Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
1.
Nature ; 571(7765): 398-402, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31292548

RESUMEN

A decline in stem cell function impairs tissue regeneration during ageing, but the role of the stem-cell-supporting niche in ageing is not well understood. The small intestine is maintained by actively cycling intestinal stem cells that are regulated by the Paneth cell niche1,2. Here we show that the regenerative potential of human and mouse intestinal epithelium diminishes with age owing to defects in both stem cells and their niche. The functional decline was caused by a decrease in stemness-maintaining Wnt signalling due to production of Notum, an extracellular Wnt inhibitor, in aged Paneth cells. Mechanistically, high activity of mammalian target of rapamycin complex 1 (mTORC1) in aged Paneth cells inhibits activity of peroxisome proliferator activated receptor α (PPAR-α)3, and lowered PPAR-α activity increased Notum expression. Genetic targeting of Notum or Wnt supplementation restored function of aged intestinal organoids. Moreover, pharmacological inhibition of Notum in mice enhanced the regenerative capacity of aged stem cells and promoted recovery from chemotherapy-induced damage. Our results reveal a role of the stem cell niche in ageing and demonstrate that targeting of Notum can promote regeneration of aged tissues.


Asunto(s)
Envejecimiento , Senescencia Celular , Esterasas/metabolismo , Mucosa Intestinal/patología , Células de Paneth/metabolismo , Regeneración , Envejecimiento/fisiología , Animales , Senescencia Celular/fisiología , Esterasas/antagonistas & inhibidores , Esterasas/biosíntesis , Femenino , Humanos , Mucosa Intestinal/fisiología , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , PPAR alfa/metabolismo , Células de Paneth/patología , Receptores Acoplados a Proteínas G/metabolismo , Nicho de Células Madre , Células Madre/patología , Proteínas Wnt/antagonistas & inhibidores , Vía de Señalización Wnt
2.
Nature ; 568(7753): 551-556, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30971823

RESUMEN

Synthetic lethality-an interaction between two genetic events through which the co-occurrence of these two genetic events leads to cell death, but each event alone does not-can be exploited for cancer therapeutics1. DNA repair processes represent attractive synthetic lethal targets, because many cancers exhibit an impairment of a DNA repair pathway, which can lead to dependence on specific repair proteins2. The success of poly(ADP-ribose) polymerase 1 (PARP-1) inhibitors in cancers with deficiencies in homologous recombination highlights the potential of this approach3. Hypothesizing that other DNA repair defects would give rise to synthetic lethal relationships, we queried dependencies in cancers with microsatellite instability (MSI), which results from deficient DNA mismatch repair. Here we analysed data from large-scale silencing screens using CRISPR-Cas9-mediated knockout and RNA interference, and found that the RecQ DNA helicase WRN was selectively essential in MSI models in vitro and in vivo, yet dispensable in models of cancers that are microsatellite stable. Depletion of WRN induced double-stranded DNA breaks and promoted apoptosis and cell cycle arrest selectively in MSI models. MSI cancer models required the helicase activity of WRN, but not its exonuclease activity. These findings show that WRN is a synthetic lethal vulnerability and promising drug target for MSI cancers.


Asunto(s)
Inestabilidad de Microsatélites , Repeticiones de Microsatélite/genética , Neoplasias/genética , Mutaciones Letales Sintéticas/genética , Helicasa del Síndrome de Werner/genética , Apoptosis/genética , Sistemas CRISPR-Cas/genética , Puntos de Control del Ciclo Celular/genética , Línea Celular Tumoral , Roturas del ADN de Doble Cadena , Humanos , Modelos Genéticos , Neoplasias/patología , Interferencia de ARN , Proteína p53 Supresora de Tumor/metabolismo , Helicasa del Síndrome de Werner/deficiencia
3.
EMBO J ; 39(19): e104319, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32915464

RESUMEN

The aryl hydrocarbon receptor (AhR), a ligand-activated transcription factor that senses xenobiotics, diet, and gut microbial-derived metabolites, is increasingly recognized as a key regulator of intestinal biology. However, its effects on the function of colonic stem and progenitor cells remain largely unexplored. Here, we observed that inducible deletion of AhR in Lgr5+ stem cells increases the percentage of colonic stem cells and enhances organoid initiating capacity and growth of sorted stem and progenitor cells, while AhR activation has the opposite effect. Moreover, intestinal-specific AhR knockout increases basal stem cell and crypt injury-induced cell proliferation and promotes colon tumorigenesis in a preclinical colitis-associated tumor model by upregulating FoxM1 signaling. Mechanistically, AhR transcriptionally suppresses FoxM1 expression. Activation of AhR in human organoids recapitulates phenotypes observed in mice, such as reduction in the percentage of colonic stem cells, promotion of stem cell differentiation, and attenuation of FoxM1 signaling. These findings indicate that the AhR-FoxM1 axis, at least in part, mediates colonic stem/progenitor cell behavior.


Asunto(s)
Colon/metabolismo , Proteína Forkhead Box M1/metabolismo , Receptores de Hidrocarburo de Aril/deficiencia , Transducción de Señal , Células Madre/metabolismo , Animales , Femenino , Proteína Forkhead Box M1/genética , Técnicas de Inactivación de Genes , Humanos , Masculino , Ratones , Ratones Transgénicos , Receptores de Hidrocarburo de Aril/metabolismo
4.
FASEB J ; 37(6): e22975, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37159340

RESUMEN

Intestinal epithelial stem cells (ISCs) are responsible for intestinal epithelial barrier renewal; thereby, ISCs play a critical role in intestinal pathophysiology research. While transgenic ISC reporter mice are available, advanced translational studies lack a large animal model. This study validates ISC isolation in a new porcine Leucine Rich Repeat Containing G Protein-Coupled Receptor 5 (LGR5) reporter line and demonstrates the use of these pigs as a novel colorectal cancer (CRC) model. We applied histology, immunofluorescence, fluorescence-activated cell sorting, flow cytometry, gene expression quantification, and 3D organoid cultures to whole tissue and single cells from the duodenum, jejunum, ileum, and colon of LGR5-H2B-GFP and wild-type pigs. Ileum and colon LGR5-H2B-GFP, healthy human, and murine biopsies were compared by mRNA fluorescent in situ hybridization (FISH). To model CRC, adenomatous polyposis coli (APC) mutation was induced by CRISPR/Cas9 editing in porcine LGR5-H2B-GFP colonoids. Crypt-base, green fluorescent protein (GFP) expressing cells co-localized with ISC biomarkers. LGR5-H2B-GFPhi cells had significantly higher LGR5 expression (p < .01) and enteroid forming efficiency (p < .0001) compared with LGR5-H2B-GFPmed/lo/neg cells. Using FISH, similar LGR5, OLFM4, HOPX, LYZ, and SOX9 expression was identified between human and LGR5-H2B-GFP pig crypt-base cells. LGR5-H2B-GFP/APCnull colonoids had cystic growth in WNT/R-spondin-depleted media and significantly upregulated WNT/ß-catenin target gene expression (p < .05). LGR5+ ISCs are reproducibly isolated in LGR5-H2B-GFP pigs and used to model CRC in an organoid platform. The known anatomical and physiologic similarities between pig and human, and those shown by crypt-base FISH, underscore the significance of this novel LGR5-H2B-GFP pig to translational ISC research.


Asunto(s)
Intestinos , Humanos , Porcinos , Animales , Ratones , Hibridación Fluorescente in Situ , Células Madre , Íleon , Colon , Proteínas Fluorescentes Verdes/genética , Receptores Acoplados a Proteínas G/genética
5.
Nature ; 560(7717): E26, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29849139

RESUMEN

In Fig. 4e of this Article, the labels for 'Control' and 'HFD' were reversed ('Control' should have been labelled blue rather than purple, and 'HFD' should have been labelled purple rather than blue). Similarly, in Fig. 4f of this Article, the labels for 'V' and 'GW' were reversed ('V' should have been labelled blue rather than purple, and 'GW' should have been labelled purple instead of blue). The original figure has been corrected online.

6.
FASEB J ; 36(10): e22546, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36106538

RESUMEN

The tricarboxylic acid (TCA) cycle is the epicenter of cellular aerobic metabolism. TCA cycle intermediates facilitate energy production and provide anabolic precursors, but also function as intra- and extracellular metabolic signals regulating pleiotropic biological processes. Despite the importance of circulating TCA cycle metabolites as signaling molecules, the source of circulating TCA cycle intermediates remains uncertain. We observe that in mice, the concentration of TCA cycle intermediates in the portal blood exceeds that in tail blood indicating that the gut is a major contributor to circulating TCA cycle metabolites. With a focus on succinate as a representative of a TCA cycle intermediate with signaling activities and using a combination of gut microbiota depletion mouse models and isotopomer tracing, we demonstrate that intestinal microbiota is not a major contributor to circulating succinate. Moreover, we demonstrate that endogenous succinate production is markedly higher than intestinal succinate absorption in normal physiological conditions. Altogether, these results indicate that endogenous succinate production within the intestinal tissue is a major physiological source of circulating succinate. These results provide a foundation for an investigation into the role of the intestine in regulating circulating TCA cycle metabolites and their potential signaling effects on health and disease.


Asunto(s)
Microbioma Gastrointestinal , Ácido Succínico , Animales , Ciclo del Ácido Cítrico/fisiología , Microbioma Gastrointestinal/fisiología , Intestinos , Ratones , Succinatos/metabolismo , Ácido Succínico/metabolismo
7.
Nature ; 531(7592): 53-8, 2016 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-26935695

RESUMEN

Little is known about how pro-obesity diets regulate tissue stem and progenitor cell function. Here we show that high-fat diet (HFD)-induced obesity augments the numbers and function of Lgr5(+) intestinal stem cells of the mammalian intestine. Mechanistically, a HFD induces a robust peroxisome proliferator-activated receptor delta (PPAR-δ) signature in intestinal stem cells and progenitor cells (non-intestinal stem cells), and pharmacological activation of PPAR-δ recapitulates the effects of a HFD on these cells. Like a HFD, ex vivo treatment of intestinal organoid cultures with fatty acid constituents of the HFD enhances the self-renewal potential of these organoid bodies in a PPAR-δ-dependent manner. Notably, HFD- and agonist-activated PPAR-δ signalling endow organoid-initiating capacity to progenitors, and enforced PPAR-δ signalling permits these progenitors to form in vivo tumours after loss of the tumour suppressor Apc. These findings highlight how diet-modulated PPAR-δ activation alters not only the function of intestinal stem and progenitor cells, but also their capacity to initiate tumours.


Asunto(s)
Transformación Celular Neoplásica/efectos de los fármacos , Neoplasias del Colon/patología , Dieta Alta en Grasa/efectos adversos , Intestinos/patología , Células Madre/efectos de los fármacos , Células Madre/patología , Animales , Recuento de Células , Autorrenovación de las Células/efectos de los fármacos , Femenino , Genes APC , Humanos , Masculino , Ratones , Obesidad/inducido químicamente , Obesidad/patología , Organoides/efectos de los fármacos , Organoides/metabolismo , Organoides/patología , PPAR delta/metabolismo , Transducción de Señal/efectos de los fármacos , Nicho de Células Madre/efectos de los fármacos , Células Madre/metabolismo , beta Catenina/metabolismo
8.
Proc Natl Acad Sci U S A ; 116(28): 14039-14048, 2019 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-31239343

RESUMEN

Most normal and tumor cells are protected from tumor necrosis factor α (TNFα)-induced apoptosis. Here, we identify the MAP3 kinase tumor progression locus-2 (TPL2) as a player contributing to the protection of a subset of tumor cell lines. The combination of TPL2 knockdown and TNFα gives rise to a synthetic lethality phenotype via receptor-interacting serine/threonine-protein kinase 1 (RIPK1)-dependent and -independent mechanisms. Whereas wild-type TPL2 rescues the phenotype, its kinase-dead mutant does not. Comparison of the molecular events initiated by small interfering RNA for TPL2 (siTPL2) ± TNFα in treatment-sensitive and -resistant lines revealed that the activation of caspase-8, downstream of miR-21-5p and cFLIP, is the dominant TPL2-dependent event. More important, comparison of the gene expression profiles of all of the tested cell lines results in the clustering of sensitive and resistant lines into distinct groups, providing proof of principle for the feasibility of generating a predictive tool for treatment sensitivity.


Asunto(s)
Carcinoma/genética , Inhibidores de Caspasas/farmacología , Quinasas Quinasa Quinasa PAM/genética , Proteínas Proto-Oncogénicas/genética , Factor de Necrosis Tumoral alfa/genética , Apoptosis/genética , Carcinoma/tratamiento farmacológico , Carcinoma/patología , Caspasa 8/genética , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Macrófagos/metabolismo , MicroARNs/genética , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , ARN Interferente Pequeño/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/antagonistas & inhibidores , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Transducción de Señal , Mutaciones Letales Sintéticas/genética
9.
J Nutr ; 151(12): 3678-3688, 2021 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-34590119

RESUMEN

BACKGROUND: Obesity increases the colorectal cancer risk, in part by elevating colonic proinflammatory cytokines. Curcumin (CUR) and supplemental vitamin B-6 each suppress colonic inflammation. OBJECTIVES: We examined whether the combination of CUR and vitamin B-6 amplifies each supplement's effects and thereby suppress obesity-promoted tumorigenesis. METHODS: Male Friend Virus B (FVB) mice (4-week-old; n = 110) received 6 weekly injections of azoxymethane beginning 1 week after arrival. Thereafter, they were randomized to receive a low-fat diet (10% energy from fat), a high-fat diet (HFD; 60% energy from fat), a HFD containing 0.2% CUR, a HFD containing supplemental vitamin B-6 (24 mg pyridoxine HCl/kg), or a HFD containing both CUR and supplemental vitamin B-6 (C + B) for 15 weeks. Colonic inflammation, assessed by fecal calprotectin, and tumor metrics were the primary endpoints. The anti-inflammatory efficacy of the combination was also determined in human colonic organoids. RESULTS: HFD-induced obesity produced a 2.6-fold increase in plasma IL-6 (P < 0.02), a 1.9-fold increase in fecal calprotectin (P < 0.05), and a 2.2-fold increase in tumor multiplicity (P < 0.05). Compared to the HFD group, the C + B combination, but not the individual agents, decreased fecal calprotectin (66%; P < 0.01) and reduced tumor multiplicity and the total tumor burden by 60%-80% (P < 0.03) in an additive fashion. The combination of C + B also significantly downregulated colonic phosphatidylinositol-4,5-bisphosphate 3-kinase, Wnt, and NF-κB signaling by 31%-47% (P < 0.05), effects largely absent with the single agents. Observations that may explain how the 2 agents work additively include a 2.8-fold increased colonic concentration of 3-hydroxyanthranillic acid (P < 0.05) and a 1.3-fold higher colonic concentration of the active coenzymatic form of vitamin B-6 (P < 0.05). In human colonic organoids, micromolar concentrations of CUR, vitamin B-6, and their combination suppressed secreted proinflammatory cytokines by 41%-93% (P < 0.03), demonstrating relevance to humans. CONCLUSIONS: In this mouse model, C + B is superior to either agent alone in preventing obesity-promoted colorectal carcinogenesis. Augmented suppression of procancerous signaling pathways may be the means by which this augmentation occurs.


Asunto(s)
Neoplasias Colorrectales , Curcumina , Animales , Masculino , Ratones , Carcinogénesis , Neoplasias Colorrectales/etiología , Neoplasias Colorrectales/prevención & control , Curcumina/farmacología , Dieta Alta en Grasa , Suplementos Dietéticos , Ratones Endogámicos C57BL , Obesidad/tratamiento farmacológico , Piridoxina , Vitamina B 6/farmacología , Vitaminas
10.
Gut ; 68(4): 684-692, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-29666172

RESUMEN

OBJECTIVE: Serrated colorectal cancer (CRC) accounts for approximately 25% of cases and includes tumours that are among the most treatment resistant and with worst outcomes. This CRC subtype is associated with activating mutations in the mitogen-activated kinase pathway gene, BRAF, and epigenetic modifications termed the CpG Island Methylator Phenotype, leading to epigenetic silencing of key tumour suppressor genes. It is still not clear which (epi-)genetic changes are most important in neoplastic progression and we begin to address this knowledge gap herein. DESIGN: We use organoid culture combined with CRISPR/Cas9 genome engineering to sequentially introduce genetic alterations associated with serrated CRC and which regulate the stem cell niche, senescence and DNA mismatch repair. RESULTS: Targeted biallelic gene alterations were verified by DNA sequencing. Organoid growth in the absence of niche factors was assessed, as well as analysis of downstream molecular pathway activity. Orthotopic engraftment of complex organoid lines, but not BrafV600E alone, quickly generated adenocarcinoma in vivo with serrated features consistent with human disease. Loss of the essential DNA mismatch repair enzyme, Mlh1, led to microsatellite instability. Sphingolipid metabolism genes are differentially regulated in both our mouse models of serrated CRC and human CRC, with key members of this pathway having prognostic significance in the human setting. CONCLUSION: We generate rapid, complex models of serrated CRC to determine the contribution of specific genetic alterations to carcinogenesis. Analysis of our models alongside patient data has led to the identification of a potential susceptibility for this tumour type.


Asunto(s)
Adenocarcinoma/genética , Adenocarcinoma/patología , Colon/patología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Organoides/patología , Proteínas Proto-Oncogénicas B-raf/genética , Adenocarcinoma/metabolismo , Alelos , Colon/metabolismo , Neoplasias Colorrectales/metabolismo , Islas de CpG/genética , Reparación de la Incompatibilidad de ADN , Análisis Mutacional de ADN , Progresión de la Enfermedad , Epigenómica , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , Humanos , Modelos Genéticos , Mutación , Organoides/metabolismo , Fenotipo , Proteínas Proto-Oncogénicas B-raf/metabolismo
11.
Am J Physiol Cell Physiol ; 317(3): C405-C419, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31216420

RESUMEN

In vitro, cell cultures are essential tools in the study of intestinal function and disease. For the past few decades, monolayer cellular cultures, such as cancer cell lines or immortalized cell lines, have been widely applied in gastrointestinal research. Recently, the development of three-dimensional cultures known as organoids has permitted the growth of normal crypt-villus units that recapitulate many aspects of intestinal physiology. Organoid culturing has also been applied to study gastrointestinal diseases, intestinal-microbe interactions, and colorectal cancer. These models are amenable to CRISPR gene editing and drug treatments, including high-throughput small-molecule testing. Three-dimensional intestinal cultures have been transplanted into mice to develop versatile in vivo models of intestinal disease, particularly cancer. Limitations of currently available organoid models include cost and challenges in modeling nonepithelial intestinal cells, such as immune cells and the microbiota. Here, we describe the development of organoid models of intestinal biology and the applications of organoids for study of the pathophysiology of intestinal diseases and cancer.


Asunto(s)
Enfermedades Gastrointestinales/patología , Tracto Gastrointestinal/patología , Tracto Gastrointestinal/fisiología , Organoides/patología , Organoides/fisiología , Animales , Células Cultivadas , Enfermedades Gastrointestinales/fisiopatología , Neoplasias Gastrointestinales/patología , Neoplasias Gastrointestinales/fisiopatología , Tracto Gastrointestinal/fisiopatología , Humanos , Mucosa Intestinal/patología , Mucosa Intestinal/fisiología , Mucosa Intestinal/fisiopatología , Organoides/fisiopatología
12.
Mater Today (Kidlington) ; 23: 16-25, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31057328

RESUMEN

Cellulose acetate (CA), viscose, or artificial silk are biocompatible human-benign derivatives of cellulose, one of the most abundant biopolymers on earth. While various optical materials have been developed from CA, optical CA nanomaterials are nonexistent. Here we report on the assembly of a new family of extremely bright fluorescent CA nanoparticles (CA-dots), which are fully suitable for in vivo imaging / targeting applications. CA-dots can encapsulate a variety of molecular fluorophores. Using various commercially available fluorophores, we demonstrate that the fluorescence of CA-dots can be tuned within the entire UV-VIS-NIR spectrum. We also demonstrate excellent specific targeting of tumors in vivo, when injected in blood in zebrafish (xenograft model of human cervical epithelial cancer), and unusually strong ex-vivo topical labeling of colon cancer in mice utilizing CA folate-functionalized nanoparticles.

13.
Int J Cancer ; 143(7): 1797-1805, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29696632

RESUMEN

Gut dysbiosis may play an etiological role in colorectal tumorigenesis. We previously observed that the abundance of Parabacteroides distasonis (Pd) in stool was inversely associated with intestinal tumor burden and IL-1ß concentrations in mice. Here, we assessed the anti-inflammatory capacity of Pd membrane fraction (PdMb) in colon cancer cell lines. In addition, we tested whether Pd could suppress colon tumorigenesis in mice. Six-week-old male A/J mice were fed a low-fat (LF) diet, high-fat (HF) diet or HF+ whole freeze-dried Pd (HF + Pd, 0.04% wt/wt) for 24 weeks. After 1 week on diet, mice received 4 weekly injections of azoxymethane. PdMb robustly suppressed the production of pro-inflammatory cytokines and lowered the abundance of MyD88 and pAkt (ser473) induced by E. coli lipopolysaccharide in colon cancer cell lines. Moreover, PdMb induced apoptosis in colon cancer cell lines and blocked TLR4 activation in a reporter line. Colon tumors were observed in 0% of LF (0 of 19), 25% of HF (5 of 20) and 0% of HF + Pd mice (0 of 20) (p = 0.005). The latter group also displayed a lower abundance of MyD88 and pAkt (ser473) in colonic mucosa than HF mice. Taken together, these data suggest that Pd has anti-inflammatory and anti-cancer properties that are likely mediated by the suppression of TLR4 and Akt signaling, as well as promotion of apoptosis. Further work is needed to confirm these findings in additional models and fully elaborate the mechanism of action.


Asunto(s)
Azoximetano/toxicidad , Bacteroidetes/fisiología , Neoplasias del Colon/prevención & control , Dieta Alta en Grasa/efectos adversos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Apoptosis , Carcinógenos/toxicidad , Proliferación Celular , Neoplasias del Colon/etiología , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Humanos , Masculino , Ratones , Ratones Endogámicos A , Células Tumorales Cultivadas
15.
Gastroenterology ; 161(2): 722-724, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33991578
17.
Gastroenterology ; 158(4): 1167-1168, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31953069
18.
Gastroenterology ; 159(2): 795-797, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32569769
20.
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda