51
|
Israel Y, Quintanilla ME, Ezquer F, Morales P, Santapau D, Berríos‐Cárcamo P, Ezquer M, Olivares B, Herrera‐Marschitz M. Aspirin and N-acetylcysteine co-administration markedly inhibit chronic ethanol intake and block relapse binge drinking: Role of neuroinflammation-oxidative stress self-perpetuation. Addict Biol 2021; 26:e12853. [PMID: 31733014 DOI: 10.1111/adb.12853] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/18/2019] [Accepted: 10/23/2019] [Indexed: 12/13/2022]
Abstract
Chronic alcohol intake leads to neuroinflammation and cell injury, proposed to result in alterations that perpetuate alcohol intake and cued relapse. Studies show that brain oxidative stress is consistently associated with alcohol-induced neuroinflammation, and literature implies that oxidative stress and neuroinflammation perpetuate each other. In line with a self-perpetuating mechanism, it is hypothesized that inhibition of either oxidative stress or neuroinflammation could reduce chronic alcohol intake and relapse. The present study conducted on alcohol-preferring rats shows that chronic ethanol intake was inhibited by 50% to 55% by the oral administration of low doses of either the antioxidant N-acetylcysteine (40 mg/kg/d) or the anti-inflammatory aspirin (ASA; 15 mg/kg/d), while the co-administration of both dugs led to a 70% to 75% (P < .001) inhibition of chronic alcohol intake. Following chronic alcohol intake, a prolonged alcohol deprivation, and subsequent alcohol re-access, relapse drinking resulted in blood alcohol levels of 95 to 100 mg/dL in 60 minutes, which were reduced by 60% by either N-acetylcysteine or aspirin and by 85% by the co-administration of both drugs (blood alcohol: 10 to 15 mg/dL; P < .001). Alcohol intake either on the chronic phase or following deprivation and re-access led to a 50% reduction of cortical glutamate transporter GLT-1 levels, while aspirin administration fully returned GLT-1 to normal levels. N-acetylcysteine administration did not alter GLT-1 levels, while N-acetylcysteine may activate the cystine/glutamate transport xCT, presynaptically inhibiting relapse. Overall, the study suggests that a neuroinflammation/oxidative stress self-perpetuation cycle maintains chronic alcohol intake and relapse drinking. The co-administration of anti-inflammatory and antioxidant agents may have translational value in alcohol-use disorders.
Collapse
Affiliation(s)
- Yedy Israel
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine University of Chile Santiago Chile
| | - María Elena Quintanilla
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine University of Chile Santiago Chile
| | - Fernando Ezquer
- Centro de Medicina Regenerativa, Facultad de Medicina, Universidad del Desarrollo Santiago Chile
| | - Paola Morales
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine University of Chile Santiago Chile
- Department of Neuroscience, Faculty of Medicine University of Chile Santiago Chile
| | - Daniela Santapau
- Centro de Medicina Regenerativa, Facultad de Medicina, Universidad del Desarrollo Santiago Chile
| | - Pablo Berríos‐Cárcamo
- Centro de Medicina Regenerativa, Facultad de Medicina, Universidad del Desarrollo Santiago Chile
| | - Marcelo Ezquer
- Centro de Medicina Regenerativa, Facultad de Medicina, Universidad del Desarrollo Santiago Chile
| | - Belen Olivares
- Centro de Química Médica, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo Santiago Chile
| | - Mario Herrera‐Marschitz
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine University of Chile Santiago Chile
| |
Collapse
|
52
|
Microbiota reprogramming for treatment of alcohol-related liver disease. Transl Res 2020; 226:26-38. [PMID: 32687975 PMCID: PMC7572584 DOI: 10.1016/j.trsl.2020.07.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/01/2020] [Accepted: 07/13/2020] [Indexed: 02/08/2023]
Abstract
In the past decade knowledge has expanded regarding the importance of the gut microbiota in maintaining intestinal homeostasis and overall health. During this same time, we have also gained appreciation for the role of the gut-liver axis in the development of liver diseases. Alcohol overconsumption is one of the leading causes of liver failure globally. However, not all people with alcohol use disorder progress to advanced stages of liver disease. With advances in technology to investigate the gut microbiome and metabolome, we are now beginning to delineate alcohol's effects on the gut microbiome in relation to liver disease. This review presents our current understanding on the role of the gut microbiota during alcohol exposure, and various therapeutic attempts that have been made to reprogram the gut microbiota with the goal of alleviating alcoholic-related liver disease.
Collapse
|
53
|
Park JH, Jung IK, Lee Y, Jin S, Yun HJ, Kim BW, Kwon HJ. Alcohol stimulates the proliferation of mouse small intestinal epithelial cells via Wnt signaling. Biochem Biophys Res Commun 2020; 534:639-645. [PMID: 33220923 DOI: 10.1016/j.bbrc.2020.11.028] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 11/08/2020] [Indexed: 02/07/2023]
Abstract
The intestinal epithelium is one of the fastest renewing tissues in mammals and is a barrier against toxic substances such as alcohol. Excessive alcohol can induce intestinal damage leading to intestinal bowel diseases. Thus, the control of small intestinal epithelial cell (IEC) regeneration is thought to be important for homeostasis in response to epithelium damage. However, reports on how epithelial cells respond to small intestinal damage are scarce. We investigated the effects of alcohol consumption on small intestinal epithelial cells of mice. To verify that alcohol altered the small intestinal epithelium, we used 8-10 weeks old male C57BL/6J mice for models of chronic and binge alcohol consumption (the NIAAA model) in addition to an organoid model. Alcohol promoted the proliferative activity of IECs and intestinal stem cells (ISCs) in intestinal crypts. Alcohol consumption increased expression of the proliferation marker cyclin D1 and activated the p44/42 MAPK (Erk1/2) signaling pathway in small intestinal epithelial cells. The Wnt target genes were markedly increased in IECs from alcohol-treated mice. In the small intestinal organoid model exposed to alcohol, the organoid area and numbers of buds increased with alcohol concentrations up to 0.5% similar to in vivo observations. These results suggest that alcohol consumption stimulates the proliferation of small intestinal epithelial cells via Wnt signaling.
Collapse
Affiliation(s)
- Jung-Ha Park
- Biopharmaceutical Engineering Major, Division of Applied Bioengineering, Dong-eui University, Busan, 47340, South Korea; Core-Facility Center for Tissue Regeneration, Dong-eui University, South Korea.
| | - In Kyo Jung
- Biopharmaceutical Engineering Major, Division of Applied Bioengineering, Dong-eui University, Busan, 47340, South Korea; Department of Chronic Disease Convergence Research, Korea National Institute of Health, Cheongju, 28159, South Korea
| | - Yongjun Lee
- Hongcheon Institute of Medicinal Herb, Hongcheon, 25142, South Korea
| | - Soojung Jin
- Core-Facility Center for Tissue Regeneration, Dong-eui University, South Korea
| | - Hee Jung Yun
- Biopharmaceutical Engineering Major, Division of Applied Bioengineering, Dong-eui University, Busan, 47340, South Korea
| | - Byung Woo Kim
- Biopharmaceutical Engineering Major, Division of Applied Bioengineering, Dong-eui University, Busan, 47340, South Korea; Blue-Bio Industry Regional Innovation Center, Dong-eui University, South Korea
| | - Hyun Ju Kwon
- Biopharmaceutical Engineering Major, Division of Applied Bioengineering, Dong-eui University, Busan, 47340, South Korea; Core-Facility Center for Tissue Regeneration, Dong-eui University, South Korea; Blue-Bio Industry Regional Innovation Center, Dong-eui University, South Korea.
| |
Collapse
|
54
|
Jochems PGM, Garssen J, Rietveld PCS, Govers C, Tomassen MMM, Wichers HJ, van Bergenhenegouwen J, Masereeuw R. Novel Dietary Proteins Selectively Affect Intestinal Health In Vitro after Clostridium difficile-Secreted Toxin A Exposure. Nutrients 2020; 12:E2782. [PMID: 32932980 PMCID: PMC7551268 DOI: 10.3390/nu12092782] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 09/01/2020] [Accepted: 09/10/2020] [Indexed: 12/25/2022] Open
Abstract
Bacterial gastroenteritis forms a burden on a global scale, both socially and economically. The Gram-positive bacterium Clostridium difficile is an inducer of gastrointestinal bacterial infections, often triggered following disruption of the microbiota by broad-spectrum antibiotics to treat other conditions. The clinical manifestatiaons, e.g., diarrhea, are driven by its toxins secretion, toxin A (TcdA) and toxin B (TcdB). Current therapies are focused on discontinuing patient medication, including antibiotics. However, relapse rates upon therapy are high (20-25%). Here, eighteen dietary proteins were evaluated for their capacity to restore gut health upon C. difficile-derived TcdA exposure. We used bioengineered intestinal tubules to assess proteins for their beneficial effects by examining the epithelial barrier, cell viability, brush-border enzyme activity, IL-6 secretion, IL-8 secretion and nitric oxide (NO) levels upon TcdA challenge. TcdA effectively disrupted the epithelial barrier, increased mitochondrial activity, but did not affect alkaline phosphatase activity, IL-6, IL-8 and NO levels. Intervention with dietary proteins did not show a protective effect on epithelial barrier integrity or mitochondrial activity. However, bovine plasma and potato protein increased alkaline phosphatase activity, egg-white protein increased IL-6 and IL-8 release and wheat, lesser mealworm and yeast protein increased NO levels after TcdA exposure. Hence, dietary proteins can influence parameters involved in intestinal physiology and immune activation suggesting that supplementation with specific dietary proteins may be of benefit during C. difficile infections.
Collapse
Affiliation(s)
- Paulus G. M. Jochems
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (P.G.M.J.); (J.G.); (P.C.S.R.); (J.v.B.)
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (P.G.M.J.); (J.G.); (P.C.S.R.); (J.v.B.)
- Nutricia Research, Global Center of Excellence Immunology, 3584 CT Utrecht, The Netherlands
| | - Pascale C. S. Rietveld
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (P.G.M.J.); (J.G.); (P.C.S.R.); (J.v.B.)
| | - Coen Govers
- Food & Biobased Research, Wageningen University & Research, 6708 WE Wageningen, The Netherlands; (C.G.); (M.M.M.T.); (H.J.W.)
| | - Monic M. M. Tomassen
- Food & Biobased Research, Wageningen University & Research, 6708 WE Wageningen, The Netherlands; (C.G.); (M.M.M.T.); (H.J.W.)
| | - Harry J. Wichers
- Food & Biobased Research, Wageningen University & Research, 6708 WE Wageningen, The Netherlands; (C.G.); (M.M.M.T.); (H.J.W.)
| | - Jeroen van Bergenhenegouwen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (P.G.M.J.); (J.G.); (P.C.S.R.); (J.v.B.)
- Nutricia Research, Global Center of Excellence Immunology, 3584 CT Utrecht, The Netherlands
| | - Rosalinde Masereeuw
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (P.G.M.J.); (J.G.); (P.C.S.R.); (J.v.B.)
| |
Collapse
|
55
|
Alcohol Addiction, Gut Microbiota, and Alcoholism Treatment: A Review. Int J Mol Sci 2020; 21:ijms21176413. [PMID: 32899236 PMCID: PMC7504034 DOI: 10.3390/ijms21176413] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 09/01/2020] [Accepted: 09/02/2020] [Indexed: 02/07/2023] Open
Abstract
Alcohol addiction is a leading risk factor for personal death and disability. In 2016, alcohol use caused 2.2% of female deaths and 6.8% of male deaths, and disability-adjusted life years (DALYs) were 2.3% in female and 8.9% in male. Individuals with alcohol use disorder are at high risk of anxiety, depression, impaired cognition performance, and illicit drug use and are comorbid with liver disease, such as alcoholic hepatitis and liver cirrhosis, which is a major cause of personal death and disability worldwide. Psychological interventions, such as cognitive behavior therapy and motivational interviewing, as well as medical treatments, such as disulfiram, naltrexone, acamprosate, and nalmefene, are used for the treatment of alcohol addiction in Europe and the United States. However, the effect of current interventions is limited, and the need for additional interventions is substantial. Alcohol use impairs the intestinal barrier and causes changes to the intestinal permeability as well as the gut microbiota composition. Emerging studies have tried to reveal the role of the gut–brain axis among individuals with alcohol use disorder with or without alcohol liver disease. Bacterial products penetrate the impaired intestinal barrier and cause central inflammation; changes to the gut microbiota impair enterohepatic circulation of bile acids; alcohol abuse causes shortage of vital nutrients such as thiamine. Several studies have suggested that probiotics, through either oral administration or fecal microbiota transplantation, increased intestinal levels of potentially beneficial bacteria such as bifidobacteria and lactobacilli, improving the levels of liver-associated enzymes in patients with mild alcoholic hepatitis, and demonstrating beneficial psychotropic effects on anxiety and depression. In addition to medications for alcohol addiction, gene editing therapy such as clustered regularly interspaced short palindromic repeats (CRISPRs) may be another potential research target. Alcohol dehydrogenase (ADH) and aldehyde dehydrogenase (ALDH), which are associated with ADH and ALDH genes, are major enzymes involved in alcohol metabolism, and gene editing approaches may have the potential to directly modify specific genes to treat alcoholism caused by genetic defects. Further research is needed to study the effect of the combined treatment for alcohol addiction.
Collapse
|
56
|
Qian G, Ho JWK. Challenges and emerging systems biology approaches to discover how the human gut microbiome impact host physiology. Biophys Rev 2020; 12:851-863. [PMID: 32638331 PMCID: PMC7429608 DOI: 10.1007/s12551-020-00724-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 07/02/2020] [Indexed: 02/07/2023] Open
Abstract
Research in the human gut microbiome has bloomed with advances in next generation sequencing (NGS) and other high-throughput molecular profiling technologies. This has enabled the generation of multi-omics datasets which holds promises for big data-enabled knowledge acquisition in the form of understanding the normal physiological and pathological involvement of gut microbiomes. Ample evidence suggests that distinct microbial compositions in the human gut are associated with different diseases. However, the biological mechanisms underlying these associations are often unclear. There is a need to move beyond statistical associations to discover how changes in the gut microbiota mechanistically affect host physiology and disease development. This review summarises state-of-the-art big data and systems biology approaches for mechanism discovery.
Collapse
Affiliation(s)
- Gordon Qian
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Joshua W K Ho
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong.
| |
Collapse
|
57
|
Quintanilla ME, Ezquer F, Morales P, Ezquer M, Olivares B, Santapau D, Herrera-Marschitz M, Israel Y. N-Acetylcysteine and Acetylsalicylic Acid Inhibit Alcohol Consumption by Different Mechanisms: Combined Protection. Front Behav Neurosci 2020; 14:122. [PMID: 32848653 PMCID: PMC7412547 DOI: 10.3389/fnbeh.2020.00122] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 06/23/2020] [Indexed: 12/20/2022] Open
Abstract
Chronic ethanol intake results in brain oxidative stress and neuroinflammation, which have been postulated to perpetuate alcohol intake and to induce alcohol relapse. The present study assessed the mechanisms involved in the inhibition of: (i) oxidative stress; (ii) neuroinflammation; and (iii) ethanol intake that follow the administration of the antioxidant N-acetylcysteine (NAC) and the anti-inflammatory acetylsalicylic acid (ASA) to animals that had consumed ethanol chronically. At doses used clinically, NAC [40 mg/kg per day orally (p.o.)] and ASA (15 mg/kg per day p.o.) significantly inhibited chronic alcohol intake and relapse intake in alcohol-preferring rats. The coadministration of both drugs reduced ethanol intake by 65% to 70%. N-acetylcysteine administration: (a) induced the Nrf2-ARE system, lowering the hippocampal oxidative stress assessed as the ratio of oxidized glutathione (GSSG)/reduced glutathione (GSH); (b) reduced the neuroinflammation assessed by astrocyte and microglial activation by immunofluorescence; and (c) inhibited chronic and relapse ethanol intake. These effects were blocked by sulfasalazine, an inhibitor of the xCT transporter, which incorporates cystine (precursor of GSH) and extrudes extracellular glutamate, an agonist of the inhibitory mGlu2/3 receptor, which lowers the synaptic glutamatergic tone. The inhibitor of mGlu2/3 receptor (LY341495) blocked the NAC-induced inhibition of both relapse ethanol intake and neuroinflammation without affecting the GSSG/GSH ratio. Unlike N-acetylcysteine, ASA inhibited chronic alcohol intake and relapse via lipoxin A4, a strong anti-inflammatory metabolite of arachidonic acid generated following the ASA acetylation of cyclooxygenases. Accordingly, the lipoxin A4 receptor inhibitor, WRW4, blocked the ASA-induced reduction of ethanol intake. Overall, via different mechanisms, NAC and ASA administered in clinically relevant doses combine their effects inhibiting ethanol intake.
Collapse
Affiliation(s)
- María Elena Quintanilla
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Fernando Ezquer
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago, Chile
| | - Paola Morales
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile.,Department of Neuroscience, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Marcelo Ezquer
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago, Chile
| | - Belen Olivares
- Centro de Química Médica, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago, Chile
| | - Daniela Santapau
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago, Chile
| | - Mario Herrera-Marschitz
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Yedy Israel
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
58
|
Bishehsari F, Moossavi S, Engen PA, Liu X, Zhang Y. Abnormal Food Timing Promotes Alcohol-Associated Dysbiosis and Colon Carcinogenesis Pathways. Front Oncol 2020; 10:1029. [PMID: 32850307 PMCID: PMC7396506 DOI: 10.3389/fonc.2020.01029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 05/26/2020] [Indexed: 12/21/2022] Open
Abstract
Background: Alcohol consumption is an established risk factor for colorectal cancer (CRC). Identifying cofactor(s) that modulate the effect of alcohol on colon inflammation and carcinogenesis could help risk stratification for CRC. Disruption of circadian rhythm by light/dark shift promotes alcohol-induced colonic inflammation and cancer. More recently, we found that abnormal food timing causes circadian rhythm disruption and promotes alcohol associated colon carcinogenesis. In this study, we examined the interaction of wrong-time feeding (WTF) and alcohol on CRC-related pathways, in relation to changes in microbial community structure. Methods: Polyposis mice (TS4Cre ×cAPC Δ468) underwent four conditions: alcohol or water and feeding during the light (wrong-time fed/WTF) or during the dark (right-time fed). Colonic cecum mucosal gene expression was analyzed by RNA-seq. Microbiota 16S ribosomal RNA sequencing analysis was used to examine colonic feces. Modeling was used to estimate the extent of the gene expression changes that could be related to the changes in the colonic microbial composition. Results: The circadian rhythm pathway was the most altered pathway by the WTF treatment, indicating that WTF is disruptive to the colonic circadian rhythm. Pathway analysis revealed interaction of WTF with alcohol in dysregulating pathways related to colon carcinogenesis. Similarly, the interaction of alcohol and WTF was detected at multiple parameters of the colonic microbiota including α and β diversity, as well as the community structure. Our modeling revealed that almost a third of total gene alterations induced by our treatments could be related to alterations in the abundance of the microbial taxa. Conclusion: These data support the promoting effect of abnormal food timing alcohol-associated CRC-related pathways in the colon and suggest colon dysbiosis as a targetable mechanism.
Collapse
Affiliation(s)
- Faraz Bishehsari
- Department of Internal Medicine, Division of Gastroenterology, Rush University Medical Center, Chicago, IL, United States
| | - Shirin Moossavi
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
- Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
- Digestive Oncology Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Phillip A. Engen
- Department of Internal Medicine, Division of Gastroenterology, Rush University Medical Center, Chicago, IL, United States
| | - Xiaohan Liu
- Department of Mathematics, Statistics, and Computer Science, University of Illinois at Chicago, Chicago, IL, United States
| | - Yue Zhang
- Department of Mathematics, Statistics, and Computer Science, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
59
|
Elshaghabee FMF, Ghadimi D, Habermann D, de Vrese M, Bockelmann W, Kaatsch HJ, Heller KJ, Schrezenmeir J. Effect of Oral Administration of Weissella confusa on Fecal and Plasma Ethanol Concentrations, Lipids and Glucose Metabolism in Wistar Rats Fed High Fructose and Fat Diet. Hepat Med 2020; 12:93-106. [PMID: 32617026 PMCID: PMC7326399 DOI: 10.2147/hmer.s254195] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 05/20/2020] [Indexed: 12/12/2022] Open
Abstract
Background and Purpose In previous investigations, Weissella confusa was shown to lack the metabolic pathway from fructose to mannitol and to produce ethanol when cultivated in the presence of fructose. Hence, we assessed the effect of oral administration of W. confusa (strain NRRL-B-14171) on blood and fecal ethanol concentrations, glucose and lipid metabolism and traits of the metabolic syndrome in Wistar rats (n=27) fed diets with two different fat and fructose levels and with or without the addition of W. confusa during a total intervention time of 15 weeks (105 days). Materials and Methods From week 1 to 6, rats were given a medium fructose and fat (MFru-MF) diet containing 28% fructose and 10% fat without the addition of W. confusa (control group, n=13) or mixed with 30 g per kg diet of lyophilized W. confusa (10.56 ± 0.20 log CFU/g; W. confusa group, n=14). From week 7 to 15, the percentage of dietary fructose and fat in the control and W. confusa group was increased to 56% and 16%, respectively (high fructose-high fat (HFru-HF) diet). Results In HFru-HF-fed rats, W. confusa was detected in feces, regardless of whether W. confusa was added to the diet or not, but not in rats receiving the MFru-MF diet without added W. confusa or in an additional control group (n=10) fed standard rat food without fructose, increased fat content and W. confusa. This indicates that fecal W. confusa may be derived from orally administered W. confusa as well as - in the case of high fructose and fat intake and obesity of rats - from the intestinal microbiota. As shown by multifactorial ANOVA, blood ethanol, the relative liver weight, serum triglycerides, and serum cholesterol as well as fecal ethanol, ADH, acetate, propionate and butyrate, but not lactate, were significantly higher in the W. confusa - compared to the control group. Discussion This is the first in vivo trial demonstrating that heterofermentative lactic acid bacteria lacking the mannitol pathway (like W. confusa) can increase fecal and blood ethanol concentrations in mammals on a high fructose-high fat diet. This may explain why W. confusa resulted in hyperlipidemia and may promote development of NAFLD in the host.
Collapse
Affiliation(s)
- Fouad M F Elshaghabee
- Max Rubner-Institute, Federal Research Institute of Nutrition and Food, Department of Microbiology and Biotechnology, Kiel, Germany
| | - Darab Ghadimi
- Max Rubner-Institute, Federal Research Institute of Nutrition and Food, Department of Microbiology and Biotechnology, Kiel, Germany
| | - Diana Habermann
- Max Rubner-Institute, Federal Research Institute of Nutrition and Food, Department of Microbiology and Biotechnology, Kiel, Germany
| | - Michael de Vrese
- Max Rubner-Institute, Federal Research Institute of Nutrition and Food, Department of Microbiology and Biotechnology, Kiel, Germany
| | - Wilhelm Bockelmann
- Max Rubner-Institute, Federal Research Institute of Nutrition and Food, Department of Microbiology and Biotechnology, Kiel, Germany
| | - Hans-Jürgen Kaatsch
- Institute of Legal Medicine, University Hospital of Schleswig-Holstein, Kiel, Germany
| | - Knut J Heller
- Max Rubner-Institute, Federal Research Institute of Nutrition and Food, Department of Microbiology and Biotechnology, Kiel, Germany
| | - Jürgen Schrezenmeir
- Max Rubner-Institute, Federal Research Institute of Nutrition and Food, Department of Physiology and Biochemistry of Nutrition, Karlsruhe, Germany.,Clinical Research Center, Kiel Innovation and Technology Center, Kiel, Germany
| |
Collapse
|
60
|
Rios-Arce ND, Schepper JD, Dagenais A, Schaefer L, Daly-Seiler CS, Gardinier JD, Britton RA, McCabe LR, Parameswaran N. Post-antibiotic gut dysbiosis-induced trabecular bone loss is dependent on lymphocytes. Bone 2020; 134:115269. [PMID: 32061677 PMCID: PMC7138712 DOI: 10.1016/j.bone.2020.115269] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 12/31/2019] [Accepted: 02/10/2020] [Indexed: 02/06/2023]
Abstract
Recent studies in mouse models have shown that gut microbiota significantly influences bone health. We demonstrated that 2-week oral treatment with broad spectrum antibiotics followed by 4 weeks of recovery of the gut microbiota results in dysbiosis (microbiota imbalance)-induced bone loss in mice. Because gut microbiota is critical for the development of the immune system and since both microbiota and the immune system can regulate bone health, in this study, we tested the role of the immune system in mediating post-antibiotic dysbiosis-induced bone loss. For this, we treated wild-type (WT) and lymphocyte deficient Rag2 knockout (KO) mice with ampicillin/neomycin cocktail in water for 2 weeks followed by 4 weeks of water without antibiotics. This led to a significant bone loss (31% decrease from control) in WT mice. Interestingly, no bone loss was observed in the KO mice suggesting that lymphocytes are required for dysbiosis-induced bone loss. Bray-Curtis diversity metrics showed similar microbiota changes in both the WT and KO post-antibiotic treated groups. However, several operational taxonomic units (OTUs) classified as Lactobacillales were significantly higher in the repopulated KO when compared to the WT mice, suggesting that these bacteria might play a protective role in preventing bone loss in the KO mice after antibiotic treatment. The effect of dysbiosis on bone was therefore examined in the WT mice in the presence or absence of oral Lactobacillus reuteri treatment for 4 weeks (post-ABX treatment). As hypothesized, mice treated with L. reuteri did not display bone loss, suggesting a bone protective role for this group of bacteria. Taken together, our studies elucidate an important role for lymphocytes in regulating post-antibiotic dysbiosis-induced bone loss.
Collapse
Affiliation(s)
- Naiomy Deliz Rios-Arce
- Comparative Medicine and Integrative Biology Program, Michigan State University, East Lansing, MI, USA; Department of Physiology, Michigan State University, East Lansing, MI, USA
| | | | - Andrew Dagenais
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Laura Schaefer
- Department of Molecular Virology and Microbiology, Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, TX, USA
| | | | | | - Robert A Britton
- Department of Molecular Virology and Microbiology, Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, TX, USA
| | - Laura R McCabe
- Department of Physiology, Michigan State University, East Lansing, MI, USA; Department of Radiology, Michigan State University, East Lansing, MI, USA; Biomedical Imaging Research Center, Michigan State University, East Lansing, MI, USA.
| | - Narayanan Parameswaran
- Comparative Medicine and Integrative Biology Program, Michigan State University, East Lansing, MI, USA; Department of Physiology, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
61
|
Addolorato G, Ponziani FR, Dionisi T, Mosoni C, Vassallo GA, Sestito L, Petito V, Picca A, Marzetti E, Tarli C, Mirijello A, Zocco MA, Lopetuso LR, Antonelli M, Rando MM, Paroni Sterbini F, Posteraro B, Sanguinetti M, Gasbarrini A. Gut microbiota compositional and functional fingerprint in patients with alcohol use disorder and alcohol-associated liver disease. Liver Int 2020; 40:878-888. [PMID: 31951082 DOI: 10.1111/liv.14383] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 01/09/2020] [Accepted: 01/10/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Alcohol use disorder (AUD) represents the most common cause of liver disease. The gut microbiota plays a critical role in the progression of alcohol-related liver damage. Aim of this study was to characterize the gut microbial composition and function in AUD patients with alcohol-associated liver disease (AALD). METHODS This study included 36 AUD patients (14 with cirrhosis) who were active drinkers and an equal number of matched controls. Stool microbial composition, serum levels of lipopolysaccharide, cytokines/chemokines and gut microbiota functional profile were assessed. RESULTS AUD patients had a decreased microbial alpha diversity as compared to controls (0.092 vs 0.130, P = .047) and a specific gut microbial signature. The reduction of Akkermansia and the increase in Bacteroides were able to identify AUD patients with an accuracy of 93.4%. Serum levels of lipopolysaccharide (4.91 vs 2.43, P = .009) and pro-inflammatory mediators (tumour necrosis factor alpha 60.85 vs 15.08, P = .001; interleukin [IL] 1beta 4.43 vs 1.72, P = .0001; monocyte chemoattractant protein 1 225.22 vs 16.43, P = .006; IL6 1.87 vs 1.23, P = .008) were significantly increased in AUD patients compared to controls and in cirrhotic patients compared to non-cirrhotic ones (IL6 3.74 vs 1.39, P = .019; IL8 57.60 vs 6.53, P = .004). The AUD-associated gut microbiota showed an increased expression of gamma-aminobutyric acid (GABA) metabolic pathways and energy metabolism. CONCLUSIONS AUD patients present a specific gut microbial fingerprint, associated with increased endotoxaemia, systemic inflammatory status and functional alterations that may be involved in the progression of the AALD and in the pathogenesis of AUD.
Collapse
Affiliation(s)
- Giovanni Addolorato
- Institute of Internal Medicine and Gastroenterology, Catholic University of Rome, Rome, Italy
- Alcohol Use Disorder and Alcohol Related Disease Unit, Department of Internal Medicine and Gastroenterology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Francesca R Ponziani
- Internal Medicine, Gastroenterology and Hepatology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Tommaso Dionisi
- Alcohol Use Disorder and Alcohol Related Disease Unit, Department of Internal Medicine and Gastroenterology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Carolina Mosoni
- Alcohol Use Disorder and Alcohol Related Disease Unit, Department of Internal Medicine and Gastroenterology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | | | - Luisa Sestito
- Alcohol Use Disorder and Alcohol Related Disease Unit, Department of Internal Medicine and Gastroenterology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Valentina Petito
- Internal Medicine, Gastroenterology and Hepatology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Anna Picca
- Department of Geriatrics, Neuroscience and Orthopedics, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Emanuele Marzetti
- Department of Geriatrics, Neuroscience and Orthopedics, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Claudia Tarli
- Alcohol Use Disorder and Alcohol Related Disease Unit, Department of Internal Medicine and Gastroenterology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Antonio Mirijello
- Department of Medical Sciences, IRCCS Casa Sollievo della Sofferenza Hospital, San Giovanni Rotondo, Italy
| | - Maria Assunta Zocco
- Internal Medicine, Gastroenterology and Hepatology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Loris R Lopetuso
- Internal Medicine, Gastroenterology and Hepatology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Mariangela Antonelli
- Alcohol Use Disorder and Alcohol Related Disease Unit, Department of Internal Medicine and Gastroenterology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Maria M Rando
- Alcohol Use Disorder and Alcohol Related Disease Unit, Department of Internal Medicine and Gastroenterology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Francesco Paroni Sterbini
- Department of Laboratory and Infectious Disease Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Brunella Posteraro
- Internal Medicine, Gastroenterology and Hepatology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Institute of Microbiology, Chatoloc University of Rome, Rome, Italy
| | - Maurizio Sanguinetti
- Department of Laboratory and Infectious Disease Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Institute of Microbiology, Chatoloc University of Rome, Rome, Italy
| | - Antonio Gasbarrini
- Institute of Internal Medicine and Gastroenterology, Catholic University of Rome, Rome, Italy
- Internal Medicine, Gastroenterology and Hepatology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| |
Collapse
|
62
|
Schepper JD, Collins F, Rios-Arce ND, Kang HJ, Schaefer L, Gardinier JD, Raghuvanshi R, Quinn RA, Britton R, Parameswaran N, McCabe LR. Involvement of the Gut Microbiota and Barrier Function in Glucocorticoid-Induced Osteoporosis. J Bone Miner Res 2020; 35:801-820. [PMID: 31886921 DOI: 10.1002/jbmr.3947] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 12/05/2019] [Accepted: 12/14/2019] [Indexed: 12/14/2022]
Abstract
Glucocorticoids (GCs) are potent immune-modulating drugs with significant side effects, including glucocorticoid-induced osteoporosis (GIO). GCs directly induce osteoblast and osteocyte apoptosis but also alter intestinal microbiota composition. Although the gut microbiota is known to contribute to the regulation of bone density, its role in GIO has never been examined. To test this, male C57/Bl6J mice were treated for 8 weeks with GC (prednisolone, GC-Tx) in the presence or absence of broad-spectrum antibiotic treatment (ABX) to deplete the microbiota. Long-term ABX prevented GC-Tx-induced trabecular bone loss, showing the requirement of gut microbiota for GIO. Treatment of GC-Tx mice with a probiotic (Lactobacillus reuteri [LR]) prevented trabecular bone loss. Microbiota analyses indicated that GC-Tx changed the abundance of Verrucomicobiales and Bacteriodales phyla and random forest analyses indicated significant differences in abundance of Porphyromonadaceae and Clostridiales operational taxonomic units (OTUs) between groups. Furthermore, transplantation of GC-Tx mouse fecal material into recipient naïve, untreated WT mice caused bone loss, supporting a functional role for microbiota in GIO. We also report that GC caused intestinal barrier breaks, as evidenced by increased serum endotoxin level (2.4-fold), that were prevented by LR and ABX treatments. Enhancement of barrier function with a mucus supplement prevented both GC-Tx-induced barrier leakage and trabecular GIO. In bone, treatment with ABX, LR or a mucus supplement reduced GC-Tx-induced osteoblast and osteocyte apoptosis. GC-Tx suppression of Wnt10b in bone was restored by the LR and high-molecular-weight polymer (MDY) treatments as well as microbiota depletion. Finally, we identified that bone-specific Wnt10b overexpression prevented GIO. Taken together, our data highlight the previously unappreciated involvement of the gut microbiota and intestinal barrier function in trabecular GIO pathogenesis (including Wnt10b suppression and osteoblast and osteocyte apoptosis) and identify the gut as a novel therapeutic target for preventing GIO. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
| | - Fraser Collins
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Naiomy Deliz Rios-Arce
- Department of Physiology, Michigan State University, East Lansing, MI, USA.,Comparative Medicine and Integrative Biology Program, Michigan State University, East Lansing, MI, USA
| | - Ho Jun Kang
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Laura Schaefer
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | | | - Ruma Raghuvanshi
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
| | - Robert A Quinn
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
| | - Robert Britton
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | | | - Laura R McCabe
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
63
|
Wang H, Liu D, Ji Y, Liu Y, Xu L, Guo Y. Dietary Supplementation of Black Rice Anthocyanin Extract Regulates Cholesterol Metabolism and Improves Gut Microbiota Dysbiosis in C57BL/6J Mice Fed a High-Fat and Cholesterol Diet. Mol Nutr Food Res 2020; 64:e1900876. [PMID: 32050056 DOI: 10.1002/mnfr.201900876] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 12/23/2019] [Indexed: 12/19/2022]
Abstract
SCOPE This study explores the beneficial effects of dietary supplementation of black rice anthocyanin extract (BRAE) on cholesterol metabolism and gut dysbiosis. METHODS AND RESULTS C57BL/6J mice are grouped into the normal chow diet group (NCD), the high-fat and the cholesterol diet group (HCD), and three treatment groups feeding HCD supplemented with various dosage of BRAE for 12 weeks. Results reveal that BRAE alleviates the increased body weight, serum triglyceride (TG), total cholesterol (TC), non-high-density lipoprotein cholesterol levels (non-HDL-C), and increased fecal sterols excretion and caecal short-chain fatty acids (SCFAs) concentration in HCD-induced hypercholesterolemic mice. Moreover, BRAE decreases hepatic TC content through the fundamental regulation of body energy balance gene, adenosine 5'-monophosphate activated protein kinase α (AMPKα). Meanwhile, BRAE improves the genes expression involved in cholesterol uptake and efflux, and preserves CYP7A1, ATP-binding cassette subfamily G member 5/8 mRNA expression, and the relative abundance of gut microbiota. Additionally, the antibiotic treatment experiment indicates that the beneficial effects of BRAE in reducing hypocholesterolemia risk largely depends on the gut microbiota homeostasis. CONCLUSION BRAE supplement could be a beneficial treatment option for preventing HCD-induced hypocholesterolemia and related metabolic syndromes.
Collapse
Affiliation(s)
- Hao Wang
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Dong Liu
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Yanglin Ji
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Yaojie Liu
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Lin Xu
- Pathology Department, Tianjin Municipal Public Security Hospital, Tianjin, 300042, China
| | - Yatu Guo
- Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Hospital, Tianjin Eye Institute, Tianjin, 300384, China
| |
Collapse
|
64
|
Kamphuis JBJ, Guiard B, Leveque M, Olier M, Jouanin I, Yvon S, Tondereau V, Rivière P, Guéraud F, Chevolleau S, Noguer-Meireles MH, Martin JF, Debrauwer L, Eutamène H, Theodorou V. Lactose and Fructo-oligosaccharides Increase Visceral Sensitivity in Mice via Glycation Processes, Increasing Mast Cell Density in Colonic Mucosa. Gastroenterology 2020; 158:652-663.e6. [PMID: 31711923 DOI: 10.1053/j.gastro.2019.10.037] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 10/28/2019] [Accepted: 10/31/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Irritable bowel syndrome (IBS) is characterized by abdominal pain, bloating, and erratic bowel habits. A diet low in fermentable oligosaccharides, disaccharides, monosaccharides, and polyols (FODMAPs) can reduce symptoms of IBS, possibly by reducing microbial fermentation products. We investigated whether ingestion of FODMAPs can induce IBS-like visceral hypersensitivity mediated by fermentation products of intestinal microbes in mice. METHODS C57Bl/6 mice were gavaged with lactose, with or without the antiglycation agent pyridoxamine, or saline (controls) daily for 3 weeks. A separate group of mice were fed a diet containing fructo-oligosaccharides, with or without pyridoxamine in drinking water, or a normal chow diet (controls) for 6 weeks. Feces were collected and analyzed by 16S ribosomal RNA gene sequencing and bacterial community analyses. Abdominal sensitivity was measured by electromyography and mechanical von Frey filament assays. Colon tissues were collected from some mice and analyzed by histology and immunofluorescence to quantify mast cells and expression of advanced glycosylation end-product specific receptor (AGER). RESULTS Mice gavaged with lactose or fed fructo-oligosaccharides had increased abdominal sensitivity compared with controls, associated with increased numbers of mast cells in colon and expression of the receptor for AGER in proximal colon epithelium. These effects were prevented by administration of pyridoxamine. Lactose and/or pyridoxamine did not induce significant alterations in the composition of the fecal microbiota. Mass spectrometric analysis of carbonyl compounds in fecal samples identified signatures associated with mice given lactose or fructo-oligosaccharides vs controls. CONCLUSIONS We found that oral administration of lactose or fructo-oligosaccharides to mice increases abdominal sensitivity, associated with increased numbers of mast cells in colon and expression of AGER; these can be prevented with an antiglycation agent. Lactose and/or pyridoxamine did not produce alterations in fecal microbiota of mice. Our findings indicate that preventing glycation reactions might reduce abdominal pain in patients with IBS with sensitivity to FODMAPs.
Collapse
Affiliation(s)
- Jasper B J Kamphuis
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Toxalim, UMR1331, INRAE/INP/UPS, Toulouse, France; Neurogastroenterology and Nutrition, Toxalim, Toulouse
| | - Bruno Guiard
- Centre de Recherches sur la Cognition Animale, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Mathilde Leveque
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Toxalim, UMR1331, INRAE/INP/UPS, Toulouse, France; Neurogastroenterology and Nutrition, Toxalim, Toulouse
| | - Maiwenn Olier
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Toxalim, UMR1331, INRAE/INP/UPS, Toulouse, France; Neurogastroenterology and Nutrition, Toxalim, Toulouse
| | - Isabelle Jouanin
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Toxalim, UMR1331, INRAE/INP/UPS, Toulouse, France; AXIOM Platform, MetaToul MetaboHUB, National Infrastructure for Metabolomics and Fluxomics, Toulouse, France
| | - Sophie Yvon
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Toxalim, UMR1331, INRAE/INP/UPS, Toulouse, France; Neurogastroenterology and Nutrition, Toxalim, Toulouse
| | - Valerie Tondereau
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Toxalim, UMR1331, INRAE/INP/UPS, Toulouse, France; Neurogastroenterology and Nutrition, Toxalim, Toulouse
| | - Pauline Rivière
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Toxalim, UMR1331, INRAE/INP/UPS, Toulouse, France; Neurogastroenterology and Nutrition, Toxalim, Toulouse
| | - Françoise Guéraud
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Toxalim, UMR1331, INRAE/INP/UPS, Toulouse, France; Prevention and Promotion of Carcinogenesis by Food team, Toxalim, Toulouse, France
| | - Sylvie Chevolleau
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Toxalim, UMR1331, INRAE/INP/UPS, Toulouse, France; AXIOM Platform, MetaToul MetaboHUB, National Infrastructure for Metabolomics and Fluxomics, Toulouse, France
| | - Maria-Helena Noguer-Meireles
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Toxalim, UMR1331, INRAE/INP/UPS, Toulouse, France; AXIOM Platform, MetaToul MetaboHUB, National Infrastructure for Metabolomics and Fluxomics, Toulouse, France
| | - Jean-François Martin
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Toxalim, UMR1331, INRAE/INP/UPS, Toulouse, France; AXIOM Platform, MetaToul MetaboHUB, National Infrastructure for Metabolomics and Fluxomics, Toulouse, France
| | - Laurent Debrauwer
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Toxalim, UMR1331, INRAE/INP/UPS, Toulouse, France; AXIOM Platform, MetaToul MetaboHUB, National Infrastructure for Metabolomics and Fluxomics, Toulouse, France
| | - Helene Eutamène
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Toxalim, UMR1331, INRAE/INP/UPS, Toulouse, France; Neurogastroenterology and Nutrition, Toxalim, Toulouse.
| | - Vassilia Theodorou
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Toxalim, UMR1331, INRAE/INP/UPS, Toulouse, France; Neurogastroenterology and Nutrition, Toxalim, Toulouse
| |
Collapse
|
65
|
Flores-Bastías O, Adriasola-Carrasco A, Karahanian E. Activation of Melanocortin-4 Receptor Inhibits Both Neuroinflammation Induced by Early Exposure to Ethanol and Subsequent Voluntary Alcohol Intake in Adulthood in Animal Models: Is BDNF the Key Mediator? Front Cell Neurosci 2020; 14:5. [PMID: 32063838 PMCID: PMC6997842 DOI: 10.3389/fncel.2020.00005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 01/10/2020] [Indexed: 12/31/2022] Open
Abstract
The concept that neuroinflammation induced by excessive alcohol intake in adolescence triggers brain mechanisms that perpetuate consumption has strengthened in recent years. The melanocortin system, composed of the melanocortin 4 receptor (MC4R) and its ligand α-melanocyte-stimulating hormone (α-MSH), has been implicated both in modulation of alcohol consumption and in ethanol-induced neuroinflammation decrease. Chronic alcohol consumption in adolescent rats causes a decrease in an α-MSH release by the hypothalamus, while the administration of synthetic agonists of MC4R causes a decrease in neuroinflammation and a decrease in voluntary alcohol consumption. However, the mechanism that connects the activation of MC4R with the decrease of both neuroinflammation and voluntary alcohol consumption has not been elucidated. Brain-derived neurotrophic factor (BDNF) has been implicated in alcohol drinking motivation, dependence and withdrawal, and its levels are reduced in alcoholics. Deficiencies in BDNF levels increased ethanol self-administration in rats. Further, BDNF triggers important anti-inflammatory effects in the brain, and this could be one of the mechanisms by which BDNF reduces chronic alcohol intake. Interestingly, MC4R signaling induces BDNF expression through the activation of the cAMP-responsive element-binding protein (CREB). We hypothesize that ethanol exposure during adolescence decreases the expression of α-MSH and hence MC4R signaling in the hippocampus, leading to a lower BDNF activity that causes dramatic changes in the brain (e.g., neuroinflammation and decreased neurogenesis) that predispose to maintain alcohol abuse until adulthood. The activation of MC4R either by α-MSH or by synthetic agonist peptides can induce the expression of BDNF, which would trigger several processes that lead to lower alcohol consumption.
Collapse
Affiliation(s)
- Osvaldo Flores-Bastías
- Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Santiago, Chile.,Research Center for the Study of Alcohol Drinking Behavior in Adolescents, Universidad Autónoma de Chile, Santiago, Chile
| | - Alfredo Adriasola-Carrasco
- Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Santiago, Chile
| | - Eduardo Karahanian
- Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Santiago, Chile.,Research Center for the Study of Alcohol Drinking Behavior in Adolescents, Universidad Autónoma de Chile, Santiago, Chile
| |
Collapse
|
66
|
Antibiotic Treatment Does Not Ameliorate the Metabolic Changes in Rats Presenting Dysbiosis After Consuming a High Fructose Diet. Nutrients 2020; 12:nu12010203. [PMID: 31941016 PMCID: PMC7019627 DOI: 10.3390/nu12010203] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 01/06/2020] [Accepted: 01/09/2020] [Indexed: 12/17/2022] Open
Abstract
High fructose consumption is one of the hallmarks of Western diets and has been found to induce MeS symptoms in parallel to gut microbial dysbiosis. However, the causality between those two is still elusive. Here, we studied whether a significant modification of gut microbial composition by antibiotics can influence the fructose-induced metabolic changes. Male Sprague-Dawley (SD) rats were divided into four groups including controls, controls + antibiotics, high fructose diet (HFrD, 60% fructose), HFrD + antibiotics (n = 7-8 in each group) for a period of 8-weeks. The high fructose diet increased blood pressure (BP), triglyceride (TG), fatty liver and the expression of hepatic genes related to lipogenesis, and fructose transport and metabolism. In addition, fructose changed the microbial composition and increased acetic and butyric acids in fecal samples but not in the blood. Antibiotic treatment significantly reduced microbial diversity and modified the microbial composition in the samples. However, minimal or no effect was seen in the metabolic phenotypes. In conclusion, high fructose consumption (60%) induced metabolic changes and dysbiosis in rats. However, antibiotic treatment did not reverse the metabolic phenotype. Therefore, the metabolic changes are probably independent of a specific microbiome profile.
Collapse
|
67
|
Jenkins SV, Robeson MS, Griffin RJ, Quick CM, Siegel ER, Cannon MJ, Vang KB, Dings RP. Gastrointestinal Tract Dysbiosis Enhances Distal Tumor Progression through Suppression of Leukocyte Trafficking. Cancer Res 2019; 79:5999-6009. [PMID: 31591154 PMCID: PMC6891208 DOI: 10.1158/0008-5472.can-18-4108] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 08/21/2019] [Accepted: 10/01/2019] [Indexed: 12/20/2022]
Abstract
The overall use of antibiotics has increased significantly in recent years. Besides fighting infections, antibiotics also alter the gut microbiota. Commensal bacteria in the gastrointestinal tract are crucial to maintain immune homeostasis, and microbial imbalance or dysbiosis affects disease susceptibility and progression. We hypothesized that antibiotic-induced dysbiosis of the gut microbiota would suppress cytokine profiles in the host, thereby leading to changes in the tumor microenvironment. The induced dysbiosis was characterized by alterations in bacterial abundance, composition, and diversity in our animal models. On the host side, antibiotic-induced dysbiosis caused elongated small intestines and ceca, and B16-F10 melanoma and Lewis lung carcinoma progressed more quickly than in control mice. Mechanistic studies revealed that this progression was mediated by suppressed TNFα levels, both locally and systemically, resulting in reduced expression of tumor endothelial adhesion molecules, particularly intercellular adhesion molecule-1 (ICAM-1) and a subsequent decrease in the number of activated and effector CD8+ T cells in the tumor. However, suppression of ICAM-1 or its binding site, the alpha subunit of lymphocyte function-associated antigen-1, was not seen in the spleen or thymus during dysbiosis. TNFα supplementation in dysbiotic mice was able to increase ICAM-1 expression and leukocyte trafficking into the tumor. Overall, these results demonstrate the importance of commensal bacteria in supporting anticancer immune surveillance, define an important role of tumor endothelial cells within this process, and suggest adverse consequences of antibiotics on cancer control. SIGNIFICANCE: Antibiotic-induced dysbiosis enhances distal tumor progression by altering host cytokine levels, resulting in suppression of tumor endothelial adhesion molecules and activated and effector CD8+ T cells in the tumor.
Collapse
Affiliation(s)
- Samir V. Jenkins
- Department of Radiation Oncology, University of Arkansas
for Medical Sciences, Little Rock, AR USA
| | - Michael S. Robeson
- Department of Biomedical Informatics, University of
Arkansas for Medical Sciences, Little Rock, AR USA
| | - Robert J. Griffin
- Department of Radiation Oncology, University of Arkansas
for Medical Sciences, Little Rock, AR USA
| | - Charles M. Quick
- Department of Pathology, University of Arkansas for Medical
Sciences, Little Rock, AR USA
| | - Eric R. Siegel
- Department of Biostatistics, University of Arkansas for
Medical Sciences, Little Rock, AR USA
| | - Martin J. Cannon
- Department of Microbiology and Immunology, University of
Arkansas for Medical Sciences, Little Rock, AR USA
| | - Kieng B. Vang
- Center for Integrative Nanotechnology Sciences, University
of Arkansas at Little Rock, Little Rock, AR USA
| | - Ruud P.M. Dings
- Department of Radiation Oncology, University of Arkansas
for Medical Sciences, Little Rock, AR USA,Corresponding Author: Ruud P.M.
Dings, 4301 W. Markham Street, Mail Slot #771, Little Rock, AR 72205.
Phone: +1 501 526 7876
| |
Collapse
|
68
|
A Simulated Microgravity Environment Causes a Sustained Defect in Epithelial Barrier Function. Sci Rep 2019; 9:17531. [PMID: 31772208 PMCID: PMC6879622 DOI: 10.1038/s41598-019-53862-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 10/31/2019] [Indexed: 02/06/2023] Open
Abstract
Intestinal epithelial cell (IEC) junctions constitute a robust barrier to invasion by viruses, bacteria and exposure to ingested agents. Previous studies showed that microgravity compromises the human immune system and increases enteropathogen virulence. However, the effects of microgravity on epithelial barrier function are poorly understood. The aims of this study were to identify if simulated microgravity alters intestinal epithelial barrier function (permeability), and susceptibility to barrier-disrupting agents. IECs (HT-29.cl19a) were cultured on microcarrier beads in simulated microgravity using a rotating wall vessel (RWV) for 18 days prior to seeding on semipermeable supports to measure ion flux (transepithelial electrical resistance (TER)) and FITC-dextran (FD4) permeability over 14 days. RWV cells showed delayed apical junction localization of the tight junction proteins, occludin and ZO-1. The alcohol metabolite, acetaldehyde, significantly decreased TER and reduced junctional ZO-1 localization, while increasing FD4 permeability in RWV cells compared with static, motion and flask control cells. In conclusion, simulated microgravity induced an underlying and sustained susceptibility to epithelial barrier disruption upon removal from the microgravity environment. This has implications for gastrointestinal homeostasis of astronauts in space, as well as their capability to withstand the effects of agents that compromise intestinal epithelial barrier function following return to Earth.
Collapse
|
69
|
Abstract
BACKGROUND Periprosthetic joint infection (PJI) is one of the most devastating complications of total joint arthroplasty. Given the mortality and morbidity associated with PJI and the challenges in treating it, there has been increased interest in risk factors that can be modified before surgery. In this study, we used a novel mouse model to consider the role of the gut microbiome as a risk factor for PJI. QUESTIONS/PURPOSES (1) Does the state of the gut microbiota before surgery influence the likelihood of developing an established infection in a mouse model of PJI? (2) How does the state of the gut microbiota before surgery influence the local and systemic response to the presence of an established infection in a mouse model of PJI? METHODS Male C57Bl/6 mice were divided into two groups: those with modified microbiome [INCREMENT]microbiome (n = 40) and untreated mice (n = 42). In [INCREMENT]microbiome mice, the gut flora were modified using oral neomycin and ampicillin from 4 weeks to 16 weeks of age. Mice received a titanium tibial implant to mimic a joint implant and a local inoculation of Staphylococcus aureus in the synovial space (10 colony forming units [CFUs]). The proportion of animals developing an established infection in each group was determined by CFU count. The local and systemic response to established infection was determined using CFU counts in surrounding joint tissues, analysis of gait, radiographs, body weight, serum markers of inflammation, and immune cell profiles and was compared with animals that received the inoculation but resisted infection. RESULTS A greater proportion of animals with disrupted gut microbiota had infection (29 of 40 [73%]) than did untreated animals (21 of 42 [50%]; odds ratio, 2.63, 95% CI, 1.04-6.61; p = 0.035). The immune response to established infection in mice with altered microbiota was muted; serum amyloid A, a marker of systemic infection in mice, was greater than in mice with disrupted gut microbiota with infection (689 µg/dL; range, 68-2437 µg/dL, p < 0.05); infection associated increases in monocytes and neutrophils in the spleen and local lymph node in untreated mice but not were not observed in mice with disrupted gut microbiota. CONCLUSIONS The findings from this in vivo mouse model suggest that the gut microbiota may influence susceptibility to PJI. CLINICAL RELEVANCE These preclinical findings support the idea that the state of the gut microbiome before surgery may influence the development of PJI and justify further preclinical and clinical studies to develop appropriate microbiome-based interventions.
Collapse
|
70
|
Abstract
Many studies have indicated that intestinal barrier dysfunction is the key mechanism of alcoholic liver disease (ALD). In this paper, we systematically review the causes of intestinal barrier dysfunction and the pathogenesis of ALD and discuss the treatment of intestinal barrier dysfunction.
Collapse
Affiliation(s)
- Zhao-Chun Chi
- Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao 266011, Shandong Province, China
| |
Collapse
|
71
|
Ezquer F, Quintanilla ME, Morales P, Santapau D, Ezquer M, Kogan MJ, Salas-Huenuleo E, Herrera-Marschitz M, Israel Y. Intranasal delivery of mesenchymal stem cell-derived exosomes reduces oxidative stress and markedly inhibits ethanol consumption and post-deprivation relapse drinking. Addict Biol 2019; 24:994-1007. [PMID: 30239077 DOI: 10.1111/adb.12675] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 07/19/2018] [Accepted: 07/30/2018] [Indexed: 01/01/2023]
Abstract
Chronic ethanol consumption leads to brain oxidative stress and neuroinflammation, conditions known to potentiate and perpetuate each other. Several studies have shown that neuroinflammation results in increases in chronic ethanol consumption. Recent reports showed that the intra-cerebroventricular administration of mesenchymal stem cells to rats consuming alcohol chronically markedly inhibited oxidative-stress, abolished neuroinflammation and greatly reduced chronic alcohol intake and post deprivation relapse-like alcohol intake. However, the intra-cerebroventricular administration of living cells is not suitable as a treatment of a chronic condition. The present study aimed at inhibiting ethanol intake by the non-invasive intranasal administration of human mesenchymal stem cell products: exosomes, microvesicles (40 to 150 nm) with marked antioxidant activity extruded from mesenchymal stem cells. The exosome membrane can fuse with the plasma membrane of cells in different tissues, thus delivering their content intracellularly. The study showed that the weekly intranasal administration of mesenchymal stem cell-derived exosomes to rats consuming alcohol chronically (1) inhibited their ethanol intake by 84 percent and blunted the relapse-like 'binge' drinking that follows an alcohol deprivation period and ethanol re-access. (2) Intranasally administered exosomes were found in the brain within 24 hours; (3) fully reversed both alcohol-induced hippocampal oxidative-stress, evidenced by a lower ratio of oxidized to reduced glutathione, and neuroinflammation, shown by a reduced astrocyte activation and microglial density; and (4) increased glutamate transporter GLT1 expression in nucleus accumbens, counteracting the inhibition of glutamate transporter activity, reportedly depressed under oxidative-stress conditions. Possible translational implications are envisaged.
Collapse
Affiliation(s)
- Fernando Ezquer
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana-Universidad del Desarrollo; Chile
| | - María Elena Quintanilla
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences; Faculty of Medicine, University of Chile; Chile
| | - Paola Morales
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences; Faculty of Medicine, University of Chile; Chile
- Department of Neuroscience; University of Chile; Chile
| | - Daniela Santapau
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana-Universidad del Desarrollo; Chile
| | - Marcelo Ezquer
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana-Universidad del Desarrollo; Chile
| | - Marcelo J. Kogan
- Faculty of Medicine, Department of Pharmacological and Toxicological Chemistry, Faculty of Chemical and Pharmaceutical Sciences; University of Chile; Chile
- Advanced Center for Chronic Diseases (ACCDis); Chile
| | - Edison Salas-Huenuleo
- Faculty of Medicine, Department of Pharmacological and Toxicological Chemistry, Faculty of Chemical and Pharmaceutical Sciences; University of Chile; Chile
- Advanced Center for Chronic Diseases (ACCDis); Chile
| | - Mario Herrera-Marschitz
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences; Faculty of Medicine, University of Chile; Chile
| | - Yedy Israel
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences; Faculty of Medicine, University of Chile; Chile
| |
Collapse
|
72
|
Waller K, James C, de Jong A, Blackmore L, Ma Y, Stagg A, Kelsell D, O'Dwyer M, Hutchins R, Alazawi W. ADAM17-Mediated Reduction in CD14 ++CD16 + Monocytes ex vivo and Reduction in Intermediate Monocytes With Immune Paresis in Acute Pancreatitis and Acute Alcoholic Hepatitis. Front Immunol 2019; 10:1902. [PMID: 31507587 PMCID: PMC6718469 DOI: 10.3389/fimmu.2019.01902] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 07/26/2019] [Indexed: 12/16/2022] Open
Abstract
Impaired immune responses and increased susceptibility to infection characterize acute inflammatory conditions such as pancreatitis and alcoholic hepatitis and are major causes of morbidity and mortality. However, the mechanisms that drive this apparent immune paresis remain poorly understood. Monocytes mediate host responses to damage and pathogens in health and disease, and three subsets of monocytes have been defined based on CD14 and CD16 expression. We sought to determine the changes in monocyte subsets in acute pancreatitis (AP) and acute alcoholic hepatitis (AAH), together with functional consequences and mechanisms that underlie this change. Peripheral blood mononuclear cells (PBMCs) from patients with AP or AAH were compared with healthy controls. Monocyte subsets were defined by HLA-DR, CD14, and CD16 expression. Changes in surface and intracellular protein expression and phosphorylation were determined by flow cytometry. Phenotype and function were assessed following stimulation with lipopolysaccharide (LPS) or other agonists in the presence of specific inhibitors of TNFα and a disintegrin and metalloproteinase 17 (ADAM17). Patients with AP and AAH had reduced CD14++CD16+ intermediate monocytes compared to controls. Reduction of intermediate monocytes was recapitulated ex vivo by stimulating healthy control PBMCs with Toll-like receptor (TLR) agonists LPS, flagellin or polyinosilic:polycytidylic acid (poly I:C). Stimulation caused shedding of CD14 and CD16, which could be reversed using the ADAM17 inhibitor, TMI005 but not direct inhibitors of TNFα, a known ADAM17-target. Culturing PBMCs from healthy controls resulted in expansion of intermediate monocytes, which did not occur when LPS was in the culture medium. Cultured intermediate monocytes showed reduced expression of CX3CR1, CCR2, TLR4, and TLR5. We found reduced migratory responses, intracellular signaling and pro-inflammatory cytokine production, and increased expression of IL-10. Stimulation with TLR agonists results in ADAM17-mediated shedding of phenotypic markers from CD16+ monocytes, leading to apparent “loss” of intermediate monocytes. Reduction in CD14++CD16− monocytes and increased CD14++CD16+ is associated with altered responses in functional assays ex vivo. Patients with AP and AAH had reduced proportions of CD14++CD16+ monocytes and reduced phosphorylation of NFκB and IL-6 production in response to bacterial LPS. Together, these processes may contribute to the susceptibility to infection observed in AP and AAH.
Collapse
Affiliation(s)
- Kathryn Waller
- Blizard Institute, Queen Mary University of London, London, United Kingdom
| | - Charlotte James
- Blizard Institute, Queen Mary University of London, London, United Kingdom
| | - Anja de Jong
- Blizard Institute, Queen Mary University of London, London, United Kingdom
| | - Laura Blackmore
- Institute of Liver Studies and Transplantation, King's College London, London, United Kingdom
| | - Yun Ma
- Institute of Liver Studies and Transplantation, King's College London, London, United Kingdom
| | - Andrew Stagg
- Blizard Institute, Queen Mary University of London, London, United Kingdom
| | - David Kelsell
- Blizard Institute, Queen Mary University of London, London, United Kingdom
| | | | - Robert Hutchins
- Hepatopancreaticobiliary Unit, Barts Health NHS Trust, London, United Kingdom
| | - William Alazawi
- Blizard Institute, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
73
|
Yvon S, Schwebel L, Belahcen L, Tormo H, Peter M, Haimoud-Lekhal DA, Eutamene H, Jard G. Effects of thermized donkey milk with lysozyme activity on altered gut barrier in mice exposed to water-avoidance stress. J Dairy Sci 2019; 102:7697-7706. [PMID: 31326167 DOI: 10.3168/jds.2019-16642] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 05/16/2019] [Indexed: 01/26/2023]
Abstract
Nutrition plays a crucial role in human gut health through the improvement of gut barrier functionality. Donkey milk represents an interesting source of natural antimicrobial factors such as lysozyme. Recently, anti-inflammatory properties of donkey milk lysozyme activity were described in a mouse model of ileitis. The current increase of donkey milk consumption highlights the necessity to propose a healthy milk compliant with microbiological standards. This study aims to define a heat treatment of donkey milk, retaining its high lysozyme activity, and to evaluate its beneficial effects on a gut barrier impairment model due to chronic stress in mice. To perform this experiment, samples of raw donkey milk were collected in 15 distinct French farms. Microbiological analysis and lysozyme content and activity were evaluated for each sample. Then, several heat treatments were carried out to define a time and temperature combination that allowed for both a reduction in the number of total micro-organisms, increasing the shelf-life of the product, and preservation of lysozyme activity. The beneficial effect of heated donkey milk on the gut barrier of mice was evaluated and compared with raw donkey milk. We found that samples of raw donkey milk showed low total mesophilic microbial counts, and no pathogens were detected. Among the different heat-treatment procedures tested, a 2-min, 72°C combination was determined to be the most optimal time and temperature combination to preserve lysozyme activity and increase the shelf-life of donkey milk. Oral administration of this heat-treated donkey milk in mice counteracted chronic stress-induced intestinal damage, illustrated by gut hyper-permeability and low-grade inflammation, similar to raw donkey milk. We have demonstrated for the first time that oral intervention with donkey milk, optimally heat-treated to retain enzymatic lysozyme activity, improves intestinal barrier damage linked to psychological stress in mice.
Collapse
Affiliation(s)
- Sophie Yvon
- Unité Mixte de Recherche 1331 Toxalim, Institut National de la Recherche Agronomique, Institut National Polytechnique-Ecole Nationale Vétérinaire de Toulouse, Institut National Polytechnique-Purpan, Université de Toulouse, Neuro-Gastroenterology and Nutrition Group, Toulouse, France
| | - Lauriane Schwebel
- Unité Mixte de Recherche 1331 Toxalim, Institut National de la Recherche Agronomique, Institut National Polytechnique-Ecole Nationale Vétérinaire de Toulouse, Institut National Polytechnique-Purpan, Université de Toulouse, Neuro-Gastroenterology and Nutrition Group, Toulouse, France
| | - Loubnah Belahcen
- Equipe Agromolécules et Agroalimentaire, Université de Toulouse, Institut National Polytechnique-Purpan, Toulouse, France
| | - Helene Tormo
- Equipe Agromolécules et Agroalimentaire, Université de Toulouse, Institut National Polytechnique-Purpan, Toulouse, France
| | - Magali Peter
- Equipe Agromolécules et Agroalimentaire, Université de Toulouse, Institut National Polytechnique-Purpan, Toulouse, France
| | - Djamila Ali Haimoud-Lekhal
- Unité Mixte de Recherche 1388 Génétique, Physiologie et Systèmes d'Elevage, Institut National de la Recherche Agronomique, Institut National Polytechnique-Purpan, Toulouse, France
| | - Hélène Eutamene
- Unité Mixte de Recherche 1331 Toxalim, Institut National de la Recherche Agronomique, Institut National Polytechnique-Ecole Nationale Vétérinaire de Toulouse, Institut National Polytechnique-Purpan, Université de Toulouse, Neuro-Gastroenterology and Nutrition Group, Toulouse, France.
| | - Gwenaelle Jard
- Equipe Agromolécules et Agroalimentaire, Université de Toulouse, Institut National Polytechnique-Purpan, Toulouse, France
| |
Collapse
|
74
|
Quintanilla ME, Ezquer F, Morales P, Santapau D, Berríos-Cárcamo P, Ezquer M, Herrera-Marschitz M, Israel Y. Intranasal mesenchymal stem cell secretome administration markedly inhibits alcohol and nicotine self-administration and blocks relapse-intake: mechanism and translational options. Stem Cell Res Ther 2019; 10:205. [PMID: 31286996 PMCID: PMC6615104 DOI: 10.1186/s13287-019-1304-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 04/30/2019] [Accepted: 06/17/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Chronic consumption of most drugs of abuse leads to brain oxidative stress and neuroinflammation, which inhibit the glutamate transporter GLT-1, proposed to perpetuate drug intake. The present study aimed at inhibiting chronic ethanol and nicotine self-administration and relapse by the non-invasive intranasal administration of antioxidant and anti-inflammatory secretome generated by adipose tissue-derived activated mesenchymal stem cells. The anti-addiction mechanism of stem cell secretome is also addressed. METHODS Rats bred for their alcohol preference ingested alcohol chronically or were trained to self-administer nicotine. Secretome of human adipose tissue-derived activated mesenchymal stem cells was administered intranasally to animals, both (i) chronically consuming alcohol or nicotine and (ii) during a protracted deprivation before a drug re-access leading to relapse intake. RESULTS The intranasal administration of secretome derived from activated mesenchymal stem cells inhibited chronic self-administration of ethanol or nicotine by 85% and 75%, respectively. Secretome administration further inhibited by 85-90% the relapse "binge" intake that occurs after a protracted drug deprivation followed by a 60-min drug re-access. Secretome administration fully abolished the oxidative stress induced by chronic ethanol or nicotine self-administration, shown by the normalization of the hippocampal oxidized/reduced glutathione ratio, and the neuroinflammation determined by astrocyte and microglial immunofluorescence. Knockdown of the glutamate transporter GLT-1 by the intracerebral administration of an antisense oligonucleotide fully abolished the inhibitory effect of the secretome on ethanol and nicotine intake. CONCLUSIONS The non-invasive intranasal administration of secretome generated by human adipose tissue-derived activated mesenchymal stem cells markedly inhibits alcohol and nicotine self-administration, an effect mediated by the glutamate GLT-1 transporter. Translational implications are envisioned.
Collapse
Affiliation(s)
- María Elena Quintanilla
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Santiago, Chile
| | - Fernando Ezquer
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana-Universidad del Desarrollo, Av. Las Condes 12438, Lo Barnechea, 7710162, Santiago, RM, Chile.
| | - Paola Morales
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Santiago, Chile
- Department of Neuroscience, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Daniela Santapau
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana-Universidad del Desarrollo, Av. Las Condes 12438, Lo Barnechea, 7710162, Santiago, RM, Chile
| | - Pablo Berríos-Cárcamo
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana-Universidad del Desarrollo, Av. Las Condes 12438, Lo Barnechea, 7710162, Santiago, RM, Chile
| | - Marcelo Ezquer
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana-Universidad del Desarrollo, Av. Las Condes 12438, Lo Barnechea, 7710162, Santiago, RM, Chile
| | - Mario Herrera-Marschitz
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Santiago, Chile
| | - Yedy Israel
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Santiago, Chile
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
75
|
Zhou YF, Zhou YJ, Ye FZ, Liu WY, Zheng MH. A Novel Use of Model for End-Stage Liver Disease (MELD) Score in Guiding Therapeutic Antibiotics Choice for Critically Ill Cirrhotic Patients. Med Sci Monit 2019; 25:5005-5014. [PMID: 31278890 PMCID: PMC6628631 DOI: 10.12659/msm.914409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 03/10/2019] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Inappropriate use of antibiotics results in antimicrobial resistance and dysbacteriosis. Among critically ill cirrhotic patients, consensus regarding the most optimal prescription strategy for antibiotics use has not been achieved. For these patients, the score for end-stage liver disease (MELD) demonstrated its value in predicting prognosis of cirrhosis. This study investigated use of the MELD score to guide antibiotics choice. MATERIAL AND METHODS We enrolled 1250 patients with cirrhosis. We collected patient information, including antibiotics administration. Linear regression analyses were performed to determine independent predictors of antibiotic administration. Survival curves were constructed based on Cox regression models. Cox proportional hazard models were used to calculate the hazard ratio, shown by forest plots. RESULTS The population was equally stratified into 4 groups based on the MELD score (Q1: MELD <10; Q2: 10≤ MELD <17; Q3: 17≤ MELD <26; Q4: 26≤ MELD). In Q1, all the HR (hazard ratio) related to the duration of antibiotics use demonstrated no statistical significance. In Q2, the HR related to the duration of antibiotics use revealed a successive decrease. In Q3, the HR showed statistical significance only with a duration of antibiotics use of 7 days or more. In Q4, all the HR were statistically significant. As for categories of antibiotics use, whatever the MELD score was, the HR continued to increase with ascending categories. CONCLUSIONS For low MELD score patients (MELD <17), changing the duration of antibiotics use was not associated with a better prognosis. For high MELD score patients (MELD ≥17), longer duration of antibiotics use was associated with a reduction in mortality. Whatever the MELD score was, an increase of number of antibiotic categories was positively associat ed with poor prognosis.
Collapse
Affiliation(s)
- Yi-Fan Zhou
- NAFLD Research Center, Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China
- First Clinical Medical Sciences School, Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China
| | - Yu-Jie Zhou
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, P.R. China
| | - Fang-Zhou Ye
- NAFLD Research Center, Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China
- First Clinical Medical Sciences School, Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China
| | - Wen-Yue Liu
- Department of Endocrinology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China
| | - Ming-Hua Zheng
- NAFLD Research Center, Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China
- Institute of Hepatology, Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China
| |
Collapse
|
76
|
Li T, Li H, Li W, Chen S, Feng T, Jiao W, Wu C, Dong J, Li Y, Li S, Feng M, Wei X. Interleukin-37 sensitize the elderly type 2 diabetic patients to insulin therapy through suppressing the gut microbiota dysbiosis. Mol Immunol 2019; 112:322-329. [PMID: 31238287 DOI: 10.1016/j.molimm.2019.06.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 06/12/2019] [Accepted: 06/14/2019] [Indexed: 12/30/2022]
Abstract
OBJECTIVE The morbidity and prevalence of type 2 diabetes mellitus (DM) are increasing in the elderly population. Interleukin 37 (IL-37) play important roles in anti-inflammatory and anti-bacteria immune responses, but its role in the development of type 2 DM in the elderly is unclear. Therefore, we investigated whether IL-37 is associated with type 2 DM in the elderly and the underlying mechanism. METHODS Hospitalized patients (aged 65-95 years) with recently diagnosed type 2 diabetes mellitus were studied retrospectively and compared with healthy subjects without glucose metabolism abnormalities. A diabetic mouse model was established by feeding ob/ob mice (C57BL/6) a high-fat, carbohydrate-free diet. Plasma glucose and insulin levels were determined by glucose oxidase assay and radioimmunoassay, respectively. The IL-37 expression level was determined by real-time PCR, western blot and ELISA (Enzyme-linked immunoassay). RESULTS Statistic analysis showed that the IL-37 level was significantly associated with type 2 DM and insulin resistance in the elderly. The patients were then divided into insulin therapy sensitive and resistant group according to their response to insulin therapy. Data showed that the IL-37 was highly expressed in the insulin therapy sensitive group. And this was related to the less severe gut microbiota dysbiosis. In the mice model, overexpressing the IL-37 could suppress the gut microbiota dysbiosis and also the diabetes development. CONCLUSION Thus our results showed that higher IL-37 was associated with increased insulin sensitive in elderly type 2 DM patients through suppressing the gut microbiota dysbiosis.
Collapse
Affiliation(s)
- Tianyi Li
- Department of Elderly Endocrinology, The First Affiliated Hospital of Zhengzhou University, 450000, Zhengzhou, Henan Province, PR China.
| | - Hua Li
- Department of Elderly Endocrinology, The First Affiliated Hospital of Zhengzhou University, 450000, Zhengzhou, Henan Province, PR China.
| | - Weifang Li
- Department of Elderly Endocrinology, The First Affiliated Hospital of Zhengzhou University, 450000, Zhengzhou, Henan Province, PR China.
| | - Sufang Chen
- Department of Elderly Endocrinology, The First Affiliated Hospital of Zhengzhou University, 450000, Zhengzhou, Henan Province, PR China.
| | - Tao Feng
- Department of Elderly Endocrinology, The First Affiliated Hospital of Zhengzhou University, 450000, Zhengzhou, Henan Province, PR China.
| | - Wenjun Jiao
- Department of Elderly Endocrinology, The First Affiliated Hospital of Zhengzhou University, 450000, Zhengzhou, Henan Province, PR China.
| | - Cong Wu
- Department of Elderly Endocrinology, The First Affiliated Hospital of Zhengzhou University, 450000, Zhengzhou, Henan Province, PR China.
| | - Jie Dong
- Department of Elderly Endocrinology, The First Affiliated Hospital of Zhengzhou University, 450000, Zhengzhou, Henan Province, PR China.
| | - Yuanyuan Li
- Department of Elderly Endocrinology, The First Affiliated Hospital of Zhengzhou University, 450000, Zhengzhou, Henan Province, PR China.
| | - Sujun Li
- Department of Elderly Endocrinology, The First Affiliated Hospital of Zhengzhou University, 450000, Zhengzhou, Henan Province, PR China.
| | - Ming Feng
- Department of Elderly Endocrinology, The First Affiliated Hospital of Zhengzhou University, 450000, Zhengzhou, Henan Province, PR China.
| | - Xiaowen Wei
- Department of Elderly Endocrinology, The First Affiliated Hospital of Zhengzhou University, 450000, Zhengzhou, Henan Province, PR China.
| |
Collapse
|
77
|
González-Arancibia C, Urrutia-Piñones J, Illanes-González J, Martinez-Pinto J, Sotomayor-Zárate R, Julio-Pieper M, Bravo JA. Do your gut microbes affect your brain dopamine? Psychopharmacology (Berl) 2019; 236:1611-1622. [PMID: 31098656 DOI: 10.1007/s00213-019-05265-5] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 04/26/2019] [Indexed: 02/08/2023]
Abstract
Increasing evidence shows changes in gut microbiota composition in association with psychiatric disorders, including anxiety and depression. Moreover, it has been reported that perturbations in gut microbe diversity and richness influence serotonergic, GABAergic, noradrenergic, and dopaminergic neurotransmission. Among these, dopamine is regarded as a main regulator of cognitive functions such as decision making, attention, memory, motivation, and reward. In this work, we will highlight findings that link alterations in intestinal microbiota and dopaminergic neurotransmission, with a particular emphasis on the mesocorticolimbic circuit, which is involved in reward to natural reinforcers, as well as abuse substances. For this, we reviewed evidence from studies carried out on germ-free animals, or in rodents subjected to intestinal dysbiosis using antibiotics, and also through the use of probiotics. All this evidence strongly supports that the microbiota-gut-brain axis is key to the physiopathology of several neuropsychiatric disorders involving those where dopaminergic neurotransmission is compromised. In addition, the gut microbiota appears as a key player when it comes to proposing novel strategies to the treatment of these psychiatric conditions.
Collapse
Affiliation(s)
- Camila González-Arancibia
- Grupo de NeuroGastroBioquímica, Laboratorio de Química Biológica y Bioquímica de Sistemas, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Av. Universidad 330, Curauma, Valparaíso, Región de Valparaíso, Chile.,Laboratorio de Neuroquímica y Neurofarmacología, Centro de Neurobiología y Fisiopatología Integrativa, Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile.,Programa de Doctorado en Ciencias mención Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Jocelyn Urrutia-Piñones
- Grupo de NeuroGastroBioquímica, Laboratorio de Química Biológica y Bioquímica de Sistemas, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Av. Universidad 330, Curauma, Valparaíso, Región de Valparaíso, Chile.,Programa de Doctorado en Ciencias mención Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Javiera Illanes-González
- Grupo de NeuroGastroBioquímica, Laboratorio de Química Biológica y Bioquímica de Sistemas, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Av. Universidad 330, Curauma, Valparaíso, Región de Valparaíso, Chile.,Programa de Doctorado en Ciencias mención Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Jonathan Martinez-Pinto
- Laboratorio de Neuroquímica y Neurofarmacología, Centro de Neurobiología y Fisiopatología Integrativa, Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Ramón Sotomayor-Zárate
- Laboratorio de Neuroquímica y Neurofarmacología, Centro de Neurobiología y Fisiopatología Integrativa, Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Marcela Julio-Pieper
- Grupo de NeuroGastroBioquímica, Laboratorio de Química Biológica y Bioquímica de Sistemas, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Av. Universidad 330, Curauma, Valparaíso, Región de Valparaíso, Chile
| | - Javier A Bravo
- Grupo de NeuroGastroBioquímica, Laboratorio de Química Biológica y Bioquímica de Sistemas, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Av. Universidad 330, Curauma, Valparaíso, Región de Valparaíso, Chile.
| |
Collapse
|
78
|
Schepper JD, Collins F, Rios-Arce ND, Raehtz S, Schaefer L, Gardinier JD, Britton R, Parameswaran N, McCabe LR. Probiotic Lactobacillus reuteri Prevents Postantibiotic Bone Loss by Reducing Intestinal Dysbiosis and Preventing Barrier Disruption. J Bone Miner Res 2019; 34:681-698. [PMID: 30690795 PMCID: PMC6557403 DOI: 10.1002/jbmr.3635] [Citation(s) in RCA: 127] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 10/24/2018] [Accepted: 11/11/2018] [Indexed: 12/23/2022]
Abstract
Antibiotic treatment, commonly prescribed for bacterial infections, depletes and subsequently causes long-term alterations in intestinal microbiota composition. Knowing the importance of the microbiome in the regulation of bone density, we investigated the effect of postantibiotic treatment on gut and bone health. Intestinal microbiome repopulation at 4-weeks postantibiotic treatment resulted in an increase in the Firmicutes:Bacteroidetes ratio, increased intestinal permeability, and notably reduced femoral trabecular bone volume (approximately 30%, p < 0.01). Treatment with a mucus supplement (a high-molecular-weight polymer, MDY-1001 [MDY]) prevented the postantibiotic-induced barrier break as well as bone loss, indicating a mechanistic link between increased intestinal permeability and bone loss. A link between the microbiome composition and bone density was demonstrated by supplementing the mice with probiotic bacteria. Specifically, Lactobacillus reuteri, but not Lactobacillus rhamnosus GG or nonpathogenic Escherichia coli, reduced the postantibiotic elevation of the Firmicutes:Bacteroidetes ratio and prevented femoral and vertebral trabecular bone loss. Consistent with causing bone loss, postantibiotic-induced dysbiosis decreased osteoblast and increased osteoclast activities, changes that were prevented by both L. reuteri and MDY. These data underscore the importance of microbial dysbiosis in the regulation of intestinal permeability and bone health, as well as identify L. reuteri and MDY as novel therapies for preventing these adverse effects. © 2018 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
| | - Fraser Collins
- Department of Physiology, Michigan State University, East Lansing, Michigan
| | - Naiomy Deliz Rios-Arce
- Department of Physiology, Michigan State University, East Lansing, Michigan
- Comparative Medicine and Integrative Biology Program, Michigan State University, East Lansing, Michigan
| | - Sandi Raehtz
- Department of Physiology, Michigan State University, East Lansing, Michigan
| | - Laura Schaefer
- Department of Molecular Virology and Microbiology, Baylor College of Medicine
| | | | - Robert Britton
- Department of Molecular Virology and Microbiology, Baylor College of Medicine
| | - Narayanan Parameswaran
- Department of Physiology, Michigan State University, East Lansing, Michigan
- equal contribution and co-senior authors
| | - Laura R McCabe
- Department of Physiology, Michigan State University, East Lansing, Michigan
- equal contribution and co-senior authors
| |
Collapse
|
79
|
Scherf KA, Lindenau AC, Valentini L, Collado MC, García-Mantrana I, Christensen M, Tomsitz D, Kugler C, Biedermann T, Brockow K. Cofactors of wheat-dependent exercise-induced anaphylaxis do not increase highly individual gliadin absorption in healthy volunteers. Clin Transl Allergy 2019; 9:19. [PMID: 30962874 PMCID: PMC6432753 DOI: 10.1186/s13601-019-0260-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 03/11/2019] [Indexed: 12/12/2022] Open
Abstract
Background In wheat-dependent exercise-induced anaphylaxis (WDEIA), cofactors such as exercise, acetylsalicylic acid (ASA), alcohol or unfavorable climatic conditions are required to elicit a reaction to wheat products. The mechanism of action of these cofactors is unknown, but an increase of gliadin absorption has been speculated. Our objectives were to study gliadin absorption with and without cofactors and to correlate plasma gliadin levels with factors influencing protein absorption in healthy volunteers.
Methods Twelve healthy probands (six males, six females; aged 20–56 years) ingested 32 g of gluten without any cofactor or in combination with cofactors aerobic and anaerobic exercise, ASA, alcohol and pantoprazole. Gliadin serum levels were measured up to 120 min afterwards and the intestinal barrier function protein zonulin in stool was collected before and after the procedure; both were measured by ELISA. Stool microbiota profile was obtained by 16S gene sequencing.
Results Within 15 min after gluten intake, gliadin concentrations in blood serum increased from baseline in all subjects reaching highly variable peak levels after 15–90 min. Addition of cofactors did not lead to substantially higher gliadin levels, although variability of levels was higher with differences between individuals (p < 0.001) and increased levels at later time points. Zonulin levels in stool were associated neither with addition of cofactors nor with peak gliadin concentrations. There were no differences in gut microbiota between the different interventions, although the composition of microbiota (p < 0.001) and the redundancy discriminant analysis (p < 0.007) differed in probands with low versus high stool zonulin levels. Conclusion The adsorption of gliadin in the gut in healthy volunteers is less dependent on cofactors than has been hypothesized. Patients with WDEIA may have a predisposition needed for the additional effect of cofactors, e.g., hyperresponsive or damaged intestinal epithelium. Alternatively, other mechanisms, such as cofactor-induced blood flow redistribution, increased activity of tissue transglutaminase, or increases in plasma osmolality and acidosis inducing basophil and mast cell histamine release may play the major role in WDEIA.
Collapse
Affiliation(s)
- Katharina Anne Scherf
- 1Leibniz-Institute for Food Systems Biology, Technical University of Munich, Lise-Meitner-Strasse 34, 85354 Freising, Germany
| | - Ann-Christin Lindenau
- 2Department of Agriculture and Food Sciences, Section of Dietetics, University of Applied Sciences Neubrandenburg, Brodaer Str. 2, 17033 Neubrandenburg, Germany
| | - Luzia Valentini
- 2Department of Agriculture and Food Sciences, Section of Dietetics, University of Applied Sciences Neubrandenburg, Brodaer Str. 2, 17033 Neubrandenburg, Germany
| | - Maria Carmen Collado
- 3Department of Biotechnology, Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Av. Catedrático Agustín Escardino 7, 46980 Valencia, Spain
| | - Izaskun García-Mantrana
- 3Department of Biotechnology, Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Av. Catedrático Agustín Escardino 7, 46980 Valencia, Spain
| | - Morten Christensen
- 4Department of Dermatology and Allergy Centre, Odense Research Center for Anaphylaxis (ORCA), Odense University Hospital, 5000 Odense, Denmark
| | - Dirk Tomsitz
- 5Department of Dermatology and Allergy Biederstein, Technical University of Munich, Biedersteiner Strasse 29, 80802 Munich, Germany
| | - Claudia Kugler
- 5Department of Dermatology and Allergy Biederstein, Technical University of Munich, Biedersteiner Strasse 29, 80802 Munich, Germany
| | - Tilo Biedermann
- 5Department of Dermatology and Allergy Biederstein, Technical University of Munich, Biedersteiner Strasse 29, 80802 Munich, Germany
| | - Knut Brockow
- 5Department of Dermatology and Allergy Biederstein, Technical University of Munich, Biedersteiner Strasse 29, 80802 Munich, Germany
| |
Collapse
|
80
|
Gene and cell therapy on the acquisition and relapse-like binge drinking in a model of alcoholism: translational options. Gene Ther 2019; 26:407-417. [DOI: 10.1038/s41434-019-0064-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 02/06/2019] [Accepted: 02/11/2019] [Indexed: 12/11/2022]
|
81
|
Liang Y, Zhan J, Liu D, Luo M, Han J, Liu X, Liu C, Cheng Z, Zhou Z, Wang P. Organophosphorus pesticide chlorpyrifos intake promotes obesity and insulin resistance through impacting gut and gut microbiota. MICROBIOME 2019; 7:19. [PMID: 30744700 PMCID: PMC6371608 DOI: 10.1186/s40168-019-0635-4] [Citation(s) in RCA: 157] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 01/29/2019] [Indexed: 05/17/2023]
Abstract
BACKGROUND Disruption of the gut microbiota homeostasis may induce low-grade inflammation leading to obesity-associated diseases. A major protective mechanism is to use the multi-layered mucus structures to keep a safe distance between gut epithelial cells and microbiota. To investigate whether pesticides would induce insulin resistance/obesity through interfering with mucus-bacterial interactions, we conducted a study to determine how long-term exposure to chlorpyrifos affected C57Bl/6 and CD-1 (ICR) mice fed high- or normal-fat diets. To further investigate the effects of chlorpyrifos-altered microbiota, antibiotic treatment and microbiota transplantation experiments were conducted. RESULTS The results showed that chlorpyrifos caused broken integrity of the gut barrier, leading to increased lipopolysaccharide entry into the body and finally low-grade inflammation, while genetic background and diet pattern have limited influence on the chlorpyrifos-induced results. Moreover, the mice given chlorpyrifos-altered microbiota had gained more fat and lower insulin sensitivity. CONCLUSIONS Our results suggest that widespread use of pesticides may contribute to the worldwide epidemic of inflammation-related diseases.
Collapse
Affiliation(s)
- Yiran Liang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, People's Republic of China
- College of Chemistry and Biological Engineering, University of Science and Technology Beijing, No. 30, Xueyuan Road, Beijing, 100083, People's Republic of China
| | - Jing Zhan
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, People's Republic of China
| | - Donghui Liu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, People's Republic of China
| | - Mai Luo
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, People's Republic of China
| | - Jiajun Han
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, People's Republic of China
| | - Xueke Liu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, People's Republic of China
| | - Chang Liu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, People's Republic of China
| | - Zheng Cheng
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, People's Republic of China
| | - Zhiqiang Zhou
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, People's Republic of China
| | - Peng Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, People's Republic of China.
| |
Collapse
|
82
|
Jiang JW, Chen XH, Ren Z, Zheng SS. Gut microbial dysbiosis associates hepatocellular carcinoma via the gut-liver axis. Hepatobiliary Pancreat Dis Int 2019; 18:19-27. [PMID: 30527903 DOI: 10.1016/j.hbpd.2018.11.002] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 09/28/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is one of the most common malignancies in the world. Gut microbiota has been demonstrated to play a critical role in liver inflammation, chronic fibrosis, liver cirrhosis, and HCC development through the gut-liver axis. DATA SOURCES Recently there have been several innovative studies investigating gut microbial dysbiosis-mediated enhancement of HCC through the gut-liver axis. Literatures from January 1998 to January 2018 were searched in the PubMed database using the keywords "gut microbiota" and "hepatocellular carcinoma" or "liver cancer", and the results of experimental and clinical studies were analyzed. RESULTS Gut microbial dysbiosis accompanies the progression of alcoholic liver disease, non-alcoholic fatty liver disease and liver cirrhosis, and promotes HCC progression in an experimental mouse model. The immune system and key factors such as Toll-like receptor 4 are involved in the process. There is evidence for gut microbial dysbiosis in hepatitis virus-related HCC patients. CONCLUSIONS Gut microbial dysbiosis is closely associated with hepatic inflammation disease and HCC through the gut-liver axis. With the enhanced understanding of the interactions between gut microbiota and liver through the gut-liver axis, new treatment strategies for HCC are being developed.
Collapse
Affiliation(s)
- Jian-Wen Jiang
- Department of Hepatobiliary and Pancreatic Surgery, Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, First Affiliated Hospital, Zhejiang University School of Medicine, #79 Qingchun Road, Hangzhou 310003, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou 310003, China; Health Management Center, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Xin-Hua Chen
- Department of Hepatobiliary and Pancreatic Surgery, Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, First Affiliated Hospital, Zhejiang University School of Medicine, #79 Qingchun Road, Hangzhou 310003, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou 310003, China
| | - Zhigang Ren
- Department of Infectious Disease, Precision Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Shu-Sen Zheng
- Department of Hepatobiliary and Pancreatic Surgery, Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, First Affiliated Hospital, Zhejiang University School of Medicine, #79 Qingchun Road, Hangzhou 310003, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou 310003, China.
| |
Collapse
|
83
|
Ezquer F, Quintanilla ME, Morales P, Ezquer M, Lespay-Rebolledo C, Herrera-Marschitz M, Israel Y. Activated mesenchymal stem cell administration inhibits chronic alcohol drinking and suppresses relapse-like drinking in high-alcohol drinker rats. Addict Biol 2019; 24:17-27. [PMID: 29044813 DOI: 10.1111/adb.12572] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 09/12/2017] [Accepted: 09/14/2017] [Indexed: 12/26/2022]
Abstract
Neuroinflammation has been reported to follow chronic ethanol intake and may perpetuate alcohol consumption. Present studies determined the effect of human mesenchymal stem cells (hMSCs), known for their anti-inflammatory action, on chronic ethanol intake and relapse-like ethanol intake in a post-deprivation condition. Rats were allowed 12-17 weeks of chronic voluntary ethanol (10% and 20% v/v) intake, after which a single dose of activated hMSCs (5 × 105 ) was injected into a brain lateral ventricle. Control animals were administered vehicle. After assessing the effect of hMSCs on chronic ethanol intake for 1 week, animals were deprived of ethanol for 2 weeks and thereafter an ethanol re-access of 60 min was allowed to determine relapse-like intake. A single administration of activated hMSCs inhibited chronic alcohol consumption by 70% (P < 0.001), an effect seen within the first 24 hours of hMSCs administration, and reduced relapse-like drinking by 80% (P < 0.001). In the relapse-like condition, control animals attain blood ethanol ('binge-like') levels >80 mg/dl. The single hMSC administration reduced relapse-like blood ethanol levels to 20 mg/dl. Chronic ethanol intake increased by 250% (P < 0.001) the levels of reactive oxygen species in hippocampus, which were markedly reduced by hMSC administration. Astrocyte glial acidic fibrillary protein immunoreactivity, a hallmark of neuroinflammation, was increased by 60-80% (P < 0.001) by chronic ethanol intake, an effect that was fully abolished by the administration of hMSCs. This study supports the neuroinflammation-chronic ethanol intake hypothesis and suggest that mesenchymal stem cell administration may be considered in the treatment of alcohol use disorders.
Collapse
Affiliation(s)
- Fernando Ezquer
- Centro de Medicina Regenerativa; Facultad de Medicina Clínica Alemana-Universidad del Desarrollo; Chile
| | | | - Paola Morales
- Molecular and Clinical Pharmacology Program; Institute of Biomedical Sciences; Chile
- Department of Neuroscience, Faculty of Medicine; University of Chile; Chile
| | - Marcelo Ezquer
- Centro de Medicina Regenerativa; Facultad de Medicina Clínica Alemana-Universidad del Desarrollo; Chile
| | | | | | - Yedy Israel
- Molecular and Clinical Pharmacology Program; Institute of Biomedical Sciences; Chile
| |
Collapse
|
84
|
Erickson EK, Grantham EK, Warden AS, Harris RA. Neuroimmune signaling in alcohol use disorder. Pharmacol Biochem Behav 2018; 177:34-60. [PMID: 30590091 DOI: 10.1016/j.pbb.2018.12.007] [Citation(s) in RCA: 147] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 10/25/2018] [Accepted: 12/20/2018] [Indexed: 02/07/2023]
Abstract
Alcohol use disorder (AUD) is a widespread disease with limited treatment options. Targeting the neuroimmune system is a new avenue for developing or repurposing effective pharmacotherapies. Alcohol modulates innate immune signaling in different cell types in the brain by altering gene expression and the molecular pathways that regulate neuroinflammation. Chronic alcohol abuse may cause an imbalance in neuroimmune function, resulting in prolonged perturbations in brain function. Likewise, manipulating the neuroimmune system may change alcohol-related behaviors. Psychiatric disorders that are comorbid with AUD, such as post-traumatic stress disorder, major depressive disorder, and other substance use disorders, may also have underlying neuroimmune mechanisms; current evidence suggests that convergent immune pathways may be involved in AUD and in these comorbid disorders. In this review, we provide an overview of major neuroimmune cell-types and pathways involved in mediating alcohol behaviors, discuss potential mechanisms of alcohol-induced neuroimmune activation, and present recent clinical evidence for candidate immune-related drugs to treat AUD.
Collapse
Affiliation(s)
- Emma K Erickson
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712-01095, USA.
| | - Emily K Grantham
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712-01095, USA
| | - Anna S Warden
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712-01095, USA
| | - R A Harris
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712-01095, USA
| |
Collapse
|
85
|
Tripathi A, Debelius J, Brenner DA, Karin M, Loomba R, Schnabl B, Knight R. Publisher Correction: The gut-liver axis and the intersection with the microbiome. Nat Rev Gastroenterol Hepatol 2018; 15:785. [PMID: 29785003 PMCID: PMC7133393 DOI: 10.1038/s41575-018-0031-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In the original version of Table 1 published online, upward arrows to indicate increased translocation of PAMPs were missing from the row entitled 'Translocation' for both the column on alcoholic liver disease and nonalcoholic fatty liver disease. This error has now been updated in the PDF and HTML version of the article.
Collapse
|
86
|
Quintanilla ME, Morales P, Ezquer F, Ezquer M, Herrera-Marschitz M, Israel Y. Commonality of Ethanol and Nicotine Reinforcement and Relapse in Wistar-Derived UChB Rats: Inhibition by N
-Acetylcysteine. Alcohol Clin Exp Res 2018; 42:1988-1999. [DOI: 10.1111/acer.13842] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 07/15/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Maria Elena Quintanilla
- Molecular and Clinical Pharmacology Program; Institute of Biomedical Sciences; University of Chile; Santiago Chile
| | - Paola Morales
- Molecular and Clinical Pharmacology Program; Institute of Biomedical Sciences; University of Chile; Santiago Chile
- Neuroscience Department; Faculty of Medicine; University of Chile; Santiago Chile
| | - Fernando Ezquer
- Facultad de Medicina Clínica; Centro de Medicina Regenerativa; Alemana-Universidad del Desarrollo; Santiago Chile
- Facultad de Medicina; Centro de Medicina Regenerativa; Clinica Alemana-Universidad del Desarrollo; Santiago Chile
| | - Marcelo Ezquer
- Facultad de Medicina Clínica; Centro de Medicina Regenerativa; Alemana-Universidad del Desarrollo; Santiago Chile
- Facultad de Medicina; Centro de Medicina Regenerativa; Clinica Alemana-Universidad del Desarrollo; Santiago Chile
| | - Mario Herrera-Marschitz
- Molecular and Clinical Pharmacology Program; Institute of Biomedical Sciences; University of Chile; Santiago Chile
| | - Yedy Israel
- Molecular and Clinical Pharmacology Program; Institute of Biomedical Sciences; University of Chile; Santiago Chile
| |
Collapse
|
87
|
Wang Y, Wang X, Li H, Xu M, Frank J, Luo J. Binge ethanol exposure induces endoplasmic reticulum stress in the brain of adult mice. Toxicol Appl Pharmacol 2018; 356:172-181. [PMID: 30114398 DOI: 10.1016/j.taap.2018.08.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 08/02/2018] [Accepted: 08/11/2018] [Indexed: 12/16/2022]
Abstract
Alcohol abuse causes brain damage and cognitive dysfunction. However, the underlying mechanisms remain elusive. Endoplasmic reticulum (ER) acts as machinery to ensure the proper folding of newly synthesized proteins. The perturbation of ER, i.e., ER stress, plays a pivotal role in some neurological disorders. Mammalian target of rapamycin (mTOR), a serine/threonine kinase, is involved in the regulation of ER stress. The current study sought to determine whether binge ethanol exposure induces ER stress in adult mouse brain and the role mTOR signaling during this process. Adult C57BL6 mice received binge ethanol exposure by daily gavage (5 g/kg, 25% ethanol w/v) for 1, 5 or 10 days. Binge ethanol exposure caused neurodegeneration and neuroinflammation after 5 days of exposure, and a concomitant increase of ER stress and inhibition of mTOR. However, ethanol exposure did not significantly alter spatial learning and memory, and spontaneous locomotor activity. Ethanol treatment induced ER stress and the death of cultured neuronal cells. Cotreatment with an ER stress inhibitor, sodium 4-phenylbutyrate (4-PBA) significantly diminished ethanol-induced ER stress and neuronal apoptosis, suggesting that ER stress contributes to ethanol-induced neurodegeneration. Furthermore, the blockage of mTOR activity by rapamycin increased ER stress in cultured neuronal cells; whereas the activation or inhibition of ER stress by tunicamycin or 4-PBA respectively had little effects on mTOR signaling. These results suggested that mTOR signaling is upstream of ER stress and may thereby mediate ethanol-induced ER stress.
Collapse
Affiliation(s)
- Yongchao Wang
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY 40536, United States
| | - Xin Wang
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY 40536, United States; Center for Health Services Research, University of Kentucky College of Medicine, Lexington, KY 40536, United States
| | - Hui Li
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY 40536, United States
| | - Mei Xu
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY 40536, United States
| | - Jacqueline Frank
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY 40536, United States
| | - Jia Luo
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY 40536, United States.
| |
Collapse
|
88
|
Hillemacher T, Bachmann O, Kahl KG, Frieling H. Alcohol, microbiome, and their effect on psychiatric disorders. Prog Neuropsychopharmacol Biol Psychiatry 2018; 85:105-115. [PMID: 29705711 DOI: 10.1016/j.pnpbp.2018.04.015] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Revised: 04/05/2018] [Accepted: 04/25/2018] [Indexed: 12/12/2022]
Abstract
There is accumulating evidence that alcohol consumption and especially alcohol withdrawal increase brain levels of known innate immune signaling molecules and cause neuroinflammation. It has been shown that microbiota play a pivotal role in this process and affect central neurochemistry and behavior. Disruption of or alterations in the intimate cross-talk between microbiome and brain may be a significant factor in many psychiatric disorders. Alterations in the composition of the microbiome, so called dysbiosis, may result in detrimental distortion of microbe-host homeostasis modulating the hypothalamic-pituitary-adrenal axis. A variety of pathologies are associated with changes in the community structure and function of the gut microbiota, suggesting a link between dysbiosis and disease etiology, including irritable bowel syndrome depression, anxiety disorders, schizophrenia, and alcoholism. Despite a paucity of clinical studies in alcohol-dependent humans, emerging data suggests that alcohol induced alterations of the microbiome may explain reward-seeking behaviors as well as anxiety, depression, and craving in withdrawal and increase the risk of developing psychiatric disorders.
Collapse
Affiliation(s)
- Thomas Hillemacher
- Department of Psychiatry and Psychotherapy, Paracelsus Medical University Nuremberg, Germany; Department of Psychiatry, Social Psychiatry, and Psychotherapy, Hannover Medical School (MHH), Germany
| | - Oliver Bachmann
- Department of Gastroenterology, Hepatology, and Endocrinology, Hannover Medical School (MHH), Germany; German Center for Infection Research (DZIF), partner site Hannover-Braunschweig, Germany
| | - Kai G Kahl
- Department of Psychiatry, Social Psychiatry, and Psychotherapy, Hannover Medical School (MHH), Germany
| | - Helge Frieling
- Department of Psychiatry, Social Psychiatry, and Psychotherapy, Hannover Medical School (MHH), Germany.
| |
Collapse
|
89
|
Barr T, Sureshchandra S, Ruegger P, Zhang J, Ma W, Borneman J, Grant K, Messaoudi I. Concurrent gut transcriptome and microbiota profiling following chronic ethanol consumption in nonhuman primates. Gut Microbes 2018; 9:338-356. [PMID: 29517944 PMCID: PMC6219653 DOI: 10.1080/19490976.2018.1441663] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 11/28/2017] [Accepted: 02/13/2018] [Indexed: 02/07/2023] Open
Abstract
Alcohol use disorder (AUD) results in increased intestinal permeability, nutrient malabsorption, and increased risk of colorectal cancer (CRC). Our understanding of the mechanisms underlying these morbidities remains limited because studies to date have relied almost exclusively on short-term heavy/binge drinking rodent models and colonic biopsies/fecal samples collected from AUD subjects with alcoholic liver disease (ALD). Consequently, the dose- and site-dependent impact of chronic alcohol consumption in the absence of overt liver disease remains poorly understood. In this study, we addressed this knowledge gap using a nonhuman primate model of voluntary ethanol self-administration where rhesus macaques consume varying amounts of 4% ethanol in water for 12 months. Specifically, we performed RNA-Seq and 16S rRNA gene sequencing on duodenum, jejunum, ileum, and colon biopsies collected from 4 controls and 8 ethanol-consuming male macaques. Our analysis revealed that chronic ethanol consumption leads to changes in the expression of genes involved in protein trafficking, metabolism, inflammation, and CRC development. Additionally, we observed differences in the relative abundance of putatively beneficial bacteria as well as those associated with inflammation and CRC. Given that the animals studied in this manuscript did not exhibit signs of ALD or CRC, our data suggest that alterations in gene expression and bacterial communities precede clinical disease and could serve as biomarkers as well as facilitate future studies aimed at developing interventions to restore gut homeostasis.
Collapse
Affiliation(s)
- Tasha Barr
- Division of Biomedical Sciences, University of California-Riverside, Riverside, CA, USA
| | - Suhas Sureshchandra
- Department of Molecular Biology and Biochemistry, University of California-Irvine, Irvine, CA, USA
| | - Paul Ruegger
- Department of Plant Pathology and Microbiology, University of California-Riverside, Riverside, CA, USA
| | - Jingfei Zhang
- Department of Statistics, University of California-Riverside, Riverside, CA, USA
| | - Wenxiu Ma
- Department of Statistics, University of California-Riverside, Riverside, CA, USA
| | - James Borneman
- Department of Plant Pathology and Microbiology, University of California-Riverside, Riverside, CA, USA
| | - Kathleen Grant
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, USA
| | - Ilhem Messaoudi
- Department of Molecular Biology and Biochemistry, University of California-Irvine, Irvine, CA, USA
| |
Collapse
|
90
|
Tripathi A, Debelius J, Brenner DA, Karin M, Loomba R, Schnabl B, Knight R. The gut-liver axis and the intersection with the microbiome. Nat Rev Gastroenterol Hepatol 2018; 15:397-411. [PMID: 29748586 PMCID: PMC6319369 DOI: 10.1038/s41575-018-0011-z] [Citation(s) in RCA: 935] [Impact Index Per Article: 133.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In the past decade, an exciting realization has been that diverse liver diseases - ranging from nonalcoholic steatohepatitis, alcoholic steatohepatitis and cirrhosis to hepatocellular carcinoma - fall along a spectrum. Work on the biology of the gut-liver axis has assisted in understanding the basic biology of both alcoholic fatty liver disease and nonalcoholic fatty liver disease (NAFLD). Of immense importance is the advancement in understanding the role of the microbiome, driven by high-throughput DNA sequencing and improved computational techniques that enable the complexity of the microbiome to be interrogated, together with improved experimental designs. Here, we review gut-liver communications in liver disease, exploring the molecular, genetic and microbiome relationships and discussing prospects for exploiting the microbiome to determine liver disease stage and to predict the effects of pharmaceutical, dietary and other interventions at a population and individual level. Although much work remains to be done in understanding the relationship between the microbiome and liver disease, rapid progress towards clinical applications is being made, especially in study designs that complement human intervention studies with mechanistic work in mice that have been humanized in multiple respects, including the genetic, immunological and microbiome characteristics of individual patients. These 'avatar mice' could be especially useful for guiding new microbiome-based or microbiome-informed therapies.
Collapse
Affiliation(s)
- Anupriya Tripathi
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
- Department of Pediatrics, University of California, San Diego, CA, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, CA, USA
| | - Justine Debelius
- Department of Pediatrics, University of California, San Diego, CA, USA
| | - David A Brenner
- NAFLD Research Center, Division of Gastroenterology, Department of Medicine, University of California, San Diego, CA, USA
| | - Michael Karin
- Department of Pediatrics, University of California, San Diego, CA, USA
- Department of Computer Science and Engineering, University of California, San Diego, CA, USA
| | - Rohit Loomba
- NAFLD Research Center, Division of Gastroenterology, Department of Medicine, University of California, San Diego, CA, USA
- Center for Microbiome Innovation, University of California, San Diego, CA, USA
| | - 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 Microbiome Innovation, University of California, San Diego, CA, USA
| | - Rob Knight
- Department of Pediatrics, University of California, San Diego, CA, USA.
- Department of Computer Science and Engineering, University of California, San Diego, CA, USA.
- Center for Microbiome Innovation, University of California, San Diego, CA, USA.
| |
Collapse
|
91
|
Cassard AM, Ciocan D. Microbiota, a key player in alcoholic liver disease. Clin Mol Hepatol 2018; 24:100-107. [PMID: 29268595 PMCID: PMC6038939 DOI: 10.3350/cmh.2017.0067] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 11/22/2017] [Indexed: 02/07/2023] Open
Abstract
Alcoholic liver disease (ALD) is a major cause of morbidity and mortality worldwide. Only 20% of heavy alcohol consumers develop alcoholic liver cirrhosis. The intestinal microbiota (IM) has been recently identified as a key player in the severity of liver injury in ALD. Common features of ALD include a decrease of gut epithelial tight junction protein expression, mucin production, and antimicrobial peptide levels. This disruption of the gut barrier, which is a prerequisite for ALD, leads to the passage of bacterial products into the blood stream (endotoxemia). Moreover, metabolites produced by bacteria, such as short chain fatty acids, volatile organic compounds (VOS), and bile acids (BA), are involved in ALD pathology. Probiotic treatment, IM transplantation, or the consumption of dietary fiber, such as pectin, which all alter the ratio of bacterial species, have been shown to improve liver injury in animal models of ALD and to be associated with an improvement in gut barrier function. Although the connections between the microbiota and the host in ALD are well established, the underlying mechanisms are still an active area of research. Targeting the microbiome through the use of prebiotic, probiotic, and postbiotic modalities could be an attractive new approach to manage ALD.
Collapse
Affiliation(s)
- Anne-Marie Cassard
- INSERM UMR996, Inflammation, Chemokines, and Immunopathology, Clamart, France
- Univ Paris-Sud, Univ Paris-Saclay, DHU Hepatinov, Labex Lermit, CHU Bicêtre, Kremlin-Bicêtre, France
| | - Dragos Ciocan
- INSERM UMR996, Inflammation, Chemokines, and Immunopathology, Clamart, France
- Univ Paris-Sud, Univ Paris-Saclay, DHU Hepatinov, Labex Lermit, CHU Bicêtre, Kremlin-Bicêtre, France
| |
Collapse
|
92
|
Cannon AR, Kuprys PV, Cobb AN, Ding X, Kothari AN, Kuo PC, Eberhardt JM, Hammer AM, Morris NL, Li X, Choudhry MA. Alcohol enhances symptoms and propensity for infection in inflammatory bowel disease patients and a murine model of DSS-induced colitis. J Leukoc Biol 2018; 104:543-555. [PMID: 29775230 DOI: 10.1002/jlb.4ma1217-506r] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 03/28/2018] [Accepted: 04/11/2018] [Indexed: 12/13/2022] Open
Abstract
Over 1.4 million Americans have been diagnosed with inflammatory bowel disease (IBD), and ulcerative colitis (UC) makes up approximately half of those diagnoses. As a disease, UC cycles between periods of remission and flare, which is characterized by intense abdominal pain, increased weight loss, intestinal inflammation, rectal bleeding, and dehydration. Interestingly, a widespread recommendation to IBD patients for avoidance of a flare period is "Don't Drink Alcohol" as recent work correlated alcohol consumption with increased GI symptoms in patients with IBD. Alcohol alone not only induces a systemic pro-inflammatory response, but can also be directly harmful to gut barrier integrity. However, how alcohol could result in the exacerbation of UC in both patients and murine models of colitis has yet to be elucidated. Therefore, we conducted a retrospective analysis of patients admitted for IBD with a documented history of alcohol use in conjunction with a newly developed mouse model of binge alcohol consumption following dextran sulfate sodium (DSS)-induced colitis. We found that alcohol negatively impacts clinical outcomes of patients with IBD, specifically increased intestinal infections, antibiotic injections, abdomen CT scans, and large intestine biopsies. Furthermore, in our mouse model of binge alcohol consumption following an induced colitis flare, we found alcohol exacerbates weight loss, clinical scores, colonic shortening and inflammation, and propensity to infection. These findings highlight alcohol's ability to potentiate symptoms and susceptibility to infection in UC and suggest alcohol as an underlying factor in perpetuating symptoms of IBD.
Collapse
Affiliation(s)
- Abigail R Cannon
- Alcohol Research Program, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA.,Burn & Shock Trauma Research Institute, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA.,Integrative Cell Biology Program, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA
| | - Paulius V Kuprys
- Alcohol Research Program, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA.,Burn & Shock Trauma Research Institute, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA.,Integrative Cell Biology Program, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA
| | - Adrienne N Cobb
- Burn & Shock Trauma Research Institute, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA.,Department of Surgery, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA
| | - Xianzhong Ding
- Department of Pathology, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA
| | - Anai N Kothari
- Burn & Shock Trauma Research Institute, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA.,Department of Surgery, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA
| | - Paul C Kuo
- Department of Surgery, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA
| | - Joshua M Eberhardt
- Department of Surgery, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA
| | - Adam M Hammer
- Alcohol Research Program, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA.,Burn & Shock Trauma Research Institute, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA.,Integrative Cell Biology Program, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA
| | - Niya L Morris
- Alcohol Research Program, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA.,Burn & Shock Trauma Research Institute, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA.,Integrative Cell Biology Program, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA
| | - Xiaoling Li
- Alcohol Research Program, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA.,Burn & Shock Trauma Research Institute, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA.,Department of Surgery, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA
| | - Mashkoor A Choudhry
- Alcohol Research Program, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA.,Burn & Shock Trauma Research Institute, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA.,Department of Surgery, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA.,Department of Pathology, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA.,Department of Microbiology and Immunology, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA.,Integrative Cell Biology Program, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA
| |
Collapse
|
93
|
Grander C, Adolph TE, Wieser V, Lowe P, Wrzosek L, Gyongyosi B, Ward DV, Grabherr F, Gerner RR, Pfister A, Enrich B, Ciocan D, Macheiner S, Mayr L, Drach M, Moser P, Moschen AR, Perlemuter G, Szabo G, Cassard AM, Tilg H. Recovery of ethanol-induced Akkermansia muciniphila depletion ameliorates alcoholic liver disease. Gut 2018; 67:891-901. [PMID: 28550049 DOI: 10.1136/gutjnl-2016-313432] [Citation(s) in RCA: 440] [Impact Index Per Article: 62.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 04/11/2017] [Accepted: 04/15/2017] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Alcoholic liver disease (ALD) is a global health problem with limited therapeutic options. Intestinal barrier integrity and the microbiota modulate susceptibility to ALD. Akkermansia muciniphila, a Gram-negative intestinal commensal, promotes barrier function partly by enhancing mucus production. The aim of this study was to investigate microbial alterations in ALD and to define the impact of A. muciniphila administration on the course of ALD. DESIGN The intestinal microbiota was analysed in an unbiased approach by 16S ribosomal DNA (rDNA) sequencing in a Lieber-DeCarli ALD mouse model, and faecal A. muciniphila abundance was determined in a cohort of patients with alcoholic steatohepatitis (ASH). The impact of A. muciniphila on the development of experimental acute and chronic ALD was determined in a preventive and therapeutic setting, and intestinal barrier integrity was analysed. RESULTS Patients with ASH exhibited a decreased abundance of faecal A. muciniphila when compared with healthy controls that indirectly correlated with hepatic disease severity. Ethanol feeding of wild-type mice resulted in a prominent decline in A. muciniphila abundance. Ethanol-induced intestinal A. muciniphila depletion could be restored by oral A. muciniphila supplementation. Furthermore, A. muciniphila administration when performed in a preventive setting decreased hepatic injury, steatosis and neutrophil infiltration. A. muciniphila also protected against ethanol-induced gut leakiness, enhanced mucus thickness and tight-junction expression. In already established ALD, A. muciniphila used therapeutically ameliorated hepatic injury and neutrophil infiltration. CONCLUSION Ethanol exposure diminishes intestinal A. muciniphila abundance in both mice and humans and can be recovered in experimental ALD by oral supplementation. A. muciniphila promotes intestinal barrier integrity and ameliorates experimental ALD. Our data suggest that patients with ALD might benefit from A. muciniphila supplementation.
Collapse
Affiliation(s)
- Christoph Grander
- Department of Internal Medicine I, Gastroenterology, Endocrinology & Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Timon E Adolph
- Department of Internal Medicine I, Gastroenterology, Endocrinology & Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Verena Wieser
- Department of Internal Medicine I, Gastroenterology, Endocrinology & Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Patrick Lowe
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Laura Wrzosek
- Department of Inflammation, Chemokines and Immunopathology, INSERM UMR996, Clamart, France
| | - Benedek Gyongyosi
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Doyle V Ward
- Center for Microbiome Research, University of Massachusetts Medical School, Worcester, Massachusetts, USA.,Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Felix Grabherr
- Department of Internal Medicine I, Gastroenterology, Endocrinology & Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Romana R Gerner
- Department of Internal Medicine I, Gastroenterology, Endocrinology & Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Alexandra Pfister
- Department of Internal Medicine I, Gastroenterology, Endocrinology & Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Barbara Enrich
- Department of Internal Medicine I, Gastroenterology, Endocrinology & Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Dragos Ciocan
- Department of Inflammation, Chemokines and Immunopathology, INSERM UMR996, Clamart, France.,Univ Paris-Sud, Univ Paris-Saclay, DHU Hepatinov, Labex Lermit, CHU Bicêtre, Kremlin-Bicêtre, France.,AP-HP, Hepatogastroenterology and Nutrition, Hôpital Antoine-Béclère, Clamart, France
| | - Sophie Macheiner
- Department of Internal Medicine I, Gastroenterology, Endocrinology & Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Lisa Mayr
- Department of Internal Medicine I, Gastroenterology, Endocrinology & Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Matthias Drach
- Department of Dermatology, University Hospital Zürich, Zürich, Switzerland
| | - Patrizia Moser
- Institute of Pathology, Medical University Innsbruck, Innsbruck, Austria
| | - Alexander R Moschen
- Department of Internal Medicine I, Gastroenterology, Endocrinology & Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Gabriel Perlemuter
- Department of Inflammation, Chemokines and Immunopathology, INSERM UMR996, Clamart, France.,Univ Paris-Sud, Univ Paris-Saclay, DHU Hepatinov, Labex Lermit, CHU Bicêtre, Kremlin-Bicêtre, France.,AP-HP, Hepatogastroenterology and Nutrition, Hôpital Antoine-Béclère, Clamart, France
| | - Gyongyi Szabo
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Anne Marie Cassard
- Department of Inflammation, Chemokines and Immunopathology, INSERM UMR996, Clamart, France.,Univ Paris-Sud, Univ Paris-Saclay, DHU Hepatinov, Labex Lermit, CHU Bicêtre, Kremlin-Bicêtre, France
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Endocrinology & Metabolism, Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
94
|
TLR4/MyD88 -mediated CCL2 production by lipopolysaccharide (endotoxin): Implications for metabolic inflammation. J Diabetes Metab Disord 2018; 17:77-84. [PMID: 30288388 PMCID: PMC6154519 DOI: 10.1007/s40200-018-0341-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 03/21/2018] [Indexed: 12/11/2022]
Abstract
Background Obese human and mice were reported to have higher circularity endotoxin (LPS) levels as compared to their lean counter parts. The current study was aimed to reveal the molecular mechanisms underlying the LPS mediated induction of CCL2 in human monocytes/macrophages. Methods Human monocytic cell line THP-1, THP-1 cells derived macrophages and primary macrophages were treated with LPS and TNF-α (positive control). CCL2 expression was determined with real-time RT-PCR and ELISA. THP-1-XBlue™ cells, THP-1-XBlue™-defMyD cells, TLR4 neutralization antibody, TLR4 siRNA and inhibitors for NF-kB and MAPK were used to study the signaling pathways. Phosphorylation of NF-kB and c-Jun was analyzed by ELISA. Results LPS upregulates CCL2 expression at both mRNA (THP-1: 23.40 ± .071 Fold, P < 0.0001; THP-1-derived macrophages: 103 ± 0.56 Fold, < 0.0001; Primary macrophages: 48 ± 1.41 Fold, P < 0.0005) and protein (THP1 monocytes:1048 ± 5.67 pg/ml, P < 0.0001; THP-1-derived macrophages; 2014 ± 2.12, P = 0.0001; Primary macrophages: 859.5 ± 3.54, P < 0.0001) levels in human monocytic cells/macrophages. Neutralization of TLR4 blocked LPS-induced CCL-2 secretion (P < 0.0001). Silencing of TLR4 by siRNA also significantly reduced LPS-induced CCL-2 production. Furthermore, MyD88-Knockout cells treated with LPS did not produce CCL-2. NF-kB and c-Jun phosphorylation was noted in LPS treated cells. Conclusion Overall, our data reveal that LPS induces CCL-2 in monocytes/macrophages via TLR4/MyD88 signaling which leads to the activation of NF-kB/AP-1 transcription factors.
Collapse
|
95
|
Hwang S, Kim SH, Rhee KJ. Gut Microbiome and Gastrointestinal Diseases. KOREAN JOURNAL OF CLINICAL LABORATORY SCIENCE 2018. [DOI: 10.15324/kjcls.2018.50.1.11] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Affiliation(s)
- Soonjae Hwang
- Department of Biomedical Laboratory Science, College of Health Sciences, Yonsei University at Wonju, Wonju, Korea
| | - Sung Hoon Kim
- Department of Biomedical Laboratory Science, College of Health Sciences, Yonsei University at Wonju, Wonju, Korea
| | - Ki-Jong Rhee
- Department of Biomedical Laboratory Science, College of Health Sciences, Yonsei University at Wonju, Wonju, Korea
| |
Collapse
|
96
|
Ezquer F, Morales P, Quintanilla ME, Santapau D, Lespay-Rebolledo C, Ezquer M, Herrera-Marschitz M, Israel Y. Intravenous administration of anti-inflammatory mesenchymal stem cell spheroids reduces chronic alcohol intake and abolishes binge-drinking. Sci Rep 2018; 8:4325. [PMID: 29567966 PMCID: PMC5864829 DOI: 10.1038/s41598-018-22750-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 02/22/2018] [Indexed: 02/06/2023] Open
Abstract
Chronic alcohol intake leads to neuroinflammation and astrocyte dysfunction, proposed to perpetuate alcohol consumption and to promote conditioned relapse-like binge drinking. In the present study, human mesenchymal stem cells (MSCs) were cultured in 3D-conditions to generate MSC-spheroids, which greatly increased MSCs anti-inflammatory ability and reduced cell volume by 90% versus conventionally 2D-cultured MSCs, enabling their intravenous administration and access to the brain. It is shown, in an animal model of chronic ethanol intake and relapse-drinking, that both the intravenous and intra-cerebroventricular administration of a single dose of MSC-spheroids inhibited chronic ethanol intake and relapse-like drinking by 80–90%, displaying significant effects over 3–5 weeks. The MSC-spheroid administration fully normalized alcohol-induced neuroinflammation, as shown by a reduced astrocyte activation, and markedly increased the levels of the astrocyte Na-glutamate (GLT-1) transporter. This research suggests that the intravenous administration of MSC-spheroids may constitute an effective new approach for the treatment of alcohol-use disorders.
Collapse
Affiliation(s)
- Fernando Ezquer
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - Paola Morales
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, University of Chile, Santiago, Chile.,Department of Neuroscience, Faculty of Medicine, University of Chile, Santiago, Chile
| | - María Elena Quintanilla
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Daniela Santapau
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - Carolyne Lespay-Rebolledo
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Marcelo Ezquer
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - Mario Herrera-Marschitz
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Yedy Israel
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, University of Chile, Santiago, Chile.
| |
Collapse
|
97
|
Intestinal dysbiosis and permeability: the yin and yang in alcohol dependence and alcoholic liver disease. Clin Sci (Lond) 2018; 132:199-212. [PMID: 29352076 DOI: 10.1042/cs20171055] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 12/07/2017] [Accepted: 12/19/2017] [Indexed: 02/07/2023]
Abstract
Alcohol dependence and alcoholic liver disease represent a major public health problem with substantial morbidity and mortality. By yet incompletely understood mechanisms, chronic alcohol abuse is associated with increased intestinal permeability and alterations of the gut microbiota composition, allowing bacterial components, bacteria, and metabolites to reach the portal and the systemic circulation. These gut-derived bacterial products are recognized by immune cells circulating in the blood or residing in remote organs such as the liver leading to the release of pro-inflammatory cytokines which are considered important mediators of the liver-gut-brain communication. Although circulating cytokines are likely not the sole factors involved, they can induce liver inflammation/damage and reach the central nervous system where they favor neuroinflammation which is associated with change in mood, cognition, and drinking behavior. In this review, the authors focus on the current evidence describing the changes that occur in the intestinal microbiota with chronic alcohol consumption in conjunction with intestinal barrier breakdown and inflammatory changes sustaining the concept of a gut-liver-brain axis in the pathophysiology of alcohol dependence and alcoholic liver disease.
Collapse
|
98
|
Cassard AM, Gérard P, Perlemuter G. Microbiota, Liver Diseases, and Alcohol. BUGS AS DRUGS 2018:187-212. [DOI: 10.1128/9781555819705.ch8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Affiliation(s)
- Anne-Marie Cassard
- INSERM U996 Inflammation, Chemokines and Immunopathology, DHU Hepatinov, Univ Paris-Sud; Université Paris-Saclay; 92140 Clamart France
| | - Philippe Gérard
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay; 78350 Jouyen-Josas France
| | - Gabriel Perlemuter
- INSERM U996 Inflammation, Chemokines and Immunopathology, DHU Hepatinov, Univ Paris-Sud; Université Paris-Saclay; 92140 Clamart France
- AP-HP, Hepatogastroenterology and Nutrition, Hôpital Antoine-Béclère; Clamart France
| |
Collapse
|
99
|
Walter TJ, Vetreno RP, Crews FT. Alcohol and Stress Activation of Microglia and Neurons: Brain Regional Effects. Alcohol Clin Exp Res 2017; 41:2066-2081. [PMID: 28941277 PMCID: PMC5725687 DOI: 10.1111/acer.13511] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 09/19/2017] [Indexed: 12/13/2022]
Abstract
Background Cycles of alcohol and stress are hypothesized to contribute to alcohol use disorders. How this occurs is poorly understood, although both alcohol and stress activate the neuroimmune system—the immune molecules and cells that interact with the nervous system. The effects of alcohol and stress on the neuroimmune system are mediated in part by peripheral signaling molecules. Alcohol and stress both enhance immunomodulatory molecules such as corticosterone and endotoxin to impact neuroimmune cells, such as microglia, and may subsequently impact neurons. In this study, we therefore examined the effects of acute and chronic ethanol (EtOH) on the corticosterone, endotoxin, and microglial and neuronal response to acute stress. Methods Male Wistar rats were treated intragastrically with acute EtOH and acutely stressed with restraint/water immersion. Another group of rats was treated intragastrically with chronic intermittent EtOH and acutely stressed following prolonged abstinence. Plasma corticosterone and endotoxin were measured, and immunohistochemical stains for the microglial marker CD11b and neuronal activation marker c‐Fos were performed. Results Acute EtOH and acute stress interacted to increase plasma endotoxin and microglial CD11b, but not plasma corticosterone or neuronal c‐Fos. Chronic EtOH caused a lasting sensitization of stress‐induced plasma endotoxin, but not plasma corticosterone. Chronic EtOH also caused a lasting sensitization of stress‐induced microglial CD11b, but not neuronal c‐Fos. Conclusions These results find acute EtOH combined with acute stress enhanced plasma endotoxin, as well as microglial CD11b in many brain regions. Chronic EtOH followed by acute stress also increased plasma endotoxin and microglial CD11b, suggesting a lasting sensitization to acute stress. Overall, these data suggest alcohol and stress interact to increase plasma endotoxin, resulting in enhanced microglial activation that could contribute to disease progression.
Collapse
Affiliation(s)
- Thomas Jordan Walter
- Bowles Center for Alcohol Studies, The School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC.,Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Ryan P Vetreno
- Bowles Center for Alcohol Studies, The School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Fulton T Crews
- Bowles Center for Alcohol Studies, The School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC.,Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| |
Collapse
|
100
|
Bachtell RK, Jones JD, Heinzerling KG, Beardsley PM, Comer SD. Glial and neuroinflammatory targets for treating substance use disorders. Drug Alcohol Depend 2017; 180:156-170. [PMID: 28892721 PMCID: PMC5790191 DOI: 10.1016/j.drugalcdep.2017.08.003] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 07/28/2017] [Accepted: 08/03/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND The plenary session at the 2016 Behavior, Biology and Chemistry: Translational Research in Addiction Conference focused on glia as potential players in the development, persistence and treatment of substance use disorders. Glia partake in various functions that are important for healthy brain activity. Drugs of abuse alter glial cell activity producing several perturbations in brain function that are thought to contribute to behavioral changes associated with substance use disorders. Consequently, drug-induced changes in glia-driven processes in the brain represent potential targets for pharmacotherapeutics treating substance use disorders. METHODS Four speakers presented preclinical and clinical research illustrating the effects that glial modulators have on abuse-related behavioral effects of psychostimulants and opioids. This review highlights some of these findings and expands its focus to include other research focused on drug-induced glia abnormalities and glia-focused treatment approaches in substance use disorders. RESULTS Preclinical findings show that drugs of abuse induce neuroinflammatory signals and disrupt glutamate homeostasis through their interaction with microglia and astrocytes. Preclinical and clinical studies testing the effects of glial modulators show general effectiveness in reducing behaviors associated with substance use disorders. CONCLUSIONS The contribution of drug-induced glial activity continues to emerge as an intriguing target for substance use disorder treatments. Clinical investigations of glial modulators have yielded promising results on substance use measures and indicate that they are generally safe and well-tolerated. However, results have not been entirely positive and more questions remain for continued exploration in the development and testing of glial-directed treatments for substance use disorders.
Collapse
Affiliation(s)
- Ryan K. Bachtell
- Department of Psychology and Neuroscience, and Center for Neuroscience, UCB 345, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Jermaine D. Jones
- Division on Substance Use Disorders, New York State Psychiatric Institute and College of Physicians and Surgeons, Columbia University, 1051 Riverside Drive, New York, NY 10032, USA
| | - Keith G. Heinzerling
- Department of Family Medicine and Center for Behavioral and Addiction Medicine, UCLA, Los Angeles, CA, USA
| | - Patrick M. Beardsley
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 410 N. 12th Street, Richmond, VA 23298, USA
| | - Sandra D. Comer
- Division on Substance Use Disorders, New York State Psychiatric Institute and College of Physicians and Surgeons, Columbia University, 1051 Riverside Drive, New York, NY 10032, USA
| |
Collapse
|