Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
2.
J Gastrointest Cancer ; 54(4): 1017-1030, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37009977

RESUMEN

BACKGROUND: Immune checkpoint inhibitors have recently replaced over chemotherapy as the first-line treatment for microsatellite instability-high or mismatch repair deficient (dMMR/MSI-H) stage 4 colorectal cancers. Considering this success, many studies have tried to replicate the use of immune checkpoint inhibitors, either as a single agent or in combination with other therapeutic agents, in the treatment of proficient mismatch repair (pMMR/MSS) stage 4 colorectal cancers. This review summarizes the seminal clinical data about the immune checkpoint inhibitors used in pMMR/MSS colorectal cancers and some future directions. RESULTS: Studies concerning the use of immune checkpoint inhibitors as a single agent or in combination with other immune checkpoint inhibitors, targeted therapy, chemotherapy, or radiotherapy have proven inefficient in the treatment of pMMR/MSS colorectal cancer. However, a small subset of patients with pMMR/MSS colorectal cancer who has a mutation in POLE and POLD1 enzymes may respond to immunotherapy. Moreover, patients without liver metastasis appear to have a better chance of response. New immune checkpoint targets are being identified, such as VISTA, TIGIT, LAG3, STING signal pathway, and BTLA, and studies are ongoing to determine their efficiency in this disease type. CONCLUSION: Immune checkpoint inhibitor-based regimens have not yet shown any meaningful positive outcomes for most pMMR/MSS colorectal cancers. A beneficial effect among a minority of these patients has been observed, but concrete biomarkers of response are lacking. Understanding the underlying mechanisms of immune resistance should guide further research for overcoming these obstacles.


Asunto(s)
Neoplasias del Colon , Neoplasias Colorrectales , Humanos , Reparación de la Incompatibilidad de ADN , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunoterapia , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Inestabilidad de Microsatélites
4.
Brief Bioinform ; 24(1)2023 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-36472568

RESUMEN

Accounting for cell type compositions has been very successful at analyzing high-throughput data from heterogeneous tissues. Differential gene expression analysis at cell type level is becoming increasingly popular, yielding biomarker discovery in a finer granularity within a particular cell type. Although several computational methods have been developed to identify cell type-specific differentially expressed genes (csDEG) from RNA-seq data, a systematic evaluation is yet to be performed. Here, we thoroughly benchmark six recently published methods: CellDMC, CARseq, TOAST, LRCDE, CeDAR and TCA, together with two classical methods, csSAM and DESeq2, for a comprehensive comparison. We aim to systematically evaluate the performance of popular csDEG detection methods and provide guidance to researchers. In simulation studies, we benchmark available methods under various scenarios of baseline expression levels, sample sizes, cell type compositions, expression level alterations, technical noises and biological dispersions. Real data analyses of three large datasets on inflammatory bowel disease, lung cancer and autism provide evaluation in both the gene level and the pathway level. We find that csDEG calling is strongly affected by effect size, baseline expression level and cell type compositions. Results imply that csDEG discovery is a challenging task itself, with room to improvements on handling low signal-to-noise ratio and low expression genes.


Asunto(s)
Perfilación de la Expresión Génica , Programas Informáticos , Perfilación de la Expresión Génica/métodos , RNA-Seq , Simulación por Computador , Relación Señal-Ruido , Análisis de Secuencia de ARN/métodos
5.
Clin Colorectal Cancer ; 21(4): 315-324, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36283915

RESUMEN

BACKGROUND: Adjuvant chemotherapy for stage III colon cancer is underutilized in the United States. The aim of this study was to assess the use of adjuvant chemotherapy in younger and medically fit patients and analyze the socioeconomic factors associated with its utilization. METHODS: Using the National Cancer Database from 2004 to 2015, we selected stage III colon cancer patients between age 18 to 65, Charlson-Deyo Comorbidity Index (CDCI) of 0 or 1, and those that survived at least 12 months after surgery. We then compared patients that underwent surgery only with those that received adjuvant chemotherapy. Multivariable logistic regression analysis was performed to identify variables associated with adjuvant chemotherapy use in the population. Overall survival was estimated by Kaplan-Meier curves. RESULTS: Of the 48,336 patients that met inclusion criteria, 43,315 (90%) received adjuvant chemotherapy. The utilization of adjuvant chemotherapy increased from 87% in 2004 to 91% in 2015. On multivariable regression analysis, the use of adjuvant chemotherapy was lower among males, Non-Hispanic Blacks and Hispanics, low-grade cancer, left-sided tumors, CDCI 1, those who travel ≥ 50 miles, yearly income < $40,227, and uninsured patients. The most common reason for the omission of adjuvant chemotherapy was the patient or caregiver's choice (40% between 2013 and 2015). The 5-year and 10-year overall survival rates were 76.7% and 63.8% respectively, in those who received adjuvant chemotherapy as compared to 65.1% and 49.3% in those who underwent surgery only (P < .001). CONCLUSION: In young and medically fit stage III colon cancer patients, most patients received guideline-compliant care in the United States. However, socioeconomic disparities adversely impacted the use of adjuvant chemotherapy. The patient or caregiver's decision was the most common reason for non-adherence to adjuvant chemotherapy and lead to poor survival outcomes. Emphasis should be placed on developing patient-centered strategies to improve adherence to chemotherapy in all patients.


Asunto(s)
Neoplasias del Colon , Masculino , Humanos , Estados Unidos/epidemiología , Persona de Mediana Edad , Adolescente , Adulto Joven , Adulto , Anciano , Estadificación de Neoplasias , Quimioterapia Adyuvante , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/epidemiología , Comorbilidad , Tasa de Supervivencia
6.
Cells ; 11(9)2022 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-35563773

RESUMEN

Recurrent chronic mucosal inflammation, a characteristic of inflammatory bowel diseases (IBD), perturbs the intestinal epithelial homeostasis resulting in formation of mucosal wounds and, in most severe cases, leads to colitis-associated colon cancer (CAC). The altered structure of epithelial cell-cell adhesions is a hallmark of intestinal inflammation contributing to epithelial injury, repair, and tumorigenesis. P-cadherin is an important adhesion protein, poorly expressed in normal intestinal epithelial cells (IEC) but upregulated in inflamed and injured mucosa. The goal of this study was to investigate the roles of P-cadherin in regulating intestinal inflammation and CAC. P-cadherin expression was markedly induced in the colonic epithelium of human IBD patients and CAC tissues. The roles of P-cadherin were investigated in P-cadherin null mice using dextran sulfate sodium (DSS)-induced colitis and an azoxymethane (AOM)/DSS induced CAC. Although P-cadherin knockout did not affect the severity of acute DSS colitis, P-cadherin null mice exhibited faster recovery after colitis. No significant differences in the number of colonic tumors were observed in P-cadherin null and control mice. Consistently, the CRISPR/Cas9-mediated knockout of P-cadherin in human IEC accelerated epithelial wound healing without affecting cell proliferation. The accelerated migration of P-cadherin depleted IEC was driven by activation of Src kinases, Rac1 GTPase and myosin II motors and was accompanied by transcriptional reprogramming of the cells. Our findings highlight P-cadherin as a negative regulator of IEC motility in vitro and mucosal repair in vivo. In contrast, this protein is dispensable for IEC proliferation and CAC development.


Asunto(s)
Cadherinas , Neoplasias Asociadas a Colitis , Colitis , Enfermedades Inflamatorias del Intestino , Animales , Cadherinas/metabolismo , Colitis/inducido químicamente , Colitis/complicaciones , Colitis/metabolismo , Sulfato de Dextran , Células Epiteliales/metabolismo , Humanos , Inflamación/metabolismo , Enfermedades Inflamatorias del Intestino/metabolismo , Mucosa Intestinal/patología , Ratones , Ratones Noqueados
7.
Bioinformatics ; 38(8): 2361-2363, 2022 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-35176143

RESUMEN

SUMMARY: Correctly annotating individual cell's type is an important initial step in single-cell RNA sequencing (scRNA-seq) data analysis. Here, we present NeuCA web server, a neural network-based scRNA-seq cell annotation tool with web-app portal and graphical user interface, for automatically assigning cell labels. NeuCA algorithm is accurate and exhaustive, maximizing the usage of measured cells for downstream analysis. NeuCA web server provides over 20 ready-to-use pre-trained classifiers for commonly used tissue types. As the first web-app tool with neural-network infrastructure implemented, NeuCA web will facilitate the research community in analyzing and annotating scRNA-seq data. AVAILABILITY AND IMPLEMENTATION: NeuCA web server is implemented with R Shiny application online at https://statbioinfo.shinyapps.io/NeuCA/. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Aplicaciones Móviles , Programas Informáticos , Computadores , Algoritmos , Redes Neurales de la Computación
8.
Cell ; 185(2): 283-298.e17, 2022 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-35021065

RESUMEN

Gasdermins are a family of structurally related proteins originally described for their role in pyroptosis. Gasdermin B (GSDMB) is currently the least studied, and while its association with genetic susceptibility to chronic mucosal inflammatory disorders is well established, little is known about its functional relevance during active disease states. Herein, we report increased GSDMB in inflammatory bowel disease, with single-cell analysis identifying epithelial specificity to inflamed colonocytes/crypt top colonocytes. Surprisingly, mechanistic experiments and transcriptome profiling reveal lack of inherent GSDMB-dependent pyroptosis in activated epithelial cells and organoids but instead point to increased proliferation and migration during in vitro wound closure, which arrests in GSDMB-deficient cells that display hyper-adhesiveness and enhanced formation of vinculin-based focal adhesions dependent on PDGF-A-mediated FAK phosphorylation. Importantly, carriage of disease-associated GSDMB SNPs confers functional defects, disrupting epithelial restitution/repair, which, altogether, establishes GSDMB as a critical factor for restoration of epithelial barrier function and the resolution of inflammation.


Asunto(s)
Células Epiteliales/metabolismo , Células Epiteliales/patología , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/patología , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Piroptosis , Secuencia de Bases , Estudios de Casos y Controles , Adhesión Celular/efectos de los fármacos , Adhesión Celular/genética , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Células Epiteliales/efectos de los fármacos , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Células HEK293 , Células HT29 , Humanos , Enfermedades Inflamatorias del Intestino/genética , Metotrexato/farmacología , Mutación/genética , Fosforilación/efectos de los fármacos , Polimorfismo de Nucleótido Simple/genética , Piroptosis/efectos de los fármacos , Piroptosis/genética , Reproducibilidad de los Resultados , Transcriptoma/efectos de los fármacos , Transcriptoma/genética , Regulación hacia Arriba/efectos de los fármacos , Cicatrización de Heridas/efectos de los fármacos , Cicatrización de Heridas/genética
9.
Cancers (Basel) ; 13(24)2021 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-34944826

RESUMEN

Colorectal cancer (CRC) is the third most common malignancy and the second most common cause of cancer-related mortality worldwide. A total of 20% of CRC patients present with distant metastases, most frequently to the liver and lung. In the primary tumor, as well as at each metastatic site, the cellular components of the tumor microenvironment (TME) contribute to tumor engraftment and metastasis. These include immune cells (macrophages, neutrophils, T lymphocytes, and dendritic cells) and stromal cells (cancer-associated fibroblasts and endothelial cells). In this review, we highlight how the TME influences tumor progression and invasion at the primary site and its function in fostering metastatic niches in the liver and lungs. We also discuss emerging clinical strategies to target the CRC TME.

10.
Adv Sci (Weinh) ; 8(19): e2004673, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34378358

RESUMEN

Colorectal cancer (CRC) metastasizes mainly to the liver, which accounts for the majority of CRC-related deaths. Here it is shown that metastatic cells undergo specific chromatin remodeling in the liver. Hepatic growth factor (HGF) induces phosphorylation of PU.1, a pioneer factor, which in turn binds and opens chromatin regions of downstream effector genes. PU.1 increases histone acetylation at the DPP4 locus. Precise epigenetic silencing by CRISPR/dCas9KRAB or CRISPR/dCas9HDAC revealed that individual PU.1-remodeled regulatory elements collectively modulate DPP4 expression and liver metastasis growth. Genetic silencing or pharmacological inhibition of each factor along this chromatin remodeling axis strongly suppressed liver metastasis. Therefore, microenvironment-induced epimutation is an important mechanism for metastatic tumor cells to grow in their new niche. This study presents a potential strategy to target chromatin remodeling in metastatic cancer and the promise of repurposing drugs to treat metastasis.


Asunto(s)
Ensamble y Desensamble de Cromatina/genética , Neoplasias Colorrectales/patología , Dipeptidil Peptidasa 4/genética , Factor de Crecimiento de Hepatocito/genética , Neoplasias Hepáticas/secundario , Proteínas Proto-Oncogénicas/genética , Transactivadores/genética , Animales , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Dipeptidil Peptidasa 4/metabolismo , Epigénesis Genética/genética , Regulación Neoplásica de la Expresión Génica/genética , Factor de Crecimiento de Hepatocito/metabolismo , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Ratones , Ratones Endogámicos BALB C , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas/metabolismo , Transactivadores/metabolismo
11.
Cancers (Basel) ; 13(4)2021 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-33670106

RESUMEN

Colorectal cancer (CRC) remains the third most common cause of cancer and the second most common cause of cancer deaths worldwide. Clinicians are largely faced with advanced and metastatic disease for which few interventions are available. One poorly understood aspect of CRC involves altered organization of the actin cytoskeleton, especially at the metastatic stage of the disease. Myosin motors are crucial regulators of actin cytoskeletal architecture and remodeling. They act as mechanosensors of the tumor environments and control key cellular processes linked to oncogenesis, including cell division, extracellular matrix adhesion and tissue invasion. Different myosins play either oncogenic or tumor suppressor roles in breast, lung and prostate cancer; however, little is known about their functions in CRC. This review focuses on the functional roles of myosins in colon cancer development. We discuss the most studied class of myosins, class II (conventional) myosins, as well as several classes (I, V, VI, X and XVIII) of unconventional myosins that have been linked to CRC development. Altered expression and mutations of these motors in clinical tumor samples and their roles in CRC growth and metastasis are described. We also evaluate the potential of using small molecular modulators of myosin activity to develop novel anticancer therapies.

12.
Nat Commun ; 12(1): 262, 2021 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-33431859

RESUMEN

The pathogenesis of ulcerative colitis (UC), a major type of inflammatory bowel disease, remains unknown. No model exists that adequately recapitulates the complexity of clinical UC. Here, we take advantage of induced pluripotent stem cells (iPSCs) to develop an induced human UC-derived organoid (iHUCO) model and compared it with the induced human normal organoid model (iHNO). Notably, iHUCOs recapitulated histological and functional features of primary colitic tissues, including the absence of acidic mucus secretion and aberrant adherens junctions in the epithelial barrier both in vitro and in vivo. We demonstrate that the CXCL8/CXCR1 axis was overexpressed in iHUCO but not in iHNO. As proof-of-principle, we show that inhibition of CXCL8 receptor by the small-molecule non-competitive inhibitor repertaxin attenuated the progression of UC phenotypes in vitro and in vivo. This patient-derived organoid model, containing both epithelial and stromal compartments, will generate new insights into the underlying pathogenesis of UC while offering opportunities to tailor interventions to the individual patient.


Asunto(s)
Colitis Ulcerosa/patología , Organoides/patología , Uniones Adherentes/metabolismo , Cadherinas/metabolismo , Progresión de la Enfermedad , Epitelio/patología , Fibroblastos/patología , Humanos , Inflamación/patología , Epiplón/trasplante , Fenotipo , Análisis de Componente Principal , Análisis de Secuencia de ARN , Sulfonamidas/farmacología , Transcriptoma/genética , beta Catenina/metabolismo
13.
iScience ; 23(11): 101719, 2020 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-33205026

RESUMEN

Colon cancer remains the third most common cause of cancer in the US, and the third most common cause of cancer death. Worldwide, colon cancer is the second most common cause of cancer and cancer deaths. At least 25% of patients still present with metastatic disease, and at least 25-30% will develop metastatic colon cancer in the course of their disease. While chemotherapy and surgery remain the mainstay of treatment, understanding the fundamental cellular niche and mechanical properties that result in metastases would facilitate both prevention and cure. Advances in biomaterials, novel 3D primary human cells, modelling using microfluidics and the ability to alter the physical environment, now offers a unique opportunity to develop and test impactful treatment.

14.
Dis Colon Rectum ; 63(11): 1559-1569, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32868555

RESUMEN

BACKGROUND: Colorectal cancer and IBD account for a large portion of the practice of colorectal surgery. Historical research models have provided insights into the underlying causes of these diseases but come with many limitations. OBJECTIVE: The aim of this study was to systematically review the literature regarding the advantage of organoid models in modeling benign and malignant colorectal pathology. DATA SOURCES: Sources included PubMed, Ovid-Medline, and Ovid Embase STUDY SELECTION:: Two reviewers completed a systematic review of the literature between January 2006 and January of 2020 for studies related to colon and intestinal organoids. Reviews, commentaries, protocols, and studies not performed in humans or mice were excluded. RESULTS: A total of 73 articles were included. Organoid models of colorectal disease have been rising in popularity to further elucidate the genetic, transcriptomic, and treatment response of these diseases at the individual level. Increasingly complex models utilizing coculture techniques are being rapidly developed that allow in vitro recapitulation of the disease microenvironment. LIMITATIONS: This review is only qualitative, and the lack of well utilized nomenclature in the organoid community may have resulted in the exclusion of articles. CONCLUSIONS: Historical disease models including cell lines, patient-derived tumor xenografts, and animal models have created a strong foundation for our understanding of colorectal pathology. Recent advances in 3-dimensional cell cultures, in the form of patient-derived epithelial organoids and induced human intestinal organoids have opened a new avenue for high-resolution analysis of pathology at the level of an individual patient. Recent research has shown the potential of organoids as a tool for personalized medicine with their ability to retain patient characteristics, including treatment response.


Asunto(s)
Neoplasias Colorrectales , Enfermedades Inflamatorias del Intestino , Modelos Animales , Organoides , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Línea Celular , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/terapia , Humanos , Enfermedades Inflamatorias del Intestino/patología , Enfermedades Inflamatorias del Intestino/terapia , Ratones , Medicina de Precisión/métodos , Utilización de Procedimientos y Técnicas
15.
Nat Commun ; 11(1): 900, 2020 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-32060280

RESUMEN

Copper levels are known to be elevated in inflamed and malignant tissues. But the mechanism underlying this selective enrichment has been elusive. In this study, we report a axis by which inflammatory cytokines, such as IL-17, drive cellular copper uptake via the induction of a metalloreductase, STEAP4. IL-17-induced elevated intracellular copper level leads to the activation of an E3-ligase, XIAP, which potentiates IL-17-induced NFκB activation and suppresses the caspase 3 activity. Importantly, this IL-17-induced STEAP4-dependent cellular copper uptake is critical for colon tumor formation in a murine model of colitis-associated tumorigenesis and STEAP4 expression correlates with IL-17 level and XIAP activation in human colon cancer. In summary, this study reveals a IL-17-STEAP4-XIAP axis through which the inflammatory response induces copper uptake, promoting colon tumorigenesis.


Asunto(s)
Colon/inmunología , Neoplasias del Colon/inmunología , Cobre/metabolismo , Proteínas Inhibidoras de la Apoptosis/inmunología , Interleucina-17/inmunología , Proteínas de la Membrana/inmunología , Animales , Carcinogénesis , Colitis/genética , Colitis/inmunología , Colitis/metabolismo , Colon/metabolismo , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Cobre/inmunología , Humanos , Proteínas Inhibidoras de la Apoptosis/genética , Interleucina-17/genética , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
16.
Integr Biol (Camb) ; 12(1): 1-11, 2020 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-31965190

RESUMEN

Tumor-initiating cells (TICs), a subpopulation of cancerous cells with high tumorigenic potential and stem-cell-like properties, drive tumor progression and are resistant to conventional therapies. Identification and isolation of TICs are limited by their low frequency and lack of robust markers. Here, we characterize the heterogeneous adhesive properties of a panel of human and murine cancer cells and demonstrate differences in adhesion strength among cells, which exhibit TIC properties and those that do not. These differences in adhesion strength were exploited to rapidly (~10 min) and efficiently isolate cancerous cells with increased tumorigenic potential in a label-free manner by use of a microfluidic technology. Isolated murine and human cancer cells gave rise to larger tumors with increased growth rate and higher frequency in both immunocompetent and immunocompromised mice, respectively. This rapid and label-free TIC isolation technology has the potential to be a valuable tool for facilitating research into TIC biology and the development of more efficient diagnostics and cancer therapies.


Asunto(s)
Carcinogénesis/patología , Adhesión Celular , Separación Celular/métodos , Hidrodinámica , Neoplasias/fisiopatología , Células Madre Neoplásicas/patología , Animales , Neoplasias de la Mama/patología , Línea Celular Tumoral , Progresión de la Enfermedad , Femenino , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Células MCF-7 , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Microfluídica , Transducción de Señal , Estrés Mecánico
18.
Neoplasia ; 21(3): 269-281, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30738331

RESUMEN

Dysfunctional inflammatory pathways are associated with an increased risk of cancer, including colorectal cancer. We have previously identified and enriched for a self-renewing, colon cancer stem cell (CCSC) subpopulation in primary sporadic colorectal cancers (CRC) and a related subpopulation in ulcerative colitis (UC) patients defined by the stem cell marker, aldehyde dehydrogenase (ALDH). Subsequent work demonstrated that CCSC-initiated tumors are dependent on the inflammatory chemokine, CXCL8, a known inducer of tumor proliferation, angiogenesis and invasion. Here, we use RNA interference to target CXCL8 and its receptor, CXCR1, to establish the existence of a functional signaling pathway promoting tumor growth initiated by sporadic and colitis CCSCs. Knocking down either CXCL8 or CXCR1 had a dramatic effect on inhibiting both in vitro proliferation and angiogenesis. Likewise, tumorigenicity was significantly inhibited due to reduced levels of proliferation and angiogenesis. Decreased expression of cycle cell regulators cyclins D1 and B1 along with increased p21 levels suggested that the reduction in tumor growth is due to dysregulation of cell cycle progression. Therapeutically targeting the CXCL8-CXCR1 signaling pathway has the potential to block sustained tumorigenesis by inhibiting both CCSC- and pCCSC-induced proliferation and angiogenesis.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Neoplasias del Colon/etiología , Neoplasias del Colon/metabolismo , Inflamación/metabolismo , Interleucina-8/metabolismo , Células Madre Neoplásicas/metabolismo , Receptores de Interleucina-8A/metabolismo , Transducción de Señal , Animales , Biomarcadores , Línea Celular Tumoral , Proliferación Celular , Colitis/complicaciones , Colitis/genética , Colitis/metabolismo , Neoplasias del Colon/patología , Modelos Animales de Enfermedad , Dosificación de Gen , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Xenoinjertos , Humanos , Inmunofenotipificación , Inflamación/complicaciones , Inflamación/genética , Interleucina-8/genética , Ratones , Modelos Biológicos , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Receptores de Interleucina-8A/genética
19.
Oncotarget ; 9(47): 28717-28730, 2018 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-29983891

RESUMEN

Ulcerative colitis (UC) is a prevalent form of inflammatory bowel disease (IBD) whose pathogenic mechanisms remain unclear. Elucidating these mechanisms is important to reduce UC symptoms and to prevent UC progression into colitis-associated colon cancer (CAC). Our goal was to develop and validate faithful, human-derived, UC models and analyze them at histologic, transcriptomic and epigenetic levels to allow mechanistic studies of UC and CAC pathogenesis. We generated patient-derived primary-organoid cultures from UC and non-IBD colonic epithelium. We phenotyped them histologically and used next-generation-sequencing approaches to profile whole transcriptomes and epigenomes of organoids and primary tissues. Tissue organization and expression of mucin 2 (MUC2) and lysozyme (LYZ) demonstrated histologic faithfulness of organoids to healthy and diseased colonic epithelium. Transcriptomic analyses showed increased expression of inflammatory pathways in UC patient-derived organoids and tissues. Profiling for active enhancers using the H3K27ac histone modification revealed UC-derived organoid enrichment for pathways indicative of gastrointestinal cancer, including S100 calcium-binding protein P (S100P), and revealed novel markers for GI cancer, including both LYZ and neuropeptide S receptor 1 (NPSR1). Immunolocalization showed increased levels of LYZ, S100P, and NPSR1 proteins in UC and CAC. In conclusion, primary colonic organoid cultures from UC and non-IBD patients can be established that faithfully represent diseased or normal colonic states. These models reveal precancerous molecular pathways that are already activated in UC. The findings demonstrate the suitability of primary organoids for dissecting UC and CAC pathogenic mechanisms and suggest new targets for therapeutic intervention.

20.
Oncotarget ; 9(16): 13048-13059, 2018 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-29560130

RESUMEN

Inflammatory bowel disease (IBD) affects one million people in the US. Ulcerative colitis (UC) is a subtype of IBD that can lead to colitis-associated cancer (CAC). In UC, the rate of CAC is 3-5-fold greater than the rate of sporadic colorectal cancer (CRC). The pathogenesis of UC and CAC are due to aberrant interactions between host immune system and microenvironment, but precise mechanisms are still unknown. In colitis and CAC, microenvironmental fibroblasts exhibit an activated, inflammatory phenotype that contributes to tumorigenesis accompanied by excessive secretion of the chemokine CXCL8. However, mechanisms regulating CXCL8 secretion are unclear. Since it is known that miRNAs regulate chemokines such as CXCL8, we queried a microRNA library for mimics affecting CXCL8 secretion. Among the identified microRNAs, miR-20a/b was further investigated as its stromal expression levels inversely correlated with the amounts of CXCL8 secreted and predicted fibroblast tumor-promoting activity. Indeed, miR-20a directly bound to the 3'UTR of CXCL8 mRNA and regulated its expression by translational repression. In vivo co-inoculation studies with CRC stem cells demonstrated that fibroblasts characterized by high miR-20a expression had reduced tumor-promoting activities. These studies reveal that in stromal fibroblasts, miR-20a modulates CXCL8 function, therefore influencing tumor latency.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...