Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cancer Prev Res (Phila) ; 15(12): 803-814, 2022 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-36049217

RESUMEN

Bacteria are believed to play an important role in intestinal tumorigenesis and contribute to both gut luminal and circulating metabolites. Celecoxib, a selective cyclooxygenase-2 inhibitor, alters gut bacteria and metabolites in association with suppressing the development of intestinal polyps in mice. The current study sought to evaluate whether celecoxib exerts its chemopreventive effects, in part, through intestinal bacteria and metabolomic alterations. Using ApcMin/+ mice, we demonstrated that treatment with broad-spectrum antibiotics (ABx) reduced abundance of gut bacteria and attenuated the ability of celecoxib to suppress intestinal tumorigenesis. Use of ABx also impaired celecoxib's ability to shift microbial populations and gut luminal and circulating metabolites. Treatment with ABx alone markedly reduced tumor number and size in ApcMin/+ mice, in conjunction with profoundly altering the metabolite profiles of the intestinal lumen and blood. Many of the metabolite changes in the gut and circulation overlapped and included shifts in microbially derived metabolites. To complement these findings in mice, we evaluated the effects of ABx on circulating metabolites in patients with colon cancer. This showed that ABx treatment led to a shift in blood metabolites, including several that were of bacterial origin. Importantly, changes in metabolites in patients given ABx overlapped with alterations found in mice that also received ABx. Taken together, these findings suggest a potential role for bacterial metabolites in mediating both the chemopreventive effects of celecoxib and intestinal tumor growth. PREVENTION RELEVANCE: This study demonstrates novel mechanisms by which chemopreventive agents exert their effects and gut microbiota impact intestinal tumor development. These findings have the potential to lead to improved cancer prevention strategies by modulating microbes and their metabolites.


Asunto(s)
Anticarcinógenos , Microbioma Gastrointestinal , Ratones , Animales , Celecoxib/farmacología , Inhibidores de la Ciclooxigenasa 2/farmacología , Metaboloma , Antibacterianos/farmacología , Anticarcinógenos/farmacología , Bacterias , Carcinogénesis
2.
Cancer Res ; 81(9): 2275-2288, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33526512

RESUMEN

Serine is a nonessential amino acid generated by the sequential actions of phosphoglycerate dehydrogenase (PHGDH), phosphoserine aminotransferase (PSAT1), and phosphoserine phosphatase (PSPH). Increased serine biosynthesis occurs in several cancers and supports tumor growth. In addition, cancer cells can harness exogenous serine to enhance their metabolism and proliferation. Here we tested the relative contributions of exogenous and endogenous sources of serine on the biology of colorectal cancer. In murine tumors, Apc status was identified as a determinant of the expression of genes controlling serine synthesis. In patient samples, PSAT1 was overexpressed in both colorectal adenomas and adenocarcinomas. Combining genetic deletion of PSAT1 with exogenous serine deprivation maximally suppressed the proliferation of colorectal cancer cells and induced profound metabolic defects including diminished nucleotide production. Inhibition of serine synthesis enhanced the transcriptional changes following exogenous serine removal as well as alterations associated with DNA damage. Both loss of PSAT1 and removal of serine from the diet were necessary to suppress colorectal cancer xenograft growth and enhance the antitumor activity of 5-fluorouracil (5-FU). Restricting endogenous and exogenous serine in vitro augmented 5-FU-induced cell death, DNA damage, and metabolic perturbations, likely accounting for the observed antitumor effect. Collectively, our results suggest that both endogenous and exogenous sources of serine contribute to colorectal cancer growth and resistance to 5-FU. SIGNIFICANCE: These findings provide insights into the metabolic requirements of colorectal cancer and reveal a novel approach for its treatment. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/9/2275/F1.large.jpg.


Asunto(s)
Antimetabolitos Antineoplásicos/administración & dosificación , Neoplasias del Colon/dietoterapia , Neoplasias del Colon/metabolismo , Dieta/métodos , Resistencia a Antineoplásicos/efectos de los fármacos , Fluorouracilo/administración & dosificación , Serina/deficiencia , Anciano , Animales , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Daño del ADN , Resistencia a Antineoplásicos/genética , Femenino , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Células HCT116 , Humanos , Masculino , Ratones , Ratones Desnudos , Ratones Transgénicos , Persona de Mediana Edad , Embarazo , Serina/genética , Transaminasas/deficiencia , Transaminasas/genética , Resultado del Tratamiento , Carga Tumoral/efectos de los fármacos , Carga Tumoral/genética , Ensayos Antitumor por Modelo de Xenoinjerto
3.
J Vis Exp ; (168)2021 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-33616099

RESUMEN

Understanding the tissue and cellular changes that occur in the acute injury response as well as during the wound healing process is of paramount importance when studying diseases of the gastrointestinal (GI) tract. The murine colonic pinch biopsy model is a useful tool to define these processes. Additionally, the interplay between gut luminal content (e.g., microbes) and the colon can be studied. However, wound induction and the ability to track wound closure over time in a reliable manner can be challenging. Moreover, tissue preparation and orientation must be carried out in a standardized way to optimally interrogate histologic and molecular changes. Here, we present a detailed method describing biopsy-induced injury and the monitoring of wound closure through repeat colonoscopies. An approach is described that ensures consistent and reproducible measurements of wound size, the ability to collect the wound bed for molecular analyses as well as visualize the wound bed upon sectioning of tissues. The ability to successfully carry out these techniques allows for studies of the acute injury response, wound healing and luminal-host interactions within the colon.


Asunto(s)
Colon/citología , Colonoscopía/métodos , Biopsia Guiada por Imagen/métodos , Cicatrización de Heridas , Animales , Colon/patología , Colon/cirugía , Ratones
4.
Am J Pathol ; 188(12): 2811-2825, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30273600

RESUMEN

Colonic inflammation, a hallmark of inflammatory bowel disease, can be influenced by host intrinsic and extrinsic factors. There continues to be a need for models of colonic inflammation that can both provide insights into disease pathogenesis and be used to investigate potential therapies. Herein, we tested the utility of colonoscopic-guided pinch biopsies in mice for studying colonic inflammation and its treatment. Gene expression profiling of colonic wound beds after injury showed marked changes, including increased expression of genes important for the inflammatory response. Interestingly, many of these gene expression changes mimicked those alterations found in inflammatory bowel disease patients. Biopsy-induced inflammation was associated with increases in neutrophils, macrophages, and natural killer cells. Injury also led to elevated levels of sphingosine-1-phosphate (S1P), a bioactive lipid that is an important mediator of inflammation mainly through its receptor, S1P1. Genetic deletion of S1P1 in the endothelium did not alter the inflammatory response but led to increased colonic bleeding. Bacteria invaded into the wound beds, raising the possibility that microbes contributed to the observed changes in mucosal gene expression. In support of this, reducing bacterial abundance markedly attenuated the inflammatory response to wounding. Taken together, this study demonstrates the utility of the pinch biopsy model of colonic injury to elucidate the molecular underpinnings of colonic inflammation and its treatment.


Asunto(s)
Colon/inmunología , Modelos Animales de Enfermedad , Inflamación/inmunología , Mucosa Intestinal/inmunología , Microbiota , Receptores de Lisoesfingolípidos/fisiología , Cirugía Asistida por Computador/métodos , Animales , Antibacterianos/farmacología , Biopsia , Células Cultivadas , Colon/lesiones , Colon/cirugía , Colonoscopía/métodos , Femenino , Perfilación de la Expresión Génica , Inflamación/metabolismo , Inflamación/microbiología , Enfermedades Inflamatorias del Intestino , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Lisofosfolípidos/metabolismo , Masculino , Ratones , Ratones Noqueados , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato
5.
Cancer Prev Res (Phila) ; 9(9): 721-31, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27432344

RESUMEN

Treatment with celecoxib, a selective COX-2 inhibitor, reduces formation of premalignant adenomatous polyps in the gastrointestinal tracts of humans and mice. In addition to its chemopreventive activity, celecoxib can exhibit antimicrobial activity. Differing bacterial profiles have been found in feces from colon cancer patients compared with those of normal subjects. Moreover, preclinical studies suggest that bacteria can modulate intestinal tumorigenesis by secreting specific metabolites. In the current study, we determined whether celecoxib treatment altered the luminal microbiota and metabolome in association with reducing intestinal polyp burden in mice. Administration of celecoxib for 10 weeks markedly reduced intestinal polyp burden in APC(Min/+) mice. Treatment with celecoxib also altered select luminal bacterial populations in both APC(Min/+) and wild-type mice, including decreased Lactobacillaceae and Bifidobacteriaceae as well as increased Coriobacteriaceae Metabolomic analysis demonstrated that celecoxib caused a strong reduction in many fecal metabolites linked to carcinogenesis, including glucose, amino acids, nucleotides, and lipids. Ingenuity Pathway Analysis suggested that these changes in metabolites may contribute to reduced cell proliferation. To this end, we showed that celecoxib reduced cell proliferation in the base of normal appearing ileal and colonic crypts of APC(Min/+) mice. Consistent with this finding, lineage tracing indicated that celecoxib treatment reduced the rate at which Lgr5-positive stem cells gave rise to differentiated cell types in the crypts. Taken together, these results demonstrate that celecoxib alters the luminal microbiota and metabolome along with reducing epithelial cell proliferation in mice. We hypothesize that these actions contribute to its chemopreventive activity. Cancer Prev Res; 9(9); 721-31. ©2016 AACR.


Asunto(s)
Celecoxib/farmacología , Inhibidores de la Ciclooxigenasa 2/farmacología , Microbioma Gastrointestinal/efectos de los fármacos , Pólipos Intestinales/patología , Metaboloma/efectos de los fármacos , Animales , Proliferación Celular/efectos de los fármacos , Heces/química , Heces/microbiología , Masculino , Ratones
6.
Am J Vet Res ; 70(11): 1333-8, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19878015

RESUMEN

OBJECTIVE: To compare the time to desaturation in healthy dogs that breathed oxygen or room air for 3 minutes before induction of anesthesia. ANIMALS: 20 healthy dogs. PROCEDURES: Dogs were sedated with morphine and acepromazine maleate. Dogs received a 3-minute treatment of room air or oxygen (100 mL/kg/min) via face mask. Arterial blood samples were collected before and after treatment to determine PaCO(2), PaO(2), pH, and SaO(2); propofol (6 mg/kg, IV) was injected during a 7-second period, and the dogs were intubated. A lingual pulse oximeter probe was placed. Dogs remained disconnected from the breathing circuit until SpO(2) equaled 90% (desaturation point) and then connected and ventilated until the SpO(2) was >or= 97%. Arterial blood samples were collected and SpO(2) was recorded every 30 seconds for 4 minutes and then every minute until the desaturation point. Times to first breath and the desaturation point were recorded. Data were collected at 0, 5, 30, 60, 90, 120, and 150 seconds. RESULTS: Mean +/- SEM time to desaturation differed significantly between dogs treated with room air (69.6 +/- 10.6 seconds) and oxygen (297.8 +/- 42.0 seconds). Lowest mean PaO(2) and SaO(2) when dogs were breathing room air were 62 +/- 6.3 mm Hg and 82.3 +/- 4%, respectively, at 30 seconds. CONCLUSIONS AND CLINICAL RELEVANCE: Preoxygenation for 3 minutes increased the time to desaturation in healthy dogs sedated with acepromazine and morphine in which anesthesia was induced with propofol.


Asunto(s)
Acepromazina/farmacología , Morfina/farmacología , Oxígeno , Cuidados Preoperatorios/veterinaria , Propofol/farmacología , Acepromazina/administración & dosificación , Anestésicos Intravenosos/administración & dosificación , Anestésicos Intravenosos/farmacología , Animales , Perros , Femenino , Hipnóticos y Sedantes/administración & dosificación , Hipnóticos y Sedantes/farmacología , Masculino , Morfina/administración & dosificación , Propofol/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...