Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
Add more filters

Publication year range
1.
J Sci Food Agric ; 104(9): 5077-5088, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38284794

ABSTRACT

BACKGROUND: During high sugar fermentation, yeast is mainly affected by high sugar stress in the early stage. It becomes jointly affected by high sugar and ethanol stress as ethanol accumulates during fermentation. Ca2+, as the second messenger of the cell, mediates various metabolic processes. In this study, the effects of the Ca2+ signal on the activities of key enzymes, expression of related genes of ethanol metabolism, and mitochondrial function were investigated. RESULTS: The results showed a significant increase in the activities of enzymes related to ethanol metabolism in yeast cells under a high sugar environment. Ca2+ significantly promoted the activities of enzymes related to mitochondrial respiratory metabolism and regulated the carbon flow between ethanol metabolism and the tricarboxylic acid cycle. The high sugar environment affected the expression of genes related to carbon metabolism, while the addition of Ca2+ stabilized the expression of related genes. CONCLUSION: Ca2+ signal participated in ethanol and mitochondrial metabolism and regulated the key enzymes and related gene expression to enhance the resistance of yeast to stress during high sugar fermentation. © 2024 Society of Chemical Industry.


Subject(s)
Calcium , Ethanol , Fermentation , Mitochondria , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/genetics , Ethanol/metabolism , Mitochondria/metabolism , Mitochondria/genetics , Calcium/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Gene Expression Regulation, Fungal , Citric Acid Cycle
2.
J Gastroenterol Hepatol ; 38(8): 1233-1240, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37423758

ABSTRACT

Alcoholic liver disease (ALD), which is a leading cause of morbidity and mortality worldwide, covers a large spectrum of liver injuries ranging from simple steatosis to steatohepatitis, advanced fibrosis, cirrhosis, and hepatocellular carcinoma. The pathogenesis of ALD includes genetic and epigenetic alterations, oxidative stress, acetaldehyde-mediated toxicity and cytokine and chemokine-induced inflammation, metabolic reprogramming, immune damage, and dysbiosis of the gut microbiota. This review discusses the progress in the pathogenesis and molecular mechanism of ALD, which could provide evidence for further research on the potential therapeutic strategies targeting these pathways.


Subject(s)
Carcinoma, Hepatocellular , Liver Diseases, Alcoholic , Liver Neoplasms , Humans , Ethanol/metabolism , Liver Diseases, Alcoholic/etiology , Liver/pathology , Carcinoma, Hepatocellular/complications , Liver Neoplasms/pathology
3.
Arch Biochem Biophys ; 718: 109148, 2022 03 30.
Article in English | MEDLINE | ID: mdl-35143783

ABSTRACT

Ethanol is known to significantly affect gluconeogenesis and lipid metabolism in the liver, primarily by altering the redox ratio in both cytosol and mitochondria. The effect of ethanol was analyzed using a comprehensive, dynamic model of liver metabolism that takes into account sub-cellular compartmentation, detailed kinetics for the citric acid cycle, ethanol and acetaldehyde oxidation, and gluconeogenesis, and inter-compartmental transport of metabolites, including the malate-aspartate shuttle. The kinetic expression for alcohol dehydrogenase takes into account inhibition by ethanol and NADH. Simulations of perfusions of the rat liver were performed with various combinations of substrates (lactate, pyruvate, and fatty acids), with subsequent addition of ethanol to the perfusate. The model successfully predicts NADH/NAD+, in both cytosol and mitochondria, the expected directional flux of reducing equivalents between the two compartments during perfusion with different gluconeogenic precursors, and the effect of ethanol on glucose and ketone body production. This model can serve as a platform for in silico experiments investigating the effects of ethanol on the many dehydrogenases, and thus the major carbohydrate and lipid metabolic pathways in the liver, as well as potential effects of various drugs that may interact with ethanol.


Subject(s)
Gluconeogenesis , NAD , Animals , Ethanol/metabolism , Fatty Acids/metabolism , Lactic Acid/metabolism , Lipid Metabolism , Liver/metabolism , NAD/metabolism , Oxidation-Reduction , Pyruvic Acid/metabolism , Rats
4.
Microb Cell Fact ; 21(1): 116, 2022 Jun 16.
Article in English | MEDLINE | ID: mdl-35710409

ABSTRACT

BACKGROUND: Microbial production of propionate from diluted streams of ethanol (e.g., deriving from syngas fermentation) is a sustainable alternative to the petrochemical production route. Yet, few ethanol-fermenting propionigenic bacteria are known, and understanding of their metabolism is limited. Anaerotignum neopropionicum is a propionate-producing bacterium that uses the acrylate pathway to ferment ethanol and CO2 to propionate and acetate. In this work, we used computational and experimental methods to study the metabolism of A. neopropionicum and, in particular, the pathway for conversion of ethanol into propionate. RESULTS: Our work describes iANEO_SB607, the first genome-scale metabolic model (GEM) of A. neopropionicum. The model was built combining the use of automatic tools with an extensive manual curation process, and it was validated with experimental data from this and published studies. The model predicted growth of A. neopropionicum on ethanol, lactate, sugars and amino acids, matching observed phenotypes. In addition, the model was used to implement a dynamic flux balance analysis (dFBA) approach that accurately predicted the fermentation profile of A. neopropionicum during batch growth on ethanol. A systematic analysis of the metabolism of A. neopropionicum combined with model simulations shed light into the mechanism of ethanol fermentation via the acrylate pathway, and revealed the presence of the electron-transferring complexes NADH-dependent reduced ferredoxin:NADP+ oxidoreductase (Nfn) and acryloyl-CoA reductase-EtfAB, identified for the first time in this bacterium. CONCLUSIONS: The realisation of the GEM iANEO_SB607 is a stepping stone towards the understanding of the metabolism of the propionate-producer A. neopropionicum. With it, we have gained insight into the functioning of the acrylate pathway and energetic aspects of the cell, with focus on the fermentation of ethanol. Overall, this study provides a basis to further exploit the potential of propionigenic bacteria as microbial cell factories.


Subject(s)
Clostridium , Propionates , Acrylates/metabolism , Clostridiales , Clostridium/metabolism , Ethanol/metabolism , Fermentation , Lactic Acid/metabolism , Propionates/metabolism
5.
Int J Mol Sci ; 23(22)2022 Nov 16.
Article in English | MEDLINE | ID: mdl-36430619

ABSTRACT

Aldehydes, particularly acetaldehyde, are carcinogenic molecules and their concentrations in foodstuffs should be controlled to avoid upper aerodigestive tract (UADT) and liver cancers. Highly reactive, acetaldehyde forms DNA and protein adducts, impairing physiological functions and leading to the development of pathological conditions. The consumption of aged beer, outside of the ethanol metabolism, exposes habitual drinkers to this carcinogen, whose concentrations can be over-increased due to post-brewing chemical and biochemical reactions. Storage-related changes are a challenge faced by the brewing industry, impacting volatile compound formation and triggering flavor instability. Aldehydes are among the volatile compounds formed during beer aging, recognized as off-flavor compounds. To track and understand aldehyde formation through multiple pathways during beer storage, consequent changes in flavor but particularly quality losses and harmful compound formation, this systematic review reunited data on volatile compound profiles through gas chromatography analyses from 2011 to 2021. Conditions to avoid flavor instability and successful methods for reducing beer staling, and consequent acetaldehyde accumulation, were raised by exploring the dynamic conversion between free and bound-state aldehydes. Future research should focus on implementing sensory analyses to investigate whether adding aldehyde-binding agents, e.g., cysteine and bisulfite, would contribute to consumer acceptance, restore beer flavor, and minimize acetaldehyde-related health damage.


Subject(s)
Acetaldehyde , Aldehydes , Humans , Aged , Beer , Carcinogens , Carcinogenesis
6.
Yeast ; 37(2): 227-236, 2020 02.
Article in English | MEDLINE | ID: mdl-31603243

ABSTRACT

The methylotrophic yeast Pichia pastoris (syn. Komagataella phaffii) is a successful host widely used in recombinant protein production. The widespread use of a methanol-regulated alcohol oxidase 1 (AOX1) promoter for recombinant protein production has directed studies particularly about methanol metabolism in this yeast. Although there is comprehensive knowledge about methanol metabolism, there are other mechanisms in P. pastoris that have not been investigated yet, such as ethanol metabolism. The gene responsible for the consumption of ethanol ADH2 (XM_002491337, known as ADH3) was identified and characterized in our previous study. In this study, the ADH genes (XM_002489969, XM_002491163, XM_002493969) in P. pastoris genome were investigated to determine their roles in ethanol production by gene disruption analysis. We report that the ADH900 (XM_002491163) is the main gene responsible for ethanol production in P. pastoris. The ADH2 gene, previously identified as the only gene responsible for ethanol consumption, also plays a minor role in ethanol production in the absence of the ADH900 gene. The investigation of the carbon source regulation mechanism has also revealed that the ADH2 gene exhibit similar expression behaviours with ADH900 on glucose, glycerol, and methanol, however, it is strongly induced by ethanol.


Subject(s)
Alcohol Dehydrogenase/genetics , Ethanol/metabolism , Genes, Fungal/genetics , Pichia/enzymology , Pichia/genetics , Pichia/metabolism , Alcohol Oxidoreductases/genetics , Culture Media , Escherichia coli/genetics , Fungal Proteins/genetics , Fungal Proteins/metabolism , Gene Expression Regulation, Fungal , Glucose/metabolism , Glycerol/metabolism , Methanol/metabolism , Pichia/growth & development , Promoter Regions, Genetic , Recombinant Proteins/metabolism , Sequence Analysis, Protein , Sequence Homology, Amino Acid
7.
Biol Lett ; 16(4): 20200070, 2020 04.
Article in English | MEDLINE | ID: mdl-32343936

ABSTRACT

Humans have a long evolutionary relationship with ethanol, pre-dating anthropogenic sources, and possess unusually efficient ethanol metabolism, through a mutation that evolved in our last common ancestor with African great apes. Increased exposure to dietary ethanol through fermenting fruits and nectars is hypothesized to have selected for this in our lineage. Yet, other mammals have frugivorous and nectarivorous diets, raising the possibility of natural ethanol exposure and adaptation in other taxa. We conduct a comparative genetic analysis of alcohol dehydrogenase class IV (ADH IV) across mammals to provide insight into their evolutionary history with ethanol. We find genetic variation and multiple pseudogenization events in ADH IV, indicating the ability to metabolize ethanol is variable. We suggest that ADH enzymes are evolutionarily plastic and show promise for revealing dietary adaptation. We further highlight the derived condition of humans and draw attention to problems with modelling the physiological responses of other mammals on them, a practice that has led to potentially erroneous conclusions about the likelihood of natural intoxication in wild animals. It is a fallacy to assume that other animals share our metabolic adaptations, rather than taking into consideration each species' unique physiology.


Subject(s)
Ethanol , Hominidae , Adaptation, Physiological , Animals , Biological Evolution , Diet/veterinary , Humans
8.
Xenobiotica ; 50(10): 1180-1201, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32338108

ABSTRACT

Ethanol, as a small-molecule organic compound exhibiting both hydrophilic and lipophilic properties, quickly pass through the biological barriers. Over 95% of absorbed ethanol undergoes biotransformation, the remaining amount is excreted unchanged, mainly with urine and exhaled air.The main route of ethyl alcohol metabolism is its oxidation to acetaldehyde, which is converted into acetic acid with the participation of cytosolic NAD+ - dependent alcohol (ADH) and aldehyde (ALDH) dehydrogenases. Oxidative biotransformation pathways of ethanol also include reactions catalyzed by the microsomal ethanol oxidizing system (MEOS), peroxisomal catalase and aldehyde (AOX) and xanthine (XOR) oxidases. The resulting acetic acid can be activated to acetyl-CoA by the acetyl-CoA synthetase (ACS).It is also possible, to a much smaller extent, non-oxidative routes of ethanol biotransformation including its esterification with fatty acids by ethyl fatty acid synthase (FAEES), re-esterification of phospholipids, especially phosphatidylcholines, with phospholipase D (PLD), coupling with sulfuric acid by alcohol sulfotransferase (SULT) and with glucuronic acid using UDP-glucuronyl transferase (UGT, syn. UDPGT).The intestinal microbiome plays a significant role in the ethanol biotransformation and in the initiation and progression of liver diseases stimulated by ethanol and its metabolite - acetaldehyde, or by lipopolysaccharide and ROS.


Subject(s)
Biotransformation/physiology , Ethanol/metabolism , Acetaldehyde , Catalase/metabolism , Humans , Metabolic Clearance Rate , Metabolic Networks and Pathways , Microsomes, Liver/metabolism , Oxidation-Reduction
9.
Microb Cell Fact ; 17(1): 82, 2018 May 31.
Article in English | MEDLINE | ID: mdl-29855316

ABSTRACT

BACKGROUND: Saccharomyces cerevisiae is a host for the industrial production of S-adenosyl-L-methionine (SAM), which has been widely used in pharmaceutical and nutritional supplement industries. It has been reported that the intracellular SAM content in S. cerevisiae can be improved by the addition of ethanol during cultivation. However, the metabolic state in ethanol-assimilating S. cerevisiae remains unclear. In this study, 13C-metabolic flux analysis (13C-MFA) was conducted to investigate the metabolic regulation responsible for the high SAM production from ethanol. RESULTS: The comparison between the metabolic flux distributions of central carbon metabolism showed that the metabolic flux levels of the tricarboxylic acid cycle and glyoxylate shunt in the ethanol culture were significantly higher than that of glucose. Estimates of the ATP balance from the 13C-MFA data suggested that larger amounts of excess ATP was produced from ethanol via increased oxidative phosphorylation. The finding was confirmed by the intracellular ATP level under ethanol-assimilating condition being similarly higher than glucose. CONCLUSIONS: These results suggest that the enhanced ATP regeneration due to ethanol assimilation was critical for the high SAM accumulation.


Subject(s)
Ethanol/metabolism , Metabolic Flux Analysis/methods , S-Adenosylmethionine/metabolism , Saccharomyces cerevisiae/metabolism
10.
Biosci Biotechnol Biochem ; 82(4): 724-731, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29616890

ABSTRACT

Alcoholic beverages are enjoyed together with meals worldwide, but their excessive intake is associated with an increased risk of various diseases. We investigated whether S-allyl-L-cysteine sulfoxide (ACSO), a sulfuric odor precursor of garlic, suppresses elevation in plasma ethanol concentration by accelerating ethanol metabolism and preventing ethanol absorption from the gut in rats. ACSO and garlic extract with a high ACSO content (Garlic-H) suppressed elevation in concentrations of ethanol and acetaldehyde in plasma and promoted the activities of alcohol dehydrogenase and aldehyde dehydrogenase. However, ACSO and Garlic-H did not affect plasma acetate so much. Furthermore, we examined the change in plasma ethanol concentration by injecting ACSO or Garlic-H into the ligated stomach or jejunum together with ethanol solution. ACSO and Garlic-H suppressed the absorption of ethanol from the stomach and jejunum, but suppression in the jejunum was less than in the stomach. In conclusion, ACSO inhibits ethanol absorption and accelerates ethanol metabolism.


Subject(s)
Alcoholic Beverages , Blood Alcohol Content , Cysteine/analogs & derivatives , Ethanol/blood , Garlic/chemistry , Intestinal Absorption/drug effects , Acetaldehyde/blood , Administration, Oral , Alcohol Dehydrogenase/metabolism , Aldehyde Dehydrogenase/metabolism , Ammonia/analysis , Animals , Arginine/analysis , Cysteine/administration & dosage , Cysteine/analysis , Cysteine/pharmacology , Ethanol/administration & dosage , Ethanol/metabolism , Jejunum , Liver/enzymology , Male , Odorants , Plant Extracts/chemistry , Pyruvic Acid/analysis , Rats, Sprague-Dawley , Stomach
11.
Proc Natl Acad Sci U S A ; 112(10): 3074-9, 2015 Mar 10.
Article in English | MEDLINE | ID: mdl-25713355

ABSTRACT

Correcting a genetic mutation that leads to a loss of function has been a challenge. One such mutation is in aldehyde dehydrogenase 2 (ALDH2), denoted ALDH2*2. This mutation is present in ∼ 0.6 billion East Asians and results in accumulation of toxic acetaldehyde after consumption of ethanol. To temporarily increase metabolism of acetaldehyde in vivo, we describe an approach in which a pharmacologic agent recruited another ALDH to metabolize acetaldehyde. We focused on ALDH3A1, which is enriched in the upper aerodigestive track, and identified Alda-89 as a small molecule that enables ALDH3A1 to metabolize acetaldehyde. When given together with the ALDH2-specific activator, Alda-1, Alda-89 reduced acetaldehyde-induced behavioral impairment by causing a rapid reduction in blood ethanol and acetaldehyde levels after acute ethanol intoxication in both wild-type and ALDH2-deficient, ALDH2*1/*2, heterozygotic knock-in mice. The use of a pharmacologic agent to recruit an enzyme to metabolize a substrate that it usually does not metabolize may represent a novel means to temporarily increase elimination of toxic agents in vivo.


Subject(s)
Aldehyde Dehydrogenase/drug effects , Aldehyde Dehydrogenase/metabolism , Ethanol/metabolism , Aldehyde Dehydrogenase, Mitochondrial , Animals , Humans , Mice , Mice, Transgenic , Protein Binding
12.
J Biol Chem ; 291(22): 11865-75, 2016 May 27.
Article in English | MEDLINE | ID: mdl-27036938

ABSTRACT

Fatty acid ethyl esters (FAEEs) are non-oxidative metabolites of ethanol that accumulate in human tissues upon ethanol intake. Although FAEEs are considered as toxic metabolites causing cellular dysfunction and tissue damage, the enzymology of FAEE metabolism remains poorly understood. In this study, we used a biochemical screen in Saccharomyces cerevisiae to identify and characterize putative hydrolases involved in FAEE catabolism. We found that Yju3p, the functional orthologue of mammalian monoacylglycerol lipase (MGL), contributes >90% of cellular FAEE hydrolase activity, and its loss leads to the accumulation of FAEE. Heterologous expression of mammalian MGL in yju3Δ mutants restored cellular FAEE hydrolase activity and FAEE catabolism. Moreover, overexpression or pharmacological inhibition of MGL in mouse AML-12 hepatocytes decreased or increased FAEE levels, respectively. FAEEs were transiently incorporated into lipid droplets (LDs) and both Yju3p and MGL co-localized with these organelles. We conclude that the storage of FAEE in inert LDs and their mobilization by LD-resident FAEE hydrolases facilitate a controlled metabolism of these potentially toxic lipid metabolites.


Subject(s)
Biological Evolution , Ethanol/metabolism , Fatty Acids/metabolism , Hepatocytes/metabolism , Monoacylglycerol Lipases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Animals , Cells, Cultured , Chromatography, Thin Layer , Gas Chromatography-Mass Spectrometry , Hepatocytes/cytology , Humans , Inactivation, Metabolic , Mice , Saccharomyces cerevisiae/growth & development
13.
Fungal Genet Biol ; 95: 30-38, 2016 10.
Article in English | MEDLINE | ID: mdl-27486067

ABSTRACT

The non-conventional yeast Yarrowia lipolytica is able to utilize a wide range of different substrates like glucose, glycerol, ethanol, acetate, proteins and various hydrophobic molecules. Although most metabolic pathways for the utilization of these substrates have been clarified by now, it was not clear whether ethanol is oxidized by alcohol dehydrogenases or by an alternative oxidation system inside the cell. In order to detect the genes that are required for ethanol utilization in Y. lipolytica, eight alcohol dehydrogenase (ADH) genes and one alcohol oxidase gene (FAO1) have been identified and respective deletion strains were tested for their ability to metabolize ethanol. As a result of this, we found that the availability of ADH1, ADH2 or ADH3 is required for ethanol utilization in Y. lipolytica. A strain with deletions in all three genes is lacking the ability to utilize ethanol as sole carbon source. Although Adh2p showed by far the highest enzyme activity in an in vitro assay, the availability of any of the three genes was sufficient to enable a decent growth. In addition to ADH1, ADH2 and ADH3, an acetyl-CoA synthetase encoding gene (ACS1) was found to be essential for ethanol utilization. As Y. lipolytica is a non-fermenting yeast, it is neither able to grow under anaerobic conditions nor to produce ethanol. To investigate whether Y. lipolytica may produce ethanol, the key genes of alcoholic fermentation in S. cerevisiae, ScADH1 and ScPDC1, were overexpressed in an ADH and an ACS1 deletion strain. However, instead of producing ethanol, the respective strains regained the ability to use ethanol as single carbon source and were still not able to grow under anaerobic conditions.


Subject(s)
Acetate-CoA Ligase/genetics , Alcohol Dehydrogenase/genetics , Ethanol/metabolism , Yarrowia/enzymology , Yarrowia/genetics , Aerobiosis , Alcohol Dehydrogenase/classification , Anaerobiosis , DNA, Fungal , Enzyme Activation/genetics , Escherichia coli/genetics , Fermentation , Gene Expression Regulation, Fungal , Genes, Fungal/genetics , Oxidation-Reduction , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Sequence Deletion , Transformation, Genetic , Yarrowia/growth & development , Yarrowia/metabolism
14.
IUBMB Life ; 68(12): 916-923, 2016 12.
Article in English | MEDLINE | ID: mdl-27714979

ABSTRACT

Ethanol is a widely used psychoactive drug whose chronic abuse is associated with organ dysfunction and disease. Although the prevalent metabolic fate of ethanol in the human body is oxidation a smaller fraction undergoes nonoxidative metabolism yielding ethyl glucuronide, ethyl sulfate, phosphatidylethanol and fatty acid ethyl esters. Nonoxidative ethanol metabolites persist in tissues and body fluids for much longer than ethanol itself and represent biomarkers for the assessment of ethanol intake in clinical and forensic settings. Of note, the nonoxidative reaction of ethanol with phospholipids and fatty acids yields bioactive compounds that affect cellular signaling pathways and organelle function and may contribute to ethanol toxicity. Thus, despite low quantitative contributions of nonoxidative pathways to overall ethanol metabolism the resultant ethanol metabolites have important biological implications. In this review we summarize the current knowledge about the enzymatic formation of nonoxidative ethanol metabolites in humans and discuss the implications of nonoxidative ethanol metabolites as biomarkers of ethanol intake and mediators of ethanol toxicity. © 2016 IUBMB Life, 68(12):916-923, 2016.


Subject(s)
Alcohol Drinking/metabolism , Ethanol/metabolism , Fatty Acids/metabolism , Glycerophospholipids/metabolism , Animals , Biomarkers/metabolism , Glucuronates/metabolism , Humans , Inactivation, Metabolic , Oxidation-Reduction , Sulfuric Acid Esters/metabolism , Tissue Distribution
15.
Biotechnol Lett ; 38(3): 463-9, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26573637

ABSTRACT

OBJECTIVES: To characterize the genes responsible for ethanol utilization in Pichia pastoris. RESULTS: ADH3 (XM_002491337) and ADH (FN392323) genes were disrupted in P. pastoris. The ADH3 mutant strain, MK115 (Δadh3), lost its ability to grow on minimal ethanol media but produced ethanol in minimal glucose medium. ADH3p was responsible for 92 % of total Adh enzyme activity in glucose media. The double knockout strain MK117 (Δadh3Δadh) also produced ethanol. The Adh activities of X33 and MK116 (Δadh) strains were not different. Thus, the ADH gene does not play a role in ethanol metabolism. CONCLUSION: The PpADH3 is the only gene responsible for consumption of ethanol in P. pastoris.


Subject(s)
Alcohol Dehydrogenase/metabolism , Ethanol/metabolism , Pichia/enzymology , Pichia/metabolism , Alcohol Dehydrogenase/genetics , Culture Media/chemistry , Gene Knockout Techniques , Pichia/genetics , Pichia/growth & development
16.
Am J Physiol Gastrointest Liver Physiol ; 309(7): G566-77, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26251470

ABSTRACT

Alcohol exposure worsens the course and outcomes of hepatitis C virus (HCV) infection. Activation of protective antiviral genes is induced by IFN-α signaling, which is altered in liver cells by either HCV or ethanol exposure. However, the mechanisms of the combined effects of HCV and ethanol metabolism in IFN-α signaling modulation are not well elucidated. Here, we explored a possibility that ethanol metabolism potentiates HCV-mediated dysregulation of IFN-α signaling in liver cells via impairment of methylation reactions. HCV-infected Huh7.5 CYP2E1(+) cells and human hepatocytes were exposed to acetaldehyde (Ach)-generating system (AGS) and stimulated with IFN-α to activate IFN-sensitive genes (ISG) via the Jak-STAT-1 pathway. We observed significant suppression of signaling events by Ach. Ach exposure decreased STAT-1 methylation via activation of protein phosphatase 2A and increased the protein inhibitor of activated STAT-1 (PIAS-1)-STAT-1 complex formation in both HCV(+) and HCV(-) cells, preventing ISG activation. Treatment with a promethylating agent, betaine, attenuated all examined Ach-induced defects. Ethanol metabolism-induced changes in ISGs are methylation related and confirmed by in vivo studies on HCV(+) transgenic mice. HCV- and Ach-induced impairment of IFN signaling temporarily increased HCV RNA levels followed by apoptosis of heavily infected cells. We concluded that Ach potentiates the suppressive effects of HCV on activation of ISGs attributable to methylation-dependent dysregulation of IFN-α signaling. A temporary increase in HCV RNA sensitizes the liver cells to Ach-induced apoptosis. Betaine reverses the inhibitory effects of Ach on IFN signaling and thus can be used for treatment of HCV(+) alcohol-abusing patients.


Subject(s)
Acetaldehyde/pharmacology , DNA Methylation/drug effects , Hepacivirus/physiology , Hepatocytes/immunology , Immunity, Innate/drug effects , Animals , Betaine/pharmacology , Cell Line , Ethanol/metabolism , Hepatocytes/virology , Humans , Immunoblotting , Immunoprecipitation , Interferon-alpha/pharmacology , Male , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction , STAT1 Transcription Factor/metabolism , Signal Transduction , Transfection
17.
Int J Cancer ; 135(10): 2424-36, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-24719202

ABSTRACT

Alcohol consumption is an established risk factor for head and neck cancer (HNC). The major carcinogen from alcohol is acetaldehyde, which may be produced by humans or by oral microorganisms through the metabolism of ethanol. To account for the different sources of acetaldehyde production, the current study examined the interplay between alcohol consumption, oral hygiene (as a proxy measure for the growth of oral microorganisms), and alcohol-metabolizing genes (ADH1B and ALDH2) in the risk of HNC. We found that both the fast (*2/*2) and the slow (*1/*1+ *1/*2) ADH1B genotypes increased the risk of HNC due to alcohol consumption, and this association differed according to the slow/non-functional ALDH2 genotypes (*1/*2+ *2/*2) or poor oral hygiene. In persons with the fast ADH1B genotype, the HNC risk associated with alcohol drinking was increased for those with the slow/non-functional ALDH2 genotypes. For those with the slow ADH1B genotypes, oral hygiene appeared to play an important role; the highest magnitude of an increased HNC risk in alcohol drinkers occurred among those with the worst oral hygiene. This is the first study to show that the association between alcohol drinking and HNC risk may be modified by the interplay between genetic polymorphisms of ADH1B and ALDH2 and oral hygiene. Although it is important to promote abstinence from or reduction of alcohol drinking to decrease the occurrence of HNC, improving oral hygiene practices may provide additional benefit.


Subject(s)
Alcohol Dehydrogenase/genetics , Alcohol Drinking/adverse effects , Aldehyde Dehydrogenase/genetics , Carcinoma, Squamous Cell/etiology , Head and Neck Neoplasms/etiology , Oral Hygiene/adverse effects , Polymorphism, Genetic/genetics , Adult , Aged , Aged, 80 and over , Aldehyde Dehydrogenase, Mitochondrial , Carcinoma, Squamous Cell/epidemiology , Case-Control Studies , Female , Follow-Up Studies , Genotype , Head and Neck Neoplasms/epidemiology , Humans , Male , Middle Aged , Polymerase Chain Reaction , Prognosis , Risk Factors , Smoking/adverse effects , Young Adult
18.
Article in English | MEDLINE | ID: mdl-39031522

ABSTRACT

BACKGROUND: Genome-wide association studies (GWAS) have identified hundreds of common variants associated with alcohol consumption. In contrast, genetic studies of alcohol consumption that use rare variants are still in their early stages. No prior studies of alcohol consumption have examined whether common and rare variants implicate the same genes and molecular networks, leaving open the possibility that the two approaches might identify distinct biology. METHODS: To address this knowledge gap, we used publicly available alcohol consumption GWAS summary statistics (GSCAN, N = 666,978) and whole exome sequencing data (Genebass, N = 393,099) to identify a set of common and rare variants for alcohol consumption. We used gene-based analysis to implicate genes from common and rare variant analyses, which we then propagated onto a shared molecular network using a network colocalization procedure. RESULTS: Gene-based analysis of each dataset implicated 294 (common variants) and 35 (rare variants) genes, including ethanol metabolizing genes ADH1B and ADH1C, which were identified by both analyses, and ANKRD12, GIGYF1, KIF21B, and STK31, which were identified in only the rare variant analysis, but have been associated with other neuropsychiatric traits. Network colocalization revealed significant network overlap between the genes identified via common and rare variants. The shared network identified gene families that function in alcohol metabolism, including ADH, ALDH, CYP, and UGT. Seventy-one of the genes in the shared network were previously implicated in neuropsychiatric or substance use disorders but not alcohol-related behaviors (e.g. EXOC2, EPM2A, and CACNG4). Differential gene expression analysis showed enrichment in the liver and several brain regions. CONCLUSIONS: Genes implicated by network colocalization identify shared biology relevant to alcohol consumption, which also underlie neuropsychiatric traits and substance use disorders that are comorbid with alcohol use, providing a more holistic understanding of two disparate sources of genetic information.

19.
Biotechnol Biofuels Bioprod ; 17(1): 115, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39160588

ABSTRACT

Industrial biotechnology heavily relies on the microbial conversion of carbohydrate substrates derived from sugar- or starch-rich crops. This dependency poses significant challenges in the face of a rising population and food scarcity. Consequently, exploring renewable, non-competing carbon sources for sustainable bioprocessing becomes increasingly important. Ethanol, a key C2 feedstock, presents a promising alternative, especially for producing acetyl-CoA derivatives. In this review, we offer an in-depth analysis of ethanol's potential as an alternative carbon source, summarizing its distinctive characteristics when utilized by microbes, microbial ethanol metabolism pathway, and microbial responses and tolerance mechanisms to ethanol stress. We provide an update on recent progress in ethanol-based biomanufacturing and ethanol biosynthesis, discuss current challenges, and outline potential research directions to guide future advancements in this field. The insights presented here could serve as valuable theoretical support for researchers and industry professionals seeking to harness ethanol's potential for the production of high-value products.

20.
Life Sci ; 343: 122508, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38382873

ABSTRACT

Alcohol intake provokes severe organ injuries including alcoholic cardiomyopathy with hallmarks of cardiac remodeling and contractile defects. This study examined the toxicity of facilitated ethanol metabolism in alcoholism-evoked changes in myocardial morphology and contractile function, insulin signaling and various cell death domains using cardiac-selective overexpression of alcohol dehydrogenase (ADH). WT and ADH mice were offered an alcohol liquid diet for 12 weeks prior to assessment of cardiac geometry, function, ER stress, apoptosis and ferroptosis. Alcohol intake provoked pronounced glucose intolerance, cardiac remodeling and contractile anomalies with apoptosis, ER stress, and ferroptosis, the effects were accentuated by ADH with the exception of global glucose intolerance. Hearts from alcohol ingesting mice displayed dampened insulin-stimulated phosphorylation of insulin receptor (tyr1146) and IRS-1 (tyrosine) along with elevated IRS-1 serine phosphorylation, the effect was augmented by ADH. Alcohol challenge dampened phosphorylation of Akt and GSK-3ß, and increased phosphorylation of c-Jun and JNK, the effects were accentuated by ADH. Alcohol challenge promoted ER stress, FK506 binding protein 5 (FKBP5), YAP, apoptosis and ferroptosis, the effects were exaggerated by ADH. Using a short-term ethanol challenge model (3 g/kg, i.p., twice in three days), we found that inhibition of FKBP5-YAP signaling or facilitated ethanol detoxification by Alda-1 alleviated ethanol cardiotoxicity. In vitro study revealed that the ethanol metabolite acetaldehyde evoked cardiac contractile anomalies, lipid peroxidation, and apoptosis, the effects of which were mitigated by Alda-1, inhibition of ER stress, FKBP5 and YAP. These data suggest that facilitated ethanol metabolism via ADH exacerbates alcohol-evoked myocardial remodeling, functional defects, and insulin insensitivity possibly through a FKBP5-YAP-associated regulation of ER stress and ferroptosis.


Subject(s)
Alcoholism , Ferroptosis , Glucose Intolerance , Tacrolimus Binding Proteins , Mice , Animals , Ethanol/pharmacology , Alcohol Dehydrogenase/metabolism , Alcohol Dehydrogenase/pharmacology , Glucose Intolerance/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Ventricular Remodeling , Mice, Transgenic , Alcoholism/complications , Alcoholism/metabolism , Myocardial Contraction , Insulin/metabolism , Myocytes, Cardiac/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL