Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Exp Clin Cancer Res ; 42(1): 56, 2023 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-36869386

RESUMEN

BACKGROUND: Colorectal cancer (CRC) can be divided into four consensus molecular subtypes (CMS), each with distinct biological features. CMS4 is associated with epithelial-mesenchymal transition and stromal infiltration (Guinney et al., Nat Med 21:1350-6, 2015; Linnekamp et al., Cell Death Differ 25:616-33, 2018), whereas clinically it is characterized by lower responses to adjuvant therapy, higher incidence of metastatic spreading and hence dismal prognosis (Buikhuisen et al., Oncogenesis 9:66, 2020). METHODS: To understand the biology of the mesenchymal subtype and unveil specific vulnerabilities, a large CRISPR-Cas9 drop-out screen was performed on 14 subtyped CRC cell lines to uncover essential kinases in all CMSs. Dependency of CMS4 cells on p21-activated kinase 2 (PAK2) was validated in independent 2D and 3D in vitro cultures and in vivo models assessing primary and metastatic outgrowth in liver and peritoneum. TIRF microscopy was used to uncover actin cytoskeleton dynamics and focal adhesion localization upon PAK2 loss. Subsequent functional assays were performed to determine altered growth and invasion patterns. RESULTS: PAK2 was identified as a key kinase uniquely required for growth of the mesenchymal subtype CMS4, both in vitro and in vivo. PAK2 plays an important role in cellular attachment and cytoskeletal rearrangements (Coniglio et al., Mol Cell Biol 28:4162-72, 2008; Grebenova et al., Sci Rep 9:17171, 2019). In agreement, deletion or inhibition of PAK2 impaired actin cytoskeleton dynamics in CMS4 cells and, as a consequence, significantly reduced invasive capacity, while it was dispensable for CMS2 cells. Clinical relevance of these findings was supported by the observation that deletion of PAK2 from CMS4 cells prevented metastatic spreading in vivo. Moreover, growth in a model for peritoneal metastasis was hampered when CMS4 tumor cells were deficient for PAK2. CONCLUSION: Our data reveal a unique dependency of mesenchymal CRC and provide a rationale for PAK2 inhibition to target this aggressive subgroup of colorectal cancer.


Asunto(s)
Neoplasias Colorrectales , Sarcoma , Humanos , Citoesqueleto de Actina , Carcinogénesis , Línea Celular
2.
Cell Stem Cell ; 26(4): 569-578.e7, 2020 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-32169167

RESUMEN

Colorectal cancer stem cells (CSCs) express Lgr5 and display extensive stem cell-like multipotency and self-renewal and are thought to seed metastatic disease. Here, we used a mouse model of colorectal cancer (CRC) and human tumor xenografts to investigate the cell of origin of metastases. We found that most disseminated CRC cells in circulation were Lgr5- and formed distant metastases in which Lgr5+ CSCs appeared. This plasticity occurred independently of stemness-inducing microenvironmental factors and was indispensable for outgrowth, but not establishment, of metastases. Together, these findings show that most colorectal cancer metastases are seeded by Lgr5- cells, which display intrinsic capacity to become CSCs in a niche-independent manner and can restore epithelial hierarchies in metastatic tumors.


Asunto(s)
Neoplasias del Colon , Neoplasias Colorrectales , Biomarcadores de Tumor , Humanos , Células Madre Neoplásicas , Receptores Acoplados a Proteínas G
3.
Nat Protoc ; 13(2): 235-247, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29300390

RESUMEN

Most currently available colorectal cancer (CRC) mouse models are not suitable for studying progression toward the metastatic stage. Recently, establishment of tumor organoid lines, either from murine CRC models or patients, and the possibility of engineering them with genome-editing technologies, have provided a large collection of tumor material faithfully recapitulating phenotypic and genetic heterogeneity of native tumors. To study tumor progression in the natural in vivo environment, we developed an orthotopic approach based on transplantation of CRC organoids into the cecal epithelium. The 20-min procedure is described in detail here and enables growth of transplanted organoids into a single tumor mass within the intestinal tract. Due to long latency, tumor cells are capable of spreading through the blood circulation and forming metastases at distant sites. This method is designed to generate tumors suitable for studying CRC progression, thereby providing the opportunity to visualize tumor cell dynamics in vivo in real time by intravital microscopy.


Asunto(s)
Neoplasias Colorrectales/fisiopatología , Edición Génica/métodos , Organoides/trasplante , Animales , Sistemas CRISPR-Cas/genética , Transformación Celular Neoplásica , Neoplasias del Colon , Colonoscopía/métodos , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Hepáticas , Ratones , Ratones Endogámicos NOD , Organoides/fisiología , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Dev Biol ; 428(2): 328-337, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28473106

RESUMEN

Homeostasis of tissues is tightly regulated at the cellular, tissue and organismal level. Interestingly, tumor cells have found ways to hijack many of these physiological processes at all the different levels. Here we review how intravital microscopy techniques have provided new insights into our understanding of tissue homeostasis and cancer progression. In addition, we highlight the different strategies that tumor cells have adopted to use these physiological processes for their own benefit. We describe how visualization of these dynamic processes in living mice has broadened to our view on cancer initiation and progression.


Asunto(s)
Neoplasias/fisiopatología , Células Madre Adultas/patología , Células Madre Adultas/fisiología , Animales , Comunicación Celular/fisiología , Movimiento Celular/fisiología , Progresión de la Enfermedad , Humanos , Inflamación/patología , Inflamación/fisiopatología , Microscopía Intravital/métodos , Neoplasias/etiología , Neoplasias/patología , Células Madre Neoplásicas/patología , Células Madre Neoplásicas/fisiología , Neovascularización Patológica , Nicho de Células Madre/fisiología , Microambiente Tumoral/fisiología , Cicatrización de Heridas/fisiología
5.
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
6.
Curr Biol ; 26(4): 428-38, 2016 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-26853366

RESUMEN

Tumor-host interactions play an increasingly recognized role in modulating tumor growth. Thus, understanding the nature and impact of this complex bidirectional communication is key to identifying successful anti-cancer strategies. It has been proposed that tumor cells compete with and kill neighboring host tissue to clear space that they can expand into; however, this has not been demonstrated experimentally. Here we use the adult fly intestine to investigate the existence and characterize the role of competitive tumor-host interactions. We show that APC(-/-)-driven intestinal adenomas compete with and kill surrounding cells, causing host tissue attrition. Importantly, we demonstrate that preventing cell competition, by expressing apoptosis inhibitors, restores host tissue growth and contains adenoma expansion, indicating that cell competition is essential for tumor growth. We further show that JNK signaling is activated inside the tumor and in nearby tissue and is required for both tumor growth and cell competition. Lastly, we find that APC(-/-) cells display higher Yorkie (YAP) activity than host cells and that this promotes tumor growth, in part via cell competition. Crucially, we find that relative, rather than absolute, Hippo activity determines adenoma growth. Overall, our data indicate that the intrinsic over-proliferative capacity of APC(-/-) cells is not uncontrolled and can be constrained by host tissues if cell competition is inhibited, suggesting novel possible therapeutic approaches.


Asunto(s)
Adenoma/etiología , Carcinogénesis , Transformación Celular Neoplásica , Drosophila melanogaster/crecimiento & desarrollo , Neoplasias Intestinales/etiología , Adenoma/fisiopatología , Animales , Proliferación Celular , Modelos Animales de Enfermedad , Drosophila melanogaster/citología , Humanos , Neoplasias Intestinales/fisiopatología , Transducción de Señal
7.
Dev Cell ; 34(3): 297-309, 2015 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-26212135

RESUMEN

Throughout their lifetime, cells may suffer insults that reduce their fitness and disrupt their function, and it is unclear how these potentially harmful cells are managed in adult tissues. We address this question using the adult Drosophila posterior midgut as a model of homeostatic tissue and ribosomal Minute mutations to reduce fitness in groups of cells. We take a quantitative approach combining lineage tracing and biophysical modeling and address how cell competition affects stem cell and tissue population dynamics. We show that healthy cells induce clonal extinction in weak tissues, targeting both stem and differentiated cells for elimination. We also find that competition induces stem cell proliferation and self-renewal in healthy tissue, promoting selective advantage and tissue colonization. Finally, we show that winner cell proliferation is fueled by the JAK-STAT ligand Unpaired-3, produced by Minute(-/+) cells in response to chronic JNK stress signaling.


Asunto(s)
Células Madre Adultas/citología , Proteínas de Drosophila/metabolismo , Quinasas Janus/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Factores de Transcripción STAT/metabolismo , Factores de Transcripción/metabolismo , Animales , Diferenciación Celular/fisiología , Linaje de la Célula , Proliferación Celular , Drosophila melanogaster/citología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ribosomas/genética
8.
Dev Cell ; 23(4): 745-55, 2012 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-23079597

RESUMEN

Maintenance of chromosomal stability depends on error-free chromosome segregation. The pseudokinase BUBR1 is essential for this, because it is a core component of the mitotic checkpoint and is required for formation of stable kinetochore-microtubule attachments. We have identified a conserved and highly phosphorylated domain (KARD) in BUBR1 that is crucial for formation of kinetochore-microtubule attachments. Deletion of this domain or prevention of its phosphorylation abolishes formation of kinetochore microtubules, which can be reverted by inhibiting Aurora B activity. Phosphorylation of KARD by PLK1 promotes direct interaction of BUBR1 with the PP2A-B56α phosphatase that counters excessive Aurora B activity at kinetochores. As a result, removal of BUBR1 from mitotic cells or inhibition of PLK1 reduces PP2A-B56α kinetochore binding and elevates phosphorylation of Aurora B substrates on the outer kinetochore. We propose that PLK1 and BUBR1 cooperate to stabilize kinetochore-microtubule interactions by regulating PP2A-B56α-mediated dephosphorylation of Aurora B substrates at the kinetochore-microtubule interface.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Cinetocoros/metabolismo , Microtúbulos/metabolismo , Proteína Fosfatasa 2/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Aurora Quinasa B , Aurora Quinasas , Células HEK293 , Células HeLa , Humanos , Fosforilación , Células Tumorales Cultivadas , Quinasa Tipo Polo 1
9.
Dev Cell ; 22(6): 1321-9, 2012 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-22698286

RESUMEN

Chromosomal stability is safeguarded by a mitotic checkpoint, of which BUB1 and Mad3/BUBR1 are core components. These paralogs have similar, but not identical, domain organization. We show that Mad3/BUBR1 and BUB1 paralogous pairs arose by nine independent gene duplications throughout evolution, followed by parallel subfunctionalization in which preservation of the ancestral, amino-terminal KEN box or kinase domain was mutually exclusive. In one exception, vertebrate BUBR1-defined by the KEN box-preserved the kinase domain but allowed nonconserved degeneration of catalytic motifs. Although BUBR1 evolved to a typical pseudokinase in some vertebrates, it retained the catalytic triad in humans. However, we show that putative catalysis by human BUBR1 is dispensable for error-free chromosome segregation. Instead, residues that interact with ATP in conventional kinases are essential for conformational stability in BUBR1. We propose that parallel evolution of BUBR1 orthologs rendered its kinase function dispensable in vertebrates, producing an unusual, triad-containing pseudokinase.


Asunto(s)
Puntos de Control de la Fase M del Ciclo Celular , Proteínas Serina-Treonina Quinasas/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Evolución Biológica , Segregación Cromosómica , Duplicación de Gen , Humanos , Lagartos , Datos de Secuencia Molecular , Mutación , Conformación Proteica , Proteínas Serina-Treonina Quinasas/genética , Alineación de Secuencia , Proteínas de Pez Cebra/genética
10.
Cancer Res ; 70(12): 4891-900, 2010 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-20516114

RESUMEN

Genetic mutations in the mitotic regulatory kinase BUBR1 are associated with the cancer-susceptible disorder mosaic variegated aneuploidy (MVA). In patients with biallelic mutations, a missense mutation pairs with a truncating mutation. Here, we show that cell lines derived from MVA patients with biallelic mutations have an impaired mitotic checkpoint, chromosome alignment defects, and low overall BUBR1 abundance. Ectopic expression of BUBR1 restored mitotic checkpoint activity, proving that BUBR1 dysfunction causes chromosome segregation errors in the patients. Combined analysis of patient cells and functional protein replacement shows that all MVA mutations fall in two distinct classes: those that impose specific defects in checkpoint activity or microtubule attachment and those that lower BUBR1 protein abundance. Low protein abundance is the direct result of the absence of transcripts from truncating mutants combined with high protein turnover of missense mutants. In this group of missense mutants, the amino acid change consistently occurs in or near the BUBR1 kinase domain. Our findings provide a molecular explanation for chromosomal instability in patients with biallelic genetic mutations in BUBR1.


Asunto(s)
Anomalías Múltiples/genética , Aneuploidia , Segregación Cromosómica/genética , Predisposición Genética a la Enfermedad , Mutación/genética , Neoplasias/genética , Proteínas Serina-Treonina Quinasas/genética , Northern Blotting , Citometría de Flujo , Genes cdc/fisiología , Células HeLa , Humanos , Immunoblotting , Mosaicismo , Neoplasias/patología , Plásmidos , Proteínas Serina-Treonina Quinasas/metabolismo , Huso Acromático/patología , Síndrome , Transfección , Células Tumorales Cultivadas
11.
Biochim Biophys Acta ; 1786(1): 24-31, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18472014

RESUMEN

Aneuploidy, an abnormal number of chromosomes, is a trait shared by most solid tumors. Chromosomal instability (CIN) manifested as aneuploidy might promote tumorigenesis and cause increased resistance to anti-cancer therapies. The mitotic checkpoint or spindle assembly checkpoint is a major signaling pathway involved in the prevention of CIN. We review current knowledge on the contribution of misregulation of mitotic checkpoint proteins to tumor formation and will address to what extent this contribution is due to chromosome segregation errors directly. We propose that both checkpoint and non-checkpoint functions of these proteins contribute to the wide array of oncogenic phenotypes seen upon their misregulation.


Asunto(s)
Aneuploidia , Mitosis/fisiología , Neoplasias/etiología , Animales , Proteínas Cdc20 , Proteínas de Ciclo Celular/fisiología , Supervivencia Celular , Inestabilidad Cromosómica , Segregación Cromosómica/fisiología , Humanos , Proteínas Mad2 , Ratones , Modelos Animales , Proteínas Nucleares/fisiología , Prometafase/fisiología , Proteínas Serina-Treonina Quinasas , Proteínas de Saccharomyces cerevisiae/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA