Your browser doesn't support javascript.
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Phytomedicine ; 101: 154100, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: covidwho-1895371

RESUMEN

BACKGROUND: A number of studies have shown that gastrointestinal manifestations co-exist with respiratory symptoms in coronavirus disease 2019 (COVID-19) patients. Xuanfei Baidu decoction (XFBD) was recommended by the National Health Commission to treat mild and moderate COVID-19 patients and proved to effectively alleviate intestinal symptoms. However, the exact mechanisms remain elusive. PURPOSE: This study aimed at exploring potential mechanisms of XFBD by utilizing a mouse model of dextran sulfate sodium (DSS)-induced acute experimental colitis, mimicking the disease conditions of intestinal microecological disorders. METHODS: The network pharmacology approach was employed to identify the potential targets and pathways of XFBD on the intestinal disorders. Mice with DSS-induced intestinal disorders were utilized to evaluate the protective effect of XFBD in vivo, including body weight, disease activity index (DAI) score, colon length, spleen weight, and serum tumor necrosis factor-α (TNF-α) level. Colon tissues were used to perform hematoxylin-eosin (H&E) staining, western blot analysis, and transcriptome sequencing. Macrophages, neutrophils and the proportions of T helper cell (Th) 1 and Th2 cells were measured by flow cytometry. Intestinal contents were collected for 16S rRNA gene sequencing. RESULTS: Network pharmacology analysis indicated that XFBD inhibited the progression of COVID-19-related intestinal diseases by repressing inflammation. In mice with DSS-induced intestinal inflammation, XFBD treatment significantly reduced weight loss, the spleen index, the disease activity index, TNF-α levels, and colonic tissue damage, and prevented colon shortening. Transcriptomics and flow cytometry results suggested that XFBD remodeled intestinal immunity by downregulating the Th1/Th2 ratio. Western blot analysis showed that XFBD exerted its anti-inflammatory effects by blocking the nuclear factor-κB (NF-κB) signaling pathway. Indicator analysis of microbiota showed that 75 operational taxonomic units (OTUs) were affected after XFBD administration. Among them, Akkermansia, Muribaculaceae, Lachnospiraceae, and Enterorhabdus were simultaneously negatively correlated with intestinal disorders' parameters, and Bacteroides, Escherichia-Shigella, Eubacterium nodatum,Turicibacter, and Clostridium sensu stricto 1, showed positive correlations with intestinal disorders' parameters. CONCLUSIONS: Our data indicate that XFBD treatment attenuated intestinal disorders associated with inhibiting inflammation, remodeling of intestinal immunity, and improving intestinal flora. These findings provide a scientific basis for the clinical use of XFBD and offer a potential therapeutic approach for the treatment of COVID-19 patients with intestinal symptoms.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Colitis Ulcerosa , Colitis , Medicamentos Herbarios Chinos , Microbioma Gastrointestinal , Linfocitos T Reguladores/inmunología , Animales , Colitis/inducido químicamente , Colitis Ulcerosa/inducido químicamente , Colitis Ulcerosa/tratamiento farmacológico , Colitis Ulcerosa/patología , Colon/patología , Sulfato de Dextran/efectos adversos , Modelos Animales de Enfermedad , Medicamentos Herbarios Chinos/uso terapéutico , Humanos , Inflamación/tratamiento farmacológico , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , ARN Ribosómico 16S , Factor de Necrosis Tumoral alfa/metabolismo
2.
Mol Med Rep ; 25(4)2022 04.
Artículo en Inglés | MEDLINE | ID: covidwho-1753714

RESUMEN

Aberrant TGF­ß/Smad7 signaling has been reported to be an important mechanism underlying the pathogenesis of ulcerative colitis. Therefore, the present study aimed to investigate the effects of a number of potential anti­colitis agents on intestinal epithelial permeability and the TGF­ß/Smad7 signaling pathway in an experimental model of colitis. A mouse model of colitis was first established before anti­TNF­α and 5­aminosalicyclic acid (5­ASA) were administered intraperitoneally and orally, respectively. Myeloperoxidase (MPO) activity, histological index (HI) of the colon and the disease activity index (DAI) scores were then detected in each mouse. Transmission electron microscopy (TEM), immunohistochemical and functional tests, including Evans blue (EB) and FITC­dextran (FD­4) staining, were used to evaluate intestinal mucosal permeability. The expression of epithelial phenotype markers E­cadherin, occludin, zona occludens (ZO­1), TGF­ß and Smad7 were measured. In addition, epithelial myosin light chain kinase (MLCK) expression and activity were measured. Anti­TNF­α and 5­ASA treatments was both found to effectively reduce the DAI score and HI, whilst decreasing colonic MPO activity, plasma levels of FD­4 and EB permeation of the intestine. Furthermore, anti­TNF­α and 5­ASA treatments decreased MLCK expression and activity, reduced the expression of Smad7 in the small intestine epithelium, but increased the expression of TGF­ß. In mice with colitis, TEM revealed partial epithelial injury in the ileum, where the number of intercellular tight junctions and the expression levels of E­cadherin, ZO­1 and occludin were decreased, all of which were alleviated by anti­TNF­α and 5­ASA treatment. In conclusion, anti­TNF­α and 5­ASA both exerted protective effects on intestinal epithelial permeability in an experimental mouse model of colitis. The underlying mechanism may be mediated at least in part by the increase in TGF­ß expression and/or the reduction in Smad7 expression, which can inhibit epithelial MLCK activity and in turn reduce mucosal permeability during the pathogenesis of ulcerative colitis.


Asunto(s)
Colitis Ulcerosa/metabolismo , Proteína smad7/genética , Proteína smad7/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Animales , Cadherinas/metabolismo , Colitis Ulcerosa/inducido químicamente , Colon/patología , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Femenino , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Mucosa Intestinal/ultraestructura , Masculino , Mesalamina/administración & dosificación , Ratones Endogámicos C57BL , Quinasa de Cadena Ligera de Miosina/metabolismo , Ocludina/metabolismo , Peroxidasa/efectos de los fármacos , Índice de Severidad de la Enfermedad , Transducción de Señal/efectos de los fármacos , Uniones Estrechas/metabolismo , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Proteína de la Zonula Occludens-1/metabolismo
3.
Cells ; 10(12)2021 11 25.
Artículo en Inglés | MEDLINE | ID: covidwho-1542428

RESUMEN

Acute respiratory distress syndrome (ARDS) is a serious lung condition characterized by severe hypoxemia leading to limitations of oxygen needed for lung function. In this study, we investigated the effect of anandamide (AEA), an endogenous cannabinoid, on Staphylococcal enterotoxin B (SEB)-mediated ARDS in female mice. Single-cell RNA sequencing data showed that the lung epithelial cells from AEA-treated mice showed increased levels of antimicrobial peptides (AMPs) and tight junction proteins. MiSeq sequencing data on 16S RNA and LEfSe analysis demonstrated that SEB caused significant alterations in the microbiota, with increases in pathogenic bacteria in both the lungs and the gut, while treatment with AEA reversed this effect and induced beneficial bacteria. AEA treatment suppressed inflammation both in the lungs as well as gut-associated mesenteric lymph nodes (MLNs). AEA triggered several bacterial species that produced increased levels of short-chain fatty acids (SCFAs), including butyrate. Furthermore, administration of butyrate alone could attenuate SEB-mediated ARDS. Taken together, our data indicate that AEA treatment attenuates SEB-mediated ARDS by suppressing inflammation and preventing dysbiosis, both in the lungs and the gut, through the induction of AMPs, tight junction proteins, and SCFAs that stabilize the gut-lung microbial axis driving immune homeostasis.


Asunto(s)
Ácidos Araquidónicos/uso terapéutico , Endocannabinoides/uso terapéutico , Microbioma Gastrointestinal , Tracto Gastrointestinal/patología , Pulmón/patología , Alcamidas Poliinsaturadas/uso terapéutico , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Síndrome de Dificultad Respiratoria/microbiología , Animales , Péptidos Antimicrobianos/metabolismo , Ácidos Araquidónicos/farmacología , Butiratos/metabolismo , Ciego/patología , Separación Celular , Colon/efectos de los fármacos , Colon/patología , Análisis Discriminante , Disbiosis/complicaciones , Disbiosis/microbiología , Endocannabinoides/farmacología , Enterotoxinas , Femenino , Tracto Gastrointestinal/efectos de los fármacos , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/patología , Activación de Linfocitos/efectos de los fármacos , Ratones Endogámicos C57BL , Neumonía/tratamiento farmacológico , Neumonía/microbiología , Alcamidas Poliinsaturadas/farmacología , Síndrome de Dificultad Respiratoria/complicaciones , Linfocitos T/efectos de los fármacos
4.
J Pediatric Infect Dis Soc ; 9(6): 781-784, 2020 Dec 31.
Artículo en Inglés | MEDLINE | ID: covidwho-1387931

RESUMEN

We describe an 8-week-old infant with severe gastrointestinal symptoms, significant hypoalbuminemia, and mild carditis following asymptomatic infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The infant's symptoms, including their temporal appearance, were consistent with multisystem inflammatory syndrome in children (MIS-C). A unique finding on colonic histology which may shed light on the pathogenesis of MIS-C was identified. The patient improved significantly following several anti-inflammatory treatments. The lag between the presentation of MIS-C and initial SARS-CoV-2 exposure, which may often be asymptomatic, together with the young age of our patient, makes this a challenging diagnosis. Clinicians should be aware of this entity, even in the neonatal and infantile age groups, to facilitate timely identification and treatment.


Asunto(s)
COVID-19/diagnóstico , Síndrome de Respuesta Inflamatoria Sistémica/diagnóstico , COVID-19/patología , COVID-19/terapia , Colon/patología , Femenino , Tracto Gastrointestinal/patología , Humanos , Lactante , SARS-CoV-2 , Síndrome de Respuesta Inflamatoria Sistémica/patología , Síndrome de Respuesta Inflamatoria Sistémica/terapia
5.
Stem Cell Reports ; 16(4): 940-953, 2021 04 13.
Artículo en Inglés | MEDLINE | ID: covidwho-1180038

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection leading to coronavirus disease 2019 (COVID-19) usually results in respiratory disease, but extrapulmonary manifestations are of major clinical interest. Intestinal symptoms of COVID-19 are present in a significant number of patients, and include nausea, diarrhea, and viral RNA shedding in feces. Human induced pluripotent stem cell-derived intestinal organoids (HIOs) represent an inexhaustible cellular resource that could serve as a valuable tool to study SARS-CoV-2 as well as other enteric viruses that infect the intestinal epithelium. Here, we report that SARS-CoV-2 productively infects both proximally and distally patterned HIOs, leading to the release of infectious viral particles while stimulating a robust transcriptomic response, including a significant upregulation of interferon-related genes that appeared to be conserved across multiple epithelial cell types. These findings illuminate a potential inflammatory epithelial-specific signature that may contribute to both the multisystemic nature of COVID-19 as well as its highly variable clinical presentation.


Asunto(s)
COVID-19/patología , Colon/patología , Mucosa Intestinal/patología , Organoides/patología , Línea Celular , Colon/virología , Células Epiteliales/virología , Humanos , Células Madre Pluripotentes Inducidas/citología , Inflamación/virología , Mucosa Intestinal/virología , Modelos Biológicos , Organoides/citología , Organoides/virología , SARS-CoV-2 , Replicación Viral/fisiología
6.
EMBO Mol Med ; 13(4): e13191, 2021 04 09.
Artículo en Inglés | MEDLINE | ID: covidwho-1068062

RESUMEN

SARS-CoV-2, the agent that causes COVID-19, invades epithelial cells, including those of the respiratory and gastrointestinal mucosa, using angiotensin-converting enzyme-2 (ACE2) as a receptor. Subsequent inflammation can promote rapid virus clearance, but severe cases of COVID-19 are characterized by an inefficient immune response that fails to clear the infection. Using primary epithelial organoids from human colon, we explored how the central antiviral mediator IFN-γ, which is elevated in COVID-19, affects epithelial cell differentiation, ACE2 expression, and susceptibility to infection with SARS-CoV-2. In mouse and human colon, ACE2 is mainly expressed by surface enterocytes. Inducing enterocyte differentiation in organoid culture resulted in increased ACE2 production. IFN-γ treatment promoted differentiation into mature KRT20+ enterocytes expressing high levels of ACE2, increased susceptibility to SARS-CoV-2 infection, and resulted in enhanced virus production in infected cells. Similarly, infection-induced epithelial interferon signaling promoted enterocyte maturation and enhanced ACE2 expression. We here reveal a mechanism by which IFN-γ-driven inflammatory responses induce a vulnerable epithelial state with robust replication of SARS-CoV-2, which may have an impact on disease outcome and virus transmission.


Asunto(s)
COVID-19/etiología , Interferón gamma/inmunología , Modelos Inmunológicos , SARS-CoV-2 , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , COVID-19/inmunología , COVID-19/patología , Diferenciación Celular/inmunología , Colon/inmunología , Colon/patología , Colon/virología , Susceptibilidad a Enfermedades , Enterocitos/metabolismo , Enterocitos/patología , Enterocitos/virología , Expresión Génica , Interacciones Microbiota-Huesped/inmunología , Humanos , Interferón gamma/administración & dosificación , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Mucosa Intestinal/virología , Ratones , Organoides/inmunología , Organoides/patología , Organoides/virología , SARS-CoV-2/genética , SARS-CoV-2/inmunología , SARS-CoV-2/patogenicidad , Replicación Viral/inmunología
7.
Chirurgia (Bucur) ; 115(5): 677-680, 2020.
Artículo en Inglés | MEDLINE | ID: covidwho-903188

RESUMEN

The spread of SARS-CoV-2 in Italy has been rapid, with over 230.000 infections and 33.000 deaths (May 31st, 2020). The full impact of COVID19 on surgery is still unknown, as its effects on healthcare strategy, hospital infrastructure, staff, regional economy and colorectal disease progression, may not be evident before several months. No systematic reports are available about a higher incidence of COVID19 infections in patients with cancer. However, available data indicate that older people are more vulnerable, particularly when there are underlying health conditions such as chemotherapy or active cancer. Herein, we present the case of a patient with rectal cancer treated with pull-through technique low anterior rectal resection and coloanal anastomosis with protective loop ileostomy, complicated with Sars-CoV-2 infection and late (31st post-operative day) colic ischemia with colo-vaginal fistula. Late intestinal ischemia is a rare complication and can be secondary to several traditional factors, but certainly small vessel thrombosis related to Coronavirus disease must be taken into consideration.


Asunto(s)
Colon/patología , Infecciones por Coronavirus/complicaciones , Isquemia/cirugía , Neumonía Viral/complicaciones , Fístula Vaginal/cirugía , Anciano , Betacoronavirus , COVID-19 , Colon/cirugía , Femenino , Humanos , Isquemia/complicaciones , Italia , Pandemias , SARS-CoV-2 , Resultado del Tratamiento , Fístula Vaginal/complicaciones
9.
J Crohns Colitis ; 15(3): 485-498, 2021 Mar 05.
Artículo en Inglés | MEDLINE | ID: covidwho-756892

RESUMEN

BACKGROUND: Patients with inflammatory bowel disease [IBD] are considered immunosuppressed, but do not seem more vulnerable for COVID-19. Nevertheless, intestinal inflammation has shown to be an important risk factor for SARS-CoV-2 infection and prognosis. Therefore, we investigated the role of intestinal inflammation on the viral intestinal entry mechanisms, including ACE2, in IBD. METHODS: We collected inflamed and uninflamed mucosal biopsies from Crohn's disease [CD] [n = 193] and ulcerative colitis [UC] [n = 158] patients, and from 51 matched non-IBD controls for RNA sequencing, differential gene expression, and co-expression analysis. Organoids from UC patients were subjected to an inflammatory mix and processed for RNA sequencing. Transmural ileal biopsies were processed for single-cell [sc] sequencing. Publicly available colonic sc-RNA sequencing data, and microarrays from tissue pre/post anti-tumour necrosis factor [TNF] therapy, were analysed. RESULTS: In inflamed CD ileum, ACE2 was significantly decreased compared with control ileum [p = 4.6E-07], whereas colonic ACE2 was higher in inflamed colon of CD/UC compared with control [p = 8.3E-03; p = 1.9E-03]. Sc-RNA sequencing confirmed this ACE2 dysregulation and exclusive epithelial ACE2 expression. Network analyses highlighted HNF4A as key regulator of ileal ACE2, and pro-inflammatory cytokines and interferon regulating factors regulated colonic ACE2. Inflammatory stimuli upregulated ACE2 in UC organoids [p = 1.7E-02], but not in non-IBD controls [p = 9.1E-01]. Anti-TNF therapy restored colonic ACE2 regulation in responders. CONCLUSIONS: Intestinal inflammation alters SARS-CoV-2 coreceptors in the intestine, with opposing dysregulations in ileum and colon. HNF4A, an IBD susceptibility gene, seems an important upstream regulator of ACE2 in ileum, whereas interferon signalling might dominate in colon.


Asunto(s)
Enzima Convertidora de Angiotensina 2/inmunología , COVID-19 , Colitis Ulcerosa , Colon , Enfermedad de Crohn , Factor Nuclear 4 del Hepatocito , Íleon , Interferones/inmunología , SARS-CoV-2/fisiología , Biopsia/métodos , COVID-19/inmunología , COVID-19/patología , COVID-19/fisiopatología , Colitis Ulcerosa/inmunología , Colitis Ulcerosa/patología , Colitis Ulcerosa/virología , Colon/inmunología , Colon/patología , Colon/virología , Enfermedad de Crohn/inmunología , Enfermedad de Crohn/patología , Enfermedad de Crohn/virología , Citocinas/inmunología , Femenino , Regulación de la Expresión Génica , Factor Nuclear 4 del Hepatocito/genética , Factor Nuclear 4 del Hepatocito/inmunología , Humanos , Íleon/inmunología , Íleon/patología , Íleon/virología , Masculino , Persona de Mediana Edad , Análisis de Secuencia de ARN , Transducción de Señal , Análisis de la Célula Individual
11.
Signal Transduct Target Ther ; 5(1): 121, 2020 07 08.
Artículo en Inglés | MEDLINE | ID: covidwho-640304

Asunto(s)
Betacoronavirus/genética , Colon/virología , Infecciones por Coronavirus/genética , Interacciones Huésped-Patógeno/genética , Neumonía Viral/genética , Receptores Virales/genética , Transcriptoma , Adenoma/etnología , Adenoma/genética , Adenoma/patología , Enzima Convertidora de Angiotensina 2 , Pueblo Asiatico , Basigina/genética , Basigina/metabolismo , Betacoronavirus/metabolismo , Betacoronavirus/patogenicidad , COVID-19 , Catepsina B/genética , Catepsina B/metabolismo , Catepsina L/genética , Catepsina L/metabolismo , Colon/metabolismo , Colon/patología , Neoplasias Colorrectales/etnología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Infecciones por Coronavirus/etnología , Infecciones por Coronavirus/patología , Infecciones por Coronavirus/virología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Células Epiteliales/virología , Furina/genética , Furina/metabolismo , Humanos , Interleucina-6/sangre , Interleucina-6/genética , Pandemias , Peptidil-Dipeptidasa A/genética , Peptidil-Dipeptidasa A/metabolismo , Neumonía Viral/etnología , Neumonía Viral/patología , Neumonía Viral/virología , Receptores Virales/metabolismo , SARS-CoV-2 , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Índice de Severidad de la Enfermedad , Análisis de la Célula Individual , Factor de Necrosis Tumoral alfa/sangre , Factor de Necrosis Tumoral alfa/genética , Internalización del Virus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA