1
|
Thoudam T, Gao H, Jiang Y, Huda N, Yang Z, Ma J, Liangpunsakul S. Mitochondrial quality control in alcohol-associated liver disease. Hepatol Commun 2024; 8:e0534. [PMID: 39445886 PMCID: PMC11512632 DOI: 10.1097/hc9.0000000000000534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 07/31/2024] [Indexed: 10/25/2024] Open
Abstract
Excessive alcohol consumption is a leading cause of alcohol-associated liver disease (ALD), a significant global health concern with limited therapeutic options. Understanding the key factors contributing to ALD pathogenesis is crucial for identifying potential therapeutic targets. Central to ALD pathogenesis is the intricate interplay between alcohol metabolism and cellular processes, particularly involving mitochondria. Mitochondria are essential organelles in the liver, critical for energy production and metabolic functions. However, they are particularly vulnerable to alcohol-induced damage due to their involvement in alcohol metabolism. Alcohol disrupts mitochondrial function, impairing ATP production and triggering oxidative stress, which leads to cellular damage and inflammation. Mitochondrial quality control mechanisms, including biogenesis, dynamics, and mitophagy, are crucial for maintaining optimal mitochondrial function. Chronic alcohol consumption disrupts mitochondrial quality control checkpoints, leading to mitochondrial dysfunction that impairs fatty acid oxidation and contributes to hepatic steatosis in ALD. Moreover, alcohol promotes the accumulation of damaged mitochondria and the release of proinflammatory components, exacerbating liver damage and inflammation. Preserving mitochondrial health presents a promising therapeutic approach to mitigate ALD progression. In this review, we provide a comprehensive overview of the effects of alcohol on mitochondrial function and quality control mechanisms, highlighting their role in ALD pathogenesis. Understanding these mechanisms may pave the way for the development of novel therapeutic interventions for ALD.
Collapse
Affiliation(s)
- Themis Thoudam
- Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Hui Gao
- Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Yanchao Jiang
- Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Nazmul Huda
- Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Zhihong Yang
- Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Jing Ma
- Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Suthat Liangpunsakul
- Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Roudebush Veterans Administration Medical Center, Indianapolis, Indiana, USA
| |
Collapse
|
2
|
Hatley M, Lam T, Ekeruo I, Taegtmeyer H. Alcohol and Atrial Fibrillation: An Update and New Perspectives. Am J Med 2024; 137:1042-1048. [PMID: 38971529 DOI: 10.1016/j.amjmed.2024.06.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 06/26/2024] [Accepted: 06/28/2024] [Indexed: 07/08/2024]
Abstract
In the Western world, sales of alcoholic beverages are skyrocketing. Alcohol (ethanol) is consumed for its transient euphoric effects but is a risk factor for the development of heart disease. Here, we review the possible association between alcohol consumption and atrial fibrillation. Using a familiar analogy, we propose that atrial fibrillation is the mere tip of an iceberg (alcohol-associated heart disease). Our concern is that the many research studies on the effects of ethanol on the heart have produced inconsistent results. These include studies of individuals drinking only moderate amounts of alcoholic beverages (aka the "French paradox") on the one hand, and paroxysmal atrial fibrillation after binge drinking ("holiday heart syndrome") on the other hand. The evidence available in the literature suggests that hypertension, structured heart disease of any form, neurohumoral stress, and cardiometabolic disorders all favor the development of atrial fibrillation triggered by alcohol. We also suggest that alcohol should be classified as a modifiable risk factor for atrial fibrillation, and also for heart disease in general.
Collapse
Affiliation(s)
- Marsha Hatley
- Division of Cardiovascular Medicine, Department of Internal Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston
| | - Truong Lam
- Division of Pediatrics, Department of Pediatrics - Research, The University of Texas MD Anderson Cancer Center, Houston
| | - Ijeoma Ekeruo
- Division of Cardiovascular Medicine, Department of Internal Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston
| | - Heinrich Taegtmeyer
- Division of Cardiovascular Medicine, Department of Internal Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston.
| |
Collapse
|
3
|
Bouchebti S, Gershon Y, Gordin A, Huchon D, Levin E. Tolerance and efficient metabolization of extremely high ethanol concentrations by a social wasp. Proc Natl Acad Sci U S A 2024; 121:e2410874121. [PMID: 39432778 DOI: 10.1073/pnas.2410874121] [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: 05/31/2024] [Accepted: 09/07/2024] [Indexed: 10/23/2024] Open
Abstract
Ethanol, a natural by-product of sugar fermentation, can be found in various fruits and nectar. Although many animals routinely consume ethanol in low concentrations as part of their natural diets, its inherent toxicity can cause severe damage. Even species particularly well adapted to ethanol consumption face detrimental effects when exposed to concentrations above 4%. Here, we investigated the metabolism of ethanol and its impact on survival and behavior in the Oriental hornet (Vespa orientalis), a social wasp that naturally consumes ethanol. We show that chronic ethanol consumption, even at concentrations as high as 80%, had no impact on hornet mortality, construction behavior, or agonistic behavior. Using 13C1 labeled ethanol, we show that hornets efficiently metabolized ingested ethanol and at a much higher rate than honey bees. The presence of multiple copies of the alcohol dehydrogenase (NADP+) gene in the Vespa genera suggests a potential mechanism for ethanol tolerance. These findings support the hypothesis that the mutualistic relationship between ethanol-producing organisms and vespid hosts may be at the origin of their remarkable capacity to utilize and metabolize ethanol.
Collapse
Affiliation(s)
- Sofia Bouchebti
- School of Zoology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Yael Gershon
- School of Zoology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Alexander Gordin
- The ADAMA Center for Novel Delivery Systems in Crop Protection, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Dorothée Huchon
- School of Zoology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
- The Steinhardt Museum of Natural History and National Research Center, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Eran Levin
- School of Zoology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
4
|
Zhang L, Zhang Y, Liu S, Song J, Suo H. Degradation effects and mechanisms of Limosilactobacillus fermentum on ethanol. Food Funct 2024; 15:10283-10299. [PMID: 39233486 DOI: 10.1039/d4fo02918f] [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: 09/06/2024]
Abstract
Acute heavy drinking can lead to a rapid increase in blood ethanol concentration, resulting in dizziness, liver damage, and other adverse effects. Although lactic acid bacteria possess the ability to degrade ethanol, the mechanisms remain unclear. For the first time, our study revealed that Limosilactobacillus fermentum DACN611, derived from traditional Chinese fermented yogurt, exhibited superior ethanol degradation capability, achieving a 90.87% ± 8.12% reduction in ethanol concentration in a 2.5% (v/v) ethanol MRS broth over 24 h, among fifty lactic acid bacteria strains. Notably, transcriptome analysis of DACN611 under ethanol stress conditions revealed that DACN611 degraded ethanol by adjusting the cell cycle, promoting protein synthesis, maintaining oxidative metabolic homeostasis, and modulating cell wall and membrane synthesis along with other metabolic pathways. Additionally, DACN611 showed excellent resistance to gastric acid and bile salts, along with a safe profile. In the acute heavy drinking Kunming mouse model, DACN611 significantly increased the latency of the loss of righting reflex (LORR) and reduced the LORR duration. Serum ethanol and acetaldehyde concentrations decreased by 35.36% and 33.56%, respectively. The gastric and hepatic activities of alcohol dehydrogenase (ADH) and acetaldehyde dehydrogenase (ALDH) increased by 1.98-fold and 1.95-fold, and 1.79-fold and 1.70-fold, respectively. In addition, DACN611 decreased serum alanine aminotransferase and aspartate aminotransferase levels, and reduced hepatic cytochrome P450 2E1 expression. It also alleviated pathological liver changes, demonstrating protective effects against alcoholic liver injury in mice. In conclusion, DACN611 significantly degraded ethanol through adaptive metabolic changes under ethanol stress conditions and the promotion of ADH and ALDH activities in gastric and hepatic tissues.
Collapse
Affiliation(s)
- Lingling Zhang
- College of Food Science, Southwest University, Chongqing, China.
- Chongqing Agricultural Product Processing Technology Innovation Platform, Southwest University, Chongqing, China
| | - Yuhong Zhang
- Institute of Food Sciences and Technology, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, China
| | - Shijian Liu
- College of Food Science, Southwest University, Chongqing, China.
- Chongqing Agricultural Product Processing Technology Innovation Platform, Southwest University, Chongqing, China
| | - Jiajia Song
- College of Food Science, Southwest University, Chongqing, China.
- Chongqing Agricultural Product Processing Technology Innovation Platform, Southwest University, Chongqing, China
| | - Huayi Suo
- College of Food Science, Southwest University, Chongqing, China.
- Chongqing Agricultural Product Processing Technology Innovation Platform, Southwest University, Chongqing, China
- National Citrus Engineering Research Center, Southwest University, Chongqing, China
| |
Collapse
|
5
|
Yang Z, Gao H, Ma J, Liang NA, Liang SP, Huda N, Jiang Y, Thoudam T, Tu W, Su J, Hesler M, Chandler K, Liangpunsakul S. Unique urine and serum metabolomic signature in patients with excessive alcohol use: An exploratory study. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2024; 48:1519-1528. [PMID: 38951043 DOI: 10.1111/acer.15398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 06/02/2024] [Accepted: 06/04/2024] [Indexed: 07/03/2024]
Abstract
BACKGROUND Excessive alcohol consumption has a multifaceted impact on the body's metabolic pathways and organ systems. The objectives of this study were to characterize global metabolomic changes and identify specific pathways that are altered in individuals with excessive alcohol use. METHODS This exploratory study included 22 healthy controls with no known history of excessive alcohol use and 38 patients identified as using alcohol excessively. A Fibrosis-4 score was used to determine the risk of underlying alcohol-associated liver disease among the excessive drinkers. RESULTS We found significantly altered urinary and serum metabolites among excessive drinkers, affecting various metabolic pathways including the metabolism of lipids, amino acids and peptides, cofactors and vitamins, carbohydrates, and nucleotides. Levels of two steroid hormones-5alpha-androstan-3beta,17beta-diol disulfate and androstenediol (3beta,17beta) disulfate-were significantly higher in both the serum and urine samples of excessive drinkers. These elevated levels may be associated with a higher risk of liver fibrosis in individuals with excessive alcohol use. CONCLUSION Alcohol consumption leads to marked alterations in multiple metabolic pathways, highlighting the systemic impact of alcohol on various tissues and organ systems. These findings provide a foundation for future mechanistic studies aimed at elucidating alcohol-induced changes in these metabolic pathways and their implications.
Collapse
Affiliation(s)
- Zhihong Yang
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Hui Gao
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Jing Ma
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | | | - Nazmul Huda
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Yanchao Jiang
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Themis Thoudam
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Wanzhu Tu
- Department of Biostatistics and Health Data Science, Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, Indiana, USA
| | - Jing Su
- Department of Biostatistics and Health Data Science, Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, Indiana, USA
| | - Maggie Hesler
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Kristina Chandler
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Suthat Liangpunsakul
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Roudebush Veterans Administration Medical Center, Indianapolis, Indiana, USA
| |
Collapse
|
6
|
Kong EQZ, Subramaniyan V, Lubau NSA. Uncovering the impact of alcohol on internal organs and reproductive health: Exploring TLR4/NF-kB and CYP2E1/ROS/Nrf2 pathways. Animal Model Exp Med 2024; 7:444-459. [PMID: 38853347 PMCID: PMC11369036 DOI: 10.1002/ame2.12436] [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: 12/25/2023] [Accepted: 05/10/2024] [Indexed: 06/11/2024] Open
Abstract
This review delves into the detrimental impact of alcohol consumption on internal organs and reproductive health, elucidating the underlying mechanisms involving the Toll-like receptor 4 (TLR4)/Nuclear factor kappa light chain enhancer of activated B cells (NF-kB) pathway and the Cytochrome P450 2E1 (CYP2E1)/reactive oxygen species (ROS)/nuclear factor erythroid 2-related factor 2 (Nrf2) pathways. The TLR4/NF-kB pathway, crucial for inflammatory and immune responses, triggers the production of pro-inflammatory agents and type-1 interferon, disrupting the balance between inflammatory and antioxidant responses when tissues are chronically exposed to alcohol. Alcohol-induced dysbiosis in gut microbes heightens gut wall permeability to pathogen-associated molecular patterns (PAMPs), leading to liver cell infection and subsequent inflammation. Concurrently, CYP2E1-mediated alcohol metabolism generates ROS, causing oxidative stress and damaging cells, lipids, proteins, and deoxyribonucleic acid (DNA). To counteract this inflammatory imbalance, Nrf2 regulates gene expression, inhibiting inflammatory progression and promoting antioxidant responses. Excessive alcohol intake results in elevated liver enzymes (ADH, CYP2E1, and catalase), ROS, NADH, acetaldehyde, and acetate, leading to damage in vital organs such as the heart, brain, and lungs. Moreover, alcohol negatively affects reproductive health by inhibiting the hypothalamic-pituitary-gonadal axis, causing infertility in both men and women. These findings underscore the profound health concerns associated with alcohol-induced damage, emphasizing the need for public awareness regarding the intricate interplay between immune responses and the multi-organ impacts of alcohol consumption.
Collapse
Affiliation(s)
- Eason Qi Zheng Kong
- Pharmacology Unit, Jeffrey Cheah School of Medicine and Health SciencesMonash University MalaysiaSubang JayaSelangorMalaysia
| | - Vetriselvan Subramaniyan
- Pharmacology Unit, Jeffrey Cheah School of Medicine and Health SciencesMonash University MalaysiaSubang JayaSelangorMalaysia
- Center for Global Health Research, Saveetha Medical CollegeSaveetha Institute of Medical and Technical SciencesChennaiTamil NaduIndia
| | - Natasha Sura Anak Lubau
- Pharmacology Unit, Jeffrey Cheah School of Medicine and Health SciencesMonash University MalaysiaSubang JayaSelangorMalaysia
| |
Collapse
|
7
|
Meijnikman AS, Nieuwdorp M, Schnabl B. Endogenous ethanol production in health and disease. Nat Rev Gastroenterol Hepatol 2024; 21:556-571. [PMID: 38831008 DOI: 10.1038/s41575-024-00937-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/23/2024] [Indexed: 06/05/2024]
Abstract
The gut microbiome exerts metabolic actions on distal tissues and organs outside the intestine, partly through microbial metabolites that diffuse into the circulation. The disruption of gut homeostasis results in changes to microbial metabolites, and more than half of the variance in the plasma metabolome can be explained by the gut microbiome. Ethanol is a major microbial metabolite that is produced in the intestine of nearly all individuals; however, elevated ethanol production is associated with pathological conditions such as metabolic dysfunction-associated steatotic liver disease and auto-brewery syndrome, in which the liver's capacity to metabolize ethanol is surpassed. In this Review, we describe the mechanisms underlying excessive ethanol production in the gut and the role of ethanol catabolism in mediating pathogenic effects of ethanol on the liver and host metabolism. We conclude by discussing approaches to target excessive ethanol production by gut bacteria.
Collapse
Affiliation(s)
| | - Max Nieuwdorp
- Department of Internal Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, Netherlands
- Department of Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, Netherlands
- Diabeter Centrum Amsterdam, Amsterdam, Netherlands
| | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, CA, USA.
- Department of Medicine, VA San Diego Healthcare System, San Diego, CA, USA.
- Center for Innovative Phage Applications and Therapeutics, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
8
|
Osik N, Lukzen NN, Yanshole VV, Tsentalovich YP. Loss of Volatile Metabolites during Concentration of Metabolomic Extracts. ACS OMEGA 2024; 9:24015-24024. [PMID: 38854568 PMCID: PMC11154959 DOI: 10.1021/acsomega.4c02439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/02/2024] [Accepted: 05/16/2024] [Indexed: 06/11/2024]
Abstract
Volatile metabolites can be lost during the preanalytical stage of metabolomic analysis. This work is aimed at the experimental and theoretical study of mechanisms of volatile substance evaporation and retention in the residues during the drying of extract solutions. We demonstrate that solvent evaporation leads to the unavoidable loss of nondissociating volatile metabolites with low boiling points and high vapor pressures (such as acetone and ethanol). The retention of dissociating volatile compounds (primarily organic acids RH) during the evaporation depends on the presence of buffer salts in solution, which are responsible for maintaining the neutral pH. An acid remains in the solution as long as it is present predominantly in the dissociated R- state. At the very last stage of solvent evaporation, buffer salts precipitate, forming a solid matrix for metabolite trapping in the residue. At the same time, buffer precipitation leads to a decrease of the solution pH, increase of the portion of RH in associated state, and acceleration of RH volatilization. The RH recovery is thus determined by the competition between the solute volatilization in the associated RH form and metabolite trapping in the solid matrix. The retention of volatile acids in the residue after extract drying can be improved either by adding buffer salts to maintain high pH or by incomplete sample drying.
Collapse
Affiliation(s)
- Nataliya
A. Osik
- International
Tomography Center Siberian Branch of Russian Academy of Sciences, Institutskaya str. 3a, Novosibirsk 630090, Russia
| | - Nikita N. Lukzen
- International
Tomography Center Siberian Branch of Russian Academy of Sciences, Institutskaya str. 3a, Novosibirsk 630090, Russia
- Novosibirsk
State University, Pirogova
str. 1, Novosibirsk 630090, Russia
| | - Vadim V. Yanshole
- International
Tomography Center Siberian Branch of Russian Academy of Sciences, Institutskaya str. 3a, Novosibirsk 630090, Russia
- Novosibirsk
State University, Pirogova
str. 1, Novosibirsk 630090, Russia
| | - Yuri P. Tsentalovich
- International
Tomography Center Siberian Branch of Russian Academy of Sciences, Institutskaya str. 3a, Novosibirsk 630090, Russia
| |
Collapse
|
9
|
Yang YJ, Kim MJ, Yang JH, Heo JW, Kim HH, Kim WH, Kim GS, Lee HJ, Kim YW, Kim KY, Park KI. Liquid Chromatography/Tandem Mass Spectrometry Analysis of Sophora flavescens Aiton and Protective Effects against Alcohol-Induced Liver Injury and Oxidative Stress in Mice. Antioxidants (Basel) 2024; 13:541. [PMID: 38790646 PMCID: PMC11117756 DOI: 10.3390/antiox13050541] [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: 03/13/2024] [Revised: 04/26/2024] [Accepted: 04/26/2024] [Indexed: 05/26/2024] Open
Abstract
In this study, we investigated the hepatoprotective effects of an ethanol extract of Sophora flavescens Aiton (ESF) on an alcohol-induced liver disease mouse model. Alcoholic liver disease (ALD) was caused by the administration of ethanol to male C57/BL6 mice who were given a Lieber-DeCarli liquid diet, including ethanol. The alcoholic fatty liver disease mice were orally administered ESF (100 and 200 mg/kg bw/day) or silymarin (50 mg/kg bw/day), which served as a positive control every day for 16 days. The findings suggest that ESF enhances hepatoprotective benefits by significantly decreasing serum levels of aspartate transaminase (AST) and alanine transaminase (ALT), markers for liver injury. Furthermore, ESF alleviated the accumulation of triglyceride (TG) and total cholesterol (TC), increased serum levels of superoxide dismutase (SOD) and glutathione (GSH), and improved serum alcohol dehydrogenase (ADH) activity in the alcoholic fatty liver disease mice model. Cells and organisms rely on the Kelch-like ECH-associated protein 1- Nuclear factor erythroid 2-related factor 2 (Keap1-Nrf2) system as a critical defensive mechanism in response to oxidative stress. Therefore, Nrf2 plays an important role in ALD antioxidant responses, and its level is decreased by increased reactive oxidation stress (ROS) in the liver. ESF increased Nrf2, which was decreased in ethanol-damaged livers. Additionally, four polyphenol compounds were identified through a qualitative analysis of the ESF using LC-MS/MS. This study confirmed ESF's antioxidative and hangover-elimination effects and suggested the possibility of using Sophora flavescens Aiton (SF) to treat ALD.
Collapse
Affiliation(s)
- Ye Jin Yang
- Departments of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (Y.J.Y.); (M.J.K.); (J.W.H.); (H.H.K.); (W.H.K.); (G.S.K.); (H.-J.L.)
| | - Min Jung Kim
- Departments of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (Y.J.Y.); (M.J.K.); (J.W.H.); (H.H.K.); (W.H.K.); (G.S.K.); (H.-J.L.)
| | - Ju-Hye Yang
- Korean Medicine (KM) Application Center, Korea Institute of Oriental Medicine, Daegu 41062, Republic of Korea;
| | - Ji Woong Heo
- Departments of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (Y.J.Y.); (M.J.K.); (J.W.H.); (H.H.K.); (W.H.K.); (G.S.K.); (H.-J.L.)
| | - Hun Hwan Kim
- Departments of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (Y.J.Y.); (M.J.K.); (J.W.H.); (H.H.K.); (W.H.K.); (G.S.K.); (H.-J.L.)
| | - Woo H. Kim
- Departments of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (Y.J.Y.); (M.J.K.); (J.W.H.); (H.H.K.); (W.H.K.); (G.S.K.); (H.-J.L.)
| | - Gon Sup Kim
- Departments of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (Y.J.Y.); (M.J.K.); (J.W.H.); (H.H.K.); (W.H.K.); (G.S.K.); (H.-J.L.)
| | - Hu-Jang Lee
- Departments of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (Y.J.Y.); (M.J.K.); (J.W.H.); (H.H.K.); (W.H.K.); (G.S.K.); (H.-J.L.)
| | - Young Woo Kim
- School of Korean Medicine, Dongguk University, Gyeongju 38066, Republic of Korea;
| | - Kwang Youn Kim
- Korean Medicine (KM) Application Center, Korea Institute of Oriental Medicine, Daegu 41062, Republic of Korea;
| | - Kwang Il Park
- Departments of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (Y.J.Y.); (M.J.K.); (J.W.H.); (H.H.K.); (W.H.K.); (G.S.K.); (H.-J.L.)
| |
Collapse
|
10
|
Lu H. Inflammatory liver diseases and susceptibility to sepsis. Clin Sci (Lond) 2024; 138:435-487. [PMID: 38571396 DOI: 10.1042/cs20230522] [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: 09/03/2023] [Revised: 01/09/2024] [Accepted: 03/12/2024] [Indexed: 04/05/2024]
Abstract
Patients with inflammatory liver diseases, particularly alcohol-associated liver disease and metabolic dysfunction-associated fatty liver disease (MAFLD), have higher incidence of infections and mortality rate due to sepsis. The current focus in the development of drugs for MAFLD is the resolution of non-alcoholic steatohepatitis and prevention of progression to cirrhosis. In patients with cirrhosis or alcoholic hepatitis, sepsis is a major cause of death. As the metabolic center and a key immune tissue, liver is the guardian, modifier, and target of sepsis. Septic patients with liver dysfunction have the highest mortality rate compared with other organ dysfunctions. In addition to maintaining metabolic homeostasis, the liver produces and secretes hepatokines and acute phase proteins (APPs) essential in tissue protection, immunomodulation, and coagulation. Inflammatory liver diseases cause profound metabolic disorder and impairment of energy metabolism, liver regeneration, and production/secretion of APPs and hepatokines. Herein, the author reviews the roles of (1) disorders in the metabolism of glucose, fatty acids, ketone bodies, and amino acids as well as the clearance of ammonia and lactate in the pathogenesis of inflammatory liver diseases and sepsis; (2) cytokines/chemokines in inflammatory liver diseases and sepsis; (3) APPs and hepatokines in the protection against tissue injury and infections; and (4) major nuclear receptors/signaling pathways underlying the metabolic disorders and tissue injuries as well as the major drug targets for inflammatory liver diseases and sepsis. Approaches that focus on the liver dysfunction and regeneration will not only treat inflammatory liver diseases but also prevent the development of severe infections and sepsis.
Collapse
Affiliation(s)
- Hong Lu
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY 13210, U.S.A
| |
Collapse
|
11
|
Popova D, Sun J, Chow HM, Hart RP. A critical review of ethanol effects on neuronal firing: A metabolic perspective. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2024; 48:450-458. [PMID: 38217065 DOI: 10.1111/acer.15266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 12/22/2023] [Accepted: 01/02/2024] [Indexed: 01/14/2024]
Abstract
Ethanol metabolism is relatively understudied in neurons, even though changes in neuronal metabolism are known to affect their activity. Recent work demonstrates that ethanol is preferentially metabolized over glucose as a source of carbon and energy, and it reprograms neurons to a state of reduced energy potential and diminished capacity to utilize glucose once ethanol is exhausted. Ethanol intake has been associated with changes in neuronal firing and specific brain activity (EEG) patterns have been linked with risk for alcohol use disorder (AUD). Furthermore, a haplotype of the inwardly rectifying potassium channel subunit, GIRK2, which plays a critical role in regulating excitability of neurons, has been linked with AUD and shown to be directly regulated by ethanol. At the same time, overexpression of GIRK2 prevents ethanol-induced metabolic changes. Based on the available evidence, we conclude that the mechanisms underlying the effects of ethanol on neuronal metabolism are a novel target for developing therapies for AUD.
Collapse
Affiliation(s)
- Dina Popova
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey, USA
| | - Jacquelyne Sun
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Hei-Man Chow
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong, Hong Kong
- Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Ronald P Hart
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey, USA
| |
Collapse
|
12
|
Bhagat B, Gupta SK, Mandal D, Gor AA, Bandyopadhyay R, Mukherjee K. Probing the p-type Chemiresistive Response of NiFe 2 O 4 Nanoparticles for Potential Utilization as Ethanol Sensor. Chem Asian J 2024; 19:e202300841. [PMID: 38100152 DOI: 10.1002/asia.202300841] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/20/2023] [Indexed: 01/24/2024]
Abstract
Detection of gas molecules and volatile organic compounds (VOCs) using efficient, low cost sensors has fetched significant attention in environmental monitoring, safety measures and medical diagnosis. In the present work, nickel ferrite (NFO) nanoparticles are explored as p-type semiconducting metal oxide (SMO) sensor for detection of five different organic vapors namely methanol, ethanol, n-propanol, iso-propanol and acetone which often cause severe damage to human body under prolonged exposure. The sensing studies in presence of the aforementioned five vapors are carried out by varying the sensor operating temperature (225-300 °C) and vapor concentrations (10-1000 ppm). Developed NFO sensor demonstrated best performance in terms of sensing (~10 ppm), response time (<10 s), excellent repeatability and selectivity towards ethanol among all other considered gas species. The repeatability of the sensor response is verified and the underlying reasons for the variation in the response of NFO sensor due to the change of operating temperature, analyte type and concentrations has been discussed. The synthesis of NFO through auto combustion method and study on their formation behaviour, oxygen vacancy evolution, band gap calculation, crystalline nature as well as microstructural features provides here the comprehensive information about the potential application of NFO nanoparticles as gas sensor.
Collapse
Affiliation(s)
- B Bhagat
- Department of Chemistry, School of Energy Technology, Pandit Deendayal Energy University, Gandhinagar, 382426, Gujarat, India
| | - Santosh K Gupta
- Radiochemistry Division, Bhabha Atomic Research Centre, Mumbai, 400085, India
- Homi Bhabha National Institute, Anushaktinagar, Mumbai, 400094, India
| | - D Mandal
- Institute of Nano Science and Technology, Mohali, India
| | - Abhishek A Gor
- Department of Physics, School of Energy Technology, Pandit Deendayal Energy University, Gandhinagar, 382426, Gujarat, India
| | - R Bandyopadhyay
- Department of Chemistry, School of Energy Technology, Pandit Deendayal Energy University, Gandhinagar, 382426, Gujarat, India
| | - K Mukherjee
- Department of Chemistry, School of Energy Technology, Pandit Deendayal Energy University, Gandhinagar, 382426, Gujarat, India
| |
Collapse
|
13
|
Jiang N, Li W, Jiang S, Xie M, Liu R. Acetylation in pathogenesis: Revealing emerging mechanisms and therapeutic prospects. Biomed Pharmacother 2023; 167:115519. [PMID: 37729729 DOI: 10.1016/j.biopha.2023.115519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/08/2023] [Accepted: 09/14/2023] [Indexed: 09/22/2023] Open
Abstract
Protein acetylation modifications play a central and pivotal role in a myriad of biological processes, spanning cellular metabolism, proliferation, differentiation, apoptosis, and beyond, by effectively reshaping protein structure and function. The metabolic state of cells is intricately connected to epigenetic modifications, which in turn influence chromatin status and gene expression patterns. Notably, pathological alterations in protein acetylation modifications are frequently observed in diseases such as metabolic syndrome, cardiovascular disorders, and cancer. Such abnormalities can result in altered protein properties and loss of function, which are closely associated with developing and progressing related diseases. In recent years, the advancement of precision medicine has highlighted the potential value of protein acetylation in disease diagnosis, treatment, and prevention. This review includes provocative and thought-provoking papers outlining recent breakthroughs in acetylation modifications as they relate to cardiovascular disease, mitochondrial metabolic regulation, liver health, neurological health, obesity, diabetes, and cancer. Additionally, it covers the molecular mechanisms and research challenges in understanding the role of acetylation in disease regulation. By summarizing novel targets and prognostic markers for the treatment of related diseases, we aim to contribute to the field. Furthermore, we discuss current hot topics in acetylation research related to health regulation, including N4-acetylcytidine and liquid-liquid phase separation. The primary objective of this review is to provide insights into the functional diversity and underlying mechanisms by which acetylation regulates proteins in disease contexts.
Collapse
Affiliation(s)
- Nan Jiang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Wenyong Li
- School of Biology and Food Engineering, Fuyang Normal University, Fuyang, Anhui 236037, China
| | - Shuanglin Jiang
- School of Biology and Food Engineering, Fuyang Normal University, Fuyang, Anhui 236037, China
| | - Ming Xie
- North China Petroleum Bureau General Hospital, Renqiu 062550, China.
| | - Ran Liu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China.
| |
Collapse
|
14
|
Dharavath RN, Pina-Leblanc C, Tang VM, Sloan ME, Nikolova YS, Pangarov P, Ruocco AC, Shield K, Voineskos D, Blumberger DM, Boileau I, Bozinoff N, Gerretsen P, Vieira E, Melamed OC, Sibille E, Quilty LC, Prevot TD. GABAergic signaling in alcohol use disorder and withdrawal: pathological involvement and therapeutic potential. Front Neural Circuits 2023; 17:1218737. [PMID: 37929054 PMCID: PMC10623140 DOI: 10.3389/fncir.2023.1218737] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 09/04/2023] [Indexed: 11/07/2023] Open
Abstract
Alcohol is one of the most widely used substances. Alcohol use accounts for 5.1% of the global disease burden, contributes substantially to societal and economic costs, and leads to approximately 3 million global deaths yearly. Alcohol use disorder (AUD) includes various drinking behavior patterns that lead to short-term or long-lasting effects on health. Ethanol, the main psychoactive molecule acting in alcoholic beverages, directly impacts the GABAergic system, contributing to GABAergic dysregulations that vary depending on the intensity and duration of alcohol consumption. A small number of interventions have been developed that target the GABAergic system, but there are promising future therapeutic avenues to explore. This review provides an overview of the impact of alcohol on the GABAergic system, the current interventions available for AUD that target the GABAergic system, and the novel interventions being explored that in the future could be included among first-line therapies for the treatment of AUD.
Collapse
Affiliation(s)
| | - Celeste Pina-Leblanc
- Campbell Family Mental Health Research Institute of CAMH, Toronto, ON, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Victor M. Tang
- Campbell Family Mental Health Research Institute of CAMH, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Addiction Division, CAMH, Toronto, ON, Canada
- Division of Neurosciences and Clinical Translation, Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Institute of Mental Health Policy Research, CAMH, Toronto, ON, Canada
| | - Matthew E. Sloan
- Campbell Family Mental Health Research Institute of CAMH, Toronto, ON, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
- Addiction Division, CAMH, Toronto, ON, Canada
- Division of Neurosciences and Clinical Translation, Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Department of Psychological Clinical Science, University of Toronto Scarborough, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Institute of Mental Health Policy Research, CAMH, Toronto, ON, Canada
| | - Yuliya S. Nikolova
- Campbell Family Mental Health Research Institute of CAMH, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Peter Pangarov
- Campbell Family Mental Health Research Institute of CAMH, Toronto, ON, Canada
| | - Anthony C. Ruocco
- Campbell Family Mental Health Research Institute of CAMH, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Temerty Centre for Therapeutic Brain Intervention, CAMH, Toronto, ON, Canada
- Department of Psychology, University of Toronto Scarborough, Toronto, ON, Canada
| | - Kevin Shield
- Institute of Mental Health Policy Research, CAMH, Toronto, ON, Canada
| | - Daphne Voineskos
- Campbell Family Mental Health Research Institute of CAMH, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Temerty Centre for Therapeutic Brain Intervention, CAMH, Toronto, ON, Canada
| | - Daniel M. Blumberger
- Campbell Family Mental Health Research Institute of CAMH, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Temerty Centre for Therapeutic Brain Intervention, CAMH, Toronto, ON, Canada
| | - Isabelle Boileau
- Campbell Family Mental Health Research Institute of CAMH, Toronto, ON, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Brain Health Imaging Centre, CAMH, Toronto, ON, Canada
| | - Nikki Bozinoff
- Campbell Family Mental Health Research Institute of CAMH, Toronto, ON, Canada
- Department of Family and Community Medicine, University of Toronto, Toronto, ON, Canada
| | - Philip Gerretsen
- Campbell Family Mental Health Research Institute of CAMH, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Brain Health Imaging Centre, CAMH, Toronto, ON, Canada
| | - Erica Vieira
- Campbell Family Mental Health Research Institute of CAMH, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Osnat C. Melamed
- Campbell Family Mental Health Research Institute of CAMH, Toronto, ON, Canada
- Department of Family and Community Medicine, University of Toronto, Toronto, ON, Canada
| | - Etienne Sibille
- Campbell Family Mental Health Research Institute of CAMH, Toronto, ON, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Lena C. Quilty
- Campbell Family Mental Health Research Institute of CAMH, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Thomas D. Prevot
- Campbell Family Mental Health Research Institute of CAMH, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
15
|
Li A, Shami GJ, Griffiths L, Lal S, Irving H, Braet F. Giant mitochondria in cardiomyocytes: cellular architecture in health and disease. Basic Res Cardiol 2023; 118:39. [PMID: 37775647 PMCID: PMC10541842 DOI: 10.1007/s00395-023-01011-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/29/2023] [Accepted: 09/15/2023] [Indexed: 10/01/2023]
Abstract
Giant mitochondria are frequently observed in different disease models within the brain, kidney, and liver. In cardiac muscle, these enlarged organelles are present across diverse physiological and pathophysiological conditions including in ageing and exercise, and clinically in alcohol-induced heart disease and various cardiomyopathies. This mitochondrial aberration is widely considered an early structural hallmark of disease leading to adverse organ function. In this thematic paper, we discuss the current state-of-knowledge on the presence, structure and functional implications of giant mitochondria in heart muscle. Despite its demonstrated reoccurrence in different heart diseases, the literature on this pathophysiological phenomenon remains relatively sparse since its initial observations in the early 60s. We review historical and contemporary investigations from cultured cardiomyocytes to human tissue samples to address the role of giant mitochondria in cardiac health and disease. Finally, we discuss their significance for the future development of novel mitochondria-targeted therapies to improve cardiac metabolism and functionality.
Collapse
Affiliation(s)
- Amy Li
- Department of Rural Clinical Sciences, La Trobe Rural Health School, La Trobe University, Bendigo, VIC, Australia.
- Centre for Healthy Futures, Torrens University Australia, Surry Hills, NSW, Australia.
- School of Medical Sciences, The University of Sydney, Camperdown, NSW, Australia.
| | - Gerald J Shami
- School of Medical Sciences (Molecular and Cellular Biomedicine), The University of Sydney, Camperdown, NSW, Australia
- Australian Centre for Microscopy and Microanalysis, The University of Sydney, Camperdown, NSW, Australia
| | - Lisa Griffiths
- Anatomical Pathology, PathWest, QEII Medical Centre, Nedlands, WA, Australia
| | - Sean Lal
- School of Medical Sciences, The University of Sydney, Camperdown, NSW, Australia
| | - Helen Irving
- Department of Rural Clinical Sciences, La Trobe Rural Health School, La Trobe University, Bendigo, VIC, Australia
| | - Filip Braet
- School of Medical Sciences (Molecular and Cellular Biomedicine), The University of Sydney, Camperdown, NSW, Australia.
- Australian Centre for Microscopy and Microanalysis, The University of Sydney, Camperdown, NSW, Australia.
| |
Collapse
|
16
|
Padovan JC, Dourado TMH, Pimenta GF, Bruder-Nascimento T, Tirapelli CR. Reactive Oxygen Species Are Central Mediators of Vascular Dysfunction and Hypertension Induced by Ethanol Consumption. Antioxidants (Basel) 2023; 12:1813. [PMID: 37891892 PMCID: PMC10604002 DOI: 10.3390/antiox12101813] [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: 09/04/2023] [Revised: 09/23/2023] [Accepted: 09/28/2023] [Indexed: 10/29/2023] Open
Abstract
Consumption of high amounts of ethanol is a risk factor for development of cardiovascular diseases such as arterial hypertension. The hypertensive state induced by ethanol is a complex multi-factorial event, and oxidative stress is a pathophysiological hallmark of vascular dysfunction associated with ethanol consumption. Increasing levels of reactive oxygen species (ROS) in the vasculature trigger important processes underlying vascular injury, including accumulation of intracellular Ca2+ ions, reduced bioavailability of nitric oxide (NO), activation of mitogen-activated protein kinases (MAPKs), endothelial dysfunction, and loss of the anticontractile effect of perivascular adipose tissue (PVAT). The enzyme nicotinamide adenine dinucleotide phosphate (NADPH) oxidase plays a central role in vascular ROS generation in response to ethanol. Activation of the renin-angiotensin-aldosterone system (RAAS) is an upstream mechanism which contributes to NADPH oxidase stimulation, overproduction of ROS, and vascular dysfunction. This review discusses the mechanisms of vascular dysfunction induced by ethanol, detailing the contribution of ROS to these processes. Data examining the association between neuroendocrine changes and vascular oxidative stress induced by ethanol are also reviewed and discussed. These issues are of paramount interest to public health as ethanol contributes to blood pressure elevation in the general population, and it is linked to cardiovascular conditions and diseases.
Collapse
Affiliation(s)
- Júlio C. Padovan
- Laboratory of Blood and Vascular Biology, The Rockefeller University, New York, NY 10065, USA;
| | - Thales M. H. Dourado
- Programa de Pós-Graduação em Farmacologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo (USP), Ribeirão Preto 14040-902, SP, Brazil; (T.M.H.D.); (G.F.P.)
- Departamento de Enfermagem Psiquiátrica e Ciências Humanas, Laboratório de Farmacologia, Escola de Enfermagem de Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-902, SP, Brazil
| | - Gustavo F. Pimenta
- Programa de Pós-Graduação em Farmacologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo (USP), Ribeirão Preto 14040-902, SP, Brazil; (T.M.H.D.); (G.F.P.)
- Departamento de Enfermagem Psiquiátrica e Ciências Humanas, Laboratório de Farmacologia, Escola de Enfermagem de Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-902, SP, Brazil
| | - Thiago Bruder-Nascimento
- Department of Pediatrics and Vascular Medicine Institute (VMI), University of Pittsburgh, Pittsburgh, PA 15260, USA;
| | - Carlos R. Tirapelli
- Departamento de Enfermagem Psiquiátrica e Ciências Humanas, Laboratório de Farmacologia, Escola de Enfermagem de Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-902, SP, Brazil
| |
Collapse
|
17
|
Lee SH, Shnitko TA, Hsu LM, Broadwater MA, Sardinas M, Wang TWW, Robinson DL, Vetreno RP, Crews FT, Shih YYI. Acute alcohol induces greater dose-dependent increase in the lateral cortical network functional connectivity in adult than adolescent rats. ADDICTION NEUROSCIENCE 2023; 7:100105. [PMID: 37576436 PMCID: PMC10421607 DOI: 10.1016/j.addicn.2023.100105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Alcohol misuse and, particularly adolescent drinking, is a major public health concern. While evidence suggests that adolescent alcohol use affects frontal brain regions that are important for cognitive control over behavior little is known about how acute alcohol exposure alters large-scale brain networks and how sex and age may moderate such effects. Here, we employ a recently developed functional magnetic resonance imaging (fMRI) protocol to acquire rat brain functional connectivity data and use an established analytical pipeline to examine the effect of sex, age, and alcohol dose on connectivity within and between three major rodent brain networks: defaul mode, salience, and lateral cortical network. We identify the intra- and inter-network connectivity differences and establish moderation models to reveal significant influences of age on acute alcohol-induced lateral cortical network connectivity. Through this work, we make brain-wide isotropic fMRI data with acute alcohol challenge publicly available, with the hope to facilitate future discovery of brain regions/circuits that are causally relevant to the impact of acute alcohol use.
Collapse
Affiliation(s)
- Sung-Ho Lee
- Center for Animal MRI, University of North Carolina, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Neurology, University of North Carolina, Chapel Hill, NC, USA
- Bowles Center for Alcohol Studies University of North Carolina, Chapel Hill, NC, USA
| | - Tatiana A. Shnitko
- Center for Animal MRI, University of North Carolina, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Neurology, University of North Carolina, Chapel Hill, NC, USA
| | - Li-Ming Hsu
- Center for Animal MRI, University of North Carolina, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Neurology, University of North Carolina, Chapel Hill, NC, USA
| | - Margaret A. Broadwater
- Center for Animal MRI, University of North Carolina, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Neurology, University of North Carolina, Chapel Hill, NC, USA
- Bowles Center for Alcohol Studies University of North Carolina, Chapel Hill, NC, USA
| | - Mabelle Sardinas
- Center for Animal MRI, University of North Carolina, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Neurology, University of North Carolina, Chapel Hill, NC, USA
| | - Tzu-Wen Winnie Wang
- Center for Animal MRI, University of North Carolina, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Neurology, University of North Carolina, Chapel Hill, NC, USA
| | - Donita L. Robinson
- Bowles Center for Alcohol Studies University of North Carolina, Chapel Hill, NC, USA
- Department of Psychiatry, University of North Carolina, Chapel Hill, NC, USA
| | - Ryan P. Vetreno
- Bowles Center for Alcohol Studies University of North Carolina, Chapel Hill, NC, USA
- Department of Psychiatry, University of North Carolina, Chapel Hill, NC, USA
| | - Fulton T. Crews
- Bowles Center for Alcohol Studies University of North Carolina, Chapel Hill, NC, USA
- Department of Psychiatry, University of North Carolina, Chapel Hill, NC, USA
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC, USA
| | - Yen-Yu Ian Shih
- Center for Animal MRI, University of North Carolina, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Neurology, University of North Carolina, Chapel Hill, NC, USA
- Bowles Center for Alcohol Studies University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
18
|
Moslemi M, Jannat B, Mahmoudzadeh M, Ghasemlou M, Abedi A. Detoxification activity of bioactive food compounds against ethanol-induced injuries and hangover symptoms: A review. Food Sci Nutr 2023; 11:5028-5040. [PMID: 37701198 PMCID: PMC10494618 DOI: 10.1002/fsn3.3520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 04/08/2023] [Accepted: 06/08/2023] [Indexed: 09/14/2023] Open
Abstract
Alcohol drinking is a popular activity among adolescents in many countries, largely due to its pleasant, relaxing effects. As a major concern, ethanol consumption put the drinkers at risk of nutrients' deficiency due to the disordered eating, anorexia, and malabsorption of nutrients. Moreover, alcohol drinking may lead to the development of hangover symptoms including diarrhea, thirsty, fatigue, and oxidative stress. A broad range of functional food components with antioxidant and/or anti-inflammatory properties including pectin, aloe vera polysaccharides, chito-oligosaccharides, and other herbal components have been explored due to their detoxification effects against ethanol. The underlying anti-hangover mechanisms include reducing the intestinal absorption of ethanol or its metabolites, increasing the activity of ethanol metabolizing enzymes, development of fatty acid β-oxidation in mitochondria, inhibition of inflammatory response, blocking the target receptors of ethanol in the body, and possession of antioxidant activity under the oxidative stress developed by ethanol consumption. Therefore, the development of bioactive food-based therapeutic formula can assist clinicians and also drinkers in the alleviation of alcohol side effects.
Collapse
Affiliation(s)
- Masoumeh Moslemi
- Halal Research Center of IRIMinistry of Health and Medical EducationTehranIran
| | - Behrooz Jannat
- Halal Research Center of IRIMinistry of Health and Medical EducationTehranIran
| | - Maryam Mahmoudzadeh
- Nutrition Research Center and Department of Food Science and Technology, Faculty of Nutrition and Food ScienceTabriz University of Medical SciencesTabrizIran
| | - Mehran Ghasemlou
- School of ScienceSTEM College, RMIT UniversityMelbourneVictoriaAustralia
| | - Abdol‐Samad Abedi
- Department of Research Deputy, National Nutrition and Food Technology Research Institute, Faculty of Nutrition Sciences and Food TechnologyShahid Beheshti University of Medical SciencesTehranIran
| |
Collapse
|
19
|
Cen Y, Lou G, Qi J, Zheng M, Liu Y. A new perspective on mesenchymal stem cell-based therapy for liver diseases: restoring mitochondrial function. Cell Commun Signal 2023; 21:214. [PMID: 37596671 PMCID: PMC10436412 DOI: 10.1186/s12964-023-01230-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 07/16/2023] [Indexed: 08/20/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have emerged as a promising alternative treatment for liver disease due to their roles in regeneration, fibrosis inhibition, and immunoregulation. Mitochondria are crucial in maintaining hepatocyte integrity and function. Mitochondrial dysfunction, such as impaired synthesis of adenosine triphosphate (ATP), decreased activity of respiratory chain complexes, and altered mitochondrial dynamics, is observed in most liver diseases. Accumulating evidence has substantiated that the therapeutic potential of MSCs is mediated not only through their cell replacement and paracrine effects but also through their regulation of mitochondrial dysfunction in liver disease. Here, we comprehensively review the involvement of mitochondrial dysfunction in the development of liver disease and how MSCs can target mitochondrial dysfunction. We also discuss recent advances in a novel method that modifies MSCs to enhance their functions in liver disease. A full understanding of MSC restoration of mitochondrial function and the underlying mechanisms will provide innovative strategies for clinical applications. Video Abstract.
Collapse
Affiliation(s)
- Yelei Cen
- The State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79# Qingchun Road, 6A-17, Hangzhou, 310003, China
| | - Guohua Lou
- The State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79# Qingchun Road, 6A-17, Hangzhou, 310003, China
| | - Jinjin Qi
- The State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79# Qingchun Road, 6A-17, Hangzhou, 310003, China
| | - Min Zheng
- The State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79# Qingchun Road, 6A-17, Hangzhou, 310003, China.
| | - Yanning Liu
- The State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79# Qingchun Road, 6A-17, Hangzhou, 310003, China.
| |
Collapse
|
20
|
López-Gil JF, Smith L, Gaya AR, Victoria-Montesinos D, Gutiérrez-Espinoza H, Herrera-Gutiérrez E, García-Hermoso A. The moderating role of recreational substance use in the association of Mediterranean diet with academic performance among adolescents. Sci Rep 2023; 13:10816. [PMID: 37402752 DOI: 10.1038/s41598-023-37529-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 06/22/2023] [Indexed: 07/06/2023] Open
Abstract
No study has examined the potential moderating role of recreational substance use in the relationship between the Mediterranean diet (MedDiet) and academic performance. The aim of this study was to test the potential moderating role of recreational substance use (i.e., alcohol, tobacco, and cannabis) in the association of adherence to the MedDiet with academic performance among adolescents. This cross-sectional study included a sample of 757 adolescents (55.6% girls) aged 12-17 years from the Valle de Ricote (Region of Murcia). The Region of Murcia is an autonomous community of Spain located in the southeast of the Iberian Peninsula, along the coast of the Mediterranean Sea. Adherence to the MedDiet was assessed by the Mediterranean Diet Quality Index for Children and Teenagers (KIDMED). Recreational substance use (i.e., tobacco, alcohol, cannabis) was self-reported by adolescents. Academic performance was assessed by the school records at the end of the academic year. The relationship between adherence to the MedDiet and academic performance was moderated by both tobacco and alcohol use (for grade point average and all school records). In conclusion, higher adherence to the MedDiet was related to greater academic performance in adolescents, but recreational substance use could moderate this association.
Collapse
Affiliation(s)
- José Francisco López-Gil
- Navarrabiomed, Hospital Universitario de Navarra, Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Navarra, Spain
- Department of Environmental Health, Harvard University T.H. Chan School of Public Health, Boston, MA, USA
- One Health Research Group, Universidad de Las Américas, Quito, Ecuador
| | - Lee Smith
- Centre for Health, Performance and Wellbeing, Anglia Ruskin University, Cambridge, UK
| | - Anelise Reis Gaya
- School of Physical Education, Physiotherapy and Dance, Post-Graduate Program in Human Movement Sciences, Federal University of Rio Grande Do Sul, Porto Alegre, Brazil
| | | | | | - Eva Herrera-Gutiérrez
- Department of Developmental and Educational Psychology, Faculty of Psychology, Espinardo Campus, University of Murcia, Murcia, Spain
| | - Antonio García-Hermoso
- Navarrabiomed, Hospital Universitario de Navarra, Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Navarra, Spain
| |
Collapse
|
21
|
Kim M, Oh S, Kim S, Ji M, Choi B, Bae JW, Lee YS, Paik MJ, Lee S. Alcohol perturbed locomotor behavior, metabolism, and pharmacokinetics of gamma-hydroxybutyric acid in rats. Biomed Pharmacother 2023; 164:114992. [PMID: 37301134 DOI: 10.1016/j.biopha.2023.114992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 05/30/2023] [Accepted: 06/06/2023] [Indexed: 06/12/2023] Open
Abstract
Gamma-hydroxybutyric acid (GHB), both a metabolic precursor and product of gamma-aminobutyric acid (GABA), is a central nervous system depressant used for the treatment of narcolepsy-associated cataplexy and alcohol withdrawal. However, administration of GHB with alcohol (ethanol) is a major cause of hospitalizations related to GHB intoxication. In this study, we investigated locomotor behavior as well as metabolic and pharmacokinetic interactions following co-administration of GHB and ethanol in rats. The locomotor behavior of rats was evaluated following the intraperitoneal administration of GHB (sodium salt, 500 mg/kg) and/or ethanol (2 g/kg). Further, time-course urinary metabolic profiling of GHB and its biomarker metabolites glutamic acid, GABA, succinic acid, 2,4-dihydroxybutyric acid (OH-BA), 3,4-OH-BA, and glycolic acid as well as pharmacokinetic analysis were performed. GHB/ethanol co-administration significantly reduced locomotor activity, compared to the individual administration of GHB or ethanol. The urinary and plasma concentrations of GHB and other target compounds, except for 2,4-OH-BA, were significantly higher in the GHB/ethanol co-administration group than the group administered only GHB. The pharmacokinetic analysis results showed that the co-administration of GHB and ethanol significantly increased the half-life of GHB while the total clearance decreased. Moreover, a comparison of the metabolite-to-parent drug area under the curve ratios demonstrated that the metabolic pathways of GHB, such α- and β-oxidation, were inhibited by ethanol. Consequently, the co-administration of GHB and ethanol aggravated the metabolism and elimination of GHB and enhanced its sedative effect. These findings will contribute to clinical interpretation of GHB intoxication.
Collapse
Affiliation(s)
- Mingyu Kim
- College of Pharmacy, Keimyung University, 1095 Dalgubeol-daero, Dalseo-gu, Daegu 42601, the Republic of Korea
| | - Songjin Oh
- College of Pharmacy, Sunchon National University, 25 Jungang-ro, Suncheon 57922, the Republic of Korea
| | - Suji Kim
- College of Pharmacy, Keimyung University, 1095 Dalgubeol-daero, Dalseo-gu, Daegu 42601, the Republic of Korea
| | - Moongi Ji
- College of Pharmacy, Sunchon National University, 25 Jungang-ro, Suncheon 57922, the Republic of Korea
| | - Byeongchan Choi
- College of Pharmacy, Sunchon National University, 25 Jungang-ro, Suncheon 57922, the Republic of Korea
| | - Jung-Woo Bae
- College of Pharmacy, Keimyung University, 1095 Dalgubeol-daero, Dalseo-gu, Daegu 42601, the Republic of Korea
| | - Yong Sup Lee
- College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, the Republic of Korea
| | - Man-Jeong Paik
- College of Pharmacy, Sunchon National University, 25 Jungang-ro, Suncheon 57922, the Republic of Korea.
| | - Sooyeun Lee
- College of Pharmacy, Keimyung University, 1095 Dalgubeol-daero, Dalseo-gu, Daegu 42601, the Republic of Korea.
| |
Collapse
|
22
|
Müller SG, Jardim NS, Zeni G, Nogueira CW. (m-CF 3-PhSe) 2 counteracts metabolic disturbances and hypothalamic inflammation in a lifestyle rodent model. Food Chem Toxicol 2023; 176:113750. [PMID: 37023972 DOI: 10.1016/j.fct.2023.113750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 03/01/2023] [Accepted: 03/26/2023] [Indexed: 04/08/2023]
Abstract
An unhealthy lifestyle is associated with metabolic disorders and neuroinflammation. In this study, the efficacy of m-trifluoromethyl-diphenyl diselenide [(m-CF3-PhSe)2] against lifestyle model-related metabolic disturbances and hypothalamic inflammation in young mice was investigated. From postnatal day 25 (PND25) to 66, male Swiss mice were subjected to a lifestyle model, an energy-dense diet (20:20% lard: corn syrup) and sporadic ethanol (3x/week). Ethanol was administrated intragastrically (i.g., 2g/kg) to mice from PND45 to 60. From PND60 to 66, mice received (m-CF3-PhSe)2 (5mg/kg/day; i.g). (m-CF3-PhSe)2 reduced relative abdominal adipose tissue weight, hyperglycemia, and dyslipidemia in mice exposed to the lifestyle-induced model. (m-CF3-PhSe)2 normalized hepatic cholesterol and triglyceride levels, and the activity of G-6-Pase increased in lifestyle-exposed mice. (m-CF3-PhSe)2 was effective in modulating hepatic glycogen levels, citrate synthase and hexokinase activities, protein levels of GLUT-2, p-IRS/IRS, p-AKT/AKT, redox homeostasis, and inflammatory profile of mice exposed to a lifestyle model. (m-CF3-PhSe)2 counteracted hypothalamic inflammation and the ghrelin receptor levels in mice exposed to the lifestyle model. (m-CF3-PhSe)2 reversed the decreased levels of GLUT-3, p-IRS/IRS, and the leptin receptor in the hypothalamus of lifestyle-exposed mice. In conclusion, (m-CF3-PhSe)2 counteracted metabolic disturbances and hypothalamic inflammation in young mice exposed to a lifestyle model.
Collapse
Affiliation(s)
- Sabrina G Müller
- Laboratório de Síntese, Reatividade e Avaliação Farmacológica e Toxicológica de Organocalcogênios, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Santa Maria, Santa Maria, CEP 97105-900, RS, Brazil
| | - Natália S Jardim
- Laboratório de Síntese, Reatividade e Avaliação Farmacológica e Toxicológica de Organocalcogênios, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Santa Maria, Santa Maria, CEP 97105-900, RS, Brazil
| | - Gilson Zeni
- Laboratório de Síntese, Reatividade e Avaliação Farmacológica e Toxicológica de Organocalcogênios, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Santa Maria, Santa Maria, CEP 97105-900, RS, Brazil
| | - Cristina W Nogueira
- Laboratório de Síntese, Reatividade e Avaliação Farmacológica e Toxicológica de Organocalcogênios, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Santa Maria, Santa Maria, CEP 97105-900, RS, Brazil.
| |
Collapse
|
23
|
Lai S, Ma Y, Hao L, Ding Q, Chang K, Zhuge H, Qiu J, Xu T, Dou X, Li S. 1-Methylnicotinamide promotes hepatic steatosis in mice: A potential mechanism in chronic alcohol-induced fatty liver disease. Biochim Biophys Acta Mol Cell Biol Lipids 2023; 1868:159286. [PMID: 36690322 DOI: 10.1016/j.bbalip.2023.159286] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 12/16/2022] [Accepted: 01/16/2023] [Indexed: 01/22/2023]
Abstract
Alcohol abuse and its related diseases are the major risk factors for human health. Alcohol-related liver disease (ALD) is a leading cause of morbidity and mortality worldwide. Although the mechanism of ALD has been widely investigated, liver metabolites associated with long-term alcohol intake-induced hepatic steatosis have not been well explored. In this study, we aimed to investigate the role and mechanisms of 1-methylnicotinamide (1-MNA), a metabolite during nicotinamide adenine dinucleotide (NAD+) metabolism, in the pathogenesis of ALD. C57BL/6 wild-type mice were subjected to chronic alcohol feeding with or without 1-MNA (50 mg/kg/day). Our data showed that 1-MNA administration significantly enhanced chronic alcohol consumption-induced hepatic steatosis. Mechanistic studies revealed that alcohol-increased hepatic protein levels of sterol regulatory element-binding transcription factor (SREBP-1c), a key enzyme that regulates lipid lipogenesis, were enhanced in mice administered with 1-MNA, regardless of alcohol feeding. Consistently, alcohol-increased mRNA and protein levels of hepatic diacylglycerol o-acyltransferase 2 (DGAT2) and very low-density lipoprotein receptor (VLDLR) were also exacerbated by 1-MNA administration. Alcohol-induced hepatic endoplasmic reticulum (ER) stress was enhanced by 1-MNA administration, which was evidenced by increased protein levels of binding immunoglobulin protein (BIP), phosphorylated- protein kinase r-like ER kinase (PERK), activating transcription factor 4 (ATF4), and C/EBP-homologous protein (CHOP) in the mouse liver. Overall, this study demonstrated that 1-MNA serves as a pathogenic factor in the development of ALD. Targeting liver 1-MNA levels may serve as a promising therapeutic approach for improving hepatic steatosis in ALD.
Collapse
Affiliation(s)
- Shanglei Lai
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China; School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Yue Ma
- Zhejiang Provincial Key Laboratory of Laboratory Animals and Safety Research, Hangzhou Medical College, Hangzhou, PR China
| | - Liuyi Hao
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Qinchao Ding
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China; Institute of Dairy Science, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Kaixin Chang
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Hui Zhuge
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Jiannan Qiu
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Tiantian Xu
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Xiaobing Dou
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China.
| | - Songtao Li
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China; Department of Clinical Nutrition, Affiliated Zhejiang Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, PR China.
| |
Collapse
|
24
|
Tarragon E. Alcohol and energy drinks: individual contribution of common ingredients on ethanol-induced behaviour. Front Behav Neurosci 2023; 17:1057262. [PMID: 36865774 PMCID: PMC9971501 DOI: 10.3389/fnbeh.2023.1057262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 01/23/2023] [Indexed: 02/16/2023] Open
Abstract
Since energy drinks (EDs) were sold to the general public as soft drinks and recreational beverages, mixing EDs with ethanol has grown in popularity, particularly among younger people. Given the research that links these drinks with higher risk behaviors and increased ethanol intake, ethanol combined with EDs (AmEDs) is a particularly worrying combination. EDs generally commonly include a variety of ingredients. Sugar, caffeine, taurine, and B-group vitamins are almost always present. Studies on the combined effect of ethanol and sugar and caffeine on ethanol-induced behaviors are extensive. Not so much in regards to taurine and vitamins. This review briefly summarises available information from research on the isolated compounds on EtOH-induced behaviors first, and secondly, the combination of AmEDs on EtOH effects. The conclusion is that additional research is needed to fully comprehend the characteristics and consequences of AmEDs on EtOH-induced behaviors.
Collapse
|
25
|
Wu X, Fan X, Miyata T, Kim A, Cajigas-Du Ross CK, Ray S, Huang E, Taiwo M, Arya R, Wu J, Nagy LE. Recent Advances in Understanding of Pathogenesis of Alcohol-Associated Liver Disease. ANNUAL REVIEW OF PATHOLOGY 2023; 18:411-438. [PMID: 36270295 PMCID: PMC10060166 DOI: 10.1146/annurev-pathmechdis-031521-030435] [Citation(s) in RCA: 41] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Alcohol-associated liver disease (ALD) is one of the major diseases arising from chronic alcohol consumption and is one of the most common causes of liver-related morbidity and mortality. ALD includes asymptomatic liver steatosis, fibrosis, cirrhosis, and alcohol-associated hepatitis and its complications. The progression of ALD involves complex cell-cell and organ-organ interactions. We focus on the impact of alcohol on dysregulation of homeostatic mechanisms and regulation of injury and repair in the liver. In particular, we discuss recent advances in understanding the disruption of balance between programmed cell death and prosurvival pathways, such as autophagy and membrane trafficking, in the pathogenesis of ALD. We also summarize current understanding of innate immune responses, liver sinusoidal endothelial cell dysfunction and hepatic stellate cell activation, and gut-liver and adipose-liver cross talk in response to ethanol. In addition,we describe the current potential therapeutic targets and clinical trials aimed at alleviating hepatocyte injury, reducing inflammatory responses, and targeting gut microbiota, for the treatment of ALD.
Collapse
Affiliation(s)
- Xiaoqin Wu
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA;
| | - Xiude Fan
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA;
| | - Tatsunori Miyata
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA;
| | - Adam Kim
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA;
| | - Christina K Cajigas-Du Ross
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA;
| | - Semanti Ray
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA;
| | - Emily Huang
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA;
| | - Moyinoluwa Taiwo
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA;
| | - Rakesh Arya
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA;
| | - Jianguo Wu
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA;
- Department of Molecular Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Laura E Nagy
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA;
- Department of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Molecular Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
26
|
Di Tommaso N, Santopaolo F, Gasbarrini A, Ponziani FR. The Gut-Vascular Barrier as a New Protagonist in Intestinal and Extraintestinal Diseases. Int J Mol Sci 2023; 24:ijms24021470. [PMID: 36674986 PMCID: PMC9864173 DOI: 10.3390/ijms24021470] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/07/2023] [Accepted: 01/09/2023] [Indexed: 01/15/2023] Open
Abstract
The intestinal barrier, with its multiple layers, is the first line of defense between the outside world and the intestine. Its disruption, resulting in increased intestinal permeability, is a recognized pathogenic factor of intestinal and extra-intestinal diseases. The identification of a gut-vascular barrier (GVB), consisting of a structured endothelium below the epithelial layer, has led to new evidence on the etiology and management of diseases of the gut-liver axis and the gut-brain axis, with recent implications in oncology as well. The gut-brain axis is involved in several neuroinflammatory processes. In particular, the recent description of a choroid plexus vascular barrier regulating brain permeability under conditions of gut inflammation identifies the endothelium as a key regulator in maintaining tissue homeostasis and health.
Collapse
Affiliation(s)
- Natalia Di Tommaso
- Internal Medicine and Gastroenterology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Francesco Santopaolo
- Internal Medicine and Gastroenterology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Antonio Gasbarrini
- Internal Medicine and Gastroenterology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Translational Medicine and Surgery Department, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Francesca Romana Ponziani
- Internal Medicine and Gastroenterology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Translational Medicine and Surgery Department, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Correspondence:
| |
Collapse
|
27
|
Krum-Hansen S, Standahl Olsen K, Anderssen E, Frantzen JO, Lund E, Paulssen RH. Associations of breast cancer related exposures and gene expression profiles in normal breast tissue-The Norwegian Women and Cancer normal breast tissue study. Cancer Rep (Hoboken) 2023; 6:e1777. [PMID: 36617746 PMCID: PMC10075301 DOI: 10.1002/cnr2.1777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 11/11/2022] [Accepted: 12/12/2022] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Normal breast tissue is utilized in tissue-based studies of breast carcinogenesis. While gene expression in breast tumor tissue is well explored, our knowledge of transcriptomic signatures in normal breast tissue is still incomplete. The aim of this study was to investigate variability of gene expression in a large sample of normal breast tissue biopsies, according to breast cancer related exposures (obesity, smoking, alcohol, hormone therapy, and parity). METHODS We analyzed gene expression profiles from 311 normal breast tissue biopsies from cancer-free, post-menopausal women, using Illumina bead chip arrays. Principal component analysis and K-means clustering was used for initial analysis of the dataset. The association of exposures and covariates with gene expression was determined using linear models for microarrays. RESULTS Heterogeneity of the breast tissue and cell composition had the strongest influence on gene expression profiles. After adjusting for cell composition, obesity, smoking, and alcohol showed the highest numbers of associated genes and pathways, whereas hormone therapy and parity were associated with negligible gene expression differences. CONCLUSION Our results provide insight into associations between major exposures and gene expression profiles and provide an informative baseline for improved understanding of exposure-related molecular events in normal breast tissue of cancer-free, post-menopausal women.
Collapse
Affiliation(s)
- Sanda Krum-Hansen
- Department of Community Medicine, UiT The Arctic University of Norway, Tromsø, Norway.,Department of Hematology and Oncology, Stavanger University Hospital, Stavanger, Norway
| | - Karina Standahl Olsen
- Department of Community Medicine, UiT The Arctic University of Norway, Tromsø, Norway
| | - Endre Anderssen
- Genomics Support Center Tromsø (GSCT), UiT The Arctic University of Norway, Tromsø, Norway
| | - Jan Ole Frantzen
- Narvik Hospital, University Hospital of North Norway, Narvik, Norway
| | - Eiliv Lund
- Department of Community Medicine, UiT The Arctic University of Norway, Tromsø, Norway
| | - Ruth H Paulssen
- Genomics Support Center Tromsø (GSCT), UiT The Arctic University of Norway, Tromsø, Norway.,Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
28
|
Koning M, Herrema H, Nieuwdorp M, Meijnikman AS. Targeting nonalcoholic fatty liver disease via gut microbiome-centered therapies. Gut Microbes 2023; 15:2226922. [PMID: 37610978 PMCID: PMC10305510 DOI: 10.1080/19490976.2023.2226922] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 08/25/2023] Open
Abstract
Humans possess abundant amounts of microorganisms, including bacteria, fungi, viruses, and archaea, in their gut. Patients with nonalcoholic fatty liver disease (NAFLD) exhibit alterations in their gut microbiome and an impaired gut barrier function. Preclinical studies emphasize the significance of the gut microbiome in the pathogenesis of NAFLD. In this overview, we explore how adjusting the gut microbiome could serve as an innovative therapeutic strategy for NAFLD. We provide a summary of current information on untargeted techniques such as probiotics and fecal microbiota transplantation, as well as targeted microbiome-focused therapies including engineered bacteria, prebiotics, postbiotics, and phages for the treatment of NAFLD.
Collapse
Affiliation(s)
- Mijra Koning
- Departments of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences Diabetes, Amsterdam, The Netherlands
| | - Hilde Herrema
- Departments of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
- Amsterdam Gastroenterology and Metabolism, Amsterdam, The Netherlands
| | - Max Nieuwdorp
- Departments of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences Diabetes, Amsterdam, The Netherlands
| | - Abraham S. Meijnikman
- Departments of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences Diabetes, Amsterdam, The Netherlands
| |
Collapse
|
29
|
The Status of Oxidative Stress in Patients with Alcohol Dependence: A Meta-Analysis. Antioxidants (Basel) 2022; 11:antiox11101919. [PMID: 36290642 PMCID: PMC9598131 DOI: 10.3390/antiox11101919] [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: 08/29/2022] [Revised: 09/18/2022] [Accepted: 09/25/2022] [Indexed: 11/19/2022] Open
Abstract
Alcohol-induced oxidative stress (OS) plays a pivotal role in the pathophysiology of alcohol dependence (AD). This meta-analysis was aimed at investigating the changes in the levels of OS biomarkers in AD patients. We included relevant literature published before 1 April 2022, from the PubMed, Web of Science, and EBSCO databases following PRISMA guidelines. Finally, 15 eligible articles were enrolled in this meta-analysis, including 860 patients and 849 controls. Compared with healthy controls, AD patients had lower activities of superoxide dismutase (SOD) and glutathione peroxidase (GPx) enzymes, and lower levels of albumin, while levels of malondialdehyde (MDA), vitamin B12, homocysteine, and bilirubin were significantly increased in serum/plasma samples of AD subjects (all p < 0.05). In male patients, the activities of SOD and GPx were increased in serum/plasma but decreased in erythrocytes (all p < 0.05). The opposite trends in the level of SOD and GPx activities in serum/plasma and erythrocytes of male patients could be used as the biomarker of alcohol-induced OS injury, and the synergistic changes of MDA, vitamin B12, albumin, bilirubin, and homocysteine levels should also be considered.
Collapse
|
30
|
Bashir K, Todaka D, Rasheed S, Matsui A, Ahmad Z, Sako K, Utsumi Y, Vu AT, Tanaka M, Takahashi S, Ishida J, Tsuboi Y, Watanabe S, Kanno Y, Ando E, Shin KC, Seito M, Motegi H, Sato M, Li R, Kikuchi S, Fujita M, Kusano M, Kobayashi M, Habu Y, Nagano AJ, Kawaura K, Kikuchi J, Saito K, Hirai MY, Seo M, Shinozaki K, Kinoshita T, Seki M. Ethanol-Mediated Novel Survival Strategy against Drought Stress in Plants. PLANT & CELL PHYSIOLOGY 2022; 63:1181-1192. [PMID: 36003026 PMCID: PMC9474946 DOI: 10.1093/pcp/pcac114] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 07/22/2022] [Accepted: 08/05/2022] [Indexed: 05/08/2023]
Abstract
Water scarcity is a serious agricultural problem causing significant losses to crop yield and product quality. The development of technologies to mitigate the damage caused by drought stress is essential for ensuring a sustainable food supply for the increasing global population. We herein report that the exogenous application of ethanol, an inexpensive and environmentally friendly chemical, significantly enhances drought tolerance in Arabidopsis thaliana, rice and wheat. The transcriptomic analyses of ethanol-treated plants revealed the upregulation of genes related to sucrose and starch metabolism, phenylpropanoids and glucosinolate biosynthesis, while metabolomic analysis showed an increased accumulation of sugars, glucosinolates and drought-tolerance-related amino acids. The phenotyping analysis indicated that drought-induced water loss was delayed in the ethanol-treated plants. Furthermore, ethanol treatment induced stomatal closure, resulting in decreased transpiration rate and increased leaf water contents under drought stress conditions. The ethanol treatment did not enhance drought tolerance in the mutant of ABI1, a negative regulator of abscisic acid (ABA) signaling in Arabidopsis, indicating that ABA signaling contributes to ethanol-mediated drought tolerance. The nuclear magnetic resonance analysis using 13C-labeled ethanol indicated that gluconeogenesis is involved in the accumulation of sugars. The ethanol treatment did not enhance the drought tolerance in the aldehyde dehydrogenase (aldh) triple mutant (aldh2b4/aldh2b7/aldh2c4). These results show that ABA signaling and acetic acid biosynthesis are involved in ethanol-mediated drought tolerance and that chemical priming through ethanol application regulates sugar accumulation and gluconeogenesis, leading to enhanced drought tolerance and sustained plant growth. These findings highlight a new survival strategy for increasing crop production under water-limited conditions.
Collapse
Affiliation(s)
- Khurram Bashir
- Plant Genomic Network Research Team, RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045 Japan
- Department of Life Sciences, SBA School of Science and Engineering, Lahore University of Management Sciences, DHA Phase 5, Lahore 54792, Pakistan
| | - Daisuke Todaka
- Plant Genomic Network Research Team, RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045 Japan
| | - Sultana Rasheed
- Plant Genomic Network Research Team, RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045 Japan
| | - Akihiro Matsui
- Plant Genomic Network Research Team, RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045 Japan
- Plant Epigenome Regulation Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama, 351-0198 Japan
| | - Zarnab Ahmad
- Plant Genomic Network Research Team, RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045 Japan
- Department of Life Sciences, SBA School of Science and Engineering, Lahore University of Management Sciences, DHA Phase 5, Lahore 54792, Pakistan
| | - Kaori Sako
- Plant Genomic Network Research Team, RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045 Japan
- Department of Advanced Bioscience, Faculty of Agriculture, Kindai University, 3327-204 Nakamachi, Nara, 631-8505, Japan
| | - Yoshinori Utsumi
- Plant Genomic Network Research Team, RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045 Japan
| | - Anh Thu Vu
- Plant Genomic Network Research Team, RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045 Japan
| | - Maho Tanaka
- Plant Genomic Network Research Team, RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045 Japan
- Plant Epigenome Regulation Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama, 351-0198 Japan
| | - Satoshi Takahashi
- Plant Genomic Network Research Team, RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045 Japan
- Plant Epigenome Regulation Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama, 351-0198 Japan
| | - Junko Ishida
- Plant Genomic Network Research Team, RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045 Japan
- Plant Epigenome Regulation Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama, 351-0198 Japan
| | - Yuuri Tsuboi
- Environmental Metabolic Analysis Research Team, RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045 Japan
| | - Shunsuke Watanabe
- Dormancy and Adaptation Research Unit, RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
- IPSiM, University of Montpellier, CNRS, INRAE, Institut Agro, Montpellier 34060, France
| | - Yuri Kanno
- Dormancy and Adaptation Research Unit, RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
| | - Eigo Ando
- Division of Biological Sciences, Graduate School of Science, Nagoya University, Chikusa, Nagoya, 464-8602 Japan
- Department of Biological Sciences, School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Kwang-Chul Shin
- Division of Biological Sciences, Graduate School of Science, Nagoya University, Chikusa, Nagoya, 464-8602 Japan
| | - Makoto Seito
- Kihara Institute for Biological Research, Yokohama City University, 641-12 Maiokacho, Totsuka Ward, Yokohama, Kanagawa, 244-0813 Japan
| | - Hinata Motegi
- Plant Genomic Network Research Team, RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045 Japan
- Kihara Institute for Biological Research, Yokohama City University, 641-12 Maiokacho, Totsuka Ward, Yokohama, Kanagawa, 244-0813 Japan
| | - Muneo Sato
- Mass Spectrometry and Microscopy Unit, RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045 Japan
- Metabolic Systems Research Team, RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045 Japan
| | - Rui Li
- Metabolic Systems Research Team, RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045 Japan
| | - Saya Kikuchi
- Gene Discovery Research Group, RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045 Japan
| | - Miki Fujita
- Mass Spectrometry and Microscopy Unit, RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045 Japan
- Gene Discovery Research Group, RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045 Japan
| | - Miyako Kusano
- Metabolomics Research Group, RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045 Japan
- Graduate School of Life and Environmental Science, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8572 Japan
| | - Makoto Kobayashi
- Metabolomics Research Group, RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045 Japan
| | - Yoshiki Habu
- Graduate School of Life and Environmental Science, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8572 Japan
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, 2-1-2 Kannondai, Tsukuba, Ibaraki, 305-8602 Japan
| | - Atsushi J Nagano
- Faculty of Agriculture, Ryukoku University, Yokotani 1-5, Seta Oe-cho, Otsu, Shiga, 520-2914, Japan
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, 997-0017 Japan
| | - Kanako Kawaura
- Kihara Institute for Biological Research, Yokohama City University, 641-12 Maiokacho, Totsuka Ward, Yokohama, Kanagawa, 244-0813 Japan
| | - Jun Kikuchi
- Environmental Metabolic Analysis Research Team, RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045 Japan
- Graduate School of Medical Life Science, Yokohama City University, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045 Japan
- Department of Applied Biosciences, Graduate School of Bioagricultural Sciences, Nagoya University, Chikusa, Nagoya, Aichi, 464-8601 Japan
| | - Kazuki Saito
- Metabolomics Research Group, RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045 Japan
| | - Masami Yokota Hirai
- Mass Spectrometry and Microscopy Unit, RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045 Japan
- Department of Applied Biosciences, Graduate School of Bioagricultural Sciences, Nagoya University, Chikusa, Nagoya, Aichi, 464-8601 Japan
- Metabolic Systems Research Team, RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045 Japan
| | - Mitsunori Seo
- Dormancy and Adaptation Research Unit, RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
| | - Kazuo Shinozaki
- Gene Discovery Research Group, RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045 Japan
| | - Toshinori Kinoshita
- Division of Biological Sciences, Graduate School of Science, Nagoya University, Chikusa, Nagoya, 464-8602 Japan
- Institute of Transformative Bio-Molecules (WPI-ITbM), Nagoya University, Chikusa, Nagoya, Aichi, 464-8601 Japan
| | | |
Collapse
|
31
|
Martín-Estal I, Fajardo-Ramírez ÓR, Bermúdez de León M, Zertuche-Mery C, Benavides-Guajardo R, García-Cruz MI, Rodríguez De Ita J, Castilla-Cortázar I, Castorena-Torres F. Effect of Ethanol Consumption on the Placenta and Liver of Partially IGF-1-Deficient Mice: The Role of Metabolism via CYP2E1 and the Antioxidant Enzyme System. BIOLOGY 2022; 11:biology11091264. [PMID: 36138743 PMCID: PMC9495332 DOI: 10.3390/biology11091264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/16/2022] [Accepted: 08/23/2022] [Indexed: 11/17/2022]
Abstract
Simple Summary Ethanol is the most consumed drug worldwide, even during pregnancy. One of its adverse outcomes is fetal growth restriction, an alteration in development due to decreased IGF-1 levels. Several studies have shown that ethanol can impair the IGF-1 signaling pathway, thus exacerbating IGF-1 adverse effects in both intrauterine and postnatal growth and development. In this manuscript, we used a partially IGF-1-deficient mouse model to demonstrate the key role of IGF-1 in fetal development, as well as ethanol’s adverse effects on CYP2E1 expression levels and the antioxidant enzyme system during pregnancy. Abstract Ethanol use during pregnancy is a risk factor for developing adverse outcomes. Its metabolism by cytochrome P450 2E1 (CYP2E1) produces radical oxygen species (ROS), promoting cellular injury and apoptosis. To date, no studies have been conducted to elucidate the teratogenic effects due to both IGF-1 deficiency and ethanol consumption in mice placentas. The aim of this study is to determine the effect of ethanol consumption on the placenta and liver of partially IGF-1-deficient mice, the role of metabolism via CYP2E1, and the antioxidant enzyme system. Heterozygous (HZ, Igf1+/−) pregnant female mice were given water or 10% ethanol. Wild-type (WT, Igf1+/+) female mice were used as controls. At gestational day 19, pregnant dams were euthanized, and maternal liver and placentas were collected. Pregnant HZ dams were smaller than controls, and this effect was higher due to ethanol consumption. Cyp2e1 gene was overexpressed in the liver of HZ pregnant dams exposed to ethanol; at the protein level, CYP2E1 is reduced in placentas from all genotypes. The antioxidant enzymatic system was altered by ethanol consumption in both the maternal liver and placenta. The results in this work hint that IGF-1 is involved in intrauterine development because its deficiency exacerbates ethanol’s effects on both metabolism and the placenta.
Collapse
Affiliation(s)
- Irene Martín-Estal
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, Monterrey 64710, N.L., Mexico
| | - Óscar R. Fajardo-Ramírez
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, Monterrey 64710, N.L., Mexico
| | - Mario Bermúdez de León
- Departamento de Biología Molecular, Centro de Investigación Biomédica del Noreste, Instituto Mexicano del Seguro Social, Monterrey 64720, N.L., Mexico
| | - Carolina Zertuche-Mery
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, Monterrey 64710, N.L., Mexico
| | - Rodolfo Benavides-Guajardo
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, Monterrey 64710, N.L., Mexico
| | - María Isabel García-Cruz
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, Monterrey 64710, N.L., Mexico
| | - Julieta Rodríguez De Ita
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, Monterrey 64710, N.L., Mexico
| | - Inma Castilla-Cortázar
- Fundación de Investigación HM Hospitales, 28015 Madrid, Spain
- Correspondence: (I.C.-C.); (F.C.-T.)
| | - Fabiola Castorena-Torres
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, Monterrey 64710, N.L., Mexico
- Correspondence: (I.C.-C.); (F.C.-T.)
| |
Collapse
|
32
|
Simon L, Molina PE. Cellular Bioenergetics: Experimental Evidence for Alcohol-induced Adaptations. FUNCTION 2022; 3:zqac039. [PMID: 36120487 PMCID: PMC9469757 DOI: 10.1093/function/zqac039] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 08/08/2022] [Accepted: 08/10/2022] [Indexed: 01/07/2023] Open
Abstract
At-risk alcohol use is associated with multisystemic effects and end-organ injury, and significantly contributes to global health burden. Several alcohol-mediated mechanisms have been identified, with bioenergetic maladaptation gaining credence as an underlying pathophysiological mechanism contributing to cellular injury. This evidence-based review focuses on the current knowledge of alcohol-induced bioenergetic adaptations in metabolically active tissues: liver, cardiac and skeletal muscle, pancreas, and brain. Alcohol metabolism itself significantly interferes with bioenergetic pathways in tissues, particularly the liver. Alcohol decreases states of respiration in the electron transport chain, and activity and expression of respiratory complexes, with a net effect to decrease ATP content. In addition, alcohol dysregulates major metabolic pathways, including glycolysis, the tricarboxylic acid cycle, and fatty acid oxidation. These bioenergetic alterations are influenced by alcohol-mediated changes in mitochondrial morphology, biogenesis, and dynamics. The review highlights similarities and differences in bioenergetic adaptations according to tissue type, pattern of (acute vs. chronic) alcohol use, and energy substrate availability. The compromised bioenergetics synergizes with other critical pathophysiological mechanisms, including increased oxidative stress and accelerates cellular dysfunction, promoting senescence, programmed cell death, and end-organ injury.
Collapse
Affiliation(s)
- Liz Simon
- Department of Physiology and Comprehensive Alcohol-HIV/AIDS Research Center, Louisiana State University Health Sciences Center, 1901 Perdido Street, New Orleans, LA 70112, USA
| | - Patricia E Molina
- Department of Physiology and Comprehensive Alcohol-HIV/AIDS Research Center, Louisiana State University Health Sciences Center, 1901 Perdido Street, New Orleans, LA 70112, USA
| |
Collapse
|
33
|
Ojeda ML, Nogales F, Del Carmen Gallego-López M, Carreras O. Binge drinking during the adolescence period causes oxidative damage-induced cardiometabolic disorders: A possible ameliorative approach with selenium supplementation. Life Sci 2022; 301:120618. [PMID: 35533761 DOI: 10.1016/j.lfs.2022.120618] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 05/03/2022] [Accepted: 05/04/2022] [Indexed: 12/15/2022]
Abstract
Binge drinking (BD) is the most common alcohol consumption model among adolescents. BD exposure during adolescence disrupts the nervous system function, being involved in the major mortality causes at this age: motor vehicle accidents, homicides and suicides. Recent studies have also shown that BD consumption during adolescence affects liver, renal and cardiovascular physiology, predisposing adolescents to future adult cardiometabolic damage. BD is a particularly pro-oxidant alcohol consumption pattern, because it leads to the production of a great source of reactive oxygen species (ROS) via the microsomal ethanol oxidizing system, also decreasing the antioxidant activity of glutathione peroxidase (GPx). Selenium (Se) is a mineral which plays a pivotal role against oxidation; it forms part of the catalytic center of different antioxidant selenoproteins such as GPxs (GPx1, GPx4, GPx3) and selenoprotein P (SelP). Specifically, GPx4 has an essential role in mitochondria, preventing their oxidation, apoptosis and NFkB-inflamative response, being this function even more relevant in heart's tissue. Se serum levels are decreased in acute and chronic alcoholic adult patients, being correlated to the severity of oxidation, liver damage and metabolic profile. Experimental studies have described that Se supplementation to alcohol exposed mice clearly decreases oxidative and liver damage. However, clinical BD effects on Se homeostasis and selenoproteins' tissue distribution related to oxidation during adolescence are not yet studied. In this narrative review we will describe the use of sodium selenite supplementation as an antioxidant therapy in adolescent BD rats in order to analyze Se homeostasis implication during BD exposure, oxidative balance, apoptosis and inflammation, mainly in liver, kidney, and heart. These biomolecular changes and the cardiovascular function will be analyzed. Se supplementation therapies could be a good strategy to prevent the oxidation, inflammation and apoptosis generated in tissues by BD during adolescence, such as liver, kidney and heart, improving cardiovascular functioning.
Collapse
Affiliation(s)
- María Luisa Ojeda
- Department of Physiology, Faculty of Pharmacy, Seville University, 41012 Seville, Spain
| | - Fátima Nogales
- Department of Physiology, Faculty of Pharmacy, Seville University, 41012 Seville, Spain.
| | | | - Olimpia Carreras
- Department of Physiology, Faculty of Pharmacy, Seville University, 41012 Seville, Spain
| |
Collapse
|
34
|
McGinnis CD, Jennings EQ, Harris PS, Galligan JJ, Fritz KS. Biochemical Mechanisms of Sirtuin-Directed Protein Acylation in Hepatic Pathologies of Mitochondrial Dysfunction. Cells 2022; 11:cells11132045. [PMID: 35805129 PMCID: PMC9266223 DOI: 10.3390/cells11132045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/06/2022] [Accepted: 06/10/2022] [Indexed: 12/12/2022] Open
Abstract
Mitochondrial protein acetylation is associated with a host of diseases including cancer, Alzheimer’s, and metabolic syndrome. Deciphering the mechanisms regarding how protein acetylation contributes to disease pathologies remains difficult due to the complex diversity of pathways targeted by lysine acetylation. Specifically, protein acetylation is thought to direct feedback from metabolism, whereby nutritional status influences mitochondrial pathways including beta-oxidation, the citric acid cycle, and the electron transport chain. Acetylation provides a crucial connection between hepatic metabolism and mitochondrial function. Dysregulation of protein acetylation throughout the cell can alter mitochondrial function and is associated with numerous liver diseases, including non-alcoholic and alcoholic fatty liver disease, steatohepatitis, and hepatocellular carcinoma. This review introduces biochemical mechanisms of protein acetylation in the regulation of mitochondrial function and hepatic diseases and offers a viewpoint on the potential for targeted therapies.
Collapse
Affiliation(s)
- Courtney D. McGinnis
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (C.D.M.); (P.S.H.)
| | - Erin Q. Jennings
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA; (E.Q.J.); (J.J.G.)
| | - Peter S. Harris
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (C.D.M.); (P.S.H.)
| | - James J. Galligan
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA; (E.Q.J.); (J.J.G.)
| | - Kristofer S. Fritz
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (C.D.M.); (P.S.H.)
- Correspondence:
| |
Collapse
|
35
|
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: 1.5] [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
|
36
|
Moscardó V, Garcia A, Bondia J, Diaz J, Ramos-Prol A, Rossetti P. Effect of Ethanol Consumption on the Accuracy of a Glucose Oxidase-Based Subcutaneous Glucose Sensor in Subjects with Type 1 Diabetes. SENSORS (BASEL, SWITZERLAND) 2022; 22:3101. [PMID: 35590791 PMCID: PMC9104985 DOI: 10.3390/s22093101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/12/2022] [Accepted: 04/15/2022] [Indexed: 06/15/2023]
Abstract
Continuous glucose monitors (CGM) have improved the management of patients with type 1 diabetes (T1D), with glucose oxidase (GOx)-based sensors being the most used. However, they are potentially subject to both electrochemical and enzymatic interferences, including those related to changes of pH. The objective of this study is to investigate the effect of ethanol, given as beer along with a mixed meal, on the accuracy of a commercial GOx-CGM. Data from 12 T1D participants in a randomized crossover trial to evaluate the effect of meal composition and alcohol consumption on postprandial glucose concentration were used. Absolute error (AE) and mean absolute relative difference (MARD) were calculated. The differences between the alcohol and nonalcohol scenarios were assessed using the Mann−Whitney U and Wilcoxon signed-rank tests. The AE in the alcohol study was low, but significantly greater as compared to the study without alcohol (p-value = 0.0418). The MARD was numerically but not significantly greater. However, both variables were greater at pH < 7.36 and significantly affected by time only in the alcohol arm. In T1D, alcohol consumption affects the accuracy of a GOx-CGM. This effect could be at least partially related to the ethanol-induced changes in pH.
Collapse
Affiliation(s)
- Vanessa Moscardó
- Facultad de Ciencia y Tecnología, Universidad Internacional de Valencia, 46002 Valencia, Spain
| | - Alia Garcia
- Department of Endocrinology, University Hospital of La Ribera, 46600 Alzira, Spain;
| | - Jorge Bondia
- Instituto Universitario de Automática e Informática Industrial, Universitat Politècnica de València, 46022 Valencia, Spain;
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)—Grupo CB17/08/00004, Instituto de Salud Carlos III, 41092 Madrid, Spain;
| | - Julián Diaz
- Hospital Francesc de Borja, 46702 Gandia, Spain; (J.D.); (A.R.-P.)
| | | | - Paolo Rossetti
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)—Grupo CB17/08/00004, Instituto de Salud Carlos III, 41092 Madrid, Spain;
- Department of Endocrinology, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain
| |
Collapse
|
37
|
Clement EJ, Law HCH, Qiao F, Noe D, Trevino JG, Woods NT. Combined Alcohol Exposure and KRAS Mutation in Human Pancreatic Ductal Epithelial Cells Induces Proliferation and Alters Subtype Signatures Determined by Multi-Omics Analysis. Cancers (Basel) 2022; 14:cancers14081968. [PMID: 35454872 PMCID: PMC9027648 DOI: 10.3390/cancers14081968] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/06/2022] [Accepted: 04/07/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Pancreatic ductal adenocarcinoma is a deadly disease wherein alcohol use increases the risk of developing this cancer. Mutations in the KRAS oncogene are required for alcohol to promote pancreatic cancer in mice, but little is known about the molecular events associated with the combined exposure of alcohol and mutant KRAS expression in pancreas cells. In this study, we use pancreas cell models with and without mutant KRAS to evaluate the impact of chronic alcohol exposure on transcription and protein expression. This study identifies numerous differentially expressed transcripts and proteins that could influence the emergence of oncogenic features, such as increased proliferation, in pancreas cells. Abstract Pancreatic Ductal adenocarcinoma (PDAC) is an aggressive cancer commonly exhibiting KRAS-activating mutations. Alcohol contributes to the risk of developing PDAC in humans, and murine models have shown alcohol consumption in the context of KRAS mutation in the pancreas promotes the development of PDAC. The molecular signatures in pancreas cells altered by alcohol exposure in the context of mutant KRAS could identify pathways related to the etiology of PDAC. In this study, we evaluated the combined effects of alcohol exposure and KRAS mutation status on the transcriptome and proteome of pancreatic HPNE cell models. These analyses identified alterations in transcription and translational processes in mutant KRAS cells exposed to alcohol. In addition, multi-omics analysis suggests an increase in the correlation between mRNA transcript and protein abundance in cells exposed to alcohol with an underlying KRAS mutation. Through differential co-expression, SERPINE1 was found to be influential for PDAC development in the context of mutant KRAS and ethanol. In terms of PDAC subtypes, alcohol conditioning of HPNE cells expressing mutant KRAS decreases the Inflammatory subtype signature and increases the Proliferative and Metabolic signatures, as we previously observed in patient samples. The alterations in molecular subtypes were associated with an increased sensitivity to chemotherapeutic agents gemcitabine, irinotecan, and oxaliplatin. These results provide a framework for distinguishing the molecular dysregulation associated with combined alcohol and mutant KRAS in a pancreatic cell line model.
Collapse
Affiliation(s)
- Emalie J. Clement
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; (E.J.C.); (H.C.-H.L.); (F.Q.)
| | - Henry C.-H. Law
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; (E.J.C.); (H.C.-H.L.); (F.Q.)
| | - Fangfang Qiao
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; (E.J.C.); (H.C.-H.L.); (F.Q.)
| | - Dragana Noe
- Mass Spectrometry and Proteomics Core Facility, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Jose G. Trevino
- Department of Surgery, Virginia Commonwealth University, Richmond, VA 23298, USA;
| | - Nicholas T. Woods
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; (E.J.C.); (H.C.-H.L.); (F.Q.)
- Correspondence:
| |
Collapse
|
38
|
Islam R, Melvin SD, Kit Yu RM, O'Connor WA, Anh Tran TK, Andrew-Priestley M, Leusch FDL, MacFarlane GR. Estrogenic mixtures induce alterations in lipidomic profiles in the gonads of female oysters. CHEMOSPHERE 2022; 291:132997. [PMID: 34822861 DOI: 10.1016/j.chemosphere.2021.132997] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 11/18/2021] [Accepted: 11/18/2021] [Indexed: 06/13/2023]
Abstract
This study aimed to reveal possible alterations to lipidomic profiles in Sydney rock oysters, Saccostrea glomerata, exposed to estrogenic mixtures (i.e., estrone, E1; 17β-estradiol, E2; estriol, E3; 17α-ethinylestradiol, EE2; bisphenol A, BPA; 4-t-octylphenol, 4-t-OP; and 4-nonylphenol, 4-NP) at "low" and "high" concentrations, typical of those detected in Australian and global receiving waters. A seven-day acute exposure window exhibited significantly lower abundances of many non-polar metabolites in digestive gland, gills, and gonads. Overall, there was a strong effect of the carrier solvent ethanol (despite a low exposure of 0.0002%), with all solvent containing treatments exhibiting lower abundances of lipidic metabolites, especially in the gill and digestive gland. No significant changes of the lipidome were exhibited in the male gonad by estrogenic exposure. However, in the female gonad, significant reductions of phospholipids and phosphatidylcholine were associated with exposure to high estrogenic mixtures. We hypothesise that the decreases in these phospholipids in the female gonad may be attributable to 1) lower algal consumption and thus lower uptake of lipidic building blocks; 2) a reduction of available substrates for phospholipid and phosphatidylcholine synthesis; and/or 3) induction of reactive oxygen species via estrogen metabolism, which may cause lipid peroxidation and lower abundance of phospholipids.
Collapse
Affiliation(s)
- Rafiquel Islam
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW, 2308, Australia; Department of Applied Chemistry and Chemical Engineering, Islamic University, Kushtia, 7003, Bangladesh
| | - Steven D Melvin
- Australian Rivers Institute, School of Environment and Science, Griffith University, QLD, 4222, Australia
| | - Richard Man Kit Yu
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW, 2308, Australia
| | - Wayne A O'Connor
- New South Wales Department of Primary Industries, Port Stephens Fisheries Institute, Taylors Beach, NSW, 2316, Australia
| | - Thi Kim Anh Tran
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW, 2308, Australia; School of Agriculture and Resources, Vinh University, Viet Nam
| | | | - Frederic D L Leusch
- Australian Rivers Institute, School of Environment and Science, Griffith University, QLD, 4222, Australia
| | - Geoff R MacFarlane
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW, 2308, Australia.
| |
Collapse
|
39
|
The association between alcohol intake shortly before arrest and survival outcomes of out-of-hospital cardiac arrest. Resuscitation 2022; 173:39-46. [DOI: 10.1016/j.resuscitation.2022.02.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 02/07/2022] [Accepted: 02/07/2022] [Indexed: 12/17/2022]
|
40
|
Je J, Song M, Baek JH, Kang JS, Chung HJ, Lee K, Park SW, Kim HJ. Combined Water Extracts from Oxidation-Treated Leaves and Branches of Hovenia dulcis Has Anti-Hangover and Liver Protective Effects in Binge Alcohol Intake of Male Mice. Nutrients 2021; 13:4404. [PMID: 34959956 PMCID: PMC8707081 DOI: 10.3390/nu13124404] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 02/02/2023] Open
Abstract
Hovenia dulcis, known as the oriental raisin tree, is used for food supplements and traditional medicine for the liver after alcohol-related symptoms. However, little information exists about the use of its leaves and branches. In this study, we established a method to use the leaves and branches to develop anti-hangover treatment and elucidated the underlying mechanisms. Oxidation-treated leaves (OL) exhibited high antioxidant content comparable to that of the peduncles and showed an anti-hangover effect in male mice. The branch extract (BE) was enriched in the flavonoid catechin, approximately five times more than OL extract. The mixture of OL and BE (OLB) was formulated in a 2:1 ratio with frozen-dried extract weight and was tested for anti-hangover effects and protective properties against binge alcohol-induced liver injury. OLB showed better anti-hangover effect than OL. In addition to this anti-hangover effect, OLB protected the liver from oxidative/nitrosative damage induced by binge alcohol intake.
Collapse
Affiliation(s)
- Jihyun Je
- Department of Pharmacology, Institute of Health Sciences, Anti-Aging Bio Cell Factory Regional Leading Research Center, Gyeongsang National University Medical School, 15 Jinju-daero 816 Beongil, Jinju 52727, Gyeongnam, Korea;
| | - Miyoung Song
- Department of Anatomy and Convergence Medical Sciences, Institute of Health Sciences, Anti-Aging Bio Cell Factory Regional Leading Research Center, Gyeongsang National University Medical School, 15 Jinju-daero 816 Beongil, Jinju 52727, Gyeongnam, Korea; (M.S.); (J.H.B.); (J.S.K.)
| | - Ji Hyeong Baek
- Department of Anatomy and Convergence Medical Sciences, Institute of Health Sciences, Anti-Aging Bio Cell Factory Regional Leading Research Center, Gyeongsang National University Medical School, 15 Jinju-daero 816 Beongil, Jinju 52727, Gyeongnam, Korea; (M.S.); (J.H.B.); (J.S.K.)
| | - Jae Soon Kang
- Department of Anatomy and Convergence Medical Sciences, Institute of Health Sciences, Anti-Aging Bio Cell Factory Regional Leading Research Center, Gyeongsang National University Medical School, 15 Jinju-daero 816 Beongil, Jinju 52727, Gyeongnam, Korea; (M.S.); (J.H.B.); (J.S.K.)
| | - Hye Jin Chung
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, 501 Jinju-daero, Jinju 52828, Gyeongnam, Korea;
| | - Kwonsu Lee
- Malgeunsan Agricultural Union Corp., 991 Wolasan-ro, Munsan-eup, Jinju 52839, Gyeongnam, Korea;
| | - Sang Won Park
- Department of Pharmacology, Institute of Health Sciences, Anti-Aging Bio Cell Factory Regional Leading Research Center, Gyeongsang National University Medical School, 15 Jinju-daero 816 Beongil, Jinju 52727, Gyeongnam, Korea;
| | - Hyun Joon Kim
- Department of Anatomy and Convergence Medical Sciences, Institute of Health Sciences, Anti-Aging Bio Cell Factory Regional Leading Research Center, Gyeongsang National University Medical School, 15 Jinju-daero 816 Beongil, Jinju 52727, Gyeongnam, Korea; (M.S.); (J.H.B.); (J.S.K.)
| |
Collapse
|
41
|
Pereska Z, Simonovska N, Babulovska A, Berat-Huseini A, Naumoski K, Kostadinoski K. Acute severe poisoning with disinfectant in senior aged patient-case report and overview of literature considering age influence on treatment decision in alcohol-based intoxications. SAGE Open Med Case Rep 2021; 9:2050313X211047717. [PMID: 34567557 PMCID: PMC8461117 DOI: 10.1177/2050313x211047717] [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: 07/04/2021] [Accepted: 09/01/2021] [Indexed: 01/12/2023] Open
Abstract
We present our experiences in the first case of severe suicidal poisoning with 70% ethanol-disinfectant in North Macedonia, in an elderly patient with immunocompromising comorbidities. A 66-year-old unconscious woman was admitted at our clinic, with a history of seropositive rheumatoid arthritis treated with methotrexate. She was in a coma, without signs of serotonin syndrome, recurrent episodes of cardio-respiratory insufficiency under supportive treatment without invasive ventilation, metabolic acidosis, increased D-dimer 3254 ng/mL. The toxicology screening confirmed low benzodiazepines levels and alcoholaemia of 526 mg/dL (5.26 g/L), due to ingestion of 70% ethanol. Considering the decreased biotransformation in the elderly, immunocompromising comorbidities, reports of fatal outcome in poisoned elderly patients with disinfectants under standard fluids supportive protocol, haemodialysis was initiated, with registered associated hypercoagulability which resulted in complete stabilization after 48 h of admission. Treatment protocols of poisoning with ethanol-based disinfectant in the elderly should consider timely performing haemodialysis at lower alcoholaemia levels than recommended.
Collapse
Affiliation(s)
- Zanina Pereska
- Faculty of Medicine, Ss Cyril and Methodius University, University Clinic of Toxicology, Skopje, North Macedonia
| | - Natasha Simonovska
- Faculty of Medicine, Ss Cyril and Methodius University, University Clinic of Toxicology, Skopje, North Macedonia
| | - Aleksandra Babulovska
- Faculty of Medicine, Ss Cyril and Methodius University, University Clinic of Toxicology, Skopje, North Macedonia
| | - Afrodita Berat-Huseini
- Faculty of Medicine, Ss Cyril and Methodius University, University Clinic of Toxicology, Skopje, North Macedonia
| | - Kiril Naumoski
- Faculty of Medicine, Ss Cyril and Methodius University, University Clinic of Toxicology, Skopje, North Macedonia
| | - Kristin Kostadinoski
- Faculty of Medicine, Ss Cyril and Methodius University, University Clinic of Toxicology, Skopje, North Macedonia
| |
Collapse
|
42
|
Brocato E, Wolstenholme JT. Neuroepigenetic consequences of adolescent ethanol exposure. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2021; 160:45-84. [PMID: 34696879 DOI: 10.1016/bs.irn.2021.06.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Adolescence is a critical developmental period characterized by ongoing brain maturation processes including myelination and synaptic pruning. Adolescents experience heightened reward sensitivity, sensation seeking, impulsivity, and diminished inhibitory self-control, which contribute to increased participation in risky behaviors, including the initiation of alcohol use. Ethanol exposure in adolescence alters memory and cognition, anxiety-like behavior, and ethanol sensitivity as well as brain myelination and dendritic spine morphology, with effects lasting into adulthood. Emerging evidence suggests that epigenetic modifications may explain these lasting effects. Focusing on the amygdala, prefrontal cortex and hippocampus, we review studies investigating the epigenetic consequences of adolescent ethanol exposure. Ethanol metabolism globally increases donor substrates for histone acetylation and histone and DNA methylation, and this chapter discusses how this can further impact epigenetic programming of the adolescent brain. Elucidation of the mechanisms through which ethanol can alter the epigenetic code at specific transcripts may provide therapeutic targets for intervention.
Collapse
Affiliation(s)
- Emily Brocato
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States
| | - Jennifer T Wolstenholme
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States; VCU-Alcohol Research Center, Virginia Commonwealth University, Richmond, VA, United States.
| |
Collapse
|
43
|
Rodriguez FD, Coveñas R. Biochemical Mechanisms Associating Alcohol Use Disorders with Cancers. Cancers (Basel) 2021; 13:cancers13143548. [PMID: 34298760 PMCID: PMC8306032 DOI: 10.3390/cancers13143548] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 07/01/2021] [Accepted: 07/14/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Of all yearly deaths attributable to alcohol consumption globally, approximately 12% are due to cancers, representing approximately 0.4 million deceased individuals. Ethanol metabolism disturbs cell biochemistry by targeting the structure and function of essential biomolecules (proteins, nucleic acids, and lipids) and by provoking alterations in cell programming that lead to cancer development and cancer malignancy. A better understanding of the metabolic and cell signaling realm affected by ethanol is paramount to designing effective treatments and preventive actions tailored to specific neoplasias. Abstract The World Health Organization identifies alcohol as a cause of several neoplasias of the oropharynx cavity, esophagus, gastrointestinal tract, larynx, liver, or female breast. We review ethanol’s nonoxidative and oxidative metabolism and one-carbon metabolism that encompasses both redox and transfer reactions that influence crucial cell proliferation machinery. Ethanol favors the uncontrolled production and action of free radicals, which interfere with the maintenance of essential cellular functions. We focus on the generation of protein, DNA, and lipid adducts that interfere with the cellular processes related to growth and differentiation. Ethanol’s effects on stem cells, which are responsible for building and repairing tissues, are reviewed. Cancer stem cells (CSCs) of different origins suffer disturbances related to the expression of cell surface markers, enzymes, and transcription factors after ethanol exposure with the consequent dysregulation of mechanisms related to cancer metastasis or resistance to treatments. Our analysis aims to underline and discuss potential targets that show more sensitivity to ethanol’s action and identify specific metabolic routes and metabolic realms that may be corrected to recover metabolic homeostasis after pharmacological intervention. Specifically, research should pay attention to re-establishing metabolic fluxes by fine-tuning the functioning of specific pathways related to one-carbon metabolism and antioxidant processes.
Collapse
Affiliation(s)
- Francisco D. Rodriguez
- Department of Biochemistry and Molecular Biology, Faculty of Chemistry, University of Salamanca, 37007 Salamanca, Spain
- Group GIR USAL: BMD (Bases Moleculares del Desarrollo), 37007 Salamanca, Spain;
- Correspondence: ; Tel.: +34-677-510-030
| | - Rafael Coveñas
- Group GIR USAL: BMD (Bases Moleculares del Desarrollo), 37007 Salamanca, Spain;
- Institute of Neurosciences of Castilla y León (INCYL), Laboratory of Neuroanatomy of the Peptidergic Systems, University of Salamanca, 37007 Salamanca, Spain
| |
Collapse
|
44
|
Ojeda ML, Nogales F, Romero-Herrera I, Carreras O. Fetal Programming Is Deeply Related to Maternal Selenium Status and Oxidative Balance; Experimental Offspring Health Repercussions. Nutrients 2021; 13:nu13062085. [PMID: 34207090 PMCID: PMC8233903 DOI: 10.3390/nu13062085] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/08/2021] [Accepted: 06/15/2021] [Indexed: 12/12/2022] Open
Abstract
Nutrients consumed by mothers during pregnancy and lactation can exert permanent effects upon infant developing tissues, which could represent an important risk factor for diseases during adulthood. One of the important nutrients that contributes to regulating the cell cycle and tissue development and functionality is the trace element selenium (Se). Maternal Se requirements increase during gestation and lactation. Se performs its biological action by forming part of 25 selenoproteins, most of which have antioxidant properties, such as glutathione peroxidases (GPxs) and selenoprotein P (SELENOP). These are also related to endocrine regulation, appetite, growth and energy homeostasis. In experimental studies, it has been found that low dietary maternal Se supply leads to an important oxidative disruption in dams and in their progeny. This oxidative stress deeply affects gestational parameters, and leads to intrauterine growth retardation and abnormal development of tissues, which is related to endocrine metabolic imbalance. Childhood pathologies related to oxidative stress during pregnancy and/or lactation, leading to metabolic programing disorders like fetal alcohol spectrum disorders (FASD), have been associated with a low maternal Se status and intrauterine growth retardation. In this context, Se supplementation therapy to alcoholic dams avoids growth retardation, hepatic oxidation and improves gestational and breastfeeding parameters in FASD pups. This review is focused on the important role that Se plays during intrauterine and breastfeeding development, in order to highlight it as a marker and/or a nutritional strategy to avoid diverse fetal programming disorders related to oxidative stress.
Collapse
|
45
|
Huang Y, Niu M, Jing J, Zhang ZT, Zhao X, Chen SS, Li SS, Shi Z, Huang A, Zou ZS, Yu YC, Xiao XH, Liangpunsakul S, Wang JB. Metabolomic Analysis Uncovers Energy Supply Disturbance as an Underlying Mechanism of the Development of Alcohol-Associated Liver Cirrhosis. Hepatol Commun 2021; 5:961-975. [PMID: 34141983 PMCID: PMC8183172 DOI: 10.1002/hep4.1699] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 01/31/2021] [Accepted: 02/07/2021] [Indexed: 12/13/2022] Open
Abstract
Alcohol-associated liver disease (ALD) is caused by alcohol metabolism's effects on the liver. The underlying mechanisms from a metabolic view in the development of alcohol-associated liver cirrhosis (ALC) are still elusive. We performed an untargeted serum metabolomic analysis in 14 controls, 16 patients with ALD without cirrhosis (NC), 27 patients with compensated cirrhosis, and 79 patients with decompensated ALC. We identified two metabolic fingerprints associated with ALC development (38 metabolites) and those associated with hepatic decompensation (64 metabolites) in ALC. The cirrhosis-associated fingerprint (eigenmetabolite) showed a better capability to differentiate ALC from NC than the aspartate aminotransferase-to-platelet ratio index score. The eigenmetabolite associated with hepatic decompensation showed an increasing trend during the disease progression and was positively correlated with the Model for End-Stage Liver Disease score. These metabolic fingerprints belong to the metabolites in lipid metabolism, amino acid pathway, and intermediary metabolites in the tricarboxylic acid cycle. Conclusion: The metabolomic fingerprints suggest the disturbance of the metabolites associated with cellular energy supply as an underlying mechanism in the development and progression of alcoholic cirrhosis.
Collapse
Affiliation(s)
- Ying Huang
- School of PharmacyHunan University of Chinese MedicineChangshaHunanChina.,China Military Institute of Chinese MedicineFifth Medical Center of Chinese PLA General HospitalBeijingChina
| | - Ming Niu
- Department of Poisoning TreatmentFifth Medical Center of Chinese PLA General HospitalBeijingChina
| | - Jing Jing
- China Military Institute of Chinese MedicineFifth Medical Center of Chinese PLA General HospitalBeijingChina
| | - Zi-Teng Zhang
- China Military Institute of Chinese MedicineFifth Medical Center of Chinese PLA General HospitalBeijingChina
| | - Xu Zhao
- China Military Institute of Chinese MedicineFifth Medical Center of Chinese PLA General HospitalBeijingChina
| | - Shuai-Shuai Chen
- China Military Institute of Chinese MedicineFifth Medical Center of Chinese PLA General HospitalBeijingChina
| | - Shan-Shan Li
- China Military Institute of Chinese MedicineFifth Medical Center of Chinese PLA General HospitalBeijingChina
| | - Zhuo Shi
- China Military Institute of Chinese MedicineFifth Medical Center of Chinese PLA General HospitalBeijingChina
| | - Ang Huang
- Center for Noninfectious Liver DiseaseFifth Medical Center of Chinese PLA General HospitalBeijingChina
| | - Zheng-Sheng Zou
- Center for Noninfectious Liver DiseaseFifth Medical Center of Chinese PLA General HospitalBeijingChina
| | - Yue-Cheng Yu
- Liver Diseases Center of General Hospital of PLA Eastern Theater Command and Bayi HospitalNanjing University of Chinese MedicineNanjingChina
| | - Xiao-He Xiao
- China Military Institute of Chinese MedicineFifth Medical Center of Chinese PLA General HospitalBeijingChina
| | - Suthat Liangpunsakul
- Division of Gastroenterology and HepatologyDepartment of MedicineIndiana University School of MedicineIndianapolisINUSA
| | - Jia-Bo Wang
- China Military Institute of Chinese MedicineFifth Medical Center of Chinese PLA General HospitalBeijingChina.,School of Chinese MedicineCapital Medical UniversityBeijingChina
| |
Collapse
|
46
|
Tanabe J, Neff S, Sutton B, Ellis S, Patten L, Brown MS, Hoffman PL, Tabakoff B, Burnham EL. Effects of acetate on cerebral blood flow, systemic inflammation, and behavior in alcohol use disorder. Alcohol Clin Exp Res 2021; 45:922-933. [PMID: 33682145 PMCID: PMC8496991 DOI: 10.1111/acer.14588] [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: 11/10/2020] [Revised: 02/19/2021] [Accepted: 02/22/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND Alcohol use disorders (AUDs) are associated with altered regulation of physiological processes in the brain. Acetate, a metabolite of ethanol, has been implicated in several processes that are disrupted in AUDs including transcriptional regulation, metabolism, inflammation, and neurotransmission. To further understand the effects of acetate on brain function in AUDs, we investigated the effects of acetate on cerebral blood flow (CBF), systemic inflammatory cytokines, and behavior in AUD. METHODS Sixteen participants with AUD were recruited from a nonmedical, clinically managed detoxification center. Each participant received acetate and placebo in a randomly assigned order of infusion and underwent 3T MR scanning using quantitative pseudo-continuous arterial spin labeling. Participants and the study team were blinded to the infusion. CBF values (ml/100 g/min) extracted from thalamus were compared between placebo and acetate using a mixed effect linear regression model accounting for infusion order. Voxel-wise CBF comparisons were set at threshold of p < 0.05 cluster-corrected for multiple comparisons, voxel-level p < 0.0001. Plasma cytokine levels and behavior were also assessed between infusions. RESULTS Fifteen men and 1 woman were enrolled with Alcohol Use Disorders Identification Test (AUDIT) scores between 13 and 38 with a mean of 28.3 ± 9.1. Compared to placebo, acetate administration increased CBF in the thalamus bilaterally (Left: 51.2 vs. 68.8, p < 0.001; Right: 53.7 vs. 69.6, p = 0.001), as well as the cerebellum, brainstem, and cortex. Older age and higher AUDIT scores were associated with increases in acetate-induced thalamic blood flow. Cytokine levels and behavioral measures did not differ between placebo and acetate infusions. CONCLUSIONS This pilot study in AUD suggests that during the first week of abstinence from alcohol, the brain's response to acetate differs by brain region and this response may be associated with the severity of alcohol dependence.
Collapse
Affiliation(s)
- Jody Tanabe
- Department of Radiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
- Department of Psychiatry, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
| | - Sarah Neff
- Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
| | - Brianne Sutton
- Department of Psychiatry, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
| | - Sam Ellis
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
| | - Luke Patten
- Department of Biostatistics and Informatics, School of Public Health; University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
| | - Mark S. Brown
- Department of Radiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
| | - Paula L. Hoffman
- Department of Pharmacology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
| | - Boris Tabakoff
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
| | - Ellen L. Burnham
- Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
| |
Collapse
|
47
|
Nogales F, Cebadero O, Romero-Herrera I, Rua RM, Carreras O, Ojeda ML. Selenite supplementation modulates the hepatic metabolic sensors AMPK and SIRT1 in binge drinking exposed adolescent rats by avoiding oxidative stress. Food Funct 2021; 12:3022-3032. [PMID: 33710180 DOI: 10.1039/d0fo02831b] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Binge drinking (BD) is the main alcohol consumption pattern among teenagers. Recently, oxidative stress (OS) generated by BD exposure has been related to hepatic metabolic deregulation and cardiovascular dysfunction. This study analyzed if BD by generating oxidative stress modulates the alteration in hepatic energy homeostasis through two important regulators of energy metabolism: the NAD+-dependent sirtuin deacetylase (SIRT1) and AMP-activated protein kinase (AMPK) and if supplementation with the antioxidant selenium (Se) improves these metabolic disorders. Four groups of adolescent rats supplemented or not with Se (0.4 ppm) and exposed to intermittent i.p. BD were used. BD rats showed an increased AST/ALT ratio, total bilirubin in serum and lipid peroxidation in the liver. The BD rats also showed a higher abdominal/thoracic ratio and increased levels of TG, gluc, and chol compared to the control group, provoking an increase in mean blood pressure (MBP). This alcohol consumption pattern decreased hepatic Se deposits, cytoplasmic GPx activity, and GSH levels as well as the expressions of two metabolic sensors and the pAMPK/AMPK ratio. Se supplementation restored antioxidant parameters and decreased lipid oxidation, avoiding OS and improving the hepatic expression of pAMPK and SIRT1, contributing to the improvement of metabolic (better lipid profile and IRS-1 expression) and vascular function (lower MBP), and to the increase of hepatic functionality (lower AST/ALT ratio). All these actions decrease cardiometabolic risk factor development in the short and long term and could disrupt the relationship between BD and MS, two problems which are currently affecting adolescents.
Collapse
Affiliation(s)
- Fátima Nogales
- Department of Physiology, Faculty of Pharmacy, Seville University, 41012 Seville, Spain.
| | | | | | | | | | | |
Collapse
|
48
|
Hade AC, Philips MA, Reimann E, Jagomäe T, Eskla KL, Traks T, Prans E, Kõks S, Vasar E, Väli M. Chronic Alcohol Use Induces Molecular Genetic Changes in the Dorsomedial Thalamus of People with Alcohol-Related Disorders. Brain Sci 2021; 11:435. [PMID: 33805312 PMCID: PMC8066746 DOI: 10.3390/brainsci11040435] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 03/25/2021] [Accepted: 03/26/2021] [Indexed: 01/12/2023] Open
Abstract
The Mediodorsal (MD) thalamus that represents a fundamental subcortical relay has been underrepresented in the studies focusing on the molecular changes in the brains of subjects with alcohol use disorder (AUD). In the current study, MD thalamic regions from AUD subjects and controls were analyzed with Affymetrix Clariom S human microarray. Long-term alcohol use induced a significant (FDR ≤ 0.05) upregulation of 2802 transcripts and downregulation of 1893 genes in the MD thalamus of AUD subjects. A significant upregulation of GRIN1 (glutamate receptor NMDA type 1) and FTO (alpha-ketoglutarate dependent dioxygenase) was confirmed in western blot analysis. Immunohistochemical staining revealed similar heterogenous distribution of GRIN1 in the thalamic nuclei of both AUD and control subjects. The most prevalent functional categories of upregulated genes were related to glutamatergic and GABAergic neurotransmission, cellular metabolism, and neurodevelopment. The prevalent gene cluster among down-regulated genes was immune system mediators. Forty-two differentially expressed genes, including FTO, ADH1B, DRD2, CADM2, TCF4, GCKR, DPP6, MAPT and CHRH1, have been shown to have strong associations (FDR p < 10-8) with AUD or/and alcohol use phenotypes in recent GWA studies. Despite a small number of subjects, we were able to detect robust molecular changes in the mediodorsal thalamus caused by alcohol emphasizing the importance of deeper brain structures such as diencephalon, in the development of AUD-related dysregulation of neurocircuitry.
Collapse
Affiliation(s)
- Andreas-Christian Hade
- Department of Pathological Anatomy and Forensic Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia; (A.-C.H.); (M.V.)
- Forensic Medical Examination Department, Estonian Forensic Science Institute, 30 Tervise Street, 13419 Tallinn, Estonia
| | - Mari-Anne Philips
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 50411 Tartu, Estonia; (T.J.); (K.-L.E.); (E.V.)
- Centre of Excellence in Genomics and Translational Medicine, University of Tartu, 50411 Tartu, Estonia
| | - Ene Reimann
- Estonian Genome Centre, Institute of Genomics, University of Tartu, 51010 Tartu, Estonia;
| | - Toomas Jagomäe
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 50411 Tartu, Estonia; (T.J.); (K.-L.E.); (E.V.)
- Centre of Excellence in Genomics and Translational Medicine, University of Tartu, 50411 Tartu, Estonia
| | - Kattri-Liis Eskla
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 50411 Tartu, Estonia; (T.J.); (K.-L.E.); (E.V.)
- Centre of Excellence in Genomics and Translational Medicine, University of Tartu, 50411 Tartu, Estonia
| | - Tanel Traks
- Department of Dermatology and Venerology, Institute of Clinical Medicine, University of Tartu, 51010 Tartu, Estonia;
| | - Ele Prans
- Department of Anaesthesiology and Intensive Care, Tartu University Hospital, 50406 Tartu, Estonia;
| | - Sulev Kõks
- Perron Institute for Neurological and Translational Science, Perth, WA 6009, Australia;
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA 6150, Australia
| | - Eero Vasar
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 50411 Tartu, Estonia; (T.J.); (K.-L.E.); (E.V.)
- Centre of Excellence in Genomics and Translational Medicine, University of Tartu, 50411 Tartu, Estonia
| | - Marika Väli
- Department of Pathological Anatomy and Forensic Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia; (A.-C.H.); (M.V.)
- Forensic Medical Examination Department, Estonian Forensic Science Institute, 30 Tervise Street, 13419 Tallinn, Estonia
| |
Collapse
|
49
|
Carrino D, Branca JJV, Becatti M, Paternostro F, Morucci G, Gulisano M, Di Cesare Mannelli L, Pacini A. Alcohol-Induced Blood-Brain Barrier Impairment: An In Vitro Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:2683. [PMID: 33799986 PMCID: PMC7967408 DOI: 10.3390/ijerph18052683] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/01/2021] [Accepted: 03/04/2021] [Indexed: 12/16/2022]
Abstract
In recent years, alcohol abuse has dramatically grown with deleterious consequence for people's health and, in turn, for health care costs. It has been demonstrated, in humans and animals, that alcohol intoxication induces neuroinflammation and neurodegeneration thus leading to brain impairments. Furthermore, it has been shown that alcohol consumption is able to impair the blood-brain barrier (BBB), but the molecular mechanisms underlining this detrimental effect have not been fully elucidated. For this reason, in this study we investigated the effects of alcohol exposure on a rat brain endothelial (RBE4) cell line, as an in vitro-validated model of brain microvascular endothelial cells. To assess whether alcohol caused a concentration-related response, the cells were treated at different times with increasing concentrations (10-1713 mM) of ethyl alcohol (EtOH). Microscopic and molecular techniques, such as cell viability assay, immunofluorescence and Western blotting, were used to examine the mechanisms involved in alcohol-induced brain endothelial cell alterations including tight junction distribution, apoptosis, and reactive oxygen species production. Our findings clearly demonstrate that alcohol causes the formation of gaps between cells by tight junction disassembly, triggered by the endoplasmic reticulum and oxidative stress, highlighted by GRP78 chaperone upregulation and increase in reactive oxygen species production, respectively. The results from this study shed light on the mechanisms underlying alcohol-induced blood-brain barrier dysfunction and a better understanding of these processes will allow us to take advantage of developing new therapeutic strategies in order to prevent the deleterious effects of alcohol.
Collapse
Affiliation(s)
- Donatello Carrino
- Department Experimental and Clinical Medicine, Anatomy and Histology Section, University of Firenze, 50134 Firenze, Italy; (D.C.); (J.J.V.B.); (F.P.); (M.G.)
| | - Jacopo Junio Valerio Branca
- Department Experimental and Clinical Medicine, Anatomy and Histology Section, University of Firenze, 50134 Firenze, Italy; (D.C.); (J.J.V.B.); (F.P.); (M.G.)
| | - Matteo Becatti
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Firenze, 50134 Firenze, Italy;
| | - Ferdinando Paternostro
- Department Experimental and Clinical Medicine, Anatomy and Histology Section, University of Firenze, 50134 Firenze, Italy; (D.C.); (J.J.V.B.); (F.P.); (M.G.)
| | - Gabriele Morucci
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy;
| | - Massimo Gulisano
- Department Experimental and Clinical Medicine, Anatomy and Histology Section, University of Firenze, 50134 Firenze, Italy; (D.C.); (J.J.V.B.); (F.P.); (M.G.)
| | - Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Pharmacology and Toxicology Section, University of Firenze, 50139 Firenze, Italy;
| | - Alessandra Pacini
- Department Experimental and Clinical Medicine, Anatomy and Histology Section, University of Firenze, 50134 Firenze, Italy; (D.C.); (J.J.V.B.); (F.P.); (M.G.)
| |
Collapse
|
50
|
Translational Approaches with Antioxidant Phytochemicals against Alcohol-Mediated Oxidative Stress, Gut Dysbiosis, Intestinal Barrier Dysfunction, and Fatty Liver Disease. Antioxidants (Basel) 2021; 10:antiox10030384. [PMID: 33806556 PMCID: PMC8000766 DOI: 10.3390/antiox10030384] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/25/2021] [Accepted: 02/25/2021] [Indexed: 12/12/2022] Open
Abstract
Emerging data demonstrate the important roles of altered gut microbiomes (dysbiosis) in many disease states in the peripheral tissues and the central nervous system. Gut dysbiosis with decreased ratios of Bacteroidetes/Firmicutes and other changes are reported to be caused by many disease states and various environmental factors, such as ethanol (e.g., alcohol drinking), Western-style high-fat diets, high fructose, etc. It is also caused by genetic factors, including genetic polymorphisms and epigenetic changes in different individuals. Gut dysbiosis, impaired intestinal barrier function, and elevated serum endotoxin levels can be observed in human patients and/or experimental rodent models exposed to these factors or with certain disease states. However, gut dysbiosis and leaky gut can be normalized through lifestyle alterations such as increased consumption of healthy diets with various fruits and vegetables containing many different kinds of antioxidant phytochemicals. In this review, we describe the mechanisms of gut dysbiosis, leaky gut, endotoxemia, and fatty liver disease with a specific focus on the alcohol-associated pathways. We also mention translational approaches by discussing the benefits of many antioxidant phytochemicals and/or their metabolites against alcohol-mediated oxidative stress, gut dysbiosis, intestinal barrier dysfunction, and fatty liver disease.
Collapse
|