1
|
Wang S, Kou GJ, Zhao XH, Huang G, Wang JX, Tian L, Zuo XL, Li YQ, Wang JY, Yu YB. Altered mucosal bacteria and metabolomics in patients with Peutz-Jeghers syndrome. Gut Pathog 2024; 16:25. [PMID: 38678229 PMCID: PMC11056063 DOI: 10.1186/s13099-024-00617-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 04/19/2024] [Indexed: 04/29/2024] Open
Abstract
BACKGROUND Peutz-Jeghers syndrome (PJS) is a rare genetic disorder characterized by the development of pigmented spots, gastrointestinal polyps and increased susceptibility to cancers. Currently, most studies have investigated intestinal microbiota through fecal microbiota, and there are few reports about mucosa-associated microbiota. It remains valuable to search for the key intestinal microbiota or abnormal metabolic pathways linked to PJS. AIM This study aimed to assess the structure and composition of mucosa-associated microbiota in patients with PJS and to explore the potential influence of intestinal microbiota disorders and metabolite changes on PJS. METHODS The bacterial composition was analyzed in 13 PJS patients and 12 controls using 16S rRNA gene sequencing (Illumina MiSeq) for bacteria. Differential analyses of the intestinal microbiota were performed from the phylum to species level. Liquid chromatography-tandem mass spectrometry (LC‒MS) was used to detect the differentially abundant metabolites of PJS patients and controls to identify different metabolites and metabolic biomarkers of small intestinal mucosa samples. RESULTS High-throughput sequencing confirmed the special characteristics and biodiversity of the mucosa microflora in patients with PJS. They had lower bacterial biodiversity than controls. The abundance of intestinal mucosal microflora was significantly lower than that of fecal microflora. In addition, lipid metabolism, amino acid metabolism, carbohydrate metabolism, nucleotide metabolism and other pathways were significantly different from those of controls, which were associated with the development of the enteric nervous system, intestinal inflammation and development of tumors. CONCLUSION This is the first report on the mucosa-associated microbiota and metabolite profile of subjects with PJS, which may be meaningful to provide a structural basis for further research on intestinal microecology in PJS.
Collapse
Affiliation(s)
- Sui Wang
- Department of Respiratory Medicine, The Second Hospital of Shandong University, Jinan, 250033, Shandong, People's Republic of China
| | - Guan-Jun Kou
- Department of Gastroenterology, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, 250012, Shandong, People's Republic of China
| | - Xiao-Han Zhao
- Department of Gastroenterology, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, 250012, Shandong, People's Republic of China
| | - Gang Huang
- Department of Gastroenterology, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, 250012, Shandong, People's Republic of China
| | - Jue-Xin Wang
- Department of Gastroenterology, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, 250012, Shandong, People's Republic of China
| | - Lin Tian
- Department of Gastroenterology, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, 250012, Shandong, People's Republic of China
| | - Xiu-Li Zuo
- Department of Gastroenterology, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, 250012, Shandong, People's Republic of China
| | - Yan-Qing Li
- Department of Gastroenterology, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, 250012, Shandong, People's Republic of China
| | - Jia-Yong Wang
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, People's Republic of China.
| | - Yan-Bo Yu
- Department of Gastroenterology, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, 250012, Shandong, People's Republic of China.
| |
Collapse
|
2
|
Jia F, Li F, Bai KS, Zou XL. Current status and advancements in research of gut microecology in acute pancreatitis. Shijie Huaren Xiaohua Zazhi 2023; 31:521-527. [DOI: 10.11569/wcjd.v31.i13.521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/22/2023] [Accepted: 06/29/2023] [Indexed: 07/08/2023] Open
Abstract
Acute pancreatitis (AP) is one of the most common acute abdominal conditions in clinical practice, with increasing incidence and substantial healthcare burden. In recent years, substantial research with high-throughput sequencing technologies has revealed the imbalance between beneficial and pathogenic microbiomes as well as their metabolites during the clinical course of AP. Furthermore, disruption of the intestinal barrier and microbial translocation have been identified as important factors exacerbating systemic inflammatory response and subsequent infectious complications in AP. Maintaining a stable gastrointestinal microecology in patients may help prevent gut-derived infection and attenuate the "second hit" of inflammation induced by AP, thereby improving patient outcomes. This article provides a systematic review of the role of intestinal microbiota and microbial metabolites in the progression of AP, as well as potential therapeutic strategies, in order to offer insights into the understanding of AP pathogenesis and the identification of novel therapeutic targets.
Collapse
Affiliation(s)
- Feng Jia
- Duerbote County Hospital of Traditional Chinese Medicine, Daqing 163000, Heilongjiang Province, China
- Key Laboratory of Intensive Care Medicine, Qunli Branch, The First Affiliated Hospital of Harbin Medical University, Harbin 150081, Heilongjiang Province, China
| | - Feng Li
- Department of Hepatobiliary and Pancreatic Surgery, Qunli Branch, The First Affiliated Hospital of Harbin Medical University, Harbin 150081, Heilongjiang Province, China
- Key Laboratory of Intensive Care Medicine, Qunli Branch, The First Affiliated Hospital of Harbin Medical University, Harbin 150081, Heilongjiang Province, China
| | - Kai-Song Bai
- Department of Hepatobiliary and Pancreatic Surgery, Qunli Branch, The First Affiliated Hospital of Harbin Medical University, Harbin 150081, Heilongjiang Province, China
- Key Laboratory of Intensive Care Medicine, Qunli Branch, The First Affiliated Hospital of Harbin Medical University, Harbin 150081, Heilongjiang Province, China
| | - Xiao-Long Zou
- Department of Hepatobiliary and Pancreatic Surgery, Qunli Branch, The First Affiliated Hospital of Harbin Medical University, Harbin 150081, Heilongjiang Province, China
- Key Laboratory of Intensive Care Medicine, Qunli Branch, The First Affiliated Hospital of Harbin Medical University, Harbin 150081, Heilongjiang Province, China
| |
Collapse
|
3
|
Mack S, Flattet Y, Bichard P, Frossard JL. Recent advances in the management of autoimmune pancreatitis in the era of artificial intelligence. World J Gastroenterol 2022; 28:6867-6874. [PMID: 36632320 PMCID: PMC9827582 DOI: 10.3748/wjg.v28.i48.6867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 10/31/2022] [Accepted: 11/16/2022] [Indexed: 12/26/2022] Open
Abstract
Autoimmune pancreatitis (AIP) is a type of immune-mediated pancreatitis subdivided into two subtypes, type 1 and type 2 AIP. Furthermore, type 1 AIP is considered to be the pancreatic manifestation of the immunoglobulin G4 (IgG4)-related disease. Nowadays, AIP is increasingly researched and recognized, although its diagnosis represents a challenge for several reasons: False positive ultrasound-guided cytological samples for a neoplastic process, difficult to interpret levels of IgG4, the absence of biological markers to diagnose type 2 AIP, and the challenging clinical identification of atypical forms. Furthermore, 60% and 78% of type 1 and type 2 AIP, respectively, are retrospectively diagnosed on surgical specimens of resected pancreas for suspected cancer. As distinguishing AIP from pancreatic ductal adenocarcinoma can be challenging, obtaining a definitive diagnosis can therefore prove difficult, since endoscopic ultrasound fine-needle aspiration or biopsy of the pancreas are suboptimal. This paper focuses on recent innovations in the management of AIP with regard to the use of artificial intelligence, new serum markers, and new therapeutic approaches, while it also outlines the current management recommendations. A better knowledge of AIP can reduce the recourse to surgery and avoid its overuse, although such an approach requires close collaboration between gastroenterologists, surgeons and radiologists. Better knowledge on AIP and IgG4-related disease remains necessary to diagnose and manage patients.
Collapse
Affiliation(s)
- Sahar Mack
- Division of Gastroenterology, Department of Medical Specialties, University Hospital of Geneva, Geneva 1205, Switzerland
| | - Yves Flattet
- Division of Gastroenterology, Department of Medical Specialties, University Hospital of Geneva, Geneva 1205, Switzerland
| | - Philippe Bichard
- Division of Gastroenterology, Department of Medical Specialties, University Hospital of Geneva, Geneva 1205, Switzerland
| | - Jean Louis Frossard
- Division of Gastroenterology, Department of Medical Specialties, University Hospital of Geneva, Geneva 1205, Switzerland
| |
Collapse
|
4
|
Wiese ML, Urban S, von Rheinbaben S, Frost F, Sendler M, Weiss FU, Bülow R, Kromrey ML, Tran QT, Lerch MM, Schauer B, Aghdassi AA. Identification of early predictors for infected necrosis in acute pancreatitis. BMC Gastroenterol 2022; 22:405. [PMID: 36057565 PMCID: PMC9440524 DOI: 10.1186/s12876-022-02490-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 08/29/2022] [Indexed: 11/10/2022] Open
Abstract
Background In acute pancreatitis, secondary infection of pancreatic necrosis is a complication that mostly necessitates interventional therapy. A reliable prediction of infected necrotizing pancreatitis would enable an early identification of patients at risk, which however, is not possible yet. Methods This study aims to identify parameters that are useful for the prediction of infected necrosis and to develop a prediction model for early detection. We conducted a retrospective analysis from the hospital information and reimbursement data system and screened 705 patients hospitalized with diagnosis of acute pancreatitis who underwent contrast-enhanced computed tomography and additional diagnostic puncture or drainage of necrotic collections. Both clinical and laboratory parameters were analyzed for an association with a microbiologically confirmed infected pancreatic necrosis. A prediction model was developed using a logistic regression analysis with stepwise inclusion of significant variables. The model quality was tested by receiver operating characteristics analysis and compared to single parameters and APACHE II score. Results We identified a total of 89 patients with necrotizing pancreatitis, diagnosed by computed tomography, who additionally received biopsy or drainage. Out of these, 59 individuals had an infected necrosis. Eleven parameters showed a significant association with an infection including C-reactive protein, albumin, creatinine, and alcoholic etiology, which were independent variables in a predictive model. This model showed an area under the curve of 0.819, a sensitivity of 0.692 (95%-CI [0.547–0.809]), and a specificity of 0.840 (95%-CI [0.631–0.947]), outperforming single laboratory markers and APACHE II score. Even in cases of missing values predictability was reliable. Conclusion A model consisting of a few single blood parameters and etiology of pancreatitis might help for differentiation between infected and non-infected pancreatic necrosis and assist medical therapy in acute necrotizing pancreatitis. Supplementary Information The online version contains supplementary material available at 10.1186/s12876-022-02490-9.
Collapse
Affiliation(s)
- Mats L Wiese
- Department of Medicine A, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, 17475, Greifswald, Germany
| | - Steffi Urban
- Department of Medicine A, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, 17475, Greifswald, Germany
| | - Sabrina von Rheinbaben
- Department of Medicine A, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, 17475, Greifswald, Germany
| | - Fabian Frost
- Department of Medicine A, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, 17475, Greifswald, Germany
| | - Matthias Sendler
- Department of Medicine A, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, 17475, Greifswald, Germany
| | - Frank Ulrich Weiss
- Department of Medicine A, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, 17475, Greifswald, Germany
| | - Robin Bülow
- Institute of Diagnostic Radiology and Neuroradiology, University Medicine Greifswald, Greifswald, Germany
| | - Marie-Luise Kromrey
- Institute of Diagnostic Radiology and Neuroradiology, University Medicine Greifswald, Greifswald, Germany
| | - Quang Trung Tran
- Department of Medicine A, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, 17475, Greifswald, Germany.,Department of Internal Medicine, University of Medicine and Pharmacy, Hue University, Hue City, Vietnam
| | - Markus M Lerch
- Department of Medicine A, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, 17475, Greifswald, Germany.,Ludwigs-Maximilians University Munich, Munich, Germany
| | - Birgit Schauer
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Ali A Aghdassi
- Department of Medicine A, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, 17475, Greifswald, Germany.
| |
Collapse
|
5
|
Suresh V, Dash P, Suklabaidya S, Murmu KC, Sasmal PK, Jogdand GM, Parida D, Sethi M, Das B, Mohapatra D, Saha S, Prasad P, Satoskar A, Senapati S. MIF confers survival advantage to pancreatic CAFs by suppressing interferon pathway-induced p53-dependent apoptosis. FASEB J 2022; 36:e22449. [PMID: 35839070 DOI: 10.1096/fj.202101953r] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 06/22/2022] [Accepted: 06/30/2022] [Indexed: 11/11/2022]
Abstract
The presence of activated pancreatic stellate cells (PSCs) in the pancreatic ductal adenocarcinoma (PDAC) microenvironment plays a significant role in cancer progression. Macrophage migration inhibitory factor (MIF) is overexpressed in PDAC tissues and expressed by both cancer and stromal cells. The pathophysiological role of MIF in PDAC-associated fibroblasts or PSCs is yet to be elucidated. Here we report that the PSCs of mouse or cancer-associated fibroblast cells (CAFs) of human expresses MIF and its receptors, whose expression gets upregulated upon LPS or TNF-α stimulation. In vitro functional experiments showed that MIF significantly conferred a survival advantage to CAFs/PSCs upon growth factor deprivation. Genetic or pharmacological inhibition of MIF also corroborated these findings. Further, co-injection of mouse pancreatic cancer cells with PSCs isolated from Mif-/- or Mif+/+ mice confirmed the pro-survival effect of MIF in PSCs and also demonstrated the pro-tumorigenic role of MIF expressed by CAFs in vivo. Differential gene expression analysis and in vitro mechanistic studies indicated that MIF expressed by activated CAFs/PSCs confers a survival advantage to these cells by suppression of interferon pathway induced p53 dependent apoptosis.
Collapse
Affiliation(s)
- Voddu Suresh
- Tumor Microenvironment and Animal Models Lab, Department of Cancer Biology, Institute of Life Sciences, Bhubaneswar, India
- Regional Centre for Biotechnology, Faridabad, India
| | - Pujarini Dash
- Tumor Microenvironment and Animal Models Lab, Department of Cancer Biology, Institute of Life Sciences, Bhubaneswar, India
| | - Sujit Suklabaidya
- Tumor Microenvironment and Animal Models Lab, Department of Cancer Biology, Institute of Life Sciences, Bhubaneswar, India
| | - Krushna Chandra Murmu
- Regional Centre for Biotechnology, Faridabad, India
- Epigenetic and Chromatin Biology Unit, Institute of Life Sciences, Bhubaneswar, India
| | - Prakash K Sasmal
- Department of General Surgery, All India Institute of Medical Sciences, Bhubaneswar, India
| | - Gajendra M Jogdand
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, India
| | - Deepti Parida
- Tumor Microenvironment and Animal Models Lab, Department of Cancer Biology, Institute of Life Sciences, Bhubaneswar, India
- Regional Centre for Biotechnology, Faridabad, India
| | - Manisha Sethi
- Tumor Microenvironment and Animal Models Lab, Department of Cancer Biology, Institute of Life Sciences, Bhubaneswar, India
- Regional Centre for Biotechnology, Faridabad, India
| | - Biswajit Das
- Tumor Microenvironment and Animal Models Lab, Department of Cancer Biology, Institute of Life Sciences, Bhubaneswar, India
| | - Debasish Mohapatra
- Tumor Microenvironment and Animal Models Lab, Department of Cancer Biology, Institute of Life Sciences, Bhubaneswar, India
- Kalinga Institute of Industrial Technology, Bhubaneswar, India
| | - Subha Saha
- Regional Centre for Biotechnology, Faridabad, India
- Epigenetic and Chromatin Biology Unit, Institute of Life Sciences, Bhubaneswar, India
| | - Punit Prasad
- Epigenetic and Chromatin Biology Unit, Institute of Life Sciences, Bhubaneswar, India
| | - Abhay Satoskar
- Department of Microbiology, The Ohio State University, Columbus, Ohio, USA
| | - Shantibhusan Senapati
- Tumor Microenvironment and Animal Models Lab, Department of Cancer Biology, Institute of Life Sciences, Bhubaneswar, India
| |
Collapse
|
6
|
Wang S, Huang G, Wang JX, Tian L, Zuo XL, Li YQ, Yu YB. Altered Gut Microbiota in Patients With Peutz-Jeghers Syndrome. Front Microbiol 2022; 13:881508. [PMID: 35910641 PMCID: PMC9326469 DOI: 10.3389/fmicb.2022.881508] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 06/06/2022] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Peutz-Jeghers syndrome (PJS) is a rare genetic disorder characterized by the development of pigmented spots and gastrointestinal polyps and increased susceptibility to cancers. It remains unknown whether gut microbiota dysbiosis is linked to PJS. AIM This study aimed to assess the structure and composition of the gut microbiota, including both bacteria and fungi, in patients with PJS and investigate the relationship between gut microbiota dysbiosis and PJS pathogenesis. METHODS The bacterial and fungal composition of the fecal microbiota was analyzed in 23 patients with PJS (cases), 17 first-degree asymptomatic relatives (ARs), and 24 healthy controls (HCs) using 16S (MiSeq) and ITS2 (pyrosequencing) sequencing for bacteria and fungi, respectively. Differential analyses of the intestinal flora were performed from the phylum to species level. RESULTS Alpha-diversity distributions of bacteria and fungi indicated that the abundance of both taxa differed between PJS cases and controls. However, while the diversity and composition of fecal bacteria in PJS cases were significantly different from those in ARs and HCs, fungal flora was more stable. High-throughput sequencing confirmed the special characteristics and biodiversity of the fecal bacterial and fungal microflora in patients with PJS. They had lower bacterial biodiversity than controls, with a higher frequency of the Proteobacteria phylum, Enterobacteriaceae family, and Escherichia-Shigella genus, and a lower frequency of the Firmicutes phylum and the Lachnospiraceae and Ruminococcaceae families. Of fungi, Candida was significantly higher in PJS cases than in controls. CONCLUSION The findings reported here confirm gut microbiota dysbiosis in patients with PJS. This is the first report on the bacterial and fungal microbiota profile of subjects with PJS, which may be meaningful to provide a structural basis for further research on intestinal microecology in PJS.
Collapse
Affiliation(s)
- Sui Wang
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, China
- Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, China
| | - Gang Huang
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, China
- Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, China
| | - Jue-Xin Wang
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, China
- Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, China
| | - Lin Tian
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, China
- Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, China
| | - Xiu-Li Zuo
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, China
- Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, China
| | - Yan-Qing Li
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, China
- Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, China
| | - Yan-Bo Yu
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, China
- Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
7
|
Lycopene Inhibits IL-6 Expression by Upregulating NQO1 and HO-1 via Activation of Nrf2 in Ethanol/Lipopolysaccharide-Stimulated Pancreatic Acinar Cells. Antioxidants (Basel) 2022; 11:antiox11030519. [PMID: 35326169 PMCID: PMC8944646 DOI: 10.3390/antiox11030519] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/05/2022] [Accepted: 03/07/2022] [Indexed: 12/16/2022] Open
Abstract
In alcoholic pancreatitis, alcohol increases gut permeability, which increases the penetration of endotoxins, such as lipopolysaccharides (LPS). LPS act as clinically significant triggers to increase pancreatic damage in alcoholic pancreatitis. Ethanol or LPS treatment increases reactive oxygen species (ROS) production in pancreatic acinar cells. ROS induce inflammatory cytokine production in pancreatic acinar cells, leading to pancreatic inflammation. The nuclear erythroid-2-related factor 2 (Nrf2) pathway is activated as a cytoprotective response to oxidative stress, and induces the expression of NAD(P)H quinone oxidoreductase 1 (NQO1) and heme oxygenase-1 (HO-1). Lycopene exerts anti-inflammatory and antioxidant effects in various cells. We previously showed that lycopene inhibits NADPH oxidase to reduce ROS and IL-6 levels, and zymogene activation in ethanol or palmitoleic acid-treated pancreatic acinar cells. In this study, we examined whether lycopene inhibits IL-6 expression by activating the Nrf2/NQO1-HO-1 pathway, and reducing intracellular and mitochondrial ROS levels, in ethanol and LPS-treated pancreatic AR42J cells. Lycopene increased the phosphorylated and nuclear-translocated Nrf2 levels by decreasing the amount of Nrf2 sequestered in the cytoplasm via a complex formation with Kelch-like ECH1-associated protein 1 (Keap1). Using exogenous inhibitors targeting Nrf2 and HO-1, we showed that the upregulation of activated Nrf2 and HO-1 results in lycopene-induced suppression of IL-6 expression and ROS production. The consumption of lycopene-rich foods may prevent the development of ethanol and LPS-associated pancreatic inflammation by activating Nrf2-mediated expression of NQO1 and HO-1, thereby decreasing ROS-mediated IL-6 expression in pancreatic acinar cells.
Collapse
|
8
|
Kozlova IV, Bykova AP. Osteosarcopenia in chronic pancreatitis. TERAPEVT ARKH 2021; 93:869-875. [DOI: 10.26442/00403660.2021.08.200971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 08/31/2021] [Indexed: 11/22/2022]
Abstract
Aim. To determine clinical features and some mechanisms of osteosarcopenia development in patients with chronic pancreatitis (CP).
Materials and methods. A casecontrol study was conducted on the basis of the Saratov State Clinical Hospital 5 in 20152018 of patients with CP. In a study of 161 patients with CP included, the control group 30 healthy individuals. Patients were divided into groups according to the etiology of CP: 79 with toxic-metabolic CP, 82 with biliary CP. To determine the risks of low-energy fractures, 154 patients were tested with the Fracture risk assessment tool (FRAX). Along with the standard examination, 30 patients with CP dual-energy X-ray absorptiometry was performed. To assess the state of skeletal muscles, body mass index was determined, hand-held dynamometry was performed, and a set of Short Physical Performance Battery (SPPB) tests was used. Along with the assessment of traditional risk factors for osteosarcopenia gender, age, state of reproductive function in women, body mass index, functional state of the pancreas (pancreas) the quantitative content of interleukins (IL)-2, 6, 8 in in colonic biopsies was analyzed by enzyme-linked immunosorbent assay (ELISA).
Results. Bone disorders, according to densitometry, was detected in 70.0% of patients with CP, in 13.3% of the control group. Presarcopenia was detected in 62 (38.5%) patients with CP, sarcopenia in 34 (21.1%), in the control group presarcopenia and sarcopenia were not detected. Sarcopenia was statistically significantly more common in toxic-metabolic CP than in biliary CP (2=11.6; p0.001). Correlations of the lumbar spine T-score and IL-6 (r=-0.29; p=0.03), IL-8 (r=-0.29; p=0.04) were revealed. Correlations between sarcopenia and the concentration of cytokines in the in the colon mucosa in CP were determined (IL-2: r=0.44; p0.001; IL-6: r=0.48; p0.001; IL-8: r=0.42; p0.001).
Conclusion. The development of osteopenia and sarcopenia syndromes in CP is interrelated and associated with both traditional risk factors and an increased concentration of cytokines in the in the colon mucosa.
Collapse
|
9
|
Zhou CH, Meng YT, Xu JJ, Fang X, Zhao JL, Zhou W, Zhao J, Han JC, Zhang L, Wang KX, Hu LH, Liao Z, Zou WB, Li ZS, Zou DW. Altered diversity and composition of gut microbiota in Chinese patients with chronic pancreatitis. Pancreatology 2020; 20:16-24. [PMID: 31806503 DOI: 10.1016/j.pan.2019.11.013] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 11/14/2019] [Accepted: 11/23/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND/OBJECTIVES Gut microbiota alterations in chronic pancreatitis (CP) are seldomly described systematically. It is unknown whether pancreatic exocrine insufficiency (PEI) and different etiologies in patients with CP are associated with gut microbiota dysbiosis. METHODS The fecal microbiota of 69 healthy controls (HCs) and 71 patients with CP were compared to investigate gut microbiome alterations in CP and the relationship among gut microbiome dysbiosis, PEI and different etiologies. Fecal microbiomes were analyzed through 16S ribosomal RNA gene profiling, based on next-generation sequencing. Pancreatic exocrine function was evaluated by determining fecal elastase 1 activity. RESULTS Patients with CP showed gut microbiota dysbiosis with decreased diversity and richness, and taxa-composition changes. On the phylum level, the gut microbiome of the CP group showed lower Firmicutes and Actinobacteria abundances than the HC group and higher Proteobacteria abundances. The abundances of Escherichia-Shigella and other genera were high in gut microbiomes in the CP group, whereas that of Faecalibacterium was low. Kyoto Encyclopedia of Genes and Genomes pathways (lipopolysaccharide biosynthesis and bacterial invasion of epithelial cells) were predicted to be enriched in the CP group. Among the top 5 phyla and 8 genera (in terms of abundance), only Fusobacteria and Eubacterium rectale group showed significant differences between CP patients, with or without PEI. Correlation analysis showed that Bifidobacterium and Lachnoclostridium correlated positively with fecal elastase 1 (r = 0.2616 and 0.2486, respectively, P < 0.05). CONCLUSIONS The current findings indicate that patients with CP have gut microbiota dysbiosis that is partly affected by pancreatic exocrine function.
Collapse
Affiliation(s)
- Chun-Hua Zhou
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University, No.168, Changhai Road, Yangpu District, Shanghai, 200433, China; Shanghai Institute of Pancreatic Diseases, Shanghai, China; Department of Gastroenterology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No.197, Rui Jin Er Road, Shanghai, 200025, China
| | - Yu-Ting Meng
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University, No.168, Changhai Road, Yangpu District, Shanghai, 200433, China; Shanghai Institute of Pancreatic Diseases, Shanghai, China
| | - Jia-Jia Xu
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University, No.168, Changhai Road, Yangpu District, Shanghai, 200433, China; Shanghai Institute of Pancreatic Diseases, Shanghai, China
| | - Xue Fang
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University, No.168, Changhai Road, Yangpu District, Shanghai, 200433, China; Shanghai Institute of Pancreatic Diseases, Shanghai, China
| | - Jiu-Long Zhao
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University, No.168, Changhai Road, Yangpu District, Shanghai, 200433, China; Shanghai Institute of Pancreatic Diseases, Shanghai, China
| | - Wei Zhou
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University, No.168, Changhai Road, Yangpu District, Shanghai, 200433, China; Shanghai Institute of Pancreatic Diseases, Shanghai, China
| | - Jianhua Zhao
- Shanghai Majorbio Bio-pharm Technology Co., Ltd., China
| | - Ji-Chen Han
- Shanghai Majorbio Bio-pharm Technology Co., Ltd., China
| | - Ling Zhang
- Department of Gastroenterology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No.197, Rui Jin Er Road, Shanghai, 200025, China
| | - Kai-Xuan Wang
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University, No.168, Changhai Road, Yangpu District, Shanghai, 200433, China; Shanghai Institute of Pancreatic Diseases, Shanghai, China
| | - Liang-Hao Hu
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University, No.168, Changhai Road, Yangpu District, Shanghai, 200433, China; Shanghai Institute of Pancreatic Diseases, Shanghai, China
| | - Zhuan Liao
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University, No.168, Changhai Road, Yangpu District, Shanghai, 200433, China; Shanghai Institute of Pancreatic Diseases, Shanghai, China
| | - Wen-Bin Zou
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University, No.168, Changhai Road, Yangpu District, Shanghai, 200433, China; Shanghai Institute of Pancreatic Diseases, Shanghai, China.
| | - Zhao-Shen Li
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University, No.168, Changhai Road, Yangpu District, Shanghai, 200433, China; Shanghai Institute of Pancreatic Diseases, Shanghai, China.
| | - Duo-Wu Zou
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University, No.168, Changhai Road, Yangpu District, Shanghai, 200433, China; Shanghai Institute of Pancreatic Diseases, Shanghai, China; Department of Gastroenterology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No.197, Rui Jin Er Road, Shanghai, 200025, China.
| |
Collapse
|
10
|
Li H, Xiu M, Wang S, Brigstock DR, Sun L, Qu L, Gao R. Role of Gut-Derived Endotoxin on Type I Collagen Production in the Rat Pancreas After Chronic Alcohol Exposure. Alcohol Clin Exp Res 2017; 42:306-314. [PMID: 29121396 DOI: 10.1111/acer.13550] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 11/02/2017] [Indexed: 12/13/2022]
Affiliation(s)
- Hongyan Li
- Department of Hepatic, Biliary Pancreatic Medicine; First Hospital of Jilin University; Changchun China
| | - Ming Xiu
- Department of Hepatic, Biliary Pancreatic Medicine; First Hospital of Jilin University; Changchun China
| | - Shuhua Wang
- Department of Surgical Gastroenterolog; First Hospital of Jilin University; Changchun China
| | | | - Li Sun
- Department of Hepatic, Biliary Pancreatic Medicine; First Hospital of Jilin University; Changchun China
| | - Limei Qu
- Department of Hepatic, Biliary Pancreatic Medicine; First Hospital of Jilin University; Changchun China
| | - Runping Gao
- Department of Hepatic, Biliary Pancreatic Medicine; First Hospital of Jilin University; Changchun China
| |
Collapse
|
11
|
Memba R, Duggan SN, Ni Chonchubhair HM, Griffin OM, Bashir Y, O'Connor DB, Murphy A, McMahon J, Volcov Y, Ryan BM, Conlon KC. The potential role of gut microbiota in pancreatic disease: A systematic review. Pancreatology 2017; 17:867-874. [PMID: 28935288 DOI: 10.1016/j.pan.2017.09.002] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 09/04/2017] [Accepted: 09/05/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND Several studies have suggested a link between microbiota imbalance and some gastrointestinal, inflammatory and neoplastic diseases. However, the role in pancreatic diseases remain unclear. To evaluate the available evidence for pancreatic diseases, we undertook a systematic review. METHODS OVID Medline (1946-2017), EMBASE (1980-2017) and the Cochrane Central Register of Controlled Trials (CENTRAL Issue 3, 2017) were searched for studies on microbiota in pancreatic disease. We also searched the reference lists of retrieved papers, and conference proceedings. We excluded animal studies, reviews, and case reports. RESULTS A total of 2833 articles were retrieved. After screening and applying the exclusion criteria, 10 studies were included. Three studies showed lower levels of Bifidobacterium or Lactobacillus and higher levels of Enterobacteriaceae in chronic pancreatitis. Two of these studies were uncontrolled, and the third (controlled) study which compared patients with endocrine and exocrine insufficiency, reported that Bacteroidetes levels were lower in those patients without diabetes, while Bifidobacteria levels were higher in those without exocrine insufficiency. Only one study investigated acute pancreatitis, showing higher levels of Enterococcus and lower levels of Bifidobacterium versus healthy participants. There was an overall association between pancreatic cancer and lower levels of Neisseria elongate, Streptococcus mitis and higher levels of Porphyromonas gingivalis and Granulicatella adiacens. CONCLUSIONS Current evidence suggests a possible link between microbiota imbalance and pancreatic cancer. Regarding acute and chronic pancreatitis, data are scarce, dysbiosis appears to be present in both conditions. However, further investigation is required to confirm these findings and to explore therapeutic possibilities.
Collapse
Affiliation(s)
- Robert Memba
- Professorial Surgical Unit, Department of Surgery, Trinity College Dublin, Tallaght Hospital, Dublin 24, Ireland.
| | - Sinead N Duggan
- Professorial Surgical Unit, Department of Surgery, Trinity College Dublin, Tallaght Hospital, Dublin 24, Ireland
| | - Hazel M Ni Chonchubhair
- Professorial Surgical Unit, Department of Surgery, Trinity College Dublin, Tallaght Hospital, Dublin 24, Ireland
| | - Oonagh M Griffin
- Professorial Surgical Unit, Department of Surgery, Trinity College Dublin, Tallaght Hospital, Dublin 24, Ireland
| | - Yasir Bashir
- Professorial Surgical Unit, Department of Surgery, Trinity College Dublin, Tallaght Hospital, Dublin 24, Ireland
| | - Donal B O'Connor
- Professorial Surgical Unit, Department of Surgery, Trinity College Dublin, Tallaght Hospital, Dublin 24, Ireland
| | - Anne Murphy
- Tallaght Hospital Library, Tallaght Hospital, Dublin, Ireland
| | - Jean McMahon
- Tallaght Hospital Library, Tallaght Hospital, Dublin, Ireland
| | - Yuri Volcov
- Department of Molecular and Translational Medicine, Trinity College Dublin, St James Hospital, Dublin 8, Ireland
| | - Barbara M Ryan
- Department of Gastroenterology, Tallaght Hospital, Dublin 24, Ireland
| | - Kevin C Conlon
- Professorial Surgical Unit, Department of Surgery, Trinity College Dublin, Tallaght Hospital, Dublin 24, Ireland
| |
Collapse
|
12
|
Husain SZ, Morinville V, Pohl J, Abu-El-Haija M, Bellin MD, Freedman S, Hegyi P, Heyman MB, Himes R, Ooi CY, Schwarzenberg SJ, Usatin D, Uc A. Toxic-metabolic Risk Factors in Pediatric Pancreatitis: Recommendations for Diagnosis, Management, and Future Research. J Pediatr Gastroenterol Nutr 2016; 62:609-617. [PMID: 26594832 PMCID: PMC4805437 DOI: 10.1097/mpg.0000000000001035] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Pancreatitis in children can result from metabolic and toxic risk factors, but the evidence linking these factors is sparse. We review the evidence for association or causality of these risk factors in pancreatitis, discuss management strategies, and their rationale. We conducted a review of the pediatric pancreatitis literature with respect to the following risk factors: hyperlipidemia, hypercalcemia, chronic renal failure, smoking exposure, alcohol, and medications. Areas of additional research were identified. Hypertriglyceridemia of 1000 mg/dL or greater poses an absolute risk for pancreatitis; persistent elevations of calcium are predisposing. Further research is necessary to determine whether end-stage renal disease leads to increased pancreatitis in children similar to adults. It is unknown whether cigarette smoking exposure, which clearly increases risk in adults, also increases risk in children. The role of alcohol in pediatric pancreatitis, whether direct or modifying, needs to be elucidated. The evidence supporting most cases of medication-induced pancreatitis is poor. Drug structure, improper handling of drug by host, and bystander status may be implicated. Other pancreatitis risk factors must be sought in all cases. The quality of evidence supporting causative role of various toxic and metabolic factors in pediatric pancreatitis is variable. Careful phenotyping is essential, including search for other etiologic risk factors. Directed therapy includes correction/removal of any agent identified, and general supportive measures. Further research is necessary to improve our understanding of these pancreatitis risk factors in children.
Collapse
Affiliation(s)
- Sohail Z Husain
- *Children's Hospital of Pittsburgh, Pittsburgh, PA †Montreal Children's Hospital, McGill University, Montreal, QC, Canada ‡University of Utah, Salt Lake City §Cincinnati Children's Hospital Medical Center, Cincinnati, OH ||University of Minnesota Masonic Children's Hospital, Minneapolis ¶Harvard Medical School, Boston, MA #First Department of Medicine, University of Szeged and HAS-SZTE Monument Gastroenterology Multidisciplinary Research Group, Szeged, Hungary **University of California at San Francisco, San Francisco ††Baylor College of Medicine, Houston, TX ‡‡Discipline of Paediatrics, School of Women's and Children's Health, Medicine, University of New South Wales and Sydney Children's Hospital Randwick, Sydney, Australia §§University of Iowa Carver College of Medicine, Iowa City
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Bagyánszki M, Bódi N. Gut region-dependent alterations of nitrergic myenteric neurons after chronic alcohol consumption. World J Gastrointest Pathophysiol 2015; 6:51-57. [PMID: 26301118 PMCID: PMC4540706 DOI: 10.4291/wjgp.v6.i3.51] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 03/27/2015] [Accepted: 06/02/2015] [Indexed: 02/06/2023] Open
Abstract
Chronic alcohol abuse damages nearly every organ in the body. The harmful effects of ethanol on the brain, the liver and the pancreas are well documented. Although chronic alcohol consumption causes serious impairments also in the gastrointestinal tract like altered motility, mucosal damage, impaired absorption of nutrients and inflammation, the effects of chronically consumed ethanol on the enteric nervous system are less detailed. While the nitrergic myenteric neurons play an essential role in the regulation of gastrointestinal peristalsis, it was hypothesised, that these neurons are the first targets of consumed ethanol or its metabolites generated in the different gastrointestinal segments. To reinforce this hypothesis the effects of ethanol on the gastrointestinal tract was investigated in different rodent models with quantitative immunohistochemistry, in vivo and in vitro motility measurements, western blot analysis, evaluation of nitric oxide synthase enzyme activity and bio-imaging of nitric oxide synthesis. These results suggest that chronic alcohol consumption did not result significant neural loss, but primarily impaired the nitrergic pathways in gut region-dependent way leading to disturbed gastrointestinal motility. The gut segment-specific differences in the effects of chronic alcohol consumption highlight the significance the ethanol-induced neuronal microenvironment involving oxidative stress and intestinal microbiota.
Collapse
|
14
|
Holmes E, Wijeyesekera A, Taylor-Robinson SD, Nicholson JK. The promise of metabolic phenotyping in gastroenterology and hepatology. Nat Rev Gastroenterol Hepatol 2015. [PMID: 26194948 DOI: 10.1038/nrgastro.2015.114] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Disease risk and treatment response are determined, at the individual level, by a complex history of genetic and environmental interactions, including those with our endogenous microbiomes. Personalized health care requires a deep understanding of patient biology that can now be measured using a range of '-omics' technologies. Patient stratification involves the identification of genetic and/or phenotypic disease subclasses that require different therapeutic strategies. Stratified medicine approaches to disease diagnosis, prognosis and therapeutic response monitoring herald a new dimension in patient care. Here, we explore the potential value of metabolic profiling as applied to unmet clinical needs in gastroenterology and hepatology. We describe potential applications in a number of diseases, with emphasis on large-scale population studies as well as metabolic profiling on the individual level, using spectrometric and imaging technologies that will leverage the discovery of mechanistic information and deliver novel health care solutions to improve clinical pathway management.
Collapse
Affiliation(s)
- Elaine Holmes
- Division of Computational and Systems Medicine, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London SW7 2AZ, UK
| | - Anisha Wijeyesekera
- Division of Computational and Systems Medicine, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London SW7 2AZ, UK
| | | | - Jeremy K Nicholson
- MRC-NIHR National Phenome Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London SW7 2AZ, UK
| |
Collapse
|
15
|
Hoque R, Mehal WZ. Inflammasomes in pancreatic physiology and disease. Am J Physiol Gastrointest Liver Physiol 2015; 308:G643-51. [PMID: 25700081 PMCID: PMC4398840 DOI: 10.1152/ajpgi.00388.2014] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 02/10/2015] [Indexed: 01/31/2023]
Abstract
In this review we summarize the role of inflammasomes in pancreatic physiology and disease with a focus on acute pancreatitis where much recent progress has been made. New findings have identified inducers of and cell specificity of inflammasome component expression in the pancreas, the contribution of inflammasome-regulated effectors to pancreatitis, and metabolic regulation of inflammasome activation, which are strong determinants of injury in pancreatitis. New areas of pancreatic biology will be highlighted in the context of our evolving understanding of gut microbiome- and injury-induced inflammasome priming, pyroptosis, and innate immune-mediated regulation of cell metabolism.
Collapse
Affiliation(s)
- Rafaz Hoque
- 1Section of Digestive Diseases, Yale University School of Medicine, New Haven, Connecticut; and
| | - Wajahat Z. Mehal
- 1Section of Digestive Diseases, Yale University School of Medicine, New Haven, Connecticut; and ,2Section of Digestive Diseases, Department of Veterans Affairs Connecticut Healthcare, West Haven, Connecticut
| |
Collapse
|
16
|
Intestinal CYP2E1: A mediator of alcohol-induced gut leakiness. Redox Biol 2014; 3:40-6. [PMID: 25462064 PMCID: PMC4297927 DOI: 10.1016/j.redox.2014.10.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 10/13/2014] [Accepted: 10/15/2014] [Indexed: 02/07/2023] Open
Abstract
Chronic alcohol use can result in many pathological effects including alcoholic liver disease (ALD). While alcohol is necessary for the development of ALD, only 20-30% of alcoholics develop alcoholic steatohepatitis (ASH) with progressive liver disease leading to cirrhosis and liver failure (ALD). This suggests that while chronic alcohol consumption is necessary it is not sufficient to induce clinically relevant liver damage in the absence of a secondary risk factor. Studies in rodent models and alcoholic patients show that increased intestinal permeability to microbial products like endotoxin play a critical role in promoting liver inflammation in ALD pathogenesis. Therefore identifying mechanisms of alcohol-induced intestinal permeability is important in identifying mechanisms of ALD and for designing new avenues for therapy. Cyp2e1 is a cytochrome P450 enzyme that metabolizes alcohol has been shown to be upregulated by chronic alcohol use and to be a major source of oxidative stress and liver injury in alcoholics and in animal and in vitro models of chronic alcohol use. Because Cyp2e1 is also expressed in the intestine and is upregulated by chronic alcohol use, we hypothesized it could play a role in alcohol-induced intestinal hyperpermeability. Our in vitro studies with intestinal Caco-2 cells and in mice fed alcohol showed that circadian clock proteins CLOCK and PER2 are required for alcohol-induced permeability. We also showed that alcohol increases Cyp2e1 protein and activity but not mRNA in Caco-2 cells and that an inhibitor of oxidative stress or siRNA knockdown of Cyp2e1 prevents the increase in CLOCK or PER2 proteins and prevents alcohol-induced hyperpermeability. With our collaborators we have also shown that Cyp2e1 knockout mice are resistant to alcohol-induced gut leakiness and liver inflammation. Taken together our data support a novel Cyp2e1-circadian clock protein mechanism for alcohol-induced gut leakiness that could provide new avenues for therapy of ALD.
Collapse
|