Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Cell Biosci ; 14(1): 38, 2024 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-38521952

RESUMEN

BACKGROUND: Bacterial cancer therapy was first trialled in patients at the end of the nineteenth century. More recently, tumour-targeting bacteria have been harnessed to deliver plasmid-expressed therapeutic interfering RNA to a range of solid tumours. A major limitation to clinical translation of this is the short-term nature of RNA interference in vivo due to plasmid instability. To overcome this, we sought to develop tumour-targeting attenuated bacteria that stably express shRNA by virtue of integration of an expression cassette within the bacterial chromosome and demonstrate therapeutic efficacy in vitro and in vivo. RESULTS: The attenuated tumour targeting Salmonella typhimurium SL7207 strain was modified to carry chromosomally integrated shRNA expression cassettes at the xylA locus. The colorectal cancer cell lines SW480, HCT116 and breast cancer cell line MCF7 were used to demonstrate the ability of these modified strains to perform intracellular infection and deliver effective RNA and protein knockdown of the target gene c-Myc. In vivo therapeutic efficacy was demonstrated using the Lgr5creERT2Apcflx/flx and BlgCreBrca2flx/flp53flx/flx orthotopic immunocompetent mouse models of colorectal and breast cancer, respectively. In vitro co-cultures of breast and colorectal cancer cell lines with modified SL7207 demonstrated a significant 50-95% (P < 0.01) reduction in RNA and protein expression with SL7207/c-Myc targeted strains. In vivo, following establishment of tumour tissue, a single intra-peritoneal administration of 1 × 106 CFU of SL7207/c-Myc was sufficient to permit tumour colonisation and significantly extend survival with no overt toxicity in control animals. CONCLUSIONS: In summary we have demonstrated that tumour tropic bacteria can be modified to safely deliver therapeutic levels of gene knockdown. This technology has the potential to specifically target primary and secondary solid tumours with personalised therapeutic payloads, providing new multi-cancer detection and treatment options with minimal off-target effects. Further understanding of the tropism mechanisms and impact on host immunity and microbiome is required to progress to clinical translation.

2.
Mol Nutr Food Res ; 66(19): e2200234, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36045438

RESUMEN

SCOPE: Black raspberries (BRBs) have colorectal cancer (CRC) chemo-preventative effects. As CRC originates from an intestinal stem cell (ISC) this study has investigated the impact of BRBs on normal and mutant ISCs. METHODS AND RESULTS: Mice with an inducible Apcfl mutation in either the ISC (Lgr5CreERT2 ) or intestinal crypt (AhCre/VillinCreERT2 ) are fed a control or 10% BRB-supplemented diet. This study uses immunohistochemistry, gene expression analysis, and organoid culture to evaluate the effect of BRBs on intestinal homeostasis. RNAscope is performed for ISC markers on CRC adjacent normal colonic tissue pre and post BRB intervention from patients. 10% BRB diet has no overt effect on murine intestinal homeostasis, despite a reduced stem cell number. Following Apc ISC deletion, BRB diet extends lifespan and reduces tumor area. In the AhCre model, BRB diet attenuates the "crypt-progenitor" phenotype and reduces ISC marker gene expression. In ex vivo culture BRBs reduce the self-renewal capacity of murine and human Apc deficient organoids. Finally, the study observes a reduction in ISC marker gene expression in adjacent normal crypts following introduction of BRBs to the human bowel. CONCLUSION: BRBs play a role in CRC chemoprevention by protectively regulating the ISC compartment and further supports the use of BRBs in CRC prevention.


Asunto(s)
Rubus , Animales , Colon/metabolismo , Dieta , Humanos , Mucosa Intestinal/metabolismo , Intestinos , Ratones , Células Madre
3.
Biosci Rep ; 41(7)2021 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-34236075

RESUMEN

Over the last few decades it has been established that the complex interaction between the host and the multitude of organisms that compose the intestinal microbiota plays an important role in human metabolic health and disease. Whilst there is no defined consensus on the composition of a healthy microbiome due to confounding factors such as ethnicity, geographical locations, age and sex, there are undoubtably populations of microbes that are consistently dysregulated in gut diseases including colorectal cancer (CRC). In this review, we discuss the most recent advances in the application of the gut microbiota, not just bacteria, and derived microbial compounds in the diagnosis of CRC and the potential to exploit microbes as novel agents in the management and treatment of CRC. We highlight examples of the microbiota, and their derivatives, that have the potential to become standalone diagnostic tools or be used in combination with current screening techniques to improve sensitivity and specificity for earlier CRC diagnoses and provide a perspective on their potential as biotherapeutics with translatability to clinical trials.


Asunto(s)
Antibacterianos/uso terapéutico , Antineoplásicos/uso terapéutico , Bacterias/efectos de los fármacos , Neoplasias Colorrectales/terapia , Trasplante de Microbiota Fecal , Microbioma Gastrointestinal/efectos de los fármacos , Probióticos/uso terapéutico , Animales , Antibacterianos/efectos adversos , Antineoplásicos/efectos adversos , Bacterias/crecimiento & desarrollo , Bacterias/metabolismo , Bacterias/patogenicidad , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/microbiología , Disbiosis , Trasplante de Microbiota Fecal/efectos adversos , Interacciones Huésped-Patógeno , Humanos , Probióticos/efectos adversos , Resultado del Tratamiento
4.
Biosci Rep ; 41(3)2021 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-33620068

RESUMEN

Inactivating mutations including both germline and somatic mutations in the adenomatous polyposis coli (APC) gene drives most familial and sporadic colorectal cancers. Understanding the metabolic implications of this mutation will aid to establish its wider impact on cellular behaviour and potentially inform clinical decisions. However, to date, alterations in lipid metabolism induced by APC mutations remain unclear. Intestinal organoids have gained widespread popularity in studying colorectal cancer and chemotherapies, because their 3D structure more accurately mimics an in vivo environment. Here, we aimed to investigate intra-cellular lipid disturbances induced by APC gene mutations in intestinal organoids using a reversed-phase ultra-high-performance liquid chromatography mass spectrometry (RP-UHPLC-MS)-based lipid profiling method. Lipids of the organoids grown from either wild-type (WT) or mice with APC mutations (Lgr5-EGFP-IRES-CreERT2Apcfl/fl) were extracted and analysed using RP-UHPLC-MS. Levels of phospholipids (e.g. PC(16:0/16:0), PC(18:1/20:0), PC(38:0), PC(18:1/22:1)), ceramides (e.g. Cer(d18:0/22:0), Cer(d42:0), Cer(d18:1/24:1)) and hexosylceramides (e.g. HexCer(d18:1/16:0), HexCer(d18:1/22:0)) were higher in Apcfl/fl organoids, whereas levels of sphingomyelins (e.g. SM(d18:1/14:0), SM(d18:1/16:0)) were lower compared with WT. These observations indicate that cellular metabolism of sphingomyelin was up-regulated, resulting in the cellular accumulation of ceramides and production of HexCer due to the absence of Apcfl/fl in the organoids. Our observations demonstrated lipid profiling of organoids and provided an enhanced insight into the effects of the APC mutations on lipid metabolism, making for a valuable addition to screening options of the organoid lipidome.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Metabolismo de los Lípidos , Organoides/metabolismo , Proteína de la Poliposis Adenomatosa del Colon/genética , Animales , Ceramidas/metabolismo , Neoplasias Colorrectales/genética , Ratones , Mutación , Esfingomielinas/metabolismo , Células Tumorales Cultivadas
5.
Trends Cancer ; 5(10): 577-579, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31706504

RESUMEN

Intestinal microbiota undergo significant changes in colorectal cancer (CRC). Zheng et al. (Nat. Biomed. Eng., 2019) observe detrimental overpopulation of Fusobacterium nucleatum in mice and patients, suppressing the beneficial butyrate-producing Clostridium butyricum. Phage-guided irinotecan-loaded dextran nanoparticles promote release of bacterial-derived butyrate, while F. nucleatum and CRC cells are eliminated. These findings describe a possible novel therapeutic strategy for CRC.


Asunto(s)
Bacteriófagos , Neoplasias Colorrectales , Microbioma Gastrointestinal , Animales , Bacterias , Fusobacterium nucleatum , Humanos , Ratones
6.
Mol Cancer Res ; 17(3): 686-696, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30552232

RESUMEN

Both alterations to the epigenome and loss of polarity have been linked to cancer initiation, progression, and metastasis. It has previously been demonstrated that loss of the epigenetic reader protein Kaiso suppresses intestinal tumorigenesis in the Apc+/min mouse model, in which altered polarity plays a key role. Thus, we investigated the link between Kaiso deficiency, polarity, and suppression of intestinal tumorigenesis. We used Kaiso-deficient mice to conditionally delete Apc within the intestinal epithelia and demonstrated upregulation of the spindle polarity genes Dlg1 and Dlgap1. To understand the role of Dlg1, we generated Villin-creApc+/minDlg1flx/flx Kaiso-/y mice to analyze gene expression, survival, tumor burden, and spindle orientation. In vivo analysis of the Dlg1-deficient intestine revealed improper orientation of mitotic spindles and a decreased rate of cellular migration. Loss of Dlg1 decreased survival in Apc+/min mice, validating its role as a tumor suppressor in the intestine. Significantly, the increased survival of Apc+/minKaisoy/- mice was shown to be dependent on Dlg1 expression. Taken together, these data indicate that maintenance of spindle polarity in the intestinal crypt requires appropriate regulation of Dlg1 expression. As Dlg1 loss leads to incorrect spindle orientation and a delay in cells transiting the intestinal crypt. We propose that the delayed exit from the crypt increase the window in which spontaneous mutations can become fixed, producing a "tumor-permissive" environment, without an increase in mutation rate. IMPLICATIONS: Loss of mitotic spindle polarity delays the exit of cells from the intestinal crypt and promotes a tumorigenic environment.


Asunto(s)
Homólogo 1 de la Proteína Discs Large/genética , Neoplasias Intestinales/genética , Huso Acromático/fisiología , Factores de Transcripción/genética , Animales , Carcinogénesis , Polaridad Celular/fisiología , Homólogo 1 de la Proteína Discs Large/metabolismo , Epigénesis Genética , Neoplasias Intestinales/metabolismo , Neoplasias Intestinales/patología , Masculino , Ratones , Huso Acromático/metabolismo , Factores de Transcripción/metabolismo
7.
Oncotarget ; 9(92): 36430-36443, 2018 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-30559928

RESUMEN

Leukocyte cell-derived chemotaxin 2 (Lect2) is a chemokine-like chemotactic factor that has been identified as a downstream target of the Wnt signalling pathway. Whilst the primary function of Lect2 is thought to be in modulating the inflammatory process, it has recently been implicated as a potential inhibitor of the Wnt pathway. Deregulation of the Wnt pathway, often due to loss of the negative regulator APC, is found in ~80% of colorectal cancer (CRC). Here we have used the ApcMin/+Lect2-/- mouse model to characterise the role of Lect2 in Wnt-driven intestinal tumourigenesis. Histopathological, immunohistochemical, PCR and flow cytometry analysis were employed to identify the role of Lect2 in the intestine. The ApcMin/+Lect2-/- mice had a reduced mean survival and a significantly increased number of adenomas in the small intestine with increased severity. Analysis of Lect2 loss indicated it had no effect on the Wnt pathway in the intestine but significant differences were observed in circulating inflammatory markers, CD4+ T cells, and T cell lineage-specification factors. In summary, in the murine intestine loss of Lect2 promotes the initiation and progression of Wnt-driven colorectal cancer. This protection is performed independently of the Wnt signalling pathway and is associated with an altered inflammatory environment during Wnt-driven tumorigenesis.

8.
J Pathol ; 245(3): 270-282, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29603746

RESUMEN

Epigenetic regulation plays a key role in the link between inflammation and cancer. Here we examine Mbd2, which mediates epigenetic transcriptional silencing by binding to methylated DNA. In separate studies the Mbd2-/- mouse has been shown (1) to be resistant to intestinal tumourigenesis and (2) to have an enhanced inflammatory/immune response, observations that are inconsistent with the links between inflammation and cancer. To clarify its role in tumourigenesis and inflammation, we used constitutive and conditional models of Mbd2 deletion to explore its epithelial and non-epithelial roles in the intestine. Using a conditional model, we found that suppression of intestinal tumourigenesis is due primarily to the absence of Mbd2 within the epithelia. Next, we demonstrated, using the DSS colitis model, that non-epithelial roles of Mbd2 are key in preventing the transition from acute to tumour-promoting chronic inflammation. Combining models revealed that prior to inflammation the altered Mbd2-/- immune response plays a role in intestinal tumour suppression. However, following inflammation the intestine converts from tumour suppressive to tumour promoting. To summarise, in the intestine the normal function of Mbd2 is exploited by cancer cells to enable tumourigenesis, while in the immune system it plays a key role in preventing tumour-enabling inflammation. Which role is dominant depends on the inflammation status of the intestine. As environmental interactions within the intestine can alter DNA methylation patterns, we propose that Mbd2 plays a key role in determining whether these interactions are anti- or pro-tumourigenic and this makes it a useful new epigenetic model for inflammation-associated carcinogenesis. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Colitis/metabolismo , Proteínas de Unión al ADN/metabolismo , Mucosa Intestinal/metabolismo , Neoplasias Intestinales/metabolismo , Animales , Transformación Celular Neoplásica/inducido químicamente , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Colitis/inducido químicamente , Colitis/genética , Colitis/patología , Metilación de ADN , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Sulfato de Dextran , Modelos Animales de Enfermedad , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Genes APC , Mucosa Intestinal/patología , Neoplasias Intestinales/inducido químicamente , Neoplasias Intestinales/genética , Neoplasias Intestinales/patología , Ratones Noqueados , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Transducción de Señal , Células TH1/metabolismo , Células TH1/patología , Células Th2/metabolismo , Células Th2/patología
9.
Cancer Immunol Res ; 5(11): 1005-1015, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28947544

RESUMEN

T-cell infiltration into tumors represents a critical bottleneck for immune-mediated control of cancer. We previously showed that this bottleneck can be overcome by depleting immunosuppressive Foxp3+ regulatory T cells (Tregs), a process that can increase frequencies of tumor-infiltrating lymphocytes through promoting the development of specialized portals for lymphocyte entry, namely high endothelial venules (HEVs). In this paper, we used a carcinogen-induced tumor model that allows for coevolution of the tumor microenvironment and the immune response to demonstrate that Treg depletion not only results in widespread disruption to HEV networks in lymph nodes (LNs) but also activates CD8+ T cells, which then drive intratumoral HEV development. Formation of these vessels contrasts with ontogenic HEV development in LNs in that the process is dependent on the TNF receptor and independent of lymphotoxin ß receptor-mediated signaling. These intratumoral HEVs do not express the chemokine CCL21, revealing a previously undescribed intratumoral blood vessel phenotype. We propose a model where Treg depletion enables a self-amplifying loop of T-cell activation, which promotes HEV development, T-cell infiltration, and ultimately, tumor destruction. The findings point to a need to test for HEV development as part of ongoing clinical studies in patients with cancer. Cancer Immunol Res; 5(11); 1005-15. ©2017 AACR.


Asunto(s)
Neoplasias/inmunología , Linfocitos T Reguladores/inmunología , Animales , Células Dendríticas/inmunología , Endotelio Vascular/inmunología , Depleción Linfocítica , Linfocitos Infiltrantes de Tumor/inmunología , Receptor beta de Linfotoxina/inmunología , Metilcolantreno , Ratones , Neoplasias/inducido químicamente , Receptores del Factor de Necrosis Tumoral/inmunología
10.
Mol Cancer ; 16(1): 84, 2017 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-28446200

RESUMEN

BACKGROUND: Cancer/testis (CT) genes have expression normally restricted to the testis, but become activated during oncogenesis, so they have excellent potential as cancer-specific biomarkers. Evidence is starting to emerge to indicate that they also provide function(s) in the oncogenic programme. Human TEX19 is a recently identified CT gene, but a functional role for TEX19 in cancer has not yet been defined. METHODS: siRNA was used to deplete TEX19 levels in various cancer cell lines. This was extended using shRNA to deplete TEX19 in vivo. Western blotting, fluorescence activated cell sorting and immunofluorescence were used to study the effect of TEX19 depletion in cancer cells and to localize TEX19 in normal testis and cancer cells/tissues. RT-qPCR and RNA sequencing were employed to determine the changes to the transcriptome of cancer cells depleted for TEX19 and Kaplan-Meier plots were generated to explore the relationship between TEX19 expression and prognosis for a range of cancer types. RESULTS: Depletion of TEX19 levels in a range of cancer cell lines in vitro and in vivo restricts cellular proliferation/self-renewal/reduces tumour volume, indicating TEX19 is required for cancer cell proliferative/self-renewal potential. Analysis of cells depleted for TEX19 indicates they enter a quiescent-like state and have subtle defects in S-phase progression. TEX19 is present in both the nucleus and cytoplasm in both cancerous cells and normal testis. In cancer cells, localization switches in a context-dependent fashion. Transcriptome analysis of TEX19 depleted cells reveals altered transcript levels of a number of cancer-/proliferation-associated genes, suggesting that TEX19 could control oncogenic proliferation via a transcript/transcription regulation pathway. Finally, overall survival analysis of high verses low TEX19 expressing tumours indicates that TEX19 expression is linked to prognostic outcomes in different tumour types. CONCLUSIONS: TEX19 is required to drive cell proliferation in a range of cancer cell types, possibly mediated via an oncogenic transcript regulation mechanism. TEX19 expression is linked to a poor prognosis for some cancers and collectively these findings indicate that not only can TEX19 expression serve as a novel cancer biomarker, but may also offer a cancer-specific therapeutic target with broad spectrum potential.


Asunto(s)
Biomarcadores de Tumor/genética , Células Germinativas/metabolismo , Neoplasias/genética , Proteínas Nucleares/genética , Testículo/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular/genética , Supervivencia sin Enfermedad , Regulación Neoplásica de la Expresión Génica/genética , Células Germinativas/patología , Humanos , Estimación de Kaplan-Meier , Masculino , Ratones , Neoplasias/patología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Pronóstico , Proteínas de Unión al ARN , Testículo/patología , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Emerg Top Life Sci ; 1(4): 385-400, 2017 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-33525777

RESUMEN

The human gut microbiome is considered an organ in its entirety and has been the subject of extensive research due to its role in physiology, metabolism, digestion, and immune regulation. Disequilibria of the normal microbiome have been associated with the development of several gastrointestinal diseases, but the exact underlying interactions are not well understood. Conventional in vivo and in vitro modelling systems fail to faithfully recapitulate the complexity of the human host-gut microbiome, emphasising the requirement for novel systems that provide a platform to study human host-gut microbiome interactions with a more holistic representation of the human in vivo microenvironment. In this review, we outline the progression and applications of new and old modelling systems with particular focus on their ability to model and to study host-microbiome cross-talk.

12.
Curr Pathobiol Rep ; 4(4): 209-219, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27882268

RESUMEN

PURPOSE OF REVIEW: Long-term culture of adult progenitor cells in 3D is a recently emerging technology that inhabits the space between 2D cell lines and organ slice culture. RECENT FINDINGS: Adaptations to defined media components in the wake of advances in ES and iPS cell culture has led to the identification of conditions that maintained intestinal cell progenitors in culture. These conditions retain cellular heterogeneity of the normal or tumour tissue, and the cultures have been shown to be genetically stable, such that substantial biobanks are being created from patient derived material. This coupled with advances in analytical tools has generated a field, characterized by the term "organoid culture", that has huge potential for advancing drug discovery, regenerative medicine, and furthering the understanding of fundamental intestinal biology. SUMMARY: In this review, we describe the approaches available for the long-term culture of intestinal cells from normal and diseased tissue, the current challenges, and how the technology is likely to develop further.

13.
BMC Cancer ; 16: 399, 2016 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-27388476

RESUMEN

BACKGROUND: Increased numbers and improperly positioned centrosomes, aneuploidy or polyploidy, and chromosomal instability are frequently observed characteristics of cancer cells. While some aspects of these events and the checkpoint mechanisms are well studied, not all players have yet been identified. As the role of proteases other than the proteasome in tumorigenesis is an insufficiently addressed question, we investigated the epigenetic control of the widely conserved protease HTRA1 and the phenotypes of deregulation. METHODS: Mouse embryonal fibroblasts and HCT116 and SW480 cells were used to study the mechanism of epigenetic silencing of HTRA1. In addition, using cell biological and genetic methods, the phenotypes of downregulation of HTRA1 expression were investigated. RESULTS: HTRA1 is epigenetically silenced in HCT116 colon carcinoma cells via the epigenetic adaptor protein MBD2. On the cellular level, HTRA1 depletion causes multiple phenotypes including acceleration of cell growth, centrosome amplification and polyploidy in SW480 colon adenocarcinoma cells as well as in primary mouse embryonic fibroblasts (MEFs). CONCLUSIONS: Downregulation of HTRA1 causes a number of phenotypes that are hallmarks of cancer cells suggesting that the methylation state of the HtrA1 promoter may be used as a biomarker for tumour cells or cells at risk of transformation.


Asunto(s)
Neoplasias del Colon/genética , Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Fibroblastos/citología , Serina Endopeptidasas/genética , Animales , Línea Celular Tumoral , Proliferación Celular , Células Cultivadas , Centrosoma/metabolismo , Neoplasias del Colon/patología , Regulación hacia Abajo , Epigénesis Genética , Fibroblastos/metabolismo , Regulación Neoplásica de la Expresión Génica , Células HCT116 , Serina Peptidasa A1 que Requiere Temperaturas Altas , Humanos , Ratones , Trasplante de Neoplasias , Poliploidía , Regiones Promotoras Genéticas
14.
Nat Commun ; 7: 10883, 2016 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-26955760

RESUMEN

Constitutive activation of Wnt/ß-catenin inhibits oligodendrocyte myelination. Tcf7l2/Tcf4, a ß-catenin transcriptional partner, is required for oligodendrocyte differentiation. How Tcf7l2 modifies ß-catenin signalling and controls myelination remains elusive. Here we define a stage-specific Tcf7l2-regulated transcriptional circuitry in initiating and sustaining oligodendrocyte differentiation. Multistage genome occupancy analyses reveal that Tcf7l2 serially cooperates with distinct co-regulators to control oligodendrocyte lineage progression. At the differentiation onset, Tcf7l2 interacts with a transcriptional co-repressor Kaiso/Zbtb33 to block ß-catenin signalling. During oligodendrocyte maturation, Tcf7l2 recruits and cooperates with Sox10 to promote myelination. In that context, Tcf7l2 directly activates cholesterol biosynthesis genes and cholesterol supplementation partially rescues oligodendrocyte differentiation defects in Tcf712 mutants. Together, we identify stage-specific co-regulators Kaiso and Sox10 that sequentially interact with Tcf7l2 to coordinate the switch at the transitions of differentiation initiation and maturation during oligodendrocyte development, and point to a previously unrecognized role of Tcf7l2 in control of cholesterol biosynthesis for CNS myelinogenesis.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Sistema Nervioso/crecimiento & desarrollo , Oligodendroglía/metabolismo , Factores de Transcripción SOXE/metabolismo , Proteína 2 Similar al Factor de Transcripción 7/metabolismo , Factores de Transcripción/metabolismo , Animales , Regulación de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Sistema Nervioso/metabolismo , Unión Proteica , Factores de Transcripción SOXE/genética , Transducción de Señal , Especificidad de la Especie , Proteína 2 Similar al Factor de Transcripción 7/genética , Factores de Transcripción/genética
15.
J Vis Exp ; (105)2015 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-26649885

RESUMEN

The epithelial surface of the mammalian intestine is a dynamic tissue that renews every 3 - 7 days. Understanding this renewal process identified a population of rapidly cycling intestinal stem cells (ISCs) characterized by their expression of the Lgr5 gene. These are supported by a quiescent stem cell population, marked by Bmi-1 expression, capable of replacing them in the event of injury. Investigating the interactions between these populations is crucial to understanding their roles in disease and cancer. The ISCs exist within crypts on the intestinal surface, these niches support the ISC in replenishing the epithelia. The interaction between active and quiescent ISCs likely involves other differentiated cells within the niche, as it has previously been demonstrated that the ''stemness'' of the Lgr5 ISC is closely tied to the presence of their neighboring Paneth cells. Using conditional cre-lox mouse models we tested the effect of deleting the majority of active ISCs in the presence or absence of the Paneth cells. Here we describe the techniques and analysis undertaken to characterize the intestine and demonstrate that the Paneth cells play a crucial role within the ISC niche in aiding recovery following substantial insult.

16.
EMBO J ; 34(18): 2321-33, 2015 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-26240067

RESUMEN

Wnt pathway deregulation is a common characteristic of many cancers. Only colorectal cancer predominantly harbours mutations in APC, whereas other cancer types (hepatocellular carcinoma, solid pseudopapillary tumours of the pancreas) have activating mutations in ß-catenin (CTNNB1). We have compared the dynamics and the potency of ß-catenin mutations in vivo. Within the murine small intestine (SI), an activating mutation of ß-catenin took much longer to achieve Wnt deregulation and acquire a crypt-progenitor cell (CPC) phenotype than Apc or Gsk3 loss. Within the colon, a single activating mutation of ß-catenin was unable to drive Wnt deregulation or induce the CPC phenotype. This ability of ß-catenin mutation to differentially transform the SI versus the colon correlated with higher expression of E-cadherin and a higher number of E-cadherin:ß-catenin complexes at the membrane. Reduction in E-cadherin synergised with an activating mutation of ß-catenin resulting in a rapid CPC phenotype within the SI and colon. Thus, there is a threshold of ß-catenin that is required to drive transformation, and E-cadherin can act as a buffer to sequester mutated ß-catenin.


Asunto(s)
Cadherinas/metabolismo , Transformación Celular Neoplásica , Neoplasias del Colon , Mutación , Proteínas de Neoplasias , Vía de Señalización Wnt , beta Catenina , Animales , Cadherinas/genética , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Ratones , Ratones Transgénicos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
17.
Proc Natl Acad Sci U S A ; 111(42): 15078-83, 2014 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-25288747

RESUMEN

An unresolved issue in genotoxic stress response is identification of induced regulatory proteins and how these activate tumor suppressor p53 to determine appropriate cell responses. Transcription factor KAISO was previously described to repress transcription following binding to methylated DNA. In this study, we show that KAISO is induced by DNA damage in p53-expressing cells and then interacts with the p53-p300 complex to increase acetylation of p53 K320 and K382 residues, although decreasing K381 acetylation. Moreover, the p53 with this particular acetylation pattern shows increased DNA binding and potently induces cell cycle arrest and apoptosis by activating transcription of CDKN1A (cyclin-dependent kinase inhibitor 1) and various apoptotic genes. Analogously, in Kaiso KO mouse embryonic fibroblast cells, p53-to-promoter binding and up-regulation of p21 and apoptosis gene expression is significantly compromised. KAISO may therefore be a critical regulator of p53-mediated cell cycle arrest and apoptosis in response to various genotoxic stresses in mammalian cells.


Asunto(s)
Apoptosis , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Acetilación , Animales , Ciclo Celular , Línea Celular , Proliferación Celular , ADN/química , Daño del ADN , Metilación de ADN , Proteína p300 Asociada a E1A/metabolismo , Femenino , Fibroblastos/citología , Células HCT116 , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Regiones Promotoras Genéticas , Unión Proteica , Elementos de Respuesta
18.
PLoS Genet ; 9(8): e1003638, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23935526

RESUMEN

Conditional deletion of Apc in the murine intestine alters crypt-villus architecture and function. This process is accompanied by multiple changes in gene expression, including upregulation of Cited1, whose role in colorectal carcinogenesis is unknown. Here we explore the relevance of Cited1 to intestinal tumorigenesis. We crossed Cited1 null mice with Apc(Min/+) and AhCre(+)Apc(fl/fl) mice and determined the impact of Cited1 deficiency on tumour growth/initiation including tumour multiplicity, cell proliferation, apoptosis and the transcriptome. We show that Cited1 is up-regulated in both human and murine tumours, and that constitutive deficiency of Cited1 increases survival in Apc(Min/+) mice from 230.5 to 515 days. However, paradoxically, Cited1 deficiency accentuated nearly all aspects of the immediate phenotype 4 days after conditional deletion of Apc, including an increase in cell death and enhanced perturbation of differentiation, including of the stem cell compartment. Transcriptome analysis revealed multiple pathway changes, including p53, PI3K and Wnt. The activation of Wnt through Cited1 deficiency correlated with increased transcription of ß-catenin and increased levels of dephosphorylated ß-catenin. Hence, immediately following deletion of Apc, Cited1 normally restrains the Wnt pathway at the level of ß-catenin. Thus deficiency of Cited1 leads to hyper-activation of Wnt signaling and an exaggerated Wnt phenotype including elevated cell death. Cited1 deficiency decreases intestinal tumourigenesis in Apc(Min/+) mice and impacts upon a number of oncogenic signaling pathways, including Wnt. This restraint imposed by Cited1 is consistent with a requirement for Cited1 to constrain Wnt activity to a level commensurate with optimal adenoma formation and maintenance, and provides one mechanism for tumour repression in the absence of Cited1.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/genética , Carcinogénesis , Perfilación de la Expresión Génica , Proteínas Nucleares/genética , Factores de Transcripción/genética , Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Animales , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis , Diferenciación Celular , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Células Madre Neoplásicas , Proteínas Nucleares/deficiencia , Proteínas Nucleares/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Transactivadores , Factores de Transcripción/deficiencia , Factores de Transcripción/metabolismo , Vía de Señalización Wnt/genética , beta Catenina/genética
19.
Stem Cells ; 31(4): 776-85, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23335179

RESUMEN

The identification of the intestinal stem cell (ISC) markers Lgr5 and Bmi-1 has furthered our understanding of how they accomplish homeostasis in this rapidly self-renewing tissue. Recent work indicates that these markers identify a cycling Lgr5(+) ISC which can be replaced by a quiescent Bmi-1(+) ISC. Currently, there is little data on how these cells interact to control intestinal crypt homeostasis and regeneration. This interaction likely involves other differentiated cells within the niche as it has previously been demonstrated that the "stemness" of the Lgr5 ISC is closely tied to the presence of their neighboring Paneth cells. To investigate this, we used two conditional mouse models to delete the transcription factor ß-catenin within the intestinal crypt. Critically these differ in their ability to drive recombination within Paneth cells and therefore allow us to compare the effect of deleting the majority of active ISCs in the presence or absence of the Paneth cells. After gene deletion, the intestines in the model in which Paneth cells were retained showed a rapid recovery and repopulation of the crypt-villus axis presumably from either a spared ISC or the hypothetical quiescent ISCs. However, in the absence of Paneth cells the recovery ability was compromised resulting in complete loss of intestinal epithelial integrity. This data indicates that the Paneth cells play a crucial role within the in vivo ISC niche in aiding recovery following substantial insult.


Asunto(s)
Intestinos/citología , Intestinos/lesiones , Células de Paneth/citología , Células de Paneth/metabolismo , Alelos , Animales , Inmunohistoquímica , Ratones , Ratones Transgénicos , Células Madre/citología , Células Madre/metabolismo , beta Catenina/deficiencia , beta Catenina/genética , beta Catenina/metabolismo
20.
Gut ; 61(9): 1306-14, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22082586

RESUMEN

BACKGROUND: Cyclooxygenase-2 (COX-2) overexpression in colorectal cancer increases levels of its pro-tumorigenic product prostaglandin E2 (PGE(2)). The recently identified colorectal tumour suppressor 15-prostaglandin dehydrogenase (15-PGDH) catalyses prostaglandin turnover and is downregulated at a very early stage in colorectal tumorigenesis; however, the mechanism responsible remains unclear. As Wnt/ß-catenin signalling is also deregulated early in colorectal neoplasia, a study was undertaken to determine whether ß-catenin represses 15-PGDH expression. METHODS: The effect of modulating Wnt/ß-catenin signalling (using ß-catenin siRNA, mutant TCF4, Wnt3A or GSK3 inhibition) on 15-PGDH mRNA, protein expression and promoter activity was determined in colorectal cell lines by immunoblotting, qRT-PCR and reporter assays. The effect of ß-catenin deletion in vivo was addressed by 15-PGDH immunostaining of ß-catenin(-/lox)-villin-creERT2 mouse tissue. 15-PGDH promoter occupancy was determined using chromatin immunoprecipitation and PGE(2) levels by ELISA. RESULTS: The study shows for the first time that ß-catenin knockdown upregulates 15-PGDH in colorectal adenoma and carcinoma cells without affecting COX-2 protein levels. A dominant negative mutant form of TCF4 (dnTCF4), unable to bind ß-catenin, also upregulated 15-PGDH; conversely, increasing ß-catenin activity using Wnt3A or GSK3 inhibition downregulated 15-PGDH. Importantly, inducible ß-catenin deletion in vivo also upregulated intestinal epithelial 15-PGDH. 15-PGDH regulation occurred at the protein, mRNA and promoter activity levels and chromatin immunoprecipitation indicated ß-catenin/TCF4 binding to the 15-PGDH promoter. ß-catenin knockdown decreased PGE(2) levels, and this was significantly rescued by 15-PGDH siRNA. CONCLUSION: These data suggest a novel role for ß-catenin in promoting colorectal tumorigenesis through very early 15-PGDH suppression leading to increased PGE(2) levels, possibly even before COX-2 upregulation.


Asunto(s)
Adenoma/enzimología , Neoplasias Colorrectales/enzimología , Hidroxiprostaglandina Deshidrogenasas/metabolismo , Mucosa Intestinal/enzimología , beta Catenina/fisiología , Animales , Línea Celular Tumoral , Ciclooxigenasa 2/metabolismo , Dinoprostona/metabolismo , Regulación hacia Abajo , Represión Enzimática , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Hidroxiprostaglandina Deshidrogenasas/biosíntesis , Hidroxiprostaglandina Deshidrogenasas/genética , Immunoblotting , Inmunohistoquímica , Ratones , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Activación Transcripcional , Regulación hacia Arriba , beta Catenina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA