1
|
Tyler R, Vizioli C, Barb J, Farokhnia M, Leggio L. Circulating Immune and Endocrine Markers in Currently Drinking and Abstinent Individuals With Alcohol Use Disorder and Controls. Addict Biol 2025; 30:e70039. [PMID: 40317574 PMCID: PMC12046569 DOI: 10.1111/adb.70039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 02/20/2025] [Accepted: 04/10/2025] [Indexed: 05/07/2025]
Abstract
Alcohol use disorder (AUD) is associated with changes in endocrine and immune system function. This study is a secondary analysis aimed at investigating changes in circulating immune and endocrine biomarkers in blood samples from three groups: (1) healthy controls (HC, N = 12), (2) AUD-currently drinking, nontreatment seeking (CD, N = 9), and (3) AUD-abstinent, treatment-seeking (AB, N = 10; abstinent for at least 6 weeks). We hypothesized that both immune and endocrine biomarker concentrations would be different in AUD groups compared to healthy controls. Immune biomarkers included IL-8, IL-18, CCL2, TNF-α, IL-1RA, IL-6, and IL-10. Endocrine biomarkers included brain-derived neurotrophic factor (BDNF), glucagon-like peptide 1 (GLP-1), ghrelin, gastric inhibitory peptide (GIP), growth hormone, leptin, and insulin. Biomarker concentrations were compared between the three groups while controlling for age and sex, and associations between biomarker concentrations and behavioral measures were explored. IL-8 concentrations were elevated in AB compared to CD and HC (F(2,29) = 6.33, p = 0.006, ƞp 2 = 0.318). BDNF concentrations were lower in AB compared to HC (F(2,30) = 4.34, p = 0.02, ƞp 2 = 0.266). GLP-1 concentrations were higher in AB compared to HC (F(2,25) = 4.22, p = 0.03, ƞp 2 = 0.287). Exploratory analyses in combined groups showed that measures of past drinking, AUD severity, and anxiety/depression positively correlated with IL-18 and TNF-α and negatively correlated with BDNF. These results demonstrate that circulating concentrations of both immune and endocrine proteins are altered in abstinent individuals with a history of severe AUD (AB group) compared to healthy controls. In contrast, no group differences were observed for any biomarker between the nontreatment seeking, currently drinking people with AUD and the HC group. Our findings highlight the importance of accounting for AUD severity, comorbidities, and treatment-seeking status, especially when studying alcohol-related biomarkers.
Collapse
Affiliation(s)
- Ryan E. Tyler
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology (CPN) Section, Translational Addiction Medicine BranchNational Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of HealthBaltimoreMarylandUSA
- National Institute of General Medical SciencesBethesdaMarylandUSA
| | - Carlotta Vizioli
- Interoceptive Disorders Unit, Office of the Clinical DirectorNational Institute of Neurological Disorders and Stroke, NIHBethesdaMarylandUSA
| | - Jennifer J. Barb
- Translational Biobehavioral and Health Disparities Branch, Clinical Center, NIHBethesdaMarylandUSA
| | - Mehdi Farokhnia
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology (CPN) Section, Translational Addiction Medicine BranchNational Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of HealthBaltimoreMarylandUSA
| | - Lorenzo Leggio
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology (CPN) Section, Translational Addiction Medicine BranchNational Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of HealthBaltimoreMarylandUSA
| |
Collapse
|
2
|
Barreto Garcia V, Gasparotto LHS, de Araujo AA, Leitão RFC, Brito GAC, Vilar NF, Lima Oliveira E, Guedes PMM, de Araújo Júnior RF. Gold Nanoparticles (AuNPs) Coadministered with a β-Blocker Prevent Liver Fibrosis Caused by Ethanol and Methamphetamine in Rats by Downregulating the Expression of M2 Macrophages. ACS OMEGA 2025; 10:14924-14939. [PMID: 40290979 PMCID: PMC12019731 DOI: 10.1021/acsomega.4c10118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 03/21/2025] [Accepted: 04/02/2025] [Indexed: 04/30/2025]
Abstract
Simultaneous abuse of ethanol and methamphetamine (METH) causes severe liver damage through oxidative stress and inflammation. This study evaluated the antifibrotic effects of gold nanoparticles (AuNPs) coadministered with the β-blocker carvedilol (CARV) against liver damage in rats. Male Wistar rats received 30% ethanol (7 g/kg) daily for 28 days, with METH (10 mg/kg) administered on the 22nd and 28th days. Liver damage was assessed using serum hepatic enzymes, glutathione (GSH) levels, malondialdehyde (MDA) formation, myeloperoxidase (MPO) inhibition, and histopathological analysis, including H&E, Picrosirius Red staining, immunofluorescence, and transmission electron microscopy. Cytokine levels were measured in liver tissue samples. In vitro, RAW 264.7 macrophages were induced to polarize into M1 and M2 phenotypes and cocultured with AuNPs + CARV-treated 3T3 cells, analyzed by rtPCR. AuNPs + CARV effectively protected the liver by modulating interactions between hepatic stellate cells (HSCs) and Kupffer cells, promoting an antifibrotic immune response driven by M1 macrophages. This was indicated by downregulation of profibrotic M2 macrophages and upregulation of M1 macrophages, shown by an increased CD86/CD163 ratio and reduced levels of IL-1β, TNF-α, TGFβ, AKT, and PI3K., pointing an attenuated liver inflammation. These results suggest that AuNPs combined with CARV could potentially serve as a therapy for alcohol and METH-induced liver fibrosis by targeting M2 macrophages.
Collapse
Affiliation(s)
- Vinícius Barreto Garcia
- Inflammation
and Cancer Research Laboratory, Department of Morphology, Federal University of Rio Grande do Norte (UFRN), Natal 59078-970, RN, Brazil
| | - Luiz H. S. Gasparotto
- Institute
of Chemistry, Federal University of Mato
Grosso (UFMT), Cuiaba 78060-900, MT, Brazil
| | - Aurigena A. de Araujo
- Department
of Pharmacology, Federal University of Rio
Grande do Norte (UFRN), Natal 59078-970, RN, Brazil
| | - Renata F. C. Leitão
- Department
of Morphology, Postgraduate Program in Morphology, Federal University of Ceará (UFC), Fortaleza 60355-636, CE, Brazil
| | - Gerly A. C. Brito
- Department
of Morphology, Postgraduate Program in Morphology, Federal University of Ceará (UFC), Fortaleza 60355-636, CE, Brazil
| | - Natalia Feitosa Vilar
- Inflammation
and Cancer Research Laboratory, Department of Morphology, Federal University of Rio Grande do Norte (UFRN), Natal 59078-970, RN, Brazil
| | - Emily Lima Oliveira
- Inflammation
and Cancer Research Laboratory, Department of Morphology, Federal University of Rio Grande do Norte (UFRN), Natal 59078-970, RN, Brazil
| | - Paulo M. M. Guedes
- Department
of Microbiology and Parasitology, Federal
University of Rio Grande do Norte (UFRN), Natal 59078-970, RN, Brazil
| | - Raimundo F. de Araújo Júnior
- Inflammation
and Cancer Research Laboratory, Department of Morphology, Federal University of Rio Grande do Norte (UFRN), Natal 59078-970, RN, Brazil
| |
Collapse
|
3
|
Sturm R, Haag F, Bergmann CB, Marzi I, Relja B. Alcohol drinking leads to sex-dependent differentiation of T cells. Eur J Trauma Emerg Surg 2025; 51:87. [PMID: 39870931 PMCID: PMC11772406 DOI: 10.1007/s00068-024-02732-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 09/28/2024] [Indexed: 01/29/2025]
Abstract
OBJECTIVE Global per capita alcohol consumption is increasing, posing significant socioeconomic and medical challenges also due to alcohol-related traumatic injuries but also its biological effects. Trauma as a leading cause of death in young adults, is often associated with an increased risk of complications, such as sepsis and multiple organ failure, due to immunological imbalances. Regulatory T cells play a crucial role in maintaining immune homeostasis by regulating the inflammatory response. Since it is crucial to understand the effects of alcohol in healthy volunteers, in order to refer findings from trauma cohorts, this study investigates the time- and dose-dependent modulation of CD4+ lymphocytes and their subsets following acute alcohol consumption, considering both general and sex-specific variations. METHODS Twelve female and ten male healthy volunteers consumed twelve alcohol mixed drinks over four hours to achieve a blood alcohol level of 1.0‰. Blood samples were collected before and at various time points (2, 4, 6, 24 and 48 h) post-consumption for flow cytometric analyses of the phenotype and activation makers CD4/CD25/CD127 of CD4+ T cells and their subtypes. RESULTS CD4+ lymphocytes significantly decreased at 4 h and increased at 6 h post-alcohol consumption. Naïve CD25-CD127+ T cells significantly decreased from 2 to 24 h in women and 2 to 48 h in men, while CD25+CD127+ effector T cells significantly increased during the same period. Natural CD25+CD127- regulatory T cells increased significantly at 4 and 6 h, with a higher increase in men. Induced regulatory T cells (CD4+CD25highCD127-) significantly increased at 2 h for all volunteers, with lower proportions of natural and induced regulatory T cells in women. CONCLUSIONS Acute alcohol consumption induces immune modulation persisting for days, impacting T cell subsets differently in men and women. The prolonged modulation in men may contribute to slightly poorer clinical outcomes, emphasizing the need to consider these effects in trauma patients with acute alcohol intoxication.
Collapse
Affiliation(s)
- Ramona Sturm
- Department of Trauma Surgery and Orthopedics, Goethe University, University Hospital, Frankfurt, Germany
| | - Florian Haag
- Clinic for Radiology and Nuclear Medicine, Heidelberg University, University Medical Centre Mannheim, Mannheim, Germany
| | - Christian B Bergmann
- Translational and Experimental Trauma Research, Department of Trauma, Hand, Plastic and Reconstructive Surgery, University Ulm, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| | - Ingo Marzi
- Department of Trauma Surgery and Orthopedics, Goethe University, University Hospital, Frankfurt, Germany
| | - Borna Relja
- Department of Trauma Surgery and Orthopedics, Goethe University, University Hospital, Frankfurt, Germany.
- Translational and Experimental Trauma Research, Department of Trauma, Hand, Plastic and Reconstructive Surgery, University Ulm, Albert-Einstein-Allee 23, 89081, Ulm, Germany.
| |
Collapse
|
4
|
Szentirmai É, Buckley K, Massie AR, Kapás L. Lipopolysaccharide-mediated effects of the microbiota on sleep and body temperature. Sci Rep 2024; 14:27378. [PMID: 39521828 PMCID: PMC11550806 DOI: 10.1038/s41598-024-78431-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Recent research suggests that microbial molecules translocated from the intestinal lumen into the host's internal environment may play a role in various physiological functions, including sleep. Previously, we identified that butyrate, a short-chain fatty acid produced by intestinal bacteria, and lipoteichoic acid, a cell wall component of gram-positive bacteria, induce sleep when their naturally occurring translocation is mimicked by direct delivery into the portal vein. Building upon these findings, we aimed to explore the sleep signaling potential of intraportally administered lipopolysaccharide (LPS), a primary component of gram-negative bacterial cell walls, in rats. Low dose of LPS (1 μg/kg) increased sleep duration and prolonged fever, without affecting systemic LPS levels. Interestingly, administering LPS systemically outside the portal region at a dose 20 times higher did not affect sleep, indicating a localized sensitivity within the hepatoportal region for the sleep and febrile effects of LPS. Furthermore, both the sleep- and fever-inducing effects of LPS were inhibited by indomethacin, a prostaglandin synthesis inhibitor, and replicated by intraportal administration of prostaglandin E2 or arachidonic acid, suggesting the involvement of the prostaglandin system in mediating these actions.
Collapse
Affiliation(s)
- Éva Szentirmai
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, 412 E Spokane Falls Blvd, Spokane, WA, 99210, USA.
- Sleep and Performance Research Center, Washington State University, Spokane, Washington, USA.
| | - Katelin Buckley
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, 412 E Spokane Falls Blvd, Spokane, WA, 99210, USA
| | - Ashley R Massie
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, 412 E Spokane Falls Blvd, Spokane, WA, 99210, USA
| | - Levente Kapás
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, 412 E Spokane Falls Blvd, Spokane, WA, 99210, USA
- Sleep and Performance Research Center, Washington State University, Spokane, Washington, USA
| |
Collapse
|
5
|
Melamud MM, Bobrik DV, Brit PI, Efremov IS, Buneva VN, Nevinsky GA, Akhmetova EA, Asadullin AR, Ermakov EA. Biochemical, Hematological, Inflammatory, and Gut Permeability Biomarkers in Patients with Alcohol Withdrawal Syndrome with and without Delirium Tremens. J Clin Med 2024; 13:2776. [PMID: 38792318 PMCID: PMC11121978 DOI: 10.3390/jcm13102776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 05/04/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024] Open
Abstract
Background: Delirium Tremens (DT) is known to be a serious complication of alcohol withdrawal syndrome (AWS). Neurotransmitter abnormalities, inflammation, and increased permeability are associated with the pathogenesis of AWS and DT. However, the biomarkers of these conditions are still poorly understood. Methods: In this work, biochemical, hematologic, inflammatory, and gut permeability biomarkers were investigated in the following three groups: healthy controls (n = 75), severe AWS patients with DT (n = 28), and mild/moderate AWS without DT (n = 97). Blood sampling was performed after resolution of the acute condition (on 5 ± 1 day after admission) to collect clinical information from patients and to investigate associations with clinical scales. Biomarker analysis was performed using automated analyzers and ELISA. Inflammatory biomarkers included the erythrocyte sedimentation rate (ESR), high-sensitivity C-reactive protein (hsCRP), and platelet-to-lymphocyte ratio (PLR). Results: Among the biochemical biomarkers, only glucose, total cholesterol, and alanine aminotransferase (ALT) changed significantly in the analyzed groups. A multiple regression analysis showed that age and ALT were independent predictors of the CIWA-Ar score. Hematologic biomarker analysis showed an increased white blood cell count, and the elevated size and greater size variability of red blood cells and platelets (MCV, RDWc, and PDWc) in two groups of patients. Gut permeability biomarkers (FABP2, LBP, and zonulin) did not change, but were associated with comorbid pathologies (alcohol liver disease and pancreatitis). The increase in inflammatory biomarkers (ESR and PLR) was more evident in AWS patients with DT. Cluster analysis confirmed the existence of a subgroup of patients with evidence of high inflammation, and such a subgroup was more frequent in DT patients. Conclusions: These findings contribute to the understanding of biomarker variability in AWS patients with and without DT and support the heterogeneity of patients by the level of inflammation.
Collapse
Affiliation(s)
- Mark M. Melamud
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (M.M.M.); (V.N.B.); (G.A.N.)
| | - Daria V. Bobrik
- Department of Psychiatry and Addiction, Bashkir State Medical University, 450008 Ufa, Russia; (D.V.B.); (E.A.A.); (A.R.A.)
| | - Polina I. Brit
- Department of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Ilia S. Efremov
- Institute of Personalized Psychiatry and Neurology, Shared Core Facilities, V.M. Bekhterev National Medical Research Centre for Psychiatry and Neurology, 192019 Saint Petersburg, Russia;
| | - Valentina N. Buneva
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (M.M.M.); (V.N.B.); (G.A.N.)
- Department of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Georgy A. Nevinsky
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (M.M.M.); (V.N.B.); (G.A.N.)
- Department of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Elvina A. Akhmetova
- Department of Psychiatry and Addiction, Bashkir State Medical University, 450008 Ufa, Russia; (D.V.B.); (E.A.A.); (A.R.A.)
- Institute of Personalized Psychiatry and Neurology, Shared Core Facilities, V.M. Bekhterev National Medical Research Centre for Psychiatry and Neurology, 192019 Saint Petersburg, Russia;
| | - Azat R. Asadullin
- Department of Psychiatry and Addiction, Bashkir State Medical University, 450008 Ufa, Russia; (D.V.B.); (E.A.A.); (A.R.A.)
- Institute of Personalized Psychiatry and Neurology, Shared Core Facilities, V.M. Bekhterev National Medical Research Centre for Psychiatry and Neurology, 192019 Saint Petersburg, Russia;
| | - Evgeny A. Ermakov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (M.M.M.); (V.N.B.); (G.A.N.)
- Department of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| |
Collapse
|
6
|
Szentirmai E, Buckley K, Massie AR, Kapas L. Lipopolysaccharide-Mediated Effects of the Microbiota on Sleep and Body Temperature. RESEARCH SQUARE 2024:rs.3.rs-3995260. [PMID: 38496422 PMCID: PMC10942547 DOI: 10.21203/rs.3.rs-3995260/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Background Recent research suggests that microbial molecules translocated from the intestinal lumen into the host's internal environment may play a role in various physiological functions, including sleep. Previously, we identified that butyrate, a short-chain fatty acid, produced by intestinal bacteria, and lipoteichoic acid, a cell wall component of gram-positive bacteria induce sleep when their naturally occurring translocation is mimicked by direct delivery into the portal vein. Building upon these findings, we aimed to explore the sleep signaling potential of intraportally administered lipopolysaccharide, a primary component of gram-negative bacterial cell walls, in rats. Results Low dose of lipopolysaccharide (1 μg/kg) increased sleep duration and prolonged fever, without affecting systemic lipopolysaccharide levels. Interestingly, administering LPS systemically outside the portal region at a dose 20 times higher did not affect sleep, indicating a localized sensitivity within the hepatoportal region, encompassing the portal vein and liver, for the sleep and febrile effects of lipopolysaccharide. Furthermore, both the sleep- and fever-inducing effects of LPS were inhibited by indomethacin, a prostaglandin synthesis inhibitor, and replicated by intraportal administration of prostaglandin E2 or arachidonic acid, suggesting the involvement of the prostaglandin system in mediating these actions. Conclusions These findings underscore the dynamic influence of lipopolysaccharide in the hepatoportal region on sleep and fever mechanisms, contributing to a complex microbial molecular assembly that orchestrates communication between the intestinal microbiota and brain. Lipopolysaccharide is a physiological component of plasma in both the portal and extra-portal circulation, with its levels rising in response to everyday challenges like high-fat meals, moderate alcohol intake, sleep loss and psychological stress. The increased translocation of lipopolysaccharide under such conditions may account for their physiological impact in daily life, highlighting the intricate interplay between microbial molecules and host physiology.
Collapse
|
7
|
Vollrath JT, Schindler CR, Herrmann E, Verboket RD, Henrich D, Marzi I, Störmann P. EVALUATION OF CYFRA 21-1, ANGIOPOETIN-2, PENTRAXIN-3, SRAGE, IL-6, AND IL-10 IN POLYTRAUMATIZED PATIENTS WITH CONCOMITANT THORACIC TRAUMA-HELPFUL MARKERS TO PREDICT PNEUMONIA? Shock 2023; 60:392-399. [PMID: 37548620 DOI: 10.1097/shk.0000000000002186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
ABSTRACT Background: Pneumonia is a frequent complication after polytrauma. This study aims to evaluate the ability of different serum markers to identify patients at risk of developing pneumonia after polytrauma. Methods: A retrospective analysis of prospectively collected data in polytraumatized patients with concomitant thoracic trauma (Injury Severity Score ≥16, Abbreviated Injury Scale Thorax ≥ 3) was performed. The study cohort was divided into patients with and without pneumonia during the clinical course. Serum levels of lung epithelial (CYFRA 21-1), endothelial (Ang-2), and inflammatory (PTX-3, sRAGE, IL-6, IL-10) markers were measured upon arrival in the trauma room and on days 2 and 5. Results: A total of 73 patients and 16 healthy controls were included in this study. Of these, 20 patients (27.4%) developed pneumonia. Polytraumatized patients showed significantly increased CYFRA 21-1 levels with a distinct peak after admission compared with healthy controls. Serum PTX-3 significantly increased on day 2 in polytraumatized patients compared with healthy controls. Injury Severity Score and demographic parameters were comparable between both groups (pneumonia vs. no pneumonia). No statistically significant difference could be observed for serum levels of CYFRA 21-1, Ang-2, PTX-3, sRAGE, IL-6, and IL-10 between the groups (pneumonia vs. no pneumonia) on all days. Logistic regression revealed a combination of IL-6, IL-10, sRAGE, and PTX-3 to be eventually helpful to identify patients at risk of developing pneumonia and our newly developed score was significantly higher on day 0 in patients developing pneumonia ( P < 0.05). Conclusion: The investigated serum markers alone are not helpful to identify polytraumatized patients at risk of developing pneumonia, while a combination of IL-6, IL-10, PTX-3, and sRAGE might be.
Collapse
Affiliation(s)
- Jan Tilmann Vollrath
- Department of Trauma, Hand and Reconstructive Surgery, Goethe University, Frankfurt, Germany
| | - Cora Rebecca Schindler
- Department of Trauma, Hand and Reconstructive Surgery, Goethe University, Frankfurt, Germany
| | - Eva Herrmann
- Institute for Biostatistics and Mathematical Modeling, Goethe University, Frankfurt, Germany
| | - René D Verboket
- Department of Trauma, Hand and Reconstructive Surgery, Goethe University, Frankfurt, Germany
| | - Dirk Henrich
- Department of Trauma, Hand and Reconstructive Surgery, Goethe University, Frankfurt, Germany
| | - Ingo Marzi
- Department of Trauma, Hand and Reconstructive Surgery, Goethe University, Frankfurt, Germany
| | - Philipp Störmann
- Department of Trauma, Hand and Reconstructive Surgery, Goethe University, Frankfurt, Germany
| |
Collapse
|
8
|
Maegele M, Aletti F, Efron PA, Relja B, Orfanos SE. NEW INSIGHTS INTO THE PATHOPHYSIOLOGY OF TRAUMA AND HEMORRHAGE. Shock 2023; 59:6-9. [PMID: 36867756 DOI: 10.1097/shk.0000000000001954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2023]
Abstract
ABSTRACT Circulatory shock from trauma and hemorrhage remains a clinical challenge with mortality still high within the first hours after impact. It represents a complex disease involving the impairment of a number of physiological systems and organs and the interaction of different pathological mechanisms. Multiple external and patient-specific factors may further modulate and complicate the clinical course. Recently, novel targets and models with complex multiscale interaction of data from different sources have been identified which offer new windows of opportunity. Future works needs to consider patient-specific conditions and outcomes to mount shock research onto the next higher level of precision and personalized medicine.
Collapse
Affiliation(s)
- Marc Maegele
- Department of Traumatology and Orthopedic Surgery, Cologne-Merheim Medical Center, Institute for Research in Operative Medicine, University Witten-Herdecke, Cologne, Germany
| | - Federico Aletti
- Instituto de Ciência e Tecnologia, Universidade Federal de São Paulo, São José dos Campos, Brazil
| | - Philip A Efron
- Department of Surgery, Division of Acute Care Surgery and Surgical Critical Care, Laboratory of Inflammation Biology and Surgical Science, UF Health Critical Care Organization, Florida
| | - Borna Relja
- Department of Radiology and Nuclear Medicine, Experimental Radiology, Otto-von-Guericke University, Magdeburg, Germany
| | - Stylianos E Orfanos
- 1st Department of Critical Care Medicine, National and Kapodistrian University of Athens, Medical School, Greece
| |
Collapse
|
9
|
Vollrath JT, Klingebiel F, Bläsius F, Greven J, Bolierakis E, Nowak AJ, Simic M, Hildebrand F, Marzi I, Relja B. I-FABP as a Potential Marker for Intestinal Barrier Loss in Porcine Polytrauma. J Clin Med 2022; 11:jcm11154599. [PMID: 35956214 PMCID: PMC9369469 DOI: 10.3390/jcm11154599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 07/28/2022] [Accepted: 08/02/2022] [Indexed: 12/01/2022] Open
Abstract
Polytrauma and concomitant hemorrhagic shock can lead to intestinal damage and subsequent multiple organ dysfunction syndrome. The intestinal fatty acid-binding protein (I-FABP) is expressed in the intestine and appears quickly in the circulation after intestinal epithelial cell damage. This porcine animal study investigates the I-FABP dynamics in plasma and urine after polytrauma. Furthermore, it evaluates to what extent I-FABP can also act as a marker of intestinal damage in a porcine polytrauma model. Eight pigs (Sus scrofa) were subjected to polytrauma which consisted of lung contusion, tibial fracture, liver laceration, and hemorrhagic shock followed by blood and fluid resuscitation and fracture fixation with an external fixator. Eight sham animals were identically instrumented but not injured. Afterwards, intensive care treatment including mechanical ventilation for 72 h followed. I-FABP levels in blood and urine were determined by ELISA. In addition, immunohistological staining for I-FABP, active caspase-3 and myeloperoxidase were performed after 72 h. Plasma and urine I-FABP levels were significantly increased shortly after trauma. I-FABP expression in intestinal tissue showed significantly lower expression in polytraumatized animals vs. sham. Caspase-3 and myeloperoxidase expression in the immunohistological examination were significantly higher in the jejunum and ileum of polytraumatized animals compared to sham animals. This study confirms a loss of intestinal barrier after polytrauma which is indicated by increased I-FABP levels in plasma and urine as well as decreased I-FABP levels in immunohistological staining of the intestine.
Collapse
Affiliation(s)
- Jan Tilmann Vollrath
- Department of Trauma, Hand and Reconstructive Surgery, Goethe University, 60596 Frankfurt, Germany
| | - Felix Klingebiel
- Department of Trauma, Hand and Reconstructive Surgery, Goethe University, 60596 Frankfurt, Germany
- Department of Trauma, University of Zurich, Universitätsspital Zurich, 8091 Zurich, Switzerland
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto von Guericke University, 39120 Magdeburg, Germany
| | - Felix Bläsius
- Department of Trauma and Reconstructive Surgery, RWTH Aachen University, 52074 Aachen, Germany
| | - Johannes Greven
- Department of Trauma and Reconstructive Surgery, RWTH Aachen University, 52074 Aachen, Germany
| | - Eftychios Bolierakis
- Department of Trauma and Reconstructive Surgery, RWTH Aachen University, 52074 Aachen, Germany
| | - Aleksander J. Nowak
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto von Guericke University, 39120 Magdeburg, Germany
| | - Marija Simic
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto von Guericke University, 39120 Magdeburg, Germany
| | - Frank Hildebrand
- Department of Trauma and Reconstructive Surgery, RWTH Aachen University, 52074 Aachen, Germany
| | - Ingo Marzi
- Department of Trauma, Hand and Reconstructive Surgery, Goethe University, 60596 Frankfurt, Germany
| | - Borna Relja
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto von Guericke University, 39120 Magdeburg, Germany
- Correspondence:
| |
Collapse
|
10
|
Zafari N, Velayati M, Fahim M, Maftouh M, Pourali G, Khazaei M, Nassiri M, Hassanian SM, Ghayour-Mobarhan M, Ferns GA, Kiani MA, Avan A. Role of gut bacterial and non-bacterial microbiota in alcohol-associated liver disease: Molecular mechanisms, biomarkers, and therapeutic prospective. Life Sci 2022; 305:120760. [PMID: 35787997 DOI: 10.1016/j.lfs.2022.120760] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/20/2022] [Accepted: 06/28/2022] [Indexed: 12/17/2022]
Abstract
Alcohol-associated liver disease (ALD) comprises a spectrum of liver diseases that include: steatosis to alcohol-associated hepatitis, cirrhosis, and ultimately hepatocellular carcinoma. The pathophysiology and potential underlying mechanisms for alcohol-associated liver disease are unclear. Moreover, the treatment of ALD remains a challenge. Intestinal microbiota include bacteria, fungi, and viruses, that are now known to be important in the development of ALD. Alcohol consumption can change the gut microbiota and function leading to liver disease. Given the importance of interactions between intestinal microbiota, alcohol, and liver injury, the gut microbiota has emerged as a potential biomarker and therapeutic target. This review focuses on the potential mechanisms by which the gut microbiota may be involved in the pathogenesis of ALD and explains how this can be translated into clinical management. We discuss the potential of utilizing the gut microbiota signature as a biomarker in ALD patients. Additionally, we present an overview of the prospect of modulating the intestinal microbiota for the management of ALD.
Collapse
Affiliation(s)
- Nima Zafari
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahla Velayati
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mostafa Fahim
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mina Maftouh
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ghazaleh Pourali
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Khazaei
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammadreza Nassiri
- Recombinant Proteins Research Group, The Research Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Ghayour-Mobarhan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex BN1 9PH, UK
| | - Mohammad Ali Kiani
- Department of Pediatrics, Akbar Hospital, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pediatric Gastroenterology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
11
|
Noack L, Bundkirchen K, Xu B, Gylstorff S, Zhou Y, Köhler K, Jantaree P, Neunaber C, Nowak AJ, Relja B. Acute Intoxication With Alcohol Reduces Trauma-Induced Proinflammatory Response and Barrier Breakdown in the Lung via the Wnt/β-Catenin Signaling Pathway. Front Immunol 2022; 13:866925. [PMID: 35663960 PMCID: PMC9159919 DOI: 10.3389/fimmu.2022.866925] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 04/07/2022] [Indexed: 11/13/2022] Open
Abstract
Background Trauma is the third leading cause of mortality worldwide. Upon admission, up to 50% of traumatized patients are acutely intoxicated with alcohol, which might lead to aberrant immune responses. An excessive and uncontrolled inflammatory response to injury is associated with damage to trauma-distant organs. We hypothesize that, along with inflammation-induced apoptosis, the activation of the Wnt/β-catenin signaling pathway would cause breakdown of the lung barrier and the development of lung injury after trauma. It remains unclear whether ethanol intoxication (EI) prior to trauma and hemorrhagic shock will attenuate inflammation and organ injury. Methods In this study, 14 male C57BL/6J mice were randomly assigned to two groups and exposed either to EtOH or to NaCl as a control by an oral gavage before receiving a femur fracture (Fx) and hemorrhagic shock, followed by resuscitation (THFx). Fourteen sham animals received either EtOH or NaCl and underwent surgical procedures without THFx induction. After 24 h, oil red O staining of fatty vacuoles in the liver was performed. Histological lung injury score (LIS) was assessed to analyze the trauma-induced RLI. Gene expression of Cxcl1, Il-1β, Muc5ac, Tnf, and Tnfrsf10b as well as CXCL1, IL-1β, and TNF protein levels in the lung tissue and bronchoalveolar lavage fluid were determined by RT-qPCR, ELISA, and immunohistological analyses. Infiltrating polymorphonuclear leukocytes (PMNLs) were examined via immunostaining. Apoptosis was detected by activated caspase-3 expression in the lung tissue. To confirm active Wnt signaling after trauma, gene expression of Wnt3a and its inhibitor sclerostin (Sost) was determined. Protein expression of A20 and RIPK4 as possible modulators of the Wnt signaling pathway was analyzed via immunofluorescence. Results Significant fatty changes in the liver confirmed the acute EI. Histopathology and decreased Muc5ac expression revealed an increased lung barrier breakdown and concomitant lung injury after THFx versus sham. EI prior trauma decreased lung injury. THFx increased not only the gene expression of pro-inflammatory markers but also the pulmonary infiltration with PMNL and apoptosis versus sham, while EI prior to THFx reduced those changes significantly. EI increased the THFx-reduced gene expression of Sost and reduced the THFx-induced expression of Wnt3a. While A20, RIPK4, and membranous β-catenin were significantly reduced after trauma, they were enhanced upon EI. Conclusion These findings suggest that acute EI alleviates the uncontrolled inflammatory response and lung barrier breakdown after trauma by suppressing the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Laurens Noack
- Department of Radiology and Nuclear Medicine, Experimental Radiology, Otto-von-Guericke University, Magdeburg, Germany
| | | | - Baolin Xu
- Department of Radiology and Nuclear Medicine, Experimental Radiology, Otto-von-Guericke University, Magdeburg, Germany.,Trauma Department, Hannover Medical School, Hannover, Germany
| | - Severin Gylstorff
- Department of Radiology and Nuclear Medicine, Experimental Radiology, Otto-von-Guericke University, Magdeburg, Germany
| | - Yuzhuo Zhou
- Department of Radiology and Nuclear Medicine, Experimental Radiology, Otto-von-Guericke University, Magdeburg, Germany.,Trauma Department, Hannover Medical School, Hannover, Germany
| | - Kernt Köhler
- Institute of Veterinary Pathology, Justus Liebig University Giessen, Giessen, Germany
| | - Phatcharida Jantaree
- Institute of Experimental Internal Medicine, Otto-von-Guericke University, Magdeburg, Germany
| | | | - Aleksander J Nowak
- Department of Radiology and Nuclear Medicine, Experimental Radiology, Otto-von-Guericke University, Magdeburg, Germany
| | - Borna Relja
- Department of Radiology and Nuclear Medicine, Experimental Radiology, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
12
|
Focus on biomarkers, confounders and new therapeutic approaches in trauma. Eur J Trauma Emerg Surg 2022; 48:1521-1523. [PMID: 35701902 PMCID: PMC9192362 DOI: 10.1007/s00068-022-01976-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
13
|
Underwood ML, Park B, Uebelhoer LS, Gu G, Kunkel LE, Korthuis PT, Cook RR, Sekaly RP, Ribeiro SP, Lancioni CL. Chronic Alcohol Exposure Among People Living with HIV Is Associated with Innate Immune Activation and Alterations in Monocyte Phenotype and Plasma Cytokine Profile. Front Immunol 2022; 13:867937. [PMID: 35371104 PMCID: PMC8971672 DOI: 10.3389/fimmu.2022.867937] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 02/23/2022] [Indexed: 12/25/2022] Open
Abstract
Despite advances in antiretroviral therapy, chronic immune activation continues to be observed among individuals with well-controlled HIV viral loads, and is associated with non-AIDS defining morbidities among people living with HIV. Alcohol use disorder impacts a significant proportion of individuals living with HIV, and alcohol exposure is known to damage the intestinal epithelium which may increase translocation of pathogens and their molecular products, driving systemic immune activation and dysregulation. The aim of this study was to determine if adults living with HIV with well-controlled viral loads, who also suffer from alcohol use disorder with and without hepatitis C virus co-infection (n=23), exhibit evidence of advanced systemic immune activation, intestinal damage, and microbial translocation, as compared to adults living with HIV who are not exposed to chronic alcohol or other substances of abuse (n=29). The impact of a 1-month intervention to treat alcohol-use disorder was also examined. Alcohol-use disorder was associated with evidence of advanced innate immune activation, alterations in monocyte phenotype including increased expression of Toll-like receptor 4, increased burden of stimulatory ligands for Toll-like receptor 4, and alterations in plasma cytokine signature, most notably elevations in soluble CD40 ligand and transforming growth factor beta. Alcohol-associated immune activation was more pronounced among individuals with hepatitis C virus co-infection. Although the 1-month intervention to treat alcohol use disorder did not result in significant reductions in the interrogated indicators of immune activation, our findings suggest that chronic alcohol exposure is a major modifiable risk factor for chronic immune activation and dysregulation among people-living with HIV.
Collapse
Affiliation(s)
- Michelle L. Underwood
- Department of Pediatrics, Oregon Health & Science University, Portland, OR, United States
| | - Byung Park
- Knight-Cancer Institute, Department of Biostatistics, Oregon Health & Science University, Portland, OR, United States
| | - Luke S. Uebelhoer
- Department of Pediatrics, Oregon Health & Science University, Portland, OR, United States
| | - Geoffrey Gu
- Undergraduate Studies, University of Southern California, Los Angeles, CA, United States
| | - Lynn E. Kunkel
- Department of Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Philip T. Korthuis
- Department of Medicine, Oregon Health & Science University, Portland, OR, United States
- Department of Public Health, Oregon Health & Science University, Portland, OR, United States
| | - Ryan R. Cook
- Department of Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Rafick Pierre Sekaly
- Department of Pathology & Translational Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - Susan Pereira Ribeiro
- Department of Pathology & Translational Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - Christina L. Lancioni
- Department of Pediatrics, Oregon Health & Science University, Portland, OR, United States
- *Correspondence: Christina L. Lancioni,
| |
Collapse
|
14
|
Janicova A, Haag F, Xu B, Garza AP, Dunay IR, Neunaber C, Nowak AJ, Cavalli P, Marzi I, Sturm R, Relja B. Acute Alcohol Intoxication Modulates Monocyte Subsets and Their Functions in a Time-Dependent Manner in Healthy Volunteers. Front Immunol 2021; 12:652488. [PMID: 34084163 PMCID: PMC8167072 DOI: 10.3389/fimmu.2021.652488] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 04/26/2021] [Indexed: 12/21/2022] Open
Abstract
Background Excessive alcohol intake is associated with adverse immune response-related effects, however, acute and chronic abuse differently modulate monocyte activation. In this study, we have evaluated the phenotypic and functional changes of monocytes in acutely intoxicated healthy volunteers (HV). Methods Twenty-two HV consumed individually adjusted amounts of alcoholic beverages until reaching a blood alcohol level of 1‰ after 4h (T4). Peripheral blood was withdrawn before and 2h (T2), 4h (T4), 6h (T6), 24h (T24), and 48h (T48) after starting the experiment and stained for CD14, CD16 and TLR4. CD14brightCD16-, CD14brightCD16+ and CD14dimCD16+ monocyte subsets and their TLR4 expression were analyzed by flow cytometry. Inflammasome activation via caspase-1 in CD14+ monocytes was measured upon an ex vivo in vitro LPS stimulation. Systemic IL-1β and adhesion capacity of isolated CD14+ monocytes upon LPS stimulation were evaluated. Results The percentage of CD14+ monocyte did not change following alcohol intoxication, whereas CD14brightCD16- monocyte subset significantly increased at T2 and T24, CD14brightCD16+ at T2, T4 and T6 and CD14dimCD16+ at T4 and T6. The relative fraction of TLR4 expressing CD14+ monocytes as well as the density of TLR4 surface presentation increased at T2 and decreased at T48 significantly. TLR4+CD14+ monocytes were significantly enhanced in all subsets at T2. TLR4 expression significantly decreased in CD14brightCD16- at T48, in CD14brightCD16+ at T24 and T48, increased in CD14dimCD16+ at T2. IL-1β release upon LPS stimulation decreased at T48, correlating with TLR4 receptor expression. Alcohol downregulated inflammasome activation following ex vivo in vitro stimulation with LPS between T2 and T48 vs. T0. The adhesion capacity of CD14+ monocytes decreased from T2 with significance at T4, T6 and T48. Following LPS administration, a significant reduction of adhesion was observed at T4 and T6. Conclusions Alcohol intoxication immediately redistributes monocyte subsets toward the pro-inflammatory phenotype with their subsequent differentiation into the anti-inflammatory phenotype. This is paralleled by a significant functional depression, suggesting an alcohol-induced time-dependent hyporesponsiveness of monocytes to pathogenic triggers.
Collapse
Affiliation(s)
- Andrea Janicova
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto von Guericke University, Magdeburg, Germany
| | - Florian Haag
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto von Guericke University, Magdeburg, Germany
| | - Baolin Xu
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto von Guericke University, Magdeburg, Germany
| | - Alejandra P Garza
- Institute of Inflammation and Neurodegeneration, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Ildiko Rita Dunay
- Institute of Inflammation and Neurodegeneration, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | | | - Aleksander J Nowak
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto von Guericke University, Magdeburg, Germany
| | - Paola Cavalli
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto von Guericke University, Magdeburg, Germany
| | - Ingo Marzi
- Department of Trauma, Hand and Reconstructive Surgery, Goethe University, Frankfurt, Germany
| | - Ramona Sturm
- Department of Trauma, Hand and Reconstructive Surgery, Goethe University, Frankfurt, Germany
| | - Borna Relja
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto von Guericke University, Magdeburg, Germany.,Department of Trauma, Hand and Reconstructive Surgery, Goethe University, Frankfurt, Germany
| |
Collapse
|