that, despite similarities in the number and distribution of proliferative cells with an Lgr5 signature in mice, small intestinal crypts contain twice as many effective stem cells as large intestinal crypts. We find that, although passively displaced by a conveyor-belt-like upward movement, small intestinal cells positioned away from the crypt base can function as long-term effective stem cells owing to Wnt-dependent retrograde cellular movement. By contrast, the near absence of retrograde movement in the large intestine restricts cell repositioning, leading to a reduction in effective stem cell number. Moreover, after suppression of the retrograde movement in the small intestine, the number of effective stem cells is reduced, and the rate of monoclonal conversion of crypts is accelerated. Together, these results show that the number of effective stem cells is determined by active retrograde movement, revealing a new channel of stem cell regulation that can be experimentally and pharmacologically manipulated.


Asunto(s)
Recuento de Células , Movimiento Celular , Intestinos , Células Madre , Animales , Mucosa Intestinal/citología , Intestino Delgado/citología , Intestinos/citología , Ratones , Receptores Acoplados a Proteínas G , Células Madre/citología , Proteínas Wnt
4.
Genes Dev ; 33(13-14): 763-781, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31123064

RESUMEN

Coordinated induction, but also repression, of genes are key to normal differentiation. Although the role of lineage-specific transcription regulators has been studied extensively, their functional integration with chromatin remodelers, one of the key enzymatic machineries that control chromatin accessibility, remains ill-defined. Here we investigate the role of Mi-2ß, a SNF-2-like nucleosome remodeler and key component of the nucleosome remodeling and histone deacetylase (NuRD) complex in early B cells. Inactivation of Mi-2ß arrested differentiation at the large pre-B-cell stage and caused derepression of cell adhesion and cell migration signaling factors by increasing chromatin access at poised enhancers and chromosome architectural elements. Mi-2ß also supported IL-7R signaling, survival, and proliferation by repressing negative effectors of this pathway. Importantly, overexpression of Bcl2, a mitochondrial prosurvival gene and target of IL-7R signaling, partly rescued the differentiation block caused by Mi-2ß loss. Mi-2ß stably associated with chromatin sites that harbor binding motifs for IKAROS and EBF1 and physically associated with these transcription factors both on and off chromatin. Notably, Mi-2ß shared loss-of-function cellular and molecular phenotypes with IKAROS and EBF1, albeit in a distinct fashion. Thus, the nucleosome remodeler Mi-2ß promotes pre-B-cell differentiation by providing repression capabilities to distinct lineage-specific transcription factor-based regulatory networks.


Asunto(s)
Linfocitos B/citología , Diferenciación Celular/genética , Cromatina/metabolismo , ADN Helicasas/genética , ADN Helicasas/metabolismo , Regulación del Desarrollo de la Expresión Génica , Animales , Linaje de la Célula , Proliferación Celular/genética , Supervivencia Celular/genética , Células Cultivadas , Ratones , Factores de Transcripción
5.
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
6.
Cell ; 143(1): 134-44, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20887898

RESUMEN

Intestinal stem cells, characterized by high Lgr5 expression, reside between Paneth cells at the small intestinal crypt base and divide every day. We have carried out fate mapping of individual stem cells by generating a multicolor Cre-reporter. As a population, Lgr5(hi) stem cells persist life-long, yet crypts drift toward clonality within a period of 1-6 months. We have collected short- and long-term clonal tracing data of individual Lgr5(hi) cells. These reveal that most Lgr5(hi) cell divisions occur symmetrically and do not support a model in which two daughter cells resulting from an Lgr5(hi) cell division adopt divergent fates (i.e., one Lgr5(hi) cell and one transit-amplifying [TA] cell per division). The cellular dynamics are consistent with a model in which the resident stem cells double their numbers each day and stochastically adopt stem or TA fates. Quantitative analysis shows that stem cell turnover follows a pattern of neutral drift dynamics.


Asunto(s)
Linaje de la Célula , Intestino Delgado/citología , Células Madre/citología , Animales , Células Clonales , Ratones , Modelos Biológicos , Receptores Acoplados a Proteínas G/metabolismo
7.
Nature ; 543(7645): 424-427, 2017 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-28273069

RESUMEN

The small intestinal epithelium self-renews every four or five days. Intestinal stem cells (Lgr5+ crypt base columnar cells (CBCs)) sustain this renewal and reside between terminally differentiated Paneth cells at the bottom of the intestinal crypt. Whereas the signalling requirements for maintaining stem cell function and crypt homeostasis have been well studied, little is known about how metabolism contributes to epithelial homeostasis. Here we show that freshly isolated Lgr5+ CBCs and Paneth cells from the mouse small intestine display different metabolic programs. Compared to Paneth cells, Lgr5+ CBCs display high mitochondrial activity. Inhibition of mitochondrial activity in Lgr5+ CBCs or inhibition of glycolysis in Paneth cells strongly affects stem cell function, as indicated by impaired organoid formation. In addition, Paneth cells support stem cell function by providing lactate to sustain the enhanced mitochondrial oxidative phosphorylation in the Lgr5+ CBCs. Mechanistically, we show that oxidative phosphorylation stimulates p38 MAPK activation by mitochondrial reactive oxygen species signalling, thereby establishing the mature crypt phenotype. Together, our results reveal a critical role for the metabolic identity of Lgr5+ CBCs and Paneth cells in supporting optimal stem cell function, and we identify mitochondria and reactive oxygen species signalling as a driving force of cellular differentiation.


Asunto(s)
Autorrenovación de las Células , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Intestino Delgado/citología , Intestino Delgado/metabolismo , Células Madre/citología , Animales , Diferenciación Celular , Medios de Cultivo Condicionados/química , Medios de Cultivo Condicionados/farmacología , Glucólisis , Homeostasis , Ácido Láctico/metabolismo , Ratones , Mitocondrias/metabolismo , Organoides/citología , Organoides/efectos de los fármacos , Organoides/metabolismo , Fosforilación Oxidativa , Células de Paneth/citología , Células de Paneth/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Células Madre/fisiología , Proteína Wnt3A/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
8.
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
9.
Nature ; 507(7492): 362-365, 2014 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-24531760

RESUMEN

The rapid turnover of the mammalian intestinal epithelium is supported by stem cells located around the base of the crypt. In addition to the Lgr5 marker, intestinal stem cells have been associated with other markers that are expressed heterogeneously within the crypt base region. Previous quantitative clonal fate analyses have led to the proposal that homeostasis occurs as the consequence of neutral competition between dividing stem cells. However, the short-term behaviour of individual Lgr5(+) cells positioned at different locations within the crypt base compartment has not been resolved. Here we establish the short-term dynamics of intestinal stem cells using the novel approach of continuous intravital imaging of Lgr5- Confetti mice. We find that Lgr5(+) cells in the upper part of the niche (termed 'border cells') can be passively displaced into the transit-amplifying domain, after the division of proximate cells, implying that the determination of stem-cell fate can be uncoupled from division. Through quantitative analysis of individual clonal lineages, we show that stem cells at the crypt base, termed 'central cells', experience a survival advantage over border stem cells. However, through the transfer of stem cells between the border and central regions, all Lgr5(+) cells are endowed with long-term self-renewal potential. These findings establish a novel paradigm for stem-cell maintenance in which a dynamically heterogeneous cell population is able to function long term as a single stem-cell pool.


Asunto(s)
Homeostasis , Mucosa Intestinal/citología , Análisis de la Célula Individual , Células Madre/citología , Animales , División Celular , Linaje de la Célula , Supervivencia Celular , Células Clonales/citología , Femenino , Masculino , Ratones , Modelos Biológicos , Imagen Molecular , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo
10.
Nucleic Acids Res ; 46(13): 6435-6454, 2018 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-29955892

RESUMEN

Model systems with defined genetic modifications are powerful tools for basic research and translational disease modelling. Fortunately, generating state-of-the-art genetic model systems is becoming more accessible to non-geneticists due to advances in genome editing technologies. As a consequence, solely relying on (transient) overexpression of (mutant) effector proteins is no longer recommended since scientific standards increasingly demand genetic modification of endogenous loci. In this review, we provide up-to-date guidelines with respect to homology-directed repair (HDR)-mediated editing of mammalian model systems, aimed at assisting researchers in designing an efficient genome editing strategy.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica , Modelos Genéticos , Proteína 9 Asociada a CRISPR , Endodesoxirribonucleasas , Reacción en Cadena de la Polimerasa , Reparación del ADN por Recombinación
11.
Mol Syst Biol ; 14(6): e8227, 2018 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-29945941

RESUMEN

Intestinal organoids accurately recapitulate epithelial homeostasis in vivo, thereby representing a powerful in vitro system to investigate lineage specification and cellular differentiation. Here, we applied a multi-omics framework on stem cell-enriched and stem cell-depleted mouse intestinal organoids to obtain a holistic view of the molecular mechanisms that drive differential gene expression during adult intestinal stem cell differentiation. Our data revealed a global rewiring of the transcriptome and proteome between intestinal stem cells and enterocytes, with the majority of dynamic protein expression being transcription-driven. Integrating absolute mRNA and protein copy numbers revealed post-transcriptional regulation of gene expression. Probing the epigenetic landscape identified a large number of cell-type-specific regulatory elements, which revealed Hnf4g as a major driver of enterocyte differentiation. In summary, by applying an integrative systems biology approach, we uncovered multiple layers of gene expression regulation, which contribute to lineage specification and plasticity of the mouse small intestinal epithelium.


Asunto(s)
Biología Computacional , Intestinos/citología , Organogénesis , Organoides/citología , Animales , Regulación de la Expresión Génica , Ratones , Organogénesis/genética , Células Madre
12.
Gastroenterology ; 153(3): 674-677.e3, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28552620

RESUMEN

The intestinal epithelium is a repetitive sheet of crypt and villus units with stem cells at the bottom of the crypts. During postnatal development, crypts multiply via fission, generating 2 daughter crypts from 1 parental crypt. In the adult intestine, crypt fission is observed at a low frequency. Using intravital microscopy in Lgr5EGFP-Ires-CreERT2 mice, we monitored individual crypt dynamics over multiple days with single-cell resolution. We discovered the existence of crypt fusion, an almost exact reverse phenomenon of crypt fission, in which 2 crypts fuse into 1 daughter crypt. Examining 819 crypts in 4 mice, we found that 3.5% ± 0.6% of all crypts were in the process of fission, whereas 4.1 ± 0.9% of all crypts were undergoing crypt fusion. As counteracting processes, crypt fission and fusion could regulate crypt numbers during the lifetime of a mouse. Identifying the mechanisms that regulate rates of crypt fission and fusion could provide insights into intestinal adaptation to altered environmental conditions and disease pathogenesis.


Asunto(s)
Mucosa Intestinal/citología , Mucosa Intestinal/diagnóstico por imagen , Células Madre/citología , Células Madre/fisiología , Animales , Fusión Celular , Femenino , Homeostasis , Mucosa Intestinal/fisiología , Microscopía Intravital , Masculino , Ratones
13.
Nature ; 469(7330): 415-8, 2011 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-21113151

RESUMEN

Homeostasis of self-renewing small intestinal crypts results from neutral competition between Lgr5 stem cells, which are small cycling cells located at crypt bottoms. Lgr5 stem cells are interspersed between terminally differentiated Paneth cells that are known to produce bactericidal products such as lysozyme and cryptdins/defensins. Single Lgr5-expressing stem cells can be cultured to form long-lived, self-organizing crypt-villus organoids in the absence of non-epithelial niche cells. Here we find a close physical association of Lgr5 stem cells with Paneth cells in mice, both in vivo and in vitro. CD24(+) Paneth cells express EGF, TGF-α, Wnt3 and the Notch ligand Dll4, all essential signals for stem-cell maintenance in culture. Co-culturing of sorted stem cells with Paneth cells markedly improves organoid formation. This Paneth cell requirement can be substituted by a pulse of exogenous Wnt. Genetic removal of Paneth cells in vivo results in the concomitant loss of Lgr5 stem cells. In colon crypts, CD24(+) cells residing between Lgr5 stem cells may represent the Paneth cell equivalents. We conclude that Lgr5 stem cells compete for essential niche signals provided by a specialized daughter cell, the Paneth cell.


Asunto(s)
Intestinos/citología , Células Madre Multipotentes/citología , Células Madre Multipotentes/metabolismo , Células de Paneth/citología , Receptores Acoplados a Proteínas G/metabolismo , Nicho de Células Madre/citología , Animales , Antígeno CD24/metabolismo , Recuento de Células , Proliferación Celular , Técnicas de Cocultivo , Humanos , Ratones , Células de Paneth/metabolismo , Nicho de Células Madre/metabolismo , Proteínas Wnt/metabolismo , Proteína Wnt3
14.
EMBO Rep ; 15(1): 62-9, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24355609

RESUMEN

The concept of 'field cancerization' describes the clonal expansion of genetically altered, but morphologically normal cells that predisposes a tissue to cancer development. Here, we demonstrate that biased stem cell competition in the mouse small intestine can initiate the expansion of such clones. We quantitatively analyze how the activation of oncogenic K-ras in individual Lgr5(+) stem cells accelerates their cell division rate and creates a biased drift towards crypt clonality. K-ras mutant crypts then clonally expand within the epithelium through enhanced crypt fission, which distributes the existing Paneth cell niche over the two new crypts. Thus, an unequal competition between wild-type and mutant intestinal stem cells initiates a biased drift that leads to the clonal expansion of crypts carrying oncogenic mutations.


Asunto(s)
Células Madre Adultas/fisiología , Neoplasias Colorrectales/patología , Proteínas Proto-Oncogénicas p21(ras)/genética , Receptores Acoplados a Proteínas G/metabolismo , Animales , Ciclo Celular , Proliferación Celular , Transformación Celular Neoplásica , Neoplasias Colorrectales/genética , Mucosa Intestinal/patología , Intestino Delgado/patología , Ratones , Ratones Transgénicos , Mutación Missense , Oncogenes , Receptores Acoplados a Proteínas G/genética , Nicho de Células Madre
15.
Nature ; 459(7244): 262-5, 2009 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-19329995

RESUMEN

The intestinal epithelium is the most rapidly self-renewing tissue in adult mammals. We have recently demonstrated the presence of about six cycling Lgr5(+) stem cells at the bottoms of small-intestinal crypts. Here we describe the establishment of long-term culture conditions under which single crypts undergo multiple crypt fission events, while simultanously generating villus-like epithelial domains in which all differentiated cell types are present. Single sorted Lgr5(+) stem cells can also initiate these cryptvillus organoids. Tracing experiments indicate that the Lgr5(+) stem-cell hierarchy is maintained in organoids. We conclude that intestinal cryptvillus units are self-organizing structures, which can be built from a single stem cell in the absence of a non-epithelial cellular niche.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Intestinos/anatomía & histología , Intestinos/citología , Organoides/citología , Receptores Acoplados a Proteínas G/metabolismo , Células Madre/citología , Células Madre/metabolismo , Animales , Linaje de la Célula , Separación Celular , Regulación del Desarrollo de la Expresión Génica , Mucosa Intestinal/metabolismo , Mesodermo/citología , Mesodermo/metabolismo , Ratones , Células Madre Multipotentes/citología , Células Madre Multipotentes/metabolismo , Organoides/crecimiento & desarrollo , Organoides/metabolismo , Células de Paneth/metabolismo , Receptores Notch/metabolismo , Regeneración , Nicho de Células Madre
16.
Science ; 384(6697): 785-792, 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38753784

RESUMEN

In response to excessive DNA damage, human cells can activate p53 to induce apoptosis. Cells lacking p53 can still undergo apoptosis upon DNA damage, yet the responsible pathways are unknown. We observed that p53-independent apoptosis in response to DNA damage coincided with translation inhibition, which was characterized by ribosome stalling on rare leucine-encoding UUA codons and globally curtailed translation initiation. A genetic screen identified the transfer RNAse SLFN11 and the kinase GCN2 as factors required for UUA stalling and global translation inhibition, respectively. Stalled ribosomes activated a ribotoxic stress signal conveyed by the ribosome sensor ZAKα to the apoptosis machinery. These results provide an explanation for the frequent inactivation of SLFN11 in chemotherapy-unresponsive tumors and highlight ribosome stalling as a signaling event affecting cell fate in response to DNA damage.


Asunto(s)
Apoptosis , Daño del ADN , Biosíntesis de Proteínas , Ribosomas , Proteína p53 Supresora de Tumor , Humanos , Línea Celular Tumoral , Codón/genética , Leucina/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Ribosomas/metabolismo , Transducción de Señal , Proteína p53 Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/genética , Quinasas Quinasa Quinasa PAM/genética , Quinasas Quinasa Quinasa PAM/metabolismo
17.
EMBO Rep ; 12(2): 113-22, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21252944

RESUMEN

The maintenance of stem-cell-driven tissue homeostasis requires a balance between the generation and loss of cell mass. Adult stem cells have a close relationship with the surrounding tissue--known as their niche--and thus, stem-cell studies should preferably be performed in a physiological context, rather than outside their natural environment. The mouse is an attractive model in which to study adult mammalian stem cells, as numerous experimental systems and genetic tools are available. In this review, we describe strategies commonly used to identify and functionally characterize adult stem cells in mice and discuss their potential, limitations and interpretations, as well as how they have informed our understanding of adult stem-cell biology. An accurate interpretation of physiologically relevant stem-cell assays is crucial to identify adult stem cells and elucidate how they self-renew and give rise to differentiated progeny.


Asunto(s)
Células Madre Adultas/citología , Células Madre Adultas/fisiología , Animales , Técnicas de Cultivo de Célula , Diferenciación Celular , Linaje de la Célula , Humanos , Coloración y Etiquetado , Nicho de Células Madre/citología , Nicho de Células Madre/fisiología , Trasplante de Células Madre
18.
Cell Rep ; 42(3): 112252, 2023 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-36920903

RESUMEN

Oncogene-induced senescence is a phenomenon in which aberrant oncogene expression causes non-transformed cells to enter a non-proliferative state. Cells undergoing oncogenic induction display phenotypic heterogeneity, with some cells senescing and others remaining proliferative. The causes of heterogeneity remain unclear. We studied the sources of heterogeneity in the responses of human epithelial cells to oncogenic BRAFV600E expression. We found that a narrow expression range of BRAFV600E generated a wide range of activities of its downstream effector ERK. In population-level and single-cell assays, ERK activity displayed a non-monotonic relationship to proliferation, with intermediate ERK activities leading to maximal proliferation. We profiled gene expression across a range of ERK activities over time and characterized four distinct ERK response classes, which we propose act in concert to generate the ERK-proliferation response. Altogether, our studies map the input-output relationships between ERK activity and proliferation, elucidating how heterogeneity can be generated during oncogene induction.


Asunto(s)
Oncogenes , Proteínas Proto-Oncogénicas B-raf , Humanos , Línea Celular Tumoral , Mutación , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo
19.
Cell Rep ; 42(4): 112324, 2023 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-37000626

RESUMEN

Patient-derived organoids (PDOs) are widely heralded as a drug-screening platform to develop new anti-cancer therapies. Here, we use a drug-repurposing library to screen PDOs of colorectal cancer (CRC) to identify hidden vulnerabilities within therapy-induced phenotypes. Using a microscopy-based screen that accurately scores drug-induced cell killing, we have tested 414 putative anti-cancer drugs for their ability to switch the EGFRi/MEKi-induced cytostatic phenotype toward cytotoxicity. A majority of validated hits (9/37) are microtubule-targeting agents that are commonly used in clinical oncology, such as taxanes and vinca-alkaloids. One of these drugs, vinorelbine, is consistently effective across a panel of >25 different CRC PDOs, independent of RAS mutational status. Unlike vinorelbine alone, its combination with EGFR/MEK inhibition induces apoptosis at all stages of the cell cycle and shows tolerability and effective anti-tumor activity in vivo, setting the basis for a clinical trial to treat patients with metastatic RAS-mutant CRC.


Asunto(s)
Antineoplásicos , Neoplasias del Colon , Neoplasias Colorrectales , Humanos , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Vinorelbina/farmacología , Vinorelbina/uso terapéutico , Reposicionamiento de Medicamentos , Línea Celular Tumoral , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/genética , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Organoides/metabolismo
20.
Cancer Res ; 82(10): 1953-1968, 2022 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-35570706

RESUMEN

Micrometastases of colorectal cancer can remain dormant for years prior to the formation of actively growing, clinically detectable lesions (i.e., colonization). A better understanding of this step in the metastatic cascade could help improve metastasis prevention and treatment. Here we analyzed liver specimens of patients with colorectal cancer and monitored real-time metastasis formation in mouse livers using intravital microscopy to reveal that micrometastatic lesions are devoid of cancer stem cells (CSC). However, lesions that grow into overt metastases demonstrated appearance of de novo CSCs through cellular plasticity at a multicellular stage. Clonal outgrowth of patient-derived colorectal cancer organoids phenocopied the cellular and transcriptomic changes observed during in vivo metastasis formation. First, formation of mature CSCs occurred at a multicellular stage and promoted growth. Conversely, failure of immature CSCs to generate more differentiated cells arrested growth, implying that cellular heterogeneity is required for continuous growth. Second, early-stage YAP activity was required for the survival of organoid-forming cells. However, subsequent attenuation of early-stage YAP activity was essential to allow for the formation of cell type heterogeneity, while persistent YAP signaling locked micro-organoids in a cellularly homogenous and growth-stalled state. Analysis of metastasis formation in mouse livers using single-cell RNA sequencing confirmed the transient presence of early-stage YAP activity, followed by emergence of CSC and non-CSC phenotypes, irrespective of the initial phenotype of the metastatic cell of origin. Thus, establishment of cellular heterogeneity after an initial YAP-controlled outgrowth phase marks the transition to continuously growing macrometastases. SIGNIFICANCE: Characterization of the cell type dynamics, composition, and transcriptome of early colorectal cancer liver metastases reveals that failure to establish cellular heterogeneity through YAP-controlled epithelial self-organization prohibits the outgrowth of micrometastases. See related commentary by LeBleu, p. 1870.


Asunto(s)
Neoplasias Colorrectales , Neoplasias Hepáticas , Animales , Neoplasias Colorrectales/patología , Humanos , Neoplasias Hepáticas/metabolismo , Ratones , Micrometástasis de Neoplasia/patología , Células Madre Neoplásicas/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA