1
|
Suolang Q, Basang Z, Silang W, Nima C, Yang Q, Da W. Study on intestinal microbial communities of three different cattle populations on Qinghai-Tibet Plateau. PLoS One 2025; 20:e0312314. [PMID: 39903702 PMCID: PMC11793767 DOI: 10.1371/journal.pone.0312314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 10/04/2024] [Indexed: 02/06/2025] Open
Abstract
The Tibetan cattle, indispensable·animals on the Qinghai-Tibet Plateau, have become a focal point for the region's economic development. As such, the hybridization of these cattle has been recognized as a pivotal strategy to enhance the local cattle industry. However, research on the gut microbiota of Tibetan hybrid cattle remains scarce. Based on this, we conducted a comparative analysis of the gut microbiota and its functional implications across three distinct cattle populations: two the hybrid cattle populations (Tibetan local cattle × Holstein cattle, TH and Tibetan local cattle × Jersey cattle, TJ) and one the Tibetan locoal cattle population (BL). Bacteroidetes and Firmicutes dominate the gut microbiota across all populations at the phylum level. In addition, the predominant phyla in BL cattle were found to be Cyanobacteria, Verrucomicrobiota, and Actinobacteria, which may be one of the important reasons for the adaptability of Tibetan local cattle to the high-altitude environment of the Qinghai-Tibet Plateau. Further analysis identified specific biomarkers associated with the immune systems of BL cattle, including Bacteroidales_RF16, Coriobacterium, and Muribaculaceae. In contrast, TH cattle are primarily dominated by Oscillospiraceae and Clostridia_UCG_014, and TJ cattle are mainly dominated by Christensenellaceae and Gammaproteobacteria. KEGG enrichment analysis revealed that BL and TH cattle showed significant enrichment in the immune system, energy metabolism, and amino acid metabolism-related pathways compared with TJ cattle. Overall, these results suggest that BL and TH cattle demonstrate enhanced adaptability compared to TJ cattle, and indicate that intestinal microbiota of cattle at different altitudes and breeds have diverse structures and functions. Our study presents a new perspective on the role of the microbiome in the hybridization and enhancement of Tibetan cattle.
Collapse
Affiliation(s)
- Quji Suolang
- Institute of Animal Husbandry and Veterinary Science, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, China
| | - Zhuzha Basang
- Institute of Animal Husbandry and Veterinary Science, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, China
| | - Wangmu Silang
- Institute of Animal Husbandry and Veterinary Science, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, China
| | - Cangjue Nima
- Institute of Animal Husbandry and Veterinary Science, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, China
| | - Qiwen Yang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Xianyang, Shaanxi, China
| | - Wa Da
- Institute of Animal Husbandry and Veterinary Science, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, China
| |
Collapse
|
2
|
Zhang Y, Wei X, Jiang S, Gao W, Wang K, Wang H, Wang H, Si N, Zhou Y, Luo K, Wang M, Liu Y, Chen L, Ni L, Zhao H. Jianwei Xiaoshi oral liquid attenuates high-calorie diet-induced dyspepsia in immature rats via regulating the pancreatic secretion pathway and maintaining the homeostasis of intestinal microbiota. Chin Med 2025; 20:6. [PMID: 39755683 DOI: 10.1186/s13020-024-01052-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 12/19/2024] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND Jianwei Xiaoshi oral liquid (JWXS), a classical traditional prescription comprising various edible medicinal plants, has demonstrated significant efficacy in treating paediatric indigestion. It originates from Jianpi Pill, which is developed in the Ming Dynasty and nourishes the spleen and regulates gastrointestinal function. However, the specific molecular mechanisms involved remain unclear. METHODS To elucidate the material base of JWXS and its underlying mechanism in treating dyspepsia, the UHPLC-Q-Orbitrap HRMS method and network pharmacology were utilized. This was followed by pharmacological experiments, transcriptomics analyses and gut microbiota studies to further investigate the effects of JWXS on dyspepsia. RESULTS A total of 105 compounds, mainly flavonoids, alkaloids, organic acids and cyclic peptides, were identified. According to the five principles of generic drug properties, 43 candidate compounds were screened out. Their efficacy was verified through gastric emptying and intestinal propulsion experiments. Transcriptomic analysis revealed that JWXS primarily alleviated dyspepsia symptoms by regulating the secretion of 8 key proteins in the pancreatic secretion pathway. The differences in the gut microbiota, as identified through 16S rRNA and ITS2 sequencing, were subsequently more pronounced than those observed in the bacterial microbiota of the model group. In total, 15 differential bacteria and 16 differential fungi were identified. Targeted metabolomics analysis of SCFAs revealed a significant decrease in valeric acid (VA), acetic acid (AA), and isovaleric acid (IVA) levels in the model group, which were restored to the corresponding levels after the administration of JWXS. Correlation analysis revealed that VA, AA, and IVA were positively correlated with Lactobacillus and Bacteroides, and negatively correlated with Aspergillus and Candida. This further suggested that JWXS might alleviate symptoms of indigestion by regulating the composition of the microbiota, increasing the variety and quantity of beneficial bacteria, reducing fungal contamination, and further increasing the levels of SCFAs in the body. CONCLUSION JWXS improved functional dyspepsia in immature rats via a mechanism involving the regulation of the secretion of 8 key proteins in the pancreatic secretion pathway and the amelioration of flora disorders.
Collapse
Affiliation(s)
- Yan Zhang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Xiaolu Wei
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Shan Jiang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Wenya Gao
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Kun Wang
- Jichuan Pharmaceutical Group Co., Ltd., Jiangsu, 22544, China
| | - Hongjie Wang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Huijun Wang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Nan Si
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yanyan Zhou
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Keke Luo
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Mengxiao Wang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yuyang Liu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Lihua Chen
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Liqi Ni
- Jichuan Pharmaceutical Group Co., Ltd., Jiangsu, 22544, China.
| | - Haiyu Zhao
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| |
Collapse
|
3
|
Akshintala VS, Boparai IS, Barakat MT, Husain SZ. Post Endoscopic Retrograde Cholangiopancreatography Pancreatitis: Novel Mechanisms and Prevention by Drugs. United European Gastroenterol J 2024. [PMID: 39711464 DOI: 10.1002/ueg2.12732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 12/24/2024] Open
Abstract
Endoscopic retrograde cholangiopancreatography (ERCP) is becoming more common than first-line therapy for pancreaticobiliary duct disorders. However, post-ERCP pancreatitis is the most common complication of ERCPs, and affects about 10% of cases. In this review, we provide an overview of the mechanisms purported to cause post-ERCP pancreatitis as well as associated risk factors. We discuss measures that are in practice for post-ERCP pancreatitis pharmaco-prophylaxis, along with advances in the pipeline. We emphasize that there is still a pressing need to narrow the incidence of post-ERCP pancreatitis and that a mechanistic approach may reveal the greatest benefit from utilizing a combination of targets.
Collapse
Affiliation(s)
- Venkata S Akshintala
- Division of Gastroenterology, Department of Internal Medicine, Johns Hopkins Medical Institutions, Baltimore, Maryland, USA
| | - Ibadat S Boparai
- Division of Gastroenterology, Department of Internal Medicine, Johns Hopkins Medical Institutions, Baltimore, Maryland, USA
| | - Monique T Barakat
- Division of Gastroenterology, Department of Pediatrics, School of Medicine, Stanford University, Stanford, California, USA
| | - Sohail Z Husain
- Division of Gastroenterology, Department of Pediatrics, School of Medicine, Stanford University, Stanford, California, USA
| |
Collapse
|
4
|
Daniel N, Farinella R, Chatziioannou AC, Jenab M, Mayén AL, Rizzato C, Belluomini F, Canzian F, Tavanti A, Keski-Rahkonen P, Hughes DJ, Campa D. Genetically predicted gut bacteria, circulating bacteria-associated metabolites and pancreatic ductal adenocarcinoma: a Mendelian randomisation study. Sci Rep 2024; 14:25144. [PMID: 39448785 PMCID: PMC11502931 DOI: 10.1038/s41598-024-77431-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 10/22/2024] [Indexed: 10/26/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has high mortality and rising incidence rates. Recent data indicate that the gut microbiome and associated metabolites may play a role in the development of PDAC. To complement and inform observational studies, we investigated associations of genetically predicted abundances of individual gut bacteria and genetically predicted circulating concentrations of microbiome-associated metabolites with PDAC using Mendelian randomisation (MR). Gut microbiome-associated metabolites were identified through a comprehensive search of Pubmed, Exposome Explorer and Human Metabolome Database. Single Nucleotide Polymorphisms (SNPs) associated by Genome-Wide Association Studies (GWAS) with circulating levels of 109 of these metabolites were collated from Pubmed and the GWAS catalogue. SNPs for 119 taxonomically defined gut genera were selected from a meta-analysis performed by the MiBioGen consortium. Two-sample MR was conducted using GWAS summary statistics from the Pancreatic Cancer Cohort Consortium (PanScan) and the Pancreatic Cancer Case-Control Consortium (PanC4), including a total of 8,769 cases and 7,055 controls. Inverse variance-weighted MR analyses were performed along with sensitivity analyses to assess potential violations of MR assumptions. Nominally significant associations were noted for genetically predicted circulating concentrations of mannitol (odds ratio per standard deviation [ORSD] = 0.97; 95% confidence interval [CI]: 0.95-0.99, p = 0.006), methionine (ORSD= 0.97; 95%CI: 0.94-1.00, p = 0.031), stearic acid (ORSD= 0.93; 95%CI: 0.87-0.99, p = 0.027), carnitine = (ORSD=1.01; 95% CI: 1.00-1.03, p = 0.027), hippuric acid (ORSD= 1.02; 95%CI: 1.00-1.04, p = 0.038) and 3-methylhistidine (ORSD= 1.05; 95%CI: 1.01-1.10, p = 0.02). Two gut microbiome genera were associated with reduced PDAC risk; Clostridium sensu stricto 1 (OR: 0.88; 95%CI: 0.78-0.99, p = 0.027) and Romboutsia (OR: 0.87; 95%CI: 0.80-0.96, p = 0.004). These results, though based only on genetically predicted gut microbiome characteristics and circulating bacteria-related metabolite concentrations, provide evidence for causal associations with pancreatic carcinogenesis.
Collapse
Affiliation(s)
- Neil Daniel
- Molecular Epidemiology of Cancer Group, UCD Conway Institute, School of Biomedical and Biomolecular Sciences, University College Dublin, Dublin, Ireland
| | | | | | - Mazda Jenab
- Nutrition and Metabolism Branch, International Agency for Research on Cancer (IARC), Lyon, France
| | - Ana-Lucia Mayén
- Nutrition and Metabolism Branch, International Agency for Research on Cancer (IARC), Lyon, France
| | | | | | - Federico Canzian
- Genomic Epidemiology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Pekka Keski-Rahkonen
- Nutrition and Metabolism Branch, International Agency for Research on Cancer (IARC), Lyon, France
| | - David J Hughes
- Molecular Epidemiology of Cancer Group, UCD Conway Institute, School of Biomedical and Biomolecular Sciences, University College Dublin, Dublin, Ireland.
| | - Daniele Campa
- Department of Biology, University of Pisa, Pisa, Italy
| |
Collapse
|
5
|
Hong J, Fu Y, Chen X, Zhang Y, Li X, Li T, Liu Y, Fan M, Lin R. Gut microbiome changes associated with chronic pancreatitis and pancreatic cancer: a systematic review and meta-analysis. Int J Surg 2024; 110:5781-5794. [PMID: 38847785 PMCID: PMC11392207 DOI: 10.1097/js9.0000000000001724] [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: 04/03/2024] [Accepted: 05/19/2024] [Indexed: 09/15/2024]
Abstract
BACKGROUND The study of changes in the microbiome in chronic pancreatitis (CP) and pancreatic ductal adenocarcinoma (PDAC) holds significant potential for developing noninvasive diagnostic tools as well as innovative interventions to alter the progression of diseases. This systematic review and meta-analysis aimed to analyze in detail the taxonomic and functional characteristics of the gut microbiome in patients with CP and PDAC. METHODS Two researchers conducted a systematic search across public databases to gather all published research up to June 2023. Diversity and gut microbiota composition are the main outcomes the authors focus on. RESULTS This meta-analysis included 14 studies, involving a total of 1511 individuals in the PDAC ( n =285), CP ( n =342), and control ( n =649) groups. Our results show a significant difference in the composition of gut microbiota between PDAC/CP patients compared to healthy controls (HC), as evidenced by a slight decrease in α-diversity, including Shannon (SMD=-0.33; P =0.002 and SMD=-0.59; P <0.001, respectively) and a statistically significant β-diversity ( P <0.05). The pooled results showed that at the phylum level, the proportion of Firmicutes was lower in PDAC and CP patients than in HC patients. At the genus level, more than two studies demonstrated that four genera were significantly increased in PDAC patients compared to HC (e.g. Escherichia-Shigella and Veillonella ). CP patients had an increase in four genera (e.g. Escherichia-Shigella and Klebsiella ) and a decrease in eight genera (e.g. Coprococcus and Bifidobacterium ) compared to HC. Functional/metabolomics results from various studies also showed differences between PDAC/CP patients and HC. In addition, this study found no significant differences in gut microbiota between PDAC and CP patients. CONCLUSIONS Current evidence suggests changes in gut microbiota is associated with PDAC/CP, commonly reflected by a reduction in beneficial species and an increase in the pathogenic species. Further studies are needed to confirm these findings and explore therapeutic possibilities.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Mengke Fan
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Rong Lin
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| |
Collapse
|
6
|
Pezzino S, Sofia M, Mazzone C, Litrico G, Greco LP, Gallo L, La Greca G, Latteri S. Innovative treatments for obesity and NAFLD: A bibliometric study on antioxidants, herbs, phytochemicals, and natural compounds. Heliyon 2024; 10:e35498. [PMID: 39220898 PMCID: PMC11365328 DOI: 10.1016/j.heliyon.2024.e35498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 07/19/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024] Open
Abstract
The increasing scientific interest in antioxidants and naturally derived compounds as potential remedies for obesity and non-alcoholic fatty liver disease (NAFLD) has led to extensive research. The objective of this bibliometric analysis is to present an updated perspective on the topic of antioxidants, herbs, phytochemicals, and natural compounds, in the control of obesity and NAFLD, to identify new areas for future research. Publications from the years 2012-2022 were retrieved using the Scopus database. The research trends were analyzed using the Biblioshiny and VOSviewer tools. The field has seen a significant increase in research activity, as indicated by an annual growth rate of 10 % in the number of published manuscripts. China, Korea, and the USA emerged as the most prominent contributors in this specific field, supported by their notable volumes of publications and citations. The density analysis revealed that the most frequently occurring authors' keywords related to herbal species are, in rank order, Camelia sinensis, Momordica charantia, Curcuma longa, Ilex paraguariensis, Panax ginseng, Moringa oleifera, Garcinia cambogia, Garcinia mangostana, Zingiber officinale, and Cinnamomum verum. In the group of antioxidants, phytochemicals, and natural compounds, the top 10 were resveratrol, curcumin, quercetin, vitamin E, alpha-lipoic acid, vitamin C, chlorogenic acid, lycopene, fucoxanthin, and berberine. The co-occurrence analysis unveiled significant themes and potential trends, including a notable interest in the impact of herbal species, antioxidants, phytochemicals, and natural compounds on obesity and NAFLD through the modulation of the gut microbiome. Another recurring theme that arises, is the ongoing investigation of molecular targets that demonstrate anti-adipogenesis properties. The analysis presented in this study provides valuable insights for researchers investigating the efficacy of antioxidants, herbs, phytochemicals, and natural compounds in addressing obesity and NAFLD. Through the use of bibliometric methods, the study offers a comprehensive overview. Furthermore, the findings of this analysis can serve as a foundation for future research in this specific domain.
Collapse
Affiliation(s)
- Salvatore Pezzino
- Department of Surgical Sciences and Advanced Technologies “G. F. Ingrassia”, Cannizzaro Hospital, University of Catania, 95123, Catania, Italy
| | - Maria Sofia
- Department of Surgical Sciences and Advanced Technologies “G. F. Ingrassia”, Cannizzaro Hospital, University of Catania, 95123, Catania, Italy
| | - Chiara Mazzone
- Department of Surgical Sciences and Advanced Technologies “G. F. Ingrassia”, Cannizzaro Hospital, University of Catania, 95123, Catania, Italy
| | - Giorgia Litrico
- Department of Surgical Sciences and Advanced Technologies “G. F. Ingrassia”, Cannizzaro Hospital, University of Catania, 95123, Catania, Italy
| | - Luigi Piero Greco
- Department of Surgical Sciences and Advanced Technologies “G. F. Ingrassia”, Cannizzaro Hospital, University of Catania, 95123, Catania, Italy
| | - Luisa Gallo
- Department of Surgical Sciences and Advanced Technologies “G. F. Ingrassia”, Cannizzaro Hospital, University of Catania, 95123, Catania, Italy
| | - Gaetano La Greca
- Department of Surgical Sciences and Advanced Technologies “G. F. Ingrassia”, Cannizzaro Hospital, University of Catania, 95123, Catania, Italy
| | - Saverio Latteri
- Department of Surgical Sciences and Advanced Technologies “G. F. Ingrassia”, Cannizzaro Hospital, University of Catania, 95123, Catania, Italy
| |
Collapse
|
7
|
Zhang L, Li D, Zhang Y, Hu W, Lv H, Zhang X, Zhang H. Changes in the gut microbiota of esophageal carcinoma patients based on 16S rRNA gene sequencing: a systematic review and meta-analysis. Front Oncol 2024; 14:1366975. [PMID: 39267834 PMCID: PMC11390416 DOI: 10.3389/fonc.2024.1366975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 07/16/2024] [Indexed: 09/15/2024] Open
Abstract
Background This study conducts a systematic review through meta-analysis, comparing the composition and diversity of the gut microbiome in patients with esophageal cancer and healthy individuals, and explores the relationship between risk factors and related factors of esophageal cancer. Methods According to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA), we comprehensively searched the databases of PubMed, Web of Science, Embase, Cochrane Library. In addition, we applied the R programming language version 4.0.3 and Stata 15.1 software for data analysis. We also implemented the Newcastle-Ottawa Scale (NOS), funnel plot analysis, Egger's test, and Begg's test to assess the risk of bias. Results In this study, a total of 328 studies were identified through the literature search. Among them, 117 duplicate studies were removed, and 202 studies were excluded based on inclusion and exclusion criteria. Finally, 9 studies were included in the analysis, involving a total of 216 patients with esophageal carcinoma and 352 healthy controls. Four studies provided Chao1 index for quantitative consolidation (ES = 637.41, 95% CI: 549.16 to 725.66, p = 0.000, I2 = 98.2%). Two studies [27, 29] reported ACE index (ES = 438.89, 95% CI: 362.42 to 515.35, p = 0.000, I2 = 97%). Seven studies [26,27,29,30,32] reported the Shannon index for quantitative consolidation (ES = 4.38, 95% CI: 3.95 to 4.81, p = 0.000, I2 = 99%). At the phylum level, the abundance of Bacteroidetes(ES = 37.8, 95% CI: 25.75 to 49.85, p = 0.000, I2 = 87.2%) and Proteobacteria(ES = 7.48, 95% CI: 5.02 to 8.85, p = 0.04, I2 = 2.4%) have statistical difference between ESCC and HC. There was no significant difference between ESCC and HC in the abundance of genera(p>0.05). Conclusions This observational meta-analysis revealed that changes in the GM were correlated with esophageal carcinoma, and variations in some advantageous GM might involve regional differences. Additionally, the study aims to facilitate early diagnosis of esophageal cancer and improve screening and diagnostic efficiency.
Collapse
Affiliation(s)
- Li Zhang
- Department of Pharmacology, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Delin Li
- Department of Medical Equipment, Jinan Mental Health Center, Jinan, China
| | - Yongsheng Zhang
- Department of Health Management, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Wenqi Hu
- Department of Health Management, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Haoyue Lv
- Department of Health Management, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Xiaodong Zhang
- Postgraduate Department, Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, China
| | - Hongyu Zhang
- Department of Health Management, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| |
Collapse
|
8
|
Sarkar P, Chintaluri S, Sarkar S, Unnisa M, Jakkampudi A, Mulukutla AP, Kumari S, Reddy DN, Talukdar R. Evaluation of the Crosstalk Between the Host Mycobiome and Bacteriome in Patients with Chronic Pancreatitis. Indian J Microbiol 2024; 64:603-617. [PMID: 39011022 PMCID: PMC11246408 DOI: 10.1007/s12088-024-01207-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 01/14/2024] [Indexed: 07/17/2024] Open
Abstract
The human microbiome is a diverse consortium of microbial kingdoms that play pivotal roles in host health and diseases. We previously reported a dysbiotic bacteriome in chronic pancreatitis patients with diabetes (CPD) compared with patients with it's nondiabetic (CPND) phenotype. In this study, we extended our exploration to elucidate the intricate interactions between the mycobiome, bacteriome, and hosts' plasma metabolome with the disease phenotypes. A total of 25 participants (CPD, n = 7; CPND, n = 10; healthy control, n = 8) were recruited for the study. We observed elevated species richness in both the bacterial and fungal profiles within the CP diabetic cohort compared to the nondiabetic CP phenotype and healthy control cohorts. Notably, the CP group displayed heterogeneous fungal diversity, with only 40% of the CP nondiabetic patients and 20% of the CP diabetic patients exhibiting common core gut fungal profiles. Specific microbial taxa alterations were identified, including a reduction in Bifidobacterium adolescentis and an increase in the prevalence of Aspergillus penicilloides and Klebsiella sp. in the disease groups. In silico analysis revealed the enrichment of pathways related to lipopolysaccharide (LPS), apoptosis, and peptidase, as well as reduced counts of the genes responsible for carbohydrate metabolism in the CP groups. Additionally, distinct plasma metabolome signatures were observed, with CPD group exhibiting higher concentrations of sugars and glycerolipids, while the CPND cohort displayed elevated levels of amino acids in their blood. The fatty acid-binding protein (FABP) concentration was notably greater in the CPD group than in the HC group (4.220 vs. 1.10 ng/ml, p = 0.04). Furthermore, compared with healthy controls, disease groups exhibited fewer correlations between key fungal taxa (Aspergillus sp., Candida sp.) and bacterial taxa (Prevotella copri, Bifidobacteria sp., Rumminococcaceae). Our study unveils, for the first time, a dysbiotic mycobiome and emphasizes unique host bacterial-mycobial interactions in CP patient with diabetes, potentially influencing disease severity. These findings provide crucial insights for future mechanistic studies aiming to unravel the determinants of disease severity in this complex clinical context. Supplementary Information The online version contains supplementary material available at 10.1007/s12088-024-01207-8.
Collapse
Affiliation(s)
- Priyanka Sarkar
- Gut Microbiome Research Group, Wellcome-DBT (Indian Alliance) Lab, Asian Healthcare Foundation, Asian Institute of Gastroenterology (AIG Hospitals), Hyderabad, India
| | - Sreelekha Chintaluri
- Gut Microbiome Research Group, Wellcome-DBT (Indian Alliance) Lab, Asian Healthcare Foundation, Asian Institute of Gastroenterology (AIG Hospitals), Hyderabad, India
| | - Subhaleena Sarkar
- Gut Microbiome Research Group, Wellcome-DBT (Indian Alliance) Lab, Asian Healthcare Foundation, Asian Institute of Gastroenterology (AIG Hospitals), Hyderabad, India
| | - Misbah Unnisa
- Department of Medical Gastroenterology, Asian Institute of Gastroenterology (AIG Hospitals), Hyderabad, India
| | - Aparna Jakkampudi
- Gut Microbiome Research Group, Wellcome-DBT (Indian Alliance) Lab, Asian Healthcare Foundation, Asian Institute of Gastroenterology (AIG Hospitals), Hyderabad, India
| | - Ambika Prasanna Mulukutla
- Gut Microbiome Research Group, Wellcome-DBT (Indian Alliance) Lab, Asian Healthcare Foundation, Asian Institute of Gastroenterology (AIG Hospitals), Hyderabad, India
| | - Sneha Kumari
- Gut Microbiome Research Group, Wellcome-DBT (Indian Alliance) Lab, Asian Healthcare Foundation, Asian Institute of Gastroenterology (AIG Hospitals), Hyderabad, India
| | - D. Nageshwar Reddy
- Department of Medical Gastroenterology, Asian Institute of Gastroenterology (AIG Hospitals), Hyderabad, India
| | - Rupjyoti Talukdar
- Gut Microbiome Research Group, Wellcome-DBT (Indian Alliance) Lab, Asian Healthcare Foundation, Asian Institute of Gastroenterology (AIG Hospitals), Hyderabad, India
- Department of Medical Gastroenterology, Asian Institute of Gastroenterology (AIG Hospitals), Hyderabad, India
| |
Collapse
|
9
|
Boicean A, Ichim C, Todor SB, Anderco P, Popa ML. The Importance of Microbiota and Fecal Microbiota Transplantation in Pancreatic Disorders. Diagnostics (Basel) 2024; 14:861. [PMID: 38732276 PMCID: PMC11082979 DOI: 10.3390/diagnostics14090861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 04/16/2024] [Accepted: 04/18/2024] [Indexed: 05/13/2024] Open
Abstract
The role of the intestinal microbiota in the diagnosis and treatment of pancreatic diseases is increasingly significant. Consequently, fecal microbiota transplantation (FMT) is emerging as a promising therapeutic avenue for various pancreatic disorders, including cancer, pancreatitis, and type 1 diabetes (T1D). This innovative procedure entails transferring gut microbiota from healthy donors to individuals affected by pancreatic ailments with the potential to restore intestinal balance and alleviate associated symptoms. FMT represents a pioneering approach to improve patient outcomes in pancreatic diseases, offering tailored treatments customized to individual microbiomes and specific conditions. Recent research highlights the therapeutic benefits of targeting the gut microbiota for personalized interventions in pancreatic disorders. However, a comprehensive understanding of the intricate interplay between gut microbiota and pancreatic physiology warrants further investigation. The necessity for additional studies and research endeavors remains crucial, especially in elucidating both adult and pediatric cases affected by pathological pancreatic conditions.
Collapse
Affiliation(s)
| | - Cristian Ichim
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (A.B.); (S.B.T.); (P.A.); (M.L.P.)
| | | | | | | |
Collapse
|
10
|
Xia Y, Sun M, Huang H, Jin WL. Drug repurposing for cancer therapy. Signal Transduct Target Ther 2024; 9:92. [PMID: 38637540 PMCID: PMC11026526 DOI: 10.1038/s41392-024-01808-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/05/2024] [Accepted: 03/19/2024] [Indexed: 04/20/2024] Open
Abstract
Cancer, a complex and multifactorial disease, presents a significant challenge to global health. Despite significant advances in surgical, radiotherapeutic and immunological approaches, which have improved cancer treatment outcomes, drug therapy continues to serve as a key therapeutic strategy. However, the clinical efficacy of drug therapy is often constrained by drug resistance and severe toxic side effects, and thus there remains a critical need to develop novel cancer therapeutics. One promising strategy that has received widespread attention in recent years is drug repurposing: the identification of new applications for existing, clinically approved drugs. Drug repurposing possesses several inherent advantages in the context of cancer treatment since repurposed drugs are typically cost-effective, proven to be safe, and can significantly expedite the drug development process due to their already established safety profiles. In light of this, the present review offers a comprehensive overview of the various methods employed in drug repurposing, specifically focusing on the repurposing of drugs to treat cancer. We describe the antitumor properties of candidate drugs, and discuss in detail how they target both the hallmarks of cancer in tumor cells and the surrounding tumor microenvironment. In addition, we examine the innovative strategy of integrating drug repurposing with nanotechnology to enhance topical drug delivery. We also emphasize the critical role that repurposed drugs can play when used as part of a combination therapy regimen. To conclude, we outline the challenges associated with repurposing drugs and consider the future prospects of these repurposed drugs transitioning into clinical application.
Collapse
Affiliation(s)
- Ying Xia
- Center for Clinical Laboratories, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, PR China
- The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, PR China
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, 550004, PR China
- Division of Gastroenterology and Hepatology, Department of Medicine and, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Ming Sun
- Center for Clinical Laboratories, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, PR China
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, 550004, PR China
| | - Hai Huang
- Center for Clinical Laboratories, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, PR China.
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, 550004, PR China.
| | - Wei-Lin Jin
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, PR China.
| |
Collapse
|
11
|
Qiu YF, Ye J, Xie JJ, Mao XT, Liu YL, Fang Q, Qian YY, Zou WB, Cao Y, Liao Z. Pancreatitis affects gut microbiota via metabolites and inflammatory cytokines: an exploratory two-step Mendelian randomisation study. Mol Genet Genomics 2024; 299:36. [PMID: 38492113 PMCID: PMC10944441 DOI: 10.1007/s00438-024-02125-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 02/09/2024] [Indexed: 03/18/2024]
Abstract
Previous studies have observed relationships between pancreatitis and gut microbiota; however, specific changes in gut microbiota abundance and underlying mechanisms in pancreatitis remain unknown. Metabolites are important for gut microbiota to fulfil their biological functions, and changes in the metabolic and immune environments are closely linked to changes in microbiota abundance. We aimed to clarify the mechanisms of gut-pancreas interactions and explore the possible role of metabolites and the immune system. To this end, we conducted two-sample Mendelian randomisation (MR) analysis to evaluate the casual links between four different types of pancreatitis and gut microbiota, metabolites, and inflammatory cytokines. A two-step MR analysis was conducted to further evaluate the probable mediating pathways involving metabolites and inflammatory cytokines in the causal relationship between pancreatitis and gut microbiota. In total, six potential mediators were identified in the causal relationship between pancreatitis and gut microbiota. Nineteen species of gut microbiota and seven inflammatory cytokines were genetically associated with the four types of pancreatitis. Metabolites involved in glucose and amino acid metabolisms were genetically associated with chronic pancreatitis, and those involved in lipid metabolism were genetically associated with acute pancreatitis. Our study identified alterations in the gut microbiota, metabolites, and inflammatory cytokines in pancreatitis at the genetic level and found six potential mediators of the pancreas-gut axis, which may provide insights into the precise diagnosis of pancreatitis and treatment interventions for gut microbiota to prevent the exacerbation of pancreatitis. Future studies could elucidate the mechanism underlying the association between pancreatitis and the gut microbiota.
Collapse
Affiliation(s)
- Yi-Fan Qiu
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University/Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Jun Ye
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University/Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Jin-Jin Xie
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University/Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Xiao-Tong Mao
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University/Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Yi-Long Liu
- College of Basic Medicine Sciences, Second Military Medical University/Naval Medical University, Shanghai, China
| | - Qian Fang
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University/Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Yang-Yang Qian
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University/Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Wen-Bin Zou
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University/Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Yu Cao
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University/Naval Medical University, 168 Changhai Road, Shanghai, 200433, China.
| | - Zhuan Liao
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University/Naval Medical University, 168 Changhai Road, Shanghai, 200433, China.
| |
Collapse
|
12
|
Lupu VV, Bratu RM, Trandafir LM, Bozomitu L, Paduraru G, Gimiga N, Ghiga G, Forna L, Ioniuc I, Petrariu FD, Puha B, Lupu A. Exploring the Microbial Landscape: Gut Dysbiosis and Therapeutic Strategies in Pancreatitis-A Narrative Review. Biomedicines 2024; 12:645. [PMID: 38540258 PMCID: PMC10967871 DOI: 10.3390/biomedicines12030645] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 03/09/2024] [Accepted: 03/12/2024] [Indexed: 01/03/2025] Open
Abstract
The gut microbiota is emerging as an important contributor to the homeostasis of the human body through its involvement in nutrition and metabolism, protection against pathogens, and the development and modulation of the immune system. It has therefore become an important research topic in recent decades. Although the association between intestinal dysbiosis and numerous digestive pathologies has been thoroughly researched, its involvement in pancreatic diseases constitutes a novelty in the specialized literature. In recent years, growing evidence has pointed to the critical involvement of the pancreas in regulating the intestinal microbiota, as well as the impact of the intestinal microbiota on pancreatic physiology, which implies the existence of a bidirectional connection known as the "gut-pancreas axis". It is theorized that any change at either of these levels triggers a response in the other component, hence leading to the evolution of pancreatitis. However, there are not enough data to determine whether gut dysbiosis is an underlying cause or a result of pancreatitis; therefore, more research is needed in this area. The purpose of this narrative review is to highlight the role of gut dysbiosis in the pathogenesis of acute and chronic pancreatitis, its evolution, and the prospect of employing the microbiota as a therapeutic intervention for pancreatitis.
Collapse
Affiliation(s)
| | - Roxana Mihaela Bratu
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (V.V.L.); (L.M.T.); (L.B.); (N.G.); (G.G.); (L.F.); (I.I.); (F.D.P.); (B.P.); (A.L.)
| | | | | | - Gabriela Paduraru
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (V.V.L.); (L.M.T.); (L.B.); (N.G.); (G.G.); (L.F.); (I.I.); (F.D.P.); (B.P.); (A.L.)
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Kesh K, Tao J, Ghosh N, Jalodia R, Singh S, Dawra R, Roy S. Prescription opioids induced microbial dysbiosis worsens severity of chronic pancreatitis and drives pain hypersensitivity. Gut Microbes 2024; 16:2310291. [PMID: 38329115 PMCID: PMC10857465 DOI: 10.1080/19490976.2024.2310291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 01/22/2024] [Indexed: 02/09/2024] Open
Abstract
Opioids, such as morphine and oxycodone, are widely used for pain management associated with chronic pancreatitis (CP); however, their impact on the progression and pain sensitivity of CP has never been evaluated. This report investigates the impact of opioid use on the severity of CP, pain sensitivity, and the gut microbiome. C57BL/6 mice were divided into control, CP, CP with morphine/oxycodone, and either morphine or oxycodone alone groups. CP was induced by administration of caerulein (50ug/kg/h, i.p. hourly x7, twice a week for 10 weeks). The mouse-to-pancreas weight ratio, histology, and Sirius red staining were performed to measure CP severity. Tail flick and paw pressure assays were used to measure thermal and mechanical pain. DNA was extracted from the fecal samples and subjected to whole-genome shotgun sequencing. Germ-free mice were used to validate the role of gut microbiome in sensitizing acute pancreatic inflammation. Opioid treatment exacerbates CP by increasing pancreatic necrosis, fibrosis, and immune-cell infiltration. Opioid-treated CP mice exhibited enhanced pain hypersensitivity and showed distinct clustering of the gut microbiome compared to untreated CP mice, with severely compromised gut barrier integrity. Fecal microbiota transplantation (FMT) from opioid-treated CP mice into germ-free mice resulted in pancreatic inflammation in response to a suboptimal caerulein dose. Together, these analyses revealed that opioids worsen the severity of CP and induce significant alterations in pain sensitivity and the gut microbiome in a caerulein CP mouse model. Microbial dysbiosis plays an important role in sensitizing the host to pancreatic inflammation.
Collapse
Affiliation(s)
- Kousik Kesh
- Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Junyi Tao
- Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Nillu Ghosh
- Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Richa Jalodia
- Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Salma Singh
- Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Rajinder Dawra
- Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Sabita Roy
- Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| |
Collapse
|
14
|
Osadchuk A, Loranskaya I, Osadchuk M. Refractory dyspepsia syndrome as one of chronic pancreatitis markers. RUSSIAN JOURNAL OF PREVENTIVE MEDICINE AND PUBLIC HEALTH 2024; 27:112. [DOI: 10.17116/profmed202427031112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
15
|
Garcia-Gutierrez E, O’Mahony AK, Dos Santos RS, Marroquí L, Cotter PD. Gut microbial metabolic signatures in diabetes mellitus and potential preventive and therapeutic applications. Gut Microbes 2024; 16:2401654. [PMID: 39420751 PMCID: PMC11492678 DOI: 10.1080/19490976.2024.2401654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 07/08/2024] [Accepted: 09/03/2024] [Indexed: 10/19/2024] Open
Abstract
Diabetes mellitus can be subdivided into several categories based on origin and clinical characteristics. The most common forms of diabetes are type 1 (T1D), type 2 diabetes (T2D) and gestational diabetes mellitus (GDM). T1D and T2D are chronic diseases affecting around 537 million adults worldwide and it is projected that these numbers will increase by 12% over the next two decades, while GDM affects up to 30% of women during pregnancy, depending on diagnosis methods. These forms of diabetes have varied origins: T1D is an autoimmune disease, while T2D is commonly associated with, but not limited to, certain lifestyle patterns and GDM can result of a combination of genetic predisposition and pregnancy factors. Despite some pathogenic differences among these forms of diabetes, there are some common markers associated with their development. For instance, gut barrier impairment and inflammation associated with an unbalanced gut microbiota and their metabolites may be common factors in diabetes development and progression. Here, we summarize the microbial signatures that have been linked to diabetes, how they are connected to diet and, ultimately, the impact on metabolite profiles resulting from host-gut microbiota-diet interactions. Additionally, we summarize recent advances relating to promising preventive and therapeutic interventions focusing on the targeted modulation of the gut microbiota to alleviate T1D, T2D and GDM.
Collapse
Affiliation(s)
- Enriqueta Garcia-Gutierrez
- Food Biosciences Department, Teagasc Food Research Centre, Fermoy, Co. Cork, Ireland
- APC Microbiome Ireland, University College Cork, Co. Cork, Ireland
- VistaMilk SFI Research Centre, Fermoy, Co. Cork, Ireland
- Departamento de Ingeniería Agronómica, Instituto de Biotecnología Vegetal, ETSIA-Universidad Politécnica de Cartagena, Cartagena, Spain
| | - A. Kate O’Mahony
- Food Biosciences Department, Teagasc Food Research Centre, Fermoy, Co. Cork, Ireland
- APC Microbiome Ireland, University College Cork, Co. Cork, Ireland
- School of Microbiology, University College Cork, Co. Cork, Ireland
| | - Reinaldo Sousa Dos Santos
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, Elche, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Laura Marroquí
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, Elche, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Paul D. Cotter
- Food Biosciences Department, Teagasc Food Research Centre, Fermoy, Co. Cork, Ireland
- APC Microbiome Ireland, University College Cork, Co. Cork, Ireland
- VistaMilk SFI Research Centre, Fermoy, Co. Cork, Ireland
| |
Collapse
|
16
|
Zhu SJ, Ding Z. Association between gut microbiota and seven gastrointestinal diseases: A Mendelian randomized study. J Gene Med 2024; 26:e3623. [PMID: 37957025 DOI: 10.1002/jgm.3623] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 09/25/2023] [Accepted: 10/16/2023] [Indexed: 11/21/2023] Open
Abstract
BACKGROUND Observational research has shed light on the ability of gut microbes to influence the onset and progression of gastrointestinal diseases. The causal relationships between specific gut microbiomes and various gastrointestinal conditions, however, remain unknown. METHODS We investigated the relationship between gut microbiota and seven specific gastrointestinal disorders using a robust two-sample Mendelian randomization (MR) approach. The inverse variance-weighted (IVW) method was used as the primary analysis tool in our study. Furthermore, we conducted multiple sensitivity analyses to strengthen the robustness of our findings and ensure the reliability of the IVW method. RESULTS Our research has discovered significant links between the composition of gut microbiota and a variety of gastrointestinal ailments. We found compelling links between 13 gut microbiota and fatty liver, four gut microbiota and cirrhosis, eight gut microbiota and hepatocellular carcinoma, four gut microbiota and cholelithiasis, 12 gut microbiota and acute pancreatitis, eight gut microbiota and chronic pancreatitis, and 11 gut microbiota and pancreatic cancer. These findings shed light on the intricate relationship between gut microbes and the emergence of these specific gastrointestinal conditions. CONCLUSIONS The findings of this extensive study not only validate the potential role of specific gut microbiota in gastrointestinal diseases, but also fill a critical gap in previous research. The discovery of these specific gut microbiota is a significant step forward because they may serve as novel and promising biomarkers for both the prevention and treatment of gastrointestinal conditions.
Collapse
Affiliation(s)
- Shuang Jing Zhu
- Hepatobiliary Surgery, Chaohu Hospital of Anhui Medical University, Hefei, China
| | - Zhen Ding
- Hepatobiliary Surgery, Chaohu Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
17
|
Merali N, Chouari T, Terroire J, Jessel MD, Liu DSK, Smith JH, Wooldridge T, Dhillon T, Jiménez JI, Krell J, Roberts KJ, Rockall TA, Velliou E, Sivakumar S, Giovannetti E, Demirkan A, Annels NE, Frampton AE. Bile Microbiome Signatures Associated with Pancreatic Ductal Adenocarcinoma Compared to Benign Disease: A UK Pilot Study. Int J Mol Sci 2023; 24:16888. [PMID: 38069211 PMCID: PMC10706407 DOI: 10.3390/ijms242316888] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/18/2023] [Accepted: 11/24/2023] [Indexed: 12/18/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a very poor survival. The intra-tumoural microbiome can influence pancreatic tumourigenesis and chemoresistance and, therefore, patient survival. The role played by bile microbiota in PDAC is unknown. We aimed to define bile microbiome signatures that can effectively distinguish malignant from benign tumours in patients presenting with obstructive jaundice caused by benign and malignant pancreaticobiliary disease. Prospective bile samples were obtained from 31 patients who underwent either Endoscopic Retrograde Cholangiopancreatography (ERCP) or Percutaneous Transhepatic Cholangiogram (PTC). Variable regions (V3-V4) of the 16S rRNA genes of microorganisms present in the samples were amplified by Polymerase Chain Reaction (PCR) and sequenced. The cohort consisted of 12 PDAC, 10 choledocholithiasis, seven gallstone pancreatitis and two primary sclerosing cholangitis patients. Using the 16S rRNA method, we identified a total of 135 genera from 29 individuals (12 PDAC and 17 benign). The bile microbial beta diversity significantly differed between patients with PDAC vs. benign disease (Permanova p = 0.0173). The separation of PDAC from benign samples is clearly seen through unsupervised clustering of Aitchison distance. We found three genera to be of significantly lower abundance among PDAC samples vs. benign, adjusting for false discovery rate (FDR). These were Escherichia (FDR = 0.002) and two unclassified genera, one from Proteobacteria (FDR = 0.002) and one from Enterobacteriaceae (FDR = 0.011). In the same samples, the genus Streptococcus (FDR = 0.033) was found to be of increased abundance in the PDAC group. We show that patients with obstructive jaundice caused by PDAC have an altered microbiome composition in the bile compared to those with benign disease. These bile-based microbes could be developed into potential diagnostic and prognostic biomarkers for PDAC and warrant further investigation.
Collapse
Affiliation(s)
- Nabeel Merali
- Minimal Access Therapy Training Unit (MATTU), Royal Surrey County Hospital NHS Foundation Trust, Egerton Road, Guildford GU2 7XX, UK
- Department of Hepato-Pancreato-Biliary (HPB) Surgery, Royal Surrey County Hospital NHS Foundation Trust, Egerton Road, Guildford GU2 7XX, UK
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey, Guildford GU2 7WG, UK
| | - Tarak Chouari
- Department of Hepato-Pancreato-Biliary (HPB) Surgery, Royal Surrey County Hospital NHS Foundation Trust, Egerton Road, Guildford GU2 7XX, UK
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey, Guildford GU2 7WG, UK
| | - Julien Terroire
- Surrey Institute for People-Centred AI, University of Surrey, Guildford GU2 7XH, UK
- Section of Statistical Multi-Omics, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey, Guildford GU2 7WG, UK
| | - Maria-Danae Jessel
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey, Guildford GU2 7WG, UK
| | - Daniel S. K. Liu
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London W12 0NN, UK
| | - James-Halle Smith
- Hepatobiliary and Pancreatic Surgery Unit, Queen Elizabeth Hospital Birmingham, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TH, UK
| | - Tyler Wooldridge
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey, Guildford GU2 7WG, UK
| | - Tony Dhillon
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey, Guildford GU2 7WG, UK
| | - José I. Jiménez
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Jonathan Krell
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London W12 0NN, UK
| | - Keith J. Roberts
- Hepatobiliary and Pancreatic Surgery Unit, Queen Elizabeth Hospital Birmingham, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TH, UK
| | - Timothy A. Rockall
- Minimal Access Therapy Training Unit (MATTU), Royal Surrey County Hospital NHS Foundation Trust, Egerton Road, Guildford GU2 7XX, UK
| | - Eirini Velliou
- Centre for 3D Models of Health and Disease, Division of Surgery and Interventional Science, University College London (UCL), London W1W 7TY, UK
| | - Shivan Sivakumar
- Oncology Department, Institute of Immunology and Immunotherapy, Birmingham Medical School, University of Birmingham, Birmingham B15 2TT, UK
| | - Elisa Giovannetti
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
- Cancer Pharmacology Lab, AIRC Start Up Unit, Fondazione Pisana per la Scienza, San Giuliano Terme PI, 56017 Pisa, Italy
| | - Ayse Demirkan
- Surrey Institute for People-Centred AI, University of Surrey, Guildford GU2 7XH, UK
- Section of Statistical Multi-Omics, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey, Guildford GU2 7WG, UK
| | - Nicola E. Annels
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey, Guildford GU2 7WG, UK
| | - Adam E. Frampton
- Minimal Access Therapy Training Unit (MATTU), Royal Surrey County Hospital NHS Foundation Trust, Egerton Road, Guildford GU2 7XX, UK
- Department of Hepato-Pancreato-Biliary (HPB) Surgery, Royal Surrey County Hospital NHS Foundation Trust, Egerton Road, Guildford GU2 7XX, UK
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey, Guildford GU2 7WG, UK
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London W12 0NN, UK
| |
Collapse
|
18
|
Jiang Z, Mou Y, Wang H, Li L, Jin T, Wang H, Liu M, Jin W. Causal effect between gut microbiota and pancreatic cancer: a two-sample Mendelian randomization study. BMC Cancer 2023; 23:1091. [PMID: 37950180 PMCID: PMC10636952 DOI: 10.1186/s12885-023-11493-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 10/08/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND Gut microbiota (GM) comprises a vast and diverse community of microorganisms, and recent studies have highlighted the crucial regulatory roles of various GM and their secreted metabolites in pancreatic cancer (PC). However, the causal relationship between GM and PC has yet to be confirmed. METHODS In the present study, we used two-sample Mendelian randomization (MR) analysis to investigate the causal effect between GM and PC, with genome-wide association study (GWAS) from MiBioGen consortium as an exposure factor and PC GWAS data from FinnGen as an outcome factor. Inverse variance weighted (IVW) was used as the primary method for this study. RESULTS At the genus level, we observed that Senegalimassilia (OR: 0.635, 95% CI: 0.403-0.998, P = 0.049) exhibited a protective effect against PC, while Odoribacter (OR:1.899, 95%CI:1.157-3.116, P = 0.011), Ruminiclostridium 9(OR:1.976,95%CI:1.128-3.461, P = 0.017), Ruminococcaceae (UCG011)(OR:1.433, 95%CI:1.072-1.916, P = 0.015), and Streptococcus(OR:1.712, 95%CI:1.071-1.736, P = 0.025) were identified as causative factors for PC. Additionally, sensitivity analysis, Cochran's Q test, the Mendelian randomization pleiotropy residual sum and outlier (MR-PRESSO), and MR-Egger regression indicated no heterogeneity, horizontal pleiotropy, or reverse causality between GM and PC. CONCLUSIONS Our analysis establishes a causal effect between specific GM and PC, which may provide new insights into the potential pathogenic mechanisms of GM in PC and the assignment of effective therapeutic strategies.
Collapse
Grants
- WKJ-ZJ-2201 Scientific research fund of national health commision of China, Key health science and technology program of Zhejiang Province
- WKJ-ZJ-2201 Scientific research fund of national health commision of China, Key health science and technology program of Zhejiang Province
- WKJ-ZJ-2201 Scientific research fund of national health commision of China, Key health science and technology program of Zhejiang Province
- WKJ-ZJ-2201 Scientific research fund of national health commision of China, Key health science and technology program of Zhejiang Province
- WKJ-ZJ-2201 Scientific research fund of national health commision of China, Key health science and technology program of Zhejiang Province
- 2022C03099 Key Project of social welfare program of Zhejiang Science and Technology Department,'Lingyan'Program
- 2022C03099 Key Project of social welfare program of Zhejiang Science and Technology Department,'Lingyan'Program
- 2022C03099 Key Project of social welfare program of Zhejiang Science and Technology Department,'Lingyan'Program
- 2022C03099 Key Project of social welfare program of Zhejiang Science and Technology Department,'Lingyan'Program
- 2022C03099 Key Project of social welfare program of Zhejiang Science and Technology Department,'Lingyan'Program
- Key Project of social welfare program of Zhejiang Science and Technology Department,’Lingyan’Program
Collapse
Affiliation(s)
- Zhichen Jiang
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Yiping Mou
- Department of General Surgery, Devision of Dastroenterology and Pancreas, Affiliated People's Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
- Key Laboratory of Gastroenterology of Zhejiang Province, Hangzhou, 310014, Zhejiang, China
| | - Huiju Wang
- Department of General Surgery, Devision of Dastroenterology and Pancreas, Affiliated People's Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
- Key Laboratory of Gastroenterology of Zhejiang Province, Hangzhou, 310014, Zhejiang, China
| | - Li Li
- Department of General Surgery, Devision of Dastroenterology and Pancreas, Affiliated People's Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
- Key Laboratory of Gastroenterology of Zhejiang Province, Hangzhou, 310014, Zhejiang, China
| | - Tianyu Jin
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - He Wang
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Mingyang Liu
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Weiwei Jin
- Department of General Surgery, Devision of Dastroenterology and Pancreas, Affiliated People's Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China.
- Key Laboratory of Gastroenterology of Zhejiang Province, Hangzhou, 310014, Zhejiang, China.
| |
Collapse
|
19
|
Wu L, Hu J, Yi X, Lv J, Yao J, Tang W, Zhang S, Wan M. Gut microbiota interacts with inflammatory responses in acute pancreatitis. Therap Adv Gastroenterol 2023; 16:17562848231202133. [PMID: 37829561 PMCID: PMC10566291 DOI: 10.1177/17562848231202133] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 09/01/2023] [Indexed: 10/14/2023] Open
Abstract
Acute pancreatitis (AP) is one of the most common acute abdominal conditions, and its incidence has been increasing for years. Approximately 15-20% of patients develop severe AP (SAP), which is complicated by critical inflammatory injury and intestinal dysfunction. AP-associated inflammation can lead to the gut barrier and function damage, causing dysbacteriosis and facilitating intestinal microbiota migration. Pancreatic exocrine deficiency and decreased levels of antimicrobial peptides in AP can also lead to abnormal growth of intestinal bacteria. Meanwhile, intestinal microbiota migration influences the pancreatic microenvironment and affects the severity of AP, which, in turn, exacerbates the systemic inflammatory response. Thus, the interaction between the gut microbiota (GM) and the inflammatory response may be a key pathogenic feature of SAP. Treating either of these factors or breaking their interaction may offer some benefits for SAP treatment. In this review, we discuss the mechanisms of interaction of the GM and inflammation in AP and factors that can deteriorate or even cure both, including some traditional Chinese medicine treatments, to provide new methods for studying AP pathogenesis and developing therapies.
Collapse
Affiliation(s)
- Linjun Wu
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
- Hospital of Chinese Traditional Medicine of Leshan, Leshan, China
| | - Jing Hu
- Department of Integrated Traditional Chinese and Western Medicine, West China
- Hospital, Sichuan University, Chengdu, China
- Hospital of Chinese Traditional Medicine of Leshan, Leshan, China
| | - Xiaolin Yi
- Department of Integrated Traditional Chinese and Western Medicine, West China
- Hospital, Sichuan University, Chengdu, China
- Intensive Care Unit, Suining Municipal Hospital of TCM, Suining, China
| | - Jianqin Lv
- Department of Integrated Traditional Chinese and Western Medicine, West China
- Hospital, Sichuan University, Chengdu, China
| | - Jiaqi Yao
- Department of Integrated Traditional Chinese and Western Medicine, West China
- Hospital, Sichuan University, Chengdu, China
| | - Wenfu Tang
- Department of Integrated Traditional Chinese and Western Medicine, West China
- Hospital, Sichuan University, Chengdu, China
| | - Shu Zhang
- Department of Emergency Medicine, Emergency Medical Laboratory, West China
- Hospital, Sichuan University, Guo Xue Road 37, Chengdu 610041, Sichuan, China
| | - Meihua Wan
- Department of Integrated Traditional Chinese and Western Medicine, West China
- Hospital, Sichuan University, Guo Xue Road 37, Chengdu 610041, China
| |
Collapse
|
20
|
Glaubitz J, Asgarbeik S, Lange R, Mazloum H, Elsheikh H, Weiss FU, Sendler M. Immune response mechanisms in acute and chronic pancreatitis: strategies for therapeutic intervention. Front Immunol 2023; 14:1279539. [PMID: 37881430 PMCID: PMC10595029 DOI: 10.3389/fimmu.2023.1279539] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 10/02/2023] [Indexed: 10/27/2023] Open
Abstract
Acute pancreatitis (AP) is one of the most common inflammatory diseases of the gastrointestinal tract and a steady rising diagnosis for inpatient hospitalization. About one in four patients, who experience an episode of AP, will develop chronic pancreatitis (CP) over time. While the initiating causes of pancreatitis can be complex, they consistently elicit an immune response that significantly determines the severity and course of the disease. Overall, AP is associated with a significant mortality rate of 1-5%, which is caused by either an excessive pro-inflammation, or a strong compensatory inhibition of bacterial defense mechanisms which lead to a severe necrotizing form of pancreatitis. At the time-point of hospitalization the already initiated immune response is the only promising common therapeutic target to treat or prevent a severe disease course. However, the complexity of the immune response requires fine-balanced therapeutic intervention which in addition is limited by the fact that a significant proportion of patients is in danger of development or progress to recurrent and chronic disease. Based on the recent literature we survey the disease-relevant immune mechanisms and evaluate appropriate and promising therapeutic targets for the treatment of acute and chronic pancreatitis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Matthias Sendler
- Department of Medicine A, University Medicine, University of Greifswald, Greifswald, Germany
| |
Collapse
|
21
|
Pan LL, Ren ZN, Yang J, Li BB, Huang YW, Song DX, Li X, Xu JJ, Bhatia M, Zou DW, Zhou CH, Sun J. Gut microbiota controls the development of chronic pancreatitis: A critical role of short-chain fatty acids-producing Gram-positive bacteria. Acta Pharm Sin B 2023; 13:4202-4216. [PMID: 37799394 PMCID: PMC10547962 DOI: 10.1016/j.apsb.2023.08.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/21/2023] [Accepted: 06/13/2023] [Indexed: 10/07/2023] Open
Abstract
Chronic pancreatitis (CP) is a progressive and irreversible fibroinflammatory disorder, accompanied by pancreatic exocrine insufficiency and dysregulated gut microbiota. Recently, accumulating evidence has supported a correlation between gut dysbiosis and CP development. However, whether gut microbiota dysbiosis contributes to CP pathogenesis remains unclear. Herein, an experimental CP was induced by repeated high-dose caerulein injections. The broad-spectrum antibiotics (ABX) and ABX targeting Gram-positive (G+) or Gram-negative bacteria (G-) were applied to explore the specific roles of these bacteria. Gut dysbiosis was observed in both mice and in CP patients, which was accompanied by a sharply reduced abundance for short-chain fatty acids (SCFAs)-producers, especially G+ bacteria. Broad-spectrum ABX exacerbated the severity of CP, as evidenced by aggravated pancreatic fibrosis and gut dysbiosis, especially the depletion of SCFAs-producing G+ bacteria. Additionally, depletion of SCFAs-producing G+ bacteria rather than G- bacteria intensified CP progression independent of TLR4, which was attenuated by supplementation with exogenous SCFAs. Finally, SCFAs modulated pancreatic fibrosis through inhibition of macrophage infiltration and M2 phenotype switching. The study supports a critical role for SCFAs-producing G+ bacteria in CP. Therefore, modulation of dietary-derived SCFAs or G+ SCFAs-producing bacteria may be considered a novel interventive approach for the management of CP.
Collapse
Affiliation(s)
- Li-Long Pan
- Wuxi Medical School, Jiangnan University, Wuxi 214122, China
| | - Zheng-Nan Ren
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
| | - Jun Yang
- Department of General Surgery, Affiliated Hospital of Jiangnan University, Wuxi 214122, China
| | - Bin-Bin Li
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
| | - Yi-Wen Huang
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
| | - Dong-Xiao Song
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
| | - Xuan Li
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
| | - Jia-Jia Xu
- Department of General Medicine, Beicai Community Health Service Center of Pudong New District, Shanghai 214001, China
| | - Madhav Bhatia
- Department of Pathology, University of Otago, Christchurch 8140, New Zealand
| | - Duo-Wu Zou
- Department of Gastroenterology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Chun-Hua Zhou
- Department of Gastroenterology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jia Sun
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
22
|
Wang Z, Guo M, Li J, Jiang C, Yang S, Zheng S, Li M, Ai X, Xu X, Zhang W, He X, Wang Y, Chen Y. Composition and functional profiles of gut microbiota reflect the treatment stage, severity, and etiology of acute pancreatitis. Microbiol Spectr 2023; 11:e0082923. [PMID: 37698429 PMCID: PMC10580821 DOI: 10.1128/spectrum.00829-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 07/13/2023] [Indexed: 09/13/2023] Open
Abstract
Acute pancreatitis (AP) is a type of digestive system disease with high mortality. Previous studies have shown that gut microbiota can participate in developing and treating acute pancreatitis by affecting the host's metabolism. In this study, we followed 20 AP patients to generate longitudinal gut microbiota profiles and activity during disease (before treatment, on the third day of treatment, and 1 month after discharge). We analyzed species composition and metabolic pathways' changes across the treatment phase, severity, and etiology. The diversity of the gut microbiome of patients with AP did not show much variation with treatment. In contrast, the metabolic functions of the gut microbiota, such as the essential chemical reactions that produce energy and maintain life, were partially reinstated after treatment. The severe AP (SAP) patients contained less beneficial bacteria (i.e., Bacteroides xylanisolvens, Clostridium lavalense, and Roseburia inulinivorans) and weaker sugar degradation function than mild AP patients before treatment. Moreover, etiology was one of the drivers of gut microbiome composition and explained the 3.54% variation in species' relative abundance. The relative abundance of pathways related to lipid synthesis was higher in the gut of hyperlipidemia AP patients than in biliary AP patients. The composition and functional profiles of the gut microbiota reflect the severity and etiology of AP. Otherwise, we also identified bacterial species associated with SAP, i.e., Oscillibacter sp. 57_20, Parabacteroides johnsonii, Bacteroides stercoris, Methanobrevibacter smithii, Ruminococcus lactaris, Coprococcus comes, and Dorea formicigenerans, which have the potential to identify the SAP at an early stage. IMPORTANCE Acute pancreatitis (AP) is a type of digestive system disease with high mortality. Previous studies have shown that gut microbiota can participate in the development and treatment of acute pancreatitis by affecting the host's metabolism. However, fewer studies acquired metagenomic sequencing data to associate species to functions intuitively and performed longitudinal analysis to explore how gut microbiota influences the development of AP. We followed 20 AP patients to generate longitudinal gut microbiota profiles and activity during disease and studied the differences in intestinal flora under different severities and etiologies. We have two findings. First, the gut microbiota profile has the potential to identify the severity and etiology of AP at an early stage. Second, gut microbiota likely acts synergistically in the development of AP. This study provides a reference for characterizing the driver flora of severe AP to identify the severity of acute pancreatitis at an early stage.
Collapse
Affiliation(s)
- Zhenjiang Wang
- Department of Gastroenterology, Zhuhai Hospital Affiliated with Jinan University (Zhuhai People’s Hospital), Zhuhai, China
| | - Mingyi Guo
- Department of Gastroenterology, Zhuhai Hospital Affiliated with Jinan University (Zhuhai People’s Hospital), Zhuhai, China
| | - Jing Li
- School of Management, University of Science and Technology of China, Hefei, Anhui, China
- Department of Research and Development, Shenzhen Byoryn Technology Co., Ltd., Shenzhen, China
| | - Chuangming Jiang
- Department of Gastroenterology, Gaolangang Branch of Zhuhai People’s Hospital (Hospital of Gaolangang), Zhuhai, China
| | - Sen Yang
- Department of Gastroenterology, Zhuhai Hospital Affiliated with Jinan University (Zhuhai People’s Hospital), Zhuhai, China
| | - Shizhuo Zheng
- Department of Gastroenterology, Zhuhai Hospital Affiliated with Jinan University (Zhuhai People’s Hospital), Zhuhai, China
| | - Mingzhe Li
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, China
| | - Xinbo Ai
- Department of Gastroenterology, Zhuhai Hospital Affiliated with Jinan University (Zhuhai People’s Hospital), Zhuhai, China
| | - Xiaohong Xu
- Department of Gastroenterology, Zhuhai Hospital Affiliated with Jinan University (Zhuhai People’s Hospital), Zhuhai, China
| | - Wenbo Zhang
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, China
| | - Xingxiang He
- Department of Gastroenterology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yinan Wang
- Department of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yuping Chen
- Department of Gastroenterology, Zhuhai Hospital Affiliated with Jinan University (Zhuhai People’s Hospital), Zhuhai, China
| |
Collapse
|
23
|
Zhao Y, Chang X, Gu X, Li Y, Zheng Y, Fang H. Predictive Analysis of Quality Markers of Atractylodis Rhizoma Based on Fingerprint and Network Pharmacology. J AOAC Int 2023; 106:1402-1413. [PMID: 37208180 DOI: 10.1093/jaoacint/qsad059] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 05/03/2023] [Accepted: 05/09/2023] [Indexed: 05/21/2023]
Abstract
BACKGROUND Atractylodes chinensis (DC.) Koidz. (A. chinensis) is a perennial herbaceous plant that is widely used as a Chinese medicine herb for gastric diseases. However, the bioactive compounds of this herbal medicine have not been defined, and quality control is imperfect. OBJECTIVE Although the method of quality evaluation method for A. chinensis by high-performance liquid chromatography (HPLC) fingerprinting has been reported in related papers, it remains unknown whether the chemical markers selected are representative of their clinical efficacy. To develop methods for qualitative analysis and improved quality evaluation of A. chinensis. METHOD In this study, HPLC was used to establish fingerprints and conduct similarity evaluation. Principal component analysis (PCA) and orthogonal partial least squares discriminant analysis (OPLS-DA) were used to reveal the differences of these fingerprints. Network pharmacology was used to analyze the corresponding targets of the active ingredients. Meantime, an active ingredient-target-pathway network was constructed to investigate the characteristics of the medical efficacy of A. chinensis and to predict potential Q-markers. RESULTS Combining network pharmacological effectiveness and composition specificity with the Q-marker concept, atractylodin (ATD), β-eudesmol, atractylenolide Ι (AT-I) and atractylenolide III (AT-III) were predicted to be potential Q-markers of A. chinensis that showed anti-inflammatory, antidepressant, anti-gastric, and antiviral effects by acting on 10 core targets and 20 key pathways. CONCLUSIONS The HPLC fingerprinting method established in this study is straightforward, and the identified four active constituents can be used as Q-markers of A. chinensis. These findings facilitate effective quality evaluation of A. chinensis and suggest this approach could be applied to evaluate the quality of other herbal medicines. HIGHLIGHTS The fingerprints of Atractylodis rhizoma were organically combined with network pharmacology to further clarify its criteria for quality control.
Collapse
Affiliation(s)
- Yanyun Zhao
- Hebei University of Chinese Medicine, Traditional Chinese Medicine Processing Technology Innovation Center of Hebei Province; International Joint Research Center on Resource Utilization and Quality Evaluation of Traditional Chinese Medicine of Hebei Province, No. 326 Xinshi South Road, Qiaoxi District, Shijiazhuang, Hebei Province 050091, P.R. China
- Inner Mongolia University, Key Laboratory of Forage and Endemic Crop Biotechnology, Ministry of Education, College of Life Sciences, No.235 West College Road, Saihan District, Hohhot Inner Mongolia 010000, P.R. China
| | - Xinxin Chang
- Hebei University of Chinese Medicine, Traditional Chinese Medicine Processing Technology Innovation Center of Hebei Province; International Joint Research Center on Resource Utilization and Quality Evaluation of Traditional Chinese Medicine of Hebei Province, No. 326 Xinshi South Road, Qiaoxi District, Shijiazhuang, Hebei Province 050091, P.R. China
| | - Xian Gu
- Hebei University of Chinese Medicine, Traditional Chinese Medicine Processing Technology Innovation Center of Hebei Province; International Joint Research Center on Resource Utilization and Quality Evaluation of Traditional Chinese Medicine of Hebei Province, No. 326 Xinshi South Road, Qiaoxi District, Shijiazhuang, Hebei Province 050091, P.R. China
| | - Yang Li
- Hebei University of Chinese Medicine, Traditional Chinese Medicine Processing Technology Innovation Center of Hebei Province; International Joint Research Center on Resource Utilization and Quality Evaluation of Traditional Chinese Medicine of Hebei Province, No. 326 Xinshi South Road, Qiaoxi District, Shijiazhuang, Hebei Province 050091, P.R. China
| | - Yuguang Zheng
- Hebei University of Chinese Medicine, Traditional Chinese Medicine Processing Technology Innovation Center of Hebei Province; International Joint Research Center on Resource Utilization and Quality Evaluation of Traditional Chinese Medicine of Hebei Province, No. 326 Xinshi South Road, Qiaoxi District, Shijiazhuang, Hebei Province 050091, P.R. China
- Hebei Chemical and Pharmaceutical College, No. 88 Fangxing Road, Yuhua District, Shijiazhuang, Hebei Province 050026, P.R. China
| | - Huiyong Fang
- Hebei University of Chinese Medicine, Traditional Chinese Medicine Processing Technology Innovation Center of Hebei Province; International Joint Research Center on Resource Utilization and Quality Evaluation of Traditional Chinese Medicine of Hebei Province, No. 326 Xinshi South Road, Qiaoxi District, Shijiazhuang, Hebei Province 050091, P.R. China
| |
Collapse
|
24
|
Mendes I, Vale N. How Can the Microbiome Induce Carcinogenesis and Modulate Drug Resistance in Cancer Therapy? Int J Mol Sci 2023; 24:11855. [PMID: 37511612 PMCID: PMC10380870 DOI: 10.3390/ijms241411855] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 07/18/2023] [Accepted: 07/21/2023] [Indexed: 07/30/2023] Open
Abstract
Over the years, cancer has been affecting the lives of many people globally and it has become one of the most studied diseases. Despite the efforts to understand the cell mechanisms behind this complex disease, not every patient seems to respond to targeted therapies or immunotherapies. Drug resistance in cancer is one of the limiting factors contributing to unsuccessful therapies; therefore, understanding how cancer cells acquire this resistance is essential to help cure individuals affected by cancer. Recently, the altered microbiome was observed to be an important hallmark of cancer and therefore it represents a promising topic of cancer research. Our review aims to provide a global perspective of some cancer hallmarks, for instance how genetic and epigenetic modifications may be caused by an altered human microbiome. We also provide information on how an altered human microbiome can lead to cancer development as well as how the microbiome can influence drug resistance and ultimately targeted therapies. This may be useful to develop alternatives for cancer treatment, i.e., future personalized medicine that can help in cases where traditional cancer treatment is unsuccessful.
Collapse
Affiliation(s)
- Inês Mendes
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal;
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- School of Life and Environmental Sciences, University of Trás-os-Montes and Alto Douro (UTAD), Edifício de Geociências, 5000-801 Vila Real, Portugal
| | - Nuno Vale
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal;
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Community Medicine, Information and Health Decision Sciences (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
| |
Collapse
|
25
|
De Lucia SS, Candelli M, Polito G, Maresca R, Mezza T, Schepis T, Pellegrino A, Zileri Dal Verme L, Nicoletti A, Franceschi F, Gasbarrini A, Nista EC. Nutrition in Acute Pancreatitis: From the Old Paradigm to the New Evidence. Nutrients 2023; 15:1939. [PMID: 37111158 PMCID: PMC10144915 DOI: 10.3390/nu15081939] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 04/12/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
The nutritional management of acute pancreatitis (AP) patients has widely changed over time. The "pancreatic rest" was the cornerstone of the old paradigm, and nutritional support was not even included in AP management. Traditional management of AP was based on intestinal rest, with or without complete parenteral feeding. Recently, evidence-based data underlined the superiority of early oral or enteral feeding with significantly decreased multiple-organ failure, systemic infections, surgery need, and mortality rate. Despite the current recommendations, experts still debate the best route for enteral nutritional support and the best enteral formula. The aim of this work is to collect and analyze evidence over the nutritional aspects of AP management to investigate its impact. Moreover, the role of immunonutrition and probiotics in modulating inflammatory response and gut dysbiosis during AP was extensively studied. However, we have no significant data for their use in clinical practice. This is the first work to move beyond the mere opposition between the old and the new paradigm, including an analysis of several topics still under debate in order to provide a comprehensive overview of nutritional management of AP.
Collapse
Affiliation(s)
- Sara Sofia De Lucia
- Department of Medical and Surgical Sciences, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCSS, 00168 Rome, Italy
| | - Marcello Candelli
- Department of Emergency, Anesthesiological and Reanimation Sciences, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCSS, 00168 Rome, Italy
| | - Giorgia Polito
- Department of Medical and Surgical Sciences, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCSS, 00168 Rome, Italy
| | - Rossella Maresca
- Department of Medical and Surgical Sciences, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCSS, 00168 Rome, Italy
| | - Teresa Mezza
- Department of Medical and Surgical Sciences, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCSS, 00168 Rome, Italy
| | - Tommaso Schepis
- Department of Medical and Surgical Sciences, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCSS, 00168 Rome, Italy
| | - Antonio Pellegrino
- Department of Medical and Surgical Sciences, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCSS, 00168 Rome, Italy
| | - Lorenzo Zileri Dal Verme
- Department of Medical and Surgical Sciences, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCSS, 00168 Rome, Italy
| | - Alberto Nicoletti
- Department of Medical and Surgical Sciences, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCSS, 00168 Rome, Italy
| | - Francesco Franceschi
- Department of Emergency, Anesthesiological and Reanimation Sciences, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCSS, 00168 Rome, Italy
| | - Antonio Gasbarrini
- Department of Medical and Surgical Sciences, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCSS, 00168 Rome, Italy
| | - Enrico Celestino Nista
- Department of Medical and Surgical Sciences, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCSS, 00168 Rome, Italy
| |
Collapse
|
26
|
Zhong H, Liu S, Zhu J, Wu L. Associations between genetically predicted levels of blood metabolites and pancreatic cancer risk. Int J Cancer 2023; 153:103-110. [PMID: 36757187 DOI: 10.1002/ijc.34466] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 01/14/2023] [Accepted: 01/30/2023] [Indexed: 02/10/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive solid malignancies, which is featured by systematic metabolism. Thus, a better understanding of metabolic dysregulation in PDAC is important to better characterize its etiology. Here, we performed a large metabolome-wide association study (MWAS) to systematically explore associations between genetically predicted metabolite levels in blood and PDAC risk. Using data from 881 subjects of European descent in the TwinsUK Project, comprehensive genetic models were built to predict serum metabolite levels. These prediction models were applied to the genetic data of 8275 cases and 6723 controls included in the PanScan (I, II and III) and PanC4 consortia. After assessing the metabolite-PDAC risk associations by a slightly modified TWAS/FUSION framework, we identified five metabolites (including two dipeptides) showing significant associations with PDAC risk at false discovery rate (FDR) <0.05. Integrated with gut microbial information, two-sample Mendelian randomization (MR) analyses were further performed to investigate the relationship among serum metabolites, gut microbiome features and PDAC. The flavonoid-degrading bacteria Flavonifractor sp90199495 was found to be associated with metabolite X-21849 and it was also shown to be associated with PDAC risk. Collectively, our study identified novel candidate metabolites for PDAC risk, which could lead to new insights into the etiology of PDAC and improved treatment options.
Collapse
Affiliation(s)
- Hua Zhong
- Cancer Epidemiology Division, Population Sciences in the Pacific Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - Shuai Liu
- Cancer Epidemiology Division, Population Sciences in the Pacific Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - Jingjing Zhu
- Cancer Epidemiology Division, Population Sciences in the Pacific Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - Lang Wu
- Cancer Epidemiology Division, Population Sciences in the Pacific Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| |
Collapse
|
27
|
Xue C, Chu Q, Zheng Q, Yuan X, Su Y, Bao Z, Lu J, Li L. Current understanding of the intratumoral microbiome in various tumors. Cell Rep Med 2023; 4:100884. [PMID: 36652905 PMCID: PMC9873978 DOI: 10.1016/j.xcrm.2022.100884] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 09/18/2022] [Accepted: 12/13/2022] [Indexed: 01/19/2023]
Abstract
It is estimated that in the future, the number of new cancer cases worldwide will exceed the 19.3 million recorded in 2020, and the number of deaths will exceed 10 million. Cancer remains the leading cause of human mortality and lagging socioeconomic development. Intratumoral microbes have been revealed to exist in many cancer types, including pancreatic, colorectal, liver, esophageal, breast, and lung cancers. Intratumoral microorganisms affect not only the host immune system, but also the effectiveness of tumor chemotherapy. This review concentrates on the characteristics and roles of intratumoral microbes in various tumors. In addition, the potential of therapies targeting intratumoral microbes, as well as the main challenges currently delaying these therapies, are explored. Furthermore, we briefly summarize existing technical methods used to characterize intratumoral microbes. We hope to provide ideas for exploring intratumoral microbes as potential biomarkers and targets for tumor diagnosis, treatment, and prognostication.
Collapse
Affiliation(s)
- Chen Xue
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Qingfei Chu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Qiuxian Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Xin Yuan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yuanshuai Su
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Zhengyi Bao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Juan Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| |
Collapse
|
28
|
Hu X, Han Z, Zhou R, Su W, Gong L, Yang Z, Song X, Zhang S, Shu H, Wu D. Altered gut microbiota in the early stage of acute pancreatitis were related to the occurrence of acute respiratory distress syndrome. Front Cell Infect Microbiol 2023; 13:1127369. [PMID: 36949815 PMCID: PMC10025409 DOI: 10.3389/fcimb.2023.1127369] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 02/20/2023] [Indexed: 03/08/2023] Open
Abstract
Background Acute respiratory distress syndrome (ARDS) is the most common cause of organ failure in acute pancreatitis (AP) patients, which associated with high mortality. Specific changes in the gut microbiota have been shown to influence progression of acute pancreatitis. We aimed to determine whether early alterations in the gut microbiota is related to and could predict ARDS occurrence in AP patients. Methods In this study, we performed 16S rRNA sequencing analysis in 65 AP patients and 20 healthy volunteers. The AP patients were further divided into two groups: 26 AP-ARDS patients and 39 AP-nonARDS patients based on ARDS occurrence during hospitalization. Results Our results showed that the AP-ARDS patients exhibited specific changes in gut microbiota composition and function as compared to subjects of AP-nonARDS group. Higher abundances of Proteobacteria phylum, Enterobacteriaceae family, Escherichia-Shigella genus, and Klebsiella pneumoniae, but lower abundances of Bifidobacterium genus were found in AP-ARDS group compared with AP-nonARDS groups. Random forest modelling analysis revealed that the Escherichia-shigella genus was effective to distinguish AP-ARDS from AP-nonARDS, which could predict ARDS occurrence in AP patients. Conclusions Our study revealed that alterations of gut microbiota in AP patients on admission were associated with ARDS occurrence after hospitalization, indicating a potential predictive and pathogenic role of gut microbiota in the development of ARDS in AP patients.
Collapse
Affiliation(s)
- Xiaomin Hu
- Department of Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Gastroenterology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ziying Han
- Department of Gastroenterology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ruilin Zhou
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Wan Su
- Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Liang Gong
- Department of Gastroenterology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zihan Yang
- Department of Gastroenterology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiao Song
- Department of Emergency Medicine, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Shuyang Zhang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Huijun Shu
- Department of Gastroenterology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Huijun Shu, ; Dong Wu,
| | - Dong Wu
- Department of Gastroenterology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Huijun Shu, ; Dong Wu,
| |
Collapse
|
29
|
Binda C, Gibiino G, Sbrancia M, Coluccio C, Cazzato M, Carloni L, Cucchetti A, Ercolani G, Sambri V, Fabbri C. Microbiota in the Natural History of Pancreatic Cancer: From Predisposition to Therapy. Cancers (Basel) 2022; 15:cancers15010001. [PMID: 36611999 PMCID: PMC9817971 DOI: 10.3390/cancers15010001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/28/2022] [Accepted: 12/13/2022] [Indexed: 12/29/2022] Open
Abstract
Early microbiome insights came from gut microbes and their role among intestinal and extraintestinal disease. The latest evidence suggests that the microbiota is a true organ, capable of several interactions throughout the digestive system, attracting specific interest in the biliopancreatic district. Despite advances in diagnostics over the last few decades and improvements in the management of this disease, pancreatic cancer is still a common cause of cancer death. Microbiota can influence the development of precancerous disease predisposing to pancreatic cancer (PC). At the same time, neoplastic tissue shows specific characteristics in terms of diversity and phenotype, determining the short- and long-term prognosis. Considering the above information, a role for microbiota has also been hypothesized in the different phases of the PC approach, providing future revolutionary therapeutic insights. Microbiota-modulating therapies could open new issues in the therapeutic landscape. The aim of this narrative review is to assess the most updated evidence on microbiome in all the steps regarding pancreatic adenocarcinoma, from early development to response to antineoplastic therapy and long-term prognosis.
Collapse
Affiliation(s)
- Cecilia Binda
- Gastroenterology and Digestive Endoscopy Unit, Forlì-Cesena Hospitals, Ausl Romagna, 47121 Forlì-Cesena, Italy
| | - Giulia Gibiino
- Gastroenterology and Digestive Endoscopy Unit, Forlì-Cesena Hospitals, Ausl Romagna, 47121 Forlì-Cesena, Italy
- Correspondence: ; Tel.: +39-3488609557
| | - Monica Sbrancia
- Gastroenterology and Digestive Endoscopy Unit, Forlì-Cesena Hospitals, Ausl Romagna, 47121 Forlì-Cesena, Italy
| | - Chiara Coluccio
- Gastroenterology and Digestive Endoscopy Unit, Forlì-Cesena Hospitals, Ausl Romagna, 47121 Forlì-Cesena, Italy
| | - Maria Cazzato
- Gastroenterology and Digestive Endoscopy Unit, Forlì-Cesena Hospitals, Ausl Romagna, 47121 Forlì-Cesena, Italy
| | - Lorenzo Carloni
- Gastroenterology and Digestive Endoscopy Unit, Forlì-Cesena Hospitals, Ausl Romagna, 47121 Forlì-Cesena, Italy
- Department of Medical and Surgical Sciences—DIMEC, Alma Mater Studiorum, University of Bologna, 40138 Bologna, Italy
| | - Alessandro Cucchetti
- Department of Medical and Surgical Sciences—DIMEC, Alma Mater Studiorum, University of Bologna, 40138 Bologna, Italy
- General and Oncologic Surgery, Morgagni—Pierantoni Hospital, AUSL Romagna, 47121 Forlì, Italy
| | - Giorgio Ercolani
- Department of Medical and Surgical Sciences—DIMEC, Alma Mater Studiorum, University of Bologna, 40138 Bologna, Italy
- General and Oncologic Surgery, Morgagni—Pierantoni Hospital, AUSL Romagna, 47121 Forlì, Italy
| | - Vittorio Sambri
- Department of Medical and Surgical Sciences—DIMEC, Alma Mater Studiorum, University of Bologna, 40138 Bologna, Italy
- Microbiology Unit, Hub Laboratory, AUSL della Romagna, 47121 Cesena, Italy
| | - Carlo Fabbri
- Gastroenterology and Digestive Endoscopy Unit, Forlì-Cesena Hospitals, Ausl Romagna, 47121 Forlì-Cesena, Italy
| |
Collapse
|
30
|
Ramos Meyers G, Samouda H, Bohn T. Short Chain Fatty Acid Metabolism in Relation to Gut Microbiota and Genetic Variability. Nutrients 2022; 14:5361. [PMID: 36558520 PMCID: PMC9788597 DOI: 10.3390/nu14245361] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 12/12/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
It is widely accepted that the gut microbiota plays a significant role in modulating inflammatory and immune responses of their host. In recent years, the host-microbiota interface has gained relevance in understanding the development of many non-communicable chronic conditions, including cardiovascular disease, cancer, autoimmunity and neurodegeneration. Importantly, dietary fibre (DF) and associated compounds digested by the microbiota and their resulting metabolites, especially short-chain fatty acids (SCFA), were significantly associated with health beneficial effects, such as via proposed anti-inflammatory mechanisms. However, SCFA metabolic pathways are not fully understood. Major steps include production of SCFA by microbiota, uptake in the colonic epithelium, first-pass effects at the liver, followed by biodistribution and metabolism at the host's cellular level. As dietary patterns do not affect all individuals equally, the host genetic makeup may play a role in the metabolic fate of these metabolites, in addition to other factors that might influence the microbiota, such as age, birth through caesarean, medication intake, alcohol and tobacco consumption, pathogen exposure and physical activity. In this article, we review the metabolic pathways of DF, from intake to the intracellular metabolism of fibre-derived products, and identify possible sources of inter-individual variability related to genetic variation. Such variability may be indicative of the phenotypic flexibility in response to diet, and may be predictive of long-term adaptations to dietary factors, including maladaptation and tissue damage, which may develop into disease in individuals with specific predispositions, thus allowing for a better prediction of potential health effects following personalized intervention with DF.
Collapse
Affiliation(s)
- Guilherme Ramos Meyers
- Nutrition and Health Research Group, Department of Precision Health, Luxembourg Institute of Health, 1 A-B, Rue Thomas Edison, 1445 Strassen, Luxembourg
- Doctoral School in Science and Engineering, University of Luxembourg, 2, Avenue de l'Université, 4365 Esch-sur-Alzette, Luxembourg
| | - Hanen Samouda
- Nutrition and Health Research Group, Department of Precision Health, Luxembourg Institute of Health, 1 A-B, Rue Thomas Edison, 1445 Strassen, Luxembourg
| | - Torsten Bohn
- Nutrition and Health Research Group, Department of Precision Health, Luxembourg Institute of Health, 1 A-B, Rue Thomas Edison, 1445 Strassen, Luxembourg
| |
Collapse
|
31
|
Devoy C, Flores Bueso Y, Tangney M. Understanding and harnessing triple-negative breast cancer-related microbiota in oncology. Front Oncol 2022; 12:1020121. [PMID: 36505861 PMCID: PMC9730816 DOI: 10.3389/fonc.2022.1020121] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 10/31/2022] [Indexed: 11/27/2022] Open
Abstract
Bacterial inhabitants of the body have the potential to play a role in various stages of cancer initiation, progression, and treatment. These bacteria may be distal to the primary tumour, such as gut microbiota, or local to the tissue, before or after tumour growth. Breast cancer is well studied in this context. Amongst breast cancer types, Triple Negative Breast Cancer (TNBC) is more aggressive, has fewer treatment options than receptor-positive breast cancers, has an overall worse prognosis and higher rates of reoccurrence. Thus, an in-depth understanding of the bacterial influence on TNBC progression and treatment is of high value. In this regard, the Gut Microbiota (GM) can be involved in various stages of tumour progression. It may suppress or promote carcinogenesis through the release of carcinogenic metabolites, sustenance of proinflammatory environments and/or the promotion of epigenetic changes in our genome. It can also mediate metastasis and reoccurrence through interactions with the immune system and has been recently shown to influence chemo-, radio-, and immune-therapies. Furthermore, bacteria have also been found to reside in normal and malignant breast tissue. Several studies have now described the breast and breast tumour microbiome, with the tumour microbiota of TNBC having the least taxonomic diversity among all breast cancer types. Here, specific conditions of the tumour microenvironment (TME) - low O2, leaky vasculature and immune suppression - are supportive of tumour selective bacterial growth. This innate bacterial ability could enable their use as delivery agents for various therapeutics or as diagnostics. This review aims to examine the current knowledge on bacterial relevance to TNBC and potential uses while examining some of the remaining unanswered questions regarding mechanisms underpinning observed effects.
Collapse
Affiliation(s)
- Ciaran Devoy
- Cancer Research@UCC, College of Medicine and Health, University College Cork, Cork, Ireland,SynBio Center, University College Cork, Cork, Ireland,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Yensi Flores Bueso
- Cancer Research@UCC, College of Medicine and Health, University College Cork, Cork, Ireland,SynBio Center, University College Cork, Cork, Ireland,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Mark Tangney
- Cancer Research@UCC, College of Medicine and Health, University College Cork, Cork, Ireland,SynBio Center, University College Cork, Cork, Ireland,APC Microbiome Ireland, University College Cork, Cork, Ireland,School of Pharmacy, College of Medicine and Health, University College Cork, Cork, Ireland,*Correspondence: Mark Tangney,
| |
Collapse
|
32
|
Maev IV, Levchenko AI, Andreev DN. Changes in the Intestinal Microbiota in Patients with Chronic Pancreatitis: Systematizing Literature Data. RUSSIAN JOURNAL OF GASTROENTEROLOGY, HEPATOLOGY, COLOPROCTOLOGY 2022; 32:17-26. [DOI: 10.22416/1382-4376-2022-32-4-17-26] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2024]
Abstract
The purpose of the review. To systematize literature data on changes in the structure of the intestinal microbiota in patients with chronic pancreatitis (CP).Key findings. The human intestinal microbiota is a dynamically changing system that is constantly undergoing qualitative and quantitative changes, especially in several pathological conditions of the digestive system. At present, the differences in the intestinal microbiota in pancreatic diseases are poorly understood. The severe CP is associated with impaired synthesis of antimicrobial peptides, bicarbonates, and digestive enzymes by the pancreas, which is a risk factor for dysbiotic changes in the intestinal microbiota, consisting in the development of small intestinal bacterial overgrowth (SIBO) and gut dysbiosis. The results of two large meta-analyses show that about a third of CP patients have SIBO. The colonic microbiota in patients with CP is also characterized by dysbiotic disorders, primarily in the reduction of alpha-diversity. Some studies have shown that these patients have an increase in Firmicutes, while Bacteroides and Faecalibacterium are reduced. In addition, as a rule, in patients with CP, the growth of Escherichia, Shigella and Streptococcus is recorded.Conclusion. In general, scientific papers have revealed significant heterogeneity in the profiles of the intestinal microbiota in patients with CP. Thus, several questions remain open, prioritizing the further study of the intestinal microbiota in patients with CP for identifying the specifics of its structure that can personalize the selection of enzyme replacement therapy and restrict the unreasonable prescription of additional pharmacotherapy (the use of proton pump inhibitors and / or antibacterial drugs).
Collapse
Affiliation(s)
- I. V. Maev
- A.I. Yevdokimov Moscow State University of Medicine and Dentistry
| | - A. I. Levchenko
- A.I. Yevdokimov Moscow State University of Medicine and Dentistry
| | - D. N. Andreev
- A.I. Yevdokimov Moscow State University of Medicine and Dentistry
| |
Collapse
|
33
|
Emerging Trends in Intestinal Knowledge Structure Associated With Acute Pancreatitis From 1981 to 2021: A Bibliometric Analysis. Pancreas 2022; 51:957-965. [PMID: 36607940 DOI: 10.1097/mpa.0000000000002140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
OBJECTIVES Bibliometric analysis has not comprehensively summarized studies of acute pancreatitis (AP)-associated intestinal diseases. This work aimed to evaluate cooperative networks of authors, countries, and institutions and explore the field's developing trends and hot topics. METHODS Original articles and reviews of AP-associated intestinal diseases were obtained from the Web of Science Core Collection on October 11, 2021. VOSviewer and CiteSpace software were used to perform co-occurrence analyses for authors, countries, and institutions and detect the highest citation burst. RESULTS A total of 1634 articles on the intestine associated with AP were identified. The United States, the University of Auckland, and Roland Andersson are the most influential country, research institute, and scholar, respectively. The World Journal of Gastroenterology (73 articles) has the highest number of publications, and Gastroenterology was the most co-cited journal. The top 5 key words are "acute pancreatitis," "bacterial translocation," "management," "gut," and "inflammatory bowel disease." We find that several emergent key words like "gut microbiota," "pathway," "gut barrier," "risk," and "oxidative stress" experienced a continuous and rapid development as new research directions. CONCLUSIONS This bibliometric study summarizes current important perspectives and offers comprehensive guidance on the AP-associated intestinal diseases, which may help researchers choose the most appropriate research directions.
Collapse
|
34
|
Bartolini I, Nannini G, Risaliti M, Matarazzo F, Moraldi L, Ringressi MN, Taddei A, Amedei A. Impact of microbiota-immunity axis in pancreatic cancer management. World J Gastroenterol 2022; 28:4527-4539. [PMID: 36157926 PMCID: PMC9476869 DOI: 10.3748/wjg.v28.i32.4527] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 06/28/2022] [Accepted: 07/27/2022] [Indexed: 02/06/2023] Open
Abstract
The microbiota impact on human diseases is well-known, and a growing body of literature is providing evidence about the complex interplay between microbiota-immune system-human physiology/pathology, including cancers. Together with the defined risk factors (e.g., smoke habits, diet, diabetes, and obesity), the oral, gut, biliary, and intrapancreatic microbiota contribute to pancreatic cancer development through different pathways including the interaction with the immune system. Unfortunately, a great majority of the pancreatic cancer patients received a diagnosis in advanced stages not amenable to be radically treated and potentially cured. Given the poor pancreatic cancer prognosis, complete knowledge of these complicated relationships could help researchers better understand the disease pathogenesis and thus provide early potential non-invasive biomarkers, new therapeutic targets, and tools for risk stratification that might result in greater therapeutic possibilities and eventually in a better and longer patient survival.
Collapse
Affiliation(s)
- Ilenia Bartolini
- Department of Experimental and Clinical Medicine, HPB Surgery Unit, Azienda Ospedaliero-Universitaria Careggi, Florence 50134, Italy
| | - Giulia Nannini
- Department of Experimental and Clinical Medicine, SOD of Interdisciplinary Internal Medicine, Azienda Ospedaliera-Universitaria Careggi, Florence 50134, Italy
| | - Matteo Risaliti
- Department of Experimental and Clinical Medicine, HPB Surgery Unit, Azienda Ospedaliero-Universitaria Careggi, Florence 50134, Italy
| | - Francesco Matarazzo
- Department of Experimental and Clinical Medicine, HPB Surgery Unit, Azienda Ospedaliero-Universitaria Careggi, Florence 50134, Italy
| | - Luca Moraldi
- Division of Oncologic Surgery, Department of Oncology, Careggi University Hospital, Firenze 50134, Italy
| | - Maria Novella Ringressi
- Department of Experimental and Clinical Medicine, HPB Surgery Unit, Azienda Ospedaliero-Universitaria Careggi, Florence 50134, Italy
| | - Antonio Taddei
- Department of Experimental and Clinical Medicine, HPB Surgery Unit, Azienda Ospedaliero-Universitaria Careggi, Florence 50134, Italy
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, SOD of Interdisciplinary Internal Medicine, Azienda Ospedaliera-Universitaria Careggi, Florence 50134, Italy
| |
Collapse
|
35
|
Dunn KA, MacDonald T, Rodrigues GJ, Forbrigger Z, Bielawski JP, Langille MG, Van Limbergen J, Kulkarni K. Antibiotic and antifungal use in pediatric leukemia and lymphoma patients are associated with increasing opportunistic pathogens and decreasing bacteria responsible for activities that enhance colonic defense. Front Cell Infect Microbiol 2022; 12:924707. [PMID: 35967843 PMCID: PMC9363618 DOI: 10.3389/fcimb.2022.924707] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 07/06/2022] [Indexed: 11/13/2022] Open
Abstract
Due to decreased immunity, both antibiotics and antifungals are regularly used in pediatric hematologic-cancer patients as a means to prevent severe infections and febrile neutropenia. The general effect of antibiotics on the human gut microbiome is profound, yielding decreased diversity and changes in community structure. However, the specific effect on pediatric oncology patients is not well-studied. The effect of antifungal use is even less understood, having been studied only in mouse models. Because the composition of the gut microbiome is associated with regulation of hematopoiesis, immune function and gastrointestinal integrity, changes within the patient gut can have implications for the clinical management of hematologic malignancies. The pediatric population is particularly challenging because the composition of the microbiome is age dependent, with some of the most pronounced changes occurring in the first three years of life. We investigated how antibiotic and antifungal use shapes the taxonomic composition of the stool microbiome in pediatric patients with leukemia and lymphoma, as inferred from both 16S rRNA and metagenome data. Associations with age, antibiotic use and antifungal use were investigated using multiple analysis methods. In addition, multivariable differential abundance was used to identify and assess specific taxa that were associated with multiple variables. Both antibiotics and antifungals were linked to a general decline in diversity in stool samples, which included a decrease in relative abundance in butyrate producers that play a critical role in host gut physiology (e.g., Faecalibacterium, Anaerostipes, Dorea, Blautia),. Furthermore, antifungal use was associated with a significant increase in relative abundance of opportunistic pathogens. Collectively, these findings have important implications for the treatment of leukemia and lymphoma patients. Butyrate is important for gastrointestinal integrity; it inhibits inflammation, reinforces colonic defense, mucosal immunity. and decreases oxidative stress. The routine use of broad-spectrum anti-infectives in pediatric oncology patients could simultaneously contribute to a decline in gastrointestinal integrity and colonic defense while promoting increases in opportunistic pathogens within the patient gut. Because the gut microbiome has been linked to both short-term clinical outcomes, and longer-lasting health effects, systematic characterization of the gut microbiome in pediatric patients during, and beyond, treatment is warranted.
Collapse
Affiliation(s)
- Katherine A. Dunn
- Department of Pediatrics, Division of Hematology and Oncology, Izaak Walton Killam (IWK) Health, Halifax, NS, Canada
- Department of Biology, Dalhousie University, Halifax, NS, Canada
- Institute for Comparative Genomics, Dalhousie University, Halifax, NS, Canada
| | - Tamara MacDonald
- Department of Pharmacy, IWK Health, Halifax, NS, Canada
- Faculty of Health Professions, Dalhousie University, Halifax, NS, Canada
| | | | - Zara Forbrigger
- Department of Pediatrics, Division of Hematology and Oncology, Izaak Walton Killam (IWK) Health, Halifax, NS, Canada
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
| | - Joseph P. Bielawski
- Department of Biology, Dalhousie University, Halifax, NS, Canada
- Institute for Comparative Genomics, Dalhousie University, Halifax, NS, Canada
- Department of Mathematics & Statistics, Dalhousie University, Halifax, NS, Canada
| | - Morgan G.I. Langille
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Johan Van Limbergen
- Department of Paediatric Gastroenterology and Nutrition, Emma Children’s Hospital, Amsterdam University Medical Centers, Amsterdam, Netherlands
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology and Metabolism, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Ketan Kulkarni
- Department of Pediatrics, Division of Hematology and Oncology, Izaak Walton Killam (IWK) Health, Halifax, NS, Canada
- *Correspondence: Ketan Kulkarni,
| |
Collapse
|
36
|
McEachron KR, Nalluri H, Beilman GJ, Kirchner VA, Pruett TL, Freeman ML, Trikudanathan G, Staley C, Bellin MD. Decreased Intestinal Microbiota Diversity Is Associated With Increased Gastrointestinal Symptoms in Patients With Chronic Pancreatitis. Pancreas 2022; 51:649-656. [PMID: 36099525 PMCID: PMC9547966 DOI: 10.1097/mpa.0000000000002096] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
OBJECTIVES Chronic pancreatitis (CP) is characterized by abdominal pain, recurrent hospitalizations, frequent exposure to antibiotics, nutritional deficiencies, and chronic opioid use. Data describing the gut microbial community structure of patients with CP is limited. We aimed to compare gut microbiota of a group of patients with severe CP being considered for total pancreatectomy with islet autotransplantation (TPIAT) with those of healthy controls and to associate these differences with severity of clinical symptoms. METHODS We collected stool from healthy donors (n = 14) and patients with CP (n = 20) undergoing workup for TPIAT, in addition to clinical metadata and a validated abdominal symptoms severity survey. RESULTS Patients with CP had significantly lower alpha diversity than healthy controls ( P < 0.001). There was a significantly increased mean relative abundance of Faecalibacterium in healthy controls compared with patients with CP ( P = 0.02). Among participants with CP, those with lower alpha diversity reported worse functional abdominal symptoms ( P = 0.006). CONCLUSIONS These findings indicate that changes in gut microbial community structure may contribute to gastrointestinal symptoms and provide basis for future studies on whether enrichment of healthy commensal bacteria such as Faecalibacterium could provide clinically meaningful improvements in outcomes for CP patients undergoing TPIAT.
Collapse
|
37
|
Nagata N, Nishijima S, Kojima Y, Hisada Y, Imbe K, Miyoshi-Akiyama T, Suda W, Kimura M, Aoki R, Sekine K, Ohsugi M, Miki K, Osawa T, Ueki K, Oka S, Mizokami M, Kartal E, Schmidt TSB, Molina-Montes E, Estudillo L, Malats N, Trebicka J, Kersting S, Langheinrich M, Bork P, Uemura N, Itoi T, Kawai T. Metagenomic Identification of Microbial Signatures Predicting Pancreatic Cancer From a Multinational Study. Gastroenterology 2022; 163:222-238. [PMID: 35398347 DOI: 10.1053/j.gastro.2022.03.054] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 02/23/2022] [Accepted: 03/29/2022] [Indexed: 12/26/2022]
Abstract
BACKGROUND & AIMS To identify gut and oral metagenomic signatures that accurately predict pancreatic ductal carcinoma (PDAC) and to validate these signatures in independent cohorts. METHODS We conducted a multinational study and performed shotgun metagenomic analysis of fecal and salivary samples collected from patients with treatment-naïve PDAC and non-PDAC controls in Japan, Spain, and Germany. Taxonomic and functional profiles of the microbiomes were characterized, and metagenomic classifiers to predict PDAC were constructed and validated in external datasets. RESULTS Comparative metagenomics revealed dysbiosis of both the gut and oral microbiomes and identified 30 gut and 18 oral species significantly associated with PDAC in the Japanese cohort. These microbial signatures achieved high area under the curve values of 0.78 to 0.82. The prediction model trained on the Japanese gut microbiome also had high predictive ability in Spanish and German cohorts, with respective area under the curve values of 0.74 and 0.83, validating its high confidence and versatility for PDAC prediction. Significant enrichments of Streptococcus and Veillonella spp and a depletion of Faecalibacterium prausnitzii were common gut signatures for PDAC in all the 3 cohorts. Prospective follow-up data revealed that patients with certain gut and oral microbial species were at higher risk of PDAC-related mortality. Finally, 58 bacteriophages that could infect microbial species consistently enriched in patients with PDAC across the 3 countries were identified. CONCLUSIONS Metagenomics targeting the gut and oral microbiomes can provide a powerful source of biomarkers for identifying individuals with PDAC and their prognoses. The identification of shared gut microbial signatures for PDAC in Asian and European cohorts indicates the presence of robust and global gut microbial biomarkers.
Collapse
Affiliation(s)
- Naoyoshi Nagata
- Department of Gastroenterological Endoscopy, Tokyo Medical University, Tokyo, Japan; Department of Gastroenterology and Hepatology, National Center for Global Health and Medicine, Tokyo, Japan.
| | - Suguru Nishijima
- Computational Bio-Big Data Open Innovation Lab, National Institute of Advanced Industrial Science and Technology, Tokyo, Japan; Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, Japan; Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany.
| | - Yasushi Kojima
- Department of Gastroenterology and Hepatology, National Center for Global Health and Medicine, Tokyo, Japan
| | - Yuya Hisada
- Department of Gastroenterology and Hepatology, National Center for Global Health and Medicine, Tokyo, Japan
| | - Koh Imbe
- Department of Gastroenterology and Hepatology, National Center for Global Health and Medicine, Tokyo, Japan
| | - Tohru Miyoshi-Akiyama
- Pathogenic Microbe Laboratory, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Wataru Suda
- Laboratory for Microbiome Sciences, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Moto Kimura
- Department of Clinical Research Strategic Planning Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
| | - Ryo Aoki
- Institute of Health Sciences, Ezaki Glico Co., Ltd., Osaka, Japan
| | - Katsunori Sekine
- Department of Gastroenterology and Hepatology, National Center for Global Health and Medicine, Kohnodai Hospital, Tokyo, Japan
| | - Mitsuru Ohsugi
- Department of Diabetes, Endocrinology, and Metabolism, Center Hospital, National Center for Global Health and Medicine, Tokyo, Japan; Diabetes and Metabolism Information Center, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Kuniko Miki
- Department of Gastroenterological Endoscopy, Tokyo Medical University, Tokyo, Japan; Department of Gastroenterology and Hepatology, National Center for Global Health and Medicine, Tokyo, Japan
| | - Tsuyoshi Osawa
- Division of Nutriomics and Oncology, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Kohjiro Ueki
- Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Shinichi Oka
- AIDS Clinical Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Masashi Mizokami
- Genome Medical Sciences Project, Research Institute, National Center for Global Health and Medicine, Chiba, Japan
| | - Ece Kartal
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Thomas S B Schmidt
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Esther Molina-Montes
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center (CNIO), Madrid, and CIBERONC, Spain
| | - Lidia Estudillo
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center (CNIO), Madrid, and CIBERONC, Spain
| | - Nuria Malats
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center (CNIO), Madrid, and CIBERONC, Spain
| | - Jonel Trebicka
- Section for Translational Hepatology, Department of Internal Medicine I, Goehte University Frankfurt, Frankfurt, Germany; European Foundation for the Study of Chronic Liver Failure, Barcelona, Spain
| | - Stephan Kersting
- Department of Surgery, University Hospital of Erlangen, Erlangen, Germany; Department of Surgery, University Clinic Greifswald, Greifswald, Germany
| | - Melanie Langheinrich
- Department of Surgery, University Hospital of Erlangen, Erlangen, Germany; Department of Surgery, University Clinic Greifswald, Greifswald, Germany
| | - Peer Bork
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany; Department of Bioinformatics, Biocenter, University of Würzburg, Würzburg, Germany
| | - Naomi Uemura
- Department of Gastroenterological Endoscopy, Tokyo Medical University, Tokyo, Japan; Department of Gastroenterology and Hepatology, National Center for Global Health and Medicine, Kohnodai Hospital, Tokyo, Japan
| | - Takao Itoi
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo, Japan
| | - Takashi Kawai
- Department of Gastroenterological Endoscopy, Tokyo Medical University, Tokyo, Japan
| |
Collapse
|
38
|
MiR155 Disrupts the Intestinal Barrier by Inducing Intestinal Inflammation and Altering the Intestinal Microecology in Severe Acute Pancreatitis. Dig Dis Sci 2022; 67:2209-2219. [PMID: 34341909 DOI: 10.1007/s10620-021-07022-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 04/21/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Intestinal dysfunction is a common complication of acute pancreatitis. MiR155 may be involved in the occurrence and development of intestinal dysfunction mediated by acute pancreatitis, but the specific mechanism is not clear. AIMS To investigate the effect of miR155 on severe acute pancreatitis (SAP)-associated intestinal dysfunction and its possible mechanism in a mice model. METHODS In this study, SAP mice model was induced by intraperitoneal injection of caerulein and LPS in combination. Adeno-associated virus (AAV) was given by tail vein injection before the SAP model. The pancreatic and intestinal histopathology changes were analyzed. Cecal tissue was collected for 16S rRNA Gene Sequencing. Intestinal barrier proteins ZO-1 and E-cad were measured by Immunohistochemistry Staining and Western Blot, respectively. Intestinal tissue miR155 and inflammatory factors TNF-α, IL-1β, and IL-6 were detected by Q-PCR. The expression levels of protein associated with TNF-α and TLR4/MYD88 pathway in the intestinal were detected. RESULTS In miR155 overexpression SAP group, the levels of tissue inflammatory factor were significantly increased, intestinal barrier proteins were significantly decreased, and the injury of intestinal was aggravated. Bacterial 16S rRNA sequencing was performed, showing miR155 promotes gut microbiota dysbiosis. The levels of TNF-α, TLR4, and MYD88 in the intestinal were detected, suggesting that miR155 may regulate gut microbiota and activate the TLR4/MYD88 pathway, thereby affecting the release of inflammatory mediators and regulating SAP-related intestinal injury. After application of miR155-sponge, imbalance of intestinal flora and destruction of intestinal barrier-related proteins have been alleviated. The release of inflammatory mediators decreased, and the histopathology injury of intestinal was improved obviously. CONCLUSION MiR155 may play an important role in SAP-associated intestinal dysfunction. MiR155 can significantly alter the intestinal microecology, aggravated intestinal inflammation through TLR4/MYD88 pathway, and disrupts the intestinal barrier in SAP mice.
Collapse
|
39
|
Tegegne BA, Kebede B. Probiotics, their prophylactic and therapeutic applications in human health development: A review of the literature. Heliyon 2022; 8:e09725. [PMID: 35785237 PMCID: PMC9240980 DOI: 10.1016/j.heliyon.2022.e09725] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/03/2022] [Accepted: 06/09/2022] [Indexed: 02/07/2023] Open
Abstract
Antibiotics do not differentiate between good and bad germs, disrupting normal microflora and causing vitamin deficiency in the human body. They also kill healthy bacteria in the gut and genital tract on a large scale, weakening the host's defense mechanism. Probiotics are a colony of bacteria that live in our intestines and are regarded as a metabolic 'organ' due to their beneficial effects on human health, including metabolism and immunological function. They are used in clinical settings to prevent and treat conditions such as diarrhoea, colon cancer, hypertension, diabetes, acute pancreatitis, Helicobacter pylori infection, ventilator-associated pneumonia, migraine and autism. Probiotics may modify immunological activity by increasing innate and adaptive immune responses, altering microbial habitat in the intestine, improving gut barrier function, competitive adherence to the mucosa and epithelium, and producing antimicrobial compounds. The aim of this study is to index that further in depth researches to be conducted on probiotics pivotal role in the prophylaxis and therapeutic usage for a variety of disease that may or may not have treatment alternatives. Key words such as probiotics, microbiota, prophylactics, and therapeutic applications were searched extensively in research databases such as PubMed, PubMed Central (PMC), Scopus, Web of Science, Research Gate, Google Scholar, and Cochrane Library. This concise narrative review article summarized primarily the history, selection, mechanism/mode of action, recent advances in prophylactic and therapeutic applications, and future directions in the use of probiotics for prophylactic and therapeutic applications.
Collapse
|
40
|
Characterization of the fecal microbiota in gastrointestinal cancer patients and healthy people. Clin Transl Oncol 2022; 24:1134-1147. [PMID: 35167015 DOI: 10.1007/s12094-021-02754-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 12/08/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND The incidence and mortality of gastrointestinal (GI) tumors are high in China. Some studies suggest that the gut microbiota is related to the occurrence and development of tumors. At present, there are no prospective studies based on the correlation between gastrointestinal tumors and gut microbiota in the Chinese population. The objective of this report is to characterize the fecal microbiota in healthy control participants and patients with esophageal cancer, gastric cancer, and colorectal cancer. METHODS Patients with locally advanced or metastatic esophageal, gastric, and colorectal cancer were enrolled, and healthy people were included as controls. 16S rRNA sequencing was used to analyze the characteristics of fecal microbiota. PICRUSt software was used for functional prediction. RESULTS Significant differences in the composition and abundance of fecal microbiota were identified between gastrointestinal cancer patients (n = 130) and healthy controls (n = 147). The abundance of Faecalibacterium prausnitzii, Clostridium clostridioforme and Bifidobacterium adolescent in tumor groups were all significantly lower than in the control group (P < 0.05). The levels of Blautia producta and R. faecis in the gastric (n = 46) and colorectal cancer (n = 44) groups were significantly lower than those in the control group (P < 0.05). The level of Butyricicoccus pullicaecorum in the esophageal cancer (n = 40) and gastric cancer groups was significantly lower than that in the control group (P < 0.05). B. fragilis, Akkermansia muciniphila, Clostridium hathewayi and Alistipes finegoldii were overabundant in the different tumor groups compared with the control (P < 0.05). We observed significant differences in functional metabolism and cell biological function between the tumor and control groups (P < 0.05). Optimal microbial markers were identified on a random forest model and achieved an area under the curve of 85.59% between 130 GI cancer samples and 147 control samples. The respective AUC values were 86.89%, 97.11%, and 79.1% in detecting esophageal cancer, gastric cancer, and colorectal cancer. CONCLUSIONS Patients with esophageal or gastric cancers had similar features of fecal bacteria as those with colorectal cancer. The metabolic function of fecal bacteria in the gastrointestinal cancer patients and the healthy controls were different. The microbial signatures may potentially be applied to distinguish GI cancer patients from healthy people as a non-invasive diagnostic biomarker.
Collapse
|
41
|
Upper Gastrointestinal Cancer and Liver Cirrhosis. Cancers (Basel) 2022; 14:cancers14092269. [PMID: 35565397 PMCID: PMC9105927 DOI: 10.3390/cancers14092269] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/26/2022] [Accepted: 04/29/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary There is a higher incidence rate of upper gastrointestinal cancer in those with liver cirrhosis. The contributing factors include gastric ulcers, congestive gastropathy, zinc deficiency, alcohol drinking, tobacco use and gut microbiota. Most of the de novo malignancies that develop after liver transplantation for cirrhotic patients are upper gastrointestinal cancers. The surgical risk of upper gastrointestinal cancers in cirrhotic patients with advanced liver cirrhosis is higher. Abstract The extended scope of upper gastrointestinal cancer can include esophageal cancer, gastric cancer and pancreatic cancer. A higher incidence rate of gastric cancer and esophageal cancer in patients with liver cirrhosis has been reported. It is attributable to four possible causes which exist in cirrhotic patients, including a higher prevalence of gastric ulcers and congestive gastropathy, zinc deficiency, alcohol drinking and tobacco use and coexisting gut microbiota. Helicobacter pylori infection enhances the development of gastric cancer. In addition, Helicobacter pylori, Porphyromonas gingivalis and Aggregatibacter actinomycetemcomitans also contribute to the development of pancreatic cancer in cirrhotic patients. Cirrhotic patients (especially those with alcoholic liver cirrhosis) who undergo liver transplantation have a higher overall risk of developing de novo malignancies. Most de novo malignancies are upper gastrointestinal malignancies. The prognosis is usually poor. Considering the surgical risk of upper gastrointestinal cancer among those with liver cirrhosis, a radical gastrectomy with D1 or D2 lymph node dissection can be undertaken in Child class A patients. D1 lymph node dissection can be performed in Child class B patients. Endoscopic submucosal dissection for gastric cancer or esophageal cancer can be undertaken safely in selected cirrhotic patients. In Child class C patients, a radical gastrectomy is potentially fatal. Pancreatic radical surgery should be avoided in those with liver cirrhosis with Child class B or a MELD score over 15. The current review focuses on the recent reports on some factors in liver cirrhosis that contribute to the development of upper gastrointestinal cancer. Quitting alcohol drinking and tobacco use is important. How to decrease the risk of the development of gastrointestinal cancer in those with liver cirrhosis remains a challenging problem.
Collapse
|
42
|
Halimi A, Gabarrini G, Sobkowiak MJ, Ateeb Z, Davanian H, Gaiser RA, Arnelo U, Valente R, Wong AY, Moro CF, Del Chiaro M, Özenci V, Chen MS. Isolation of pancreatic microbiota from cystic precursors of pancreatic cancer with intracellular growth and DNA damaging properties. Gut Microbes 2022; 13:1983101. [PMID: 34816784 PMCID: PMC8632270 DOI: 10.1080/19490976.2021.1983101] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Emerging research suggests gut microbiome may play a role in pancreatic cancer initiation and progression, but cultivation of the cancer microbiome remains challenging. This pilot study aims to investigate the possibility to cultivate pancreatic microbiome from pancreatic cystic lesions associated with invasive cancer. Intra-operatively acquired pancreatic cyst fluid samples showed culture-positivity mainly in the intraductal papillary mucinous neoplasm (IPMN) group of lesions. MALDI-TOF MS profiling analysis shows Gammaproteobacteria and Bacilli dominate among individual bacteria isolates. Among cultivated bacteria, Gammaproteobacteria, particularly Klebsiella pneumoniae, but also Granulicatella adiacens and Enterococcus faecalis, demonstrate consistent pathogenic properties in pancreatic cell lines tested in ex vivo co-culture models. Pathogenic properties include intracellular survival capability, cell death induction, or causing DNA double-strand breaks in the surviving cells resembling genotoxic effects. This study provides new insights into the role of the pancreatic microbiota in the intriguing link between pancreatic cystic lesions and cancer.
Collapse
Affiliation(s)
- Asif Halimi
- Division of Surgery, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden,Department of Surgical and Perioperative Sciences, Surgery, Umeå University, Umeå, Sweden
| | - Giorgio Gabarrini
- Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
| | | | - Zeeshan Ateeb
- Division of Surgery, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Haleh Davanian
- Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
| | | | - Urban Arnelo
- Division of Surgery, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden,Department of Surgical and Perioperative Sciences, Surgery, Umeå University, Umeå, Sweden
| | - Roberto Valente
- Division of Surgery, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden,Department of Surgical and Perioperative Sciences, Surgery, Umeå University, Umeå, Sweden
| | - Alicia Y.W. Wong
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden,Department of Clinical Microbiology F 72, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Carlos Fernández Moro
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden,Department of Clinical Pathology/Cytology, Karolinska University Hospital, Huddinge, Sweden
| | - Marco Del Chiaro
- Department of Surgery, University of Colorado Denver – Anschutz Medical Campus, Aurora, CO, USA
| | - Volkan Özenci
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden,Department of Clinical Microbiology F 72, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Margaret Sällberg Chen
- Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden,CONTACT Margaret Sällberg Chen Department of Dental Medicine, Karolinska Institutet, Huddinge14141, Sweden
| |
Collapse
|
43
|
A Comprehensive Review of the Current and Future Role of the Microbiome in Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2022; 14:cancers14041020. [PMID: 35205769 PMCID: PMC8870349 DOI: 10.3390/cancers14041020] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/11/2022] [Accepted: 02/15/2022] [Indexed: 02/07/2023] Open
Abstract
Simple Summary This review summarizes the current literature related to the microbiome and pancreatic ductal adenocarcinoma (PDAC). The aim of this review is to explore the current role of the microbiome in the disease process, screening/diagnostics and to postulate the future role with regards to therapeutic strategies including chemotherapy, immunotherapy and surgery. We further explore the future of microbiome modulation (faecal microbiome transplants, bacterial consortiums, anti-microbials and probiotics), their applications and how we can improve the future of microbiome modulation in a bid to improve PDAC outcomes. Abstract Pancreatic ductal adenocarcinoma (PDAC) is expected to become the second most common cause of cancer death in the USA by 2030, yet progress continues to lag behind that of other cancers, with only 9% of patients surviving beyond 5 years. Long-term survivorship of PDAC and improving survival has, until recently, escaped our understanding. One recent frontier in the cancer field is the microbiome. The microbiome collectively refers to the extensive community of bacteria and fungi that colonise us. It is estimated that there is one to ten prokaryotic cells for each human somatic cell, yet, the significance of this community in health and disease has, until recently, been overlooked. This review examines the role of the microbiome in PDAC and how it may alter survival outcomes. We evaluate the possibility of employing microbiomic signatures as biomarkers of PDAC. Ultimately this review analyses whether the microbiome may be amenable to targeting and consequently altering the natural history of PDAC.
Collapse
|
44
|
Zhang T, Gao G, Sakandar HA, Kwok LY, Sun Z. Gut Dysbiosis in Pancreatic Diseases: A Causative Factor and a Novel Therapeutic Target. Front Nutr 2022; 9:814269. [PMID: 35242797 PMCID: PMC8885515 DOI: 10.3389/fnut.2022.814269] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 01/21/2022] [Indexed: 12/12/2022] Open
Abstract
Pancreatic-related disorders such as pancreatitis, pancreatic cancer, and type 1 diabetes mellitus (T1DM) impose a substantial challenge to human health and wellbeing. Even though our understanding of the initiation and progression of pancreatic diseases has broadened over time, no effective therapeutics is yet available for these disorders. Mounting evidence suggests that gut dysbiosis is closely related to human health and disease, and pancreatic diseases are no exception. Now much effort is under way to explore the correlation and eventually potential causation between the gut microbiome and the course of pancreatic diseases, as well as to develop novel preventive and/or therapeutic strategies of targeted microbiome modulation by probiotics, prebiotics, synbiotics, postbiotics, and fecal microbiota transplantation (FMT) for these multifactorial disorders. Attempts to dissect the intestinal microbial landscape and its metabolic profile might enable deep insight into a holistic picture of these complex conditions. This article aims to review the subtle yet intimate nexus loop between the gut microbiome and pancreatic diseases, with a particular focus on current evidence supporting the feasibility of preventing and controlling pancreatic diseases via microbiome-based therapeutics and therapies.
Collapse
Affiliation(s)
- Tao Zhang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Guangqi Gao
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Hafiz Arbab Sakandar
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Lai-Yu Kwok
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Zhihong Sun
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China
- *Correspondence: Zhihong Sun
| |
Collapse
|
45
|
Tao J, Cheema H, Kesh K, Dudeja V, Dawra R, Roy S. Chronic pancreatitis in a caerulein-induced mouse model is associated with an altered gut microbiome. Pancreatology 2022; 22:30-42. [PMID: 34949545 PMCID: PMC8748396 DOI: 10.1016/j.pan.2021.12.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 10/29/2021] [Accepted: 12/12/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND Chronic pancreatitis (CP) is an inflammatory disease of the pancreas with loss of exocrine/endocrine functions as well as development of fibrosis. Dysbiosis of gut microbiome has been shown to be involved in the pathogenesis of many disease processes. Therefore, we aim to investigate the alteration in gut microbiome associated with CP in caerulein-induced mouse model. METHODS CP was induced in C57Bl/6 by using caerulein injections (50 μg/kg/h, i.p., x7, twice weekly for 10 weeks). Stool samples were collected either one week after end of injection (10-week CP) or 6 weeks (16-week CP). DNA was extracted from stool samples and V4 region of 16S rDNA was sequenced for microbiome analysis. RESULTS CP was strongly associated with the alteration in the composition of the gut microbiome, evidenced by differences in α and β diversity. When β diversity was measured using both weighted and unweighted UniFrac distances, stool from control mice is significantly different from mice on 10-week or 16-week CP (q < 0.01). The α-diversity measured by Faith's phylogenetic diversity was lowest in stool from healthy control and highest in stool from mice with 16-week CP (p < 0.001). Bacteria taxa differentially enriched in CP samples were detected using linear discriminant analysis. Bacteria from genera Bifidobacterium, Akkermansia, and Desulfovibrio were enriched in samples from 10-week CP mice. Bacteria from genera Allobaculum, Prevotella, and Bacteroides were enriched in samples from 16-week CP mice. CONCLUSION Together, these analyses reveal pronounced alteration in the gut microbiome composition, diversity, and function when mice develop CP.
Collapse
Affiliation(s)
- J Tao
- Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, 33136, USA
| | - H Cheema
- Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, 33136, USA
| | - K Kesh
- Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, 33136, USA
| | - V Dudeja
- Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, 33136, USA
| | - R Dawra
- Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, 33136, USA.
| | - S Roy
- Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, 33136, USA.
| |
Collapse
|
46
|
Futagami S, Wakabayashi M. Pancreatic dysfunction and duodenal inflammatory responses coordinate with refractory epigastric pain including functional dyspepsia "A narrative review". J NIPPON MED SCH 2022; 89:255-262. [DOI: 10.1272/jnms.jnms.2022_89-311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
47
|
Xu F, Yang C, Tang M, Wang M, Cheng Z, Chen D, Chen X, Liu K. The Role of Gut Microbiota and Genetic Susceptibility in the Pathogenesis of Pancreatitis. Gut Liver 2021; 16:686-696. [PMID: 34911043 PMCID: PMC9474482 DOI: 10.5009/gnl210362] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/06/2021] [Accepted: 09/17/2021] [Indexed: 11/22/2022] Open
Abstract
Pancreatitis is one of the most common inflammatory diseases of the pancreas caused by autodigestion induced by excessive premature protease activation. However, recognition of novel pathophysiological mechanisms remains a still challenge. Both genetic and environmental factors contribute to the pathogenesis of pancreatitis, and the gut microbiota is a potential source of an environmental effect. In recent years, several new frontiers in gut microbiota and genetic risk assessment research have emerged and improved the understanding of the disease. These investigations showed that the disease progression of pancreatitis could be regulated by the gut microbiome, either through a translocation influence or in a host immune response manner. Meanwhile, the onset of the disease is also associated with the heritage of a pathogenic mutation, and the disease progression could be modified by genetic risk factors. In this review, we focused on the recent advances in the role of gut microbiota in the pathogenesis of pancreatitis, and the genetic susceptibility in pancreatitis.
Collapse
Affiliation(s)
- Fumin Xu
- Department of Gastroenterology, Daping Hospital, Army Medical University, Chongqing, China
| | - Chunmei Yang
- Department of Gastroenterology, Daping Hospital, Army Medical University, Chongqing, China
| | - Mingcheng Tang
- Department of Gastroenterology, Daping Hospital, Army Medical University, Chongqing, China
| | - Ming Wang
- Department of Gastroenterology, Daping Hospital, Army Medical University, Chongqing, China
| | - Zhenhao Cheng
- Department of Gastroenterology, Daping Hospital, Army Medical University, Chongqing, China
| | - Dongfeng Chen
- Department of Gastroenterology, Daping Hospital, Army Medical University, Chongqing, China
| | - Xiao Chen
- Department of Nuclear Medicine, Daping Hospital, Army Medical University, Chongqing, China
| | - Kaijun Liu
- Department of Gastroenterology, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
48
|
Kunovský L, Dítě P, Jabandžiev P, Eid M, Poredská K, Vaculová J, Sochorová D, Janeček P, Tesaříková P, Blaho M, Trna J, Hlavsa J, Kala Z. Causes of Exocrine Pancreatic Insufficiency Other Than Chronic Pancreatitis. J Clin Med 2021; 10:jcm10245779. [PMID: 34945075 PMCID: PMC8708123 DOI: 10.3390/jcm10245779] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/30/2021] [Accepted: 12/02/2021] [Indexed: 12/12/2022] Open
Abstract
Exocrine pancreatic insufficiency (EPI), an important cause of maldigestion and malnutrition, results from primary pancreatic disease or is secondary to impaired exocrine pancreatic function. Although chronic pancreatitis is the most common cause of EPI, several additional causes exist. These include pancreatic tumors, pancreatic resection procedures, and cystic fibrosis. Other diseases and conditions, such as diabetes mellitus, celiac disease, inflammatory bowel disease, and advanced patient age, have also been shown to be associated with EPI, but the exact etiology of EPI has not been clearly elucidated in these cases. The causes of EPI can be divided into loss of pancreatic parenchyma, inhibition or inactivation of pancreatic secretion, and postcibal pancreatic asynchrony. Pancreatic enzyme replacement therapy (PERT) is indicated for the conditions described above presenting with clinically clear steatorrhea, weight loss, or symptoms related to maldigestion and malabsorption. This review summarizes the current literature concerning those etiologies of EPI less common than chronic pancreatitis, the pathophysiology of the mechanisms of EPI associated with each diagnosis, and treatment recommendations.
Collapse
Affiliation(s)
- Lumír Kunovský
- Department of Gastroenterology and Internal Medicine, University Hospital Brno, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic; (L.K.); (P.D.); (K.P.); (J.V.)
- Department of Surgery, University Hospital Brno, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic; (D.S.); (P.J.); (Z.K.)
| | - Petr Dítě
- Department of Gastroenterology and Internal Medicine, University Hospital Brno, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic; (L.K.); (P.D.); (K.P.); (J.V.)
- Department of Gastroenterology and Internal Medicine, University Hospital Ostrava, Faculty of Medicine, University of Ostrava, 70852 Ostrava, Czech Republic;
| | - Petr Jabandžiev
- Department of Pediatrics, University Hospital Brno, Faculty of Medicine, Masaryk University, 61300 Brno, Czech Republic;
- Central European Institute of Technology, Masaryk University, 62500 Brno, Czech Republic
| | - Michal Eid
- Department of Hematology, Oncology and Internal Medicine, University Hospital Brno, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic;
| | - Karolina Poredská
- Department of Gastroenterology and Internal Medicine, University Hospital Brno, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic; (L.K.); (P.D.); (K.P.); (J.V.)
| | - Jitka Vaculová
- Department of Gastroenterology and Internal Medicine, University Hospital Brno, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic; (L.K.); (P.D.); (K.P.); (J.V.)
| | - Dana Sochorová
- Department of Surgery, University Hospital Brno, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic; (D.S.); (P.J.); (Z.K.)
| | - Pavel Janeček
- Department of Surgery, University Hospital Brno, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic; (D.S.); (P.J.); (Z.K.)
| | - Pavla Tesaříková
- Department of Internal Medicine, Hospital Boskovice, 68001 Boskovice, Czech Republic;
| | - Martin Blaho
- Department of Gastroenterology and Internal Medicine, University Hospital Ostrava, Faculty of Medicine, University of Ostrava, 70852 Ostrava, Czech Republic;
| | - Jan Trna
- Department of Gastroenterology and Internal Medicine, University Hospital Brno, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic; (L.K.); (P.D.); (K.P.); (J.V.)
- Department of Internal Medicine, Hospital Boskovice, 68001 Boskovice, Czech Republic;
- Department of Gastroenterology and Digestive Endoscopy, Masaryk Memorial Cancer Institute Brno, 60200 Brno, Czech Republic
- Correspondence: (J.T.); (J.H.)
| | - Jan Hlavsa
- Department of Surgery, University Hospital Brno, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic; (D.S.); (P.J.); (Z.K.)
- Correspondence: (J.T.); (J.H.)
| | - Zdeněk Kala
- Department of Surgery, University Hospital Brno, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic; (D.S.); (P.J.); (Z.K.)
| |
Collapse
|
49
|
Daniluk J, Daniluk U, Rogalski P, Dabrowski A, Swidnicka-Siergiejko A. Microbiome-Friend or Foe of Pancreatic Cancer? J Clin Med 2021; 10:jcm10235624. [PMID: 34884327 PMCID: PMC8658245 DOI: 10.3390/jcm10235624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 11/15/2021] [Accepted: 11/27/2021] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma is one of the deadliest human neoplasms. Despite the development of new surgical and adjuvant therapies, the prognosis remains very poor, with the overall survival rate not exceeding 9%. There is now increasing evidence that the human microbiome, which is involved in many physiological functions, including the regulation of metabolic processes and the modulation of the immune system, is possibly linked to pancreatic oncogenesis. However, the exact mechanisms of action are poorly understood. Our review summarizes the current understanding of how the microbiome affects pancreatic cancer development and progression. We discuss potential pathways of microbe translocation to the pancreas, as well as the mechanism of their action. We describe the role of the microbiome as a potential marker of pancreatic cancer diagnosis, progression, and survival. Finally, we discuss the possibilities of modifying the microbiome to improve treatment effectiveness for this deadly disease.
Collapse
Affiliation(s)
- Jaroslaw Daniluk
- Department of Gastroenterology and Internal Medicine, Medical University of Bialystok, 15-276 Bialystok, Poland; (P.R.); (A.D.); (A.S.-S.)
- Correspondence: ; Tel.: +48-(85)-746-82-34; Fax: +48-(85)-746-85-06
| | - Urszula Daniluk
- Department of Pediatrics, Gastroenterology, Hepatology, Nutrition and Allergology, Medical University of Bialystok, 15-274 Bialystok, Poland;
| | - Pawel Rogalski
- Department of Gastroenterology and Internal Medicine, Medical University of Bialystok, 15-276 Bialystok, Poland; (P.R.); (A.D.); (A.S.-S.)
| | - Andrzej Dabrowski
- Department of Gastroenterology and Internal Medicine, Medical University of Bialystok, 15-276 Bialystok, Poland; (P.R.); (A.D.); (A.S.-S.)
| | - Agnieszka Swidnicka-Siergiejko
- Department of Gastroenterology and Internal Medicine, Medical University of Bialystok, 15-276 Bialystok, Poland; (P.R.); (A.D.); (A.S.-S.)
| |
Collapse
|
50
|
Patel BK, Patel KH, Bhatia M, Iyer SG, Madhavan K, Moochhala SM. Gut microbiome in acute pancreatitis: A review based on current literature. World J Gastroenterol 2021; 27:5019-5036. [PMID: 34497432 PMCID: PMC8384740 DOI: 10.3748/wjg.v27.i30.5019] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 06/04/2021] [Accepted: 06/23/2021] [Indexed: 02/06/2023] Open
Abstract
The gut microbiome is a complex microbial community, recognized for its potential role in physiology, health, and disease. The available evidence supports the role of gut dysbiosis in pancreatic disorders, including acute pancreatitis (AP). In AP, the presence of gut barrier damage resulting in increased mucosal permeability may lead to translocation of intestinal bacteria, necrosis of pancreatic and peripancreatic tissue, and infection, often accompanied by multiple organ dysfunction syndrome. Preserving gut microbial homeostasis may reduce the systemic effects of AP. A growing body of evidence suggests the possible involvement of the gut microbiome in various pancreatic diseases, including AP. This review discusses the possible role of the gut microbiome in AP. It highlights AP treatment and supplementation with prebiotics, synbiotics, and probiotics to maintain gastrointestinal microbial balance and effectively reduce hospitalization, morbidity and mortality in an early phase. It also addresses novel therapeutic areas in the gut microbiome, personalized treatment, and provides a roadmap of human microbial contributions to AP that have potential clinical benefit.
Collapse
Affiliation(s)
- Bharati Kadamb Patel
- Department of Surgery, National University of Singapore, Singapore 119228, Singapore
| | - Kadamb H Patel
- School of Applied Sciences, Temasek Polytechnic, Singapore 529757, Singapore
| | - Madhav Bhatia
- Department of Pathology, University of Otago, Christchurch 8140, New Zealand
| | - Shridhar Ganpati Iyer
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
- National University Hospital, National University of Singapore, Singapore 119228, Singapore
| | - Krishnakumar Madhavan
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
- National University Hospital, National University of Singapore, Singapore 119228, Singapore
| | - Shabbir M Moochhala
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| |
Collapse
|