Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
mBio ; 15(8): e0172024, 2024 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-38995074

RESUMEN

Infection with the apicomplexan parasite Cryptosporidium is a leading cause of diarrheal disease. Cryptosporidiosis is of particular importance in infants and shows a strong association with malnutrition, both as a risk factor and as a consequence. Cryptosporidium invades and replicates within the small intestine epithelial cells. This is a highly dynamic tissue that is developmentally stratified along the villus axis. New cells emerge from a stem cell niche in the crypt and differentiate into mature epithelial cells while moving toward the villus tip, where they are ultimately shed. Here, we studied the impact of Cryptosporidium infection on this dynamic architecture. Tracing DNA synthesis in pulse-chase experiments in vivo, we quantified the genesis and migration of epithelial cells along the villus. We found proliferation and epithelial migration to be elevated in response to Cryptosporidium infection. Infection also resulted in significant cell loss documented by imaging and molecular assays. Consistent with these observations, single-cell RNA sequencing of infected intestines showed a gain of young and a loss of mature cells. Interestingly, enhanced epithelial cell loss was not a function of enhanced apoptosis of infected cells. To the contrary, Cryptosporidium-infected cells were less likely to be apoptotic than bystanders, and experiments in tissue culture demonstrated that infection provided enhanced resistance to chemically induced apoptosis to the host but not bystander cells. Overall, this study suggests that Cryptosporidium may modulate cell apoptosis and documents pronounced changes in tissue homeostasis due to parasite infection, which may contribute to its long-term impact on the developmental and nutritional state of children. IMPORTANCE: The intestine must balance its roles in digestion and nutrient absorption with the maintenance of an effective barrier to colonization and breach by numerous potential pathogens. An important component of this balance is its constant turnover, which is modulated by a gain of cells due to proliferation and loss due to death or extrusion. Here, we report that Cryptosporidium infection changes the dynamics of this process increasing both gain and loss of enterocytes speeding up the villus elevator. This leads to a much more immature epithelium and a reduction of the number of those cells typically found toward the villus apex best equipped to take up key nutrients including carbohydrates and lipids. These changes in the cellular architecture and physiology of the small intestine may be linked to the profound association between cryptosporidiosis and malnutrition.


Asunto(s)
Criptosporidiosis , Cryptosporidium , Células Epiteliales , Criptosporidiosis/parasitología , Animales , Células Epiteliales/parasitología , Cryptosporidium/genética , Cryptosporidium/fisiología , Ratones , Mucosa Intestinal/parasitología , Apoptosis , Humanos , Proliferación Celular , Movimiento Celular , Intestino Delgado/parasitología
2.
Cell Metab ; 36(8): 1726-1744.e10, 2024 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-38986617

RESUMEN

The intestinal tract generates significant reactive oxygen species (ROS), but the role of T cell antioxidant mechanisms in maintaining intestinal homeostasis is poorly understood. We used T cell-specific ablation of the catalytic subunit of glutamate cysteine ligase (Gclc), which impaired glutathione (GSH) production, crucially reducing IL-22 production by Th17 cells in the lamina propria, which is critical for gut protection. Under steady-state conditions, Gclc deficiency did not alter cytokine secretion; however, C. rodentium infection induced increased ROS and disrupted mitochondrial function and TFAM-driven mitochondrial gene expression, resulting in decreased cellular ATP. These changes impaired the PI3K/AKT/mTOR pathway, reducing phosphorylation of 4E-BP1 and consequently limiting IL-22 translation. The resultant low IL-22 levels led to poor bacterial clearance, severe intestinal damage, and high mortality. Our findings highlight a previously unrecognized, essential role of Th17 cell-intrinsic GSH in promoting mitochondrial function and cellular signaling for IL-22 protein synthesis, which is critical for intestinal integrity and defense against gastrointestinal infections.


Asunto(s)
Glutatión , Interleucina-22 , Interleucinas , Mitocondrias , Células Th17 , Animales , Interleucinas/metabolismo , Mitocondrias/metabolismo , Glutatión/metabolismo , Células Th17/metabolismo , Células Th17/inmunología , Ratones , Transducción de Señal , Especies Reactivas de Oxígeno/metabolismo , Ratones Endogámicos C57BL , Citrobacter rodentium , Intestinos/patología , Intestinos/inmunología , Inflamación/metabolismo , Inflamación/patología , Infecciones por Enterobacteriaceae/inmunología , Infecciones por Enterobacteriaceae/metabolismo , Infecciones por Enterobacteriaceae/patología , Ratones Noqueados , Serina-Treonina Quinasas TOR/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología
3.
Infect Immun ; 92(9): e0048923, 2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-38591884

RESUMEN

Certain Escherichia coli (E. coli) strains are attaching and effacing (A/E) lesion pathogens that primarily infect intestinal epithelial cells. They cause actin restructuring and polymerization within the host cell to create an actin-rich protrusion below the site of adherence, termed the pedestal. Although there is clarity on the pathways initiating pedestal formation, the underlying purpose(s) of the pedestal remains ambiguous. The conservation of pedestal-forming activity across multiple pathogens and redundancy in formation pathways indicate a pathogenic advantage. However, few decisive conclusions have been drawn, given that the results vary between model systems. Some research argues that the pedestal increases the colonization capability of the bacterium. These studies utilize A/E pathogens specifically deficient in pedestal formation to evaluate adhesion and intestinal colonization following infection. There have been many proposed mechanisms for the colonization benefit conferred by the pedestal. One suggested benefit is that the pedestal allows for direct cytosolic anchoring through incorporation of the established host cortical actin, causing a stable link between the pathogen and cell structure. The pedestal may confer enhanced motility, as enteropathogenic E. coli (EPEC) and enterohemorrhagic E. coli (EHEC) are better able to migrate on the surface of host cells and infect neighboring cells in the presence of the pedestal. Additionally, some research suggests that the pedestal improves effector delivery. This review will investigate the purpose of pedestal formation using evidence from recent literature and will critically evaluate the methodology and model systems. Most importantly, we will contextualize the proposed functions to reconcile potential synergistic effects.


Asunto(s)
Adhesión Bacteriana , Infecciones por Escherichia coli , Escherichia coli , Humanos , Adhesión Bacteriana/fisiología , Escherichia coli/patogenicidad , Infecciones por Escherichia coli/microbiología , Actinas/metabolismo , Animales , Interacciones Huésped-Patógeno , Células Epiteliales/microbiología
4.
Virus Res ; 342: 199338, 2024 04.
Artículo en Inglés | MEDLINE | ID: mdl-38373599

RESUMEN

The role of aichivirus A1 (AiV-A1) in acute gastroenteritis remains controversial and in vitro data illustrating its pathogenesis in suitable human models are scarce. Here, we demonstrate that AiV-A1 isolate A846/88 replicates in ApoA1- (absorptive) and Ki-67-positive (proliferative) enterocytes in stem cell-derived human small intestinal epithelium (HIE) as well as in patient biopsy samples, but not in any of the tested human cell lines. The infection did not result in tissue damage and did not trigger type I and type III interferon (IFN) signalling, whereas the control, human coxsackievirus B3 (strain Nancy), triggered both IFNs. To investigate the tissue tropism, we infected a human tracheal/bronchial epithelium model (HTBE) with AiV-A1 isolates A846/88 and kvgh99012632/2010 and, as a control, with rhinovirus A2 (RV-A2). AiV-A1 isolate kvgh99012632/2010, but not isolate A846/88, replicated in HTBE and induced type III IFN and ISGs signalling. By using various pharmacological inhibitors, we elaborated that cellular entry of AiV-A1 depends on clathrin, dynamin, and lipid rafts and is strongly reliant on endosome acidification. Viral particles co-localised with Rab5a-positive endosomes and promoted leakage of endosomal content. Our data shed light on the early events of AiV-A1 infection and reveal that different isolates exhibit distinct tissue tropism. This supports its clinical importance as a human pathogen with the potential to evolve toward broader tissue specificity.


Asunto(s)
Bronquios , Mucosa Intestinal , Humanos , Enterocitos , Línea Celular , Clatrina
5.
Immunology ; 170(2): 167-179, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37132045

RESUMEN

Mucosa-associated invariant T cells (MAIT) are a class of innate-like T lymphocytes mainly presenting CD8+ phenotype with a semi-invariant αß T-cell receptor, which specifically recognises MR1-presented biosynthetic derivatives of riboflavin synthesis produced by various types of microbiomes. As innate-like T lymphocytes, MAIT can be activated by a variety of cytokines, leading to immediate immune responses to infection and tumour cues. As an organ that communicates with the external environment, the digestive tract, especially the gastrointestinal tract, contains abundant microbial populations. Communication between MAIT and local microbiomes is important for the homeostasis of mucosal immunity. In addition, accumulating evidence suggests changes in the abundance and structure of the microbial community during inflammation and tumorigenesis plays a critical role in disease progress partly through their impact on MAIT development and function. Therefore, it is essential for the understanding of MAIT response and their interaction with microbiomes in the digestive tract. Here, we summarised MAIT characteristics in the digestive tract and its alteration facing inflammation and tumour, raising that targeting MAIT can be a candidate for treatment of gastrointestinal diseases.


Asunto(s)
Células T Invariantes Asociadas a Mucosa , Neoplasias , Humanos , Citocinas , Tracto Gastrointestinal , Inflamación
6.
Int J Surg Case Rep ; 105: 108063, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37004449

RESUMEN

BACKGROUND: There is one reported case of inferior mesenteric arteriovenous malformation presenting as ischemic colitis after an episode of gastrointestinal infection. We documented this case to emphasize the possible association between ischemic colitis and vascular malformations. In addition, this is the number 15th case in the literature about this association. CASE SUMMARY: A 40 years old male patient presented with abdominal pain and diarrhea of 10 days duration after he was diagnosed and managed as a case of Clostridium Difficile infection and amebiasis. Computed tomography angiography revealed a vascular malformation of the inferior mesenteric artery, repeated colonoscopy showed ulceration and sloughing of the mucosa, he underwent Hartmann's procedure due to colonic ischemia diagnosed by the previous measures. Later on he had a colostomy closure and end to end colorectal anastomosis. CONCLUSION: There is a possible association between acute gastrointestinal infection and ischemic colitis in the setting of arteriovenous malformation.

7.
World J Clin Cases ; 10(22): 7890-7898, 2022 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-36158489

RESUMEN

BACKGROUND: Leukemic hematopoietic cells acquire enhanced self-renewal capacity and impaired differentiation. The emergence of symptomatic leukemia also requires the acquisition of a clonal proliferative advantage. Untreated leukemia patients usually experience an aggressive process. However, spontaneous remission occasionally occurs in patients with acute myeloid leukemia (AML), most frequently after recovery from a febrile episode, and this is generally attributed to the triggering of antineoplastic immunity. There may be another explanation for the spontaneous remission as implicated in this paper. CASE SUMMARY: A 63-year-old Chinese man presented with high fever, abdominal pain and urticaria-like skin lesions. He was diagnosed with AML-M4 with t(8;21) (q22;q22)/RUNX1-RUNX1T1 based on morphological, immunological, cytogenetic and molecular analyses. He had a complex chromosome rea-rrangement of 48,XY,t(8;21)(q22;q22),+13,+13[9]/49,idem,+mar[9]/49,idem,+8[2]. He also had a mutated tyrosine kinase domain in fms-like tyrosine kinase 3 gene. He was treated with antibiotics and glucocorticoids for gastrointestinal infection and urticaria-like skin lesions. The infection and skin lesions were quickly resolved. Unexpectedly, he achieved hematological remission along with resolution of the febrile episode, gastrointestinal symptoms and skin lesions. Notably, after relapse, repeating these treatments resulted in a return to hematological remission. Unfortunately, he demonstrated strong resistance to antibiotic and glucocorticoid treatment after the second relapse and died of sepsis from bacterial infection with multidrug resistance. The main clinical feature of this patient was that symptomatic AML emerged with flaring of the gut inflammatory disorder and it subsided after resolution of the inflammation. Learning from the present case raises the possibility that in a subgroup of AML patients, the proliferative advantage of leukemia cells may critically require the presence of inflammatory stresses. CONCLUSION: Inflammatory stresses, most likely arising from gastrointestinal infection, may sustain the growth and survival advantage of leukemic cells.

8.
Gut Microbes ; 14(1): 2111950, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35984745

RESUMEN

Melioidosis is a disease caused by the Gram-negative bacillus Burkholderia pseudomallei (Bpm), commonly found in soil and water of endemic areas. Naturally acquired human melioidosis infections can result from either exposure through percutaneous inoculation, inhalation, or ingestion of soil-contaminated food or water. Our prior studies recognized Bpm as an effective enteric pathogen, capable of establishing acute or chronic gastrointestinal infections following oral inoculation. However, the specific mechanisms and virulence factors involved in the pathogenesis of Bpm during intestinal infection are unknown. In our current study, we standardized an in vitro intestinal infection model using primary intestinal epithelial cells (IECs) and demonstrated that Bpm requires a functional T6SS for full virulence. Further, we performed dual RNA-seq analysis on Bpm-infected IECs to evaluate differentially expressed host and bacterial genes in the presence or absence of a T6SS. Our results showed a dysregulation in the TNF-α signaling via NF-κB pathway in the absence of the T6SS, with some of the genes involved in inflammatory processes and cell death also affected. Analysis of the bacterial transcriptome identified virulence factors and regulatory proteins playing a role during infection, with association to the T6SS. By using a Bpm transposon mutant library and isogenic mutants, we showed that deletion of the bicA gene, encoding a putative T3SS/T6SS regulator, ablated intracellular survival and plaque formation by Bpm and impacted survival and virulence when using murine models of acute and chronic gastrointestinal infection. Overall, these results highlight the importance of the type 6 secretion system in the gastrointestinal pathogenesis of Bpm.


Asunto(s)
Burkholderia pseudomallei , Microbioma Gastrointestinal , Melioidosis , Sistemas de Secreción Tipo VI , Factores de Virulencia , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Burkholderia pseudomallei/genética , Burkholderia pseudomallei/metabolismo , Melioidosis/metabolismo , Melioidosis/microbiología , Ratones , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , RNA-Seq , Suelo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Sistemas de Secreción Tipo VI/genética , Sistemas de Secreción Tipo VI/metabolismo , Factores de Virulencia/genética , Factores de Virulencia/metabolismo , Agua
9.
Front Pediatr ; 10: 935483, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36034546

RESUMEN

Background: Non-pharmaceutical interventions (NPI) during the COVID-19 pandemic aimed at prevention of SARS-CoV-2 transmission also influenced transmission of viruses other than SARS-CoV-2. The aim of this study was to describe and compare the burden of common viral respiratory and gastrointestinal infections in children admitted to Berlin University Children's Hospital (BCH) before and during the COVID-19 pandemic at different levels of public NPI measures. Methods: In this retrospective study, we analyzed the frequency of detection of common human respiratory and gastrointestinal viruses from January 2016 through January 2022 in all patients admitted to BCH. We compared virus detection before and during the COVID-19 pandemic at different levels of public NPI measures. Results: The frequency of detection of seasonal enveloped and non-enveloped viruses [Boca-, Corona-, Influenza-, Metapneumo-, Parainfluenza-, Rota-, and Respiratory Syncytial Viruses (RSV)] was diminished during the COVID-19 pandemic, whereas detection rates of non-seasonal viruses (Rhino-/Entero-, and Adenoviruses) were stable during the pandemic. After withdrawal of major NPI measures, we observed an out of season surge of the detection rates of Boca-, Corona-, Parainfluenzaviruses, and RSV. In contrast, no increased detection frequency was observed for Influenza-, Metapneumo-, and Rotaviruses as of January 2022. Conclusion: Corona-, Boca-, Parainfluenzaviruses, and RSV returned as frequently detected pathogens after withdrawal of major NPI measures. The out of season rise might be attributed to an "immune-debt" due to missing contact to viral antigens resulting in waning of population immunity during the COVID-19 pandemic.

10.
Med ; 3(6): 371-387.e9, 2022 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-35434682

RESUMEN

Background: COVID-19 manifests with respiratory, systemic, and gastrointestinal (GI) symptoms.1, SARS-CoV-2 RNA is detected in respiratory and fecal samples, and recent reports demonstrate viral replication in both the lung and intestinal tissue.2, 3, 4 Although much is known about early fecal RNA shedding, little is known about long-term shedding, especially in those with mild COVID-19. Furthermore, most reports of fecal RNA shedding do not correlate these findings with GI symptoms.5. Methods: We analyzed the dynamics of fecal RNA shedding up to 10 months after COVID-19 diagnosis in 113 individuals with mild to moderate disease. We also correlated shedding with disease symptoms. Findings: Fecal SARS-CoV-2 RNA is detected in 49.2% [95% confidence interval, 38.2%-60.3%] of participants within the first week after diagnosis. Whereas there was no ongoing oropharyngeal SARS-CoV-2 RNA shedding in subjects at 4 months, 12.7% [8.5%-18.4%] of participants continued to shed SARS-CoV-2 RNA in the feces at 4 months after diagnosis and 3.8% [2.0%-7.3%] shed at 7 months. Finally, we found that GI symptoms (abdominal pain, nausea, vomiting) are associated with fecal shedding of SARS-CoV-2 RNA. Conclusions: The extended presence of viral RNA in feces, but not in respiratory samples, along with the association of fecal viral RNA shedding with GI symptoms suggest that SARS-CoV-2 infects the GI tract and that this infection can be prolonged in a subset of individuals with COVID-19. Funding: This research was supported by a Stanford ChemH-IMA grant; fellowships from the AACR and NSF; and NIH R01-AI148623, R01-AI143757, and UL1TR003142.


Asunto(s)
COVID-19 , Enfermedades Transmisibles , Enfermedades Gastrointestinales , COVID-19/diagnóstico , Prueba de COVID-19 , Heces , Enfermedades Gastrointestinales/diagnóstico , Humanos , Pulmón , ARN Viral/genética , SARS-CoV-2/genética
11.
J Gastroenterol Hepatol ; 37(7): 1222-1234, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35385602

RESUMEN

Human colonic spirochetosis (CS) is usually due toBrachyspira pilosicolior Brachyspira aalborgiinfection. While traditionally considered to be commensal bacteria, there are scattered case reports and case series of gastrointestinal (GI) symptoms in CS and reports of colonic polyps with adherent spirochetes. We performed a systematic review and meta-analysis investigating the association between CS and GI symptoms and conditions including the irritable bowel syndrome (IBS) and colonic polyps. Following PRISMA 2020 guidelines, a systematic search of Medline, CINAHL, EMBASE, and Web of Science was performed using specific keywords for CS and GI disease. Pooled odds ratios (ORs) and 95% confidence intervals (CIs) were calculated using a random-effects model. Of 75 studies identified in the search, 8 case-control studies met the inclusion criteria for meta-analysis and 67 case series studies met the inclusion criteria for pooled prevalence analysis. CS was significantly associated with diarrhea (n = 141/127, cases/controls, OR: 4.19, 95% CI: 1.72-10.21, P = 0.002) and abdominal pain (n = 64/65, OR: 3.66, 95% CI: 1.43-9.35, P = 0.007). CS cases were significantly more likely to have Rome III-diagnosed IBS (n = 79/48, OR: 3.84, 95% CI: 1.44-10.20, P = 0.007), but not colonic polyps (n = 127/843, OR: 8.78, 95% CI: 0.75-103.36, P = 0.084). In conclusion, we found evidence of associations between CS and both diarrhea and IBS, but not colonic polyps. CS is likely underestimated due to suboptimal diagnostic methods and may be an overlooked risk factor for a subset of IBS patients with diarrhea.


Asunto(s)
Infecciones Bacterianas , Síndrome del Colon Irritable , Diarrea/etiología , Humanos , Intestinos , Síndrome del Colon Irritable/diagnóstico , Síndrome del Colon Irritable/epidemiología , Prevalencia
12.
Biomedicines ; 10(2)2022 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-35203691

RESUMEN

Clostridium butyricum is a human commensal bacterium with beneficial effects including butyrate production, spore formation, increasing levels of beneficial bacteria, and inhibition of pathogenic bacteria. Owing to its preventive and ameliorative effects on gastrointestinal infections, C. butyricum MIYAIRI 588 (CBM 588) has been used as a probiotic in clinical and veterinary medicine for decades. This review summarizes the effects of C. butyricum, including CBM 588, on bacterial gastrointestinal infections. Further, the characteristics of the causative bacteria, examples of clinical and veterinary use, and mechanisms exploited in basic research are presented. C. butyricum is widely effective against Clostoridioides difficile, the causative pathogen of nosocomial infections; Helicobacter pylori, the causative pathogen of gastric cancer; and antibiotic-resistant Escherichia coli. Accordingly, its mechanism is gradually being elucidated. As C. butyricum is effective against gastrointestinal infections caused by antibiotics-induced dysbiosis, it can inhibit the transmission of antibiotic-resistant genes and maintain homeostasis of the gut microbiome. Altogether, C. butyricum is expected to be one of the antimicrobial-resistance (AMR) countermeasures for the One-health approach.

13.
Infect Immun ; 90(2): e0022221, 2022 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-34978927

RESUMEN

Hypoxia-inducible transcription factor 1 (HIF-1) has been shown to enhance microbial killing and ameliorate the course of bacterial infections. While the impact of HIF-1 on inflammatory diseases of the gut has been studied intensively, its function in bacterial infections of the gastrointestinal tract remains largely elusive. With the help of a publicly available gene expression data set, we inferred significant activation of HIF-1 after oral infection of mice with Salmonella enterica serovar Typhimurium. Immunohistochemistry and Western blot analyses confirmed marked HIF-1α protein stabilization, especially in the intestinal epithelium. This prompted us to analyze conditional Hif1a-deficient mice to examine cell type-specific functions of HIF-1 in this model. Our results demonstrate enhanced noncanonical induction of HIF-1 activity upon Salmonella infection in the intestinal epithelium as well as in macrophages. Surprisingly, Hif1a deletion in intestinal epithelial cells did not impact inflammatory gene expression, bacterial spread, or disease outcomes. In contrast, Hif1a deletion in myeloid cells enhanced intestinal Cxcl2 expression and reduced the cecal Salmonella load. In vitro, HIF-1α-deficient macrophages showed overall impaired transcription of mRNA encoding proinflammatory factors; however, the intracellular survival of Salmonella was not impacted by HIF-1α deficiency.


Asunto(s)
Infecciones por Salmonella , Salmonella typhimurium , Animales , Células Epiteliales/microbiología , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Mucosa Intestinal/microbiología , Macrófagos , Ratones , Infecciones por Salmonella/genética , Salmonella typhimurium/genética
14.
Pediatr Int ; 64(1): e14937, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34324763

RESUMEN

BACKGROUND: Coronavirus disease 2019 (COVID-19) has drastically changed the recommended activities and environment for patients worldwide. Our aim was to assess the impact of COVID-19 on pediatric hospitalizations in Kitami, Japan. METHODS: A retrospective, single-center study was conducted on hospitalized patients aged 0-14 years at the Japanese Red Cross Kitami Hospital. We compared the incidence of pediatric patients hospitalized in 2020 with those in 2017-2019. RESULTS: The number of pediatric hospitalized patients dropped significantly in 2020 compared to that in 2017-2019 (median 43.0 vs 78.5 per month, P < 0.001). The patients were significantly older in 2020 (4.3 vs 3.4 years, P < 0.001). Hospitalization from respiratory (8.5 vs 30.5, P < 0.001) and gastrointestinal infections (3.0 vs 6.0, P = 0.004) significantly decreased. Admission due to respiratory syncytial virus (0.0 vs 4.0, P < 0.001), human metapneumovirus (0.0 vs 1.0, P = 0.005), influenza (0.0 vs 0.0, P = 0.009), adenovirus (0.0 vs 1.0, P = 0.003), and rotavirus infection (0.0 vs 0.0, P = 0.025) also decreased significantly. The <1-5 age groups significantly decreased (<1 year old, 6.5 vs 12.5, P < 0.001; 1-3 years old, 13.0 vs 29.5, P < 0.001; 4-5 years old, 5.5 vs 11.5, P < 0.001). Hospitalization due to foreign body ingestions increased significantly in 2020 (1.0 vs 0.0, P = 0.010). CONCLUSIONS: The COVID-19 control measures inadvertently reduced the number of hospitalized pediatric patients, especially younger children with respiratory and gastrointestinal infections.


Asunto(s)
COVID-19 , Enfermedades Transmisibles , Virus Sincitial Respiratorio Humano , Infecciones del Sistema Respiratorio , COVID-19/epidemiología , Niño , Preescolar , Enfermedades Transmisibles/epidemiología , Hospitalización , Humanos , Lactante , Japón/epidemiología , Pandemias , Infecciones del Sistema Respiratorio/epidemiología , Estudios Retrospectivos
15.
Cureus ; 13(9): e17893, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34660091

RESUMEN

A 45-year-old female who was a teacher by profession with a history of chronic asymptomatic anemia in the past presented to our hospital with complaints of intermittent fever for two weeks, cough with expectoration, dyspnea on exertion, and left upper limb edema for four days. She had a history of abdominal pain after food intake. She gave a history of having anemia for the past 23 years. Evaluation after admission showed raised inflammatory markers, marked thrombocytosis, and severe iron deficiency anemia. Further imaging in the form of a CT of the abdomen and thorax showed that she had a left-sided pleural effusion which showed an exudative picture, splenomegaly with a splenic infarct with a splenic abscess, and a suprarenal abdominal aorta thrombus. She was also found to have deep vein thrombosis (DVT) of the left subclavian and proximal internal jugular vein in a ultrasonogram (USG) Doppler. The workup done ruled out congenital and acquired causes of thrombosis and after extensive evaluation the patient was found to have unexplained thrombosis. The cause of unexplained thrombosis is the point of interest in this patient. Despite extensive workup, no precise cause for thrombosis, which was both arterial and venous in nature could be found out initially. Hence by exclusion, the possibility of secondary thrombocytosis causing the thrombosis was considered. Over the next few years, this patient underwent repeat esophageal endoscopies, colonoscopies, and capsule studies all without being able to pinpoint a diagnosis. Eventually three years later, a CT enteroscopy with biopsy showed the diagnosis of Crohn's disease and the patient was started on appropriate immunosuppressive treatment for the same. There have been multiple case reports of thrombocytosis causing arterial or venous thrombus but not many have recorded both arterial and venous thrombosis in the same patient.

16.
mBio ; 12(5): e0212721, 2021 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-34488445

RESUMEN

Interferon (IFN) signaling is key to mucosal immunity in the gastrointestinal tract, but cellular regulatory elements that determine interferon gamma (IFN-γ)-mediated antimicrobial defense in intestinal epithelial cells are not fully understood. We report here that a long noncoding RNA (lncRNA), GenBank accession no. XR_001779380, was increased in abundance in murine intestinal epithelial cells following infection by Cryptosporidium, an important opportunistic pathogen in AIDS patients and a common cause of diarrhea in young children. Expression of XR_001779380 in infected intestinal epithelial cells was triggered by TLR4/NF-κB/Cdc42 signaling and epithelial-specific transcription factor Elf3. XR_001779380 primed epithelial cells for IFN-γ-mediated gene transcription through facilitating Stat1/Swi/Snf-associated chromatin remodeling. Interactions between XR_001779380 and Prdm1, which is expressed in neonatal but not adult intestinal epithelium, attenuated Stat1/Swi/Snf-associated chromatin remodeling induced by IFN-γ, contributing to suppression of IFN-γ-mediated epithelial defense in neonatal intestine. Our data demonstrate that XR_001779380 is an important regulator in IFN-γ-mediated gene transcription and age-associated intestinal epithelial antimicrobial defense. IMPORTANCE Epithelial cells along the mucosal surface provide the front line of defense against luminal pathogen infection in the gastrointestinal tract. These epithelial cells represent an integral component of a highly regulated communication network that can transmit essential signals to cells in the underlying intestinal mucosa that, in turn, serve as targets of mucosal immune mediators. LncRNAs are recently identified long noncoding transcripts that can regulate gene transcription through their interactions with other effect molecules. In this study, we demonstrated that lncRNA XR_001779380 was upregulated in murine intestinal epithelial cells following infection by a mucosal protozoan parasite Cryptosporidium. Expression of XR_001779380 in infected cells primed host epithelial cells for IFN-γ-mediated gene transcription, relevant to age-dependent intestinal antimicrobial defense. Our data provide new mechanistic insights into how intestinal epithelial cells orchestrate intestinal mucosal defense against microbial infection.


Asunto(s)
Criptosporidiosis/inmunología , Cryptosporidium parvum/fisiología , Interferón gamma/inmunología , Mucosa Intestinal/inmunología , ARN Largo no Codificante/inmunología , Factores de Edad , Animales , Criptosporidiosis/genética , Criptosporidiosis/parasitología , Cryptosporidium parvum/genética , Células Epiteliales/inmunología , Células Epiteliales/parasitología , Humanos , Inmunidad Mucosa , Interferón gamma/genética , Mucosa Intestinal/parasitología , Ratones , FN-kappa B/genética , FN-kappa B/inmunología , ARN Largo no Codificante/genética , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología
17.
J Med Microbiol ; 70(9)2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34516365

RESUMEN

Introduction. The identification of enteropathogens is critical for the clinical management of patients with suspected gastrointestinal infection. The FLOW multiplex PCR system (FMPS) is a semi-automated platform (FLOW System, Roche) for multiplex real-time PCR analysis.Hypothesis/Gap Statement. FMPS has greater sensitivity for the detection of enteric pathogens than standard methods such as culture, biochemical identification, immunochromatography or microscopic examination.Aim.The diagnostic performance of the FMPS was evaluated and compared to that of traditional microbiological procedures.Methodology. A total of 10 659 samples were collected and analysed over a period of 7 years. From 2013 to 2018 (every July to September), samples were processed using standard microbiological culture methods. In 2019, the FMPS was implemented using real-time PCR to detect the following enteropathogens: Shigella spp., Salmonella spp., Campylobacter spp., Giardia intestinalis, Entamoeba histolytica, Blastocystis hominis, Cryptosporidum spp., Dientamoeba fragilis, adenovirus, norovirus and rotavirus. Standard microbiological culture methods (2013-2018) included stool culture, microscopy and immunochromatography.Results. A total of 1078 stool samples were analysed prospectively using the FMPS from July to September (2019): bacterial, parasitic and viral pathogens were identified in 15.3, 9.71 and 5.29 % of cases, respectively. During the same period of 6 years (2013-2018), the proportion of positive identifications using standard microbiological methods from 2013 to 2018 was significantly lower. A major significant recovery improvement was observed for all bacteria species tested: Shigella spp./enteroinvasive Escherichia coli (EIEC) (P <0.05), Salmonella spp. (P <0.05) and Campylobacter spp. (P <0.05). Marked differences were also observed for the parasites G. intestinalis, Cryptosporidium spp. and D. fragilis.Conclusion. These results support the value of multiplex real-time PCR analysis for the detection of enteric pathogens in laboratory diagnosis with outstanding performance in identifying labile micro-organisms. The identification of unsuspected micro-organisms for less specific clinical presentations may also impact on clinical practice and help optimize patient management.


Asunto(s)
Gastroenteritis/diagnóstico , Reacción en Cadena de la Polimerasa Multiplex , Reacción en Cadena en Tiempo Real de la Polimerasa , Adenoviridae/aislamiento & purificación , Blastocystis hominis/aislamiento & purificación , Campylobacter/aislamiento & purificación , Cryptosporidium/aislamiento & purificación , Dientamoeba/aislamiento & purificación , Entamoeba histolytica/aislamiento & purificación , Heces/microbiología , Heces/parasitología , Heces/virología , Gastroenteritis/microbiología , Gastroenteritis/parasitología , Giardia lamblia/aislamiento & purificación , Humanos , Norovirus/aislamiento & purificación , Rotavirus/aislamiento & purificación , Salmonella/aislamiento & purificación , Shigella/aislamiento & purificación
18.
Viruses ; 13(6)2021 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-34071855

RESUMEN

Laboratory cultivation of viruses is critical for determining requirements for viral replication, developing detection methods, identifying drug targets, and developing antivirals. Several viruses have a history of recalcitrance towards robust replication in laboratory cell lines, including human noroviruses and hepatitis B and C viruses. These viruses have tropism for tissue components of the enterohepatic circulation system: the intestine and liver, respectively. The purpose of this review is to discuss how key enterohepatic signaling molecules, bile acids (BAs), and BA receptors are involved in the replication of these viruses and how manipulation of these factors was useful in the development and/or optimization of culture systems for these viruses. BAs have replication-promoting activities through several key mechanisms: (1) affecting cellular uptake, membrane lipid composition, and endocytic acidification; (2) directly interacting with viral capsids to influence binding to cells; and (3) modulating the innate immune response. Additionally, expression of the Na+-taurocholate cotransporting polypeptide BA receptor in continuous liver cell lines is critical for hepatitis B virus entry and robust replication in laboratory culture. Viruses are capable of hijacking normal cellular functions, and understanding the role of BAs and BA receptors, components of the enterohepatic system, is valuable for expanding our knowledge on the mechanisms of norovirus and hepatitis B and C virus replication.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Bilis/metabolismo , Enfermedades Gastrointestinales/virología , Virus de la Hepatitis B/fisiología , Norovirus/fisiología , Replicación Viral/efectos de los fármacos , Ácidos y Sales Biliares/farmacología , Humanos , Hígado/metabolismo , Hígado/virología , Internalización del Virus/efectos de los fármacos
19.
Viruses ; 13(6)2021 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-34071878

RESUMEN

Historically, knowledge of human host-enteric pathogen interactions has been elucidated from studies using cancer cells, animal models, clinical data, and occasionally, controlled human infection models. Although much has been learned from these studies, an understanding of the complex interactions between human viruses and the human intestinal epithelium was initially limited by the lack of nontransformed culture systems, which recapitulate the relevant heterogenous cell types that comprise the intestinal villus epithelium. New investigations using multicellular, physiologically active, organotypic cultures produced from intestinal stem cells isolated from biopsies or surgical specimens provide an exciting new avenue for understanding human specific pathogens and revealing previously unknown host-microbe interactions that affect replication and outcomes of human infections. Here, we summarize recent biologic discoveries using human intestinal organoids and human enteric viral pathogens.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Enfermedades Gastrointestinales/virología , Tracto Gastrointestinal/virología , Interacciones Huésped-Patógeno , Organoides/virología , Virus/patogenicidad , Humanos , Células Madre , Virus/genética
20.
Gastroenterol Clin North Am ; 50(2): 415-430, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34024449

RESUMEN

Infectious gastroenteritis is common after transplantation and can lead to increased morbidity and mortality. A wide range of organisms can lead to gastroenteritis in this patient population. Clostridioides difficile, cytomegalovirus, and norovirus are the most common pathogens. Newer diagnostic methods, especially multiplex polymerase chain reaction, have increased the diagnostic yield of infectious etiologies. In this review, we describe the epidemiology and risk factors for common infectious pathogens leading to gastroenteritis.


Asunto(s)
Gastroenteritis , Trasplante de Células Madre Hematopoyéticas , Norovirus , Trasplante de Órganos , Diarrea , Gastroenteritis/diagnóstico , Gastroenteritis/epidemiología , Gastroenteritis/etiología , Humanos , Norovirus/genética , Trasplante de Órganos/efectos adversos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA