Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Virol ; 97(11): e0119423, 2023 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-37861336

RESUMEN

IMPORTANCE: Severe COVID-19 and post-acute sequelae often afflict patients with underlying co-morbidities. There is a pressing need for highly effective treatment, particularly in light of the emergence of SARS-CoV-2 variants. In a previous study, we demonstrated that DCLK1, a protein associated with cancer stem cells, is highly expressed in the lungs of COVID-19 patients and enhances viral production and hyperinflammatory responses. In this study, we report the pivotal role of DCLK1-regulated mechanisms in driving SARS-CoV-2 replication-transcription processes and pathogenic signaling. Notably, pharmacological inhibition of DCLK1 kinase during SARS-CoV-2 effectively impedes these processes and counteracts virus-induced alternations in global cell signaling. These findings hold significant potential for immediate application in treating COVID-19.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , COVID-19 , Quinasas Similares a Doblecortina , Humanos , Quinasas Similares a Doblecortina/antagonistas & inhibidores , Quinasas Similares a Doblecortina/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , SARS-CoV-2/metabolismo , Transducción de Señal , Replicación Viral/efectos de los fármacos
2.
J Virol ; 96(17): e0096722, 2022 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-35943255

RESUMEN

Host factors play critical roles in SARS-CoV-2 infection-associated pathology and the severity of COVID-19. In this study, we systematically analyzed the roles of SARS-CoV-2-induced host factors, doublecortin-like kinase 1 (DCLK1), and S100A9 in viral pathogenesis. In autopsied subjects with COVID-19 and pre-existing chronic liver disease, we observed high levels of DCLK1 and S100A9 expression and immunosuppressive (DCLK1+S100A9+CD206+) M2-like macrophages and N2-like neutrophils in lungs and livers. DCLK1 and S100A9 expression were rarely observed in normal controls, COVID-19-negative subjects with chronic lung disease, or COVID-19 subjects without chronic liver disease. In hospitalized patients with COVID-19, we detected 2 to 3-fold increased levels of circulating DCLK1+S100A9+ mononuclear cells that correlated with disease severity. We validated the SARS-CoV-2-dependent generation of these double-positive immune cells in coculture. SARS-CoV-2-induced DCLK1 expression correlated with the activation of ß-catenin, a known regulator of the DCLK1 promoter. Gain and loss of function studies showed that DCLK1 kinase amplified live virus production and promoted cytokine, chemokine, and growth factor secretion by peripheral blood mononuclear cells. Inhibition of DCLK1 kinase blocked pro-inflammatory caspase-1/interleukin-1ß signaling in infected cells. Treatment of SARS-CoV-2-infected cells with inhibitors of DCLK1 kinase and S100A9 normalized cytokine/chemokine profiles and attenuated DCLK1 expression and ß-catenin activation. In conclusion, we report previously unidentified roles of DCLK1 in augmenting SARS-CoV-2 viremia, inflammatory cytokine expression, and dysregulation of immune cells involved in innate immunity. DCLK1 could be a potential therapeutic target for COVID-19, especially in patients with underlying comorbid diseases associated with DCLK1 expression. IMPORTANCE High mortality in COVID-19 is associated with underlying comorbidities such as chronic liver diseases. Successful treatment of severe/critical COVID-19 remains challenging. Herein, we report a targetable host factor, DCLK1, that amplifies SARS-CoV-2 production, cytokine secretion, and inflammatory pathways via activation of ß-catenin(p65)/DCLK1/S100A9/NF-κB signaling. Furthermore, we observed in the lung, liver, and blood an increased prevalence of immune cells coexpressing DCLK1 and S100A9, a myeloid-derived proinflammatory protein. These cells were associated with increased disease severity in COVID-19 patients. Finally, we used a novel small-molecule inhibitor of DCLK1 kinase (DCLK1-IN-1) and S100A9 inhibitor (tasquinimod) to decrease virus production in vitro and normalize hyperinflammatory responses known to contribute to disease severity in COVID-19.


Asunto(s)
COVID-19 , Quinasas Similares a Doblecortina , COVID-19/metabolismo , COVID-19/patología , Calgranulina B/metabolismo , Quimiocinas/metabolismo , Citocinas/metabolismo , Quinasas Similares a Doblecortina/antagonistas & inhibidores , Quinasas Similares a Doblecortina/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Leucocitos Mononucleares/metabolismo , Quinolonas/farmacología , SARS-CoV-2 , beta Catenina/metabolismo
3.
Gastroenterology ; 151(1): 152-164.e11, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27059389

RESUMEN

BACKGROUND & AIMS: Core 1- and core 3-derived mucin-type O-linked oligosaccharides (O-glycans) are major components of the colonic mucus layer. Defective forms of colonic O-glycans, such as the Thomsen-nouveau (Tn) antigen, frequently are observed in patients with ulcerative colitis and colorectal cancer, but it is not clear if they contribute to their pathogenesis. We investigated whether and how impaired O-glycosylation contributes to the development of colitis-associated colorectal cancer using mice lacking intestinal core 1- and core 3-derived O-glycans. METHODS: We generated mice that lack core 1- and core 3-derived intestinal O-glycans (DKO mice) and analyzed them, along with mice that singly lack intestinal epithelial core 1 O-glycans (IEC C1galt1(-/-) mice) or core 3 O-glycans (C3Gnt(-/-) mice). Intestinal tissues were collected at different time points and analyzed for levels of mucin and Tn antigen, development of colitis, and tumor formation using imaging, quantitative polymerase chain reaction, immunoblot, and enzyme-linked immunosorbent assay techniques. We also used cellular and genetic approaches, as well as intestinal microbiota depletion, to identify inflammatory mediators and pathways that contribute to disease in DKO and wild-type littermates (controls). RESULTS: Intestinal tissues from DKO mice contained higher levels of Tn antigen and had more severe spontaneous chronic colitis than tissues from IEC C1galt1(-/-) mice, whereas spontaneous colitis was absent in C3GnT(-/-) and control mice. IEC C1galt1(-/-) mice and DKO mice developed spontaneous colorectal tumors, although the onset of tumors in the DKO mice occurred earlier (age, 8-9 months) than that in IEC C1galt1(-/-) mice (15 months old). Antibiotic depletion of the microbiota did not cause loss of Tn antigen but did reduce the development of colitis and cancer formation in DKO mice. Colon tissues from DKO mice, but not control mice, contained active forms of caspase 1 and increased caspase 11, which were reduced after antibiotic administration. Supernatants from colon tissues of DKO mice contained increased levels of interleukin-1ß and interleukin-18, compared with those from control mice. Disruption of the caspase 1 and caspase 11 genes in DKO mice (DKO/Casp1/11(-/-) mice) decreased the development of colitis and cancer, characterized by reduced colonic thickening, hyperplasia, inflammatory infiltrate, and tumors compared with DKO mice. CONCLUSIONS: Impaired expression of O-glycans causes colonic mucus barrier breach and subsequent microbiota-mediated activation of caspase 1-dependent inflammasomes in colonic epithelial cells of mice. These processes could contribute to colitis-associated colon cancer in humans.


Asunto(s)
Antígenos de Carbohidratos Asociados a Tumores/metabolismo , Colitis/complicaciones , Neoplasias Colorrectales/etiología , Mucinas/metabolismo , Polisacáridos/metabolismo , Animales , Colitis/inducido químicamente , Colitis/metabolismo , Microbioma Gastrointestinal/fisiología , Glicosilación , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Ratones , Ratones Noqueados
4.
Gut ; 64(3): 459-68, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24906974

RESUMEN

OBJECTIVE: Commensal bacteria and innate immunity play a major role in the development of colorectal cancer (CRC). We propose that selected commensals polarise colon macrophages to produce endogenous mutagens that initiate chromosomal instability (CIN), lead to expression of progenitor and tumour stem cell markers, and drive CRC through a bystander effect. DESIGN: Primary murine colon epithelial cells were repetitively exposed to Enterococcus faecalis-infected macrophages, or purified trans-4-hydroxy-2-nonenal (4-HNE)-an endogenous mutagen and spindle poison produced by macrophages. CIN, gene expression, growth as allografts in immunodeficient mice were examined for clones and expression of markers confirmed using interleukin (IL) 10 knockout mice colonised by E. faecalis. RESULTS: Primary colon epithelial cells exposed to polarised macrophages or 4-hydroxy-2-nonenal developed CIN and were transformed after 10 weekly treatments. In immunodeficient mice, 8 of 25 transformed clones grew as poorly differentiated carcinomas with 3 tumours invading skin and/or muscle. All tumours stained for cytokeratins confirming their epithelial cell origin. Gene expression profiling of clones showed alterations in 3 to 7 cancer driver genes per clone. Clones also strongly expressed stem/progenitor cell markers Ly6A and Ly6E. Although not differentially expressed in clones, murine allografts positively stained for the tumour stem cell marker doublecortin-like kinase 1. Doublecortin-like kinase 1 and Ly6A/E were expressed by epithelial cells in colon biopsies for areas of inflamed and dysplastic tissue from E. faecalis-colonised IL-10 knockout mice. CONCLUSIONS: These results validate a novel mechanism for CRC that involves endogenous CIN and cellular transformation arising through a microbiome-driven bystander effect.


Asunto(s)
Efecto Espectador , Transformación Celular Neoplásica/metabolismo , Colon/microbiología , Neoplasias del Colon/etiología , Infecciones por Bacterias Grampositivas/complicaciones , Células Madre Neoplásicas/metabolismo , Aldehídos/metabolismo , Animales , Efecto Espectador/fisiología , Línea Celular , Colon/metabolismo , Neoplasias del Colon/metabolismo , Enterococcus faecalis , Femenino , Infecciones por Bacterias Grampositivas/metabolismo , Células HCT116 , Humanos , Hibridación Fluorescente in Situ , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones , Ratones Endogámicos NOD , Trasplante de Neoplasias
5.
Cancer ; 119(12): 2291-9, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23580435

RESUMEN

BACKGROUND: Population-based studies comprehensively describing incidence patterns of human papillomavirus (HPV)-related preinvasive and invasive neoplasms prior to widespread HPV vaccination are sparse. METHODS: Age-adjusted incidence rates (IRs), IR ratios (IRRs), and annual percent changes (APCs) in IRs were calculated for potentially HPV-related tumors diagnosed in the Surveillance, Epidemiology and End Results (SEER) Program during 1978 through 2007. RESULTS: Overall IRs for preinvasive tumors were significantly higher than for invasive squamous cell tumors of cervix (IRR = 3.42), vulva (IRR = 1.87), and vagina (IRR = 1.19) and significantly lower for adenomatous cervical tumors (IRR = 0.43), and squamous cell tumors of penis (IRR = 0.64), anus (males, IRR = 0.53; females, IRR = 0.14), and head and neck (H&N) (males, IRR = 0.01; females, IRR = 0.02). Incidence of preinvasive squamous tumors of cervix, vagina, and penis rose rapidly over time and decreased for invasive neoplasms. The most rapid increases occurred for preinvasive (males, APC = 16.0; females, APC = 7.3) and invasive anal tumors (males, APC = 3.6; females, APC = 2.3). IR patterns were generally similar among evaluable racial/ethnic groups, with the exception of H&N invasive tumor IRs which increased exclusively among white males. CONCLUSIONS: Contrary to the opposing trends of preinvasive and invasive squamous tumors of cervix, vagina, and penis, preinvasive and invasive anal tumor IRs increased significantly over time by sex, age, and racial/ethnic groups. Successful HPV vaccination programs are needed to measurably reduce incidence of HPV-related neoplasms in the future, particularly for cancer sites with rising incidence rates for which effective screening modalities are limited. Cancer 2013;119:2291-2299. © 2013 American Cancer Society.


Asunto(s)
Infecciones por Papillomavirus/epidemiología , Adolescente , Adulto , Anciano , Neoplasias del Ano/epidemiología , Neoplasias del Ano/virología , Carcinoma de Células Escamosas/epidemiología , Carcinoma de Células Escamosas/virología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neoplasias del Pene/epidemiología , Neoplasias del Pene/virología , Programa de VERF , Estados Unidos/epidemiología , Neoplasias del Cuello Uterino/epidemiología , Neoplasias del Cuello Uterino/virología , Neoplasias Vaginales/epidemiología , Neoplasias Vaginales/virología , Neoplasias de la Vulva/epidemiología , Neoplasias de la Vulva/virología , Adulto Joven
6.
Gastroenterology ; 142(3): 543-551.e7, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22108198

RESUMEN

BACKGROUND & AIMS: Enterococcus faecalis is a human intestinal commensal that produces extracellular superoxide and promotes chromosome instability via macrophage-induced bystander effects. We investigated the ability of 4-hydroxy-2-nonenal (4-HNE), a diffusible breakdown product of ω-6 polyunsaturated fatty acids, to mediate these effects. METHODS: 4-HNE was purified from E faecalis-infected macrophages; its genotoxicity was assessed in human colon cancer (HCT116) and primary murine colon epithelial (YAMC) cell lines. RESULTS: 4-HNE induced G(2)-M cell cycle arrest, led to formation γH2AX foci, and disrupted the mitotic spindle in both cell lines. Binucleate tetraploid cells that formed after incubation with 4-HNE were associated with the activation of stathmin and microtubule catastrophe. Silencing glutathione S-transferase α4, a scavenger of 4-HNE, increased the susceptibility of epithelial cells to 4-HNE-induced genotoxicity. Interleukin-10 knockout mice colonized with superoxide-producing E faecalis developed inflammation and colorectal cancer, whereas colonization with a superoxide-deficient strain resulted in inflammation but not cancer. 4-HNE-protein adducts were found in the lamina propria and macrophages in areas of colorectal inflammation. CONCLUSIONS: 4-HNE can act as an autochthonous mitotic spindle poison in normal colonic epithelial and colon cancer cells. This finding links the macrophage-induced bystander effects to colorectal carcinogenesis.


Asunto(s)
Aldehídos/metabolismo , Comunicación Autocrina , Efecto Espectador , Colon/microbiología , Daño del ADN , Enterococcus faecalis/patogenicidad , Células Epiteliales/microbiología , Infecciones por Bacterias Grampositivas/microbiología , Macrófagos/microbiología , Animales , Biopsia , Técnicas de Cocultivo , Colon/metabolismo , Colon/patología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/microbiología , Neoplasias Colorrectales/patología , Modelos Animales de Enfermedad , Enterococcus faecalis/genética , Enterococcus faecalis/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/patología , Puntos de Control de la Fase G2 del Ciclo Celular , Glutatión Transferasa/genética , Glutatión Transferasa/metabolismo , Infecciones por Bacterias Grampositivas/genética , Infecciones por Bacterias Grampositivas/metabolismo , Infecciones por Bacterias Grampositivas/patología , Células HCT116 , Histonas/metabolismo , Humanos , Interleucina-10/deficiencia , Interleucina-10/genética , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Interferencia de ARN , Huso Acromático/metabolismo , Huso Acromático/patología , Estatmina/metabolismo , Tetraploidía , Factores de Tiempo , Transfección
7.
bioRxiv ; 2023 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-38187520

RESUMEN

DNA methylation data has been used to make "epigenetic clocks" which attempt to measure chronological and biological aging. These models rely on data derived from bisulfite-based measurements, which exploit a semi-selective deamination and a genomic reference to determine methylation states. Here, we demonstrate how another hallmark of aging, genomic instability, influences methylation measurements in both bisulfite sequencing and methylation arrays. We found that non-methylation factors lead to "pseudomethylation" signals that are both confounding of epigenetic clocks and uniquely age predictive. Quantifying these covariates in aging studies will be critical to building better clocks and designing appropriate studies of epigenetic aging.

8.
Cancers (Basel) ; 14(11)2022 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-35681791

RESUMEN

Inflammation is an essential hallmark of cancer. Macrophages are key innate immune effector cells in chronic inflammation, parainflammation, and inflammaging. Parainflammation is a form of subclinical inflammation associated with a persistent DNA damage response. Inflammaging represents low-grade inflammation due to the dysregulation of innate and adaptive immune responses that occur with aging. Whether induced by infection, injury, or aging, immune dysregulation and chronic macrophage polarization contributes to cancer initiation through the production of proinflammatory chemokines/cytokines and genotoxins and by modulating immune surveillance. This review presents pre-clinical and clinical evidence for polarized macrophages as endogenous cellular carcinogens in the context of chronic inflammation, parainflammation, and inflammaging. Emerging strategies for cancer prevention, including small molecule inhibitors and probiotic approaches, that target macrophage function and phenotype are also discussed.

9.
Mycoses ; 54(5): e481-5, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21554419

RESUMEN

Invasive mould infections (IMI) are associated with significant morbidity and mortality. In vitro studies have demonstrated that hydroxymethylglutaryl-CoA (HMG-CoA) reductase inhibitors (statins) have activity against several pathogenic moulds including Zygomycetes and Aspergillus spp. The aim of our study was to determine if statin use is a preventive factor for the development of IMI. This was a retrospective case-control study of 10 United States Veterans Affairs Medical Centers that comprise the Veterans Integrated Service Network (VISN) 16. Cases with IMI and controls were identified from 2001 to 2008. Controls were matched by age, facility, history of transplantation, presence of chronic steroid use and presence of human immunodeficiency virus infection (HIV). Two hundred and thirty-eight patients were included. Independent variables associated with the development of IMI were history of solid malignant tumours (OR 2.63, 1.41-4.87) and hypertension (OR 2.29, 1.13-4.68). Statin use within 3 months of index date was not an independent variable for prevention or development of IMI. No level of exposure to a statin drug appeared to influence the development of infection. This retrospective case-control study suggests that despite evidence of in vitro activity, statins may not decrease risk of IMI. Prospective, controlled trials may be necessary to investigate any potential clinical benefit.


Asunto(s)
Anticolesterolemiantes/administración & dosificación , Antifúngicos/administración & dosificación , Quimioprevención/métodos , Micosis/prevención & control , Estudios de Casos y Controles , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Estados Unidos
10.
Dis Model Mech ; 14(5)2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33969420

RESUMEN

Sporadic colorectal cancer (CRC) is a leading cause of worldwide cancer mortality. It arises from a complex milieu of host and environmental factors, including genetic and epigenetic changes in colon epithelial cells that undergo mutation, selection, clonal expansion, and transformation. The gut microbiota has recently gained increasing recognition as an additional important factor contributing to CRC. Several gut bacteria are known to initiate CRC in animal models and have been associated with human CRC. In this Review, we discuss the factors that contribute to CRC and the role of the gut microbiota, focusing on a recently described mechanism for cancer initiation, the so-called microbiota-induced bystander effect (MIBE). In this cancer mechanism, microbiota-driven parainflammation is believed to act as a source of endogenous mutation, epigenetic change and induced pluripotency, leading to the cancerous transformation of colon epithelial cells. This theory links the gut microbiota to key risk factors and common histologic features of sporadic CRC. MIBE is analogous to the well-characterized radiation-induced bystander effect. Both phenomena drive DNA damage, chromosomal instability, stress response signaling, altered gene expression, epigenetic modification and cellular proliferation in bystander cells. Myeloid-derived cells are important effectors in both phenomena. A better understanding of the interactions between the gut microbiota and mucosal immune effector cells that generate bystander effects can potentially identify triggers for parainflammation, and gain new insights into CRC prevention.


Asunto(s)
Efecto Espectador , Carcinogénesis/patología , Neoplasias Colorrectales/microbiología , Microbioma Gastrointestinal , Inflamación/patología , Animales , Neoplasias Colorrectales/complicaciones , Neoplasias Colorrectales/genética , Disbiosis/complicaciones , Disbiosis/microbiología , Humanos , Inflamación/complicaciones
11.
Breast Cancer Res ; 12(4): R54, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20653964

RESUMEN

INTRODUCTION: Adenoid cystic carcinoma of the breast (breast-ACC) is a rare and special type of basal-like tumor for which scant population-based descriptive data exist. We sought to provide new population-based information on breast-ACC incidence, relative survival, and associated cancer risk in the United States. METHODS: Using data from the Surveillance, Epidemiology and End Results Program, we calculated age-adjusted incidence rates (IRs), IR ratios (IRRs), and relative survival for breast-ACC, and standardized incidence ratios (SIRs) for other cancers. RESULTS: Overall 338 women (IR = 0.92/1 million person-years) were diagnosed with breast-ACC during 1977 to 2006. Blacks had 39% lower IRs than Whites (IRR = 0.61, 95% confidence interval = 0.37 to 0.96), and IRs remained constant over the 30-year period. Ninety-five percent of cases presented with localized stage (n = 320; IR = 0.87), and the highest IRs were observed for estrogen receptor (ER)-negative/progesterone receptor (PR)-negative tumors (IR = 0.56). Like other typically ER-negative tumors, age-specific IRs increased until midlife and then plateaued. Five-year, 10-year, and 15-year relative survival was 98.1%, 94.9%, and 91.4%, respectively. The risk of female breast cancer was not increased following (SIR = 0.89, 95% confidence interval = 0.43 to 1.64) or preceding (SIR = 0.71, 95% confidence interval = 0.28 to 1.46) breast-ACC. Similarly, no association was observed for breast-ACC and risk of all other cancers combined, solid tumors, or lymphohematopoietic malignancies. CONCLUSIONS: Breast-ACC among women is characterized by ER-negative/PR-negative expression, rare regional lymph node involvement, a favorable prognosis with excellent survival, and absence of associated cancers. These findings reinforce the importance of tailored treatments for breast-ACC and lend credence to the apparent heterogeneity of basal-like breast cancers.


Asunto(s)
Neoplasias de la Mama/epidemiología , Carcinoma Adenoide Quístico/epidemiología , Programa de VERF/estadística & datos numéricos , Adolescente , Adulto , Negro o Afroamericano/estadística & datos numéricos , Anciano , Estudios de Cohortes , Femenino , Humanos , Incidencia , Persona de Mediana Edad , Análisis de Supervivencia , Estados Unidos/epidemiología , Población Blanca/estadística & datos numéricos , Adulto Joven
12.
Am J Pathol ; 174(1): 317-29, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19119184

RESUMEN

Alterations in genes encoding transforming growth factor-beta-signaling components contribute to colon cancer in humans. Similarly, mice deficient in the transforming growth factor-beta signaling molecule, Smad3, develop colon cancer, but only after a bacterial trigger occurs, resulting in chronic inflammation. To determine whether Smad3-null lymphocytes contribute to increased cancer susceptibility, we crossed Smad3-null mice with mice deficient in both B and T lymphocytes (Rag2(-/-) mice). Helicobacter-infected Smad3/Rag2-double knockout (DKO) mice had more diffuse inflammation and increased incidence of adenocarcinoma compared with Helicobacter-infected Smad3(-/-) or Rag2(-/-) mice alone. Adoptive transfer of WT CD4(+)CD25(+) T-regulatory cells provided significant protection of Smad3/Rag2-DKO from bacterial-induced typhlocolitis, dysplasia, and tumor development, whereas Smad3(-/-) T-regulatory cells provided no protection. Immunohistochemistry, real-time reverse transcriptase-polymerase chain reaction, and Western blot analyses of colonic tissues from Smad3/Rag2-DKO mice 1 week after Helicobacter infection revealed an influx of macrophages, enhanced nuclear factor-kappaB activation, increased Bcl(XL)/Bcl-2 expression, increased c-Myc expression, accentuated epithelial cell proliferation, and up-regulated IFN-gamma, IL-1alpha, TNF-alpha, IL-1beta, and IL-6 transcription levels. These results suggest that the loss of Smad3 increases susceptibility to colon cancer by at least two mechanisms: deficient T-regulatory cell function, which leads to excessive inflammation after a bacterial trigger; and increased expression of proinflammatory cytokines, enhanced nuclear factor-kappaB activation, and increased expression of both pro-oncogenic and anti-apoptotic proteins that result in increased cell proliferation/survival of epithelial cells in colonic tissues.


Asunto(s)
Adenocarcinoma/genética , Neoplasias del Colon/genética , Proteínas de Unión al ADN/deficiencia , Infecciones por Helicobacter/complicaciones , Proteína smad3/deficiencia , Factor de Crecimiento Transformador beta/metabolismo , Adenocarcinoma/inmunología , Adenocarcinoma/microbiología , Animales , Western Blotting , Neoplasias del Colon/inmunología , Neoplasias del Colon/microbiología , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Citometría de Flujo , Infecciones por Helicobacter/inmunología , Inmunohistoquímica , Inflamación/inmunología , Inflamación/microbiología , Inflamación/patología , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína smad3/genética , Linfocitos T Reguladores/inmunología
13.
J Am Acad Dermatol ; 63(1): 71-8, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20447723

RESUMEN

BACKGROUND: Primary cutaneous adenoid cystic carcinoma (PCACC) is a rare appendageal tumor of uncertain origin. Details on epidemiologic features of PCACC are sparse and largely based on clinical reports. OBJECTIVE: We sought to develop an understanding of PCACC incidence, survival, and associated cancers using population-based data. METHODS: We used the Surveillance, Epidemiology, and End Results program to calculate age-adjusted incidence rates (IRs), IR ratios, 95% confidence intervals, standardized incidence ratios (SIRs), and 5-year relative survival of PCACC diagnosed during 1976 to 2005. RESULTS: In a population of 723,174,580 person-years, the overall PCACC IR was 0.23 per 1 million person-years (n = 152), with similar IRs among male and female patients (IR = 0.24). Most cases of PCACC presented at a localized stage and arose on the face/head/neck. Among 122 of the 2-month survivors of PCACC and more than 2.4 million 2-month cancer survivors, risk of associated cancers overall was not significantly increased (SIR = 1.17 [n = 24] and SIR = 1.43 [n = 16], respectively). However, PCACC was associated with significantly increased risks of subsequent lymphohematopoietic (n = 6; SIR = 3.70) and thyroid (n = 2; SIR = 15.25) cancers, whereas the converse associations were not observed. Five-year relative survival was excellent (96.1%; n = 122) with more favorable survival noted for PCACC involving the face/head/neck than the trunk. LIMITATIONS: A pathologic review of reported cases was not undertaken. CONCLUSION: PCACC is a rare appendageal tumor that affects male and female individuals equally, primarily presents at localized stage, predominates in the face/head/neck, and is associated with favorable survival. Immunosuppression does not appear to contribute to the development of PCACC, and the observed associated cancer patterns will need to be confirmed in larger studies.


Asunto(s)
Carcinoma Adenoide Quístico/epidemiología , Neoplasias Cutáneas/epidemiología , Carcinoma Adenoide Quístico/mortalidad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neoplasias Primarias Múltiples/epidemiología , Neoplasias Cutáneas/mortalidad , Tasa de Supervivencia , Estados Unidos/epidemiología
14.
Food Res Int ; 129: 108788, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32036912

RESUMEN

Probiotics are naturally occurring microorganisms that confer health benefits by altering host commensal microbiota, modulating immunity, enhancing intestinal barrier function, or altering pain perception. Enterococci are human and animal intestinal commensals that are used as probiotics and in food production. These microorganisms, however, express many virulence traits including cytolysin, proteases, aggregation substance, capsular polysaccharide, enterococcal surface protein, biofilm formation, extracellular superoxide, intestinal translocation, and resistance to innate immunity that can lead to serious hospital-acquired infections. In addition, enterococci are facile in acquiring antibiotic resistance genes to many clinically important antibiotics encoded on a wide variety of conjugative plasmids, transposons, and bacteriophages. The pathogenicity and disease burden caused by enterococci render them poor choices as probiotics. No large, randomized, placebo-controlled clinical trials have demonstrated the safety and efficacy of any enterococcal probiotic. As a result, no enterococcal probiotic has been approved by the United States Food and Drug Administration for the treatment, cure, or amelioration of human disease. In 2007, the European Food Safety Authority concluded that enterococci do not meet the standard for "Qualified Presumption of Safety". Enterococcal strains used or proposed for use as probiotics should be carefully screened for efficacy and safety.


Asunto(s)
Enterococcus/metabolismo , Contaminación de Alimentos/análisis , Probióticos/efectos adversos , Citotoxinas/genética , Citotoxinas/metabolismo , Farmacorresistencia Microbiana/genética , Enterococcus/genética , Enterococcus/aislamiento & purificación , Enterococcus faecalis/aislamiento & purificación , Enterococcus faecalis/metabolismo , Microbiología de Alimentos , Inocuidad de los Alimentos , Sitios Genéticos , Inmunidad Innata , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Perforina/genética , Perforina/metabolismo , Factores de Riesgo , Estados Unidos , United States Food and Drug Administration , Factores de Virulencia/genética
15.
Sci Rep ; 10(1): 10578, 2020 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-32601309

RESUMEN

Chronic liver injury is a risk factor for cirrhosis and hepatocellular carcinoma (HCC). The molecular mechanisms that regulate the decision between normal injury repair and neoplastic initiation are unclear. Doublecortin-like kinase 1 (DCLK1), a tumor stem cell marker, is induced during cirrhosis and HCC. Here, we demonstrate that DCLK1-overexpressing primary human hepatocytes formed spheroids in suspension cultures. Spheroids derived from DCLK1-overexpressing hepatoma cells showed high level expression of active ß-catenin, α-fetoprotein, and SOX9, suggesting that DCLK1 overexpression induces clonogenicity and dedifferentiated phenotypes in hepatoma cells. DCLK1 overexpression in hepatoma cells also increased phosphorylation of GSK-3ß at Ser9. This was associated with an induction of a 48-kDa active ß-catenin with a preserved hypophosphorylated N-terminus that interacted with nuclear TCF-4 resulting in luciferase reporter activity and cyclin D1 expression. DCLK1 downregulation inhibited 48-kDa ß-catenin expression. The proteasome inhibitor bortezomib did not block the 48-kDa ß-catenin, instead, caused a threefold accumulation, suggesting a proteasome-independent mechanism. Liver tissues from patients with cirrhosis and HCC revealed epithelial co-staining of DCLK1 and active ß-catenin, and cleaved E-cadherin. Repopulated DCLK1-overexpressing primary human hepatocytes in humanized FRG mouse livers demonstrated active ß-catenin. In conclusion, DCLK1 regulates oncogenic signaling and clonogenicity of hepatocytes by a novel non-canonical/atypical ß-catenin-dependent mechanism.


Asunto(s)
Hepatocitos/citología , Hepatocitos/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , beta Catenina/metabolismo , Animales , Carcinogénesis , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Quinasas Similares a Doblecortina , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Células Hep G2 , Hepatocitos/enzimología , Hepatocitos/patología , Xenoinjertos , Humanos , Cirrosis Hepática/patología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Ratones , Ratones Endogámicos C57BL , Células Madre Neoplásicas/metabolismo , Factor de Transcripción SOX9/metabolismo , Esferoides Celulares , alfa-Fetoproteínas/metabolismo
16.
Sci Transl Med ; 11(487)2019 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-30971455

RESUMEN

Multidrug-resistant enterococcal strains emerged in the early 1980s and are now among the leading causes of drug-resistant bacterial infection worldwide. We used functional genomics to study an early bacterial outbreak in patients in a Wisconsin hospital between 1984 and 1988 that was caused by multidrug-resistant Enterococcus faecalis The goal was to determine how a clonal lineage of E. faecalis became adapted to growth and survival in the human bloodstream. Genome sequence analysis revealed a progression of increasingly fixed mutations and repeated independent occurrences of mutations in a relatively small set of genes. Repeated independent mutations suggested selection within the host during the course of infection in response to pressures such as host immunity and antibiotic treatment. We observed repeated independent mutations in a small number of loci, including a little studied polysaccharide utilization pathway and the cydABDC locus. Functional studies showed that mutating these loci rendered E. faecalis better able to withstand antibiotic pressure and innate immune defenses in the human bloodstream. We also observed a shift in mutation pattern that corresponded to the introduction of carbapenem antibiotics in 1987. This work identifies pathways that allow enterococci to survive the transition from the human gut into the bloodstream, enabling them to cause severe bacteremia associated with high mortality.


Asunto(s)
Adaptación Fisiológica , Antibacterianos/farmacología , Enterococcus faecium/fisiología , Infecciones por Bacterias Grampositivas/sangre , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata/efectos de los fármacos , Adaptación Fisiológica/efectos de los fármacos , Carbapenémicos/farmacología , Brotes de Enfermedades , Farmacorresistencia Microbiana/efectos de los fármacos , Enterococcus faecium/genética , Sitios Genéticos , Variación Genética , Genoma Bacteriano , Infecciones por Bacterias Grampositivas/microbiología , Hospitales , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Lipopolisacáridos/metabolismo , Mutación/genética , N-Glicosil Hidrolasas/metabolismo , Operón/genética , Estrés Fisiológico/efectos de los fármacos , Ácidos Teicoicos/metabolismo
17.
J Med Microbiol ; 57(Pt 10): 1193-1204, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18809545

RESUMEN

Enterococcus faecalis is an intestinal commensal that cannot synthesize porphyrins and only expresses a functional respiratory chain when provided with exogenous haematin. In the absence of haematin, E. faecalis reverts to fermentative metabolism and produces extracellular superoxide that can damage epithelial-cell DNA. The acute response of the colonic mucosa to haematin-starved E. faecalis was identified by gene array. E. faecalis was inoculated into murine colons using a surgical ligation model that preserved tissue architecture and homeostasis. The mucosa was exposed to haematin-starved E. faecalis and compared with a control consisting of the same strain grown with haematin. At 1 h post-inoculation, 6 mucosal genes were differentially regulated and this increased to 42 genes at 6 h. At 6 h, a highly significant biological interaction network was identified with functions that included nuclear factor-kappaB (NF-kappaB) signalling, apoptosis and cell-cycle regulation. Colon biopsies showed no histological abnormalities by haematoxylin and eosin staining. Immunohistochemical staining, however, detected NF-kappaB activation in tissue macrophages using antibodies to the nuclear localization sequence for p65 and the F4/80 marker for murine macrophages. Similarly, haematin-starved E. faecalis strongly activated NF-kappaB in murine macrophages in vitro. Furthermore, primary and transformed colonic epithelial cells activated the G2/M checkpoint in vitro following exposure to haematin-starved E. faecalis. Modulation of this cell-cycle checkpoint was due to extracellular superoxide produced as a result of the respiratory block in haematin-starved E. faecalis. These results demonstrate that the uniquely dichotomous metabolism of E. faecalis can significantly modulate gene expression in the colonic mucosa for pathways associated with inflammation, apoptosis and cell-cycle regulation.


Asunto(s)
Colon/metabolismo , Colon/microbiología , Enterococcus faecalis/metabolismo , Regulación de la Expresión Génica/fisiología , Hemina , Animales , Ciclo Celular/fisiología , Línea Celular Tumoral , Colon/citología , Enterococcus faecalis/genética , Humanos , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , FN-kappa B/genética , FN-kappa B/metabolismo , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/metabolismo , Netrina-1 , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
18.
Oncotarget ; 8(60): 102176-102190, 2017 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-29254234

RESUMEN

The colonic microbiome contributes to the initiation of colorectal cancer through poorly characterized mechanisms. We have shown that commensal-polarized macrophages induce gene mutation, chromosomal instability, and endogenous transformation through microbiome-induced bystander effects (MIBE). In this study we show that MIBE activates Wnt/ß-catenin signaling and pluripotent transcription factors associated with dedifferentiation, reprogramming, and the development of colorectal cancer stem cells (CSCs). Exposure of murine primary colon epithelial cells (YAMC) to Enterococcus faecalis-infected macrophages increased Wnt3α expression while suppressing Wnt inhibitor factor 1 (Wif1). Wnt/ß-catenin activation was confirmed by increased active ß-catenin and Tcf4. in vivo, active ß-catenin was evident in colon biopsies from E. faecalis-colonized Il10 knockout mice compared to sham-colonized mice. This effect was mediated, in part, by 4-hydroxy-2-nonenal and tumor necrosis factor α. MIBE also activated pluripotent transcription factors c-Myc, Klf4, Oct4, and Sox2 in YAMC cells and colons from E. faecalis-colonized Il10 knockout mice. These transcription factors are associated with cellular reprogramming, dedifferentiation, and induction of colorectal CSC progenitors. In support of this was an increase in the expression of Dclk1 and CD44, two colorectal CSC markers, in YAMC cells that were exposed to MIBE. Finally, compared to normal colon biopsies and hyperplastic polyps, DCLK1 expression increased in human tubular adenomas and invasive colorectal cancers. Blocking ß-catenin/TCF4 signaling using FH535 and CTNNB1-specific small interfering RNA decreased DCLK1 expression in HCT116 human colon cancer cells. These findings provide mechanism for microbiome-induced colorectal cancer and identify new potential targets for colorectal cancer prevention.

19.
Free Radic Biol Med ; 105: 3-15, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27810411

RESUMEN

Colorectal cancer (CRC) is a leading cause of cancer death and archetype for cancer as a genetic disease. However, the mechanisms for genetic change and their interactions with environmental risk factors have been difficult to unravel. New hypotheses, models, and methods are being used to investigate a complex web of risk factors that includes the intestinal microbiome. Recent research has clarified how the microbiome can generate genomic change in CRC. Several phenotypes among a small group of selected commensals have helped us better understand how mutations and chromosomal instability (CIN) are induced in CRC (e.g., toxin production, metabolite formation, radical generation, and immune modulation leading to a bystander effect). This review discusses recent hypotheses, models, and mechanisms by which the intestinal microbiome contributes to the initiation and progression of sporadic and colitis-associated forms of CRC. Overall, it appears the microbiome can initiate and/or promote CRC at all stages of tumorigenesis by acting as an inducer of DNA damage and CIN, regulating cell growth and death, generating epigenetic changes, and modulating host immune responses. Understanding how the microbiome interacts with other risk factors to define colorectal carcinogenesis will ultimately lead to more accurate risk prediction. A deeper understanding of CRC etiology will also help identify new targets for prevention and treatment and help accelerate the decline in mortality for this common cancer.


Asunto(s)
Neoplasias Colorrectales/microbiología , Microbioma Gastrointestinal/inmunología , Animales , Transformación Celular Neoplásica/inmunología , Inestabilidad Cromosómica , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/inmunología , Daño del ADN , Enterobacteriaceae/inmunología , Enterobacteriaceae/fisiología , Transición Epitelial-Mesenquimal , Humanos , Células Madre Neoplásicas/microbiología , Factores de Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA