Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Immunol ; 212(5): 881-893, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38189569

RESUMEN

MicroRNAs (miRNAs) are small noncoding RNA molecules that negatively regulate gene expression. Within the intestinal epithelium, miRNAs play a critical role in gut homeostasis, and aberrant miRNA expression has been implicated in various disorders associated with intestinal inflammation and barrier disruption. In this study, we sought to profile changes in intestinal epithelial cell miRNA expression after alcohol and burn injury and elucidate their impact on inflammation and barrier integrity. Using a mouse model of acute ethanol intoxication and burn injury, we found that small intestinal epithelial cell expression of miR-146a is significantly decreased 1 d following injury. Using in vitro studies, we show that reduced miR-146a promotes intestinal epithelial cell inflammation by promoting p38 MAPK signaling via increased levels of its target TRAF6 (TNFR-associated factor 6). Furthermore, we demonstrate that in vivo miR-146a overexpression significantly inhibits intestinal inflammation 1 d following combined injury and potentially supports intestinal barrier homeostasis. Overall, this study highlights the important impact that miRNA expression can have on intestinal homeostasis and the valuable potential of harnessing aberrant miRNA expression as a therapeutic target to control intestinal inflammation.


Asunto(s)
Quemaduras , MicroARNs , Humanos , MicroARNs/metabolismo , Etanol , Inflamación/genética , Células Epiteliales/metabolismo , Quemaduras/complicaciones
2.
Shock ; 59(4): 621-626, 2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-36645886

RESUMEN

ABSTRACT: Background: Traumatic brain injury (TBI) is a significant cause of morbidity and mortality in the United States, with an annual cost of 60 billion dollars. There is evidence suggesting that in the post-TBI period, the gastrointestinal tract plays a central role in driving organ and immune dysfunction and may be the source of increased circulating proinflammatory mediators. In this study, we examined systemic inflammation and bacterial dysbiosis in patients who sustained a TBI with or without polytrauma. Using a mouse model of TBI, we further show how neuroinflammation after TBI is potentially linked to disruptions in gut homeostasis such as intestinal transit and inflammation. Methods: During a study of trauma patients performed from September 1, 2018, to September 1, 2019, at a single, level 1 trauma center, TBI patients aged 21 to 95 years were enrolled. Patients were categorized as TBI based on evidence of acute abnormal findings on head computed tomographic scan, which was a combination of isolated TBI and TBI with polytrauma. Blood and stool samples were collected between 24 h and 3 days after admission. Twelve plasma samples and 10 fecal samples were used for this study. Healthy control samples were obtained from a healthy control biobank. We examined systemic inflammation and bacterial changes in patients who sustained a TBI. In addition, TBI was induced in 9- to 10-week-old male mice; we assessed neuroinflammation, and intestine transit (motility) and bacterial changes 24 h after TBI. Results: When compared with healthy controls, TBI patients had increased systemic inflammation as evidenced by increased levels of IFN-γ and MCP-1 and a trend toward an increase of IL-6 and IL-8 ( P = 0.0551 and P = 0.0549), respectively. The anti-inflammatory cytokine, IL-4, was also decreased in TBI patients. Although there was a trend of an increase in copy number of Enterobacteriaceae and a decrease in copy number of Lactobacillus in both patients and mice after TBI, these trends were not found to be significantly different. However, TBI significantly increased the copy number of another potential pathogenic bacteria Bilophila wadsworthia in TBI patients compared with healthy controls. After a moderate TBI, mice had increased expression of TNF-α, IL-6 and IL-1ß, CXCL1, s100a9, and Ly6G and decreased IL-10 in the brain lesion after TBI. This accompanied decreased transit and increased TNF-α in the small intestine of mice after TBI. Conclusions: Our findings suggest that TBI increases systemic inflammation, intestinal dysfunction, and neuroinflammation. More studies are needed to confirm whether changes in intestinal motility play a role in post-TBI neuroinflammation and cognitive deficit.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Traumatismo Múltiple , Masculino , Humanos , Interleucina-6 , Factor de Necrosis Tumoral alfa , Enfermedades Neuroinflamatorias , Lesiones Traumáticas del Encéfalo/complicaciones , Inflamación , Traumatismo Múltiple/complicaciones
3.
J Leukoc Biol ; 112(6): 1471-1484, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35916052

RESUMEN

Ulcerative colitis (UC) is characterized by cycles of active disease flare and inactive disease remission. During UC remission, IL-22 is up-regulated, acting as a hallmark of entrance into UC remission. Recently, we found that in our mouse model of binge alcohol and dextran sodium sulfate (DSS)-induced colitis, alcohol increases severity of UC pathology. In this study, we assessed not only whether alcohol influenced IL-22 expression and thereby perpetuates UC, but also whether recombinant IL-22 (rIL-22) or treatment with a probiotic could alleviate exacerbated symptoms of UC. Levels of large intestine IL-22 were significantly decreased ∼6.9-fold in DSS ethanol compared with DSS vehicle. Examination of lamina propria (LP) cells in the large intestine revealed IL-22+ γδ T cells in DSS vehicle-treated mice were significantly increased, while IL-22+ γδ T cells in DSS ethanol mice were unable to mount this IL-22 response. We administered rIL-22 and found it restored weight loss of DSS ethanol-treated mice. Colonic shortening and increased Enterobacteriaceae were also attenuated. Administration of Lactobacillus delbrueckii attenuated weight loss (p < 0.01), colon length (p < 0.001), mitigated increases in Enterobacteriaceae, increased levels of IL-22, and increased levels of p-STAT3 back to that of DSS vehicle group in DSS ethanol mice. In contrast, sole administration of L. delbrueckii supernatant was not sufficient to reduce UC exacerbation following alcohol. Our findings suggest L. delbrueckii contributes to repair mechanisms by increasing levels of IL-22, resulting in phosphorylation of STAT3, thus attenuating the alcohol-induced increases in intestinal damage after colitis.


Asunto(s)
Colitis Ulcerosa , Colitis , Lactobacillus delbrueckii , Ratones , Animales , Sulfato de Dextran/toxicidad , Colitis/patología , Colon/patología , Colitis Ulcerosa/inducido químicamente , Colitis Ulcerosa/patología , Modelos Animales de Enfermedad , Etanol/efectos adversos , Pérdida de Peso , Ratones Endogámicos C57BL , Interleucina-22
4.
Immunohorizons ; 6(8): 600-613, 2022 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-35973751

RESUMEN

Alcohol intoxication combined with burn injury can lead to life-threatening complications, including sepsis, multiple organ failure, and death. After an acute burn, the gastrointestinal system becomes hypoxic because of fluid loss and reduction of intestinal blood flow. This can cause perturbations in the intestinal epithelial barrier, immune function, and the composition of the gut microbiome. Increased gut permeability leads to proinflammatory signaling, contributing to further damage to the intestinal barrier. Recent studies have suggested that IL-27 plays an anti-inflammatory role, which may be beneficial in intestinal barrier repair. Therefore, in this study, we examined the effect of ethanol and burn injury on IL-27 in the small intestine, as well as the potential beneficial role of IL-27 in restoring the intestinal barrier after intoxication and burn. Male C57BL/6 mice were gavaged with 2.9 g/kg ethanol before receiving a ∼12.5% total body surface area scald burn with or without rIL-27 in resuscitation fluid. Our results demonstrate that IL-27-producing cells are reduced in the small intestine after injury. When IL-27 is supplemented in resuscitation fluid, we were able to restore intestinal barrier integrity and transit, mediated through increased intestinal epithelial cell proliferation, reduced inflammatory cytokines, and increased anti-inflammatory cytokine IL-10. We also observed increased gene expression of tight junction proteins. These findings suggest that IL-27 may be a contributor to maintaining proper intestinal barrier function after injury through multiple mechanisms, including preventing excess inflammation and promoting intestinal epithelial cell proliferation and tight junction integrity.


Asunto(s)
Intoxicación Alcohólica , Quemaduras , Interleucina-27 , Interleucinas , Intoxicación Alcohólica/complicaciones , Intoxicación Alcohólica/metabolismo , Animales , Quemaduras/complicaciones , Quemaduras/metabolismo , Citocinas/metabolismo , Etanol , Interleucinas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL
5.
Immunohorizons ; 6(1): 64-75, 2022 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-35058308

RESUMEN

Our previous studies have shown that ethanol intoxication combined with burn injury increases intestinal bacterial growth, disrupts the intestinal barrier, and enhances bacterial translocation. Additionally, studies show that Th17 effector cytokines IL-17 and IL-22, which are dependent on IL-23, play important roles in maintaining intestine mucosal barrier integrity. Recent findings suggest neutrophils are a significant source of IL-17 and IL-22. We determined the effect of ethanol and burn injury on neutrophil IL-17 and IL-22 production, as well as their ability to phagocytose and in bacterial clearance, and whether these effects are modulated by IL-23. Mice were given ethanol 4 h prior to receiving ∼12.5% total body surface area burn and were euthanized day 1 after injury. We observed that intoxication combined with burn injury significantly decreases blood neutrophil phagocytosis and bacteria killing, as well as their ability to produce IL-17 and IL-22, compared with sham vehicle mice. The treatment of neutrophils with rIL-23 significantly increases IL-22 and IL-17 release and promotes expression of IL-23R, retinoic acid-related orphan receptor γt, Lipocalin2, and Nod-like receptor 2 following ethanol and burn injury. Furthermore, IL-22- and IL-17-producing neutrophils have enhanced neutrophil extracellular trap formation and bacterial killing ability, which are dependent on IL-23. Finally, although we observed that peritoneal neutrophils harvested after casein treatment are functionally different from blood neutrophils, both blood and peritoneal neutrophils exhibited the same response to rIL-23 treatment. Together these findings suggest that IL-23 promotes neutrophil IL-22 and IL-17 production and their ability to kill bacteria following ethanol and burn injury.


Asunto(s)
Intoxicación Alcohólica/metabolismo , Quemaduras/metabolismo , Interleucina-17/metabolismo , Interleucinas/metabolismo , Neutrófilos/metabolismo , Intoxicación Alcohólica/microbiología , Animales , Quemaduras/patología , Modelos Animales de Enfermedad , Infecciones por Escherichia coli/microbiología , Etanol/toxicidad , Trampas Extracelulares/metabolismo , Mucosa Intestinal/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Fagocitosis , Interleucina-22
6.
J Neuroinflammation ; 18(1): 263, 2021 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-34758843

RESUMEN

BACKGROUND: Inflammatory bowel disease (IBD) is established to drive pathological sequelae in organ systems outside the intestine, including the central nervous system (CNS). Many patients exhibit cognitive deficits, particularly during disease flare. The connection between colonic inflammation and neuroinflammation remains unclear and characterization of the neuroinflammatory phenotype in the brain during colitis is ill-defined. METHODS: Transgenic mice expressing a bioluminescent reporter of active caspase-1 were treated with 2% dextran sodium sulfate (DSS) for 7 days to induce acute colitis, and colonic, systemic and neuroinflammation were assessed. In some experiments, mice were prophylactically treated with paquinimod (ABR-215757) to inhibit S100A9 inflammatory signaling. As a positive control for peripheral-induced neuroinflammation, mice were injected with lipopolysaccharide (LPS). Colonic, systemic and brain inflammatory cytokines and chemokines were measured by cytokine bead array (CBA) and Proteome profiler mouse cytokine array. Bioluminescence was quantified in the brain and caspase activation was confirmed by immunoblot. Immune cell infiltration into the CNS was measured by flow cytometry, while light sheet microscopy was used to monitor changes in resident microglia localization in intact brains during DSS or LPS-induced neuroinflammation. RNA sequencing was performed to identify transcriptomic changes occurring in the CNS of DSS-treated mice. Expression of inflammatory biomarkers were quantified in the brain and serum by qRT-PCR, ELISA and WB. RESULTS: DSS-treated mice exhibited clinical hallmarks of colitis, including weight loss, colonic shortening and inflammation in the colon. We also detected a significant increase in inflammatory cytokines in the serum and brain, as well as caspase and microglia activation in the brain of mice with ongoing colitis. RNA sequencing of brains isolated from DSS-treated mice revealed differential expression of genes involved in the regulation of inflammatory responses. This inflammatory phenotype was similar to the signature detected in LPS-treated mice, albeit less robust and transient, as inflammatory gene expression returned to baseline following cessation of DSS. Pharmacological inhibition of S100A9, one of the transcripts identified by RNA sequencing, attenuated colitis severity and systemic and neuroinflammation. CONCLUSIONS: Our findings suggest that local inflammation in the colon drives systemic inflammation and neuroinflammation, and this can be ameliorated by inhibition of the S100 alarmin, S100A9.


Asunto(s)
Encéfalo/fisiopatología , Calgranulina B/genética , Colitis/inducido químicamente , Colitis/prevención & control , Enfermedades Neuroinflamatorias/prevención & control , Enfermedades Neuroinflamatorias/fisiopatología , Quinolinas/uso terapéutico , Animales , Biomarcadores , Caspasa 1/metabolismo , Quimiocinas/metabolismo , Colitis/fisiopatología , Citocinas/metabolismo , Sulfato de Dextran , Humanos , Lipopolisacáridos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
7.
J Leukoc Biol ; 109(6): 1121-1130, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-32964503

RESUMEN

Ethanol remains a confounder in postburn pathology, which is associated with an impaired intestinal barrier. Previously, we demonstrated that ethanol and burn injury reduce intestinal oxygen delivery (hypoxia) and alters microRNA (miR) expression in small intestinal epithelial cells. Hypoxia has been shown to influence expression of miRs and miR biogenesis components. Therefore, we examined whether hypoxia influences expression of miR biogenesis components (drosha, dicer, and argonaute-2 [ago-2]) and miRs (-7a and -150) and whether these changes impacted other parameters following ethanol and burn injury. Mice were gavaged with ethanol (∼2.9 g/kg) 4 h before receiving a ∼12.5% total body surface full thickness burn. Mice were resuscitated at the time of injury with normal saline with or without 5 mg/kg PX-478, a hypoxia-inducible factor-1α inhibitor. One day following injury mice were euthanized, and the expression of miRs and their biogenesis components as well as bacterial growth, tight junction proteins, intestinal transit, and permeability were assessed. Ethanol combined with burn injury significantly reduced expression of drosha, ago-2, miRs (-7a and -150), occludin, zonula occludens-1, claudin-4, zonula occludens-1, mucins-2 and -4, and intestinal transit compared to shams. Furthermore, there was an increase in intestinal permeability, total bacteria, and Enterobacteriaceae populations following the combined injury compared to shams. PX-478 treatment improved expression of drosha, ago-2, miRs (-7a and -150), occludin, claudin-4, zonula occludens-1, and mucin-2. PX-478 treatment also improved intestinal transit and reduced dysbiosis and permeability. These data suggest that PX-478 improves miR biogenesis and miR expression, and restores barrier integrity while reducing bacterial dysbiosis following ethanol and burn injury.


Asunto(s)
Quemaduras/tratamiento farmacológico , Inhibidores Enzimáticos/farmacología , Etanol/efectos adversos , Mucosa Intestinal/efectos de los fármacos , Compuestos de Mostaza/farmacología , Fenilpropionatos/farmacología , Sustancias Protectoras/farmacología , Intoxicación Alcohólica , Animales , Proteínas Argonautas/genética , Biomarcadores , Quemaduras/etiología , Quemaduras/metabolismo , Susceptibilidad a Enfermedades , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Masculino , Ratones , MicroARNs/genética , ARN Mensajero/genética , ARN Ribosómico 16S/genética
8.
Gut Microbes ; 12(1): 1-16, 2020 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-33180663

RESUMEN

Alcohol can potentiate disease in a mouse model of dextran sodium sulfate (DSS) colitis; however, the underlying mechanism remains to be established. In this study, we assessed whether the potentiated disease could be related to Enterobacteriaceae and Lactobacillus, as changes in their relative abundance can impact intestinal health. We also assessed whether the intestinal barrier is compromised after alcohol and DSS as it may increase bacterial translocation and liver inflammation. Mice were administered DSS followed by binge ethanol or water vehicle, generating four experimental groups: (Control+Vehicle, Control+Ethanol, DSS+Vehicle, DSS+Ethanol). DNA was isolated from colon and cecal contents followed by qPCR for levels of Enterobacteriaceae and Lactobacillus. Colon and liver sections were taken for histology. Intestinal epithelial cells were isolated from the colon for RNA expression. DSS+Ethanol cecal contents exhibited a 1 log increase in Enterobacteriaceae (p < .05), a 0.5 log decrease in Lactobacillus, and a 1.5 log decrease (p < .05) in the Lactobacillus:Enterobacteriaceae ratio compared to DSS+Vehicle, with similar trends in colon contents. These changes correlated with shorter colons and more weight loss. Irrespective of ethanol administration, DSS compromised the mucosal barrier integrity, however only DSS+Ethanol exhibited significant increases in circulating endotoxin. Furthermore, the livers of DSS+Ethanol mice had significantly increased levels of triglycerides, mononuclear cells, yet exhibited significantly depressed expression of liver inflammatory pathways, suggestive of tolerance induction, compared to mice receiving DSS+Vehicle. Our results suggest that ethanol after DSS colitis increases the intestinal burden of Enterobacteriaceae which may contribute to intestinal and liver damage, and the induction of immune tolerance.


Asunto(s)
Colitis/inmunología , Enterobacteriaceae/aislamiento & purificación , Etanol/farmacología , Tolerancia Inmunológica/inmunología , Mucosa Intestinal/inmunología , Lactobacillus/aislamiento & purificación , Animales , Carga Bacteriana , Colitis/inducido químicamente , Colitis/microbiología , Sulfato de Dextran , Modelos Animales de Enfermedad , Endotoxinas/sangre , Mucosa Intestinal/microbiología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Uniones Estrechas/fisiología , Triglicéridos/sangre
9.
Biochim Biophys Acta Mol Basis Dis ; 1866(11): 165901, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32711051

RESUMEN

6-Formylindolo (3, 2-b) Carbazole (FICZ) is a ligand of aryl hydrocarbon receptor (AHR) which regulates Th17 release of IL-17 and IL-22 production. Earlier, we showed that ethanol combined with burn injury suppresses Th17 responses and disrupts intestinal barrier leading to increased gut bacterial growth and translocation. Since IL-22 is known for its role in intestinal barrier maintenance, we determined whether treatment of mice with FICZ restores T cell IL-22 release and protects intestine barrier following ethanol and burn injury. Wildtype and Rag1-/- mice were gavaged with ~2.9 g/kg ethanol or water, and given a ~12.5% total body surface area burn. Mice were given FICZ (5 µg) in resuscitation fluid. FICZ treatment of wildtype mice normalized IL-22 and IL-17 in lamina propria and spleen T cells, as well as increased CYP1A1 expression in spleen T cells. This was accompanied by improved gut motility, decreased copy number of small intestine total bacteria and Enterobacteriaceae, attenuation of intestinal tissue levels of IL-6, KC, IL-18, decreased apoptosis, and prevention of gut leakiness following ethanol and burn injury. However, FICZ treatment of Rag1-/- mice did not improve any of the parameters listed after ethanol and burn injury. Additional data generated using mice treated with recombinant IL-22 alone or in combination with anti-IL-18 antibody suggest that full protection of gut barrier integrity requires both IL-18 inhibition and IL-22 restoration following ethanol and burn injury. Together our findings suggest that AHR ligand FICZ may have better therapeutic potential for maintenance of gut barrier function after ethanol and burn injury.


Asunto(s)
Quemaduras/metabolismo , Carbazoles/uso terapéutico , Citocinas/metabolismo , Etanol/farmacología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Animales , Quemaduras/tratamiento farmacológico , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1A1/metabolismo , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Interleucina-17/metabolismo , Interleucinas/metabolismo , Mucosa Intestinal/microbiología , Intestino Delgado/efectos de los fármacos , Intestino Delgado/metabolismo , Intestino Delgado/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Membrana Mucosa/efectos de los fármacos , Membrana Mucosa/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo , Interleucina-22
10.
J Immunol ; 203(9): 2497-2507, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31562211

RESUMEN

Inflammasomes are multiprotein complexes that coordinate cellular inflammatory responses and mediate host defense. Following recognition of pathogens and danger signals, inflammasomes assemble and recruit and activate caspase-1, the cysteine protease that cleaves numerous downstream targets, including pro-IL-1ß and pro-IL-18 into their biologically active form. In this study, we sought to develop a biosensor that would allow us to monitor the initiation, progression, and resolution of inflammation in living animals. To this end, we inserted a known caspase-1 target sequence into a circularly permuted luciferase construct that becomes bioluminescent upon protease cleavage. This biosensor was activated in response to various inflammatory stimuli in human monocytic cell lines and murine bone marrow-derived macrophages. Next, we generated C57BL/6 transgenic mice constitutively expressing the caspase-1 biosensor. We were able to monitor the spatiotemporal dynamics of caspase-1 activation and onset of inflammation in individual animals in the context of a systemic bacterial infection, colitis, and acute graft-versus-host disease. These data established a model whereby the development and progression of inflammatory responses can be monitored in the context of these and other mouse models of disease.


Asunto(s)
Técnicas Biosensibles/métodos , Caspasa 1/análisis , Inflamación/etiología , Animales , Apoptosis , Colitis/enzimología , Progresión de la Enfermedad , Enfermedad Injerto contra Huésped/enzimología , Células HEK293 , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Infecciones Estafilocócicas/enzimología , Células THP-1
11.
J Leukoc Biol ; 104(3): 543-555, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29775230

RESUMEN

Over 1.4 million Americans have been diagnosed with inflammatory bowel disease (IBD), and ulcerative colitis (UC) makes up approximately half of those diagnoses. As a disease, UC cycles between periods of remission and flare, which is characterized by intense abdominal pain, increased weight loss, intestinal inflammation, rectal bleeding, and dehydration. Interestingly, a widespread recommendation to IBD patients for avoidance of a flare period is "Don't Drink Alcohol" as recent work correlated alcohol consumption with increased GI symptoms in patients with IBD. Alcohol alone not only induces a systemic pro-inflammatory response, but can also be directly harmful to gut barrier integrity. However, how alcohol could result in the exacerbation of UC in both patients and murine models of colitis has yet to be elucidated. Therefore, we conducted a retrospective analysis of patients admitted for IBD with a documented history of alcohol use in conjunction with a newly developed mouse model of binge alcohol consumption following dextran sulfate sodium (DSS)-induced colitis. We found that alcohol negatively impacts clinical outcomes of patients with IBD, specifically increased intestinal infections, antibiotic injections, abdomen CT scans, and large intestine biopsies. Furthermore, in our mouse model of binge alcohol consumption following an induced colitis flare, we found alcohol exacerbates weight loss, clinical scores, colonic shortening and inflammation, and propensity to infection. These findings highlight alcohol's ability to potentiate symptoms and susceptibility to infection in UC and suggest alcohol as an underlying factor in perpetuating symptoms of IBD.


Asunto(s)
Consumo de Bebidas Alcohólicas/efectos adversos , Infecciones/epidemiología , Enfermedades Inflamatorias del Intestino/patología , Adulto , Anciano , Animales , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad
12.
J Leukoc Biol ; 102(3): 915-923, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28684599

RESUMEN

T cells play a critical role in host defense against intestinal bacteria. We have shown that ethanol combined with burn injury suppresses Peyer's patch (PP) Th17 cytokines 1 d after injury. We assessed the mechanism of suppressed Th17 effector functions. Mice were gavaged with ethanol 4 h before burn injury and euthanized 1, 3, and 7 d after injury. Mesenteric lymph nodes (MLNs), PPs, and spleen Th1 and Th17 cytokines were assessed. A significant decrease in IL-17, IL-22, IL-2, and IFN-γ were observed in all 3 lymphoid organs 1 and 3 d after injury. We used splenic cells to study the role of IL-6, IL-23, TGF-ß, and aryl hydrocarbon receptor (AHR) in suppressing Th17 cytokines. We also assessed whether the AHR agonist 6-formylindolo (3, 2-b) carbazole (FICZ) modulates Th17 cytokines. We found a significant decrease in IL-6 and TGF-ß after ethanol and burn; IL-23 was undetectable. The reconstitution of IL-23 in culture medium increased IL-17 by 2-fold and IL-22 by 20-fold in cells from burn ethanol mice. The restoration of IL-6 and TGF-ß combined did not influence the release of Th17 cytokines. We observed that AHR was necessary for IL-23 restoration of IL-22 after ethanol and burn injury. The AHR agonist FICZ enhanced IL-22, but not IL-17. None of these treatments influenced the release of Th1 cytokines. Together, these results suggest that IL-23 plays a critical role in regulation of Th17 cytokines. Furthermore, IL-6 and TGF-ß do not appear to influence IL-23-mediated restoration of Th17 cytokines after ethanol and burn injury.


Asunto(s)
Trastornos Inducidos por Alcohol , Quemaduras , Interleucina-23 , Interleucina-6/inmunología , Células Th17/inmunología , Factor de Crecimiento Transformador beta/inmunología , Trastornos Inducidos por Alcohol/tratamiento farmacológico , Trastornos Inducidos por Alcohol/inmunología , Trastornos Inducidos por Alcohol/patología , Animales , Quemaduras/tratamiento farmacológico , Quemaduras/inmunología , Quemaduras/patología , Modelos Animales de Enfermedad , Interleucina-23/inmunología , Interleucina-23/farmacología , Masculino , Ratones , Células TH1/inmunología , Células TH1/patología , Células Th17/patología
13.
Shock ; 48(6): 657-665, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28498296

RESUMEN

Intestine barrier disruption and bacterial translocation can contribute to sepsis and multiple organ failure, leading causes of mortality in burn-injured patients. In addition, findings suggest that ethanol (alcohol) intoxication at the time of injury worsens symptoms associated with burn injury. We have previously shown that interleukin-22 (IL-22) protects from intestinal leakiness and prevents overgrowth of gram-negative bacteria following ethanol and burn injury, but how IL-22 mediates these effects has not been established. Here, utilizing a mouse model of ethanol and burn injury, we show that the combined insult results in a significant loss of proliferating cells within small intestine crypts and increases Enterobacteriaceae copies, despite elevated levels of the antimicrobial peptide lipocalin-2. IL-22 administration restored numbers of proliferating cells within crypts, significantly increased Reg3ß, Reg3γ, lipocalin-2 AMP transcript levels in intestine epithelial cells, and resulted in complete reduction of Enterobacteriaceae in the small intestine. Knockout of signal transducer and activator of transcription factor-3 (STAT3) in intestine epithelial cells resulted in complete loss of IL-22 protection, demonstrating that STAT3 is required for intestine barrier protection following ethanol combined with injury. Together, these findings suggest that IL-22/STAT3 signaling is critical to gut barrier integrity and targeting this pathway may be of beneficial clinical relevance following burn injury.


Asunto(s)
Intoxicación Alcohólica , Traslocación Bacteriana/efectos de los fármacos , Quemaduras , Disbiosis , Enterobacteriaceae/inmunología , Interleucinas/inmunología , Mucosa Intestinal , Enfermedad Aguda , Intoxicación Alcohólica/complicaciones , Intoxicación Alcohólica/inmunología , Intoxicación Alcohólica/microbiología , Intoxicación Alcohólica/patología , Animales , Quemaduras/complicaciones , Quemaduras/inmunología , Quemaduras/microbiología , Quemaduras/patología , Disbiosis/etiología , Disbiosis/inmunología , Disbiosis/patología , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Ratones , Ratones Noqueados , Interleucina-22
14.
Biochim Biophys Acta Mol Basis Dis ; 1863(10 Pt B): 2645-2653, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28404517

RESUMEN

Ethanol exposure at the time of burn injury is a major contributor to post-burn pathogenesis. Many of the adverse effects associated with ethanol and burn injury are linked to an impaired intestinal barrier. The combined insult causes intestinal inflammation, resulting in tissue damage, altered tight junction expression, and increased intestinal permeability. MicroRNAs play a critical role in maintaining intestinal homeostasis including intestinal inflammation and barrier function. Specifically, miR-150 regulates inflammatory mediators which can contribute to gut barrier disruption. The present study examined whether ethanol and burn injury alter expression of microRNA processing enzymes (Drosha, Dicer, and Argonaute-2) and miR-150 in the small intestine. Male mice were gavaged with ethanol (~2.9g/kg) 4h prior to receiving a ~12.5% total body surface area full thickness burn. One or three days after injury, mice were euthanized and small intestinal epithelial cells (IECs) were isolated and analyzed for expression of microRNA biogenesis components and miR-150. Dicer mRNA and protein levels were not changed following the combined insult. Drosha and Argonaute-2 mRNA and protein levels were significantly reduced in IECs one day after injury; which accompanied reduced miR-150 expression. To further determine the role of miR-150 in intestinal inflammation, young adult mouse colonocytes were transfected with a miR-150 plasmid and stimulated with LPS (100ng/ml). miR-150 overexpression significantly reduced IL-6 and KC protein levels compared to vector control cells challenged with LPS. These results suggest that altered microRNA biogenesis and associated decrease in miR-150 likely contribute to increased intestinal inflammation following ethanol and burn injury.


Asunto(s)
Quemaduras/inmunología , Etanol/efectos adversos , Regulación de la Expresión Génica/inmunología , Mucosa Intestinal/inmunología , Intestino Delgado/inmunología , MicroARNs/inmunología , Animales , Proteínas Argonautas/inmunología , Proteínas Argonautas/metabolismo , Quemaduras/metabolismo , Quemaduras/patología , Quimiocina CXCL1/inmunología , Quimiocina CXCL1/metabolismo , ARN Helicasas DEAD-box/inmunología , ARN Helicasas DEAD-box/metabolismo , Etanol/farmacología , Mediadores de Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Interleucina-6/inmunología , Interleucina-6/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Intestino Delgado/metabolismo , Intestino Delgado/patología , Masculino , Ratones , MicroARNs/metabolismo , ARN Mensajero/inmunología , ARN Mensajero/metabolismo , Ribonucleasa III/inmunología , Ribonucleasa III/metabolismo
15.
Alcohol ; 54: 73-7, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27522326

RESUMEN

On September 27, 2015 the 20th annual Alcohol and Immunology Research Interest Group (AIRIG) meeting was held as a satellite symposium at the annual meeting of the Society for Leukocyte Biology in Raleigh, NC. The 2015 meeting focused broadly on adverse effects of alcohol and alcohol-use disorders in multiple organ systems. Divided into two plenary sessions, AIRIG opened with the topic of pulmonary inflammation as a result of alcohol consumption, which was followed by alcohol's effect on multiple organs, including the brain and liver. With presentations showing the diverse range of underlying pathology and mechanisms associated with multiple organs as a result of alcohol consumption, AIRIG emphasized the importance of continued alcohol research, as its detrimental consequences are not limited to one or even two organs, but rather extend to the entire host as a whole.


Asunto(s)
Etanol/efectos adversos , Inflamación/inducido químicamente , Animales , Humanos , Pulmón/efectos de los fármacos , Pulmón/patología
16.
J Burn Care Res ; 37(5): 283-92, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27388883

RESUMEN

Gut barrier disruption is often implicated in pathogenesis associated with burn and other traumatic injuries. In this study, the authors examined whether therapeutic intervention with mesalamine (5-aminosalicylic acid [5-ASA]), a common anti-inflammatory treatment for patients with inflammatory bowel disease, reduces intestinal inflammation and maintains normal barrier integrity after burn injury. Male C57BL/6 mice were administered an approximately 20% TBSA dorsal scald burn and resuscitated with either 1 ml normal saline or 100 mg/kg of 5-ASA dissolved in saline. The authors examined intestinal transit and permeability along with the levels of small intestine epithelial cell proinflammatory cytokines and tight junction protein expression 1 day after burn injury in the presence or absence of 5-ASA. A significant decrease in intestinal transit was observed 1 day after burn injury, which accompanied a significant increase in gut permeability. The authors found a substantial increase in the levels of interleukin (IL)-6 (by ~1.5-fold) and IL-18 (by ~2.5-fold) in the small intestine epithelial cells 1 day after injury. Furthermore, burn injury decreases the expression of the tight junction proteins claudin-4, claudin-8, and occludin. Treatment with 5-ASA after burn injury prevented the burn-induced increase in permeability, partially restored normal intestinal transit, normalized the levels of the proinflammatory cytokines IL-6 and IL-18, and restored tight junction protein expression of claudin-4 and occludin compared with that of sham levels. Together these findings suggest that 5-ASA can potentially be used as treatment to decrease intestinal inflammation and normalize intestinal function after burn injury.


Asunto(s)
Quemaduras/terapia , Mucosa Intestinal/efectos de los fármacos , Mesalamina/uso terapéutico , Animales , Claudina-4/metabolismo , Claudinas/metabolismo , Interleucinas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ocludina/metabolismo , Permeabilidad
17.
Shock ; 45(1): 73-81, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26368926

RESUMEN

Alcohol intoxication at the time of burn injury exacerbates postburn pathogenesis. Recent findings suggest gut barrier integrity is compromised after combined alcohol and burn insult, which could contribute to these complications. Tight junction proteins and mucins play critical roles in keeping the gut barrier intact. Therefore, the goal of this study was to examine the effects of alcohol and burn injury on claudin and mucin expression in the intestines. We also evaluated if the combined insult differentially influences their expression in the small and large intestines. Male C57BL/6 mice were given a single dose of 2.9 g/kg ethanol before an approximately 12.5% body area burn. One and three days after injury, we profiled expression of several tight junction proteins, mucin, and bacterial 16S rRNA genes in the small and large intestines, using qPCR. We observed >50% decrease in claudin-4 and claudin-8 genes in both ileal and colonic epithelial cells 1 day after injury. Claudin-2 was significantly upregulated, and occludin was downregulated in the small intestine 1 day after injury. Mucin-3 expression was substantially elevated (>50%) in the small intestine, whereas mucin-2 and mucin-4 were considerably diminished in the colon (>50%) 1 day after injury. Most of the parameters were normalized to sham levels on day 3, except for mucin-3 and claudin-8, which remained decreased in the large intestine. Neither alcohol nor burn alone resulted in changes in junction or mucin gene expression compared to shams. This was accompanied with increases in the family of Gram-negative bacteria, Enterobacteriaceae, in both the small and the large intestines 1 day after injury. These findings suggest that alcohol and burn injury disrupts the normal gut microbiota and alters tight junction and mucin expression in the small and large intestines.


Asunto(s)
Intoxicación Alcohólica/metabolismo , Quemaduras/metabolismo , Claudinas/biosíntesis , Intestino Grueso/metabolismo , Intestino Delgado/metabolismo , Mucinas/biosíntesis , Intoxicación Alcohólica/genética , Intoxicación Alcohólica/microbiología , Animales , Carga Bacteriana , Quemaduras/genética , Quemaduras/microbiología , Claudinas/genética , Enterobacteriaceae/aislamiento & purificación , Heces/microbiología , Regulación de la Expresión Génica , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Intestino Grueso/microbiología , Intestino Delgado/microbiología , Masculino , Ratones Endogámicos C57BL , Mucinas/genética , ARN Mensajero/genética , Uniones Estrechas/metabolismo
18.
Alcohol ; 49(8): 767-72, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26520175

RESUMEN

On November 21, 2014 the 19th annual Alcohol and Immunology Research Interest Group (AIRIG) meeting was held at Loyola University Chicago Health Sciences Campus in Maywood, Illinois. The meeting focused broadly on inflammatory cell signaling responses in the context of alcohol and alcohol-use disorders, and was divided into four plenary sessions focusing on the gut and liver, lung infections, general systemic effects of alcohol, and neuro-inflammation. One common theme among many talks was the differential roles of macrophages following both chronic and acute alcohol intoxication. Macrophages were shown to play significant roles in regulating inflammation, oxidative stress, and viral infection following alcohol exposure in the liver, lungs, adipose tissue, and brain. Other work examined the role of alcohol on disease progression in a variety of pathologies including psoriasis, advanced stage lung disease, and cancer.


Asunto(s)
Intoxicación Alcohólica/inmunología , Alcoholismo/inmunología , Macrófagos/inmunología , Tejido Adiposo/inmunología , Intoxicación Alcohólica/complicaciones , Alcoholismo/complicaciones , Animales , Asma/complicaciones , Asma/inmunología , Encéfalo/inmunología , Congresos como Asunto , Progresión de la Enfermedad , Microbioma Gastrointestinal/inmunología , Humanos , Inflamación , Hígado/inmunología , Pulmón/inmunología , Enfermedades Pulmonares/complicaciones , Enfermedades Pulmonares/inmunología , Neoplasias/complicaciones , Neoplasias/inmunología , Estrés Oxidativo/inmunología , Neumonía Viral/complicaciones , Neumonía Viral/inmunología , Psoriasis/complicaciones , Psoriasis/inmunología , Transducción de Señal , Virosis/inmunología
19.
PLoS One ; 10(7): e0129996, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26154283

RESUMEN

Sepsis remains one of the leading causes of death in burn patients who survive the initial insult of injury. Disruption of the intestinal epithelial barrier has been shown after burn injury; this can lead to the translocation of bacteria or their products (e.g., endotoxin) from the intestinal lumen to the circulation, thereby increasing the risk for sepsis in immunocompromised individuals. Since the maintenance of the epithelial barrier is largely dependent on the intestinal microbiota, we examined the diversity of the intestinal microbiome of severely burned patients and a controlled mouse model of burn injury. We show that burn injury induces a dramatic dysbiosis of the intestinal microbiome of both humans and mice and allows for similar overgrowths of Gram-negative aerobic bacteria. Furthermore, we show that the bacteria increasing in abundance have the potential to translocate to extra-intestinal sites. This study provides an insight into how the diversity of the intestinal microbiome changes after burn injury and some of the consequences these gut bacteria can have in the host.


Asunto(s)
Traslocación Bacteriana , Quemaduras/microbiología , Microbioma Gastrointestinal , Adulto , Animales , Quemaduras/patología , Enterobacteriaceae/fisiología , Femenino , Humanos , Intestino Delgado/microbiología , Ganglios Linfáticos/patología , Masculino , Ratones , Persona de Mediana Edad , Permeabilidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...