Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Alcohol Clin Exp Res (Hoboken) ; 47(1): 36-44, 2023 01.
Article in English | MEDLINE | ID: mdl-36446606

ABSTRACT

Alcohol misuse contributes to the dysregulation of immune responses and multiorgan dysfunction across various tissues, which are associated with higher risk of morbidity and mortality in people with alcohol use disorders. Organ-specific immune cells, including microglia in the brain, alveolar macrophages in the lungs, and Kupffer cells in the liver, play vital functions in host immune defense through tissue repair and maintenance of homeostasis. However, binge drinking and chronic alcohol misuse impair these immune cells' abilities to regulate inflammatory signaling and metabolism, thus contributing to multiorgan dysfunction. Further complicating these delicate systems, immune cell dysfunction associated with alcohol misuse is exacerbated by aging and gut barrier leakage. This critical review describes recent advances in elucidating the potential mechanisms by which alcohol misuse leads to derangements in host immunity and highlights current gaps in knowledge that may be the focus of future investigations.


Subject(s)
Alcoholism , Humans , Alcoholism/metabolism , Ethanol/metabolism , Liver , Macrophages, Alveolar/metabolism , Lung
2.
Front Immunol ; 13: 865492, 2022.
Article in English | MEDLINE | ID: mdl-35634337

ABSTRACT

Excessive alcohol use increases the risk of developing respiratory infections partially due to impaired alveolar macrophage (AM) phagocytic capacity. Previously, we showed that chronic ethanol (EtOH) exposure led to mitochondrial derangements and diminished oxidative phosphorylation in AM. Since oxidative phosphorylation is needed to meet the energy demands of phagocytosis, EtOH mediated decreases in oxidative phosphorylation likely contribute to impaired AM phagocytosis. Treatment with the peroxisome proliferator-activated receptor gamma (PPARγ) ligand, pioglitazone (PIO), improved EtOH-mediated decreases in oxidative phosphorylation. In other models, hypoxia-inducible factor-1 alpha (HIF-1α) has been shown to mediate the switch from oxidative phosphorylation to glycolysis; however, the role of HIF-1α in chronic EtOH mediated derangements in AM has not been explored. We hypothesize that AM undergo a metabolic shift from oxidative phosphorylation to a glycolytic phenotype in response to chronic EtOH exposure. Further, we speculate that HIF-1α is a critical mediator of this metabolic switch. To test these hypotheses, primary mouse AM (mAM) were isolated from a mouse model of chronic EtOH consumption and a mouse AM cell line (MH-S) were exposed to EtOH in vitro. Expression of HIF-1α, glucose transporters (Glut1 and 4), and components of the glycolytic pathway (Pfkfb3 and PKM2), were measured by qRT-PCR and western blot. Lactate levels (lactate assay), cell energy phenotype (extracellular flux analyzer), glycolysis stress tests (extracellular flux analyzer), and phagocytic function (fluorescent microscopy) were conducted. EtOH exposure increased expression of HIF-1α, Glut1, Glut4, Pfkfb3, and PKM2 and shifted AM to a glycolytic phenotype. Pharmacological stabilization of HIF-1α via cobalt chloride treatment in vitro mimicked EtOH-induced AM derangements (increased glycolysis and diminished phagocytic capacity). Further, PIO treatment diminished HIF-1α levels and reversed glycolytic shift following EtOH exposure. These studies support a critical role for HIF-1α in mediating the glycolytic shift in energy metabolism of AM during excessive alcohol use.


Subject(s)
Glycolysis , Macrophages, Alveolar , Animals , Ethanol/adverse effects , Glucose Transporter Type 1 , Hypoxia , Lactic Acid , Mice
3.
J Leukoc Biol ; 110(5): 979-986, 2021 11.
Article in English | MEDLINE | ID: mdl-33577717

ABSTRACT

The gastrointestinal (GI) tract is a highly dynamic structure essential for digestion, nutrient absorption, and providing an interface to prevent gut bacterial translocation. In order to maintain the barrier function, the gut utilizes many defense mechanisms including proliferation, apoptosis, and apical junctional complexes. Disruption of any of these parameters due to injury or disease could negatively impact the intestinal barrier function and homeostasis resulting in increased intestine inflammation, permeability, bacterial dysbiosis, and tissue damage. MicroRNAs are small noncoding RNA sequences that are master regulators of normal cellular homeostasis. These regulatory molecules affect cellular signaling pathways and potentially serve as candidates for providing a mechanism of impaired gut barrier integrity following GI-related pathologic conditions, ethanol exposure, or trauma such as burn injury. MicroRNAs influence cellular apoptosis, proliferation, apical junction complex expression, inflammation, and the microbiome. Due to their widespread functional affiliations, altered expression of microRNAs are associated with many pathologic conditions. This review explores the role of microRNAs in regulation of intestinal barrier integrity. The studies reviewed demonstrate that microRNAs largely impact intestine barrier function and provide insight behind the observed adverse effects following ethanol and burn injury. Furthermore, these studies suggest that microRNAs are excellent candidates for therapeutic intervention or for biomarkers to manage gut barrier integrity following trauma such as burn injury and other GI-related pathologic conditions.


Subject(s)
Intestinal Mucosa , Permeability , RNA, Messenger , Tight Junctions , Animals , Humans , Intestines
4.
J Leukoc Biol ; 109(6): 1121-1130, 2021 06.
Article in English | MEDLINE | ID: mdl-32964503

ABSTRACT

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.


Subject(s)
Burns/drug therapy , Enzyme Inhibitors/pharmacology , Ethanol/adverse effects , Intestinal Mucosa/drug effects , Mustard Compounds/pharmacology , Phenylpropionates/pharmacology , Protective Agents/pharmacology , Alcoholic Intoxication , Animals , Argonaute Proteins/genetics , Biomarkers , Burns/etiology , Burns/metabolism , Disease Susceptibility , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Male , Mice , MicroRNAs/genetics , RNA, Messenger/genetics , RNA, Ribosomal, 16S/genetics
5.
Alcohol ; 90: 27-38, 2021 02.
Article in English | MEDLINE | ID: mdl-33278514

ABSTRACT

Excessive alcohol users have increased risk of developing respiratory infections in part due to oxidative stress-induced alveolar macrophage (AM) phagocytic dysfunction. Chronic ethanol exposure increases cellular oxidative stress in AMs via upregulation of NADPH oxidase (Nox) 4, and treatment with the peroxisome proliferator-activated receptor gamma (PPARγ) ligand, rosiglitazone, decreases ethanol-induced Nox4. However, the mechanism by which ethanol induces Nox4 expression and the PPARγ ligand reverses this defect has not been elucidated. Since microRNA (miR)-92a has been predicted to target Nox4 for destabilization, we hypothesized that ethanol exposure decreases miR-92a expression and leads to Nox4 upregulation. Previous studies have implicated mitochondrial-derived oxidative stress in AM dysfunction. We further hypothesized that ethanol increases mitochondrial-derived AM oxidative stress and dysfunction via miR-92a, and that treatment with the PPARγ ligand, pioglitazone, could reverse these derangements. To test these hypotheses, a mouse AM cell line, MH-S cells, was exposed to ethanol in vitro, and primary AMs were isolated from a mouse model of chronic ethanol consumption to measure Nox4, mitochondrial target mRNA (qRT-PCR) and protein levels (confocal microscopy), mitochondria-derived reactive oxygen species (confocal immunofluorescence), mitochondrial fission (electron microscopy), and mitochondrial bioenergetics (extracellular flux analyzer). Ethanol exposure increased Nox4, enhanced mitochondria-derived oxidative stress, augmented mitochondrial fission, and impaired mitochondrial bioenergetics. Transfection with a miR-92a mimic in vitro or pioglitazone treatment in vivo diminished Nox4 levels, resulting in improvements in these ethanol-mediated derangements. These findings demonstrate that pioglitazone may provide a novel therapeutic approach to mitigate ethanol-induced AM mitochondrial derangements.


Subject(s)
Ethanol , Macrophages, Alveolar/pathology , NADPH Oxidase 4/metabolism , Animals , Cell Line , Ethanol/toxicity , Macrophages, Alveolar/metabolism , Mice , NADPH Oxidase 4/genetics , Oxidative Stress , Reactive Oxygen Species/metabolism
6.
Alcohol ; 87: 89-95, 2020 09.
Article in English | MEDLINE | ID: mdl-32353591

ABSTRACT

On November 15, 2019, the 24th annual Alcohol and Immunology Research Interest Group (AIRIG) meeting was held as a satellite conference during the annual Society for Leukocyte Biology meeting in Boston, Massachusetts. The 2019 meeting focused on alcohol, immunity, and organ damage, and included two plenary sessions. The first session highlighted new research exploring the mechanisms of alcohol-induced inflammation and liver disease, including effects on lipidomics and lipophagy, regulatory T cells, epigenetics, epithelial cells, and age-related changes in the gut. The second session covered alcohol-induced injury of other organs, encompassing diverse areas of research ranging from neurodegeneration, to lung barrier function, to colon carcinogenesis, to effects on viral infection. The discussions also highlighted current laboratory and clinical research used to identify biomarkers of alcohol use and disease.


Subject(s)
Alcohol Drinking , Alcohol Drinking/adverse effects , Alcoholism/diagnosis , Biomarkers , Boston , Congresses as Topic , Ethanol/toxicity , Humans , Inflammation
7.
Biomolecules ; 8(4)2018 12 10.
Article in English | MEDLINE | ID: mdl-30544759

ABSTRACT

Excess alcohol consumption is a global crisis contributing to over 3 million alcohol-related deaths per year worldwide and economic costs exceeding $200 billion dollars, which include productivity losses, healthcare, and other effects (e.g., property damages). Both clinical and experimental models have shown that excessive alcohol consumption results in multiple organ injury. Although alcohol metabolism occurs primarily in the liver, alcohol exposure can lead to pathophysiological conditions in multiple organs and tissues, including the brain, lungs, adipose, liver, and intestines. Understanding the mechanisms by which alcohol-mediated organ dysfunction occurs could help to identify new therapeutic approaches to mitigate the detrimental effects of alcohol misuse. Hypoxia-inducible factor (HIF)-1 is a transcription factor comprised of HIF-1α and HIF-1ß subunits that play a critical role in alcohol-mediated organ dysfunction. This review provides a comprehensive analysis of recent studies examining the relationship between HIF-1α and alcohol consumption as it relates to multiple organ injury and potential therapies to mitigate alcohol's effects.


Subject(s)
Aryl Hydrocarbon Receptor Nuclear Translocator/genetics , Ethanol/adverse effects , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Multiple Organ Failure/genetics , Alcohol Drinking/adverse effects , Alcohol Drinking/genetics , Ethanol/toxicity , Humans , Liver/drug effects , Liver/pathology , Multiple Organ Failure/chemically induced , Multiple Organ Failure/pathology
8.
J Leukoc Biol ; 104(3): 543-555, 2018 09.
Article in English | MEDLINE | ID: mdl-29775230

ABSTRACT

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.


Subject(s)
Alcohol Drinking/adverse effects , Infections/epidemiology , Inflammatory Bowel Diseases/pathology , Adult , Aged , Animals , Dextran Sulfate/toxicity , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged
9.
J Leukoc Biol ; 102(3): 915-923, 2017 09.
Article in English | MEDLINE | ID: mdl-28684599

ABSTRACT

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.


Subject(s)
Alcohol-Induced Disorders , Burns , Interleukin-23 , Interleukin-6/immunology , Th17 Cells/immunology , Transforming Growth Factor beta/immunology , Alcohol-Induced Disorders/drug therapy , Alcohol-Induced Disorders/immunology , Alcohol-Induced Disorders/pathology , Animals , Burns/drug therapy , Burns/immunology , Burns/pathology , Disease Models, Animal , Interleukin-23/immunology , Interleukin-23/pharmacology , Male , Mice , Th1 Cells/immunology , Th1 Cells/pathology , Th17 Cells/pathology
10.
Shock ; 48(6): 657-665, 2017 12.
Article in English | MEDLINE | ID: mdl-28498296

ABSTRACT

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.


Subject(s)
Alcoholic Intoxication , Bacterial Translocation/drug effects , Burns , Dysbiosis , Enterobacteriaceae/immunology , Interleukins/immunology , Intestinal Mucosa , Acute Disease , Alcoholic Intoxication/complications , Alcoholic Intoxication/immunology , Alcoholic Intoxication/microbiology , Alcoholic Intoxication/pathology , Animals , Burns/complications , Burns/immunology , Burns/microbiology , Burns/pathology , Dysbiosis/etiology , Dysbiosis/immunology , Dysbiosis/pathology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Mice , Mice, Knockout , Interleukin-22
11.
Biochim Biophys Acta Mol Basis Dis ; 1863(10 Pt B): 2645-2653, 2017 10.
Article in English | MEDLINE | ID: mdl-28404517

ABSTRACT

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.


Subject(s)
Burns/immunology , Ethanol/adverse effects , Gene Expression Regulation/immunology , Intestinal Mucosa/immunology , Intestine, Small/immunology , MicroRNAs/immunology , Animals , Argonaute Proteins/immunology , Argonaute Proteins/metabolism , Burns/metabolism , Burns/pathology , Chemokine CXCL1/immunology , Chemokine CXCL1/metabolism , DEAD-box RNA Helicases/immunology , DEAD-box RNA Helicases/metabolism , Ethanol/pharmacology , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Interleukin-6/immunology , Interleukin-6/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestine, Small/metabolism , Intestine, Small/pathology , Male , Mice , MicroRNAs/metabolism , RNA, Messenger/immunology , RNA, Messenger/metabolism , Ribonuclease III/immunology , Ribonuclease III/metabolism
12.
Alcohol ; 54: 73-7, 2016 08.
Article in English | MEDLINE | ID: mdl-27522326

ABSTRACT

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.


Subject(s)
Ethanol/adverse effects , Inflammation/chemically induced , Animals , Humans , Lung/drug effects , Lung/pathology
13.
J Burn Care Res ; 37(5): 283-92, 2016.
Article in English | MEDLINE | ID: mdl-27388883

ABSTRACT

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.


Subject(s)
Burns/therapy , Intestinal Mucosa/drug effects , Mesalamine/therapeutic use , Animals , Claudin-4/metabolism , Claudins/metabolism , Interleukins/metabolism , Male , Mice , Mice, Inbred C57BL , Occludin/metabolism , Permeability
14.
Shock ; 45(1): 73-81, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26368926

ABSTRACT

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.


Subject(s)
Alcoholic Intoxication/metabolism , Burns/metabolism , Claudins/biosynthesis , Intestine, Large/metabolism , Intestine, Small/metabolism , Mucins/biosynthesis , Alcoholic Intoxication/genetics , Alcoholic Intoxication/microbiology , Animals , Bacterial Load , Burns/genetics , Burns/microbiology , Claudins/genetics , Enterobacteriaceae/isolation & purification , Feces/microbiology , Gene Expression Regulation , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestine, Large/microbiology , Intestine, Small/microbiology , Male , Mice, Inbred C57BL , Mucins/genetics , RNA, Messenger/genetics , Tight Junctions/metabolism
15.
Alcohol Res ; 37(2): 209-22, 2015.
Article in English | MEDLINE | ID: mdl-26695746

ABSTRACT

Alcohol (ethanol) is one of the most globally abused substances, and is one of the leading causes of premature death in the world. As a result of its complexity and direct contact with ingested alcohol, the intestine represents the primary source from which alcohol-associated pathologies stem. The gut is the largest reservoir of bacteria in the body, and under healthy conditions, it maintains a barrier preventing bacteria from translocating out of the intestinal lumen. The intestinal barrier is compromised following alcohol exposure, which can lead to life-threatening systemic complications including sepsis and multiple organ failure. Furthermore, alcohol is a major confounding factor in pathology associated with trauma. Experimental data from both human and animal studies suggest that alcohol perturbs the intestinal barrier and its function, which is exacerbated by a "second hit" from traumatic injury. This article highlights the role of alcohol-mediated alterations of the intestinal epithelia and its defense against bacteria within the gut, and the impact of alcohol on intestinal immunity, specifically on T cells and neutrophils. Finally, it discusses how the gut microbiome both contributes to and protects the intestines from dysbiosis after alcohol exposure and trauma.


Subject(s)
Alcohol Drinking/immunology , Alcoholism/immunology , Burns/immunology , Dysbiosis/immunology , Gastrointestinal Microbiome/immunology , Intestinal Mucosa/immunology , Alcoholism/complications , Alcoholism/microbiology , Bacterial Translocation/immunology , Burns/complications , Dysbiosis/complications , Humans , Neutrophils/immunology , Sepsis/complications , Sepsis/immunology , T-Lymphocytes/immunology
16.
Alcohol ; 49(8): 767-72, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26520175

ABSTRACT

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.


Subject(s)
Alcoholic Intoxication/immunology , Alcoholism/immunology , Macrophages/immunology , Adipose Tissue/immunology , Alcoholic Intoxication/complications , Alcoholism/complications , Animals , Asthma/complications , Asthma/immunology , Brain/immunology , Congresses as Topic , Disease Progression , Gastrointestinal Microbiome/immunology , Humans , Inflammation , Liver/immunology , Lung/immunology , Lung Diseases/complications , Lung Diseases/immunology , Neoplasms/complications , Neoplasms/immunology , Oxidative Stress/immunology , Pneumonia, Viral/complications , Pneumonia, Viral/immunology , Psoriasis/complications , Psoriasis/immunology , Signal Transduction , Virus Diseases/immunology
17.
PLoS One ; 10(7): e0129996, 2015.
Article in English | MEDLINE | ID: mdl-26154283

ABSTRACT

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.


Subject(s)
Bacterial Translocation , Burns/microbiology , Gastrointestinal Microbiome , Adult , Animals , Burns/pathology , Enterobacteriaceae/physiology , Female , Humans , Intestine, Small/microbiology , Lymph Nodes/pathology , Male , Mice , Middle Aged , Permeability
18.
Alcohol ; 49(5): 507-11, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25921594

ABSTRACT

The intestine is segmented into functionally discrete compartments (duodenum, jejunum, ileum, and colon). The present study examined whether alcohol combined with burn injury differently influences cytokine levels in different parts of the intestine. Male mice were gavaged with alcohol (∼2.9 g/kg) 4 h prior to receiving a ∼12.5% total body surface area full thickness burn. Mice were sacrificed 1, 3, and 7 days after injury. The intestine segments (duodenum, jejunum, ileum, and colon) were harvested, homogenized, and analyzed for inflammatory mediators (IL-6, IL-18, and KC) using their respective ELISAs. KC levels were significantly increased in the jejunum, ileum, and colon following alcohol and burn injury as compared to shams. The increase in KC was ∼28-fold higher in the colon as compared to the levels observed in duodenum following alcohol and burn injury. Both IL-6 and IL-18 levels were significantly elevated in both the ileum and colon following the combined insult. There was a ∼7-fold increase in IL-6 levels in the colon as compared with the duodenum after the combined insult. Levels of IL-18 were increased by ∼1.5-fold in the colon as compared to the ileum following alcohol and burn injury. The data suggest that pro-inflammatory mediators are differentially expressed in the intestine following alcohol and burn injury.


Subject(s)
Alcoholic Intoxication/metabolism , Burns/metabolism , Inflammation Mediators/metabolism , Interleukin-18/metabolism , Intestinal Mucosa/metabolism , Alcoholic Intoxication/complications , Animals , Burns/complications , Chemokines/metabolism , Interleukin-6/metabolism , Male , Mice
19.
Alcohol ; 49(1): 1-6, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25468277

ABSTRACT

Loyola University Chicago, Health Sciences Campus in Maywood, Illinois hosted the 18th annual Alcohol and Immunology Research Interest Group (AIRIG) meeting on November 22, 2013. This year's meeting emphasized alcohol's effect on inflammatory responses in diverse disease states and injury conditions. The meeting consisted of three plenary sessions demonstrating the adverse effects of alcohol, specifically, liver inflammation, adverse systemic effects, and alcohol's role in infection and immunology. Researchers also presented insight on modulation of microRNAs and stress proteins following alcohol consumption. Additionally, researchers revealed sex- and concentration-dependent differences in alcohol-mediated pathologies.


Subject(s)
Alcohol Drinking/metabolism , Alcoholism/metabolism , Congresses as Topic , Inflammation Mediators/metabolism , Public Opinion , Alcohol Drinking/immunology , Alcoholism/diagnosis , Alcoholism/immunology , Animals , Humans , Inflammation/diagnosis , Inflammation/immunology , Inflammation/metabolism , Inflammation Mediators/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...