Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 769
Filtrar
1.
PLoS One ; 19(4): e0300835, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38652719

RESUMO

BACKGROUND: Previous observational studies have demonstrated a connection between the risk of Type 2 diabetes mellitus (T2DM) and gastrointestinal problems brought on by Helicobacter pylori (H. pylori) infection. However, little is understood about how these factors impact on T2DM. METHOD: This study used data from the GWAS database on H. pylori antibodies, gastroduodenal ulcers, chronic gastritis, gastric cancer, T2DM and information on potential mediators: obesity, glycosylated hemoglobin (HbA1c) and blood glucose levels. Using univariate Mendelian randomization (MR) and multivariate MR (MVMR) analyses to evaluate the relationship between H. pylori and associated gastrointestinal diseases with the risk of developing of T2DM and explore the presence of mediators to ascertain the probable mechanisms. RESULTS: Genetic evidence suggests that H. pylori IgG antibody (P = 0.006, b = 0.0945, OR = 1.0995, 95% CI = 1.023-1.176), H. pylori GroEL antibody (P = 0.028, OR = 1.033, 95% CI = 1.004-1.064), gastroduodenal ulcers (P = 0.019, OR = 1.036, 95% CI = 1.006-1.068) and chronic gastritis (P = 0.005, OR = 1.042, 95% CI = 1.012-1.074) are all linked to an increased risk of T2DM, additionally, H. pylori IgG antibody is associated with obesity (P = 0.034, OR = 1.03, 95% CI = 1.002-1.055). The results of MVMR showed that the pathogenic relationship between H. pylori GroEL antibody and gastroduodenal ulcer in T2DM is mediated by blood glucose level and obesity, respectively. CONCLUSION: Our study found that H. pylori IgG antibody, H. pylori GroEL antibody, gastroduodenal ulcer and chronic gastritis are all related to t T2DM, and blood glucose level and obesity mediate the development of H. pylori GroEL antibody and gastroduodenal ulcer on T2DM, respectively. These findings may inform new prevention and intervention strategies for T2DM.


Assuntos
Diabetes Mellitus Tipo 2 , Infecções por Helicobacter , Helicobacter pylori , Análise da Randomização Mendeliana , Humanos , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/microbiologia , Diabetes Mellitus Tipo 2/genética , Infecções por Helicobacter/complicações , Infecções por Helicobacter/microbiologia , Anticorpos Antibacterianos/sangue , Gastroenteropatias/microbiologia , Gastroenteropatias/complicações , Obesidade/complicações , Obesidade/microbiologia , Estudo de Associação Genômica Ampla , Úlcera Péptica/microbiologia , Úlcera Péptica/epidemiologia , Gastrite/microbiologia , Gastrite/complicações , Chaperonina 60/genética , Fatores de Risco
2.
Rev. Hosp. Ital. B. Aires (2004) ; 43(3): 153-159, sept. 2023.
Artigo em Espanhol | LILACS, UNISALUD, BINACIS | ID: biblio-1519056

RESUMO

El consumo de probióticos, prebióticos y posbióticos, o su combinación, puede contribuir a mantener una microbiota intestinal saludable ya que permite la regulación de su disbiosis en el caso de algunas enfermedades o trastornos, principalmente en los trastornos gastrointestinales funcionales (TGIF). El microbioma intestinal es protagonista esencial en la fisiopatología de los TGIF a través de sus funciones metabólicas y nutricionales, el mantenimiento de la integridad de la mucosa intestinal y la regulación de la respuesta inmunitaria. Las investigaciones realizadas hasta la fecha indican que los probióticos, prebióticos y posbióticos pueden tener efectos inmunomoduladores directos y clínicamente relevantes. Existen pruebas del uso de esta familia de bióticos en individuos sanos para mejorar la salud general y aliviar los síntomas en una serie de enfermedades como los cólicos infantiles. La colonización y establecimiento de la microbiota comienza en el momento del nacimiento; los primeros 2-3 años de vida son fundamentales para el desarrollo de una comunidad microbiana abundante y diversa. Diversos estudios científicos realizados mediante técnicas tradicionales dependientes de cultivo y más recientemente por técnicas moleculares han observado diferencias en las poblaciones bacterianas de bebés sanos y aquellos que sufren TGIF, estos últimos caracterizados por un aumento de especies patógenas y una menor población de bifidobacterias y lactobacilos, en comparación con los primeros. En tal contexto, se considera que la microbiota intestinal como protagonista en el desarrollo de esos trastornos, entre ellos los cólicos infantiles, a través de sus funciones metabólicas, nutricionales, de mantenimiento de la integridad de la mucosa intestinal y regulación de la respuesta inmunitaria. Esto ha abierto la puerta al estudio de la utilización de prebióticos, probióticos y posbióticos en el tratamiento y/o prevención de los TGIF infantiles. El parto vaginal y de término así como la lactancia son fundamentales en la constitución de una microbiota saludable. Como herramientas de apoyo, existen estudios de eficacia que sustentan la administración de esta familia de bióticos, principalmente en los casos en que la lactancia no sea posible o esté limitada. (AU)


The consumption of probiotics, prebiotics, and postbiotics, or a combination of them, can contribute to maintaining a healthy intestinal microbiota as it allows the regulation of its dysbiosis in the case of some diseases or disorders, mainly in functional gastrointestinal disorders (FGIDs). The gut microbiome is an essential player in the pathophysiology of FGIDs through its metabolic and nutritional functions, the maintenance of intestinal mucosal integrity, and the regulation of the immune response. Research results thus far indicate that probiotics, prebiotics, and postbiotics may have direct and clinically relevant immunomodulatory effects. There is evidence regarding the prescription of this family of biotics in healthy individuals to improve overall health and alleviate symptoms in many conditions like infantile colic. The colonization and microbiota establishment begins at birth; the first 2-3 years of life are critical for developing an abundant and diverse microbial community. Several scientific studies performed by traditional culture-dependent techniques and more recently by molecular techniques have observed differences in the bacterial populations of healthy infants and those suffering from FGIDs, the latter characterized by an increase in pathogenic species and a lower population of bifidobacteria and lactobacilli, compared to the former. In this context, the intestinal microbiota plays a leading role in the onset of these disorders, including infantile colic, through its metabolic and nutritional functions, maintenance of the integrity of the intestinal mucosa, and regulation of the immune response. That has opened the door to the study of prebiotics, probiotics, and postbiotics usage in the treatment and or prevention of infantile FGIDs. Vaginal and term delivery and breastfeeding are fundamental in the constitution of a healthy microbiota. As supportive tools, there are efficacy studies that support the administration of this family of biotics, mainly in cases where lactation is not possible or is limited.


Assuntos
Humanos , Cólica/microbiologia , Probióticos , Prebióticos , Simbióticos , Microbioma Gastrointestinal , Gastroenteropatias/microbiologia , Lactação , Cólica/dietoterapia , Cólica/fisiopatologia , Cólica/prevenção & controle , Alimento Funcional , Gastroenteropatias/dietoterapia , Gastroenteropatias/fisiopatologia , Gastroenteropatias/prevenção & controle
3.
Artigo em Inglês | MEDLINE | ID: mdl-37094909

RESUMO

Plant-based diets (PBDs), rich in high-quality plant foods, offer multiple benefits for the overall and gastrointestinal health. Recently, it has been demostrated that the positive effects of PBDs on gastrointestinal health can be mediated by the gut microbiota, in particular, by inducing a greater diversity of bacteria. This review summarizes current knowledge on the relationship between nutrition, the gut microbiota, and host metabolic status. We discussed how dietary habits modify the composition and physiological activity of the gut microbiota and how gut dysbiosis affects the most prevalent gastrointestinal diseases, including inflammatory bowel diseases, functional bowel disorders, liver disorders, and gastrointestinal cancer. The beneficial role of PBDs is being increasingly recognized as potentially useful in the management of most diseases of the gastrointestinal tract.


Assuntos
Gastroenteropatias , Doenças Inflamatórias Intestinais , Humanos , Dieta , Gastroenteropatias/microbiologia , Dieta Vegetariana , Disbiose
4.
World J Gastroenterol ; 29(1): 19-42, 2023 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-36683718

RESUMO

The gut microbiota is currently considered an external organ of the human body that provides important mechanisms of metabolic regulation and protection. The gut microbiota encodes over 3 million genes, which is approximately 150 times more than the total number of genes present in the human genome. Changes in the qualitative and quantitative composition of the microbiome lead to disruption in the synthesis of key bacterial metabolites, changes in intestinal barrier function, and inflammation and can cause the development of a wide variety of diseases, such as diabetes, obesity, gastrointestinal disorders, cardiovascular issues, neurological disorders and oncological concerns. In this review, I consider issues related to the role of the microbiome in the regulation of intestinal barrier function, its influence on physiological and pathological processes occurring in the body, and potential new therapeutic strategies aimed at restoring the gut microbiome. Herewith, it is important to understand that the gut microbiota and human body should be considered as a single biological system, where change of one element will inevitably affect its other components. Thus, the study of the impact of the intestinal microbiota on health should be considered only taking into account numerous factors, the role of which has not yet been fully elucidated.


Assuntos
Gastroenteropatias , Microbioma Gastrointestinal , Microbiota , Probióticos , Humanos , Gastroenteropatias/microbiologia , Inflamação/microbiologia , Microbioma Gastrointestinal/fisiologia , Disbiose/microbiologia
5.
Cancer Chemother Pharmacol ; 89(2): 275-281, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34854953

RESUMO

PURPOSE: Irinotecan can cause high levels of diarrhea caused by toxic injury to the gastrointestinal microenvironment. Toll-like receptor 4 (TLR4) and the gut microbiome have previously been implicated in gastrointestinal toxicity and diarrhea; however, the link between these two factors has not been definitively determined. We used a tumor-bearing, intestinal epithelial cell (IEC) TLR4 knockout model (Tlr4ΔIEC) to assess microbiome changes following irinotecan treatment. We then determined if a fecal microbiota transplant (FMT) between Tlr4ΔIEC and wild-type (WT) mice altered irinotecan-induced gastrointestinal toxicity. METHODS: MC-38 colorectal cancer cells were injected into WT and Tlr4ΔIEC mice. Fecal samples were collected prior to tumor inoculation, prior to irinotecan treatment and at cull. 16S rRNA gene sequencing was used to assess changes in the microbiome. Next, FMT was used to transfer the microbiome phenotype between Tlr4ΔIEC and WT mice prior to irinotecan treatment. Gastrointestinal toxicity symptoms were assessed. RESULTS: In study 1, there were no compositional differences in the microbiome between Tlr4ΔIEC and WT mice at baseline. However, predicted functional capacity of the microbiome was different between WT and Tlr4ΔIEC at baseline and post-irinotecan. In study 2, Tlr4ΔIEC mice were protected from grade 3 diarrhea. Additionally, WT mice who did not receive FMT had more colonic damage in the colon compared to controls (P = 0.013). This was not seen in Tlr4ΔIEC mice or WT mice who received FMT (P > 0.05). CONCLUSION: Tlr4ΔIEC and WT had no baseline compositional microbiome differences, but functional differences at baseline and following irinotecan. FMT altered some aspects of irinotecan-induced gastrointestinal toxicity.


Assuntos
Neoplasias Colorretais/tratamento farmacológico , Microbioma Gastrointestinal/efeitos dos fármacos , Irinotecano/farmacologia , Receptor 4 Toll-Like/genética , Animais , Linhagem Celular Tumoral , Neoplasias Colorretais/genética , Transplante de Microbiota Fecal/métodos , Gastroenteropatias/induzido quimicamente , Gastroenteropatias/microbiologia , Microbioma Gastrointestinal/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , RNA Ribossômico 16S , Inibidores da Topoisomerase I/farmacologia
6.
BMC Microbiol ; 21(1): 247, 2021 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-34525965

RESUMO

BACKGROUND: Infants with cystic fibrosis (CF) suffer from gastrointestinal (GI) complications, including pancreatic insufficiency and intestinal inflammation, which have been associated with impaired nutrition and growth. Recent evidence identified altered fecal microbiota taxonomic compositions in infants with CF relative to healthy infants that were characterized by differences in the abundances of taxa associated with GI health and nutrition. Furthermore, these taxonomic differences were more pronounced in low length infants with CF, suggesting a potential link to linear growth failure. We hypothesized that these differences would entail shifts in the microbiome's functional capacities that could contribute to inflammation and nutritional failure in infants with CF. RESULTS: To test this hypothesis, we compared fecal microbial metagenomic content between healthy infants and infants with CF, supplemented with an analysis of fecal metabolomes in infants with CF. We identified notable differences in CF fecal microbial functional capacities, including metabolic and environmental response functions, compared to healthy infants that intensified during the first year of life. A machine learning-based longitudinal metagenomic age analysis of healthy and CF fecal metagenomic functional profiles further demonstrated that these differences are characterized by a CF-associated delay in the development of these functional capacities. Moreover, we found metagenomic differences in functions related to metabolism among infants with CF that were associated with diet and antibiotic exposure, and identified several taxa as potential drivers of these functional differences. An integrated metagenomic and metabolomic analysis further revealed that abundances of several fecal GI metabolites important for nutrient absorption, including three bile acids, correlated with specific microbes in infants with CF. CONCLUSIONS: Our results highlight several metagenomic and metabolomic factors, including bile acids and other microbial metabolites, that may impact nutrition, growth, and GI health in infants with CF. These factors could serve as promising avenues for novel microbiome-based therapeutics to improve health outcomes in these infants.


Assuntos
Fibrose Cística/complicações , Fibrose Cística/microbiologia , Disbiose/complicações , Fezes/microbiologia , Gastroenteropatias/etiologia , Metaboloma , Metagenoma , Gastroenteropatias/microbiologia , Gastroenteropatias/fisiopatologia , Humanos , Lactente , Estudos Longitudinais , Metabolômica/métodos , Estudos Prospectivos
7.
Am J Trop Med Hyg ; 105(5): 1222-1226, 2021 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-34491224

RESUMO

Gastrointestinal basidiobolomycosis (GIB) is a rare, life-threatening fungal infection affecting immunocompetent individuals in tropical and subtropical regions. A diverse presentation of GIB has been reported, but no report has yet been published on intussusception. We describe a 23-month-old immunocompetent boy from a subtropical area in Iran who presented with intussusception. Prolonged fever, an abdominal mass, hepatomegaly, high erythrocyte sedimentation rate, and peripheral eosinophilia strongly suggested GIB. Accordingly, GIB was diagnosed based on the characteristic histopathology (the Splendore-Hoeppli phenomenon) detected in a liver sample taken via biopsy. Exploratory laparotomy showed several organs, including the colon, gall bladder, liver, and abdominal wall, were involved. Antifungal therapy with trimethoprim/sulfamethoxazole, liposomal amphotericin B, a saturated solution of potassium iodide, and surgical resection of involved tissues were used with improved outcome. The presence of non-septate fungal hyphal elements in the colonic mucosa led to the thickening of the bowel wall, leading to secondary intussusception.


Assuntos
Antifúngicos/uso terapêutico , Gastroenteropatias/tratamento farmacológico , Gastroenteropatias/cirurgia , Intussuscepção/tratamento farmacológico , Mucormicose/tratamento farmacológico , Mucormicose/cirurgia , Combinação Trimetoprima e Sulfametoxazol/uso terapêutico , Gastroenteropatias/diagnóstico , Gastroenteropatias/microbiologia , Humanos , Lactente , Intussuscepção/diagnóstico , Intussuscepção/microbiologia , Intussuscepção/cirurgia , Irã (Geográfico) , Masculino , Mucorales/isolamento & purificação , Mucormicose/complicações , Mucormicose/diagnóstico , Resultado do Tratamento
8.
Pharmacol Res ; 171: 105775, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34273489

RESUMO

Nucleotide-binding oligomerization domain 1 (NOD1), a pattern recognition receptor (PRR) that detects bacterial peptidoglycan fragments and other danger signals, has been linked to inflammatory pathologies. NOD1, which is expressed by immune and non-immune cells, is activated after recognizing microbe-associated molecular patterns (MAMPs). This recognition triggers host defense responses and both immune memory and tolerance can also be achieved during these processes. Since the gut microbiota is currently considered a master regulator of human physiology central in health and disease and the intestine metabolizes a wide range of nutrients, drugs and hormones, it is a fact that dysbiosis can alter tissues and organs homeostasis. These systemic alterations occur in response to gastrointestinal immune adaptations that are not yet fully understood. Even if previous evidence confirms the connection between the microbiota, the immune system and metabolic disorders, much remains to be discovered about the contribution of NOD1 to low-grade inflammatory pathologies such as obesity, diabetes and cardiovascular diseases. This review compiles the most recent findings in this area, while providing a dynamic and practical framework with future approaches for research and clinical applications on targeting NOD1. This knowledge can help to rate the consequences of the disease and to stratify the patients for therapeutic interventions.


Assuntos
Microbioma Gastrointestinal , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/microbiologia , Proteína Adaptadora de Sinalização NOD1/imunologia , Animais , Encefalopatias/imunologia , Encefalopatias/microbiologia , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/microbiologia , Gastroenteropatias/imunologia , Gastroenteropatias/microbiologia , Humanos , Inflamação/imunologia , Inflamação/microbiologia , Proteína Adaptadora de Sinalização NOD2/imunologia
9.
Nutrients ; 13(7)2021 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-34209683

RESUMO

Gut microbiota dysbiosis is a common feature in colorectal cancer (CRC) and inflammatory bowel diseases (IBD). Adoption of the Mediterranean diet (MD) has been proposed as a therapeutic approach for the prevention of multiple diseases, and one of its mechanisms of action is the modulation of the microbiota. We aimed to determine whether MD can be used as a preventive measure against cancer and inflammation-related diseases of the gut, based on its capacity to modulate the local microbiota. A joint meta-analysis of publicly available 16S data derived from subjects following MD or other diets and from patients with CRC, IBD, or other gut-related diseases was conducted. We observed that the microbiota associated with MD was enriched in bacteria that promote an anti-inflammatory environment but low in taxa with pro-inflammatory properties capable of altering intestinal barrier functions. We found an opposite trend in patients with intestinal diseases, including cancer. Some of these differences were maintained even when MD was compared to healthy controls without a defined diet. Our findings highlight the unique effects of MD on the gut microbiota and suggest that integrating MD principles into a person's lifestyle may serve as a preventive method against cancer and other gut-related diseases.


Assuntos
Neoplasias Colorretais/prevenção & controle , Dieta Mediterrânea , Disbiose/prevenção & controle , Gastroenteropatias/prevenção & controle , Microbioma Gastrointestinal/fisiologia , Adulto , Colo/microbiologia , Neoplasias Colorretais/microbiologia , Disbiose/microbiologia , Feminino , Gastroenteropatias/microbiologia , Humanos , Doenças Inflamatórias Intestinais/microbiologia , Doenças Inflamatórias Intestinais/prevenção & controle , Masculino , Pessoa de Meia-Idade
10.
Curr Oncol Rep ; 23(8): 92, 2021 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-34125319

RESUMO

PURPOSE OF REVIEW: We review the emerging evidence regarding the relationship between the microbiota of the gastrointestinal and female reproductive tracts and gynecologic cancer. RECENT FINDINGS: The microbiome has essential roles in maintaining health. In recent years, the microbiota of the gastrointestinal and female reproductive tracts have been linked to many diseases, including gynecologic cancer. Alterations to the bacterial populations in a microbiota, or dysbiosis, have been shown to favor a pro-carcinogenic state through altered immune responses, dysregulated hormone metabolism, and modulation of the cell cycle. Pre-clinical and clinical studies have emerged, demonstrating that specific bacteria or microbial communities may be associated with increased risk for uterine, ovarian, and cervical cancers. Notably, numerous studies have linked a non-Lactobacillus-dominant vaginal microbiota, composed of anaerobic bacteria, with HPV infection, persistence, and development of invasive cervical cancer. Similarly, next-generation high-throughput sequencing techniques have enabled the characterization of unique microbiotas in patients with malignant and benign gynecologic conditions, shedding light on new associations between bacterial species and gynecologic cancers. Harnessing the power of the microbiome for early diagnosis, therapeutic intervention and modulation creates tremendous potential to optimize gynecologic cancer outcomes in the future.


Assuntos
Microbioma Gastrointestinal , Neoplasias dos Genitais Femininos/microbiologia , Genitália Feminina/microbiologia , Feminino , Gastroenteropatias/microbiologia , Trato Gastrointestinal/microbiologia , Neoplasias dos Genitais Femininos/metabolismo , Neoplasias dos Genitais Femininos/terapia , Genitália Feminina/metabolismo , Humanos
11.
BMC Infect Dis ; 21(1): 539, 2021 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-34098893

RESUMO

BACKGROUND: In sub-Saharan Africa, acute respiratory infections (ARI), acute gastrointestinal infections (GI) and acute febrile disease of unknown cause (AFDUC) have a large disease burden, especially among children, while respective aetiologies often remain unresolved. The need for robust infectious disease surveillance to detect emerging pathogens along with common human pathogens has been highlighted by the ongoing novel coronavirus disease 2019 (COVID-19) pandemic. The African Network for Improved Diagnostics, Epidemiology and Management of Common Infectious Agents (ANDEMIA) is a sentinel surveillance study on the aetiology and clinical characteristics of ARI, GI and AFDUC in sub-Saharan Africa. METHODS: ANDEMIA includes 12 urban and rural health care facilities in four African countries (Côte d'Ivoire, Burkina Faso, Democratic Republic of the Congo and Republic of South Africa). It was piloted in 2018 in Côte d'Ivoire and the initial phase will run from 2019 to 2021. Case definitions for ARI, GI and AFDUC were established, as well as syndrome-specific sampling algorithms including the collection of blood, naso- and oropharyngeal swabs and stool. Samples are tested using comprehensive diagnostic protocols, ranging from classic bacteriology and antimicrobial resistance screening to multiplex real-time polymerase chain reaction (PCR) systems and High Throughput Sequencing. In March 2020, PCR testing for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and analysis of full genomic information was included in the study. Standardised questionnaires collect relevant clinical, demographic, socio-economic and behavioural data for epidemiologic analyses. Controls are enrolled over a 12-month period for a nested case-control study. Data will be assessed descriptively and aetiologies will be evaluated using a latent class analysis among cases. Among cases and controls, an integrated analytic approach using logistic regression and Bayesian estimation will be employed to improve the assessment of aetiology and associated risk factors. DISCUSSION: ANDEMIA aims to expand our understanding of ARI, GI and AFDUC aetiologies in sub-Saharan Africa using a comprehensive laboratory diagnostics strategy. It will foster early detection of emerging threats and continued monitoring of important common pathogens. The network collaboration will be strengthened and site diagnostic capacities will be reinforced to improve quality management and patient care.


Assuntos
Doenças Transmissíveis/diagnóstico , Doenças Transmissíveis/epidemiologia , Programas de Rastreamento , Vigilância de Evento Sentinela , Teorema de Bayes , Burkina Faso , Estudos de Casos e Controles , Côte d'Ivoire , República Democrática do Congo , Febre/epidemiologia , Febre/microbiologia , Gastroenteropatias/epidemiologia , Gastroenteropatias/microbiologia , Humanos , Reação em Cadeia da Polimerase em Tempo Real , Infecções Respiratórias/epidemiologia , África do Sul
12.
Am J Clin Oncol ; 44(8): 402-408, 2021 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-34107499

RESUMO

BACKGROUND AND OBJECTIVE: Immune-mediated diarrhea and colitis (IMDC) is a common adverse event in cancer patients receiving immune checkpoint inhibitors (ICIs). Gastrointestinal (GI) infections can co-occur with IMDC, and its impact on the course and outcome of IMDC remains unclear. PATIENTS AND METHODS: We retrospectively reviewed cancer patients who received ICIs and developed IMDC between January 2015 and September 2019. GI multiplex panel is used to assess GI infection. The study group included patients with positive infection except those who are only positive for Clostridioides difficile or cytomegalovirus. The control group is IMDC patients with negative infection using frequency matching. Patients' disease course and outcome were compared between groups. RESULTS: A total of 72 patients with IMDC were included: 22 in the study group and 50 as control. Escherichia coli of different pathotypes was observed in 17 patients. Five patients had viral infections, for example, adenovirus, norovirus, and sapovirus. Patients with GI infections more frequently had grade 3 or 4 colitis (43% vs. 18%, P=0.041). Overall, GI infections were not associated with different risks of IMDC recurrence or overall survival. Antibiotics treatment did not affect the requirement for infliximab or vedolizumab but relate to a higher risk of IMDC recurrence (50.0% vs. 0.0%, P=0.015). CONCLUSIONS: In our study, concomitant GI infections are associated with more severe symptoms in IMDC patients. Antimicrobial treatment did not circumvent the need for immunosuppressive therapy for IMDC or improve the clinical outcome. Concomitant GI infection was not associated with a higher risk of IMDC recurrence or poor overall survival.


Assuntos
Colite/induzido quimicamente , Diarreia/induzido quimicamente , Gastroenteropatias/microbiologia , Gastroenteropatias/virologia , Inibidores de Checkpoint Imunológico/efeitos adversos , Idoso , Antibacterianos/efeitos adversos , Antibacterianos/uso terapêutico , Infecções por Clostridium/induzido quimicamente , Colite/mortalidade , Colite/terapia , Diarreia/mortalidade , Diarreia/terapia , Infecções por Escherichia coli/tratamento farmacológico , Infecções por Escherichia coli/etiologia , Feminino , Gastroenteropatias/tratamento farmacológico , Humanos , Inibidores de Checkpoint Imunológico/uso terapêutico , Masculino , Pessoa de Meia-Idade , Neoplasias/tratamento farmacológico , Neoplasias/microbiologia , Estudos Retrospectivos , Fatores de Risco , Resultado do Tratamento , Viroses/etiologia , Viroses/virologia
13.
Eur J Clin Microbiol Infect Dis ; 40(10): 2153-2160, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33963926

RESUMO

Aetiological diagnosis of gastrointestinal infections is challenging since a wide range of bacteria, parasites and viruses can be causal agents and derived clinical manifestations appear quite similar. Our aim was to evaluate contribution of the novel QIAstat-DxGastrointestinal Panel (GIP) to aetiological diagnosis of gastrointestinal infections and rational antimicrobial prescription in a reference paediatric hospital. Evaluation included comparison of diagnostic yield and agreement of results of QIAstat-Dx GIP and conventional microbiological methods. Parallel testing was performed on stool samples collected prospectively from children admitted to Sant Joan de Deu Barcelona Hospital (Spain) during the period February-March 2019. Influence of the panel test use on antimicrobial prescription was assessed using a pre-post study design. Eighty-six (68.8%) out of 125 specimens were positive by QIAstat-Dx GIP versus 44 (35.2%) positive by a composite of conventional methods (p<0.001). Global agreement of panel test results with rotavirus-adenovirus antigen detection (92.8%) and a two-step antigen/toxin and PCR-based algorithm for toxigenic Clostridioides difficile detection (87.5%) was greater than that with bacterial culture (76.0%) and parasite microscopic identification (64.3%). Panel test results orientated antimicrobial prescription changes in 18 (14.4%) patients, including antimicrobial start in 11 cases initially untreated, targeted antimicrobial prescription in 5 and discontinuation in 2 cases empirically treated. Results showed that QIAstat-Dx GIP significantly expanded aetiological diagnosis of gastrointestinal infections compared to conventional microbiological methods while orientating a more judicious use of antimicrobial drugs in hospitalised children.


Assuntos
Bactérias/isolamento & purificação , Infecções Bacterianas/microbiologia , Gastroenteropatias/microbiologia , Técnicas de Diagnóstico Molecular/métodos , Bactérias/classificação , Bactérias/genética , Infecções Bacterianas/diagnóstico , Criança , Pré-Escolar , Feminino , Gastroenteropatias/diagnóstico , Hospitais Pediátricos/estatística & dados numéricos , Humanos , Lactente , Masculino , Técnicas de Diagnóstico Molecular/instrumentação , Estudos Prospectivos , Espanha
14.
Diagn Pathol ; 16(1): 40, 2021 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-33952310

RESUMO

AIMS: Patients with COVID-19 can also have enteric symptoms. Here we analyzed the histopathology of intestinal detachment tissue from a patient with COVID-19. METHODS: The enteric tissue was examined by hematoxylin & eosin stain, PAS (Periodic acid-Schiff) staining, Gram staining, Ziehl-Neelsen stain and Grocott's Methenamine Silver (GMS) Stain. The distribution of CD3, CD4, CK20 and CD68, cytomegalovirus (CMV) and Herpes Simplex Virus (HSV) antigen were determined by immunohistochemistry. In situ hybridization (ISH) of SARS-CoV-2 and Epstein-Barr virus-encoded small RNA (EBER) were also performed. RESULTS: We observed mucosal epithelium shedding, intestinal mucosal erosion, focal inflammatory necrosis with hemorrhage, massive neutrophil infiltration, macrophage proliferation accompanied by minor lymphocyte infiltration. Fungal spores and gram positive cocci but not mycobacteria tuberculosis were identified. Immunohistochemistry staining showed abundant CD68+ macrophages but few lymphocytes infiltration. HSV, CMV and EBV were negative. ISH of SARS-CoV-2 RNA showed positive signal which mostly overlapped with CD68 positivity. CONCLUSIONS: The in situ detection of SARS-CoV-2 RNA in intestinal macrophages implicates a possible route for gastrointestinal infection. Further study is needed to further characterize the susceptibility of enteric cells to SARS-CoV-2 infection.


Assuntos
COVID-19/patologia , Gastroenteropatias/patologia , Mucosa Intestinal/patologia , Macrófagos/virologia , RNA Viral/isolamento & purificação , SARS-CoV-2/isolamento & purificação , Idoso , Biomarcadores/metabolismo , COVID-19/diagnóstico , COVID-19/imunologia , COVID-19/microbiologia , Teste para COVID-19 , Gastroenteropatias/diagnóstico , Gastroenteropatias/imunologia , Gastroenteropatias/microbiologia , Humanos , Imuno-Histoquímica , Hibridização In Situ , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Macrófagos/metabolismo , Masculino
15.
Molecules ; 26(7)2021 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-33917379

RESUMO

Polyphenols are classified as an organic chemical with phenolic units that display an array of biological functions. However, polyphenols have very low bioavailability and stability, which make polyphenols a less bioactive compound. Many researchers have indicated that several factors might affect the efficiency and the metabolism (biotransformation) of various polyphenols, which include the gut microbiota, structure, and physical properties as well as its interactions with other dietary nutrients (macromolecules). Hence, this mini-review covers the two-way interaction between polyphenols and gut microbiota (interplay) and how polyphenols are metabolized (biotransformation) to produce various polyphenolic metabolites. Moreover, the protective effects of numerous polyphenols and their metabolites against various gastrointestinal disorders/diseases including gastritis, gastric cancer, colorectal cancer, inflammatory bowel disease (IBD) like ulcerative colitis (UC), Crohn's disease (CD), and irritable bowel syndrome (IBS) like celiac disease (CED) are discussed. For this review, the authors chose only a few popular polyphenols (green tea polyphenol, curcumin, resveratrol, quercetin), and a discussion of their proposed mechanism underpinning the gastroprotection was elaborated with a special focus on clinical evidence. Overall, this contribution would help the general population and science community to identify a potent polyphenol with strong antioxidant, anti-inflammatory, anti-cancer, prebiotic, and immunomodulatory properties to combat various gut-related diseases or disorders (complementary therapy) along with modified lifestyle pattern and standard gastroprotective drugs. However, the data from clinical trials are much limited and hence many large-scale clinical trials should be performed (with different form/metabolites and dose) to confirm the gastroprotective activity of the above-mentioned polyphenols and their metabolites before recommendation.


Assuntos
Gastroenteropatias/tratamento farmacológico , Polifenóis/uso terapêutico , Substâncias Protetoras/uso terapêutico , Estômago/patologia , Animais , Gastroenteropatias/microbiologia , Gastroenteropatias/fisiopatologia , Microbioma Gastrointestinal/efeitos dos fármacos , Humanos , Metaboloma/efeitos dos fármacos , Polifenóis/química , Polifenóis/farmacologia , Estômago/efeitos dos fármacos
16.
J Gastroenterol Hepatol ; 36(4): 832-840, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33880762

RESUMO

For a long time, gut bacteria have been recognized for their important roles in the occurrence and progression of gastrointestinal diseases like colorectal cancer, and the ever-increasing amounts of microbiome data combined with other high-quality clinical and imaging datasets are leading the study of gastrointestinal diseases into an era of biomedical big data. The "omics" technologies used for microbiome analysis continuously evolve, and the machine learning or artificial intelligence technologies are key to extract the relevant information from microbiome data. This review intends to provide a focused summary of recent research and applications of microbiome big data and to discuss the use of artificial intelligence to combat gastrointestinal diseases.


Assuntos
Inteligência Artificial/tendências , Big Data , Gastroenteropatias/etiologia , Gastroenteropatias/microbiologia , Microbioma Gastrointestinal , Armazenamento e Recuperação da Informação/métodos , Conjuntos de Dados como Assunto
17.
J Med Chem ; 64(9): 5710-5729, 2021 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-33891818

RESUMO

Helicobacter pylori (Hp) is a human pathogen that lives in the gastric mucosa of approximately 50% of the world's population causing gastritis, peptic ulcers, and gastric cancer. An increase in resistance to current drugs has sparked the search for new Hp drug targets and therapeutics. One target is the disruption of nucleic acid production, which can be achieved by impeding the synthesis of 6-oxopurine nucleoside monophosphates, the precursors of DNA and RNA. These metabolites are synthesized by Hp xanthine-guanine-hypoxanthine phosphoribosyltransferase (XGHPRT). Here, nucleoside phosphonates have been evaluated, which inhibit the activity of this enzyme with Ki values as low as 200 nM. The prodrugs of these compounds arrest the growth of Hp at a concentration of 50 µM in cell-based assays. The kinetic properties of HpXGHPRT have been determined together with its X-ray crystal structure in the absence and presence of 9-[(N-3-phosphonopropyl)-aminomethyl-9-deazahypoxanthine, providing a basis for new antibiotic development.


Assuntos
Antibacterianos/química , Proteínas de Bactérias/metabolismo , Pentosiltransferases/metabolismo , Sequência de Aminoácidos , Antibacterianos/metabolismo , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Proteínas de Bactérias/química , Sítios de Ligação , Cristalografia por Raios X , Gastroenteropatias/tratamento farmacológico , Gastroenteropatias/microbiologia , Gastroenteropatias/patologia , Infecções por Helicobacter/tratamento farmacológico , Infecções por Helicobacter/patologia , Helicobacter pylori/efeitos dos fármacos , Helicobacter pylori/enzimologia , Humanos , Hipoxantina Fosforribosiltransferase/química , Hipoxantina Fosforribosiltransferase/metabolismo , Hipoxantinas/química , Hipoxantinas/metabolismo , Hipoxantinas/farmacologia , Hipoxantinas/uso terapêutico , Cinética , Simulação de Dinâmica Molecular , Organofosfonatos/química , Organofosfonatos/metabolismo , Organofosfonatos/farmacologia , Organofosfonatos/uso terapêutico , Pentosiltransferases/química , Pró-Fármacos/química , Pró-Fármacos/metabolismo , Pró-Fármacos/farmacologia , Pró-Fármacos/uso terapêutico , Alinhamento de Sequência , Relação Estrutura-Atividade
18.
Medicine (Baltimore) ; 100(11): e25138, 2021 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-33725995

RESUMO

BACKGROUND: Gastrointestinal complications and malnutrition are common problems that affect postoperative rehabilitation and survival of patients with esophageal cancer. Evidence has shown that probiotics have a positive effect on improving gastrointestinal complications and nutritional status of patients with esophageal cancer after surgery, but there is a lack of prospective studies on this topic. We designed this prospective randomized controlled trial to evaluate the effects of probiotics on gastrointestinal complications and nutritional status in patients with postoperative esophageal cancer. METHODS: This is a prospective, randomized, double-blind, placebo-controlled trial. It was approved by the Clinical Research Ethics Committee of our hospital. 192 patients will be randomly divided into probiotics group and the placebo group in a 1:1 ratio. After operation, probiotics and placebo will be given orally for 8 weeks. The indexes of nutritional status and incidence of digestive tract complications will be recorded and the data will be analyzed by SPSS 18.0 software. DISCUSSION: This study will evaluate the effect of probiotics on gastrointestinal complications and nutritional status of postoperative patients with esophageal cancer. The results of this study will provide clinical basis for the use of probiotics in postoperative treatment of esophageal cancer. TRIAL REGISTRATION: OSF Registration number: D DOI 10.17605/OSF.IO/QHW86.


Assuntos
Neoplasias Esofágicas/cirurgia , Esofagectomia/efeitos adversos , Gastroenteropatias/terapia , Desnutrição/terapia , Complicações Pós-Operatórias , Probióticos/uso terapêutico , Adulto , Método Duplo-Cego , Neoplasias Esofágicas/microbiologia , Feminino , Gastroenteropatias/etiologia , Gastroenteropatias/microbiologia , Humanos , Masculino , Desnutrição/etiologia , Desnutrição/microbiologia , Pessoa de Meia-Idade , Estado Nutricional , Período Pós-Operatório , Estudos Prospectivos , Ensaios Clínicos Controlados Aleatórios como Assunto , Resultado do Tratamento
19.
Am J Chin Med ; 49(2): 237-268, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33622213

RESUMO

Intestinal flora is essential for maintaining host health and plays a unique role in transforming Traditional Chinese Medicine (TCM). TCM, as a bodyguard, has saved countless lives and maintained human health in the long history, especially in this COVID-19 pandemic. Pains of diseases have been removed from the effective TCM therapy, such as TCM preparation, moxibustion, and acupuncture. With the development of life science and technology, the wisdom and foresight of TCM has been more displayed. Furthermore, TCM has been also inherited and developed in innovation to better realize the modernization and globalization. Nowadays, intestinal flora transforming TCM and TCM targeted intestinal flora treating diseases have been important findings in life science. More and more TCM researches showed the significance of intestinal flora. Intestinal flora is also a way to study TCM to elucidate the profound theory of TCM. Processing, compatibility, and properties of TCM are well demonstrated by intestinal flora. Thus, it is no doubt that intestinal flora is a core in TCM study. The interaction between intestinal flora and TCM is so crucial for host health. Therefore, it is necessary to sum up the latest results in time. This paper systematically depicted the profile of TCM and the importance of intestinal flora in host. What is more, we comprehensively summarized and discussed the latest progress of the interplay between TCM and intestinal flora to better reveal the core connotation of TCM.


Assuntos
Medicamentos de Ervas Chinesas/uso terapêutico , Disbiose/microbiologia , Microbioma Gastrointestinal , Medicina Tradicional Chinesa , Doenças Autoimunes/microbiologia , Doenças Autoimunes/terapia , COVID-19 , Doenças Cardiovasculares/microbiologia , Doenças Cardiovasculares/terapia , Diabetes Mellitus/microbiologia , Diabetes Mellitus/terapia , Eletroacupuntura , Gastroenteropatias/microbiologia , Gastroenteropatias/terapia , Humanos , Doenças Metabólicas/microbiologia , Doenças Metabólicas/terapia , Neoplasias/microbiologia , Neoplasias/terapia , Hepatopatia Gordurosa não Alcoólica/microbiologia , Hepatopatia Gordurosa não Alcoólica/terapia , Obesidade/microbiologia , Obesidade/terapia , Insuficiência Renal Crônica/microbiologia , Insuficiência Renal Crônica/terapia , SARS-CoV-2
20.
J Med Case Rep ; 15(1): 60, 2021 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-33557941

RESUMO

BACKGROUND: To investigate the potential beneficial effect of fecal microbiota transplantation (FMT) on gastrointestinal symptoms, gut dysbiosis and immune status in discharged COVID-19 patients. CASE PRESENTATION: A total of 11 COVID-19 patients were recruited in April, 2020, about one month on average after they were discharged from the hospital. All subjects received FMT for 4 consecutive days by oral capsule administrations with 10 capsules for each day. In total, 5 out of 11 patients reported to be suffered from gastrointestinal symptoms, which were improved after FMT. After FMT, alterations of B cells were observed, which was characterized as decreased naive B cell (P = 0.012) and increased memory B cells (P = 0.001) and non-switched B cells (P = 0.012).The microbial community richness indicated by operational taxonomic units number, observed species and Chao1 estimator was marginally increased after FMT. Gut microbiome composition of discharged COVID-19 patients differed from that of the general population at both phylum and genera level, which was characterized with a lower proportion of Firmicutes (41.0%) and Actinobacteria (4.0%), higher proportion of Bacteroidetes (42.9%) and Proteobacteria (9.2%). FMT can partially restore the gut dysbiosis by increasing the relative abundance of Actinobacteria (15.0%) and reducing Proteobacteria (2.8%) at the phylum level. At the genera level, Bifidobacterium and Faecalibacterium had significantly increased after FMT. CONCLUSIONS: After FMT, altered peripheral lymphocyte subset, restored gut microbiota and alleviated gastrointestinal disorders were observe, suggesting that FMT may serve as a potential therapeutic and rehabilitative intervention for the COVID-19.


Assuntos
Subpopulações de Linfócitos B , COVID-19/complicações , Disbiose/terapia , Transplante de Microbiota Fecal , Gastroenteropatias/terapia , Microbioma Gastrointestinal , Idoso , Bacteroidetes , Bifidobacterium , COVID-19/imunologia , Disbiose/microbiologia , Faecalibacterium , Feminino , Gastroenteropatias/microbiologia , Humanos , Masculino , Pessoa de Meia-Idade , Alta do Paciente , Proteobactérias , SARS-CoV-2 , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA