Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 164
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Dig Dis Sci ; 69(2): 562-569, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38135813

RESUMEN

BACKGROUND: Long-COVID is a condition post SARS-CoV-2 infection with persistent or recurring symptoms affecting multiple organs, and may involve viral persistence, changes to the microbiome, coagulopathies, and alterations to neuro-immune interactions. These factors can disrupt the Gut-Brain Axis, which is a complex system involving bidirectional communication between the central nervous system and the gastrointestinal (GI) system. As a result of these disruptions, individuals with long-COVID may develop post-infectious functional GI disorders, which can cause a range of symptoms affecting the digestive system. AIM: To understand frequency of GI manifestations of Long-COVID and to determine association with sleep or neurological symptoms in a predominantly minority population. METHODS: We included patients with positive SARS-CoV-2 PCR (n = 747) who were hospitalized from Feb. 2020 to May 2021 at Howard University Hospital and followed between 6 and 12 months from discharge. GI, sleep, and neurological symptoms (via the Montreal Cognitive Assessment (MoCA) scoring system) were assessed using a standardized questionnaire. Linear regression analysis, χ2 and Fisher's exact test were utilized to determine the statistical significance of correlations of GI/Neuro/COVID. RESULTS: The mean age of patients was 58, with 51.6% females and a predominant African American ethnicity (73.6%, n = 550). A total of 108 patients died during their initial hospital stay, with the remaining 639 patients followed-up. Three hundred fifty (350) patients responded to the questionnaire (57 patients died during the follow-up period). Overall, 39 (13.3%) patients reported GI-related symptoms, out of which 19 (6.4%) had persistent symptoms and 20 (6.8%) developed new onset GI symptoms. Nausea and vomiting were the most common 24/39 (61.5%), followed by abdominal pain 7/39 (18%), diarrhea 5/39 (12.8%), and others 3/39 (7.6%). Patients who presented with vomiting during acute SARS-CoV-2 infection were more likely to have Long-COVID GI manifestations (P = 0.023). Use of ACE inhibitors, abnormal lymphocyte count and elevated ferritin are other variables that showed significant associations with Long-COVID GI manifestations (P = 0.03, 0.006 and 0.03, respectively). During follow-up, a total of 28 (9.5%) patients reported difficulty with sleep and 79 (27%) patients had abnormal MoCA assessment. With further analysis, there was a trend between presentation of GI symptoms on admission with abnormal MoCA assessment, and an association between abnormal LFTs and history of liver disease during hospitalization with subsequent sleep problems. Baseline characteristics, clinical comorbidities, other laboratory values, hospital length of stay, mechanical ventilation, medications during hospitalization, re-admission and Flu or COVID-19 vaccination have not shown any association with Long-COVID GI symptoms in our cohort. CONCLUSION: Dyspeptic symptoms were common GI manifestations in the acute and post COVID periods. GI symptoms, abnormal LFTs and a history of liver disease during the acute infectious phase associates with abnormal MoCA and sleep problems during follow-up. Further large population studies are needed to determine if COVID-19 leads to a GI symptoms-associated Long-COVID phenotypes and other symptoms through the Gut-Brain-Axis.


Asunto(s)
COVID-19 , Enfermedades Gastrointestinales , Hepatopatías , Trastornos del Sueño-Vigilia , Femenino , Humanos , Masculino , COVID-19/complicaciones , COVID-19/diagnóstico , COVID-19/epidemiología , SARS-CoV-2 , Estudios de Seguimiento , Síndrome Post Agudo de COVID-19 , Estudios Prospectivos , Vacunas contra la COVID-19 , Enfermedades Gastrointestinales/diagnóstico , Enfermedades Gastrointestinales/epidemiología , Enfermedades Gastrointestinales/etiología , Hepatopatías/complicaciones , Vómitos , Trastornos del Sueño-Vigilia/etiología , Trastornos del Sueño-Vigilia/complicaciones
2.
Dig Dis Sci ; 68(9): 3504-3513, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37402979

RESUMEN

In this installment of the "Paradigm Shifts in Perspective" series, the authors, all scientists who have been involved in colorectal cancer (CRC) research for most or all of their careers, have watched the field develop from early pathological descriptions of tumor formation to the current understanding of tumor pathogenesis that informs personalized therapies. We outline how our understanding of the pathogenetic basis of CRC began with seemingly isolated discoveries-initially with the mutations in RAS and the APC gene, the latter of which was initially found in the context of intestinal polyposis, to the more complex process of multistep carcinogenesis, to the chase for tumor suppressor genes, which led to the unexpected discovery of microsatellite instability (MSI). These discoveries enabled the authors to better understand how the DNA mismatch repair (MMR) system not only recognizes DNA damage but also responds to damage by DNA repair or by triggering apoptosis in the injured cell. This work served, in part, to link the earlier findings on the pathogenesis of CRC to the development of immune checkpoint inhibitors, which has been transformative-and curative-for certain types of CRCs and other cancers as well. These discoveries also highlight the circuitous routes that scientific progress takes, which can include thoughtful hypothesis testing and at other times recognizing the importance of seemingly serendipitous observations that substantially change the flow and direction of the discovery process. What has happened over the past 37 years was not predictable when this journey began, but it does speak to the power of careful scientific experimentation, following the facts, perseverance in the face of opposition, and the willingness to think outside of established paradigms.


Asunto(s)
Neoplasias Colorrectales Hereditarias sin Poliposis , Neoplasias Colorrectales , Humanos , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/terapia , Neoplasias Colorrectales Hereditarias sin Poliposis/genética , Mutación , Inestabilidad de Microsatélites , Reparación de la Incompatibilidad de ADN/genética
3.
Annu Rev Med ; 71: 59-69, 2020 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-31451044

RESUMEN

Fecal (or stool) DNA examination is a noninvasive strategy recommended by several medical professional societies for colorectal cancer (CRC) screening in average-risk individuals. Fecal DNA tests assay stool for human DNA shed principally from the colon. Colonic lesions such as adenomatous and serrated polyps and cancers exfoliate cells containing neoplastically altered DNA that may be detected by sensitive assays that target specific genetic and epigenetic biomarkers to discriminate neoplastic lesions from non-neoplastic tissue. Cross-sectional validation studies confirmed initial case-control studies' assessment of performance of an optimized multitarget stool DNA (mt-sDNA) test, leading to approval by the US Food and Drug Administration in 2014. Compared to colonoscopy, mt-sDNA showed sensitivity of 92% for detection of CRC, much higher than the 74% sensitivity of another recommended noninvasive strategy, fecal immunochemical testing (FIT). Detections of advanced adenomas and sessile serrated polyps were higher with mt-sDNA than FIT (42% versus 24% and 42% versus 5%, respectively), but overall specificity for all lesions was lower (87% versus 95%). The mt-sDNA test increases patient life-years gained in CRC screening simulations, but its cost relative to other screening strategies needs to be reduced by 80-90% or its sensitivity for polyp detection enhanced to be cost effective. Noninvasive CRC screening strategies such as fecal DNA, however, have the potential to significantly increase national screening rates due to their noninvasive nature and convenience for patients.


Asunto(s)
Pólipos del Colon/patología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/prevención & control , ADN de Neoplasias/análisis , Detección Precoz del Cáncer/métodos , Heces , Anciano , Análisis Costo-Beneficio , Estudios Transversales , Detección Precoz del Cáncer/economía , Femenino , Humanos , Masculino , Persona de Mediana Edad , Lesiones Precancerosas/patología , Sensibilidad y Especificidad
4.
Annu Rev Med ; 71: 85-102, 2020 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-31756137

RESUMEN

The prevalence of genetic predisposition to cancer is greater than initially appreciated, yet most affected individuals remain undiagnosed. Deleterious germline variants in cancer predisposition genes are implicated in 1 in 10 cases of advanced cancer. Next-generation sequencing technologies have made germline and tumor DNA sequencing more accessible and less expensive. Expanded access to clinical genetic testing will improve identification of individuals with genetic predisposition to cancer and provide opportunities to effectively reduce morbidity through precision cancer therapies and surveillance. Cross-disciplinary clinical education in genomic medicine is needed to translate advances in genomic medicine into improved health outcomes.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias Colorrectales/genética , Predisposición Genética a la Enfermedad/epidemiología , Pruebas Genéticas/métodos , Mutación de Línea Germinal/genética , Neoplasias Ováricas/genética , Neoplasias de la Mama/epidemiología , Neoplasias de la Mama/patología , Neoplasias Colorrectales/epidemiología , Neoplasias Colorrectales/patología , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Incidencia , Masculino , National Institutes of Health (U.S.) , Neoplasias Ováricas/epidemiología , Neoplasias Ováricas/patología , Pronóstico , Medición de Riesgo , Análisis de Supervivencia , Estados Unidos
6.
BMC Infect Dis ; 22(1): 552, 2022 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-35715729

RESUMEN

BACKGROUND AND AIMS: Initial reports on US COVID-19 showed different outcomes in different races. In this study we use a diverse large cohort of hospitalized COVID-19 patients to determine predictors of mortality. METHODS: We analyzed data from hospitalized COVID-19 patients (n = 5852) between March 2020- August 2020 from 8 hospitals across the US. Demographics, comorbidities, symptoms and laboratory data were collected. RESULTS: The cohort contained 3,662 (61.7%) African Americans (AA), 286 (5%) American Latinx (LAT), 1,407 (23.9%), European Americans (EA), and 93 (1.5%) American Asians (AS). Survivors and non-survivors mean ages in years were 58 and 68 for AA, 58 and 77 for EA, 44 and 61 for LAT, and 51 and 63 for AS. Mortality rates for AA, LAT, EA and AS were 14.8, 7.3, 16.3 and 2.2%. Mortality increased among patients with the following characteristics: age, male gender, New York region, cardiac disease, COPD, diabetes mellitus, hypertension, history of cancer, immunosuppression, elevated lymphocytes, CRP, ferritin, D-Dimer, creatinine, troponin, and procalcitonin. Use of mechanical ventilation (p = 0.001), shortness of breath (SOB) (p < 0.01), fatigue (p = 0.04), diarrhea (p = 0.02), and increased AST (p < 0.01), significantly correlated with death in multivariate analysis. Male sex and EA and AA race/ethnicity had higher frequency of death. Diarrhea was among the most common GI symptom amongst AAs (6.8%). When adjusting for comorbidities, significant variables among the demographics of study population were age (over 45 years old), male sex, EA, and patients hospitalized in New York. When adjusting for disease severity, significant variables were age over 65 years old, male sex, EA as well as having SOB, elevated CRP and D-dimer. Glucocorticoid usage was associated with an increased risk of COVID-19 death in our cohort. CONCLUSION: Among this large cohort of hospitalized COVID-19 patients enriched for African Americans, our study findings may reflect the extent of systemic organ involvement by SARS-CoV-2 and subsequent progression to multi-system organ failure. High mortality in AA in comparison with LAT is likely related to high frequency of comorbidities and older age among AA. Glucocorticoids should be used carefully considering the poor outcomes associated with it. Special focus in treating patients with elevated liver enzymes and other inflammatory biomarkers such as CRP, troponin, ferritin, procalcitonin, and D-dimer are required to prevent poor outcomes.


Asunto(s)
COVID-19 , Negro o Afroamericano , Anciano , Biomarcadores , Diarrea , Ferritinas , Hospitalización , Humanos , Masculino , Persona de Mediana Edad , Polipéptido alfa Relacionado con Calcitonina , Estudios Retrospectivos , SARS-CoV-2 , Troponina
7.
Br J Cancer ; 124(2): 315-332, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32901135

RESUMEN

There are well-established disparities in cancer incidence and outcomes by race/ethnicity that result from the interplay between structural, socioeconomic, socio-environmental, behavioural and biological factors. However, large research studies designed to investigate factors contributing to cancer aetiology and progression have mainly focused on populations of European origin. The limitations in clinicopathological and genetic data, as well as the reduced availability of biospecimens from diverse populations, contribute to the knowledge gap and have the potential to widen cancer health disparities. In this review, we summarise reported disparities and associated factors in the United States of America (USA) for the most common cancers (breast, prostate, lung and colon), and for a subset of other cancers that highlight the complexity of disparities (gastric, liver, pancreas and leukaemia). We focus on populations commonly identified and referred to as racial/ethnic minorities in the USA-African Americans/Blacks, American Indians and Alaska Natives, Asians, Native Hawaiians/other Pacific Islanders and Hispanics/Latinos. We conclude that even though substantial progress has been made in understanding the factors underlying cancer health disparities, marked inequities persist. Additional efforts are needed to include participants from diverse populations in the research of cancer aetiology, biology and treatment. Furthermore, to eliminate cancer health disparities, it will be necessary to facilitate access to, and utilisation of, health services to all individuals, and to address structural inequities, including racism, that disproportionally affect racial/ethnic minorities in the USA.


Asunto(s)
Disparidades en el Estado de Salud , Grupos Minoritarios/estadística & datos numéricos , Neoplasias/etnología , Etnicidad/estadística & datos numéricos , Femenino , Humanos , Masculino , Estados Unidos/etnología
8.
Gastroenterology ; 158(2): 354-367, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31682851

RESUMEN

Colorectal cancer (CRC) disproportionately affects people from low socioeconomic backgrounds and some racial minorities. Disparities in CRC incidence and outcomes might result from differences in exposure to risk factors such as unhealthy diet and sedentary lifestyle; limited access to risk-reducing behaviors such as chemoprevention, screening, and follow-up of abnormal test results; or lack of access to high-quality treatment resources. These factors operate at the individual, provider, health system, community, and policy levels to perpetuate CRC disparities. However, CRC disparities can be eliminated. Addressing the complex factors that contribute to development and progression of CRC with multicomponent, adaptive interventions, at multiple levels of the care continuum, can reduce gaps in mortality. These might be addressed with a combination of health care and community-based interventions and policy changes that promote healthy behaviors and ensure access to high-quality and effective measures for CRC prevention, diagnosis, and treatment. Improving resources and coordinating efforts in communities where people of low socioeconomic status live and work would increase access to evidence-based interventions. Research is also needed to understand the role and potential mechanisms by which factors in diet, intestinal microbiome, and/or inflammation contribute to differences in colorectal carcinogenesis. Studies of large cohorts with diverse populations are needed to identify epidemiologic and molecular factors that contribute to CRC development in different populations.


Asunto(s)
Neoplasias Colorrectales/epidemiología , Disparidades en el Estado de Salud , Disparidades en Atención de Salud/estadística & datos numéricos , Factores Socioeconómicos , Neoplasias Colorrectales/diagnóstico , Neoplasias Colorrectales/prevención & control , Detección Precoz del Cáncer/estadística & datos numéricos , Accesibilidad a los Servicios de Salud , Estilo de Vida Saludable , Humanos , Incidencia , Tamizaje Masivo/estadística & datos numéricos , Grupos Minoritarios/estadística & datos numéricos , Factores de Riesgo
9.
BMC Cancer ; 20(1): 350, 2020 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-32334542

RESUMEN

BACKGROUND: ARID1A has been described as a tumor suppressor gene, participating in chromatin re-modeling, epithelial-mesenchymal-transition and many other cellular and molecular processes. It has been cited as a contribute in tumorigenesis. The role of ARID1A in CRC is not yet defined. AIM: To investigate the role of ARID1A methylation and CNV in its expression in CRC cell lines and to examine the relationship between ARID1A status with survival and clinicopathologic characteristics in patients with CRC. METHODS: We used RT-PCR to determine both CNV and expression of ARID1A from six CRC cell lines. We used MSP to evaluate methylation of ARID1A. IHC was used to assess ARID1A protein expression. We also evaluated MSI and EMAST status in 18 paired CRC and adjacent normal tissues. 5AzadC was used to assess effect of DNA demethylation on ARID1A expression. Statistical analysis was performed to establish correlations between ARID1A expression and other parameters. RESULTS: Among the 18 CRC tumors studied, 7 (38.8%) and 5 tumors (27.7%) showed no or low ARID1A expression, respectively. We observed no significant difference in ARID1A expression for overall patient survival, and no difference between clinicopathological parameters including MSI and EMAST. However, lymphatic invasion was more pronounced in the low/no ARID1A expression group when compared to moderate and high expression group (33% VS. 16.6% respectively. ARID1A promoter methylation was observed in 4/6 (66%) cell lines and correlated with ARID1A mRNA expression level ranging from very low in SW48, to more pronounced in HCT116 and HT-29/219. Treatment with the methyltransferase inhibitor 5-Azacytidine (5-aza) resulted in a 25.4-fold and 6.1-fold increase in ARID1A mRNA expression in SW48 and SW742 cells, respectively, while there was no change in SW480 and LS180 cells. No ARID1A CNV was observed in the CRC cell lines. CONCLUSION: ARID1A expression is downregulated in CRC tissues which correlates with it being a tumor suppressor protein. This finding confirms ARID1A loss of expression in CRC development. Our in-vitro results suggest high methylation status associates with reduced ARID1A expression and contributes to CRC tumorigenesis. However, there was no significant association between ARID1A loss of expression and clinicopathological characteristics. Future in-vivo analysis is warranted to further establish ARID1A role in colorectal neoplastic transformation.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Colorrectales/patología , Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Regulación Neoplásica de la Expresión Génica , Regiones Promotoras Genéticas , Factores de Transcripción/metabolismo , Antimetabolitos Antineoplásicos/farmacología , Azacitidina/farmacología , Biomarcadores de Tumor/genética , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Proteínas de Unión al ADN/genética , Femenino , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Tasa de Supervivencia , Factores de Transcripción/genética , Células Tumorales Cultivadas
10.
Dig Dis Sci ; 65(9): 2686-2690, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-31832971

RESUMEN

BACKGROUND: Colorectal and endometrial lesions increase with age. It is not known if these two precursor lesions in sporadic cases associate with each other. AIM: To determine the association between colorectal polyps and endometrial polyps (EP) in African Americans. METHODS: We reviewed records of patients referred to gynecology clinics and had colonoscopy at Howard University Hospital from January 2004 to December 2015. We defined cases as all patients who had EP and underwent colonoscopy. For controls, we used EP-free patients who underwent colonoscopy. Logistic regression analysis was used to assess the association between colon polyps and EP. RESULTS: The median age was 60 years in 118 Cases and 57 years in 664 Controls. The overall colorectal polyps prevalence in the two groups was not statistically different (54% in controls vs. 52% in cases, P = 0.60). Sessile serrated adenoma/polyps (SSPs) were more frequent in cases (8% vs. 2% in controls, P = 0.003). Sigmoid and rectal locations were more prevalent in controls than cases. In multivariate analysis and after adjusting for age, diabetes mellitus (DM), and BMI, SSPs were associated with EP occurrence with an odds ratio of 4.6 (CI 1.2-16.7, P = 0.022). CONCLUSION: Colorectal polyp prevalence was similar in EP patients compared to EP-free controls. However, we observed a significant association between higher-risk SSPs in patients with EP. The prevalence of smoking and DM was higher in these patients. Females with EP might benefit from a screening for colonic lesions in an age-independent manner.


Asunto(s)
Negro o Afroamericano , Pólipos del Colon/etnología , Neoplasias Colorrectales/etnología , Pólipos/etnología , Enfermedades Uterinas/etnología , Anciano , Pólipos del Colon/diagnóstico , Neoplasias Colorrectales/diagnóstico , Diabetes Mellitus/etnología , Femenino , Humanos , Persona de Mediana Edad , Pólipos/diagnóstico , Prevalencia , Factores Raciales , Estudios Retrospectivos , Medición de Riesgo , Factores de Riesgo , Fumar/efectos adversos , Fumar/etnología , Enfermedades Uterinas/diagnóstico
11.
Gastroenterology ; 164(6): 1012-1016, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36997444
14.
Gastroenterology ; 153(4): 910-923, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28807841

RESUMEN

Cancer from the gastrointestinal tract and its associated excretory organs will occur in more than 300,000 Americans in 2017, with colorectal cancer responsible for >40% of that burden; there will be more than 150,000 deaths from this group of cancers in the same time period. Disparities among subgroups related to the incidence and mortality of these cancers exist. The epidemiology and risk factors associated with each cancer bear out differences for racial groups in the United States. Esophageal adenocarcinoma is more frequent in non-Hispanic whites, whereas esophageal squamous cell carcinoma with risk factors of tobacco and alcohol is more frequent among blacks. Liver cancer has been most frequent among Asian/Pacific Islanders, chiefly due to hepatitis B vertical transmission, but other racial groups show increasing rates due to hepatitis C and emergence of cirrhosis from non-alcoholic fatty liver disease. Gastric cancer incidence remains highest among Asian/Pacific Islanders likely due to gene-environment interaction. In addition to esophageal squamous cell carcinoma, cancers of the small bowel, pancreas, and colorectum show the highest rates among blacks, where the explanations for the disparity are not as obvious and are likely multifactorial, including socioeconomic and health care access, treatment, and prevention (vaccination and screening) differences, dietary and composition of the gut microbiome, as well as biologic and genetic influences. Cognizance of these disparities in gastrointestinal cancer risk, as well as approaches that apply precision medicine methods to populations with the increased risk, may reduce the observed disparities for digestive cancers.


Asunto(s)
Neoplasias del Sistema Digestivo/etnología , Disparidades en el Estado de Salud , Estilo de Vida/etnología , Grupos Raciales , Neoplasias del Sistema Digestivo/diagnóstico , Neoplasias del Sistema Digestivo/mortalidad , Neoplasias del Sistema Digestivo/terapia , Femenino , Humanos , Incidencia , Masculino , Pronóstico , Medición de Riesgo , Factores de Riesgo , Distribución por Sexo , Factores de Tiempo , Estados Unidos/epidemiología
18.
JAMA ; 329(10): 839-840, 2023 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-36800187

RESUMEN

A 56-year-old man with no significant medical history has declined screening colonoscopy at previous physician visits but wants to undergo a less invasive test because his father died of colorectal cancer at age 80. What would you do next?


Asunto(s)
Neoplasias Colorrectales , Detección Precoz del Cáncer , Heces , Humanos , Colonoscopía , Neoplasias Colorrectales/diagnóstico , Detección Precoz del Cáncer/métodos , Heces/química , Tamizaje Masivo/métodos , Sangre Oculta
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA