1
|
Minty M, Germain A, Sun J, Kaglan G, Servant F, Lelouvier B, Misselis E, Neagoe RM, Rossella M, Cardellini M, Burcelin R, Federici M, Fernandez-Real JM, Blasco-Baque V. Identifying the location-dependent adipose tissue bacterial DNA signatures in obese patients that predict body weight loss. Gut Microbes 2025; 17:2439105. [PMID: 39714075 DOI: 10.1080/19490976.2024.2439105] [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: 02/01/2024] [Revised: 11/08/2024] [Accepted: 11/26/2024] [Indexed: 12/24/2024] Open
Abstract
Recent sets of evidence have described profiles of 16S rDNA sequences in host tissues, notably in fat pads that are significantly overrepresented and can serve as signatures of metabolic disease. However, these recent and original observations need to be further detailed and functionally defined. Here, using state-of-the-art targeted DNA sequencing and discriminant predictive approaches, we describe, from the longitudinal FLORINASH cohort of patients who underwent bariatric surgery, visceral, and subcutaneous fat pad-specific bacterial 16SrRNA signatures. The corresponding Porphyromonadaceae, Campylobacteraceae, Prevotellaceae, Actimomycetaceae, Veillonellaceae, Anaerivoracaceae, Fusobacteriaceae, and the Clostridium family XI 16SrRNA DNA segment profiles are signatures of the subcutaneous adipose depot while Pseudomonadaceae and Micrococcacecae, 16SrRNA DNA sequence profiles characterize the visceral adipose depot. In addition, we have further identified that a specific pre-bariatric surgery adipose tissue bacterial DNA signature predicts the efficacy of body weight loss in obese patients 5-10 years after the surgery. 16SrRNA signatures discriminate (ROC ~ 1) the patients who did not maintain bodyweight loss and those who did. Second, from the 16SrRNA sequences we infer potential pathways suggestive of catabolic biochemical activities that could be signatures of subcutaneous adipose depots that predict body weight loss.
Collapse
Affiliation(s)
- Matthieu Minty
- Institut National de la Santé et de la Recherche Médicale (INSERM), InCOMM Intestine ClinicOralOmics Metabolism & Microbiota UMR1297 Inserm / Université Toulouse III, Toulouse, France
- Université Paul Sabatier (UPS), Unité Mixte de Recherche (UMR) 1297, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Toulouse, Cedex, France
| | - Alberic Germain
- Institut National de la Santé et de la Recherche Médicale (INSERM), InCOMM Intestine ClinicOralOmics Metabolism & Microbiota UMR1297 Inserm / Université Toulouse III, Toulouse, France
- Université Paul Sabatier (UPS), Unité Mixte de Recherche (UMR) 1297, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Toulouse, Cedex, France
| | - Jiuwen Sun
- Institut National de la Santé et de la Recherche Médicale (INSERM), InCOMM Intestine ClinicOralOmics Metabolism & Microbiota UMR1297 Inserm / Université Toulouse III, Toulouse, France
- Université Paul Sabatier (UPS), Unité Mixte de Recherche (UMR) 1297, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Toulouse, Cedex, France
| | - Gracia Kaglan
- Institut National de la Santé et de la Recherche Médicale (INSERM), InCOMM Intestine ClinicOralOmics Metabolism & Microbiota UMR1297 Inserm / Université Toulouse III, Toulouse, France
- Université Paul Sabatier (UPS), Unité Mixte de Recherche (UMR) 1297, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Toulouse, Cedex, France
| | | | | | - Emiri Misselis
- Institut National de la Santé et de la Recherche Médicale (INSERM), InCOMM Intestine ClinicOralOmics Metabolism & Microbiota UMR1297 Inserm / Université Toulouse III, Toulouse, France
- Université Paul Sabatier (UPS), Unité Mixte de Recherche (UMR) 1297, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Toulouse, Cedex, France
| | - Radu Mircea Neagoe
- Science and Technology "George Emil Palade" Tîrgu Mures, Second Department of Surgery, Emergency Mureş County Hospital, University of Medicine Pharmacy, Târgu Mureș, Romania
| | - Menghini Rossella
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Marina Cardellini
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Rémy Burcelin
- Institut National de la Santé et de la Recherche Médicale (INSERM), InCOMM Intestine ClinicOralOmics Metabolism & Microbiota UMR1297 Inserm / Université Toulouse III, Toulouse, France
- Université Paul Sabatier (UPS), Unité Mixte de Recherche (UMR) 1297, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Toulouse, Cedex, France
| | - Massimo Federici
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - José Manuel Fernandez-Real
- Department of Diabetes, Endocrinology and Nutrition, University Hospital of Girona 'Dr Josep Trueta'
- Institut d'Investigacio Biomedica de Girona IdibGi, CIBER Fisiopatologia de la Obesidad y Nutricion, Girona, Spain
| | - Vincent Blasco-Baque
- Institut National de la Santé et de la Recherche Médicale (INSERM), InCOMM Intestine ClinicOralOmics Metabolism & Microbiota UMR1297 Inserm / Université Toulouse III, Toulouse, France
- Université Paul Sabatier (UPS), Unité Mixte de Recherche (UMR) 1297, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Toulouse, Cedex, France
| |
Collapse
|
2
|
Wang J, Bao S, An Q, Li C, Feng J. Roles of extracellular vesicles from different origins in metabolic-associated fatty liver disease: progress and perspectives. Front Immunol 2025; 16:1544012. [PMID: 40129979 PMCID: PMC11930831 DOI: 10.3389/fimmu.2025.1544012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 02/19/2025] [Indexed: 03/26/2025] Open
Abstract
Metabolic-Associated Fatty Liver Disease (MAFLD) is the most common chronic liver disease worldwide, associated with systemic metabolic dysregulation. It can progress from simple hepatic steatosis (MAFL) to more severe conditions like Metabolic-Associated Steatohepatitis (MASH), fibrosis, cirrhosis, and Hepatocellular Carcinoma (HCC). There is a critical lack of reliable non-invasive diagnostic methods and effective pharmaceutical treatments for MAFLD/MASH, emphasizing the need for further research. Extracellular vesicles (EVs) are nanoscale structures that play important roles in cell signaling by delivering bioactive molecules. However, there is a significant gap in literature regarding the roles of EVs from hosts, plants, and microbiota in MAFLD. This review explores the potential of EVs from various sources-host, plants, and microbiota-as biomarkers, therapeutic agents, drug carriers, and treatment targets for MAFLD. Firstly, the roles of host-derived extracellular vesicles (EVs) in MAFLD, with a focus on cell-type specific EVs and their components-proteins, miRNAs, and lipids-for disease diagnosis and monitoring were discussed. Moreover, it highlighted the therapeutic potential of mesenchymal stem cell (MSC)-derived EVs in reducing lipid accumulation and liver injury, and immune cell-derived EVs in mitigating inflammation and fibrosis. The review also discussed the use of host-derived EVs as drug carriers and therapeutic targets due to their ability to deliver bioactive molecules that impact disease mechanisms. Additionally, it summarized research on plant-derived EVs, which help reduce liver lipid accumulation, inflammation, and enhance gut barrier function in MAFLD. Also, the review explored microbial-derived EVs as novel therapeutic targets, particularly in relation to insulin resistance, liver inflammation, and dysfunction in MAFLD. Overall, by exploring the diverse roles of EVs from host, plant, and microbiota sources in MAFLD, this review offers valuable insights into their potential as non-invasive biomarkers and novel therapeutic strategies, which could pave the way for more effective diagnostic and treatment options for this increasingly prevalent liver disease. Notably, the challenges of translating EVs into clinical practice were also thoroughly discussed, aiming to provide possible directions and strategies for future research.
Collapse
Affiliation(s)
- Jing Wang
- School of Public Health, Gansu University of Chinese Medicine, Lanzhou, China
| | - Shuoqiang Bao
- School of Public Health, Gansu University of Chinese Medicine, Lanzhou, China
| | - Qi An
- School of Public Health, Gansu University of Chinese Medicine, Lanzhou, China
| | - Caihong Li
- School of Public Health, Gansu University of Chinese Medicine, Lanzhou, China
| | - Juan Feng
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen, China
| |
Collapse
|
3
|
Wang QW, Zheng H, Yang Y, Chang X, Du Z, Hang ZN, Li ZS, Liao Z. Distinct microbial signatures and their predictive value in recurrent acute pancreatitis: insights from 5-region 16S rRNA gene sequencing. Front Immunol 2025; 16:1558983. [PMID: 40093002 PMCID: PMC11906328 DOI: 10.3389/fimmu.2025.1558983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Accepted: 02/13/2025] [Indexed: 03/19/2025] Open
Abstract
Background Recurrent acute pancreatitis (RAP) poses significant clinical challenges, with 32.3% developing to chronic pancreatitis within 5 years. The underlying microbial factors contributing to RAP remain poorly understood. This study aims to identify blood microbial signatures associated with RAP and explore the potential microbial predictors for RAP. Methods In this prospective cohort, 90 acute pancreatitis patients are classified into non-recurrent acute pancreatitis (NRAP, n=68) and RAP (n=22) groups based on the number of pancreatitis episodes. Microbial composition of blood samples is analyzed using 5-region (5R) 16S rRNA gene sequencing. Key microbial taxa and functional predictions are made. A random forest model is used to assess the predictive value of microbial features for RAP. The impact of Staphylococcus hominis (S. hominis) on RAP is further evaluated in an experimental mouse model. Results Linear discriminant analysis effect size (LEfSe) analysis highlights significant microbial differences, with Paracoccus aminovorans, Corynebacterium glucuronolyticum and S. hominis being prominent in RAP. Functional predictions indicate enrichment of metabolic pathways in the RAP group. Random forest analysis identifies key microbial taxa with an AUC value of 0.759 for predicting RAP. Experimental validation shows that S. hominis exacerbates pancreatic inflammation in mice. Conclusions This study identifies distinct clinical and microbial features associated with RAP, emphasizing the role of specific bacterial taxa in pancreatitis recurrence. The findings suggest that microbial profiling could enhance the diagnosis and management of RAP, paving the way for personalized therapeutic approaches.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Zhao-Shen Li
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Zhuan Liao
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
4
|
Chen H, Yao X, Yang C, Zhang Y, Dong H, Zhai J, Fan D, Zhou Q. Distinctive circulating microbial metagenomic signatures in the plasma of patients with lung cancer and their potential value as molecular biomarkers. J Transl Med 2025; 23:186. [PMID: 39953591 PMCID: PMC11829562 DOI: 10.1186/s12967-025-06209-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 02/05/2025] [Indexed: 02/17/2025] Open
Abstract
Lung cancer (LC) remains the leading cause of cancer death globally. Recent reports have suggested that circulating microbial nucleic acids have potential as promising biomarkers for cancer liquid biopsies. However, circulating microbial profiles and their potential clinical value in LC patients remained unexplored. In this study, plasma samples from 76 LC patients, 9 liver cancer patients, 11 pancreatic cancer patients, and 53 healthy controls (HCs) were collected and underwent metagenomic analyses by whole genome sequencing. The composition and relative abundance of the microbial profiles were significantly different between the LC patients and HCs. A distinct plasma-based microbial profile was observed in LC patients. By differential analysis using MaAslin, 40 significant species between LC patients and HCs were identified. Five species were selected as optimal circulating microbial biomarkers for LC. The constructed classifier based on these five species showed an AUC of 0.9592, 0.9131, and 0.8077 in the discovery, validation, and additional validation cohorts, respectively. Furthermore, metagenomic profiles of 25 lung tumor tissue and plasma paired samples were analyzed and compared. The microbial diversity was significantly increased in plasma compared with the tumor tissue. Among the 13 shared core microbial species, 10 had no difference between the tumor tissue and paired plasma. In conclusion, circulating microbial nucleic acids in the plasma have potential as biomarkers for LC liquid biopsies. The microbiome in the tumor tissue was one of the possible sources of circulating microbial nucleic acids.
Collapse
Affiliation(s)
- Huijuan Chen
- Beijing CapitalBio Medlab Co. Ltd, Beijing, 100176, China
- South China Hospital of Shenzhen University, Shenzhen, 518111, China
| | - XueNa Yao
- Beijing CapitalBio Medlab Co. Ltd, Beijing, 100176, China
| | - Chunyan Yang
- Beijing SeqTide Gene Technology Co. Ltd, Beijing, 10017, China
- Beijing Best HealthCare Medical Technology Co. Ltd, Beijing, 100176, China
- School of Life Science and Technology, Harbin Institute of Technology, 92 West Dazhi Street, Harbin, 150001, China
| | - Yiran Zhang
- Beijing SeqTide Gene Technology Co. Ltd, Beijing, 10017, China
| | - Henan Dong
- Beijing Best HealthCare Medical Technology Co. Ltd, Beijing, 100176, China
| | - Jincheng Zhai
- Beijing Best HealthCare Medical Technology Co. Ltd, Beijing, 100176, China
| | - Dongjie Fan
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China.
| | - Qiming Zhou
- School of Life Science and Technology, Harbin Institute of Technology, 92 West Dazhi Street, Harbin, 150001, China.
| |
Collapse
|
5
|
Saeed H, Díaz LA, Gil-Gómez A, Burton J, Bajaj JS, Romero-Gomez M, Arrese M, Arab JP, Khan MQ. Microbiome-centered therapies for the management of metabolic dysfunction-associated steatotic liver disease. Clin Mol Hepatol 2025; 31:S94-S111. [PMID: 39604327 PMCID: PMC11925441 DOI: 10.3350/cmh.2024.0811] [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: 09/13/2024] [Accepted: 11/20/2024] [Indexed: 11/29/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a significant global health issue, affecting over 30% of the population worldwide due to the rising prevalence of metabolic risk factors such as obesity and type 2 diabetes mellitus. This spectrum of liver disease ranges from isolated steatosis to more severe forms such as steatohepatitis, fibrosis, and cirrhosis. Recent studies highlight the role of gut microbiota in MASLD pathogenesis, showing that dysbiosis significantly impacts metabolic health and the progression of liver disease. This review critically evaluates current microbiome-centered therapies in MASLD management, including prebiotics, probiotics, synbiotics, fecal microbiota transplantation, and emerging therapies such as engineered bacteria and bacteriophage therapy. We explore the scientific rationale, clinical evidence, and potential mechanisms by which these interventions influence MASLD. The gut-liver axis is crucial in MASLD, with notable changes in microbiome composition linked to disease progression. For instance, specific microbial profiles and reduced alpha diversity are associated with MASLD severity. Therapeutic strategies targeting the microbiome could modulate disease progression by improving gut permeability, reducing endotoxin-producing bacteria, and altering bile acid metabolism. Although promising, these therapies require further research to fully understand their mechanisms and optimize their efficacy. This review integrates findings from clinical trials and experimental studies, providing a comprehensive overview of microbiome-centered therapies' potential in managing MASLD. Future research should focus on personalized strategies, utilizing microbiome features, blood metabolites, and customized dietary interventions to enhance the effectiveness of these therapies.
Collapse
Affiliation(s)
- Huma Saeed
- Division of Infectious Diseases, Department of Medicine, University of Western Ontario, London, ON, Canada
| | - Luis Antonio Díaz
- MASLD Research Center, Division of Gastroenterology and Hepatology, University of California San Diego, San Diego, CA, USA
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Antonio Gil-Gómez
- SeLiver Group, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - Jeremy Burton
- Department of Microbiology & Immunology, Western University, London, ON, Canada
| | - Jasmohan S Bajaj
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Manuel Romero-Gomez
- SeLiver Group, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
- Departamento de Medicina, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
- UCM Digestive diseases, Hospital Universitario Virgen del Rocío, Sevilla, Spain
| | - Marco Arrese
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan Pablo Arab
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Mohammad Qasim Khan
- Division of Gastroenterology, Department of Medicine, University of Western Ontario, London, ON, Canada
- Department of Epidemiology and Biostatistics, University of Western Ontario, London, ON, Canada
| |
Collapse
|
6
|
Pasta A, Formisano E, Calabrese F, Marabotto E, Furnari M, Bodini G, Torres MCP, Pisciotta L, Giannini EG, Zentilin P. From Dysbiosis to Hepatic Inflammation: A Narrative Review on the Diet-Microbiota-Liver Axis in Steatotic Liver Disease. Microorganisms 2025; 13:241. [PMID: 40005608 PMCID: PMC11857840 DOI: 10.3390/microorganisms13020241] [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: 12/31/2024] [Revised: 01/16/2025] [Accepted: 01/21/2025] [Indexed: 02/27/2025] Open
Abstract
The gut microbiota has emerged as a critical player in metabolic and liver health, with its influence extending to the pathogenesis and progression of steatotic liver diseases. This review delves into the gut-liver axis, a dynamic communication network linking the gut microbiome and liver through metabolic, immunological, and inflammatory pathways. Dysbiosis, characterized by altered microbial composition, contributes significantly to the development of hepatic steatosis, inflammation, and fibrosis via mechanisms such as gut barrier dysfunction, microbial metabolite production, and systemic inflammation. Dietary patterns, including the Mediterranean diet, are highlighted for their role in modulating the gut microbiota, improving gut-liver axis integrity, and attenuating liver injury. Additionally, emerging microbiota-based interventions, such as fecal microbiota transplantation and bacteriophage therapy, show promise as therapeutic strategies for steatotic liver disease. However, challenges such as population heterogeneity, methodological variability, and knowledge gaps hinder the translational application of current findings. Addressing these barriers through standardized approaches and integrative research will pave the way for microbiota-targeted therapies to mitigate the global burden of steatotic liver disease.
Collapse
Affiliation(s)
- Andrea Pasta
- Gastroenterology Unit, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy; (A.P.); (F.C.); (E.M.); (M.F.); (G.B.); (M.C.P.T.); (E.G.G.)
| | - Elena Formisano
- Dietetics and Clinical Nutrition Unit, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy; (E.F.); (L.P.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Francesco Calabrese
- Gastroenterology Unit, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy; (A.P.); (F.C.); (E.M.); (M.F.); (G.B.); (M.C.P.T.); (E.G.G.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Elisa Marabotto
- Gastroenterology Unit, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy; (A.P.); (F.C.); (E.M.); (M.F.); (G.B.); (M.C.P.T.); (E.G.G.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Manuele Furnari
- Gastroenterology Unit, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy; (A.P.); (F.C.); (E.M.); (M.F.); (G.B.); (M.C.P.T.); (E.G.G.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Giorgia Bodini
- Gastroenterology Unit, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy; (A.P.); (F.C.); (E.M.); (M.F.); (G.B.); (M.C.P.T.); (E.G.G.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Maria Corina Plaz Torres
- Gastroenterology Unit, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy; (A.P.); (F.C.); (E.M.); (M.F.); (G.B.); (M.C.P.T.); (E.G.G.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Livia Pisciotta
- Dietetics and Clinical Nutrition Unit, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy; (E.F.); (L.P.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Edoardo Giovanni Giannini
- Gastroenterology Unit, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy; (A.P.); (F.C.); (E.M.); (M.F.); (G.B.); (M.C.P.T.); (E.G.G.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Patrizia Zentilin
- Gastroenterology Unit, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy; (A.P.); (F.C.); (E.M.); (M.F.); (G.B.); (M.C.P.T.); (E.G.G.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| |
Collapse
|
7
|
Chen WY, Zhang JH, Chen LL, Byrne CD, Targher G, Luo L, Ni Y, Zheng MH, Sun DQ. Bioactive metabolites: A clue to the link between MASLD and CKD? Clin Mol Hepatol 2025; 31:56-73. [PMID: 39428978 PMCID: PMC11791555 DOI: 10.3350/cmh.2024.0782] [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: 09/09/2024] [Revised: 10/15/2024] [Accepted: 10/18/2024] [Indexed: 10/22/2024] Open
Abstract
Metabolites produced as intermediaries or end-products of microbial metabolism provide crucial signals for health and diseases, such as metabolic dysfunction-associated steatotic liver disease (MASLD). These metabolites include products of the bacterial metabolism of dietary substrates, modification of host molecules (such as bile acids [BAs], trimethylamine-N-oxide, and short-chain fatty acids), or products directly derived from bacteria. Recent studies have provided new insights into the association between MASLD and the risk of developing chronic kidney disease (CKD). Furthermore, alterations in microbiota composition and metabolite profiles, notably altered BAs, have been described in studies investigating the association between MASLD and the risk of CKD. This narrative review discusses alterations of specific classes of metabolites, BAs, fructose, vitamin D, and microbiota composition that may be implicated in the link between MASLD and CKD.
Collapse
Affiliation(s)
- Wen-Ying Chen
- MAFLD Research Center, Department of Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jia-Hui Zhang
- Department of Pediatric Laboratory, Affiliated Children’s Hospital of Jiangnan University, Wuxi Children’s Hospital, Wuxi, Jiangsu, China
| | - Li-Li Chen
- MAFLD Research Center, Department of Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Christopher D. Byrne
- Southampton National Institute for Health and Care Research Biomedical Research Centre, University Hospital Southampton and University of Southampton, Southampton General Hospital, Southampton, UK
| | - Giovanni Targher
- Department of Medicine, University of Verona, Verona, Italy
- Metabolic Diseases Research Unit, IRCCS Sacro Cuore - Don Calabria Hospital, Negrar di Valpolicella, Italy
| | - Liang Luo
- Intensive Care Medicine, Jiangnan University Medical Center, Wuxi, China
| | - Yan Ni
- Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Ming-Hua Zheng
- MAFLD Research Center, Department of Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Institute of Hepatology, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Diagnosis and Treatment for the Development of Chronic Liver Disease in Zhejiang Province, Wenzhou, China
| | - Dan-Qin Sun
- Urologic Nephrology Center, Jiangnan University Medical Center, Wuxi, China
- Affiliated Wuxi Clinical College of Nantong University, Wuxi, China
- Department of Nephrology, Wuxi No.2 People’s Hospital, Wuxi, China
| |
Collapse
|
8
|
Gillard J, Roumain M, Picalausa C, Thibaut MM, Clerbaux LA, Tailleux A, Staels B, Muccioli GG, Bindels LB, Leclercq IA. A gut microbiota-independent mechanism shapes the bile acid pool in mice with MASH. JHEP Rep 2024; 6:101148. [PMID: 39741697 PMCID: PMC11686050 DOI: 10.1016/j.jhepr.2024.101148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 06/02/2024] [Accepted: 06/13/2024] [Indexed: 01/03/2025] Open
Abstract
Background & Aims An imbalance between primary and secondary bile acids contributes to the development of metabolic dysfunction-associated steatohepatitis (MASH). The precise mechanisms underlying changes in the bile acid pool in MASH remain to be identified. As gut bacteria convert primary bile acids to secondary bile acids, we investigated the contribution of the gut microbiota and its metabolizing activities to bile acid alterations in MASH. Methods To disentangle the influence of MASH from environmental and dietary factors, high-fat diet fed foz/foz mice were compared with their high-fat diet fed wildtype littermates. We developed functional assays (stable isotope labeling and in vitro experiments) to extend the analyses beyond a mere study of gut microbiota composition (16S rRNA gene sequencing). Key findings were confirmed in C57BL/6J mice were fed a Western and high-fructose diet, as an independent mouse model of MASH. Results Although mice with MASH exhibited lower levels of secondary 7α-dehydroxylated bile acids (3.5-fold lower, p = 0.0008), the gut microbial composition was similar in mice with and without MASH. Similar gut microbial bile salt hydrolase and 7α-dehydroxylating activities could not explain the low levels of secondary 7α-dehydroxylated bile acids. Furthermore, the 7α-dehydroxylating activity was unaffected by Clostridium scindens administration in mice with a non-standardized gut microbiota. By exploring alternative mechanisms, we identified an increased bile acid 7α-rehydroxylation mediated by liver CYP2A12 and CYP2A22 enzymes (4.0-fold higher, p <0.0001), that reduces secondary 7α-dehydroxylated bile acid levels in MASH. Conclusions This study reveals a gut microbiota-independent mechanism that alters the level of secondary bile acids and contributes to the development of MASH in mice. Impact and implications Although changes in bile acid levels are implicated in the development of metabolic dysfunction-associated steatohepatitis (MASH), the precise mechanisms underpinning these alterations remain elusive. In this study, we investigated the mechanisms responsible for the changes in bile acid levels in mouse models of MASH. Our results support that neither the composition nor the metabolic activity of the gut microbiota can account for the alterations in the bile acid pool. Instead, we identified hepatic 7α-rehydroxylation of secondary bile acids as a gut microbiota-independent factor contributing to the reduced levels of secondary bile acids in mice with MASH. Further investigation is warranted to understand bile acid metabolism and its physiological implications in clinical MASH. Nonetheless, our findings hold promise for exploring novel therapeutic interventions for MASH.
Collapse
Affiliation(s)
- Justine Gillard
- Laboratory of Hepato-Gastroenterology, Institute of Experimental and Clinical Research, Université Catholique de Louvain, Brussels, Belgium
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Martin Roumain
- Bioanalysis and Pharmacology of Bioactive Lipids, Louvain Drug Research Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Corinne Picalausa
- Laboratory of Hepato-Gastroenterology, Institute of Experimental and Clinical Research, Université Catholique de Louvain, Brussels, Belgium
| | - Morgane M. Thibaut
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Laure-Alix Clerbaux
- Laboratory of Hepato-Gastroenterology, Institute of Experimental and Clinical Research, Université Catholique de Louvain, Brussels, Belgium
| | - Anne Tailleux
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Bart Staels
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Giulio G. Muccioli
- Bioanalysis and Pharmacology of Bioactive Lipids, Louvain Drug Research Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Laure B. Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université Catholique de Louvain, Brussels, Belgium
- Welbio department, WEL Research Institute, Wavre, Belgium
| | - Isabelle A. Leclercq
- Laboratory of Hepato-Gastroenterology, Institute of Experimental and Clinical Research, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
9
|
Jo HE, Khom S, Lee S, Cho SH, Park SY, You GR, Kim H, Kim NI, Jeong JH, Yoon JH, Yun M. Stage dependent microbial dynamics in hepatocellular carcinoma and adjacent normal liver tissues. Sci Rep 2024; 14:26092. [PMID: 39478014 PMCID: PMC11525880 DOI: 10.1038/s41598-024-77260-6] [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: 06/19/2024] [Accepted: 10/21/2024] [Indexed: 11/02/2024] Open
Abstract
The interactive pathway of the gut-liver axis underscores the significance of microbiome modulation in the pathogenesis and progression of various liver diseases, including hepatocellular carcinoma (HCC). This study aims to investigate the disparities in the composition and functionality of the hepatic microbiota between tumor tissues and adjacent normal liver tissues, and their implications in the etiology of HCC. We conducted a comparative analysis of the hepatic microbiome between adjacent normal liver tissues and tumor tissues from HCC patients. Samples were categorized according to the modified Union for International Cancer Control (mUICC) staging system into Non-tumor, mUICC stage I, mUICC stage II, and mUICC stage III groups. Microbial richness and community composition were analyzed, and phylogenetic profiles were examined to identify significantly altered microbial taxa among the groups. Predicted metabolic pathways were analyzed using PICRUSt2. Our analysis did not reveal significant differences in microbial richness and community composition with the development of HCC. However, phylogenetic profiling identified significantly altered microbial taxa among the groups. Sphingobium, known for degrading polychlorinated biphenyls (PCBs), exhibited a significantly negative correlation with clinical indices in HCC patients. Conversely, Sphingomonas, a gut bacterium associated with various liver diseases, showed a positive correlation. Predicted metabolic pathways suggested a correlation between atrazine degradation and valine, leucine, and isoleucine biosynthesis with mUICC stage and tumor size. Our results underscore the critical link between hepatic microbial composition and function and the HCC tumor stage, suggesting a potentially pivotal role in the development of HCC. These findings highlight the importance of targeting the hepatic microbiome for therapeutic strategies in HCC.
Collapse
Affiliation(s)
- Hee Eun Jo
- Technology Innovation Research Division, World Institute of Kimchi, Gwangju, 61755, Republic of Korea
- Department of Biomedical Sciences and Department of Microbiology, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
| | - Sophallika Khom
- Department of Gastroenterology and Hepatology, Chonnam National University Hospital and Medical School, Gwangju, 61469, Republic of Korea
| | - Sumi Lee
- Department of Gastroenterology and Hepatology, Chonnam National University Hospital and Medical School, Gwangju, 61469, Republic of Korea
| | - Su Hyeon Cho
- Department of Gastroenterology and Hepatology, Chonnam National University Hospital and Medical School, Gwangju, 61469, Republic of Korea
| | - Shin Young Park
- Department of Gastroenterology and Hepatology, Chonnam National University Hospital and Medical School, Gwangju, 61469, Republic of Korea
| | - Ga Ram You
- Department of Gastroenterology and Hepatology, Hwasun Chonnam National University Hospital and Medical School, Jeonnam, 58128, Republic of Korea
| | - Hyosin Kim
- Department of Surgery, Chonnam National University Hospital and Medical School, Gwangju, 61469, Republic of Korea
| | - Nah Ihm Kim
- Deparment of Pathology, Chonnam National University Hospital and Medical School, Gwangju, 61469, Republic of Korea
| | - Jae-Ho Jeong
- Department of Biomedical Sciences and Department of Microbiology, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea.
| | - Jae Hyun Yoon
- Department of Gastroenterology and Hepatology, Chonnam National University Hospital and Medical School, Gwangju, 61469, Republic of Korea.
| | - Misun Yun
- Technology Innovation Research Division, World Institute of Kimchi, Gwangju, 61755, Republic of Korea.
| |
Collapse
|
10
|
Koliarakis I, Lagkouvardos I, Vogiatzoglou K, Tsamandouras I, Intze E, Messaritakis I, Souglakos J, Tsiaoussis J. Circulating Bacterial DNA in Colorectal Cancer Patients: The Potential Role of Fusobacterium nucleatum. Int J Mol Sci 2024; 25:9025. [PMID: 39201711 PMCID: PMC11354820 DOI: 10.3390/ijms25169025] [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: 07/15/2024] [Revised: 08/12/2024] [Accepted: 08/16/2024] [Indexed: 09/03/2024] Open
Abstract
Intestinal dysbiosis is a major contributor to colorectal cancer (CRC) development, leading to bacterial translocation into the bloodstream. This study aimed to evaluate the presence of circulated bacterial DNA (cbDNA) in CRC patients (n = 75) and healthy individuals (n = 25). DNA extracted from peripheral blood was analyzed using PCR, with specific primers targeting 16S rRNA, Escherichia coli (E. coli), and Fusobacterium nucleatum (F. nucleatum). High 16S rRNA and E. coli detections were observed in all patients and controls. Only the detection of F. nucleatum was significantly higher in metastatic non-excised CRC, compared to controls (p < 0.001), non-metastatic excised CRC (p = 0.023), and metastatic excised CRC (p = 0.023). This effect was mainly attributed to the presence of the primary tumor (p = 0.006) but not the presence of distant metastases (p = 0.217). The association of cbDNA with other clinical parameters or co-morbidities was also evaluated, revealing a higher detection of E. coli in CRC patients with diabetes (p = 0.004). These results highlighted the importance of bacterial translocation in CRC patients and the potential role of F. nucleatum as an intratumoral oncomicrobe in CRC.
Collapse
Affiliation(s)
- Ioannis Koliarakis
- Department of Anatomy, School of Medicine, University of Crete, 70013 Heraklion, Greece;
| | - Ilias Lagkouvardos
- Department of Clinical Microbiology, School of Medicine, University of Crete, 70013 Heraklion, Greece; (I.L.); (E.I.)
| | - Konstantinos Vogiatzoglou
- Laboratory of Translational Oncology, Medical School, University of Crete, 70013 Heraklion, Greece; (K.V.); (I.M.); (J.S.)
| | - Ioannis Tsamandouras
- Department of Otorhinolaryngology—Head and Neck Surgery, University General Hospital of Heraklion, 71110 Heraklion, Greece;
| | - Evangelia Intze
- Department of Clinical Microbiology, School of Medicine, University of Crete, 70013 Heraklion, Greece; (I.L.); (E.I.)
| | - Ippokratis Messaritakis
- Laboratory of Translational Oncology, Medical School, University of Crete, 70013 Heraklion, Greece; (K.V.); (I.M.); (J.S.)
- Department of Microbiology, German Oncology Center, Yiannoukas Labs LTD, Bioiatriki Group, Limassol 4108, Cyprus
| | - John Souglakos
- Laboratory of Translational Oncology, Medical School, University of Crete, 70013 Heraklion, Greece; (K.V.); (I.M.); (J.S.)
- Department of Medical Oncology, University Hospital of Heraklion, 71110 Heraklion, Greece
| | - John Tsiaoussis
- Department of Anatomy, School of Medicine, University of Crete, 70013 Heraklion, Greece;
| |
Collapse
|
11
|
Jingushi K, Kawashima A, Tanikawa S, Saito T, Yamamoto A, Uemura T, Sassi N, Ishizuya Y, Yamamoto Y, Kato T, Hatano K, Hase H, Nonomura N, Tsujikawa K. Cutibacterium acnes-derived extracellular vesicles promote tumor growth in renal cell carcinoma. Cancer Sci 2024; 115:2578-2587. [PMID: 38682309 PMCID: PMC11309925 DOI: 10.1111/cas.16202] [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: 02/07/2024] [Revised: 04/08/2024] [Accepted: 04/17/2024] [Indexed: 05/01/2024] Open
Abstract
Bacterial flora are present in various parts of the human body, including the intestine, and are thought to be involved in the etiology of various diseases such as multiple sclerosis, intestinal diseases, cancer, and uterine diseases. In recent years, the presence of bacterial 16S rRNA genes has been revealed in blood, which was previously thought to be a sterile environment, and characteristic blood microbiomes have been detected in various diseases. However, the mechanism and the origin of the bacterial information are unknown. In this study, we performed 16S rRNA metagenomic analysis of bacterial DNA in serum extracellular vesicles from five healthy donors and seven patients with renal cell carcinoma and detected Cutibacterium acnes DNA as a characteristic bacterial DNA in the serum extracellular vesicles of patients with renal cell carcinoma. In addition, C. acnes DNA was significantly reduced in postoperative serum extracellular vesicles from patients with renal cell carcinoma compared with that in preoperative serum extracellular vesicles from these patients and was also detected in tumor tissue and extracellular vesicles from tumor tissue-associated microbiota, suggesting an association between C. acnes extracellular vesicles and renal cell carcinoma. C. acnes extracellular vesicles were taken up by renal carcinoma cells to enhance their proliferative potential. C. acnes extracellular vesicles also exhibited tumor-promoting activity in a mouse model of renal cancer allografts with enhanced angiogenesis. These results suggest that extracellular vesicles released by C. acnes localized in renal cell carcinoma tissues act in a tumor-promoting manner.
Collapse
Affiliation(s)
- Kentaro Jingushi
- Laboratory of Molecular and Cellular PhysiologyGraduate School of Pharmaceutical Sciences, Osaka UniversitySuitaOsakaJapan
| | - Atsunari Kawashima
- Department of UrologyGraduate School of Medicine, Osaka UniversitySuitaJapan
| | - Sayaka Tanikawa
- Laboratory of Molecular and Cellular PhysiologyGraduate School of Pharmaceutical Sciences, Osaka UniversitySuitaOsakaJapan
| | - Takuro Saito
- Department of SurgeryGraduate School of Medicine, Osaka UniversitySuitaOsakaJapan
- Department of Clinical Research in Tumor ImmunologyGraduate School of Medicine, Osaka UniversitySuitaOsakaJapan
| | - Akinaru Yamamoto
- Department of UrologyGraduate School of Medicine, Osaka UniversitySuitaJapan
| | - Toshihiro Uemura
- Department of UrologyGraduate School of Medicine, Osaka UniversitySuitaJapan
| | - Nesrine Sassi
- Department of UrologyGraduate School of Medicine, Osaka UniversitySuitaJapan
| | - Yu Ishizuya
- Department of UrologyGraduate School of Medicine, Osaka UniversitySuitaJapan
| | - Yoshiyuki Yamamoto
- Department of UrologyGraduate School of Medicine, Osaka UniversitySuitaJapan
| | - Taigo Kato
- Department of UrologyGraduate School of Medicine, Osaka UniversitySuitaJapan
| | - Koji Hatano
- Department of UrologyGraduate School of Medicine, Osaka UniversitySuitaJapan
| | - Hiroaki Hase
- Laboratory of Molecular and Cellular PhysiologyGraduate School of Pharmaceutical Sciences, Osaka UniversitySuitaOsakaJapan
| | - Norio Nonomura
- Department of UrologyGraduate School of Medicine, Osaka UniversitySuitaJapan
| | - Kazutake Tsujikawa
- Laboratory of Molecular and Cellular PhysiologyGraduate School of Pharmaceutical Sciences, Osaka UniversitySuitaOsakaJapan
| |
Collapse
|
12
|
Xu T, Lyu L, Zheng J, Li L. Advances in omics-based biomarker discovery for biliary tract malignancy Diagnosis:A narrative review. Mol Cell Probes 2024; 76:101970. [PMID: 38964426 DOI: 10.1016/j.mcp.2024.101970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 06/30/2024] [Accepted: 07/01/2024] [Indexed: 07/06/2024]
Abstract
Biliary tract neoplasms, which originate from the intrahepatic or extrahepatic biliary epithelium, are relatively rare but diagnostically challenging types of tumours, and their morbidity and mortality have increased in recent years. Due to ineffective early diagnostic methods, once detected, patients are in an advanced stage with a poor prognosis and few treatment options. With the development of omics technologies, the associations between microorganisms, bile acid and salts, noncoding RNAs and biliary tract malignancies have been gradually revealed, providing new methods for the discovery of diagnostic biomarkers. Here, we review the research advances in microbiomics, transcriptomics, metabolomics, and proteomics in the discovery of diagnostic biomarkers for biliary tract malignancies.
Collapse
Affiliation(s)
- Tao Xu
- Department of Gastroenterology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 102200, China.
| | - Lingna Lyu
- Department of Hepatology and Gastroenterology, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China.
| | - Junfu Zheng
- Department of Gastroenterology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 102200, China.
| | - Lei Li
- Department of Gastroenterology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 102200, China.
| |
Collapse
|
13
|
Yuan X, Yang X, Xu Z, Li J, Sun C, Chen R, Wei H, Chen L, Du H, Li G, Yang Y, Chen X, Cui L, Fu J, Wu J, Chen Z, Fang X, Su Z, Zhang M, Wu J, Chen X, Zhou J, Luo Y, Zhang L, Wang R, Luo F. The profile of blood microbiome in new-onset type 1 diabetes children. iScience 2024; 27:110252. [PMID: 39027370 PMCID: PMC11255850 DOI: 10.1016/j.isci.2024.110252] [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: 10/24/2023] [Revised: 02/09/2024] [Accepted: 06/10/2024] [Indexed: 07/20/2024] Open
Abstract
Blood microbiome signatures in patients with type 1 diabetes (T1D) remain unclear. We profile blood microbiome using 16S rRNA gene sequencing in 77 controls and 64 children with new-onset T1D, and compared it with the gut and oral microbiomes. The blood microbiome of patients with T1D is characterized by increased diversity and perturbed microbial features, with a significant increase in potentially pathogenic bacteria compared with controls. Thirty-six representative genera of blood microbiome were identified by random forest analysis, providing strong discriminatory power for T1D with an AUC of 0.82. PICRUSt analysis suggested that bacteria capable of inducing inflammation were more likely to enter the bloodstream in T1D. The overlap of the gut and oral microbiome with the blood microbiome implied potential translocation of bacteria from the gut and oral cavity to the bloodstream. Our study raised the necessity of further mechanistic investigations into the roles of blood microbiome in T1D.
Collapse
Affiliation(s)
- Xiaoxiao Yuan
- Department of Pediatric Endocrinology and Inherited Metabolic Diseases, Children’s Hospital of Fudan University, Shanghai 201102, China
| | - Xin Yang
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 200120, China
- Department of Food Science and Technology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
- Section of Endocrinology, Internal Medicine, School of Medicine, Yale University, New Haven, CT 06511, United States
| | - Zhenran Xu
- Department of Pediatric Endocrinology and Inherited Metabolic Diseases, Children’s Hospital of Fudan University, Shanghai 201102, China
| | - Jie Li
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 200120, China
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong 999077, China
| | - ChengJun Sun
- Department of Pediatric Endocrinology and Inherited Metabolic Diseases, Children’s Hospital of Fudan University, Shanghai 201102, China
| | - Ruimin Chen
- Fuzhou Children’s Hospital of Fujian Medical University, Fuzhou 350000, China
| | - Haiyan Wei
- Department of Endocrinology and Inherited Metabolic, Children’s Hospital Affiliated to Zhengzhou University, Zhengzhou 450000, China
| | - Linqi Chen
- Children’s Hospital of Soochow University, Suzhou 215000, China
| | - Hongwei Du
- The First Hospital of Jilin University, Jilin 130000, China
| | - Guimei Li
- Department of Pediatric Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Yu Yang
- The Affiliated Children’s Hospital of Nanchang University, Nanchang 330006, China
| | - Xiaojuan Chen
- Department of Endocrinology, Genetics and Metabolism, The Children’s Hospital of Shanxi Province, Taiyuan 030013, China
| | - Lanwei Cui
- The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Junfen Fu
- Department of Endocrinology, Children’s Hospital, Zhejiang University School of Medicine, Hangzhou 310005, China
| | - Jin Wu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Zhihong Chen
- Department of Neuroendocrinology Pediatrics, Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Xin Fang
- Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Zhe Su
- Shenzhen Children’s Hospital, Shenzhen 518038, China
| | - Miaoying Zhang
- Department of Pediatric Endocrinology and Inherited Metabolic Diseases, Children’s Hospital of Fudan University, Shanghai 201102, China
| | - Jing Wu
- Department of Pediatric Endocrinology and Inherited Metabolic Diseases, Children’s Hospital of Fudan University, Shanghai 201102, China
| | - Xin Chen
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 200120, China
| | - Jiawei Zhou
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 200120, China
| | - Yue Luo
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Lei Zhang
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 200120, China
| | - Ruirui Wang
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 200120, China
| | - Feihong Luo
- Department of Pediatric Endocrinology and Inherited Metabolic Diseases, Children’s Hospital of Fudan University, Shanghai 201102, China
| |
Collapse
|
14
|
Santacroce L, Charitos IA, Colella M, Palmirotta R, Jirillo E. Blood Microbiota and Its Products: Mechanisms of Interference with Host Cells and Clinical Outcomes. Hematol Rep 2024; 16:440-453. [PMID: 39051416 PMCID: PMC11270377 DOI: 10.3390/hematolrep16030043] [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: 04/11/2024] [Revised: 06/01/2024] [Accepted: 06/18/2024] [Indexed: 07/27/2024] Open
Abstract
In healthy conditions, blood was considered a sterile environment until the development of new analytical approaches that allowed for the detection of circulating bacterial ribosomal DNA. Currently, debate exists on the origin of the blood microbiota. According to advanced research using dark field microscopy, fluorescent in situ hybridization, flow cytometry, and electron microscopy, so-called microbiota have been detected in the blood. Conversely, others have reported no evidence of a common blood microbiota. Then, it was hypothesized that blood microbiota may derive from distant sites, e.g., the gut or external contamination of blood samples. Alteration of the blood microbiota's equilibrium may lead to dysbiosis and, in certain cases, disease. Cardiovascular, respiratory, hepatic, kidney, neoplastic, and immune diseases have been associated with the presence of Gram-positive and Gram-negative bacteria and/or their products in the blood. For instance, lipopolysaccharides (LPSs) and endotoxins may contribute to tissue damage, fueling chronic inflammation. Blood bacteria can interact with immune cells, especially with monocytes that engulf microorganisms and T lymphocytes via spontaneous binding to their membranes. Moreover, LPSs, extracellular vesicles, and outer membrane vesicles interact with red blood cells and immune cells, reaching distant organs. This review aims to describe the composition of blood microbiota in healthy individuals and those with disease conditions. Furthermore, special emphasis is placed on the interaction of blood microbiota with host cells to better understand disease mechanisms.
Collapse
Affiliation(s)
- Luigi Santacroce
- Section of Microbiology and Virology, Interdisciplinary Department of Medicine, School of Medicine, University of Bari ‘Aldo Moro’, 70124 Bari, Italy (R.P.); (E.J.)
| | - Ioannis Alexandros Charitos
- Istituti Clinici Scientifici Maugeri IRCCS, Pneumology and Respiratory Rehabilitation Unit, Institute of Bari, 70124 Bari, Italy;
| | - Marica Colella
- Section of Microbiology and Virology, Interdisciplinary Department of Medicine, School of Medicine, University of Bari ‘Aldo Moro’, 70124 Bari, Italy (R.P.); (E.J.)
- Doctoral School, eCampus University, 22060 Novedrate, Italy
| | - Raffaele Palmirotta
- Section of Microbiology and Virology, Interdisciplinary Department of Medicine, School of Medicine, University of Bari ‘Aldo Moro’, 70124 Bari, Italy (R.P.); (E.J.)
| | - Emilio Jirillo
- Section of Microbiology and Virology, Interdisciplinary Department of Medicine, School of Medicine, University of Bari ‘Aldo Moro’, 70124 Bari, Italy (R.P.); (E.J.)
| |
Collapse
|
15
|
Kotlyarov S. Importance of the gut microbiota in the gut-liver axis in normal and liver disease. World J Hepatol 2024; 16:878-882. [PMID: 38948437 PMCID: PMC11212653 DOI: 10.4254/wjh.v16.i6.878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 05/01/2024] [Accepted: 05/17/2024] [Indexed: 06/20/2024] Open
Abstract
The gut microbiota is of growing interest to clinicians and researchers. This is because there is a growing understanding that the gut microbiota performs many different functions, including involvement in metabolic and immune processes that are systemic in nature. The liver, with its important role in detoxifying and metabolizing products from the gut, is at the forefront of interactions with the gut microbiota. Many details of these interactions are not yet known to clinicians and researchers, but there is growing evidence that normal gut microbiota function is important for liver health. At the same time, factors affecting the gut microbiota, including nutrition or medications, may also have an effect through the gut-liver axis.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University, Ryazan 390026, Russia.
| |
Collapse
|
16
|
Cho EJ, Kim B, Yu SJ, Hong SK, Choi Y, Yi NJ, Lee KW, Suh KS, Yoon JH, Park T. Urinary microbiome-based metagenomic signature for the noninvasive diagnosis of hepatocellular carcinoma. Br J Cancer 2024; 130:970-975. [PMID: 38278977 PMCID: PMC10951239 DOI: 10.1038/s41416-024-02582-1] [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: 03/17/2023] [Revised: 01/08/2024] [Accepted: 01/10/2024] [Indexed: 01/28/2024] Open
Abstract
BACKGROUND Gut microbial dysbiosis is implicated in chronic liver disease and hepatocellular carcinoma (HCC), but the role of microbiomes from various body sites remains unexplored. We assessed disease-specific alterations in the urinary microbiome in HCC patients, investigating their potential as diagnostic biomarkers. METHODS We performed cross-sectional analyses of urine samples from 471 HCC patients and 397 healthy controls and validated the results in an independent cohort of 164 HCC patients and 164 healthy controls. Urinary microbiomes were analyzed by 16S rRNA gene sequencing. A microbial marker-based model distinguishing HCC from controls was built based on logistic regression, and its performance was tested. RESULTS Microbial diversity was significantly reduced in the HCC patients compared with the controls. There were significant differences in the abundances of various bacteria correlated with HCC, thus defining a urinary microbiome-derived signature of HCC. We developed nine HCC-associated genera-based models with robust diagnostic accuracy (area under the curve [AUC], 0.89; balanced accuracy, 81.2%). In the validation, this model detected HCC with an AUC of 0.94 and an accuracy of 88.4%. CONCLUSIONS The urinary microbiome might be a potential biomarker for the detection of HCC. Further clinical testing and validation of these results are needed in prospective studies.
Collapse
Affiliation(s)
- Eun Ju Cho
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Boram Kim
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, 08826, Korea
| | - Su Jong Yu
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Suk Kyun Hong
- Department of Surgery, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - YoungRok Choi
- Department of Surgery, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Nam-Joon Yi
- Department of Surgery, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Kwang-Woong Lee
- Department of Surgery, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Kyung-Suk Suh
- Department of Surgery, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Jung-Hwan Yoon
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, 03080, Korea.
| | - Taesung Park
- Department of Statistics, Seoul National University, Seoul, 08826, Korea.
| |
Collapse
|
17
|
Lu J, Zeng Y, Zhong H, Guo W, Zhang Y, Mai W, Qin Y, Su X, Zhang B, Wu W, Zhu Y, Huang Q, Ye Y. Dual-Stimuli-Responsive Gut Microbiota-Targeting Nitidine Chloride-CS/PT-NPs Improved Metabolic Status in NAFLD. Int J Nanomedicine 2024; 19:2409-2428. [PMID: 38476281 PMCID: PMC10929648 DOI: 10.2147/ijn.s452194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
Background and Purpose Nitidine chloride (NC) is a botanical drug renowned for its potent anti-inflammatory, antimalarial, and hepatocellular carcinoma-inhibiting properties; however, its limited solubility poses challenges to its development and application. To address this issue, we have devised a colon-targeted delivery system (NC-CS/PT-NPs) aimed at modulating the dysbiosis of the gut microbiota by augmenting the interaction between NC and the intestinal microbiota, thereby exerting an effect against nonalcoholic fatty liver disease. Methods The NC-CS/PT-NPs were synthesized using the ion gel method. Subsequently, the particle size distribution, morphology, drug loading efficiency, and release behavior of the NC-CS/PT-NPs were characterized. Furthermore, the impact of NC-CS/PT-NPs on non-alcoholic fatty liver disease (NAFLD) induced by a high-fat diet (HFD) in mice was investigated through serum biochemical analysis, ELISA, and histochemical staining. Additionally, the influence of NC-CS/PT-NPs on intestinal microbiota was analyzed using 16S rDNA gene sequencing. Results The nanoparticles prepared in this study have an average particle size of (255.9±5.10) nm, with an encapsulation rate of (72.83±2.13) % and a drug loading of (4.65±0.44) %. In vitro release experiments demonstrated that the cumulative release rate in the stomach and small intestine was lower than 22.0%, while it reached 66.75% in the colon. In vivo experiments conducted on HFD-induced NAFLD mice showed that treatment with NC-CS/PT-NPs inhibited weight gain, decreased serum aspartate aminotransferase (AST), Alanine aminotransferase (ALT) and lipid levels, improved liver and intestinal inflammation, and altered the diversity of gut microbiota in mice. Conclusion This study provides new evidence for the treatment of NAFLD through the regulation of gut microbiota using active ingredients from traditional Chinese medicine.
Collapse
Affiliation(s)
- Jianmei Lu
- Department of Pharmacy, Guangxi Medical University, Nanning, People’s Republic of China
- The Second Nanning People’s Hospital, Nanning, People’s Republic of China
| | - Yongzhu Zeng
- Department of Pharmacy, Guangxi Medical University, Nanning, People’s Republic of China
| | - Huashuai Zhong
- Department of Pharmacy, Guangxi Medical University, Nanning, People’s Republic of China
| | - Wei Guo
- Department of Pharmacy, Guangxi Medical University, Nanning, People’s Republic of China
| | - Yuyan Zhang
- Department of Pharmacy, Guangxi Medical University, Nanning, People’s Republic of China
| | - Wanting Mai
- The Second Affiliated Hospital of Guangxi Medical University, Nanning, People’s Republic of China
| | - Yucui Qin
- Maternity and Child Health Care of Guangxi Zhuang Autonomous Region, Nanning, People’s Republic of China
| | - Xiaodan Su
- The Second Affiliated Hospital of Guangxi Medical University, Nanning, People’s Republic of China
| | - Bo Zhang
- Scientific Research Center, Guilin Medical University, Guilin, People’s Republic of China
| | - Weisen Wu
- Department of Pharmacy, Guangxi Medical University, Nanning, People’s Republic of China
| | - Yu Zhu
- Department of Pharmacy, Guangxi Medical University, Nanning, People’s Republic of China
| | - Qiujie Huang
- Department of Pharmacy, Guangxi University of Traditional Chinese Medicine, Nanning, People’s Republic of China
| | - Yong Ye
- Department of Pharmacy, Guangxi Medical University, Nanning, People’s Republic of China
- Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation, Nanning, People’s Republic of China
- Guangxi Key Laboratory of Pharmaceutical Precision Detection and Screening, Nanning, People’s Republic of China
- Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, Nanning, People’s Republic of China
| |
Collapse
|
18
|
Yu J, Zhang L, Gao D, Wang J, Li Y, Sun N. Comparison of metagenomic next-generation sequencing and blood culture for diagnosis of bloodstream infections. Front Cell Infect Microbiol 2024; 14:1338861. [PMID: 38328669 PMCID: PMC10847245 DOI: 10.3389/fcimb.2024.1338861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 01/08/2024] [Indexed: 02/09/2024] Open
Abstract
Objectives This study aimed to evaluate the clinical performance of plasma cell-free DNA (cfDNA) next-generation sequencing (NGS) for pathogen detection in patients with sepsis. Methods A total of 43 pairs of blood and plasma samples form 33 blood culture-positive patients were used as testing samples in metagenomic NGS (mNGS) and NGS of 16S ribosomal RNA gene amplicons (16S rRNA NGS). The results of routine tests, including microbial culture, complete blood count, and biochemical tests, were collected from electronic medical records. Results Using blood as an mNGS testing sample, the proportion of host DNA was 99.9%, with only three bacteria and no fungi detected. When using plasma in mNGS, the proportion of host DNA was approximately 97%, with 84 bacteria and two fungi detected. Notably, 16S rRNA NGS detected 15 and 16 bacteria in 43 pairs of blood and plasma samples, respectively. Blood culture detected 49 bacteria (23 gram-negative bacilli and 26 gram-positive cocci) and four fungi, with 14 bacteria considered contaminants by clinical microbiologists. For all blood cultures, plasma cfDNA mNGS detected 78.26% (19/23) gram-negative rods, 17% (2/12) gram-positive cocci, and no fungi. Compared to blood cultures, the sensitivity and specificity of plasma cfDNA mNGS for detecting bacteria and fungi were 62.07% and 57.14%, respectively. Conclusion Compared to blood, plasma is more suitable for the detection of bloodstream infections using mNGS and is less affected by host DNA. The positive detection rate of plasma cfDNA mNGS for bloodstream infections caused by gram-negative bacteria was higher than that caused by gram-positive cocci.
Collapse
Affiliation(s)
- Juan Yu
- Department of Clinical Laboratory, Nanjing Lishui People’s Hospital, Nanjing, China
- Department of Clinical Laboratory Science, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Li Zhang
- Department of Clinical Laboratory, Nanjing Lishui People’s Hospital, Nanjing, China
| | - Deyu Gao
- Department of Clinical Laboratory Science, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jie Wang
- Clinical Medicine Research Center, The Affiliated Suqian First People’s Hospital of Nanjing Medical University, Suqian, China
| | - Yi Li
- Department of Clinical Laboratory Science, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Ning Sun
- Department of Clinical Laboratory Science, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
19
|
Lu S, Liang Y, Li L, Miao R, Liao S, Zou Y, Yang C, Ouyang D. Predicting potential microbe-disease associations based on auto-encoder and graph convolution network. BMC Bioinformatics 2023; 24:476. [PMID: 38097930 PMCID: PMC10722760 DOI: 10.1186/s12859-023-05611-7] [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: 12/11/2023] [Indexed: 12/17/2023] Open
Abstract
The increasing body of research has consistently demonstrated the intricate correlation between the human microbiome and human well-being. Microbes can impact the efficacy and toxicity of drugs through various pathways, as well as influence the occurrence and metastasis of tumors. In clinical practice, it is crucial to elucidate the association between microbes and diseases. Although traditional biological experiments accurately identify this association, they are time-consuming, expensive, and susceptible to experimental conditions. Consequently, conducting extensive biological experiments to screen potential microbe-disease associations becomes challenging. The computational methods can solve the above problems well, but the previous computational methods still have the problems of low utilization of node features and the prediction accuracy needs to be improved. To address this issue, we propose the DAEGCNDF model predicting potential associations between microbes and diseases. Our model calculates four similar features for each microbe and disease. These features are fused to obtain a comprehensive feature matrix representing microbes and diseases. Our model first uses the graph convolutional network module to extract low-rank features with graph information of microbes and diseases, and then uses a deep sparse Auto-Encoder to extract high-rank features of microbe-disease pairs, after which the low-rank and high-rank features are spliced to improve the utilization of node features. Finally, Deep Forest was used for microbe-disease potential relationship prediction. The experimental results show that combining low-rank and high-rank features helps to improve the model performance and Deep Forest has better classification performance than the baseline model.
Collapse
Affiliation(s)
- Shanghui Lu
- Faculty of Innovation Enginee, Macau University of Science and Technology, Avenida Wai Long, Taipa, 999078, Macao, Macao Special Administrative Region of China, China
- School of Mathematics and Physics, Hechi University, No. 42, Longjiang, Hechi, 546300, Guangxi, China
| | - Yong Liang
- Faculty of Innovation Enginee, Macau University of Science and Technology, Avenida Wai Long, Taipa, 999078, Macao, Macao Special Administrative Region of China, China.
- Peng Cheng Laboratory, Shenzhen, 518055, Guangdong, China.
| | - Le Li
- Faculty of Innovation Enginee, Macau University of Science and Technology, Avenida Wai Long, Taipa, 999078, Macao, Macao Special Administrative Region of China, China
| | - Rui Miao
- Basic Teaching Department, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, Guangdong, China
| | - Shuilin Liao
- Faculty of Innovation Enginee, Macau University of Science and Technology, Avenida Wai Long, Taipa, 999078, Macao, Macao Special Administrative Region of China, China
| | - Yongfu Zou
- School of Mathematics and Physics, Hechi University, No. 42, Longjiang, Hechi, 546300, Guangxi, China
| | - Chengjun Yang
- School of Artificial Intelligence and Manufacturing, Hechi University, No. 42, Longjiang, Hechi, 546300, Guangxi, China
| | - Dong Ouyang
- School of Biomedical Engineering, Guangdong Medical University, No. 1, Xincheng, Zhanjiang, 523808, Guangdong, China
| |
Collapse
|
20
|
Deng ZC, Yang JC, Huang YX, Zhao L, Zheng J, Xu QB, Guan L, Sun LH. Translocation of gut microbes to epididymal white adipose tissue drives lipid metabolism disorder under heat stress. SCIENCE CHINA. LIFE SCIENCES 2023; 66:2877-2895. [PMID: 37480471 DOI: 10.1007/s11427-022-2320-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 03/08/2023] [Indexed: 07/24/2023]
Abstract
Heat stress induces multi-organ damage and serious physiological dysfunction in mammals, and gut bacteria may translocate to extra-intestinal tissues under heat stress pathology. However, whether gut bacteria translocate to the key metabolic organs and impair function as a result of heat stress remains unknown. Using a heat stress-induced mouse model, heat stress inhibited epididymal white adipose tissue (eWAT) expansion and induced lipid metabolic disorder but did not damage other organs, such as the heart, liver, spleen, or muscle. Microbial profiling analysis revealed that heat stress shifted the bacterial community in the cecum and eWAT but not in the inguinal white adipose tissue, blood, heart, liver, spleen, or muscle. Notably, gut-vascular barrier function was impaired, and the levels of some bacteria, particularly Lactobacillus, were higher in the eWAT, as confirmed by catalyzed reporter deposition fluorescence in situ hybridization (CARD-FISH) staining when mice were under heat stress. Moreover, integrated multi-omics analysis showed that the eWAT microbiota was associated with host lipid metabolism, and the expression of genes involved in the lipid metabolism in eWAT was upregulated under heat stress. A follow-up microbial supplementation study after introducing Lactobacillus plantarum to heat-stressed mice revealed that the probiotic ameliorated heat stress-induced loss of eWAT and dyslipidemia and reduced gut bacterial translocation to the eWAT by improving gut barrier function. Overall, our findings suggest that gut bacteria, particularly Lactobacillus spp., play a crucial role in heat stress-induced lipid metabolism disorder and that there is therapeutic potential for using probiotics, such as Lactobacillus plantarum.
Collapse
Affiliation(s)
- Zhang-Chao Deng
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jia-Cheng Yang
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yu-Xuan Huang
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Ling Zhao
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jinshui Zheng
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Qing-Biao Xu
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Leluo Guan
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, T6G 2P5, Canada
| | - Lv-Hui Sun
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
21
|
Vasudevan D, Ramakrishnan A, Velmurugan G. Exploring the diversity of blood microbiome during liver diseases: Unveiling Novel diagnostic and therapeutic Avenues. Heliyon 2023; 9:e21662. [PMID: 37954280 PMCID: PMC10638009 DOI: 10.1016/j.heliyon.2023.e21662] [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: 06/06/2023] [Revised: 10/07/2023] [Accepted: 10/25/2023] [Indexed: 11/14/2023] Open
Abstract
Liver diseases are a group of major metabolic and immune or inflammation related diseases caused due to various reasons including infection, abnormalities in immune system, genetic defects, and lifestyle habits. However, the cause-effect relationship is not completely understood in liver disease. The role of microbiome, particularly, the role of gut and oral microbiome in liver diseases has been extensively studied in recent years. More interestingly, the presence of blood microbiome and tissue microbiome has been identified in many liver diseases. The translocation of microbes from the gut into the portal circulation has been attributed to be the major reason for the presence of blood microbial components and its clinical implications in liver disorders. Besides microbial translocation, Pathogen associated Molecular Patterns (PAMPs) derived from gut microbiota might also translocate. The presence of blood microbiome in liver disease has been reviewed earlier. However, the role of blood microbiome as a biomarker and therapeutic target in liver diseases has not been analysed earlier. In this review, we confabulate the origin and physiology of blood microbiome and blood microbial components in relation to the progression and pathogenesis of liver disease. In conclusion, we discuss the translational perspectives targeting the blood microbial components in the diagnosis and therapy of liver disease.
Collapse
Affiliation(s)
- Dinakaran Vasudevan
- Chemomicrobiomics Laboratory, Department of Biochemistry and Microbiology, KMCH Research Foundation, Coimbatore, 641014, Tamil Nadu, India
- Gut Microbiome Division, SKAN Research Trust, Bengaluru, 560034, Karnataka, India
| | - Arulraj Ramakrishnan
- Chemomicrobiomics Laboratory, Department of Biochemistry and Microbiology, KMCH Research Foundation, Coimbatore, 641014, Tamil Nadu, India
- Liver Unit, Kovai Medical Center and Hospital, Coimbatore, 641014, Tamil Nadu, India
| | - Ganesan Velmurugan
- Chemomicrobiomics Laboratory, Department of Biochemistry and Microbiology, KMCH Research Foundation, Coimbatore, 641014, Tamil Nadu, India
| |
Collapse
|
22
|
Wang S, Friedman SL. Found in translation-Fibrosis in metabolic dysfunction-associated steatohepatitis (MASH). Sci Transl Med 2023; 15:eadi0759. [PMID: 37792957 PMCID: PMC10671253 DOI: 10.1126/scitranslmed.adi0759] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 09/15/2023] [Indexed: 10/06/2023]
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) is a severe form of liver disease that poses a global health threat because of its potential to progress to advanced fibrosis, leading to cirrhosis and liver cancer. Recent advances in single-cell methodologies, refined disease models, and genetic and epigenetic insights have provided a nuanced understanding of MASH fibrogenesis, with substantial cellular heterogeneity in MASH livers providing potentially targetable cell-cell interactions and behavior. Unlike fibrogenesis, mechanisms underlying fibrosis regression in MASH are still inadequately understood, although antifibrotic targets have been recently identified. A refined antifibrotic treatment framework could lead to noninvasive assessment and targeted therapies that preserve hepatocellular function and restore the liver's architectural integrity.
Collapse
Affiliation(s)
- Shuang Wang
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Scott L. Friedman
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| |
Collapse
|
23
|
Tang Z, Yu S, Pan Y. The gut microbiome tango in the progression of chronic kidney disease and potential therapeutic strategies. J Transl Med 2023; 21:689. [PMID: 37789439 PMCID: PMC10546717 DOI: 10.1186/s12967-023-04455-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 08/19/2023] [Indexed: 10/05/2023] Open
Abstract
Chronic kidney disease (CKD) affects more than 10% population worldwide and becomes a huge burden to the world. Recent studies have revealed multifold interactions between CKD and gut microbiome and their pathophysiological implications. The gut microbiome disturbed by CKD results in the imbalanced composition and quantity of gut microbiota and subsequent changes in its metabolites and functions. Studies have shown that both the dysbiotic gut microbiota and its metabolites have negative impacts on the immune system and aggravate diseases in different ways. Herein, we give an overview of the currently known mechanisms of CKD progression and the alterations of the immune system. Particularly, we summarize the effects of uremic toxins on the immune system and review the roles of gut microbiota in promoting the development of different kidney diseases. Finally, we discuss the current sequencing technologies and novel therapies targeting the gut microbiome.
Collapse
Affiliation(s)
- Zijing Tang
- Department of Nephrology, Shanghai Ninth People's Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shiyan Yu
- Shanghai Institute of Precision Medicine, Shanghai Ninth People's Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yu Pan
- Department of Nephrology, Shanghai Ninth People's Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
24
|
Cai X, Peng Y, Gong Y, Huang X, Liu L, Chen Y, Du J, Dai Z, Qian Y, Xu L. Variations of bile bacterial community alongside gallstone disease progression and key taxa involved in poor outcomes after endoscopic surgery. Eur J Med Res 2023; 28:313. [PMID: 37660138 PMCID: PMC10474685 DOI: 10.1186/s40001-023-01308-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 08/21/2023] [Indexed: 09/04/2023] Open
Abstract
Gallstone disease is a prevalent biliary disease worldwide, and bacteria play vital roles in the disease development and progression, as well as the prognosis after endoscopic surgery. However, there have been limited studies to explore the key taxa involved. In this study, bile samples from healthy controls (HCs, liver donors without hepatobiliary disease) and three diseased groups, namely patients with gallbladder stones (GBS), patients with common bile duct stones (CBDS), and patients with stricture in the common bile duct (SCBD), were collected and analyzed. Bacterial community characterization based on 16S rRNA amplicon sequencing showed that bacterial diversities did not change significantly alongside gallstone disease development and progression. The predominant phyla in each group were Proteobacteria, Firmicutes, Bacteroidota, and Fusobacteriota, representing over 80% in abundance of the biliary bacteria community. Specifically, the abundance of Proteobacteria decreased greatly while that of Firmicutes and Bacteroidota increased greatly in the diseased groups when compared to that in HCs. Moreover, linear discriminant analysis identified several genera highly represented in the diseased groups. Among them, Klebsiella, Prevotella, Pseudomonas and Veillonella are persistent in both the HCs group and the diseased groups, indicating an enrichment of local bile bacteria in the diseased bile; while Lachnoanerobaculum, Atopobium, Oribacterium, and Stomatobaculum, those aligned to oral cavity taxa, are persistent in the diseased groups but are transient in the HCs group, and their abundances sequentially increased with the disease development and progression (HCs→GBS→CBDS→SCBD), implying a translocation and colonization of the oral cavity bacteria in the diseased bile. Moreover, co-occurrence network analysis revealed that bacterial infection (e.g., Photobacterium and Plesiomonas) from the intestine was developed during endoscopic surgery with reduced bile bacteria diversity. The results of this study revealed that the bile bacterial community is relatively stable and dominated by a few persistent taxa. Moreover, we hypothesized that translocation and colonization of specific bacteria from the oral cavity happens alongside gallstone disease development and progression, and bacterial infection from the intestinal tract results in poor outcomes after endoscopic surgery.
Collapse
Affiliation(s)
- Xunchao Cai
- Department of Gastroenterology and Hepatology, Shenzhen University General Hospital, Shenzhen, 518055, China
| | - Yao Peng
- Department of Gastroenterology and Hepatology, Shenzhen University General Hospital, Shenzhen, 518055, China
| | - Yajie Gong
- Department of Gastroenterology and Hepatology, Shenzhen University General Hospital, Shenzhen, 518055, China
| | - Xiuting Huang
- Department of Gastroenterology and Hepatology, Shenzhen University General Hospital, Shenzhen, 518055, China
| | - Lu Liu
- Department of Gastroenterology and Hepatology, Shenzhen University General Hospital, Shenzhen, 518055, China
| | - Yifan Chen
- Department of Gastroenterology and Hepatology, Shenzhen University General Hospital, Shenzhen, 518055, China
| | - Jingfeng Du
- Department of Gastroenterology and Hepatology, Shenzhen University General Hospital, Shenzhen, 518055, China
| | - Zhongming Dai
- Department of Gastroenterology and Hepatology, Shenzhen University General Hospital, Shenzhen, 518055, China
| | - Yun Qian
- Department of Gastroenterology and Hepatology, Shenzhen University General Hospital, Shenzhen, 518055, China
| | - Long Xu
- Department of Gastroenterology and Hepatology, Shenzhen University General Hospital, Shenzhen, 518055, China.
- Marshall Laboratory of Biomedical Engineering, Shenzhen University, Shenzhen, 518055, China.
| |
Collapse
|
25
|
Grander C, Grabherr F, Tilg H. Non-alcoholic fatty liver disease: pathophysiological concepts and treatment options. Cardiovasc Res 2023; 119:1787-1798. [PMID: 37364164 PMCID: PMC10405569 DOI: 10.1093/cvr/cvad095] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/30/2022] [Accepted: 06/23/2023] [Indexed: 06/28/2023] Open
Abstract
The prevalence of non-alcoholic fatty liver disease (NAFLD) is continually increasing due to the global obesity epidemic. NAFLD comprises a systemic metabolic disease accompanied frequently by insulin resistance and hepatic and systemic inflammation. Whereas simple hepatic steatosis is the most common disease manifestation, a more progressive disease course characterized by liver fibrosis and inflammation (i.e. non-alcoholic steatohepatitis) is present in 10-20% of affected individuals. NAFLD furthermore progresses in a substantial number of patients towards liver cirrhosis and hepatocellular carcinoma. Whereas this disease now affects almost 25% of the world's population and is mainly observed in obesity and type 2 diabetes, NAFLD also affects lean individuals. Pathophysiology involves lipotoxicity, hepatic immune disturbances accompanied by hepatic insulin resistance, a gut dysbiosis, and commonly hepatic and systemic insulin resistance defining this disorder a prototypic systemic metabolic disorder. Not surprisingly many affected patients have other disease manifestations, and indeed cardiovascular disease, chronic kidney disease, and extrahepatic malignancies are all contributing substantially to patient outcome. Weight loss and lifestyle change reflect the cornerstone of treatment, and several medical treatment options are currently under investigation. The most promising treatment strategies include glucagon-like peptide 1 receptor antagonists, sodium-glucose transporter 2 inhibitors, Fibroblast Growth Factor analogues, Farnesoid X receptor agonists, and peroxisome proliferator-activated receptor agonists. Here, we review epidemiology, pathophysiology, and therapeutic options for NAFLD.
Collapse
Affiliation(s)
- Christoph Grander
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University Innsbruck, Anichstrasse 35, Innsbruck 6020, Austria
| | - Felix Grabherr
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University Innsbruck, Anichstrasse 35, Innsbruck 6020, Austria
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University Innsbruck, Anichstrasse 35, Innsbruck 6020, Austria
| |
Collapse
|
26
|
Effenberger M, Waschina S, Bronowski C, Sturm G, Tassiello O, Sommer F, Zollner A, Watschinger C, Grabherr F, Gstir R, Grander C, Enrich B, Bale R, Putzer D, Djanani A, Moschen AR, Zoller H, Rupp J, Schreiber S, Burcelin R, Lass-Flörl C, Trajanoski Z, Oberhuber G, Rosenstiel P, Adolph TE, Aden K, Tilg H. A gut bacterial signature in blood and liver tissue characterizes cirrhosis and hepatocellular carcinoma. Hepatol Commun 2023; 7:e00182. [PMID: 37314752 DOI: 10.1097/hc9.0000000000000182] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 04/18/2023] [Indexed: 06/15/2023] Open
Abstract
BACKGROUND HCC is the leading cause of cancer in chronic liver disease. A growing body of experimental mouse models supports the notion that gut-resident and liver-resident microbes control hepatic immune responses and, thereby, crucially contribute to liver tumorigenesis. However, a comprehensive characterization of the intestinal microbiome in fueling the transition from chronic liver disease to HCC in humans is currently missing. METHODS Here, we profiled the fecal, blood, and liver tissue microbiome of patients with HCC by 16S rRNA sequencing and compared profiles to nonmalignant cirrhotic and noncirrhotic NAFLD patients. RESULTS We report a distinct bacterial profile, defined from 16S rRNA gene sequences, with reduced α-and β-diversity in the feces of patients with HCC and cirrhosis compared to NAFLD. Patients with HCC and cirrhosis exhibited an increased proportion of fecal bacterial gene signatures in the blood and liver compared to NAFLD. Differential analysis of the relative abundance of bacterial genera identified an increased abundance of Ruminococcaceae and Bacteroidaceae in blood and liver tissue from both HCC and cirrhosis patients compared to NAFLD. Fecal samples from cirrhosis and HCC patients both showed a reduced abundance for several taxa, including short-chain fatty acid-producing genera, such as Blautia and Agathobacter. Using paired 16S rRNA and transcriptome sequencing, we identified a direct association between gut bacterial genus abundance and host transcriptome response within the liver tissue. CONCLUSIONS Our study indicates perturbations of the intestinal and liver-resident microbiome as a critical determinant of patients with cirrhosis and HCC.
Collapse
Affiliation(s)
- Maria Effenberger
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Silvio Waschina
- Institute for Human Nutrition and Food Science, Division of Nutriinformatics, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Christina Bronowski
- Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Gregor Sturm
- Biocenter, Institute of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
| | - Oronzo Tassiello
- Institute for Human Nutrition and Food Science, Division of Nutriinformatics, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Felix Sommer
- Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Andreas Zollner
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Christina Watschinger
- Department of Internal Medicine I, Gastroenterology, Nephrology, Metabolism & Endocrinology, Johannes Kepler University, Linz, Austria
| | - Felix Grabherr
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Ronald Gstir
- Institute of Hygiene and Medical Microbiology, ECMM, Medical University of Innsbruck, Innsbruck, Austria
| | - Christoph Grander
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Barbara Enrich
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Reto Bale
- Department of Radiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Daniel Putzer
- Department of Radiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Angela Djanani
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Alexander R Moschen
- Department of Internal Medicine I, Gastroenterology, Nephrology, Metabolism & Endocrinology, Johannes Kepler University, Linz, Austria
- Christian Doppler Laboratory for Mucosal Immunology, Johannes Kepler University, Linz, Austria
| | - Heinz Zoller
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Jan Rupp
- Department of Infectious Diseases and Microbiology, University Hospital Schleswig-Holstein, Luebeck, Germany
| | - Stefan Schreiber
- Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
- Department of Internal Medicine I, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Remy Burcelin
- INSERM 1297 and University Paul Sabatier: Institut des Maladies Métaboliques et Cardiovasculaires, France and Université Paul Sabatier, Toulouse, France
| | - Cornelia Lass-Flörl
- Institute of Hygiene and Medical Microbiology, ECMM, Medical University of Innsbruck, Innsbruck, Austria
| | - Zlatko Trajanoski
- Biocenter, Institute of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
| | - Georg Oberhuber
- INNPATH, Institute of Pathology, University Hospital of Innsbruck, Innsbruck, Austria
| | - Philip Rosenstiel
- Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Timon E Adolph
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Konrad Aden
- Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
- Department of Internal Medicine I, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
27
|
Sciarra F, Franceschini E, Campolo F, Venneri MA. The Diagnostic Potential of the Human Blood Microbiome: Are We Dreaming or Awake? Int J Mol Sci 2023; 24:10422. [PMID: 37445600 DOI: 10.3390/ijms241310422] [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: 05/03/2023] [Revised: 06/01/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
Human blood has historically been considered a sterile environment. Recently, a thriving microbiome dominated by Firmicutes, Actinobacteria, Proteobacteria, and Bacteroidetes phyla was detected in healthy blood. The localization of these microbes is restricted to some blood cell populations, particularly the peripheral blood mononuclear cells and erythrocytes. It was hypothesized that the blood microbiome originates from the skin-oral-gut axis. In addition, many studies have evaluated the potential of blood microbiome dysbiosis as a prognostic marker in cardiovascular diseases, cirrhosis, severe liver fibrosis, severe acute pancreatitis, type 2 diabetes, and chronic kidney diseases. The present review aims to summarize current findings and most recent evidence in the field.
Collapse
Affiliation(s)
- Francesca Sciarra
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Edoardo Franceschini
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Federica Campolo
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Mary Anna Venneri
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy
| |
Collapse
|
28
|
Gui W, Hole MJ, Molinaro A, Edlund K, Jørgensen KK, Su H, Begher-Tibbe B, Gaßler N, Schneider CV, Muthukumarasamy U, Mohs A, Liao L, Jaeger J, Mertens CJ, Bergheim I, Strowig T, Hengstler JG, Hov JR, Marschall HU, Trautwein C, Schneider KM. Colitis ameliorates cholestatic liver disease via suppression of bile acid synthesis. Nat Commun 2023; 14:3304. [PMID: 37280200 DOI: 10.1038/s41467-023-38840-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 05/18/2023] [Indexed: 06/08/2023] Open
Abstract
Primary sclerosing cholangitis (PSC) is a chronic cholestatic liver disease characterized by chronic inflammation and progressive fibrosis of the biliary tree. The majority of PSC patients suffer from concomitant inflammatory bowel disease (IBD), which has been suggested to promote disease development and progression. However, the molecular mechanisms by which intestinal inflammation may aggravate cholestatic liver disease remain incompletely understood. Here, we employ an IBD-PSC mouse model to investigate the impact of colitis on bile acid metabolism and cholestatic liver injury. Unexpectedly, intestinal inflammation and barrier impairment improve acute cholestatic liver injury and result in reduced liver fibrosis in a chronic colitis model. This phenotype is independent of colitis-induced alterations of microbial bile acid metabolism but mediated via hepatocellular NF-κB activation by lipopolysaccharide (LPS), which suppresses bile acid metabolism in-vitro and in-vivo. This study identifies a colitis-triggered protective circuit suppressing cholestatic liver disease and encourages multi-organ treatment strategies for PSC.
Collapse
Affiliation(s)
- Wenfang Gui
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, 52074, Germany
| | - Mikal Jacob Hole
- Norwegian PSC Research Center, Section of Gastroenterology and Research Institute of Internal Medicine, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Antonio Molinaro
- Department of Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
- Norwegian PSC Research Center, Department of Transplantation Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Karolina Edlund
- Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Dortmund, 44139, Germany
| | - Kristin K Jørgensen
- Norwegian PSC Research Center, Department of Transplantation Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Department of Gastroenterology, Akershus University Hospital, Lørenskog, Norway
| | - Huan Su
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, 52074, Germany
| | - Brigitte Begher-Tibbe
- Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Dortmund, 44139, Germany
| | - Nikolaus Gaßler
- Institute for Legal Medicine, Section Pathology, University Hospital, Jena, 07747, Germany
| | - Carolin V Schneider
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, 52074, Germany
| | - Uthayakumar Muthukumarasamy
- Helmholtz Centre for Infection Research, Braunschweig, Germany and Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, 97080, Germany
| | - Antje Mohs
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, 52074, Germany
| | - Lijun Liao
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, 52074, Germany
- Department of Anesthesiology and Pain Management, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Julius Jaeger
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, 52074, Germany
| | - Christian J Mertens
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, 52074, Germany
| | - Ina Bergheim
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, Vienna, A-1090, Austria
| | - Till Strowig
- Helmholtz Centre for Infection Research, Braunschweig, Germany and Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, 97080, Germany
| | - Jan G Hengstler
- Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, Dortmund, 44139, Germany
| | - Johannes R Hov
- Norwegian PSC Research Center, Section of Gastroenterology and Research Institute of Internal Medicine, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Hanns-Ulrich Marschall
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 41345, Sweden
| | - Christian Trautwein
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, 52074, Germany.
| | - Kai Markus Schneider
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, 52074, Germany.
| |
Collapse
|
29
|
Morris I, Croes CA, Boes M, Kalkhoven E. Advanced omics techniques shed light on CD1d-mediated lipid antigen presentation to iNKT cells. Biochim Biophys Acta Mol Cell Biol Lipids 2023; 1868:159292. [PMID: 36773690 DOI: 10.1016/j.bbalip.2023.159292] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 01/26/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023]
Abstract
Invariant natural killer T cells (iNKT cells) can be activated through binding antigenic lipid/CD1d complexes to their TCR. Antigenic lipids are processed, loaded, and displayed in complex with CD1d by lipid antigen presenting cells (LAPCs). The mechanism of lipid antigen presentation via CD1d is highly conserved with recent work showing adipocytes are LAPCs that, besides having a role in lipid storage, can activate iNKT cells and play an important role in systemic metabolic disease. Recent studies shed light on parameters potentially dictating cytokine output and how obesity-associated metabolic disease may affect such parameters. By following a lipid antigen's journey, we identify five key areas which may dictate cytokine skew: co-stimulation, structural properties of the lipid antigen, stability of lipid antigen/CD1d complexes, intracellular and extracellular pH, and intracellular and extracellular lipid environment. Recent publications indicate that the combination of advanced omics-type approaches and machine learning may be a fruitful way to interconnect these 5 areas, with the ultimate goal to provide new insights for therapeutic exploration.
Collapse
Affiliation(s)
- Imogen Morris
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, 3584, CG, Utrecht, the Netherlands
| | - Cresci-Anne Croes
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, 6708WE Wageningen, the Netherlands
| | - Marianne Boes
- Center for Translational Immunology, University Medical Centre Utrecht, Utrecht University, Lundlaan 6, 3584, EA, Utrecht, the Netherlands; Department of Paediatric Immunology, University Medical Center Utrecht, Utrecht University, Lundlaan 6, 3584, EA, Utrecht, the Netherlands
| | - Eric Kalkhoven
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, 3584, CG, Utrecht, the Netherlands.
| |
Collapse
|
30
|
Ullah Goraya M, Li R, Gu L, Deng H, Wang G. Blood Stream Microbiota Dysbiosis Establishing New Research Standards in Cardio-Metabolic Diseases, A Meta-Analysis Study. Microorganisms 2023; 11:microorganisms11030777. [PMID: 36985350 PMCID: PMC10052040 DOI: 10.3390/microorganisms11030777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/09/2023] [Accepted: 03/11/2023] [Indexed: 03/19/2023] Open
Abstract
AIMS Scientists have recently discovered a link between the circulating microbiome and homeostasis, as well as the pathogenesis of a number of metabolic diseases. It has been demonstrated that low-grade chronic inflammation is one of the primary mechanisms that has long been implicated in the risk of cardio-metabolic disease (CMDs) and its progression. Currently, the dysbiosis of circulating bacteria is considered as a key regulator for chronic inflammation in CMDs, which is why we have conducted this systemic review focused on circulating bacterial dysbiosis. METHODS A systemic review of clinical and research-based studies was conducted via PubMed, Scopus, Medline, and Web of Science. Literature was considered for risk of bias and patterns of intervention effects. A randomized effect model was used to evaluate the dysbiosis of circulating microbiota and clinical outcomes. We conducted a meta-analysis considering the circulating bacteria in both healthy people and people with cardio-metabolic disorders, in reports published mainly from 2008 to 2022, according to the PRISMA guidelines. RESULTS We searched 627 studies and, after completing the risk of bias and selection, 31 studies comprising of 11,132 human samples were considered. This meta-analysis found that dysbiosis of phyla Proteobacteria, Firmicutes, and Bacteroidetes was associated with metabolic diseases. CONCLUSIONS In most instances, metabolic diseases are linked to higher diversity and elevated bacterial DNA levels. Bacteroides abundance was higher in healthy people than with metabolic disorders. However, more rigorous studies are required to determine the role of bacterial dysbiosis in cardio-metabolic diseases. Understanding the relationship between dysbiosis and cardio-metabolic diseases, we can use the bacteria as therapeutics for the reversal of dysbiosis and targets for therapeutics use in cardio-metabolic diseases. In the future, circulating bacterial signatures can be used as biomarkers for the early detection of metabolic diseases.
Collapse
Affiliation(s)
| | - Rui Li
- Correspondence: (R.L.); (G.W.)
| | | | | | | |
Collapse
|
31
|
Pirola CJ, Sookoian S. Advances in our understanding of the molecular heterogeneity of fatty liver disease: toward informed treatment decision making. Expert Rev Gastroenterol Hepatol 2023; 17:317-324. [PMID: 36912694 DOI: 10.1080/17474124.2023.2191190] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/14/2023]
Abstract
INTRODUCTION nonalcoholic fatty liver disease (NAFLD) is a complex disorder resulting from intricate relationships with diverse cardiometabolic risk factors and environmental factors. NAFLD may result in severe chronic liver damage and potentially declining liver function. AREAS COVERED Accumulated knowledge over the last decade indicates that the disease trajectory presents substantial heterogeneity. In addition, overlapping features with the diseases of the metabolic syndrome, combined with heterogeneity in disease mechanisms, further complicates NAFLD diagnosis and prognosis, and hampers progress in biomarker and pharmacological discoveries. Here, we explore solving the heterogeneous clinical landscape of NAFLD by cluster analysis of molecular signatures that serve as a proxy for disease stratification into molecular sub-types. First, we collected information on NAFLD and metabolic syndrome-associated protein-coding genes by data mining the literature. Next, we performed pathways enrichment and cluster analyses to decipher and dissect the different patterns of phenotypic heterogeneity. Our approach showed unique biological pathways for every clinical subtype/group, namely NAFLD + obesity, NAFLD + arterial hypertension, NAFLD + dyslipidemia, and NAFLD + type 2 diabetes. EXPERT OPINION Patients with NAFLD may be benefited by a better understanding of the disease biology, which involves 'dissection' of the molecular sub-phenotypes that drive the disease progression.
Collapse
Affiliation(s)
- Carlos J Pirola
- Systems Biology of Complex Diseases, Centro de Altos Estudios En Ciencias Humanas Y de la Salud (CAECIHS), Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas Y Técnicas (CONICET), Buenos Aires, Argentina
| | - Silvia Sookoian
- Clinical and Molecular Hepatology, Centro de Altos Estudios En Ciencias Humanas Y de la Salud (CAECIHS), Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas Y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
32
|
Cheng HS, Tan SP, Wong DMK, Koo WLY, Wong SH, Tan NS. The Blood Microbiome and Health: Current Evidence, Controversies, and Challenges. Int J Mol Sci 2023; 24:5633. [PMID: 36982702 PMCID: PMC10059777 DOI: 10.3390/ijms24065633] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/14/2023] [Accepted: 03/14/2023] [Indexed: 03/18/2023] Open
Abstract
Blood is conventionally thought to be sterile. However, emerging evidence on the blood microbiome has started to challenge this notion. Recent reports have revealed the presence of genetic materials of microbes or pathogens in the blood circulation, leading to the conceptualization of a blood microbiome that is vital for physical wellbeing. Dysbiosis of the blood microbial profile has been implicated in a wide range of health conditions. Our review aims to consolidate recent findings about the blood microbiome in human health and to highlight the existing controversies, prospects, and challenges around this topic. Current evidence does not seem to support the presence of a core healthy blood microbiome. Common microbial taxa have been identified in some diseases, for instance, Legionella and Devosia in kidney impairment, Bacteroides in cirrhosis, Escherichia/Shigella and Staphylococcus in inflammatory diseases, and Janthinobacterium in mood disorders. While the presence of culturable blood microbes remains debatable, their genetic materials in the blood could potentially be exploited to improve precision medicine for cancers, pregnancy-related complications, and asthma by augmenting patient stratification. Key controversies in blood microbiome research are the susceptibility of low-biomass samples to exogenous contamination and undetermined microbial viability from NGS-based microbial profiling, however, ongoing initiatives are attempting to mitigate these issues. We also envisage future blood microbiome research to adopt more robust and standardized approaches, to delve into the origins of these multibiome genetic materials and to focus on host-microbe interactions through the elaboration of causative and mechanistic relationships with the aid of more accurate and powerful analytical tools.
Collapse
Affiliation(s)
- Hong Sheng Cheng
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore 308232, Singapore; (S.H.W.); (N.S.T.)
| | - Sin Pei Tan
- Radiotherapy and Oncology Department, Hospital Sultan Ismail, Jalan Mutiara Emas Utama, Taman Mount Austin, Johor Bahru 81100, Malaysia
| | - David Meng Kit Wong
- School of Biological Sciences, Nanyang Technological University Singapore, Singapore 637551, Singapore
| | - Wei Ling Yolanda Koo
- School of Biological Sciences, Nanyang Technological University Singapore, Singapore 637551, Singapore
| | - Sunny Hei Wong
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore 308232, Singapore; (S.H.W.); (N.S.T.)
| | - Nguan Soon Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore 308232, Singapore; (S.H.W.); (N.S.T.)
- School of Biological Sciences, Nanyang Technological University Singapore, Singapore 637551, Singapore
| |
Collapse
|
33
|
Sumida K, Pierre JF, Yuzefpolskaya M, Colombo PC, Demmer RT, Kovesdy CP. Gut Microbiota-Targeted Interventions in the Management of Chronic Kidney Disease. Semin Nephrol 2023; 43:151408. [PMID: 37619529 PMCID: PMC10783887 DOI: 10.1016/j.semnephrol.2023.151408] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/26/2023]
Abstract
Recent advances in microbiome research have informed the potential role of the gut microbiota in the regulation of metabolic, cardiovascular, and renal systems, and, when altered, in the pathogenesis of various cardiometabolic disorders, including chronic kidney disease (CKD). The improved understanding of gut dysbiosis in cardiometabolic pathologies in turn has led to a vigorous quest for developing therapeutic strategies. These therapeutic strategies aim to investigate whether interventions targeting gut dysbiosis can shift the microbiota toward eubiosis and if these shifts, in turn, translate into improvements in (or prevention of) CKD and its related complications, such as premature cardiovascular disease. Existing evidence suggests that multiple interventions (eg, plant-based diets; prebiotic, probiotic, and synbiotic supplementation; constipation treatment; fecal microbiota transplantation; and intestinal dialysis) might result in favorable modulation of the gut microbiota in patients with CKD, and thereby potentially contribute to improving clinical outcomes in these patients. In this review, we summarize the current understanding of the characteristics and roles of the gut microbiota in CKD and discuss the potential of emerging gut microbiota-targeted interventions in the management of CKD.
Collapse
Affiliation(s)
- Keiichi Sumida
- Division of Nephrology, Department of Medicine, University of Tennessee Health Science Center, Memphis, TN.
| | - Joseph F Pierre
- Department of Nutritional Sciences, College of Agriculture and Life Science, University of Wisconsin-Madison, Madison, WI
| | - Melana Yuzefpolskaya
- Division of Cardiology, Department of Medicine, New York Presbyterian Hospital, Columbia University, New York, NY
| | - Paolo C Colombo
- Division of Cardiology, Department of Medicine, New York Presbyterian Hospital, Columbia University, New York, NY
| | - Ryan T Demmer
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN; Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY
| | - Csaba P Kovesdy
- Division of Nephrology, Department of Medicine, University of Tennessee Health Science Center, Memphis, TN
| |
Collapse
|
34
|
Champion C, Neagoe RM, Effernberger M, Sala DT, Servant F, Christensen JE, Arnoriaga-Rodriguez M, Amar J, Lelouvier B, Loubieres P, Azalbert V, Minty M, Thomas C, Blasco-Baque V, Gamboa F, Tilg H, Cardellini M, Federici M, Fernández-Real JM, Loubes JM, Burcelin R. Human liver microbiota modeling strategy at the early onset of fibrosis. BMC Microbiol 2023; 23:34. [PMID: 36717776 PMCID: PMC9885577 DOI: 10.1186/s12866-023-02774-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 01/13/2023] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Gut microbiota is involved in the development of liver diseases such as fibrosis. We and others identified that selected sets of gut bacterial DNA and bacteria translocate to tissues, notably the liver, to establish a non-infectious tissue microbiota composed of microbial DNA and a low frequency live bacteria. However, the precise set of bacterial DNA, and thereby the corresponding taxa associated with the early stages of fibrosis need to be identified. Furthermore, to overcome the impact of different group size and patient origins we adapted innovative statistical approaches. Liver samples with low liver fibrosis scores (F0, F1, F2), to study the early stages of the disease, were collected from Romania(n = 36), Austria(n = 10), Italy(n = 19), and Spain(n = 17). The 16S rRNA gene was sequenced. We considered the frequency, sparsity, unbalanced sample size between cohorts to identify taxonomic profiles and statistical differences. RESULTS Multivariate analyses, including adapted spectral clustering with L1-penalty fair-discriminant strategies, and predicted metagenomics were used to identify that 50% of liver taxa associated with the early stage fibrosis were Enterobacteriaceae, Pseudomonadaceae, Xanthobacteriaceae and Burkholderiaceae. The Flavobacteriaceae and Xanthobacteriaceae discriminated between F0 and F1. Predicted metagenomics analysis identified that the preQ0 biosynthesis and the potential pathways involving glucoryranose and glycogen degradation were negatively associated with liver fibrosis F1-F2 vs F0. CONCLUSIONS Without demonstrating causality, our results suggest first a role of bacterial translocation to the liver in the progression of fibrosis, notably at the earliest stages. Second, our statistical approach can identify microbial signatures and overcome issues regarding sample size differences, the impact of environment, and sets of analyses. TRIAL REGISTRATION TirguMECCH ROLIVER Prospective Cohort for the Identification of Liver Microbiota, registration 4065/2014. Registered 01 01 2014.
Collapse
Affiliation(s)
- Camille Champion
- grid.7429.80000000121866389Institut National de La Santé Et de La Recherche Médicale (INSERM), Toulouse, France ,grid.15781.3a0000 0001 0723 035XUnité Mixte de Recherche (UMR) 1297, Institut Des Maladies Métaboliques Et Cardiovasculaires (I2MC), Team 2: ‘Intestinal Risk FactorsDiabetesDyslipidemia’, Université Paul Sabatier (UPS), F-31432 Toulouse Cedex 4, France ,grid.15781.3a0000 0001 0723 035XInstitut de Mathématiques de Toulouse, Université Paul Sabatier, Toulouse, France
| | - Radu M. Neagoe
- Second Department of Surgery, Emergency Mureş County Hospital, University of Medicine Pharmacy, Science and Technology “George Emil Palade” Tîrgu Mures, Târgu Mureș, Romania
| | - Maria Effernberger
- grid.5361.10000 0000 8853 2677Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Daniela T. Sala
- Second Department of Surgery, Emergency Mureş County Hospital, University of Medicine Pharmacy, Science and Technology “George Emil Palade” Tîrgu Mures, Târgu Mureș, Romania
| | | | - Jeffrey E. Christensen
- grid.7429.80000000121866389Institut National de La Santé Et de La Recherche Médicale (INSERM), Toulouse, France ,grid.15781.3a0000 0001 0723 035XUnité Mixte de Recherche (UMR) 1297, Institut Des Maladies Métaboliques Et Cardiovasculaires (I2MC), Team 2: ‘Intestinal Risk FactorsDiabetesDyslipidemia’, Université Paul Sabatier (UPS), F-31432 Toulouse Cedex 4, France
| | - Maria Arnoriaga-Rodriguez
- grid.411295.a0000 0001 1837 4818Department of Diabetes, Endocrinology and Nutrition, University Hospital of Girona ‘Dr Josep Trueta’, Girona, Spain ,grid.429182.4Institut d’Investigacio Biomedica de Girona IdibGi, Girona, Spain ,CIBER Fisiopatologia de La Obesidad Y Nutricion, Girona, Spain
| | - Jacques Amar
- grid.7429.80000000121866389Institut National de La Santé Et de La Recherche Médicale (INSERM), Toulouse, France ,grid.15781.3a0000 0001 0723 035XUnité Mixte de Recherche (UMR) 1297, Institut Des Maladies Métaboliques Et Cardiovasculaires (I2MC), Team 2: ‘Intestinal Risk FactorsDiabetesDyslipidemia’, Université Paul Sabatier (UPS), F-31432 Toulouse Cedex 4, France ,grid.414295.f0000 0004 0638 3479Therapeutics Department, Rangueil Hospital, Toulouse, France
| | | | - Pascale Loubieres
- grid.7429.80000000121866389Institut National de La Santé Et de La Recherche Médicale (INSERM), Toulouse, France ,grid.15781.3a0000 0001 0723 035XUnité Mixte de Recherche (UMR) 1297, Institut Des Maladies Métaboliques Et Cardiovasculaires (I2MC), Team 2: ‘Intestinal Risk FactorsDiabetesDyslipidemia’, Université Paul Sabatier (UPS), F-31432 Toulouse Cedex 4, France
| | - Vincent Azalbert
- grid.7429.80000000121866389Institut National de La Santé Et de La Recherche Médicale (INSERM), Toulouse, France ,grid.15781.3a0000 0001 0723 035XUnité Mixte de Recherche (UMR) 1297, Institut Des Maladies Métaboliques Et Cardiovasculaires (I2MC), Team 2: ‘Intestinal Risk FactorsDiabetesDyslipidemia’, Université Paul Sabatier (UPS), F-31432 Toulouse Cedex 4, France
| | - Matthieu Minty
- grid.7429.80000000121866389Institut National de La Santé Et de La Recherche Médicale (INSERM), Toulouse, France ,grid.15781.3a0000 0001 0723 035XUnité Mixte de Recherche (UMR) 1297, Institut Des Maladies Métaboliques Et Cardiovasculaires (I2MC), Team 2: ‘Intestinal Risk FactorsDiabetesDyslipidemia’, Université Paul Sabatier (UPS), F-31432 Toulouse Cedex 4, France
| | - Charlotte Thomas
- grid.7429.80000000121866389Institut National de La Santé Et de La Recherche Médicale (INSERM), Toulouse, France ,grid.15781.3a0000 0001 0723 035XUnité Mixte de Recherche (UMR) 1297, Institut Des Maladies Métaboliques Et Cardiovasculaires (I2MC), Team 2: ‘Intestinal Risk FactorsDiabetesDyslipidemia’, Université Paul Sabatier (UPS), F-31432 Toulouse Cedex 4, France
| | - Vincent Blasco-Baque
- grid.7429.80000000121866389Institut National de La Santé Et de La Recherche Médicale (INSERM), Toulouse, France ,grid.15781.3a0000 0001 0723 035XUnité Mixte de Recherche (UMR) 1297, Institut Des Maladies Métaboliques Et Cardiovasculaires (I2MC), Team 2: ‘Intestinal Risk FactorsDiabetesDyslipidemia’, Université Paul Sabatier (UPS), F-31432 Toulouse Cedex 4, France
| | - Fabrice Gamboa
- grid.15781.3a0000 0001 0723 035XInstitut de Mathématiques de Toulouse, Université Paul Sabatier, Toulouse, France
| | - Herbert Tilg
- grid.5361.10000 0000 8853 2677Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Marina Cardellini
- grid.6530.00000 0001 2300 0941Department of Systems Medicine, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy
| | - Massimo Federici
- grid.6530.00000 0001 2300 0941Department of Systems Medicine, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy
| | - Jose-Manuel Fernández-Real
- grid.411295.a0000 0001 1837 4818Department of Diabetes, Endocrinology and Nutrition, University Hospital of Girona ‘Dr Josep Trueta’, Girona, Spain ,grid.429182.4Institut d’Investigacio Biomedica de Girona IdibGi, Girona, Spain ,CIBER Fisiopatologia de La Obesidad Y Nutricion, Girona, Spain
| | - Jean Michel Loubes
- grid.15781.3a0000 0001 0723 035XInstitut de Mathématiques de Toulouse, Université Paul Sabatier, Toulouse, France
| | - Rémy Burcelin
- grid.7429.80000000121866389Institut National de La Santé Et de La Recherche Médicale (INSERM), Toulouse, France ,grid.15781.3a0000 0001 0723 035XUnité Mixte de Recherche (UMR) 1297, Institut Des Maladies Métaboliques Et Cardiovasculaires (I2MC), Team 2: ‘Intestinal Risk FactorsDiabetesDyslipidemia’, Université Paul Sabatier (UPS), F-31432 Toulouse Cedex 4, France
| |
Collapse
|
35
|
Hui D, Liu L, Azami NLB, Song J, Huang Y, Xu W, Wu C, Xie D, Jiang Y, Bian Y, Sun M. The spleen-strengthening and liver-draining herbal formula treatment of non-alcoholic fatty liver disease by regulation of intestinal flora in clinical trial. Front Endocrinol (Lausanne) 2023; 13:1107071. [PMID: 36743913 PMCID: PMC9892935 DOI: 10.3389/fendo.2022.1107071] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 12/29/2022] [Indexed: 01/20/2023] Open
Abstract
OBJECTIVE As a metabolic disease, one important feature of non-alcoholic fatty liver disease (NAFLD) is the disturbance of the intestinal flora. Spleen-strengthening and liver-draining formula (SLF) is a formula formed according to the theory of "One Qi Circulation" (Qing Dynasty, 1749) of Traditional Chinese Medicine (TCM), which has shown significant therapeutic effect in patients with NAFLD in a preliminary clinical observation. In this study, we aim to explore the mechanism of SLF against NAFLD, especially its effect on glucolipid metabolism, from the perspective of intestinal flora. METHODS A prospective, randomized, controlled clinical study was designed to observe the efficacy and safety of SLF in the treatment of NAFLD. The study participants were randomly and evenly divided into control group and treatment group (SLF group). The control group made lifestyle adjustments, while the SLF group was treated with SLF on top of the control group. Both groups were participated in the study for 12 consecutive weeks. Furthermore, the feces of the two groups were collected before and after treatment. The intestinal flora of each group and healthy control (HC) were detected utilizing 16S rRNA gene sequencing. RESULTS Compared with the control group, the SLF group showed significant improvements in liver function, controlled attenuation parameter (CAP), and liver stiffness measurement (LSM), meanwhile, patients had significantly lower lipid and homeostasis model assessment of insulin resistance (HOMA-IR) with better security. Intestinal flora 16S rRNA gene sequencing results indicated reduced flora diversity and altered species abundance in patients with NAFLD. At the phylum level, Desulfobacterota levels were reduced. Although Firmicutes and Bacteroidetes did not differ significantly between HC and NAFLD, when grouped by alanine transaminase (ALT) and aspartate transaminase (AST) levels in NAFLD, Firmicutes levels were significantly higher in patients with ALT or AST abnormalities, while Bacteroidetes was significantly lower. Clinical correlation analysis showed that Firmicutes positively correlated with gender, age, ALT, AST, LSM, and Fibroscan-AST (FAST) score, while the opposite was true for Bacteroidetes. At the genus level, the levels of Alistipes, Bilophila, Butyricimonas, Coprococcus, Lachnospiraceae_NK4A136 group Phascolarctobacterium, Ruminococcus, UCG-002, and UCG-003 were reduced, whereas abundance of Tyzzerella increased. There was no statistically significant difference in Firmicutes and Bacteroidota levels in the SLF group before and after treatment, but both bacteria tended to retrace. At the genus level, Coprococcus (Lachnospiraceae family), Lachnospiraceae_NK4A136 group (Lachnospiraceae family), and Ruminococcus (Ruminococcaceae family) were significantly higher in the SLF group after treatment, and there was also a tendency for Bilophila (Desulfovibrionaceae family) to be back-regulated toward HC. CONCLUSIONS SLF can improve liver function and glucolipid metabolism in patients with NAFLD and lower down liver fat content to some extent. SLF could be carried out by regulating the disturbance of intestinal flora, especially Coprococcus, Lachnospiraceae_NK4A136 group, and Ruminococcus genus.
Collapse
Affiliation(s)
- Dengcheng Hui
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lu Liu
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Nisma Lena Bahaji Azami
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jingru Song
- Department of Gastroenterology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yanping Huang
- Department of Good Clinical Practice Office, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wan Xu
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chao Wu
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Dong Xie
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yulang Jiang
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yanqin Bian
- Arthritis Institute of Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Mingyu Sun
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Liver Diseases, Key Laboratory of Liver and Kidney Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
36
|
Khan I, Khan I, Usman M, Xiao Wei Z, Ping X, Khan S, Khan F, Jianye Z, Zhiqiang L, Lizhe A. Circulating microbiota and metabolites: Insights into cardiovascular diseases. J Clin Lab Anal 2022; 36:e24779. [DOI: 10.1002/jcla.24779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 10/10/2022] [Accepted: 11/08/2022] [Indexed: 12/05/2022] Open
Affiliation(s)
- Ikram Khan
- Department of Microbiology, School of Life Sciences Lanzhou University Lanzhou Gansu China
- School of Stomatology Northwest Minzu University Lanzhou Gansu China
| | - Imran Khan
- Department of Microbiology Khyber Medical University Peshawar Peshawar Khyber Pakhtunkhwa Pakistan
| | - Muhammad Usman
- State Key Laboratory of Grassland Agro‐ecosystem, Key Laboratory of Grassland Livestock Industry Innovation, Ministry of Agriculture and Ruler Affairs, Collegeof Pastoral Agriculture Sciences and Technology Lanzhou University Lanzhou Gansu China
| | - Zhang Xiao Wei
- Department of Cardiology Lanzhou University Second Hospital Lanzhou Gansu China
| | - Xie Ping
- Department of Cardiology Gansu Provincial Hospital Lanzhou China
| | - Sarmir Khan
- Department of Reproductive Medicine, Academy of Medical Sciences The First Affiliated Hospital of Zheng University Zhengzhou Henan China
| | - Feroz Khan
- Department of Zoology, Wildlife, and Fisheries PirMehr Ali Shah Arid Agriculture University Rawalpindi Pakistan
| | - Zhou Jianye
- School of Stomatology Northwest Minzu University Lanzhou Gansu China
| | - Li Zhiqiang
- School of Stomatology Northwest Minzu University Lanzhou Gansu China
| | - An Lizhe
- Department of Microbiology, School of Life Sciences Lanzhou University Lanzhou Gansu China
| |
Collapse
|
37
|
Tilg H, Adolph TE, Trauner M. Gut-liver axis: Pathophysiological concepts and clinical implications. Cell Metab 2022; 34:1700-1718. [PMID: 36208625 DOI: 10.1016/j.cmet.2022.09.017] [Citation(s) in RCA: 290] [Impact Index Per Article: 96.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/17/2022] [Accepted: 09/16/2022] [Indexed: 02/07/2023]
Abstract
Bidirectional crosstalk along the gut-liver axis controls gastrointestinal health and disease and exploits environmental and host mediators. Nutrients, microbial antigens, metabolites, and bile acids regulate metabolism and immune responses in the gut and liver, which reciprocally shape microbial community structure and function. Perturbation of such host-microbe interactions is observed in a variety of experimental liver diseases and is facilitated by an impaired intestinal barrier, which is fueling hepatic inflammation and disease progression. Clinical evidence describes perturbation of the gut-liver crosstalk in non-alcoholic fatty liver disease, alcoholic liver disease, and primary sclerosing cholangitis. In liver cirrhosis, a common sequela of these diseases, the intestinal microbiota and microbial pathogen-associated molecular patterns constitute liver inflammation and clinical complications, such as hepatic encephalopathy. Understanding the intricate metabolic interplay between the gut and liver in health and disease opens an avenue for targeted therapies in the future, which is probed in controlled clinical trials.
Collapse
Affiliation(s)
- Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University, Innsbruck, Austria.
| | - Timon E Adolph
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University, Innsbruck, Austria
| | - Michael Trauner
- Division of Gastroenterology & Hepatology, Department of Internal Medicine III, Medical University, Vienna, Austria
| |
Collapse
|
38
|
Abstract
We are host to an assembly of microorganisms that vary in structure and function along the length of the gut and from the lumen to the mucosa. This ecosystem is collectively known as the gut microbiota and significant efforts have been spent during the past 2 decades to catalog and functionally describe the normal gut microbiota and how it varies during a wide spectrum of disease states. The gut microbiota is altered in several cardiometabolic diseases and recent work has established microbial signatures that may advance disease. However, most research has focused on identifying associations between the gut microbiota and human diseases states and to investigate causality and potential mechanisms using cells and animals. Since the gut microbiota functions on the intersection between diet and host metabolism, and can contribute to inflammation, several microbially produced metabolites and molecules may modulate cardiometabolic diseases. Here we discuss how the gut bacterial composition is altered in, and can contribute to, cardiometabolic disease, as well as how the gut bacteria can be targeted to treat and prevent metabolic diseases.
Collapse
Affiliation(s)
- Louise E Olofsson
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Sweden
| | - Fredrik Bäckhed
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Sweden.,Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health Sciences, University of Copenhagen, Denmark.,Region Västra Götaland, Sahlgrenska University Hospital, Department of Clinical Physiology, Gothenburg, Sweden
| |
Collapse
|
39
|
Zhang YL, Li ZJ, Gou HZ, Song XJ, Zhang L. The gut microbiota–bile acid axis: A potential therapeutic target for liver fibrosis. Front Cell Infect Microbiol 2022; 12:945368. [PMID: 36189347 PMCID: PMC9519863 DOI: 10.3389/fcimb.2022.945368] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 09/01/2022] [Indexed: 11/15/2022] Open
Abstract
Liver fibrosis involves the proliferation and deposition of extracellular matrix on liver tissues owing to various etiologies (including viral, alcohol, immune, and metabolic factors), ultimately leading to structural and functional abnormalities in the liver. If not effectively treated, liver fibrosis, a pivotal stage in the path to chronic liver disease, can progress to cirrhosis and eventually liver cancer; unfortunately, no specific clinical treatment for liver fibrosis has been established to date. In liver fibrosis cases, both the gut microbiota and bile acid metabolism are disrupted. As metabolites of the gut microbiota, bile acids have been linked to the progression of liver fibrosis via various pathways, thus implying that the gut microbiota–bile acid axis might play a critical role in the progression of liver fibrosis and could be a target for its reversal. Therefore, in this review, we examined the involvement of the gut microbiota–bile acid axis in liver fibrosis progression to the end of discovering new targets for the prevention, diagnosis, and therapy of chronic liver diseases, including liver fibrosis.
Collapse
Affiliation(s)
- Yu-Lin Zhang
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| | - Zhen-Jiao Li
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| | - Hong-Zhong Gou
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| | - Xiao-Jing Song
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| | - Lei Zhang
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
- *Correspondence: Lei Zhang,
| |
Collapse
|
40
|
Goraya MU, Li R, Mannan A, Gu L, Deng H, Wang G. Human circulating bacteria and dysbiosis in non-infectious diseases. Front Cell Infect Microbiol 2022; 12:932702. [PMID: 36093202 PMCID: PMC9448904 DOI: 10.3389/fcimb.2022.932702] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 08/01/2022] [Indexed: 02/05/2023] Open
Abstract
Blood microorganisms were once thought to indicate infection. Blood in healthy people appears to be devoid of growing bacteria; nonetheless, intracellular dormant forms of bacteria have been reported previously. With breakthroughs in sequencing and bioinformatics, the presence of bacterial DNA in healthy human blood initiated the controversy of human blood microbiota (HBM). Recently, bacteria-specific DNA and culturable bacteria were found in healthy human blood. Researchers wanted to study the phenomena of a "healthy blood microbiota" by providing a thorough description of bacterially produced nucleic acids using many complementing molecular and traditional microbiological approaches. Because blood is a relatively limited and particular environment, culturability and plate count issues can be overcome using enhanced cultured procedures. However, more evidence is required to confirm that healthy human blood contains normal microbiota. Cavities, mouth and intestinal microbiota, trauma, surgery, and animal/insect bites can introduce bacteria into human blood. All these factors strengthen the concept of transient blood bacteria too. The presence of blood bacteria may be caused by temporary immunological clearance and absorption by dendritic or M cells. This review provides an extensive and comprehensive analysis that suggests that healthy blood bacteria may not be typical microbiota but transient circulatory microorganisms. In this study, we look at how contaminants (Escherichia, Shigella, Pseudomonads, etc.) from the skin, laboratory environments, and reagents can affect the interpretation of blood-derived microbial information and the relationship between the circulating bacteria and non-communicable diseases. Circulating transient bacteria may play a role in the pathogenesis of non-infectious diseases such as diabetes and CVD. Contamination-free hematological studies can aid in understanding the disease mechanisms, therapy, and biomarkers.
Collapse
Affiliation(s)
- Mohsan Ullah Goraya
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, China
| | - Rui Li
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, China
| | - Abdul Mannan
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
| | - Liming Gu
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, China
| | - Huixiong Deng
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, China
| | - Gefei Wang
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, China
| |
Collapse
|
41
|
Prasad R, Patton MJ, Floyd JL, Fortmann S, DuPont M, Harbour A, Wright J, Lamendella R, Stevens BR, Oudit GY, Grant MB. Plasma Microbiome in COVID-19 Subjects: An Indicator of Gut Barrier Defects and Dysbiosis. Int J Mol Sci 2022; 23:9141. [PMID: 36012406 PMCID: PMC9409329 DOI: 10.3390/ijms23169141] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/10/2022] [Accepted: 08/12/2022] [Indexed: 12/16/2022] Open
Abstract
The gut is a well-established route of infection and target for viral damage by SARS-CoV-2. This is supported by the clinical observation that about half of COVID-19 patients exhibit gastrointestinal (GI) complications. We aimed to investigate whether the analysis of plasma could provide insight into gut barrier dysfunction in patients with COVID-19 infection. Plasma samples of COVID-19 patients (n = 146) and healthy individuals (n = 47) were collected during hospitalization and routine visits. Plasma microbiome was analyzed using 16S rRNA sequencing and gut permeability markers including fatty acid binding protein 2 (FABP2), peptidoglycan (PGN), and lipopolysaccharide (LPS) in both patient cohorts. Plasma samples of both cohorts contained predominately Proteobacteria, Firmicutes, Bacteroides, and Actinobacteria. COVID-19 subjects exhibit significant dysbiosis (p = 0.001) of the plasma microbiome with increased abundance of Actinobacteria spp. (p = 0.0332), decreased abundance of Bacteroides spp. (p = 0.0003), and an increased Firmicutes:Bacteroidetes ratio (p = 0.0003) compared to healthy subjects. The concentration of the plasma gut permeability marker FABP2 (p = 0.0013) and the gut microbial antigens PGN (p < 0.0001) and LPS (p = 0.0049) were significantly elevated in COVID-19 patients compared to healthy subjects. These findings support the notion that the intestine may represent a source for bacteremia and contribute to worsening COVID-19 outcomes. Therapies targeting the gut and prevention of gut barrier defects may represent a strategy to improve outcomes in COVID-19 patients.
Collapse
Affiliation(s)
- Ram Prasad
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, 1670 University BLVD, VH490, Birmingham, AL 35294, USA
| | - Michael John Patton
- Hugh Kaul Precision Medicine Institute, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jason Levi. Floyd
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, 1670 University BLVD, VH490, Birmingham, AL 35294, USA
| | - Seth Fortmann
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, 1670 University BLVD, VH490, Birmingham, AL 35294, USA
| | - Mariana DuPont
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, 1670 University BLVD, VH490, Birmingham, AL 35294, USA
| | - Angela Harbour
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, 1670 University BLVD, VH490, Birmingham, AL 35294, USA
| | | | | | - Bruce R. Stevens
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL 32611, USA
| | - Gavin Y. Oudit
- Division of Cardiology, Department of Medicine, University of Alberta, Mazankowski Alberta Heart Institute, Edmonton, AB T6G 2B7, Canada
| | - Maria B. Grant
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, 1670 University BLVD, VH490, Birmingham, AL 35294, USA
| |
Collapse
|
42
|
Chen S, Jin Y, Wang S, Xing S, Wu Y, Tao Y, Ma Y, Zuo S, Liu X, Hu Y, Chen H, Luo Y, Xia F, Xie C, Yin J, Wang X, Liu Z, Zhang N, Zech Xu Z, Lu ZJ, Wang P. Cancer type classification using plasma cell-free RNAs derived from human and microbes. eLife 2022; 11:e75181. [PMID: 35816095 PMCID: PMC9273212 DOI: 10.7554/elife.75181] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 06/26/2022] [Indexed: 11/23/2022] Open
Abstract
The utility of cell-free nucleic acids in monitoring cancer has been recognized by both scientists and clinicians. In addition to human transcripts, a fraction of cell-free nucleic acids in human plasma were proven to be derived from microbes and reported to have relevance to cancer. To obtain a better understanding of plasma cell-free RNAs (cfRNAs) in cancer patients, we profiled cfRNAs in ~300 plasma samples of 5 cancer types (colorectal cancer, stomach cancer, liver cancer, lung cancer, and esophageal cancer) and healthy donors (HDs) with RNA-seq. Microbe-derived cfRNAs were consistently detected by different computational methods when potential contaminations were carefully filtered. Clinically relevant signals were identified from human and microbial reads, and enriched Kyoto Encyclopedia of Genes and Genomes pathways of downregulated human genes and higher prevalence torque teno viruses both suggest that a fraction of cancer patients were immunosuppressed. Our data support the diagnostic value of human and microbe-derived plasma cfRNAs for cancer detection, as an area under the ROC curve of approximately 0.9 for distinguishing cancer patients from HDs was achieved. Moreover, human and microbial cfRNAs both have cancer type specificity, and combining two types of features could distinguish tumors of five different primary locations with an average recall of 60.4%. Compared to using human features alone, adding microbial features improved the average recall by approximately 8%. In summary, this work provides evidence for the clinical relevance of human and microbe-derived plasma cfRNAs and their potential utilities in cancer detection as well as the determination of tumor sites.
Collapse
Affiliation(s)
- Shanwen Chen
- Division of General Surgery, Peking University First HospitalBeijingChina
- Translational Cancer Research Center, Peking University First HospitalBeijingChina
| | - Yunfan Jin
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua UniversityBeijingChina
| | - Siqi Wang
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua UniversityBeijingChina
| | - Shaozhen Xing
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua UniversityBeijingChina
| | - Yingchao Wu
- Division of General Surgery, Peking University First HospitalBeijingChina
| | - Yuhuan Tao
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua UniversityBeijingChina
| | - Yongchen Ma
- Division of General Surgery, Peking University First HospitalBeijingChina
| | - Shuai Zuo
- Division of General Surgery, Peking University First HospitalBeijingChina
| | - Xiaofan Liu
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua UniversityBeijingChina
| | - Yichen Hu
- State Key Laboratory of Food Science and Technology, Nanchang UniversityNanchangChina
| | - Hongyan Chen
- 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 CollegeBeijingChina
| | - Yuandeng Luo
- Institute of Hepatobiliary Surgery, The First Hospital Affiliated to Army Medical UniversityChongqingChina
| | - Feng Xia
- Institute of Hepatobiliary Surgery, The First Hospital Affiliated to Army Medical UniversityChongqingChina
| | - Chuanming Xie
- Institute of Hepatobiliary Surgery, The First Hospital Affiliated to Army Medical UniversityChongqingChina
| | - Jianhua Yin
- Department of Epidemiology, Faculty of Navy Medicine, Navy Medical UniversityShanghaiChina
| | - Xin Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer /Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Zhihua 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 CollegeBeijingChina
| | - Ning Zhang
- Translational Cancer Research Center, Peking University First HospitalBeijingChina
| | - Zhenjiang Zech Xu
- State Key Laboratory of Food Science and Technology, Nanchang UniversityNanchangChina
- Shenzhen Stomatology Hospital (Pingshan), Southern Medical UniversityShenzhenChina
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical UniversityGuangzhouChina
| | - Zhi John Lu
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua UniversityBeijingChina
| | - Pengyuan Wang
- Division of General Surgery, Peking University First HospitalBeijingChina
| |
Collapse
|
43
|
Yang G, Zhuang L, Sun T, Yeo YH, Tao L, Zhang W, Ma W, Wu L, Yang Z, Yang Y, Xue D, Zhang J, Feng R, Matthias P. E, Dooley S, Seki E, Liu P, Liu C. Serum Glial Cell Line-Derived Neurotrophic Factor (sGDNF) Is a Novel Biomarker in Predicting Cirrhosis in Patients with Chronic Hepatitis B. Can J Gastroenterol Hepatol 2022; 2022:1048104. [PMID: 35855954 PMCID: PMC9288342 DOI: 10.1155/2022/1048104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 05/22/2022] [Accepted: 05/25/2022] [Indexed: 11/23/2022] Open
Abstract
OBJECTIVES We assessed the potential of glial cell line-derived neurotrophic factor (GDNF) as a useful biomarker to predict cirrhosis in chronic hepatitis B (CHB) patients. METHODS A total of 735 patients from two medical centers (385 CHB patients and 350 healthy controls) were included to determine the association of serum and tissue GDNF levels with biopsy-proven cirrhosis. The diagnostic accuracy of serum GDNF (sGDNF) was estimated and compared with other indices of cirrhosis. RESULTS We showed significantly higher levels of sGDNF in CHB patients with fibrosis (28.4 pg/ml vs. 11.6 pg/ml in patients without) and patients with cirrhosis (33.8 pg/ml vs. 23.5 pg/ml in patients without). The areas under receiver operating curve (AUROCs) of sGDNF were 0.83 (95% confidence interval (CI): 0.80-0.87) for predicting liver fibrosis and 0.84 (95% CI: 0.79-0.89) for cirrhosis. Findings from the serum protein level and hepatic mRNA expression were consistent. Using the best cutoff to predict cirrhosis, we categorized the patients into sGDNF-high and sGDNF-low groups. The sGDNF-high group had significantly larger Masson's trichrome and reticulin staining-positive area, higher Scheuer score, and METAVIR fibrosis stage (all p < 0.001) but not steatosis. On multivariable regression, sGDNF was independently associated with cirrhosis with an odds ratio of 6.98 (95% CI: 1.10-17.94). Finally, we demonstrated that sGDNF outperformed AST to platelet ratio index, FIB-4, fibroscore, forn index, and fibrometer in differentiating F4 vs. F3. CONCLUSION Using serum, tissue mRNA, and biopsy data, our study revealed a significant potential of sGDNF as a novel noninvasive biomarker for cirrhosis in CHB patients.
Collapse
Affiliation(s)
- Guangyue Yang
- Laboratory of Liver Disease, Department of Infectious Disease, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
- Experimental Center, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| | - Liping Zhuang
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Tiantian Sun
- Laboratory of Liver Disease, Department of Infectious Disease, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
- Experimental Center, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| | - Yee Hui Yeo
- Division of General Internal Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Le Tao
- Laboratory of Liver Disease, Department of Infectious Disease, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
- Experimental Center, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| | - Wei Zhang
- Laboratory of Liver Disease, Department of Infectious Disease, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
- Experimental Center, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| | - Wenting Ma
- Laboratory of Liver Disease, Department of Infectious Disease, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
- Experimental Center, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| | - Liu Wu
- Laboratory of Liver Disease, Department of Infectious Disease, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
- Experimental Center, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| | - Zongguo Yang
- Department of Integrative Medicine, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Yanqin Yang
- Department of Pathology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| | - Dongying Xue
- Laboratory of Liver Disease, Department of Infectious Disease, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| | - Jie Zhang
- Laboratory of Liver Disease, Department of Infectious Disease, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| | - Rilu Feng
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Ebert Matthias P.
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Steven Dooley
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Ekihiro Seki
- Division of Digestive and Liver Diseases, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Ping Liu
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, 528 Zhangheng Road, Pudong New District, Shanghai 201203, China
| | - Cheng Liu
- Laboratory of Liver Disease, Department of Infectious Disease, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
- Experimental Center, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| |
Collapse
|
44
|
Schneider KM, Mohs A, Gui W, Galvez EJC, Candels LS, Hoenicke L, Muthukumarasamy U, Holland CH, Elfers C, Kilic K, Schneider CV, Schierwagen R, Strnad P, Wirtz TH, Marschall HU, Latz E, Lelouvier B, Saez-Rodriguez J, de Vos W, Strowig T, Trebicka J, Trautwein C. Imbalanced gut microbiota fuels hepatocellular carcinoma development by shaping the hepatic inflammatory microenvironment. Nat Commun 2022; 13:3964. [PMID: 35803930 PMCID: PMC9270328 DOI: 10.1038/s41467-022-31312-5] [Citation(s) in RCA: 115] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 06/13/2022] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer-related deaths worldwide, and therapeutic options for advanced HCC are limited. Here, we observe that intestinal dysbiosis affects antitumor immune surveillance and drives liver disease progression towards cancer. Dysbiotic microbiota, as seen in Nlrp6-/- mice, induces a Toll-like receptor 4 dependent expansion of hepatic monocytic myeloid-derived suppressor cells (mMDSC) and suppression of T-cell abundance. This phenotype is transmissible via fecal microbiota transfer and reversible upon antibiotic treatment, pointing to the high plasticity of the tumor microenvironment. While loss of Akkermansia muciniphila correlates with mMDSC abundance, its reintroduction restores intestinal barrier function and strongly reduces liver inflammation and fibrosis. Cirrhosis patients display increased bacterial abundance in hepatic tissue, which induces pronounced transcriptional changes, including activation of fibro-inflammatory pathways as well as circuits mediating cancer immunosuppression. This study demonstrates that gut microbiota closely shapes the hepatic inflammatory microenvironment opening approaches for cancer prevention and therapy.
Collapse
Affiliation(s)
- Kai Markus Schneider
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Antje Mohs
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Wenfang Gui
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Eric J C Galvez
- Helmholtz Centre for Infection Research, Braunschweig, Germany and Hannover Medical School, Hannover, Germany
| | | | - Lisa Hoenicke
- Helmholtz Centre for Infection Research, Braunschweig, Germany and Hannover Medical School, Hannover, Germany
| | - Uthayakumar Muthukumarasamy
- Helmholtz Centre for Infection Research, Braunschweig, Germany and Hannover Medical School, Hannover, Germany
| | - Christian H Holland
- Institute for Computational Biomedicine, Bioquant, Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Heidelberg, Germany
- Joint Research Centre for Computational Biomedicine (JRC-COMBINE), RWTH Aachen University, Faculty of Medicine, Aachen, Germany
| | - Carsten Elfers
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Konrad Kilic
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Carolin Victoria Schneider
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
- The Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Robert Schierwagen
- European Foundation for the Study of Chronic Liver Failure (EF-CLIF), 08021, Barcelona, Spain
- Translational Hepatology, Department of Internal Medicine I, Goethe University Frankfurt, 60323, Frankfurt, Germany
| | - Pavel Strnad
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Theresa H Wirtz
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Hanns-Ulrich Marschall
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Eicke Latz
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127, Bonn, Germany
- Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA, 01655, USA
- German Center for Neurodegenerative Diseases, 53127, Bonn, Germany
| | | | - Julio Saez-Rodriguez
- Institute for Computational Biomedicine, Bioquant, Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Heidelberg, Germany
- Joint Research Centre for Computational Biomedicine (JRC-COMBINE), RWTH Aachen University, Faculty of Medicine, Aachen, Germany
| | - Willem de Vos
- Laboratory of Microbiology, Wageningen University, 6708 WE, Wageningen, The Netherlands
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, P.O. Box 63, 00014, Helsinki, Finland
| | - Till Strowig
- Helmholtz Centre for Infection Research, Braunschweig, Germany and Hannover Medical School, Hannover, Germany
| | - Jonel Trebicka
- European Foundation for the Study of Chronic Liver Failure (EF-CLIF), 08021, Barcelona, Spain
- Translational Hepatology, Department of Internal Medicine I, Goethe University Frankfurt, 60323, Frankfurt, Germany
| | - Christian Trautwein
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany.
| |
Collapse
|
45
|
Human Blood Bacteriome: Eubiotic and Dysbiotic States in Health and Diseases. Cells 2022; 11:cells11132015. [PMID: 35805098 PMCID: PMC9265464 DOI: 10.3390/cells11132015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 06/07/2022] [Accepted: 06/21/2022] [Indexed: 01/27/2023] Open
Abstract
The human gut microbiome is acknowledged as being associated with homeostasis and the pathogenesis of several diseases. Conventional culture techniques are limited in that they cannot culture the commensals; however, next-generation sequencing has facilitated the discovery of the diverse and delicate microbial relationship in body sites and blood. Increasing evidence regarding the blood microbiome has revolutionized the concept of sterility and germ theory in circulation. Among the types of microbial communities in the blood, bacteriomes associated with many health conditions have been thoroughly investigated. Blood bacterial profiles in healthy subjects are identified as the eubiotic blood bacteriome, whereas the dysbiotic blood bacteriome represents the change in bacterial characteristics in subjects with diseases showing deviations from the eubiotic profiles. The blood bacterial characteristics in each study are heterogeneous; thus, the association between eubiotic and dysbiotic blood bacteriomes and health and disease is still debatable. Thereby, this review aims to summarize and discuss the evidence concerning eubiotic and dysbiotic blood bacteriomes characterized by next-generation sequencing in human studies. Knowledge pertaining to the blood bacteriome will transform the concepts around health and disease in humans, facilitating clinical implementation in the near future.
Collapse
|
46
|
Whole-Genome Sequencing Reveals Age-Specific Changes in the Human Blood Microbiota. J Pers Med 2022; 12:jpm12060939. [PMID: 35743724 PMCID: PMC9225573 DOI: 10.3390/jpm12060939] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/03/2022] [Accepted: 06/06/2022] [Indexed: 11/17/2022] Open
Abstract
Based on several reports that indicate the presence of blood microbiota in patients with diseases, we became interested in identifying the presence of bacteria in the blood of healthy individuals. Using 37 samples from 5 families, we extracted sequences that were not mapped to the human reference genome and mapped them to the bacterial reference genome for characterization. Proteobacteria account for more than 95% of the blood microbiota. The results of clustering by means of principal component analysis showed similar patterns for each age group. We observed that the class Gammaproteobacteria was significantly higher in the elderly group (over 60 years old), whereas the arcsine square root-transformed relative abundance of the classes Alphaproteobacteria, Deltaproteobacteria, and Clostridia was significantly lower (p < 0.05). In addition, the diversity among the groups showed a significant difference (p < 0.05) in the elderly group. This result provides meaningful evidence of a consistent phenomenon that chronic diseases associated with aging are accompanied by metabolic endotoxemia and chronic inflammation.
Collapse
|
47
|
Guo X, Tang P, Hou C, Chong L, Zhang X, Liu P, Chen L, Liu Y, Zhang L, Li R. Integrated Microbiome and Host Transcriptome Profiles Link Parkinson’s Disease to Blautia Genus: Evidence From Feces, Blood, and Brain. Front Microbiol 2022; 13:875101. [PMID: 35722294 PMCID: PMC9204254 DOI: 10.3389/fmicb.2022.875101] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 04/20/2022] [Indexed: 01/01/2023] Open
Abstract
A link between the gut microbiome and Parkinson’s disease (PD) has been intensively studied, and more than 100 differential genera were identified across the studies. However, the predominant genera contributing to PD remain poorly understood. Inspired by recent advances showing microbiota distribution in the blood and brain, we, here, comprehensively investigated currently available fecal microbiome data (1,914 samples) to identify significantly altered genera, which were further validated by comparison to the results from microbiome analysis of blood (85 samples) and brain (268 samples). Our data showed that the composition of fecal microbiota was different from that of blood and brain. We found that Blautia was the unique genus consistently depleted across feces, blood, and brain samples of PD patients (P < 0.05), despite using rigorous criteria to remove contaminants. Moreover, enrichment analyses revealed that host genes correlated with Blautia genus abundance were mainly involved in mitochondrial function and energy metabolism, and mapped to neurodegenerative diseases (NDDs) and metabolic diseases. A random forest classifier constructed with fecal microbiota data demonstrated that Blautia genus was an important feature contributing to discriminating PD patients from controls [receiver operating characteristic (ROC)-area under curve (AUC) = 0.704, precision-recall curve (PRC)-AUC = 0.787]. Through the integration of microbiome and transcriptome, our study depicted microbial profiles in the feces, blood, and brain of PD patients, and identified Blautia genus as a potential genus linked to PD. Further studies are greatly encouraged to determine the role of Blautia genus in the pathogenesis of PD.
Collapse
Affiliation(s)
- Xingzhi Guo
- Department of Geriatric Neurology, Shaanxi Provincial People’s Hospital, Xi’an, China
- Shaanxi Provincial Clinical Research Center for Geriatric Medicine, Xi’an, China
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an, China
| | - Peng Tang
- Department of Geriatric Neurology, Shaanxi Provincial People’s Hospital, Xi’an, China
- Shaanxi Provincial Clinical Research Center for Geriatric Medicine, Xi’an, China
| | - Chen Hou
- Department of Geriatric Neurology, Shaanxi Provincial People’s Hospital, Xi’an, China
- Shaanxi Provincial Clinical Research Center for Geriatric Medicine, Xi’an, China
| | - Li Chong
- Department of Geriatric Neurology, Shaanxi Provincial People’s Hospital, Xi’an, China
- Shaanxi Provincial Clinical Research Center for Geriatric Medicine, Xi’an, China
| | - Xin Zhang
- Department of Geriatric Neurology, Shaanxi Provincial People’s Hospital, Xi’an, China
- Shaanxi Provincial Clinical Research Center for Geriatric Medicine, Xi’an, China
| | - Peng Liu
- Department of Geriatric Neurology, Shaanxi Provincial People’s Hospital, Xi’an, China
- Shaanxi Provincial Clinical Research Center for Geriatric Medicine, Xi’an, China
| | - Li Chen
- Department of Geriatric Neurology, Shaanxi Provincial People’s Hospital, Xi’an, China
- Shaanxi Provincial Clinical Research Center for Geriatric Medicine, Xi’an, China
| | - Yue Liu
- Department of Geriatric Neurology, Shaanxi Provincial People’s Hospital, Xi’an, China
- Shaanxi Provincial Clinical Research Center for Geriatric Medicine, Xi’an, China
| | - Lina Zhang
- Department of Geriatric Neurology, Shaanxi Provincial People’s Hospital, Xi’an, China
- Shaanxi Provincial Clinical Research Center for Geriatric Medicine, Xi’an, China
| | - Rui Li
- Department of Geriatric Neurology, Shaanxi Provincial People’s Hospital, Xi’an, China
- Shaanxi Provincial Clinical Research Center for Geriatric Medicine, Xi’an, China
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an, China
- *Correspondence: Rui Li,
| |
Collapse
|
48
|
Khan I, Khan I, Kakakhel MA, Xiaowei Z, Ting M, Ali I, Fei Y, Jianye Z, Zhiqiang L, Lizhe A. Comparison of Microbial Populations in the Blood of Patients With Myocardial Infarction and Healthy Individuals. Front Microbiol 2022; 13:845038. [PMID: 35694288 PMCID: PMC9176212 DOI: 10.3389/fmicb.2022.845038] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 03/31/2022] [Indexed: 01/03/2023] Open
Abstract
Increased bacterial translocation in the gut and bloodstream infections are both major comorbidities of heart failure and myocardial infarction (MI). However, the alterations in the microbiome of the blood of patients with MI remain unclear. To test this hypothesis, we conducted this case-control study to explore the microbiota compositions in the blood of Chinese patients with MI. Using high-throughput Illumina HiSeq sequencing targeting the V3–V4 region of the 16S ribosomal RNA (rRNA) gene, the microbiota communities in the blood of 29 patients with MI and 29 healthy controls were examined. In addition, the relationship between the blood microbiome and clinical features of MI was investigated. This study revealed a significant reduction in alpha diversity (Shannon index) in the MI group compared with the healthy controls. Also, a significant difference was detected in the structure and richness between the patients with MI and healthy controls. The members of the phylum Actinobacteria, class Actinobacteria, order Bifdobacteriales, family Bifidobacteriaceae, and genus Bifidobacterium were significantly abundant in the MI group, while the members of the phylum Bacteroidetes, class Bacteroidia, and order Bacteroidales were significantly enriched in the healthy controls (p < 0.05). Moreover, the functional analysis revealed a significant variation between both groups. For instance, the enrichment of genes involved in the metabolism pathways of three amino acids decreased, that is, nucleotide transport and metabolism, coenzyme transport and metabolism, and lipid transport and metabolism, among others. Our study will contribute to a better knowledge of the microbiota of blood, which will further lead to improved MI diagnosis and therapy. Further study is needed to determine the role of the blood microbiota in human health and disease.
Collapse
Affiliation(s)
- Ikram Khan
- School of Life Sciences, Lanzhou University, Lanzhou, China
- School of Stomatology, Northwest Minzu University, Lanzhou, China
| | - Imran Khan
- Department of Microbiology, Khyber Medical University Peshawar, Peshawar, Pakistan
| | | | | | - Mao Ting
- Lanzhou University Second Hospital, Lanzhou, China
| | - Ikram Ali
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Yu Fei
- School of Stomatology, Northwest Minzu University, Lanzhou, China
| | - Zhou Jianye
- School of Stomatology, Northwest Minzu University, Lanzhou, China
| | - Li Zhiqiang
- School of Stomatology, Northwest Minzu University, Lanzhou, China
- *Correspondence: Li Zhiqiang
| | - An Lizhe
- School of Life Sciences, Lanzhou University, Lanzhou, China
- An Lizhe
| |
Collapse
|
49
|
Sumida K, Han Z, Chiu CY, Mims TS, Bajwa A, Demmer RT, Datta S, Kovesdy CP, Pierre JF. Circulating Microbiota in Cardiometabolic Disease. Front Cell Infect Microbiol 2022; 12:892232. [PMID: 35592652 PMCID: PMC9110890 DOI: 10.3389/fcimb.2022.892232] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 04/11/2022] [Indexed: 12/14/2022] Open
Abstract
The rapid expansion of microbiota research has significantly advanced our understanding of the complex interactions between gut microbiota and cardiovascular, metabolic, and renal system regulation. Low-grade chronic inflammation has long been implicated as one of the key mechanisms underlying cardiometabolic disease risk and progression, even before the insights provided by gut microbiota research in the past decade. Microbial translocation into the bloodstream can occur via different routes, including through the oral and/or intestinal mucosa, and may contribute to chronic inflammation in cardiometabolic disease. Among several gut-derived products identifiable in the systemic circulation, bacterial endotoxins and metabolites have been extensively studied, however recent advances in microbial DNA sequencing have further allowed us to identify highly diverse communities of microorganisms in the bloodstream from an -omics standpoint, which is termed "circulating microbiota." While detecting microorganisms in the bloodstream was historically considered as an indication of infection, evidence on the circulating microbiota is continually accumulating in various patient populations without clinical signs of infection and even in otherwise healthy individuals. Moreover, both quantitative and compositional alterations of the circulating microbiota have recently been implicated in the pathogenesis of chronic inflammatory conditions, potentially through their immunostimulatory, atherogenic, and cardiotoxic properties. In this mini review, we aim to provide recent evidence on the characteristics and roles of circulating microbiota in several cardiometabolic diseases, such as type 2 diabetes, cardiovascular disease, and chronic kidney disease, with highlights of our emerging findings on circulating microbiota in patients with end-stage kidney disease undergoing hemodialysis.
Collapse
Affiliation(s)
- Keiichi Sumida
- Division of Nephrology, Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States,*Correspondence: Keiichi Sumida,
| | - Zhongji Han
- Division of Nephrology, Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Chi-Yang Chiu
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Tahliyah S. Mims
- Department of Nutritional Sciences, College of Agriculture and Life Science, University of Wisconsin-Madison, Madison, WI, United States
| | - Amandeep Bajwa
- Transplant Research Institute, James D. Eason Transplant Institute, Department of Surgery, School of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Ryan T. Demmer
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, United States,Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, United States
| | - Susmita Datta
- Department of Biostatistics, University of Florida, Gainesville, FL, United States
| | - Csaba P. Kovesdy
- Division of Nephrology, Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States,Nephrology Section, Memphis Veterans Affairs (VA) Medical Center, Memphis, TN, United States
| | - Joseph F. Pierre
- Department of Nutritional Sciences, College of Agriculture and Life Science, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
50
|
Luo Z, Ji Y, Zhang D, Gao H, Jin Z, Yang M, Ying W. Microbial DNA enrichment promotes liver steatosis and fibrosis in the course of non-alcoholic steatohepatitis. Acta Physiol (Oxf) 2022; 235:e13827. [PMID: 35500155 DOI: 10.1111/apha.13827] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/27/2022] [Accepted: 04/29/2022] [Indexed: 12/13/2022]
Abstract
AIM Low-grade inflammation is the hallmark of non-alcoholic fatty liver diseases (NAFLD) and non-alcoholic steatohepatitis (NASH). The leakage of microbiota-derived products can contribute to liver inflammation during NAFLD/NASH development. Here, we assessed the roles of gut microbial DNA-containing extracellular vesicles (mEVs) in regulating liver cellular abnormalities in the course of NAFLD/NASH. METHODS We performed studies with Vsig4-/- , C3-/- , cGAS-/- , and their wild-type littermate mice. Vsig4+ macrophage population and bacterial DNA abundance were examined in both mouse and human liver by either flow cytometric or immunohistochemistry analysis. Gut mEVs were adoptively transferred into Vsig4-/- , C3-/- , cGAS-/- , or littermate WT mice, and hepatocyte inflammation and HSC fibrogenic activation were measured in these mice. RESULTS Non-alcoholic fatty liver diseases and non-alcoholic steatohepatitis development was concomitant with a diminished liver Vsig4+ macrophage population and a marked bacterial DNA enrichment in both hepatocytes and HSCs. In the absence of Vsig4+ macrophages, gut mEVs translocation led to microbial DNA accumulation in hepatocytes and HSCs, resulting elevated hepatocyte inflammation and HSC fibrogenic activation. In contrast, in lean WT mice, Vsig4+ macrophages remove gut mEVs from bloodstream through a C3-dependent opsonization mechanism and prevent the infiltration of gut mEVs into hepatic cells. Additionally, Vsig4-/- mice more quickly developed significant liver steatosis and fibrosis than WT mice after Western diet feeding. In vitro treatment with NASH mEVs triggered hepatocyte inflammation and HSC fibrogenic activation. Microbial DNAs are key cargo for the effects of gut mEVs by activating cGAS/STING. CONCLUSION Accumulation of microbial DNAs fuels the development of NAFLD/NASH-associated liver abnormalities.
Collapse
Affiliation(s)
- Zhenlong Luo
- Division of Endocrinology & Metabolism Department of Medicine University of California San Diego California USA
- Department of Gastroenterology Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Yudong Ji
- Division of Endocrinology & Metabolism Department of Medicine University of California San Diego California USA
- Department of Anesthesiology Institute of Anesthesiology and Critical Care Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Dinghong Zhang
- Division of Endocrinology & Metabolism Department of Medicine University of California San Diego California USA
| | - Hong Gao
- Division of Endocrinology & Metabolism Department of Medicine University of California San Diego California USA
| | - Zhongmou Jin
- Division of Biological Sciences University of California San Diego California USA
| | - Meixiang Yang
- Pediatric Diabetes Research Center Department of Pediatrics University of California San Diego California USA
- Zhuhai Institute of Translational Medicine Zhuhai People’s Hospital Affiliated with Jinan University Biomedical Translational Research Institute Jinan University Guangzhou China
| | - Wei Ying
- Division of Endocrinology & Metabolism Department of Medicine University of California San Diego California USA
| |
Collapse
|