Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Sci Adv ; 10(14): eadj7666, 2024 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-38569041

RESUMEN

Inflammation-associated fibroblasts (IAFs) are associated with progression and drug resistance of chronic inflammatory diseases such as inflammatory bowel disease (IBD), but their direct impact on epithelial cells is unknown. Here, we developed an in vitro model whereby human colon fibroblasts are induced by specific cytokines and recapitulate key features of IAFs in vivo. When cocultured with patient-derived colon organoids (colonoids), IAFs induced rapid colonoid expansion and barrier disruption due to swelling and rupture of individual epithelial cells. Colonoids cocultured with IAFs also show increased DNA damage, mitotic errors, and proliferation arrest. These IAF-induced epithelial defects are mediated by a paracrine pathway involving prostaglandin E2 and its receptor EP4, leading to protein kinase A -dependent activation of the cystic fibrosis transmembrane conductance regulator. EP4-specific chemical inhibitors effectively prevented IAF-induced colonoid swelling and restored normal proliferation and genome stability. These findings reveal a mechanism by which IAFs could promote and perpetuate IBD and suggest a therapeutic avenue to mitigate inflammation-associated epithelial injury.


Asunto(s)
Enfermedades Inflamatorias del Intestino , Prostaglandinas , Humanos , Epitelio/metabolismo , Inflamación , Enfermedades Inflamatorias del Intestino/etiología , Enfermedades Inflamatorias del Intestino/metabolismo , Fibroblastos/metabolismo
2.
bioRxiv ; 2023 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-37808771

RESUMEN

Inflammation-associated fibroblasts (IAFs) are associated with the progression and drug resistance of chronic inflammatory diseases such as inflammatory bowel disease (IBD), but their direct impact on epithelial function and architecture is unknown. In this study, we developed an in vitro model whereby human colon fibroblasts are induced to become IAFs by specific cytokines and recapitulate key features of IAFs in vivo. When co-cultured with patient-derived colon organoids (colonoids), IAFs induced rapid colonoid swelling and barrier disruption due to swelling and rupture of individual epithelial cells. Epithelial cells co-cultured with IAFs also exhibit increased DNA damage, mitotic errors, and proliferation arrest. These IAF-induced epithelial defects are mediated through a paracrine pathway involving prostaglandin E2 (PGE2) and the PGE2 receptor EP4, leading to PKA-dependent activation of the CFTR chloride channel. Importantly, EP4-specific chemical inhibitors effectively prevented colonoid swelling and restored normal proliferation and genome stability of IAF-exposed epithelial cells. These findings reveal a mechanism by which IAFs could promote and perpetuate IBD and suggest a potential treatment to mitigate inflammation-associated epithelial injury.

3.
bioRxiv ; 2023 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-36778374

RESUMEN

Epithelial metaplasia is a common adaptation to chronic inflammatory processes and can be associated with increased risk of dysplasia and cancer. The distal colon of patients with inflammatory bowel disease (IBD) commonly shows crypt architectural distortion and Paneth cell metaplasia (PCM), and IBD patients also carry increased risk of colitis-associated dysplasia and cancer (CAC). Loss of SATB2 expression (Special AT-rich binding 2 protein, a colon-restricted chromatin remodeler) has recently been shown to distinguish colitis-associated dysplasia and CAC from sporadic disease. Here we report non-diffuse heterogeneous patterns of SATB2 loss across non-dysplastic distal colon biopsies from IBD patients (n=20). This cohort was specifically curated to include biopsies with well-developed histologic features of villiform growth and PCM. Notably, CDX2 was strongly expressed and P53 showed a wild-type immunolabeling pattern across our non-dysplastic cohort, regardless of SATB2 immunolabeling pattern. Our findings fit with recent murine studies in which colon-specific Satb2 deletion resulted in histologic conversion of colonic mucosa to small intestinal-like mucosa, including emergence of villi and Paneth cells. Taken together, we show that SATB2 loss is associated with a pre-neoplastic metaplastic response to chronic injury in human IBD and chronic colitis, reframing PCM more broadly as small intestinal metaplasia. We propose that inflammation-associated SATB2 loss mediates a remodeled chromatin landscape permissive for dysplasia and CAC.

4.
Neuron ; 110(18): 2916-2928, 2022 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-35985323

RESUMEN

Neuroblastoma is a leading cause of cancer-related death in children. Accumulated data suggest that differentiation arrest of the neural-crest-derived sympathoadrenal lineage contributes to neuroblastoma formation. The developmental arrest of these cell types explains many biological features of the disease, including its cellular heterogeneity, mutational spectrum, spontaneous regression, and response to drugs that induce tumor cell differentiation. In this review, we provide evidence that supports the notion that arrested neural-crest-derived progenitor cells give rise to neuroblastoma and discuss how this concept could be exploited for clinical management of the disease.


Asunto(s)
Neuroblastoma , Diferenciación Celular , Niño , Humanos , Cresta Neural/metabolismo , Cresta Neural/patología , Neuroblastoma/metabolismo , Neuroblastoma/patología
5.
Neoplasia ; 23(5): 488-501, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33906087

RESUMEN

Although much is known about the gene mutations required to drive colorectal cancer (CRC) initiation, the tissue-specific selective microenvironments in which neoplasia arises remains less characterized. Here, we determined whether modulation of intestinal stem cell niche morphogens alone can exert a neoplasia-relevant selective pressure on normal colonic epithelium. Using adult stem cell-derived murine colonic epithelial organoids (colonoids), we employed a strategy of sustained withdrawal of epidermal growth factor (EGF) and epidermal growth factor receptor (EGFR) inhibition to select for and expand survivors. EGFR-signaling-independent (iEGFR) colonoids emerged over rounds of selection and expansion. Colonoids derived from a mouse model of chronic mucosal injury showed an enhanced ability to adapt to EGFR inhibition. Whole-exome and transcriptomic analyses of iEGFR colonoids demonstrated acquisition of deleterious mutations and altered expression of genes implicated in EGF signaling, pyroptosis, and CRC. iEGFR colonoids acquired dysplasia-associated cytomorphologic changes, an increased proliferative rate, and the ability to survive independently of other required niche factors. These changes were accompanied by emergence of aneuploidy and chromosomal instability; further, the observed mitotic segregation errors were significantly associated with loss of interkinetic nuclear migration, a fundamental and dynamic process underlying intestinal epithelial homeostasis. This study provides key evidence that chromosomal instability and other phenotypes associated with neoplasia can be induced ex vivo via adaptation to EGF withdrawal in normal and stably euploid colonic epithelium, without introducing cancer-associated driver mutations. In addition, prior mucosal injury accelerates this evolutionary process.


Asunto(s)
Inestabilidad Cromosómica , Colon/metabolismo , Mucosa Intestinal/metabolismo , Adaptación Biológica , Aneuploidia , Animales , Proliferación Celular , Células Cultivadas , Colon/patología , Receptores ErbB/genética , Receptores ErbB/metabolismo , Edición Génica , Regulación de la Expresión Génica , Genes APC , Humanos , Mucosa Intestinal/patología , Ratones , Mutación , Organoides , Técnicas de Cultivo de Tejidos
6.
Nat Commun ; 11(1): 913, 2020 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-32060267

RESUMEN

Aggressive cancers often have activating mutations in growth-controlling oncogenes and inactivating mutations in tumor-suppressor genes. In neuroblastoma, amplification of the MYCN oncogene and inactivation of the ATRX tumor-suppressor gene correlate with high-risk disease and poor prognosis. Here we show that ATRX mutations and MYCN amplification are mutually exclusive across all ages and stages in neuroblastoma. Using human cell lines and mouse models, we found that elevated MYCN expression and ATRX mutations are incompatible. Elevated MYCN levels promote metabolic reprogramming, mitochondrial dysfunction, reactive-oxygen species generation, and DNA-replicative stress. The combination of replicative stress caused by defects in the ATRX-histone chaperone complex, and that induced by MYCN-mediated metabolic reprogramming, leads to synthetic lethality. Therefore, ATRX and MYCN represent an unusual example, where inactivation of a tumor-suppressor gene and activation of an oncogene are incompatible. This synthetic lethality may eventually be exploited to improve outcomes for patients with high-risk neuroblastoma.


Asunto(s)
Proteína Proto-Oncogénica N-Myc/genética , Neuroblastoma/metabolismo , Proteína Nuclear Ligada al Cromosoma X/genética , Animales , Preescolar , Estudios de Cohortes , Femenino , Amplificación de Genes , Humanos , Lactante , Masculino , Ratones , Mitocondrias/genética , Mitocondrias/metabolismo , Mutación , Proteína Proto-Oncogénica N-Myc/metabolismo , Neuroblastoma/genética , Especies Reactivas de Oxígeno/metabolismo , Proteína Nuclear Ligada al Cromosoma X/metabolismo
7.
Cancer Cell ; 36(5): 512-527.e9, 2019 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-31631027

RESUMEN

ATRX alterations occur at high frequency in neuroblastoma of adolescents and young adults. Particularly intriguing are the large N-terminal deletions of ATRX (Alpha Thalassemia/Mental Retardation, X-linked) that generate in-frame fusion (IFF) proteins devoid of key chromatin interaction domains, while retaining the SWI/SNF-like helicase region. We demonstrate that ATRX IFF proteins are redistributed from H3K9me3-enriched chromatin to promoters of active genes and identify REST as an ATRX IFF target whose activation promotes silencing of neuronal differentiation genes. We further show that ATRX IFF cells display sensitivity to EZH2 inhibitors, due to derepression of neurogenesis genes, including a subset of REST targets. Taken together, we demonstrate that ATRX structural alterations are not loss-of-function and put forward EZH2 inhibitors as a potential therapy for ATRX IFF neuroblastoma.


Asunto(s)
Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Regulación Neoplásica de la Expresión Génica , Neuroblastoma/tratamiento farmacológico , Proteínas Represoras/genética , Proteína Nuclear Ligada al Cromosoma X/genética , Animales , Secuencia de Bases/genética , Diferenciación Celular/genética , Línea Celular Tumoral , Cromatina/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Epigénesis Genética , Femenino , Histonas/metabolismo , Humanos , Masculino , Ratones , Neuroblastoma/genética , Neuroblastoma/patología , Neuroblastoma/cirugía , Neurogénesis/efectos de los fármacos , Neurogénesis/genética , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Regiones Promotoras Genéticas , Dominios Proteicos/genética , Eliminación de Secuencia , Proteína Nuclear Ligada al Cromosoma X/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Genetics ; 197(4): 1365-76, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24913681

RESUMEN

Tumorigenicity studies often employ outbred nude mice, in the absence of direct evidence that this mixed genetic background will negatively affect experimental outcome. Here we show that outbred nude mice carry two different alleles of Pla2g2a, a genetic modifier of intestinal tumorigenesis in mice. Here, we identify previous unreported linked polymorphisms in the promoter, noncoding and coding sequences of Pla2g2a and show that outbred nude mice from different commercial providers are heterogeneous for this polymorphic Pla2g2a allele. This heterogeneity even extends to mice obtained from a single commercial provider, which display mixed Pla2g2a genotypes. Notably, we demonstrated that the polymorphic Pla2g2a allele affects orthotopic xenograft establishment of human colon cancer cells in outbred nude mice. This finding establishes a non-cell-autonomous role for Pla2g2a in suppressing intestinal tumorigenesis. Using in vitro reporter assays and pharmacological inhibitors, we show promoter polymorphisms and nonsense-mediated RNA decay (NMD) as underlying mechanisms that lead to low Pla2g2a mRNA levels in tumor-sensitive mice. Together, this study provides mechanistic insight regarding Pla2g2a polymorphisms and demonstrates a non-cell-autonomous role for Pla2g2a in suppressing tumors. Moreover, our direct demonstration that mixed genetic backgrounds of outbred nude mice can significantly affect baseline tumorigenicity cautions against future use of outbred mice for tumor xenograft studies.


Asunto(s)
Carcinogénesis/genética , Fosfolipasas A2 Grupo II/genética , Polimorfismo Genético , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Alelos , Animales , Clonación Molecular , Genotipo , Fosfolipasas A2 Grupo II/metabolismo , Células HCT116 , Humanos , Intestinos/patología , Ratones , Ratones Desnudos , Degradación de ARNm Mediada por Codón sin Sentido , Plásmidos/genética , Regiones Promotoras Genéticas , ARN Mensajero/genética , ARN Mensajero/metabolismo
9.
Carcinogenesis ; 35(8): 1881-90, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24894865

RESUMEN

Mutation of tumor suppressor adenomatous polyposis coli (APC) initiates most colorectal cancers and chronic colitis increases risk. APC is a nucleo-cytoplasmic shuttling protein, best known for antagonizing Wnt signaling by forming a cytoplasmic complex that marks ß-catenin for degradation. Using our unique mouse model with compromised nuclear Apc import (Apc(mNLS)), we show that Apc(mNLS/mNLS) mice have increased susceptibility to tumorigenesis induced with azoxymethane (AOM) and dextran sodium sulfate (DSS). The AOM-DSS-induced colon adenoma histopathology, proliferation, apoptosis, stem cell number and ß-catenin and Kras mutation spectra were similar in Apc(mNLS/mNLS) and Apc(+/+) mice. However, AOM-DSS-treated Apc(mNLS/mNLS) mice showed more weight loss, more lymphoid follicles and edema, and increased colon shortening than treated Apc(+/+) mice, indicating a colitis predisposition. To test this directly, we induced acute colitis with a 7 day DSS treatment followed by 5 days of recovery. Compared with Apc(+/+) mice, DSS-treated Apc(mNLS/mNLS) mice developed more severe colitis based on clinical grade and histopathology. Apc(mNLS/mNLS) mice also had higher lymphocytic infiltration and reduced expression of stem cell markers, suggesting an increased propensity for chronic inflammation. Moreover, colons from DSS-treated Apc(mNLS/mNLS) mice showed fewer goblet cells and reduced Muc2 expression. Even in untreated Apc(mNLS/mNLS) mice, there were significantly fewer goblet cells in jejuna, and a modest decrease in colonocyte Muc2 expression compared with Apc(+/+) mice. Colonocytes from untreated Apc(mNLS/mNLS) mice also showed increased expression of inflammatory mediators cyclooxygenase-2 (Cox-2) and macrophage inflammatory protein-2 (MIP-2). These findings reveal novel functions for nuclear Apc in goblet cell differentiation and protection against inflammation-induced colon tumorigenesis.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Núcleo Celular/metabolismo , Transformación Celular Neoplásica/patología , Colitis/complicaciones , Neoplasias Colorrectales/patología , Modelos Animales de Enfermedad , Inflamación/patología , Proteína de la Poliposis Adenomatosa del Colon/genética , Animales , Apoptosis , Azoximetano/toxicidad , Western Blotting , Carcinógenos/toxicidad , Núcleo Celular/genética , Proliferación Celular , Transformación Celular Neoplásica/metabolismo , Colitis/inducido químicamente , Colitis/patología , Neoplasias Colorrectales/etiología , Neoplasias Colorrectales/metabolismo , Ciclooxigenasa 2 , Sulfato de Dextran/toxicidad , Inflamación/etiología , Inflamación/metabolismo , Ratones , Mutación/genética , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , beta Catenina/genética , beta Catenina/metabolismo
10.
Cancer Res ; 73(8): 2389-99, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23580574

RESUMEN

Adenomatous polyposis coli (APC) is best known for its crucial role in colorectal cancer suppression. Rodent models with various Apc mutations have enabled experimental validation of different Apc functions in tumors and normal tissues. Since the development of the first mouse model with a germline Apc mutation in the early 1990s, 20 other Apc mouse and rat models have been generated. This article compares and contrasts currently available Apc rodent models with particular emphasis on providing potential explanations for their reported variation in three areas: (i) intestinal polyp multiplicity, (ii) intestinal polyp distribution, and (iii) extraintestinal phenotypes.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/genética , Poliposis Adenomatosa del Colon/genética , Poliposis Adenomatosa del Colon/patología , Mutación de Línea Germinal , Fenotipo , Animales , Transformación Celular Neoplásica/genética , Modelos Animales de Enfermedad , Estudios de Asociación Genética , Humanos , Pólipos Intestinales/genética , Pólipos Intestinales/patología , Ratones , Ratas , Roedores
11.
Biochim Biophys Acta ; 1836(1): 80-9, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23333833

RESUMEN

Mutation of tumor suppressor gene adenomatous polyposis coli (APC) is an initiating step in most colon cancers. This review summarizes Apc models in mice and rats, with particular concentration on those most recently developed, phenotypic variation among different models, and genotype/phenotype correlations.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/fisiología , Poliposis Adenomatosa del Colon/etiología , Modelos Animales de Enfermedad , Poliposis Adenomatosa del Colon/patología , Animales , Genotipo , Humanos , Ratones , Fenotipo , Ratas
12.
Bio Protoc ; 2(22)2012 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-27148556

RESUMEN

The gastrointestinal (GI) tract is lined by a single layer of epithelial cells which function in secretion, absorption, and digestion. In addition, most GI tract tumors develop from epithelial cells (carcinomas). This protocol describes isolation of the surface epithelium from the underlying stroma, muscular layer and submucosa in the GI tract. In this protocol, epithelial cell adhesions are weekend by chelating Ca +2 ions followed by mechanical separation of the cells by vortexing. Analysis of protein levels and gene expression patterns in isolated epithelial cells versus whole GI tissue minimizes the potential for confounding contributions from contaminating stromal cells.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA