1
|
Petitclerc I, Perron J, Dugas C, Mayer T, Raymond F, Di Marzo V, Veilleux A, Robitaille J. Association between gestational diabetes mellitus, maternal health and diet, and gut microbiota in mother-infant dyads. BMC Pregnancy Childbirth 2025; 25:486. [PMID: 40275186 PMCID: PMC12023395 DOI: 10.1186/s12884-025-07584-2] [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: 01/08/2025] [Accepted: 04/08/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Gestational diabetes mellitus (GDM) increasingly affects women and predisposes both mothers and their infants to short- and long-term health consequences. Emerging research links GDM to maternal gut microbiota dysbiosis. However, the impact of GDM on the infant gut microbiota remains unclear. This cross-sectional study aims to explore potential associations between GDM and the gut microbiota in mothers and their infants, as well as correlations between maternal diet, cardiometabolic profile, and gut microbiota composition. METHODS Gut microbiota taxonomic composition was characterized by 16S rRNA gene sequencing on fecal samples collected at 2 months postpartum from 28 mothers, including 17 with (GDM+) and 11 without (GDM-) GDM, as well as 30 infants, 17 GDM + and 13 GDM-. Variations in overall composition and specific taxa between GDM + and GDM- were assessed. Correlations between maternal cardiometabolic profile, dietary intakes, and taxa were performed. RESULTS GDM was associated with the overall composition of gut microbiota between GDM + and GDM- in the maternal group, but not in infants. No statistically significant difference in alpha diversity between groups was found in either mothers or infants. However, 14 taxa showed significantly different abundance between GDM + and GDM- mothers, and 4 taxa differed in infants. Specific taxa at the family rank were correlated with maternal dietary and cardiometabolic variables in both mothers and infants. CONCLUSIONS GDM exposition was associated with gut microbiota composition in both mothers and infants at two months postpartum. This study enhances our understanding of how maternal health could be linked with the gut microbiota of mothers and their infants. TRIAL REGISTRATION NCT02872402 (2016-08-04, https://clinicaltrials.gov/study/NCT02872402?term=NCT02872402&rank=1 ) and NCT04263675 (2020-02-07, https://clinicaltrials.gov/study/NCT04263675?term=NCT04263675&rank=1 ).
Collapse
Affiliation(s)
- Isabelle Petitclerc
- Centre de recherche Nutrition, Santé et Société (NUTRISS), Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec City, QC, G1V 0A6, Canada
- School of Nutrition, Université Laval, Quebec City, QC, G1V 0A6, Canada
| | - Julie Perron
- Centre de recherche Nutrition, Santé et Société (NUTRISS), Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec City, QC, G1V 0A6, Canada
| | - Camille Dugas
- Centre de recherche Nutrition, Santé et Société (NUTRISS), Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec City, QC, G1V 0A6, Canada
| | - Thomas Mayer
- Centre de recherche Nutrition, Santé et Société (NUTRISS), Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec City, QC, G1V 0A6, Canada
- Canada Research Excellence Chair in the Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), Université Laval, Quebec City, QC, G1V 0A6, Canada
| | - Frédéric Raymond
- Centre de recherche Nutrition, Santé et Société (NUTRISS), Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec City, QC, G1V 0A6, Canada
- School of Nutrition, Université Laval, Quebec City, QC, G1V 0A6, Canada
- Canada Research Excellence Chair in the Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), Université Laval, Quebec City, QC, G1V 0A6, Canada
| | - Vincenzo Di Marzo
- Centre de recherche Nutrition, Santé et Société (NUTRISS), Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec City, QC, G1V 0A6, Canada
- School of Nutrition, Université Laval, Quebec City, QC, G1V 0A6, Canada
- Canada Research Excellence Chair in the Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), Université Laval, Quebec City, QC, G1V 0A6, Canada
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (IUCPQ), Université Laval, Quebec City, QC, G1V 4G5, Canada
| | - Alain Veilleux
- Centre de recherche Nutrition, Santé et Société (NUTRISS), Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec City, QC, G1V 0A6, Canada
- School of Nutrition, Université Laval, Quebec City, QC, G1V 0A6, Canada
- Canada Research Excellence Chair in the Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), Université Laval, Quebec City, QC, G1V 0A6, Canada
| | - Julie Robitaille
- Centre de recherche Nutrition, Santé et Société (NUTRISS), Institute of Nutrition and Functional Foods (INAF), Université Laval, Quebec City, QC, G1V 0A6, Canada.
- School of Nutrition, Université Laval, Quebec City, QC, G1V 0A6, Canada.
| |
Collapse
|
2
|
Diacova T, Cifelli CJ, Davis CD, Holscher HD, Kable ME, Lampe JW, Latulippe ME, Swanson KS, Karl JP. Best Practices and Considerations for Conducting Research on Diet-Gut Microbiome Interactions and Their Impact on Health in Adult Populations: An Umbrella Review. Adv Nutr 2025; 16:100419. [PMID: 40180180 DOI: 10.1016/j.advnut.2025.100419] [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: 08/07/2024] [Revised: 03/19/2025] [Accepted: 03/27/2025] [Indexed: 04/05/2025] Open
Abstract
Diet modulates gut microbiome composition and function. However, determining causal links between diet-gut microbiome interactions and human health is complicated by inconsistencies in the evidence, arising partially from variability in research methods and reporting. Widespread adoption of standardized best practices would advance the field but require those practices to be identified, consolidated, and discussed. This umbrella review aimed to identify recommended best practices, define existing gaps, and collate considerations for conducting research on diet-gut microbiome interactions and their impact on human health outcomes. Reviews meeting inclusion criteria and published after 2013 were identified using a systematic search. Recommendations, considerations, and gaps relating to the best practices associated with study design, participant selection, dietary intervention/assessment, biological sample collection, and data analysis and reporting were extracted and consolidated. Eight narrative reviews were included. Several general points of agreement were identified, and a recurring theme was that best practices are dependent upon the research aims, outcomes, and feasibility. Multiple gaps were also identified. Some, such as suboptimal diet assessment methods and lack of validated dietary intake biomarkers, are particularly relevant to nutrition science. Others, including defining a "healthy" gut microbiome and the absence of standardized sample and data collection/analysis protocols, were relevant specifically to gut microbiome research. Gaps specific to diet-gut microbiome research include the underrepresentation of microbiome-modulating dietary components in food databases, lack of knowledge regarding interventions eliciting changes in the gut microbiome to confer health benefits, lack of in situ measurement methods, and the need to further develop and refine statistical approaches for integrating diet and gut microbiome data. Future research and cross-disciplinary exchange will address these gaps and evolve the best practices. In the interim, the best practices and considerations discussed herein, and the publications from which that information was extracted provide a roadmap for conducting diet-gut microbiome research. This trial was registered at PROSPERO as CRD42023437645.
Collapse
Affiliation(s)
- Tatiana Diacova
- Graduate Group in Nutritional Biology, University of California Davis, Davis, CA, United States
| | | | - Cindy D Davis
- Agricultural Research Service, United States Department of Agriculture, Beltsville, MD, United States
| | - Hannah D Holscher
- Department of Food Science and Human Nutrition, University of Illinois Urbana-Champaign, Urbana, IL, United States; Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, United States
| | - Mary E Kable
- Agricultural Research Service, United States Department of Agriculture, Beltsville, MD, United States
| | - Johanna W Lampe
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Marie E Latulippe
- Institute for the Advancement of Food and Nutrition Sciences, Washington, DC, United States
| | - Kelly S Swanson
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, United States; Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, United States
| | - J Philip Karl
- Military Nutrition Division, U.S. Army Research Institute of Environmental Medicine, Natick, MA, United States.
| |
Collapse
|
3
|
Westmark CJ. Soy-based purified ingredient diet affects mouse gut permeability and the microbiome in fragile X mice. Front Mol Neurosci 2025; 18:1520211. [PMID: 40190341 PMCID: PMC11968763 DOI: 10.3389/fnmol.2025.1520211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 02/28/2025] [Indexed: 04/09/2025] Open
Abstract
Introduction Gastrointestinal problems including vomiting, reflux, flatulence, diarrhea, constipation and colic are common comorbidities in fragile X syndrome. There is accumulating evidence suggesting that leaky gut syndrome causes neurological phenotypes. Although fragile X messenger ribonucleoprotein is ubiquitously expressed, there is a dearth of knowledge regarding its role outside of the brain including effects on gut dysfunction in fragile X. The aim of this study was to generate novel data on gastrointestinal barrier function and the gut microbiome in response to Fmr1 genotype, sex and diet in mice. Methods Fmr1KO male mice and littermate controls in an FVB background were maintained on two purified ingredient diets (AIN-93G with casein protein versus soy protein isolate) versus two standard chows (Teklad 2019 with wheat, corn and yeast protein versus Purina 5015 with wheat, soy, corn, yeast and whey protein sources). Gut permeability was quantified by FITC-dextran levels in blood plasma. The cecal microbiome was identified by 16S rRNA sequencing. In addition, gut permeability was tested in Fmr1KO mice in the C57BL/6 J background maintained on casein- and soy protein isolate-based AIN-93G versus Teklad 2019. Results Knockout of the Fmr1 gene in FVB mice did not affect gut permeability. Soy protein isolate-based AIN-93G increased gut permeability. Beta-diversity of the cecal microbiome was significantly altered as a function of the four test diets. Akkermansia_muciniphila was increased in Fmr1KO mice fed AIN-93G while unnamed species within the genus Anaerovorax and family Ruminococcaceae were increased and the order Clostridales decreased in Fmr1KO mice fed AIN-93G/soy. Fmr1KO mice in the C57BL/6 J background exhibited increased gut permeability in response to soy protein. Discussion These findings regarding the effects of diet on gut permeability and the microbiome have important implications for experimental design. Single-source diets are ubiquitously used to maintain laboratory animals for medical research and feed details are frequently not reported in publications. Diet/phenotype interactions could have a large impact on inter-laboratory replicability in premedical research. For infants with fragile X, early-life diet could impact the severity of disease outcomes.
Collapse
Affiliation(s)
- Cara J. Westmark
- Department of Neurology, University of Wisconsin, Madison, WI, United States
- Molecular Environmental Toxicology Center, University of Wisconsin, Madison, WI, United States
| |
Collapse
|
4
|
Chavarria X, Park HS, Oh S, Kang D, Choi JH, Kim M, Cho YH, Yi MH, Kim JY. Using gut microbiome metagenomic hypervariable features for diabetes screening and typing through supervised machine learning. Microb Genom 2025; 11:001365. [PMID: 40063675 PMCID: PMC11893737 DOI: 10.1099/mgen.0.001365] [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: 08/08/2024] [Accepted: 01/24/2025] [Indexed: 03/14/2025] Open
Abstract
Diabetes mellitus is a complex metabolic disorder and one of the fastest-growing global public health concerns. The gut microbiota is implicated in the pathophysiology of various diseases, including diabetes. This study utilized 16S rRNA metagenomic data from a volunteer citizen science initiative to investigate microbial markers associated with diabetes status (positive or negative) and type (type 1 or type 2 diabetes mellitus) using supervised machine learning (ML) models. The diversity of the microbiome varied according to diabetes status and type. Differential microbial signatures between diabetes types and negative group revealed an increased presence of Brucellaceae, Ruminococcaceae, Clostridiaceae, Micrococcaceae, Barnesiellaceae and Fusobacteriaceae in subjects with diabetes type 1, and Veillonellaceae, Streptococcaceae and the order Gammaproteobacteria in subjects with diabetes type 2. The decision tree, elastic net, random forest (RF) and support vector machine with radial kernel ML algorithms were trained to screen and type diabetes based on microbial profiles of 76 subjects with type 1 diabetes, 366 subjects with type 2 diabetes and 250 subjects without diabetes. Using the 1000 most variable features, tree-based models were the highest-performing algorithms. The RF screening models achieved the best performance, with an average area under the receiver operating characteristic curve (AUC) of 0.76, although all models lacked sensitivity. Reducing the dataset to 500 features produced an AUC of 0.77 with sensitivity increasing by 74% from 0.46 to 0.80. Model performance improved for the classification of negative-status and type 2 diabetes. Diabetes type models performed best with 500 features, but the metric performed poorly across all model iterations. ML has the potential to facilitate early diagnosis of diabetes based on microbial profiles of the gut microbiome.
Collapse
Affiliation(s)
- Xavier Chavarria
- Department of Tropical Medicine, Institute of Tropical Medicine, Arthropods of Medical Importance Resource Bank, Yonsei University College of Medicine, Yonsei-ro 50-1, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Hyun Seo Park
- Department of Tropical Medicine, Institute of Tropical Medicine, Arthropods of Medical Importance Resource Bank, Yonsei University College of Medicine, Yonsei-ro 50-1, Seodaemun-gu, Seoul 03722, Republic of Korea
- Department of Systems Biology, Yonsei University College of Life Science and Biotechnology, Yonsei-ro 50-1, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Singeun Oh
- Department of Tropical Medicine, Institute of Tropical Medicine, Arthropods of Medical Importance Resource Bank, Yonsei University College of Medicine, Yonsei-ro 50-1, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Dongjun Kang
- Department of Tropical Medicine, Institute of Tropical Medicine, Arthropods of Medical Importance Resource Bank, Yonsei University College of Medicine, Yonsei-ro 50-1, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Jun Ho Choi
- Department of Tropical Medicine, Institute of Tropical Medicine, Arthropods of Medical Importance Resource Bank, Yonsei University College of Medicine, Yonsei-ro 50-1, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Myungjun Kim
- Department of Tropical Medicine, Institute of Tropical Medicine, Arthropods of Medical Importance Resource Bank, Yonsei University College of Medicine, Yonsei-ro 50-1, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Yoon Hee Cho
- Department of Tropical Medicine, Institute of Tropical Medicine, Arthropods of Medical Importance Resource Bank, Yonsei University College of Medicine, Yonsei-ro 50-1, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Myung-hee Yi
- Department of Tropical Medicine, Institute of Tropical Medicine, Arthropods of Medical Importance Resource Bank, Yonsei University College of Medicine, Yonsei-ro 50-1, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Ju Yeong Kim
- Department of Tropical Medicine, Institute of Tropical Medicine, Arthropods of Medical Importance Resource Bank, Yonsei University College of Medicine, Yonsei-ro 50-1, Seodaemun-gu, Seoul 03722, Republic of Korea
| |
Collapse
|
5
|
Guan Y, Chen Y, Lin R, Mo T, Li S, Cao Y, Yin T, Diao L, Li Y. Endometriosis: A new perspective on epigenetics and oxidative stress. J Reprod Immunol 2025; 169:104462. [PMID: 40010026 DOI: 10.1016/j.jri.2025.104462] [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: 10/08/2024] [Revised: 01/07/2025] [Accepted: 02/20/2025] [Indexed: 02/28/2025]
Abstract
As a complex chronic gynecological disorder characterized by multifaceted etiology involving genetics, environment, immunity and inflammation, endometriosis (EM) has long been a significant concern for women of reproductive age worldwide. This review aimed to comprehensively examine the interplay between epigenetics and oxidative stress (OS) in the pathogenesis of EM. Through the integration of cutting-edge research, the response of OS signals to epigenetic modifications was explored. The microbiome exerts an influence on this causal regulatory relationship, and these mechanisms collectively contribute to the pathophysiology of EM. Specifically, this article highlighted the roles of epigenetics and OS in EM and underscored the importance of the microbiome as a regulatory link. A discussion was also held on the future directions of biomarkers and precision medicine, including the application prospects of epigenetic and OS markers in the diagnosis and treatment decision-making of EM, and innovations in therapeutic strategies like targeting epigenetic modifications and antioxidant therapies. Moreover, this review emphasized the potential of multi-omics integrated analysis to deepen the understanding of the disease, guide future therapeutic strategies and promote personalized medicine.
Collapse
Affiliation(s)
- Yu Guan
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Yawen Chen
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Shenzhen Zhongshan Obstetrics & Gynecology Hospital (formerly Shenzhen Zhongshan Urology Hospital), China; Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-implantation, China
| | - Rong Lin
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Shenzhen Zhongshan Obstetrics & Gynecology Hospital (formerly Shenzhen Zhongshan Urology Hospital), China; Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-implantation, China
| | - Tinghui Mo
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Shiyu Li
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Ying Cao
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Tailang Yin
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China.
| | - Lianghui Diao
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Shenzhen Zhongshan Obstetrics & Gynecology Hospital (formerly Shenzhen Zhongshan Urology Hospital), China; Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-implantation, China.
| | - Yuye Li
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Shenzhen Zhongshan Obstetrics & Gynecology Hospital (formerly Shenzhen Zhongshan Urology Hospital), China; Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-implantation, China.
| |
Collapse
|
6
|
Francini E, Badillo Pazmay GV, Fumarola S, Procopio AD, Olivieri F, Marchegiani F. Bi-Directional Relationship Between Bile Acids (BAs) and Gut Microbiota (GM): UDCA/TUDCA, Probiotics, and Dietary Interventions in Elderly People. Int J Mol Sci 2025; 26:1759. [PMID: 40004221 PMCID: PMC11855466 DOI: 10.3390/ijms26041759] [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: 12/20/2024] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
The gut microbiota (GM), the set of microorganisms that colonizes our intestinal tract, can undergo many changes, some of which are age related. Several studies have shown the importance of maintaining a healthy GM for a good quality of life. In the elderly, maintaining a good GM may become a real defense against infection by pathogens, such as C. difficile. In addition to the GM, bile acids (BAs) have been shown to provide an additional defense mechanism against the proliferation of pathogenic bacteria and to regulate bacterial colonization of the gut. BAs are molecules produced in the host liver and secreted with the bile into the digestive tract, and they are necessary for the digestion of dietary lipids. In the gut, host-produced BAs are metabolized by commensal bacteria to secondary BAs. In general GM and host organisms interact in many ways. This review examines the relationship between GM, BAs, aging, and possible new approaches such as dietary interventions, administration of ursodesoxycholic acid/tauroursodesoxycholic acid (UDCA/TUDCA), and probiotics to enrich the microbial consortia of the GM in the elderly and achieve a eubiotic state necessary for maintaining good health. The presence of Firmicutes and Actinobacteria together with adequate levels of secondary BAs would provide protection and improve the frailty state in the elderly. In fact, an increase in secondary BAs has been observed in centenarians who have reached old age without serious health issues, which may justify their active role in achieving longevity.
Collapse
Affiliation(s)
- Emanuele Francini
- Clinic of Laboratory and Precision Medicine, IRCCS INRCA, 60121 Ancona, Italy; (E.F.); (A.D.P.)
| | - Gretta V. Badillo Pazmay
- Advanced Technology Center for Aging Research, IRCCS INRCA, 60121 Ancona, Italy; (G.V.B.P.); (S.F.); (F.O.)
| | - Stefania Fumarola
- Advanced Technology Center for Aging Research, IRCCS INRCA, 60121 Ancona, Italy; (G.V.B.P.); (S.F.); (F.O.)
| | - Antonio Domenico Procopio
- Clinic of Laboratory and Precision Medicine, IRCCS INRCA, 60121 Ancona, Italy; (E.F.); (A.D.P.)
- Laboratory of Experimental Pathology, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, 60100 Ancona, Italy
| | - Fabiola Olivieri
- Advanced Technology Center for Aging Research, IRCCS INRCA, 60121 Ancona, Italy; (G.V.B.P.); (S.F.); (F.O.)
- Laboratory of Experimental Pathology, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, 60100 Ancona, Italy
| | - Francesca Marchegiani
- Clinic of Laboratory and Precision Medicine, IRCCS INRCA, 60121 Ancona, Italy; (E.F.); (A.D.P.)
| |
Collapse
|
7
|
Porcari S, Mullish BH, Asnicar F, Ng SC, Zhao L, Hansen R, O'Toole PW, Raes J, Hold G, Putignani L, Hvas CL, Zeller G, Koren O, Tun H, Valles-Colomer M, Collado MC, Fischer M, Allegretti J, Iqbal T, Chassaing B, Keller J, Baunwall SM, Abreu M, Barbara G, Zhang F, Ponziani FR, Costello SP, Paramsothy S, Kao D, Kelly C, Kupcinskas J, Youngster I, Franceschi F, Khanna S, Vehreschild M, Link A, De Maio F, Pasolli E, Miguez AB, Brigidi P, Posteraro B, Scaldaferri F, Stojanovic MR, Megraud F, Malfertheiner P, Masucci L, Arumugam M, Kaakoush N, Segal E, Bajaj J, Leong R, Cryan J, Weersma RK, Knight R, Guarner F, Shanahan F, Cani PD, Elinav E, Sanguinetti M, de Vos WM, El-Omar E, Dorè J, Marchesi J, Tilg H, Sokol H, Segata N, Cammarota G, Gasbarrini A, Ianiro G. International consensus statement on microbiome testing in clinical practice. Lancet Gastroenterol Hepatol 2025; 10:154-167. [PMID: 39647502 DOI: 10.1016/s2468-1253(24)00311-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/10/2024] [Accepted: 09/13/2024] [Indexed: 12/10/2024]
Abstract
There is growing interest in the potential exploitation of the gut microbiome as a diagnostic tool in medicine, but evidence supporting its clinical usefulness is scarce. An increasing number of commercial providers offer direct-to-consumer microbiome diagnostic tests without any consensus on their regulation or any proven value in clinical practice, which could result in considerable waste of individual and health-care resources and potential drawbacks in the clinical management of patients. We convened an international multidisciplinary expert panel to standardise best practices of microbiome testing for clinical implementation, including recommendations on general principles and minimum requirements for their provision, indications, pre-testing protocols, method of analyses, reporting of results, and potential clinical value. We also evaluated current knowledge gaps and future directions in this field. We aimed to establish a framework to regulate the provision of microbiome testing and minimise the use of inappropriate tests and pave the way for the evidence-based development and use of human microbiome diagnostics in clinical medicine.
Collapse
Affiliation(s)
- Serena Porcari
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Benjamin H Mullish
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, St Mary's Hospital Campus, Imperial College London, London, UK; Departments of Gastroenterology and Hepatology, St Mary's Hospital, Imperial College Healthcare NHS Trust, London, UK
| | | | - Siew C Ng
- Microbiota I-Center (MagIC), Hong Kong Special Administrative Region, China; Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; Li Ka Shing Institute of Health Sciences, State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Liping Zhao
- Department of Biochemistry and Microbiology, New Jersey Institute of Food, Nutrition and Health, Rutgers University, New Brunswick, NY, USA
| | - Richard Hansen
- Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee, UK
| | - Paul W O'Toole
- APC Microbiome Ireland, Department of Medicine, University College Cork, Cork, Ireland; School of Microbiology, University College Cork, Cork, Ireland
| | - Jeroen Raes
- Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Leuven, Belgium; VIB, Center for Microbiology, Leuven, Belgium
| | - Georgina Hold
- Microbiome Research Centre, University of New South Wales, Sydney, Australia
| | - Lorenza Putignani
- Unit of Microbiomics and Unit of Human Microbiome, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Christian Lodberg Hvas
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus Denmark
| | - Georg Zeller
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, Leiden, Netherlands; Center for Microbiome Analyses and Therapeutics, Leiden University Medical Center, Leiden, Netherlands; Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Omry Koren
- Azrieli Faculty of Medicine Bar-Ilan University, Safed, Israel
| | - Hein Tun
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Mireia Valles-Colomer
- Department CIBIO, University of Trento, Trento, Italy; MELIS Department, Pompeu Fabra University, Barcelona, Spain
| | - Maria Carmen Collado
- Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Valencia, Spain
| | - Monika Fischer
- Division of Gastroenterology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jessica Allegretti
- Division of Gastroenterology, Brigham and Women's Hospital, Boston, MA, USA
| | - Tariq Iqbal
- Department of Gastroenterology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK; Microbiome Treatment Centre, University of Birmingham, Edgbaston, UK
| | - Benoit Chassaing
- Microbiome-Host Interactions, Institut Pasteur, Université Paris Cité, INSERM, Paris, France
| | - Josbert Keller
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, Netherlands; Public Health Laboratory, Faculty of Medicine, University of Birmingham, Birmingham, UK
| | - Simon Mark Baunwall
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Maria Abreu
- Division of Gastroenterology, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Giovanni Barbara
- IRCCS Azienda Ospedaliero, University of Bologna, Bologna, Italy; Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Faming Zhang
- Medical Center for Digestive Diseases, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, China; Key Lab of Holistic Integrative Enterology, Nanjing Medical University, Nanjing, China
| | - Francesca Romana Ponziani
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Liver Unit-Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Sam P Costello
- Department of Gastroenterology, The Queen Elizabeth Hospital, Adelaide, South Australia, Australia; Faculty of Health and Medical Sciences, Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Sudarshan Paramsothy
- Department of Gastroenterology and Hepatology, Concord Repatriation General Hospital, Sydney, Australia; Concord Clinical School, University of Sydney, Sydney, Australia
| | - Dina Kao
- Edmonton FMT program, Division of Gastroenterology, University of Alberta, Edmonton, AB, Canada
| | - Colleen Kelly
- Division of Gastroenterology, Brigham and Women's Hospital, Boston, MA, USA
| | - Juozas Kupcinskas
- Gastroenterology Department and Institute for Digestive Research; Lithuanian University of Health Sciences Kaunas, Lithuania
| | - Ilan Youngster
- Division of Pediatrics and the Center for Microbiome Research, Shamir Medical Center, Israel, Faculty of Medical & Health Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | - Francesco Franceschi
- Department of Emergency Medicine, Fondazione Policlinico Universitario, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Sahil Khanna
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Maria Vehreschild
- Goethe University Frankfurt, University Hospital Frankfurt, Department II of Internal Medicine, Infectious Diseases, Frankfurt am Main, Germany
| | - Alexander Link
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University, Magdeburg, Germany
| | - Flavio De Maio
- Department of Laboratory and Infectious Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Edoardo Pasolli
- University of Naples Federico II, Department of Agricultural Sciences, Portici, Italy
| | | | - Patrizia Brigidi
- Microbiomics Unit, Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Brunella Posteraro
- Department of Basic Biotechnological Sciences, Intensive and Perioperative Clinics, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Franco Scaldaferri
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Mirjana Rajilic Stojanovic
- Department for Biochemical Engineering and Biotechnology, University of Belgrade, Belgrade, Serbia; Faculty of Technology and Metallurgy, University of Belgrade, Belgrade, Serbia
| | | | - Peter Malfertheiner
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University, Magdeburg, Germany; LMU, University Clinic, Medical Department II, Munich, Germany
| | - Luca Masucci
- Department of Laboratory and Infectious Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Manimozhiyan Arumugam
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nadeem Kaakoush
- School of Biomedical Sciences, University of New South Wales, Sydney, Australia
| | - Eran Segal
- Computer Science and Applied Mathematics Department, Weizmann Institute of Science, Rehovot, Israel
| | - Jasmohan Bajaj
- Division of Gastroenterology, Hepatology, and Nutrition, Virginia Commonwealth University and Richmond VA Medical Center, Richmond, VA, USA
| | - Rupert Leong
- Department of Gastroenterology, Concord Repatriation General Hospital, Sydney, Australia; MQ Health, Macquarie University Hospital, Sydney, Australia
| | - John Cryan
- APC Microbiome Ireland, Department of Medicine, University College Cork, Cork, Ireland
| | - Rinse K Weersma
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Center Groningen, Groningen, Netherlands
| | - Robert Knight
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
| | | | - Fergus Shanahan
- APC Microbiome Ireland, Department of Medicine, University College Cork, Cork, Ireland
| | - Patrice D Cani
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group, UCLouvain, Université Catholique de Louvain, Brussels, Belgium; Institute of Experimental and Clinical Research (IREC), UCLouvain, Université Catholique de Louvain, Brussels, Belgium; Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO department, WEL Research Institute, Wavre, Belgium
| | - Eran Elinav
- Systems Immunology Department, Weizmann Institute of Science, Rehovot, Israel; Cancer-Microbiome Division, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Maurizio Sanguinetti
- Department of Laboratory and Infectious Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Willem M de Vos
- Laboratory of Microbiology, Wageningen University, Netherlands; Human Microbiome Research Program, University of Helsinki, Finland
| | - Emad El-Omar
- Microbiome Research Centre, St George & Sutherland Clinical Campuses, School of Clinical Medicine, University of New South Wales, Sydney, Australia
| | - Joel Dorè
- MR Micalis Institut, INRA, Paris-Saclay University, Jouy-En-Josas, France
| | - Julian Marchesi
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, St Mary's Hospital Campus, Imperial College London, London, UK
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Harry Sokol
- MR Micalis Institut, INRA, Paris-Saclay University, Jouy-En-Josas, France; Gastroenterology Department, Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint-Antoine Hospital, Paris, France; Paris Center for Microbiome MedICIsne (PaCeMM) FHU, Paris, France
| | - Nicola Segata
- Department CIBIO, University of Trento, Trento, Italy; Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Giovanni Cammarota
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Antonio Gasbarrini
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Gianluca Ianiro
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.
| |
Collapse
|
8
|
D’Ambrosio R, Cavallo S, Brunetti R, Pellicanò R, Vaccaro E, Borriello G, Paradiso R, Serpe FP, Lambiase S, Bruzzese F, Palma G, Rea D, Barbieri A, D’Amore M, Dimatteo M, degli Uberti B, Paciello O, Baldi L. The Use of Antimicrobials in Animal Husbandry as a Potential Factor for the Increased Incidence of Colorectal Cancer: Food Safety and Kinetics in a Murine Model. Animals (Basel) 2025; 15:315. [PMID: 39943084 PMCID: PMC11815752 DOI: 10.3390/ani15030315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 01/17/2025] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
The aim of this research was to investigate the effects of the prolonged use of the broad-spectrum antimicrobial widely used in animal husbandry. By means of a mouse model, a translational study was carried out on immunocompetent mice (with a complete immune system). This study highlighted the effect of antimicrobial residues taken in with food on the growth time of cancer and on alterations to the gut microbiota. This project considered the fight against antimicrobial resistance from a One Health perspectivethrough collaboration between human medicine and veterinary medicine. Regarding food safety, antimicrobial residues in products of animal origin are rarely detected; they therefore constitute a negligible factor in determining colorectal cancer.
Collapse
Affiliation(s)
- Rosa D’Ambrosio
- Istituto Zooprofilattico Sperimentale del Mezzogiorno (IZSM), 80055 Portici, Italy; (R.D.); (S.C.); (R.P.); (G.B.); (R.P.); (F.P.S.); (S.L.); (M.D.); (B.d.U.); (L.B.)
| | - Stefania Cavallo
- Istituto Zooprofilattico Sperimentale del Mezzogiorno (IZSM), 80055 Portici, Italy; (R.D.); (S.C.); (R.P.); (G.B.); (R.P.); (F.P.S.); (S.L.); (M.D.); (B.d.U.); (L.B.)
| | - Roberta Brunetti
- Istituto Zooprofilattico Sperimentale del Mezzogiorno (IZSM), 80055 Portici, Italy; (R.D.); (S.C.); (R.P.); (G.B.); (R.P.); (F.P.S.); (S.L.); (M.D.); (B.d.U.); (L.B.)
| | - Roberta Pellicanò
- Istituto Zooprofilattico Sperimentale del Mezzogiorno (IZSM), 80055 Portici, Italy; (R.D.); (S.C.); (R.P.); (G.B.); (R.P.); (F.P.S.); (S.L.); (M.D.); (B.d.U.); (L.B.)
| | - Emanuela Vaccaro
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80137 Napoli, Italy; (E.V.); (O.P.)
| | - Giorgia Borriello
- Istituto Zooprofilattico Sperimentale del Mezzogiorno (IZSM), 80055 Portici, Italy; (R.D.); (S.C.); (R.P.); (G.B.); (R.P.); (F.P.S.); (S.L.); (M.D.); (B.d.U.); (L.B.)
| | - Rubina Paradiso
- Istituto Zooprofilattico Sperimentale del Mezzogiorno (IZSM), 80055 Portici, Italy; (R.D.); (S.C.); (R.P.); (G.B.); (R.P.); (F.P.S.); (S.L.); (M.D.); (B.d.U.); (L.B.)
| | - Francesco Paolo Serpe
- Istituto Zooprofilattico Sperimentale del Mezzogiorno (IZSM), 80055 Portici, Italy; (R.D.); (S.C.); (R.P.); (G.B.); (R.P.); (F.P.S.); (S.L.); (M.D.); (B.d.U.); (L.B.)
| | - Sara Lambiase
- Istituto Zooprofilattico Sperimentale del Mezzogiorno (IZSM), 80055 Portici, Italy; (R.D.); (S.C.); (R.P.); (G.B.); (R.P.); (F.P.S.); (S.L.); (M.D.); (B.d.U.); (L.B.)
| | - Francesca Bruzzese
- Experimental Animal Unit, Istituto Nazionale Tumori-IRCCS-Fondazione “G. Pascale”, 80131 Naples, Italy; (F.B.); (G.P.)
| | - Giuseppe Palma
- Experimental Animal Unit, Istituto Nazionale Tumori-IRCCS-Fondazione “G. Pascale”, 80131 Naples, Italy; (F.B.); (G.P.)
| | - Domenica Rea
- Laboratory Medicine Unit, Istituto Nazionale Tumori- IRCCS- Fondazione “G. Pascale”, 80131 Naples, Italy;
| | - Antonio Barbieri
- ASL Salerno UOC Laboratorio d’Analisi, Vallo della Lucania, 84078 Salerno, Italy
| | - Marianna D’Amore
- Istituto Zooprofilattico Sperimentale del Mezzogiorno (IZSM), 80055 Portici, Italy; (R.D.); (S.C.); (R.P.); (G.B.); (R.P.); (F.P.S.); (S.L.); (M.D.); (B.d.U.); (L.B.)
| | - Maria Dimatteo
- Istituto Zooprofilattico Sperimentale del Mezzogiorno (IZSM), 80055 Portici, Italy; (R.D.); (S.C.); (R.P.); (G.B.); (R.P.); (F.P.S.); (S.L.); (M.D.); (B.d.U.); (L.B.)
| | - Barbara degli Uberti
- Istituto Zooprofilattico Sperimentale del Mezzogiorno (IZSM), 80055 Portici, Italy; (R.D.); (S.C.); (R.P.); (G.B.); (R.P.); (F.P.S.); (S.L.); (M.D.); (B.d.U.); (L.B.)
| | - Orlando Paciello
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80137 Napoli, Italy; (E.V.); (O.P.)
| | - Loredana Baldi
- Istituto Zooprofilattico Sperimentale del Mezzogiorno (IZSM), 80055 Portici, Italy; (R.D.); (S.C.); (R.P.); (G.B.); (R.P.); (F.P.S.); (S.L.); (M.D.); (B.d.U.); (L.B.)
| |
Collapse
|
9
|
Guccione C, Patel L, Tomofuji Y, McDonald D, Gonzalez A, Sepich-Poore GD, Sonehara K, Zakeri M, Chen Y, Dilmore AH, Damle N, Baranzini SE, Hightower G, Nakatsuji T, Gallo RL, Langmead B, Okada Y, Curtius K, Knight R. Incomplete human reference genomes can drive false sex biases and expose patient-identifying information in metagenomic data. Nat Commun 2025; 16:825. [PMID: 39827261 PMCID: PMC11742726 DOI: 10.1038/s41467-025-56077-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 01/07/2025] [Indexed: 01/22/2025] Open
Abstract
As next-generation sequencing technologies produce deeper genome coverages at lower costs, there is a critical need for reliable computational host DNA removal in metagenomic data. We find that insufficient host filtration using prior human genome references can introduce false sex biases and inadvertently permit flow-through of host-specific DNA during bioinformatic analyses, which could be exploited for individual identification. To address these issues, we introduce and benchmark three host filtration methods of varying throughput, with concomitant applications across low biomass samples such as skin and high microbial biomass datasets including fecal samples. We find that these methods are important for obtaining accurate results in low biomass samples (e.g., tissue, skin). Overall, we demonstrate that rigorous host filtration is a key component of privacy-minded analyses of patient microbiomes and provide computationally efficient pipelines for accomplishing this task on large-scale datasets.
Collapse
Affiliation(s)
- Caitlin Guccione
- Division of Biomedical Informatics, Department of Medicine, University of California San Diego, La Jolla, CA, USA
- Bioinformatics and Systems Biology Program, University of California San Diego, La Jolla, CA, USA
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Lucas Patel
- Bioinformatics and Systems Biology Program, University of California San Diego, La Jolla, CA, USA
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Medical Scientist Training Program, University of California, San Diego, La Jolla, CA, USA
| | - Yoshihiko Tomofuji
- Department of Genome Informatics, Graduate School of Medicine, the University of Tokyo, Tokyo, 113-8654, Japan
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, 565-0871, Japan
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
| | - Daniel McDonald
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Antonio Gonzalez
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Gregory D Sepich-Poore
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Kyuto Sonehara
- Department of Genome Informatics, Graduate School of Medicine, the University of Tokyo, Tokyo, 113-8654, Japan
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, 565-0871, Japan
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
| | - Mohsen Zakeri
- Department of Computer Science, Johns Hopkins University, Baltimore, MD, USA
| | - Yang Chen
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Biomedical Sciences Graduate Program, University of California San Diego, La Jolla, CA, USA
- Department of Dermatology, University of California San Diego, La Jolla, CA, USA
| | - Amanda Hazel Dilmore
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Biomedical Sciences Graduate Program, University of California San Diego, La Jolla, CA, USA
| | - Neil Damle
- Halıcıoğlu Data Science Institute, University of California San Diego, La Jolla, CA, USA
- Department of Cognitive Science, University of California San Diego, La Jolla, CA, USA
| | - Sergio E Baranzini
- Weill Institute for Neurosciences. Department of Neurology. University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - George Hightower
- Department of Dermatology, University of California San Diego, La Jolla, CA, USA
- Rady Children's Hospital, San Diego, CA, USA
| | - Teruaki Nakatsuji
- Department of Dermatology, University of California San Diego, La Jolla, CA, USA
| | - Richard L Gallo
- Department of Dermatology, University of California San Diego, La Jolla, CA, USA
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
| | - Ben Langmead
- Department of Computer Science, Johns Hopkins University, Baltimore, MD, USA
| | - Yukinori Okada
- Department of Genome Informatics, Graduate School of Medicine, the University of Tokyo, Tokyo, 113-8654, Japan
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, 565-0871, Japan
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
- Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, 565-0871, Japan
- Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Osaka University, Suita, 565-0871, Japan
| | - Kit Curtius
- Division of Biomedical Informatics, Department of Medicine, University of California San Diego, La Jolla, CA, USA.
- VA San Diego Healthcare System, San Diego, CA, USA.
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA.
| | - Rob Knight
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA.
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, USA.
- Halıcıoğlu Data Science Institute, University of California San Diego, La Jolla, CA, USA.
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA.
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
10
|
Bokor M, Chiacchiaro E, Phadtare S. Collection and Processing of Samples for Next-Generation Sequencing to Study the Gut Microbiome. Methods Mol Biol 2025; 2866:59-70. [PMID: 39546197 DOI: 10.1007/978-1-0716-4192-7_4] [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] [Indexed: 11/17/2024]
Abstract
16S rRNA gene sequencing is commonly used for identification and quantitation of microorganisms in complex biological mixtures, such as the human gut microbiome. The 16S rRNA gene is an excellent target gene for sequencing DNA in a heterogenous sample, as it is a highly conserved part of the transcriptional machinery. Universal PCR primers are used to amplify the conserved regions of 16S. Thus, it is possible to amplify the gene in a wide range of different microorganisms from a single sample. As the 16S rRNA gene consists of both conserved and variable regions, sequencing of the variable regions can be used to differentiate between different bacterial species.
Collapse
Affiliation(s)
- Maxwell Bokor
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA
| | - Emily Chiacchiaro
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA
| | - Sangita Phadtare
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA.
| |
Collapse
|
11
|
Carroll A, Bell MJ, Bleach ECL, Turner D, Williams LK. Impact of dairy calf management practices on the intestinal tract microbiome pre-weaning. J Med Microbiol 2025; 74. [PMID: 39879083 DOI: 10.1099/jmm.0.001957] [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] [Indexed: 01/31/2025] Open
Abstract
Introduction. Microbiota in the gastrointestinal tract (GIT) consisting of the rumen and hindgut (the small intestine, cecum and colon) in dairy calves play a vital role in their growth and development. This review discusses the development of dairy calf intestinal microbiomes with an emphasis on the impact that husbandry and rearing management have on microbiome development, health and growth of pre-weaned dairy calves.Discussion. The diversity and composition of the microbes that colonize the lower GIT (small and large intestine) can have a significant impact on the growth and development of the calf, through influence on nutrient metabolism, immune modulation, resistance or susceptibility to infection, production outputs and behaviour modification in adult life. The colonization of the calf intestinal microbiome dynamically changes from birth, increasing microbial richness and diversity until weaning, where further dynamic and drastic microbiome change occurs. In dairy calves, neonatal microbiome development prior to weaning is influenced by direct and indirect factors, some of which could be considered stressors, such as maternal interaction, environment, diet, husbandry and weaning practices. The specific impact of these can dictate intestinal microbial colonization, with potential lifelong consequences.Conclusion. Evidence suggests the potential detrimental effect that sudden changes and stress may have on calf health and growth due to management and husbandry practices, and the importance of establishing a stable yet diverse intestinal microbiome population at an early age is essential for calf success. The possibility of improving the health of calves through intestinal microbiome modulation and using alternative strategies including probiotic use, faecal microbiota transplantation and novel approaches of microbiome tracking should be considered to support animal health and sustainability of dairy production systems.
Collapse
Affiliation(s)
- Aisling Carroll
- Animal and Agriculture Department, Hartpury University, Gloucester, GL19 3BE, UK
| | - Matt J Bell
- Animal and Agriculture Department, Hartpury University, Gloucester, GL19 3BE, UK
| | - Emma C L Bleach
- Animal Science Research Centre, Harper Adams University, Edgmond, Newport, TF10 8NB, Shropshire, UK
| | - Dann Turner
- University of the West of England, Bristol, Coldharbour Lane, BS16 1QY, UK
| | - Lisa K Williams
- Animal and Agriculture Department, Hartpury University, Gloucester, GL19 3BE, UK
| |
Collapse
|
12
|
Muruganandam A, Migliorini F, Jeyaraman N, Vaishya R, Balaji S, Ramasubramanian S, Maffulli N, Jeyaraman M. Molecular Mimicry Between Gut Microbiome and Rheumatoid Arthritis: Current Concepts. Med Sci (Basel) 2024; 12:72. [PMID: 39728421 PMCID: PMC11677576 DOI: 10.3390/medsci12040072] [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: 10/25/2024] [Revised: 12/02/2024] [Accepted: 12/09/2024] [Indexed: 12/28/2024] Open
Abstract
Rheumatoid arthritis (RA) represents an autoimmune condition impacted by a combination of genetic and environmental factors, with the gut microbiome (GMB) being one of the influential environmental factors. Patients with RA display notable modifications in the composition of their GMB, characterised by decreased diversity and distinct bacterial alterations. The GMB, comprising an extensive array of approximately 35,000 bacterial species residing within the gastrointestinal tract, has garnered considerable attention as a pivotal contributor to both human health and the pathogenesis of diseases. This article provides an in-depth exploration of the intricate involvement of the GMB in the context of RA. The oral-GMB axis highlights the complex role of bacteria in RA pathogenesis by producing antibodies to citrullinated proteins (ACPAs) through molecular mimicry. Dysbiosis affects Tregs, cytokine levels, and RA disease activity, suggesting that regulating cytokines could be a strategy for managing inflammation in RA. The GMB also has significant implications for drug responses and toxicity, giving rise to the field of pharmacomicrobiomics. The composition of the microbiota can impact the efficacy and toxicity of drugs, while the microbiota's metabolites can influence drug response. Recent research has identified specific bacteria, metabolites, and immune responses associated with RA, offering potential targets for personalised management. However, several challenges, including the variation in microbial composition, establishing causality, accounting for confounding factors, and translating findings into clinical practice, need to be addressed. Microbiome-targeted therapy is still in its early stages and requires further research and standardisation for effective implementation.
Collapse
Affiliation(s)
- Anandanarayan Muruganandam
- Department of Orthopaedics, Faculty of Medicine—Sri Lalithambigai Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai 600095, India;
| | - Filippo Migliorini
- Department of Orthopedics and Trauma Surgery, Academic Hospital of Bolzano (SABES-ASDAA), 39100 Bolzano, Italy
- Department of Life Sciences, Health, and Health Professions, Link Campus University, 00165 Rome, Italy
| | - Naveen Jeyaraman
- Department of Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai 600077, India;
| | - Raju Vaishya
- Department of Orthopaedics and Joint Replacement Surgery, Indraprastha Apollo Hospital, New Delhi 110076, India;
| | - Sangeetha Balaji
- Department of Orthopaedics, Government Medical College, Omandurar Government Estate, Chennai 600002, India; (S.B.); (S.R.)
| | - Swaminathan Ramasubramanian
- Department of Orthopaedics, Government Medical College, Omandurar Government Estate, Chennai 600002, India; (S.B.); (S.R.)
| | - Nicola Maffulli
- Department of Trauma and Orthopaedic Surgery, Faculty of Medicine and Psychology, University La Sapienza, 00185 Roma, Italy;
- School of Pharmacy and Bioengineering, Keele University Faculty of Medicine, Stoke on Trent ST4 7QB, UK
- Centre for Sports and Exercise Medicine, Barts and the London School of Medicine and Dentistry, Mile End Hospital, Queen Mary University of London, London E1 4DG, UK
| | - Madhan Jeyaraman
- Department of Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai 600077, India;
| |
Collapse
|
13
|
Murray PE, Coffman JA, Garcia-Godoy F. Oral Pathogens' Substantial Burden on Cancer, Cardiovascular Diseases, Alzheimer's, Diabetes, and Other Systemic Diseases: A Public Health Crisis-A Comprehensive Review. Pathogens 2024; 13:1084. [PMID: 39770344 PMCID: PMC11677847 DOI: 10.3390/pathogens13121084] [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: 11/23/2024] [Revised: 11/28/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
This review synthesizes the findings from 252 studies to explore the relationship between the oral pathogens associated with periodontitis, dental caries, and systemic diseases. Individuals with oral diseases, such as periodontitis, are between 1.7 and 7.5 times (average 3.3 times) more likely to develop systemic diseases or suffer adverse pregnancy outcomes, underscoring the critical connection between dental and overall health. Oral conditions such as periodontitis and dental caries represent a significant health burden, affecting 26-47% of Americans. The most important oral pathogens, ranked by publication frequency, include the herpes virus, C. albicans, S. mutans, P. gingivalis, F. nucleatum, A. actinomycetemcomitans, P. intermedia, T. denticola, and T. forsythia. The systemic diseases and disorders linked to oral infections, ranked similarly, include cancer, respiratory, liver, bowel, fever, kidney, complications in pregnancy, cardiovascular bacteremia, diabetes, arthritis, autoimmune, bladder, dementia, lupus, and Alzheimer's diseases. Evidence supports the efficacy of dental and periodontal treatments in eliminating oral infections and reducing the severity of systemic diseases. The substantial burden that oral pathogens have on cancer, cardiovascular diseases, Alzheimer's, diabetes, and other systemic diseases poses a significant public health crisis.
Collapse
Affiliation(s)
| | - Jonathan A Coffman
- College of Pharmacy, American University of Health Sciences, Signal Hill, CA 90755, USA
| | - Franklin Garcia-Godoy
- College of Dentistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
14
|
Kramer CS, Monsegue A, Morwani-Mangnani J, Grootswagers P, Beekman M, Slagboom PE, Verdijk LB, de Groot LCPGM. Design of the VOILA-intervention study: A 12-week nutrition and resistance exercise intervention in metabolic or mobility compromised Dutch older adults and the response on immune-metabolic, gut and muscle health parameters. Mech Ageing Dev 2024; 222:112002. [PMID: 39490538 DOI: 10.1016/j.mad.2024.112002] [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: 07/25/2024] [Revised: 10/16/2024] [Accepted: 10/24/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Exercise and nutrition interventions can slow ageing-induced decline in physiology. However, effects are heterogeneous and usually studied separately per outcome domain. In the VOILA study, we simultaneously study various health outcomes relevant for older adults and the inter-individual heterogeneity in response to a lifestyle intervention. METHODS VOILA is a 12-week lifestyle intervention in 3 groups of older adults (≥60 years), with compromised mobility (n=50), compromised metabolic health (n=50), or recovering from total knee replacement (TKR, n=70, of which 20 randomized to standard care only). The intervention includes high-intensity resistance exercise training thrice weekly, nutritional counselling, and nutritional supplements every morning and evening (including 20-25 g whey protein and (evening only) 5.5 g Biotis™ GOS). We measure immune-metabolic, gut health, muscle mass and physical functioning at baseline and after completion of the intervention/standard care. An additional reference group of healthy older adults (n=50) will undergo baseline measurements only. DISCUSSION Improvements in various physiological systems are expected, but with differences between groups/individuals. This study will provide insights into how the physiological state of older adults influences the extent of lifestyle-induced health improvements to create better tailored interventions to attenuate biological ageing and improve the health span of subgroups and individuals.
Collapse
Affiliation(s)
- C S Kramer
- Wageningen University & Research, Wageningen Campus, Agrotechnology and Food Sciences Group, Division of Human Nutrition and Health, PO Box 17, Wageningen 6700 AA, the Netherlands.
| | - A Monsegue
- Maastricht University Medical Center+, Department of Human Biology, NUTRIM Institute of nutrition and translational research in metabolism, PO Box 616, Maastricht 6200 MD, the Netherlands.
| | - J Morwani-Mangnani
- Leiden University Medical Centre, Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Einthovenweg 20, Leiden 2333 ZC, the Netherlands.
| | - P Grootswagers
- Wageningen University & Research, Wageningen Campus, Agrotechnology and Food Sciences Group, Division of Human Nutrition and Health, PO Box 17, Wageningen 6700 AA, the Netherlands.
| | - M Beekman
- Leiden University Medical Centre, Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Einthovenweg 20, Leiden 2333 ZC, the Netherlands.
| | - P E Slagboom
- Leiden University Medical Centre, Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Einthovenweg 20, Leiden 2333 ZC, the Netherlands.
| | - L B Verdijk
- Maastricht University Medical Center+, Department of Human Biology, NUTRIM Institute of nutrition and translational research in metabolism, PO Box 616, Maastricht 6200 MD, the Netherlands.
| | - L C P G M de Groot
- Wageningen University & Research, Wageningen Campus, Agrotechnology and Food Sciences Group, Division of Human Nutrition and Health, PO Box 17, Wageningen 6700 AA, the Netherlands.
| |
Collapse
|
15
|
Pahirah N, Narkwichean A, Taweechotipatr M, Wannaiampikul S, Duang-Udom C, Laosooksathit W. Comparison of Gut Microbiomes Between Neonates Born by Cesarean Section and Vaginal Delivery: Prospective Observational Study. BIOMED RESEARCH INTERNATIONAL 2024; 2024:8302361. [PMID: 39640900 PMCID: PMC11620805 DOI: 10.1155/bmri/8302361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 11/02/2024] [Accepted: 11/14/2024] [Indexed: 12/07/2024]
Abstract
Background: Balanced diversity and abundance of gut microbiome play important roles in human health, including neonatal health. Though not established, there is evidence that the delivery route could alter the diversity of neonatal gut microbiomes. Objective: The objective of the study was to investigate the differences in the gut microbiomes of neonates delivered via cesarean section compared to those born by vaginal delivery and to identify the predominant microbial taxa present in each group. Study Design: A prospective observational study of 281 healthy neonates born between February 2021 and April 2023 at Her Royal Highness Maha Chakri Sirindhorn Medical Center, Srinakharinwirot University, Thailand, was performed. The study population was divided into two groups: 139 neonates born via vaginal delivery and 141 neonates born via cesarean section. The microbiota composition of each neonate's fecal sample was identified by using 16S ribosomal ribonucleic acid metagenomic sequencing. Results: Neonates delivered vaginally exhibited a gut microbiome with higher abundance and diversity than those delivered by cesarean delivery. Bifidobacterium was the dominant genus in both groups. Bifidobacterium breve was the dominant species and was significantly higher in cesarean-delivered neonates compared to those delivered vaginally (24.0% and 9.2%, respectively) (p < 0.001). However, the taxonomy of only 89 (64.0%) and 44 (31.43%) fecal samples could be identified from the vaginal and cesarean delivery groups, respectively. Conclusion: Route of delivery is associated with neonatal gut microbiome abundance and diversity. Neonates delivered via vaginal delivery exhibited higher diversity but lower abundance of the dominant species in the gut microbiome. Trial Registration: Thai Clinical Trials Registry identifier: TCTR20221024003.
Collapse
Affiliation(s)
- Nichapat Pahirah
- Department of Obstetrics and Gynecology, Faculty of Medicine, Srinakharinwirot University, Nakhon Nayok, Thailand
| | - Amarin Narkwichean
- Department of Obstetrics and Gynecology, Faculty of Medicine, Srinakharinwirot University, Nakhon Nayok, Thailand
| | - Malai Taweechotipatr
- Department of Microbiology, Faculty of Medicine, Srinakharinwirot University, Bangkok, Thailand
| | - Sivaporn Wannaiampikul
- Department of Biochemistry, Faculty of Medicine, Srinakharinwirot University, Bangkok, Thailand
| | | | - Wipada Laosooksathit
- Department of Obstetrics and Gynecology, Faculty of Medicine, Srinakharinwirot University, Nakhon Nayok, Thailand
| |
Collapse
|
16
|
van de Put M, van den Belt M, de Wit N, Kort R. Rationale and design of a randomized placebo-controlled nutritional trial embracing a citizen science approach. Nutr Res 2024; 131:96-110. [PMID: 39378660 DOI: 10.1016/j.nutres.2024.07.008] [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: 02/14/2024] [Revised: 07/24/2024] [Accepted: 07/24/2024] [Indexed: 10/10/2024]
Abstract
Modulation of the gut microbiota through specific dietary interventions shows potential for maintenance and optimization of health. A dietary fiber diet and fermented foods diet appear to alter the gut microbiota, but evidence is limited. Therefore, we designed the Gut Health Enhancement by Eating Favorable Food study, a 21-week randomized controlled trial studying effects of dietary fibers and fermented foods on gut microbiota diversity and composition, while also stimulating dietary behavior changes through a citizen science (CS) approach. We hypothesized that a high-fermented food diet would increase microbial diversity, whereas a high-dietary fiber diet would stimulate the growth of specific fiber-degrading bacteria. The following elements of CS were adopted: education on the gut microbiota, tailored dietary intervention, remote data collection by participants, sharing of personal gut microbiota outcomes with participants, and vlogs by participants for dissemination of results. Here we describe the study protocol and report the flow of participants, baseline characteristics, and compliance rates. Completed in March 2024, the trial included 147 healthy adults randomized to a high-dietary fiber intervention, high-fermented food intervention, or control group. Each group received an additional study product after 2 weeks: dried chicory root, a fermented beverage, or maltodextrin (placebo). A 3-month follow-up assessed the participants' ability to sustain dietary changes. The recruitment of participants was successful, reflected by 1448 applications. The compliance with the dietary guidelines and study products was >90%. This study shows that including elements of CS in an randomized controlled trial is feasible and may help recruitment and compliance.
Collapse
Affiliation(s)
- Marieke van de Put
- Amsterdam Institute for Life and Environment, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Maartje van den Belt
- Wageningen Food and Biobased Research, Wageningen University & Research, 6708 WG Wageningen, The Netherlands
| | - Nicole de Wit
- Wageningen Food and Biobased Research, Wageningen University & Research, 6708 WG Wageningen, The Netherlands
| | - Remco Kort
- Amsterdam Institute for Life and Environment, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands; ARTIS-Micropia, Plantage Kerklaan 38-40, 1018 CZ Amsterdam, The Netherlands.
| |
Collapse
|
17
|
Kasselman LJ, Peltier MR, De Leon J, Reiss AB. Cognitive Function and the Consumption of Probiotic Foods: A National Health and Nutrition Examination Survey Study. Nutrients 2024; 16:3631. [PMID: 39519464 PMCID: PMC11547479 DOI: 10.3390/nu16213631] [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: 09/25/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
Background/Objectives: Impaired cognition is a key trait of the diseases of aging and is an important quality of life factor for older adults and their families. Over the past decade, there has been an increasing appreciation for the role of the microbiome in cognition, as well as emerging evidence that probiotics, such as those in yogurt and other dairy products, can have a positive impact on cognitive function. However, it is unclear to what extent the consumption of yogurt is associated with improved cognitive function in older adults. Methods: Therefore, we compared the scores for the Wechsler Adult Intelligence Scale, Digit-Symbol Substitution Test between respondents who self-reported daily yogurt/dairy consumption with those who claimed they did not in an NHANES. Results: We found that cognitive scores were significantly higher (40.03 ± 0.64 vs. 36.28 ± 1.26, p = 0.017) in respondents reporting daily yogurt/dairy consumption, though only a trend remained after adjusting for sociodemographic covariates (p = 0.074). Conclusions: Further studies are required to confirm that this is a cause-effect relationship and whether changing diets is a low-cost means of protecting aging populations from cognitive decline and improving their quality of life.
Collapse
Affiliation(s)
- Lora J. Kasselman
- Department of Medical Sciences, Hackensack Meridian School of Medicine, Nutley, NJ 07110, USA;
- Hackensack Meridian Health Research Institute, Hackensack, NJ 07601, USA
| | - Morgan R. Peltier
- Department of Psychiatry, Hackensack Meridian School of Medicine, Nutley, NJ 07110, USA;
- Department of Psychiatry, Jersey Shore University Medical Center, Neptune City, NJ 07753, USA
| | - Joshua De Leon
- Department of Medicine, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA;
| | - Allison B. Reiss
- Department of Medicine, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA;
| |
Collapse
|
18
|
Masnikosa R, Cvetković Z, Pirić D. Tumor Biology Hides Novel Therapeutic Approaches to Diffuse Large B-Cell Lymphoma: A Narrative Review. Int J Mol Sci 2024; 25:11384. [PMID: 39518937 PMCID: PMC11545713 DOI: 10.3390/ijms252111384] [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: 07/14/2024] [Revised: 10/13/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is a malignancy of immense biological and clinical heterogeneity. Based on the transcriptomic or genomic approach, several different classification schemes have evolved over the years to subdivide DLBCL into clinically (prognostically) relevant subsets, but each leaves unclassified samples. Herein, we outline the DLBCL tumor biology behind the actual and potential drug targets and address the challenges and drawbacks coupled with their (potential) use. Therapeutic modalities are discussed, including small-molecule inhibitors, naked antibodies, antibody-drug conjugates, chimeric antigen receptors, bispecific antibodies and T-cell engagers, and immune checkpoint inhibitors. Candidate drugs explored in ongoing clinical trials are coupled with diverse toxicity issues and refractoriness to drugs. According to the literature on DLBCL, the promise for new therapeutic targets lies in epigenetic alterations, B-cell receptor and NF-κB pathways. Herein, we present putative targets hiding in lipid pathways, ferroptosis, and the gut microbiome that could be used in addition to immuno-chemotherapy to improve the general health status of DLBCL patients, thus increasing the chance of being cured. It may be time to devote more effort to exploring DLBCL metabolism to discover novel druggable targets. We also performed a bibliometric and knowledge-map analysis of the literature on DLBCL published from 2014-2023.
Collapse
Affiliation(s)
- Romana Masnikosa
- Department of Physical Chemistry, Vinca Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia;
| | - Zorica Cvetković
- Department of Hematology, Clinical Hospital Centre Zemun, Vukova 9, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Dr Subotića 8, 11000 Belgrade, Serbia
| | - David Pirić
- Department of Physical Chemistry, Vinca Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia;
| |
Collapse
|
19
|
Guccione C, Patel L, Tomofuji Y, McDonald D, Gonzalez A, Sepich-Poore GD, Sonehara K, Zakeri M, Chen Y, Dilmore AH, Damle N, Baranzini SE, Nakatsuji T, Gallo RL, Langmead B, Okada Y, Curtius K, Knight R. Incomplete human reference genomes can drive false sex biases and expose patient-identifying information in metagenomic data. RESEARCH SQUARE 2024:rs.3.rs-4721159. [PMID: 39502785 PMCID: PMC11537348 DOI: 10.21203/rs.3.rs-4721159/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/13/2024]
Abstract
As next-generation sequencing technologies produce deeper genome coverages at lower costs, there is a critical need for reliable computational host DNA removal in metagenomic data. We find that insufficient host filtration using prior human genome references can introduce false sex biases and inadvertently permit flow-through of host-specific DNA during bioinformatic analyses, which could be exploited for individual identification. To address these issues, we introduce and benchmark three host filtration methods of varying throughput, with concomitant applications across low biomass samples such as skin and high microbial biomass datasets including fecal samples. We find that these methods are important for obtaining accurate results in low biomass samples (e.g., tissue, skin). Overall, we demonstrate that rigorous host filtration is a key component of privacy-minded analyses of patient microbiomes and provide computationally efficient pipelines for accomplishing this task on large-scale datasets.
Collapse
Affiliation(s)
- Caitlin Guccione
- Division of Biomedical Informatics, Department of Medicine, University of California San Diego, La Jolla, California 92093, USA
- Bioinformatics and Systems Biology Program, University of California San Diego, La Jolla, California 92093, USA
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Lucas Patel
- Bioinformatics and Systems Biology Program, University of California San Diego, La Jolla, California 92093, USA
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Medical Scientist Training Program, University of California, San Diego, La Jolla, California, USA
| | - Yoshihiko Tomofuji
- Department of Genome Informatics, Graduate School of Medicine, the University of Tokyo, Tokyo 113-8654, Japan
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita 565-0871, Japan
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan
| | - Daniel McDonald
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Antonio Gonzalez
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | | | - Kyuto Sonehara
- Department of Genome Informatics, Graduate School of Medicine, the University of Tokyo, Tokyo 113-8654, Japan
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita 565-0871, Japan
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan
| | - Mohsen Zakeri
- Department of Computer Science, Johns Hopkins University
| | - Yang Chen
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Biomedical Sciences Graduate Program, University of California San Diego, La Jolla, CA, USA
- Halicioğlu Data Science Institute, University of California San Diego, La Jolla, CA, USA
| | - Amanda Hazel Dilmore
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Biomedical Sciences Graduate Program, University of California San Diego, La Jolla, CA, USA
| | - Neil Damle
- Halicioğlu Data Science Institute, University of California San Diego, La Jolla, CA, USA
- Department of Cognitive Science, University of California San Diego, La Jolla, CA, USA
| | - Sergio E. Baranzini
- Weill Institute for Neurosciences. Department of Neurology. University of California, San Francisco (UCSF), San Francisco, CA 94158, USA
| | - Teruaki Nakatsuji
- Department of Dermatology, University of California San Diego, La Jolla, CA, USA
| | - Richard L. Gallo
- Department of Dermatology, University of California San Diego, La Jolla, CA, USA
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
| | - Ben Langmead
- Department of Computer Science, Johns Hopkins University
| | - Yukinori Okada
- Department of Genome Informatics, Graduate School of Medicine, the University of Tokyo, Tokyo 113-8654, Japan
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita 565-0871, Japan
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan
- Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita 565-0871, Japan
- Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Osaka University, Suita 565-0871, Japan
| | - Kit Curtius
- Division of Biomedical Informatics, Department of Medicine, University of California San Diego, La Jolla, California 92093, USA
- VA San Diego Healthcare System, San Diego, CA, USA
| | - Rob Knight
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Halicioğlu Data Science Institute, University of California San Diego, La Jolla, CA, USA
- Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita 565-0871, Japan
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
20
|
González A, Fullaondo A, Odriozola A. Host genetics and microbiota data analysis in colorectal cancer research. ADVANCES IN GENETICS 2024; 112:31-81. [PMID: 39396840 DOI: 10.1016/bs.adgen.2024.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Colorectal cancer (CRC) is a heterogeneous disease with a complex aetiology influenced by a myriad of genetic and environmental factors. Despite advances in CRC research, it is a major burden of disease, with the second highest incidence and third leading cause of cancer deaths worldwide. To individualise diagnosis, prognosis, and treatment of CRC, developing new strategies combining precision medicine and bioinformatic procedures is promising. Precision medicine is based on omics technologies and aims to individualise the management of CRC based on patient host genetic characteristics and microbiota. Bioinformatics is central to the application of personalised medicine because it enables the analysis of large datasets generated by these technologies. At the level of host genetics, bioinformatics allows the identification of mutations, genes, molecular pathways, biomarkers and drugs relevant to colorectal carcinogenesis. At the microbiota level, bioinformatics is fundamental to analysing microbial communities' composition and functionality and developing biomarkers and personalised microbiota-based therapies. This paper explores the host and microbiota genetic data analysis in CRC research.
Collapse
Affiliation(s)
- Adriana González
- Hologenomics Research Group, Department of Genetics, Physical Anthropology, and Animal Physiology, University of the Basque Country, Spain
| | - Asier Fullaondo
- Hologenomics Research Group, Department of Genetics, Physical Anthropology, and Animal Physiology, University of the Basque Country, Spain
| | - Adrian Odriozola
- Hologenomics Research Group, Department of Genetics, Physical Anthropology, and Animal Physiology, University of the Basque Country, Spain.
| |
Collapse
|
21
|
Wang X, Li H, Yang Y, Wu Z, Wang Z, Li D, Xia W, Zou S, Liu Y, Wang F. Geographic and environmental impacts on gut microbiome in Himalayan langurs ( Semnopithecus schistaceus) and Xizang macaques ( Macaca mulatta vestita). Front Microbiol 2024; 15:1452101. [PMID: 39296299 PMCID: PMC11408304 DOI: 10.3389/fmicb.2024.1452101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 08/20/2024] [Indexed: 09/21/2024] Open
Abstract
Introduction Gut microbiome plays a crucial role in the health of wild animals. Their structural and functional properties not only reflect the host's dietary habits and habitat conditions but also provide essential support for ecological adaptation in various environments. Methods This study investigated the gut microbiome of Himalayan langurs (Semnopithecus schistaceus) and Xizang macaques (Macaca mulatta vestita) across different geographic regions using 16S rRNA gene and metagenomic sequencing. Results Results showed distinct clustering patterns in gut microbiota based on geographic location. Soil had an insignificant impact on host gut microbiome. Himalayan langurs from mid-altitude regions exhibited higher levels of antibiotic resistance genes associated with multidrug resistance, while Xizang macaques from high-altitude regions showed a broader range of resistance genes. Variations in carbohydrate-active enzymes and KEGG pathways indicated unique metabolic adaptations to different environments. Discussion These findings provide valuable insights into the health and conservation of these primates and the broader implications of microbial ecology and functional adaptations in extreme conditions.
Collapse
Affiliation(s)
- Xueyu Wang
- Key Laboratory of Conservation Biology of Rhinopithecus roxellana (Department of Education of Sichuan Province), China West Normal University, Nanchong, China
| | - Hong Li
- Key Laboratory of Conservation Biology of Rhinopithecus roxellana (Department of Education of Sichuan Province), China West Normal University, Nanchong, China
| | - Yumin Yang
- Institute of Agricultural Resources and Environment, Sichuan Academy of Agricultural Sciences, Chengdu, China
| | - Zhijiu Wu
- Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Zhixiang Wang
- Key Laboratory of Conservation Biology of Rhinopithecus roxellana (Department of Education of Sichuan Province), China West Normal University, Nanchong, China
| | - Dayong Li
- Key Laboratory of Conservation Biology of Rhinopithecus roxellana (Department of Education of Sichuan Province), China West Normal University, Nanchong, China
| | - Wancai Xia
- Key Laboratory of Conservation Biology of Rhinopithecus roxellana (Department of Education of Sichuan Province), China West Normal University, Nanchong, China
| | - Shuzhen Zou
- Key Laboratory of Conservation Biology of Rhinopithecus roxellana (Department of Education of Sichuan Province), China West Normal University, Nanchong, China
| | - Yujia Liu
- Key Laboratory of Conservation Biology of Rhinopithecus roxellana (Department of Education of Sichuan Province), China West Normal University, Nanchong, China
| | - Fan Wang
- Key Laboratory of Conservation Biology of Rhinopithecus roxellana (Department of Education of Sichuan Province), China West Normal University, Nanchong, China
| |
Collapse
|
22
|
Helal P, Xia W, Sardar P, Conway‐Morris A, Conway‐Morris A, Pedicord VA, Serfontein J. Changes in the Firmicutes to Bacteriodetes ratio in the gut microbiome in individuals with anorexia nervosa following inpatient treatment: A systematic review and a case series. Brain Behav 2024; 14:e70014. [PMID: 39295072 PMCID: PMC11410858 DOI: 10.1002/brb3.70014] [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: 04/18/2024] [Revised: 07/30/2024] [Accepted: 08/09/2024] [Indexed: 09/21/2024] Open
Abstract
OBJECTIVE Anorexia nervosa has the highest mortality rate among psychiatric illnesses. Current treatments remain ineffective for a large fraction of patients. This may be due to unclear mechanisms behind its development and maintenance. Studies exploring the role of the gut microbiome have revealed inconsistent evidence of dysbiosis. This article aims to investigate changes in the gut microbiome, particularly, mean differences in the Firmicutes to Bacteroidetes ratio, in adolescent and adult individuals with anorexia nervosa following inpatient treatment. METHODS Longitudinal studies investigating gut microbiome composition in inpatient populations of anorexia nervosa before and after treatment were systematically reviewed. Additionally, gut microbiome compositions were characterized in three acute anorexia nervosa inpatients early after admission and after 4-12 weeks of treatment. RESULTS Review results indicated an increase in the Firmicutes to Bacteroidetes ratio in individuals with anorexia nervosa after treatment. These however did not match values of their healthy counterparts. In the case-series samples, the reverse occurred with samples taken 4 weeks after treatment. In the patient who provided an extra sample 12 weeks after treatment, similar results to the studies included in the review were observed. Furthermore, Firmicutes to Bacteroidetes ratio values in the case-series samples were notably higher in the two patients who had chronic anorexia nervosa. DISCUSSION Differences in methodologies, small sample sizes, and insufficient data limited the generalizability of the outcomes of the reviewed studies. Results suggest a potentially unique microbiome signature in individuals with chronic anorexia nervosa, which may explain different outcomes in this group of patients.
Collapse
Affiliation(s)
- Passent Helal
- Adult Eating Disorders Service, Ward S3 InpatientsAddenbrooke's HospitalCambridgeUK
| | - Wangmingyu Xia
- Department of Medicine, Cambridge Institute of Therapeutic Immunology & Infectious DiseaseUniversity of CambridgeCambridgeUK
| | - Puspendu Sardar
- Department of Medicine, Cambridge Institute of Therapeutic Immunology & Infectious DiseaseUniversity of CambridgeCambridgeUK
| | - Anna Conway‐Morris
- Adult Eating Disorder Service, Ward S3 OutpatientsAddenbrooke's HospitalCambridgeUK
- School of PsychiatryNHS EnglandFulbournCambridgeUK
| | - Andrew Conway‐Morris
- Division of Anaesthesia, Department of MedicineUniversity of CambridgeCambridgeUK
- Division of Immunology, Department of PathologyUniversity of CambridgeCambridgeUK
- John V Farman Intensive Care UnitAddenbrooke's HospitalCambridgeUK
| | - Virginia A. Pedicord
- Department of Medicine, Cambridge Institute of Therapeutic Immunology & Infectious DiseaseUniversity of CambridgeCambridgeUK
| | - Jaco Serfontein
- Adult Eating Disorders Service, Ward S3 InpatientsAddenbrooke's HospitalCambridgeUK
| |
Collapse
|
23
|
Gerasimova Y, Ali H, Nadeem U. Challenges for pathologists in implementing clinical microbiome diagnostic testing. J Pathol Clin Res 2024; 10:e70002. [PMID: 39289163 PMCID: PMC11407905 DOI: 10.1002/2056-4538.70002] [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: 12/21/2023] [Revised: 08/11/2024] [Accepted: 08/26/2024] [Indexed: 09/19/2024]
Abstract
Recent research has established that the microbiome plays potential roles in the pathogenesis of numerous chronic diseases, including carcinomas. This discovery has led to significant interest in clinical microbiome testing among physicians, translational investigators, and the lay public. As novel, inexpensive methodologies to interrogate the microbiota become available, research labs and commercial vendors have offered microbial assays. However, these tests still have not infiltrated the clinical laboratory space. Here, we provide an overview of the challenges of implementing microbiome testing in clinical pathology. We discuss challenges associated with preanalytical and analytic sample handling and collection that can influence results, choosing the appropriate testing methodology for the clinical context, establishing reference ranges, interpreting the data generated by testing and its value in making patient care decisions, regulation, and cost considerations of testing. Additionally, we suggest potential solutions for these problems to expedite the establishment of microbiome testing in the clinical laboratory.
Collapse
Affiliation(s)
- Yulia Gerasimova
- Department of Infectious Diseases, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Haroon Ali
- Department of Medicine, Woodland Heights Medical Center, Lufkin, TX, USA
| | - Urooba Nadeem
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
24
|
Qi X, Sun H, Liu J, Cong M, Zhang X, Yan Y, Xia Z, Liu T, Zhao J. Phenylethanol Glycoside from Cistanche tubulosa Attenuates BSA-Induced Liver Fibrosis in Rats by Modulating the Gut Microbiota-Liver Axis. Pharmaceuticals (Basel) 2024; 17:1149. [PMID: 39338312 PMCID: PMC11435394 DOI: 10.3390/ph17091149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/01/2024] [Accepted: 08/27/2024] [Indexed: 09/30/2024] Open
Abstract
This study aimed to investigate the effect of phenylethanol glycoside from Cistanche tubulosa (CPhGs) on the prevention of bovine serum albumin (BSA)-induced hepatic fibrosis in rats. Investigation of the mechanisms of the anti-hepatic fibrosis effect was focused on CPhGs' influence on the "gut-liver" regulation, including the gut microbiota, intestinal barrier, systemic lipopolysaccharide (LPS) concentration, and LPS-related signaling pathway. The results show that CPhGs restored the diversity of gut microbiota, increased the relative abundance of Bacteroidetes, and decreased the relative abundance of Firmicutes and Proteobacteria in the fibrotic rats. In addition, CPhGs promoted the enrichment of probiotics such as Blautia, Oscillospira, Ruminococcus, Odoribacter, Bacteroides, and Parabacteroides in intestines of these rats. Furthermore, CPhGs reduced histopathological injury in the intestine and restored the tight junctions of the intestine by increasing the expression of ZO-1, occludin, and E-cadherin. CPhGs efficiently reduced serum LPS and liver lipopolysaccharide-binding protein (LBP) levels and inhibited the LPS-TLR4/MyD88/NF-κB pathway, which is related to protein expression in the liver. Correlation analysis confirmed that these beneficial bacteria were negatively associated with pathological damage, while LPS and harmful bacteria were positively associated with liver injury. Our fecal microbiota transplantation (FMT) experiment confirmed that gut microbiota is an important part of disease progression and that CPhGs is useful for the prevention and treatment of hepatic fibrosis. Our data demonstrate that the anti-hepatic fibrosis mechanism of CPhGs was mediated by regulation of the "gut-liver" axis. These results can stimulate consideration for its use in clinical practices.
Collapse
Affiliation(s)
- Xinxin Qi
- School of Public Health, Xinjiang Medical University, Urumqi 830011, China
| | - Hongguang Sun
- School of Public Health, Xinjiang Medical University, Urumqi 830011, China
| | - Jincun Liu
- School of Public Health, Xinjiang Medical University, Urumqi 830011, China
| | - Meili Cong
- School of Public Health, Xinjiang Medical University, Urumqi 830011, China
- Animal Laboratory Center, Xinjiang Medical University, Urumqi 830017, China
| | - Xinxuan Zhang
- School of Public Health, Xinjiang Medical University, Urumqi 830011, China
| | - Yuxin Yan
- School of Public Health, Xinjiang Medical University, Urumqi 830011, China
| | - Zhaolin Xia
- School of Public Health, Xinjiang Medical University, Urumqi 830011, China
| | - Tao Liu
- School of Public Health, Xinjiang Medical University, Urumqi 830011, China
| | - Jun Zhao
- School of Public Health, Xinjiang Medical University, Urumqi 830011, China
- Xinjiang Key Laboratory for Uighur Medicine, Institute of Materia Medica of Xinjiang, Urumqi 830004, China
| |
Collapse
|
25
|
Jin DM, Morton JT, Bonneau R. Meta-analysis of the human gut microbiome uncovers shared and distinct microbial signatures between diseases. mSystems 2024; 9:e0029524. [PMID: 39078158 PMCID: PMC11334437 DOI: 10.1128/msystems.00295-24] [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: 02/26/2024] [Accepted: 05/08/2024] [Indexed: 07/31/2024] Open
Abstract
Microbiome studies have revealed gut microbiota's potential impact on complex diseases. However, many studies often focus on one disease per cohort. We developed a meta-analysis workflow for gut microbiome profiles and analyzed shotgun metagenomic data covering 11 diseases. Using interpretable machine learning and differential abundance analysis, our findings reinforce the generalization of binary classifiers for Crohn's disease (CD) and colorectal cancer (CRC) to hold-out cohorts and highlight the key microbes driving these classifications. We identified high microbial similarity in disease pairs like CD vs ulcerative colitis (UC), CD vs CRC, Parkinson's disease vs type 2 diabetes (T2D), and schizophrenia vs T2D. We also found strong inverse correlations in Alzheimer's disease vs CD and UC. These findings, detected by our pipeline, provide valuable insights into these diseases. IMPORTANCE Assessing disease similarity is an essential initial step preceding a disease-based approach for drug repositioning. Our study provides a modest first step in underscoring the potential of integrating microbiome insights into the disease similarity assessment. Recent microbiome research has predominantly focused on analyzing individual diseases to understand their unique characteristics, which by design excludes comorbidities in individuals. We analyzed shotgun metagenomic data from existing studies and identified previously unknown similarities between diseases. Our research represents a pioneering effort that utilizes both interpretable machine learning and differential abundance analysis to assess microbial similarity between diseases.
Collapse
Affiliation(s)
- Dong-Min Jin
- Center for Genomics and Systems Biology, New York University, New York, New York, USA
| | - James T. Morton
- Center for Computational Biology, Flatiron Institute, Simons Foundation, New York, New York, USA
| | - Richard Bonneau
- Center for Genomics and Systems Biology, New York University, New York, New York, USA
- Genentech, New York, New York, USA
| |
Collapse
|
26
|
Sample collection time affects microbiome study results. Nat Metab 2024; 6:1207-1208. [PMID: 38951659 DOI: 10.1038/s42255-024-01077-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/03/2024]
|
27
|
Inchingolo F, Inchingolo AM, Latini G, Ferrante L, de Ruvo E, Campanelli M, Longo M, Palermo A, Inchingolo AD, Dipalma G. Difference in the Intestinal Microbiota between Breastfeed Infants and Infants Fed with Artificial Milk: A Systematic Review. Pathogens 2024; 13:533. [PMID: 39057760 PMCID: PMC11280328 DOI: 10.3390/pathogens13070533] [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: 05/29/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 07/28/2024] Open
Abstract
The gut microbiota (GM) plays a crucial role in human health, particularly during the first years of life. Differences in GM between breastfed and formula (F)-fed infants may influence long-term health outcomes. This systematic review aims to compare the gut microbiota of breastfed infants with that of F-fed infants and to evaluate the clinical implications of these differences. We searched databases on Scopus, Web of Science, and Pubmed with the following keywords: "gut microbiota", "gut microbiome", and "neonatal milk". The inclusion criteria were articles relating to the analysis of the intestinal microbiome of newborns in relation to the type of nutrition, clinical studies or case series, excluding reviews, meta-analyses, animal models, and in vitro studies. The screening phase ended with the selection of 13 publications for this work. Breastfed infants showed higher levels of beneficial bacteria such as Bifidobacterium and Lactobacillus, while F-fed infants had a higher prevalence of potentially pathogenic bacteria, including Clostridium difficile and Enterobacteriaceae. Infant feeding type influences the composition of oral GM significantly. Breastfeeding promotes a healthier and more diverse microbial ecosystem, which may offer protective health benefits. Future research should explore strategies to improve the GM of F-fed infants and understand the long-term health implications.
Collapse
Affiliation(s)
- Francesco Inchingolo
- Interdisciplinary Department of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy or (A.M.I.); or (G.L.); or (L.F.); or (E.d.R.); (M.L.); or (A.D.I.); or (G.D.)
| | - Angelo Michele Inchingolo
- Interdisciplinary Department of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy or (A.M.I.); or (G.L.); or (L.F.); or (E.d.R.); (M.L.); or (A.D.I.); or (G.D.)
| | - Giulia Latini
- Interdisciplinary Department of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy or (A.M.I.); or (G.L.); or (L.F.); or (E.d.R.); (M.L.); or (A.D.I.); or (G.D.)
| | - Laura Ferrante
- Interdisciplinary Department of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy or (A.M.I.); or (G.L.); or (L.F.); or (E.d.R.); (M.L.); or (A.D.I.); or (G.D.)
| | - Elisabetta de Ruvo
- Interdisciplinary Department of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy or (A.M.I.); or (G.L.); or (L.F.); or (E.d.R.); (M.L.); or (A.D.I.); or (G.D.)
| | - Merigrazia Campanelli
- Interdisciplinary Department of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy or (A.M.I.); or (G.L.); or (L.F.); or (E.d.R.); (M.L.); or (A.D.I.); or (G.D.)
| | - Marialuisa Longo
- Interdisciplinary Department of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy or (A.M.I.); or (G.L.); or (L.F.); or (E.d.R.); (M.L.); or (A.D.I.); or (G.D.)
| | - Andrea Palermo
- College of Medicine and Dentistry, Birmingham B4 6BN, UK;
| | - Alessio Danilo Inchingolo
- Interdisciplinary Department of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy or (A.M.I.); or (G.L.); or (L.F.); or (E.d.R.); (M.L.); or (A.D.I.); or (G.D.)
| | - Gianna Dipalma
- Interdisciplinary Department of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy or (A.M.I.); or (G.L.); or (L.F.); or (E.d.R.); (M.L.); or (A.D.I.); or (G.D.)
| |
Collapse
|
28
|
Krothapalli M, Buddendorff L, Yadav H, Schilaty ND, Jain S. From Gut Microbiota to Brain Waves: The Potential of the Microbiome and EEG as Biomarkers for Cognitive Impairment. Int J Mol Sci 2024; 25:6678. [PMID: 38928383 PMCID: PMC11203453 DOI: 10.3390/ijms25126678] [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: 04/22/2024] [Revised: 06/09/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Alzheimer's disease (AD) is a prevalent neurodegenerative disorder and a leading cause of dementia. Aging is a significant risk factor for AD, emphasizing the importance of early detection since symptoms cannot be reversed once the advanced stage is reached. Currently, there is no established method for early AD diagnosis. However, emerging evidence suggests that the microbiome has an impact on cognitive function. The gut microbiome and the brain communicate bidirectionally through the gut-brain axis, with systemic inflammation identified as a key connection that may contribute to AD. Gut dysbiosis is more prevalent in individuals with AD compared to their cognitively healthy counterparts, leading to increased gut permeability and subsequent systemic inflammation, potentially causing neuroinflammation. Detecting brain activity traditionally involves invasive and expensive methods, but electroencephalography (EEG) poses as a non-invasive alternative. EEG measures brain activity and multiple studies indicate distinct patterns in individuals with AD. Furthermore, EEG patterns in individuals with mild cognitive impairment differ from those in the advanced stage of AD, suggesting its potential as a method for early indication of AD. This review aims to consolidate existing knowledge on the microbiome and EEG as potential biomarkers for early-stage AD, highlighting the current state of research and suggesting avenues for further investigation.
Collapse
Affiliation(s)
- Mahathi Krothapalli
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida, Tampa, FL 33612, USA; (M.K.); (L.B.); (H.Y.)
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL 33612, USA;
| | - Lauren Buddendorff
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida, Tampa, FL 33612, USA; (M.K.); (L.B.); (H.Y.)
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL 33612, USA;
| | - Hariom Yadav
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida, Tampa, FL 33612, USA; (M.K.); (L.B.); (H.Y.)
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL 33612, USA;
| | - Nathan D. Schilaty
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL 33612, USA;
- Center for Neuromusculoskeletal Research, University of South Florida, Tampa, FL 33612, USA
| | - Shalini Jain
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida, Tampa, FL 33612, USA; (M.K.); (L.B.); (H.Y.)
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL 33612, USA;
| |
Collapse
|
29
|
Panzer JJ, Maples C, Meyer MP, Tillotson G, Theis KR, Chopra T. Gut microbiome alpha diversity decreases in relation to body weight, antibiotic exposure, and infection with multidrug-resistant organisms. Am J Infect Control 2024; 52:707-711. [PMID: 38176539 DOI: 10.1016/j.ajic.2023.12.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 12/23/2023] [Accepted: 12/27/2023] [Indexed: 01/06/2024]
Abstract
BACKGROUND The human gastrointestinal tract is home to a dense and diverse microbiome, predominated by bacteria. Despite the conservation of critical functionality across most individuals, the composition of the gut microbiome is highly individualized, leading to differential responses to perturbations such as oral antibiotics or multidrug-resistant organism (MDRO) infection. Herein, subject responses to these perturbations based on their body weight were evaluated. METHODS Fecal samples were collected from 45 subjects at the Detroit Medical Center to evaluate the effects of perturbations on subjects' gut microbiome composition. Bacterial profiling was completed using 16S rRNA gene sequencing. RESULTS Subjects with multiple MDROs, subjects weighing greater than 80 kg infected with MDRO E coli, and subjects weighing less than 80 kg with exposure to vancomycin and carbapenem antibiotics during hospitalization had significantly decreased gut microbiome richness. CONCLUSIONS Both administration of oral antibiotics and MDRO infections decreased gut microbiome alpha diversity, but the magnitude of these gut microbiome perturbations was body weight dependent.
Collapse
Affiliation(s)
- Jonathan J Panzer
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI
| | - Catherine Maples
- Department of Infectious Diseases, Wayne State University, Detroit, MI
| | | | | | - Kevin R Theis
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI.
| | - Teena Chopra
- Department of Infectious Diseases, Wayne State University, Detroit, MI.
| |
Collapse
|
30
|
Paraschiv AC, Vacaras V, Nistor C, Vacaras C, Strilciuc S, Muresanu DF. The effect of multiple sclerosis therapy on gut microbiota dysbiosis: a longitudinal prospective study. MICROBIAL CELL (GRAZ, AUSTRIA) 2024; 11:106-115. [PMID: 38638559 PMCID: PMC11026063 DOI: 10.15698/mic2024.03.819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 02/27/2024] [Accepted: 03/01/2024] [Indexed: 04/20/2024]
Abstract
Gut microbiota has complex immune functions, related to different pathologies, including multiple sclerosis (MS).This study evaluated the influence of treatments on gut microbiota in people with MS (PwMS). The research comprised 60 participants, including 39 PwMS and 21 healthy controls (HC). Among the PwMS, 20 were prescribed a disease-modifying therapy (DMT), either interferon beta1a or teriflunomide, while 19 received a combination of classical DMT and an immunoglobulin Y (IgY) supplement. For each participant, two sets of gut samples were collected: one at the study's outset and another after two months. Alpha and beta diversity analyses revealed no significant differences between groups. In comparison to the HC, the MS group exhibited an increase in Prevotella stercorea and a decrease in Faecalibacterium prausnitzii. Following treatment, individuals with MS showed enrichment in Lachnospiraceae and Streptococcus. The second sample, compared to the first one, demonstrated an increase in Bifidobacterium angulatum and a decrease in Oscillospira for individuals with MS. Gut microbiota diversity in PwMS is not significantly different to HC.However, specific taxonomic changes indicate the presence of a dysbiosis state. The use of DMTs and immunoglobulin Y supplements may contribute to alterations in microbial composition, potentially leading to the restoration of a healthier microbiome.
Collapse
Affiliation(s)
- Andreea-Cristina Paraschiv
- Department of Neurosciences, Faculty of Medicine, Iuliu Hațieganu University of Medicine and PharmacyCluj Napoca, 400012Romania
| | - Vitalie Vacaras
- Department of Neurosciences, Faculty of Medicine, Iuliu Hațieganu University of Medicine and PharmacyCluj Napoca, 400012Romania
- Neurology Department, Cluj Emergency County HospitalClujNapoca, 400012Romania
| | - Cristina Nistor
- Department of Neurosciences, Faculty of Medicine, Iuliu Hațieganu University of Medicine and PharmacyCluj Napoca, 400012Romania
- Neurology Department, Cluj Emergency County HospitalClujNapoca, 400012Romania
| | - Cristiana Vacaras
- Faculty of Medicine, Iuliu Hațieganu University of Medicine and PharmacyCluj Napoca, 400012Romania
| | - Stefan Strilciuc
- Department of Neurosciences, Faculty of Medicine, Iuliu Hațieganu University of Medicine and PharmacyCluj Napoca, 400012Romania
| | - Dafin F Muresanu
- Department of Neurosciences, Faculty of Medicine, Iuliu Hațieganu University of Medicine and PharmacyCluj Napoca, 400012Romania
- Neurology Department, Cluj Emergency County HospitalClujNapoca, 400012Romania
| |
Collapse
|
31
|
Wilson DR, Binford L, Hickson S. The Gut Microbiome and Mental Health. J Holist Nurs 2024; 42:79-87. [PMID: 37082808 DOI: 10.1177/08980101231170487] [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] [Indexed: 04/22/2023]
Abstract
The gut microbiome has been well researched in the past few years and may be a target for treating mental illness. Trillions of bacteria in the digestive system work with the brain, immune function, and endocrine pathways. This gut microbiome ecosystem mediates the interaction between the human being and the environment making its inclusion in holistic nursing essential. Changes in normal balance of the gut microbiome occur with diet, antibiotics and other medications, stress, cancer treatment, geography and environment, and current illnesses. When the microbiome is challenged a "dysbiotic" state leads to inadequate production of needed neurotransmitters such as serotonin and dopamine. Research has shown links between the dysbiosis, and the inflammatory response system that are known to contribute to depression, anxiety, and schizophrenia. Understanding the role of the gut microbiome can be beneficial to holistic nurses, providing a new tool to prevent, treat, or reduce symptoms of mental illness and improve general immune function. This innocuous holistic approach to mental wellness is becoming an important evidenced-based approach.
Collapse
Affiliation(s)
- Debra Rose Wilson
- Lenora C. Reuther Chair of Excellence. Austin Peay State University, Clarksville TN, Walden University, USA
| | | | | |
Collapse
|
32
|
Gradisteanu Pircalabioru G, Raileanu M, Dionisie MV, Lixandru-Petre IO, Iliescu C. Fast detection of bacterial gut pathogens on miniaturized devices: an overview. Expert Rev Mol Diagn 2024; 24:201-218. [PMID: 38347807 DOI: 10.1080/14737159.2024.2316756] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 02/06/2024] [Indexed: 03/23/2024]
Abstract
INTRODUCTION Gut microbes pose challenges like colon inflammation, deadly diarrhea, antimicrobial resistance dissemination, and chronic disease onset. Development of early, rapid and specific diagnosis tools is essential for improving infection control. Point-of-care testing (POCT) systems offer rapid, sensitive, low-cost and sample-to-answer methods for microbe detection from various clinical and environmental samples, bringing the advantages of portability, automation, and simple operation. AREAS COVERED Rapid detection of gut microbes can be done using a wide array of techniques including biosensors, immunological assays, electrochemical impedance spectroscopy, mass spectrometry and molecular biology. Inclusion of Internet of Things, machine learning, and smartphone-based point-of-care applications is an important aspect of POCT. In this review, the authors discuss various fast diagnostic platforms for gut pathogens and their main challenges. EXPERT OPINION Developing effective assays for microbe detection can be complex. Assay design must consider factors like target selection, real-time and multiplex detection, sample type, reagent stability and storage, primer/probe design, and optimizing reaction conditions for accuracy and sensitivity. Mitigating these challenges requires interdisciplinary collaboration among scientists, clinicians, engineers, and industry partners. Future efforts are essential to enhance sensitivity, specificity, and versatility of POCT systems for gut microbe detection and quantification, advancing infectious disease diagnostics and management.
Collapse
Affiliation(s)
- Gratiela Gradisteanu Pircalabioru
- eBio-hub Research Centre, National University of Science and Technology "Politehnica" Bucharest, Bucharest, Romania
- Division of Earth, Environmental and Life Sciences, The Research Institute of University of Bucharest (ICUB), Bucharest, Romania
- Academy of Romanian Scientists, Bucharest, Romania
| | - Mina Raileanu
- eBio-hub Research Centre, National University of Science and Technology "Politehnica" Bucharest, Bucharest, Romania
- Department of Life and Environmental Physics, Horia Hulubei National Institute of Physics and Nuclear Engineering, Magurele, Romania
| | - Mihai Viorel Dionisie
- eBio-hub Research Centre, National University of Science and Technology "Politehnica" Bucharest, Bucharest, Romania
| | - Irina-Oana Lixandru-Petre
- eBio-hub Research Centre, National University of Science and Technology "Politehnica" Bucharest, Bucharest, Romania
| | - Ciprian Iliescu
- eBio-hub Research Centre, National University of Science and Technology "Politehnica" Bucharest, Bucharest, Romania
- Academy of Romanian Scientists, Bucharest, Romania
- Microsystems in Biomedical and Environmental Applications, National Research and Development Institute for Microtechnology, Bucharest, Romania
| |
Collapse
|
33
|
Jin DM, Morton JT, Bonneau R. Meta-analysis of the human gut microbiome uncovers shared and distinct microbial signatures between diseases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.27.582333. [PMID: 38464323 PMCID: PMC10925178 DOI: 10.1101/2024.02.27.582333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Microbiome studies have revealed gut microbiota's potential impact on complex diseases. However, many studies often focus on one disease per cohort. We developed a meta-analysis workflow for gut microbiome profiles and analyzed shotgun metagenomic data covering 11 diseases. Using interpretable machine learning and differential abundance analysis, our findings reinforce the generalization of binary classifiers for Crohn's disease (CD) and colorectal cancer (CRC) to hold-out cohorts and highlight the key microbes driving these classifications. We identified high microbial similarity in disease pairs like CD vs ulcerative colitis (UC), CD vs CRC, Parkinson's disease vs type 2 diabetes (T2D), and schizophrenia vs T2D. We also found strong inverse correlations in Alzheimer's disease vs CD and UC. These findings detected by our pipeline provide valuable insights into these diseases.
Collapse
Affiliation(s)
- Dong-Min Jin
- Center for Genomics and Systems Biology, New York University, New York, NY, USA
| | - James T. Morton
- Center for Computational Biology, Flatiron Institute, Simons Foundation, New York, NY, USA
| | - Richard Bonneau
- Center for Genomics and Systems Biology, New York University, New York, NY, USA
- Genentech, New York, NY, USA
| |
Collapse
|
34
|
Huang H, Liu Y, Wen Z, Chen C, Wang C, Li H, Yang X. Gut microbiota in patients with prostate cancer: a systematic review and meta-analysis. BMC Cancer 2024; 24:261. [PMID: 38402385 PMCID: PMC10893726 DOI: 10.1186/s12885-024-12018-x] [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: 11/08/2023] [Accepted: 02/18/2024] [Indexed: 02/26/2024] Open
Abstract
BACKGROUND Increasing evidence indicates that gut microbiota are closely related to prostate cancer. This study aims to assess the gut microbiota composition in patients with prostate cancer compared to healthy participants, thereby advancing understanding of gut microbiota's role in prostate cancer. METHODS A systematic search was conducted across PubMed, Web of Science, and Embase databases, in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. The methodological quality of included studies was evaluated using the Newcastle-Ottawa Scale (NOS), and pertinent data were analyzed. The kappa score assessed interrater agreement. RESULTS This study encompassed seven research papers, involving 250 prostate cancer patients and 192 controls. The kappa was 0.93. Meta-analysis results showed that alpha-diversity of gut microbiota in prostate cancer patients was significantly lower than in the control group. In terms of gut microbiota abundance, the ratio of Proteobacteria, Bacteroidia, Clostridia, Bacteroidales, Clostridiales, Prevotellaceae, Lachnospiraceae, Prevotella, Escherichia-Shigella, Faecalibacterium, and Bacteroides was higher in prostate cancer patients. Conversely, the abundance ratio of Actinobacteria, Bacteroidetes, Firmicutes, Selenomonadales, Veillonella, and Megasphaera was higher in the control group. CONCLUSION Our study reveals differences in alpha-diversity and abundance of gut microbiota between patients with prostate cancer and controls, indicating gut microbiota dysbiosis in those with prostate cancer. However, given the limited quality and quantity of selected studies, further research is necessary to validate these findings.
Collapse
Affiliation(s)
- Haotian Huang
- Department of Urology, Afliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Yang Liu
- Department of Urology, Afliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Zhi Wen
- Department of Urology, Afliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Caixia Chen
- Department of Urology, Afliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Chongjian Wang
- Department of Urology, Afliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Hongyuan Li
- Department of Urology, Afliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xuesong Yang
- Department of Urology, Afliated Hospital of North Sichuan Medical College, Nanchong, China.
| |
Collapse
|
35
|
Wang XA, Li JP, Lee MS, Yang SF, Chang YS, Chen L, Li CW, Chao YH. A common trajectory of gut microbiome development during the first month in healthy neonates with limited inter-individual environmental variations. Sci Rep 2024; 14:3264. [PMID: 38332050 PMCID: PMC10853277 DOI: 10.1038/s41598-024-53949-6] [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: 09/04/2023] [Accepted: 02/07/2024] [Indexed: 02/10/2024] Open
Abstract
The early development of the gut microbiome is governed by multiple factors and has significantly long-term effects on later-in-life health. To minimize inter-individual variations in the environment, we determined developmental trajectories of the gut microbiome in 28 healthy neonates during their stay at a postpartum center. Stool samples were collected at three time points: the first-pass meconium within 24 h of life, and at 7 and 28 days of age. Illumina sequencing of the V3-V4 region of 16S rRNA was used to investigate microbiota profiles. We found that there was a distinct microbiota structure at each time point, with a significant shift during the first week. Proteobacteria was most abundant in the first-pass meconium; Firmicutes and Actinobacteria increased with age and were substituted as the major components. Except for a short-term influence of different delivery modes on the microbiota composition, early microbiome development was not remarkably affected by gravidity, maternal intrapartum antibiotic treatment, premature rupture of membranes, or postnatal phototherapy. Hence, our data showed a similar developmental trajectory of the gut microbiome during the first month in healthy neonates when limited in environmental variations. Environmental factors external to the host were crucial in the early microbiome development.
Collapse
Affiliation(s)
- Xing-An Wang
- Department of Pediatrics, Chung Shan Medical University Hospital, No. 110, Sec. 1, Chien-Kuo N. Road, Taichung, 402, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Ju-Pi Li
- Department of Pediatrics, Chung Shan Medical University Hospital, No. 110, Sec. 1, Chien-Kuo N. Road, Taichung, 402, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Maw-Sheng Lee
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Obstetrics and Gynecology, Lee Women's Hospital, Taichung, Taiwan
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Yi-Sheng Chang
- Department of Research and Development, AllBio Life Incorporation, Taichung, Taiwan
| | - Ling Chen
- Department of Research and Development, AllBio Life Incorporation, Taichung, Taiwan
| | - Chang-Wei Li
- Department of Research and Development, AllBio Life Incorporation, Taichung, Taiwan
| | - Yu-Hua Chao
- Department of Pediatrics, Chung Shan Medical University Hospital, No. 110, Sec. 1, Chien-Kuo N. Road, Taichung, 402, Taiwan.
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan.
- Department of Clinical Pathology, Chung Shan Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
36
|
Zhao W, Kodancha P, Das S. Gut Microbiome Changes in Anorexia Nervosa: A Comprehensive Review. PATHOPHYSIOLOGY 2024; 31:68-88. [PMID: 38390943 PMCID: PMC10885100 DOI: 10.3390/pathophysiology31010006] [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: 11/24/2023] [Revised: 01/03/2024] [Accepted: 01/18/2024] [Indexed: 02/24/2024] Open
Abstract
Anorexia nervosa (AN) remains a challenging condition in psychiatric management and its pathogenesis is not yet fully understood. An imbalance in the gut microbiota composition may contribute to its pathophysiology. This review aims to explore the link between the human gut microbiota and AN (objective 1) or refeeding syndrome in AN (objective 2). The online databases MEDLINE and PsycINFO were searched for relevant studies. A total of 14 studies met the inclusion and exclusion criteria and only answered objective 1. A total of 476 AN patients, 554 healthy-weight (HC) controls, and 0 patients with other psychiatric disorders were included. Compared to HC, there were consistently reduced abundances of Faecalibacterium prausnitzii and Roseburia inulinivorans, and increased Methanobrevibacter smithii, in AN patients. Changes in alpha diversity were inconsistent, while beta diversity increased in four of six studies. Our model suggests that an imbalance in gut microbiota composition leads to reduced short-chain fatty acids, contributing to a proinflammatory state in AN, which is also common in other psychiatric comorbidities. Microbial changes may also contribute to the semistarvation state through endocrine changes and altered energy utilization.
Collapse
Affiliation(s)
- Wendi Zhao
- Department of Psychiatry, University of Melbourne, Parkville, Melbourne 3052, Australia
| | | | - Soumitra Das
- Unit of Psychiatry, Western Health, Melbourne 3021, Australia
| |
Collapse
|
37
|
Álvarez-Herms J, González-Benito A, Corbi F, Odriozola A. What if gastrointestinal complications in endurance athletes were gut injuries in response to a high consumption of ultra-processed foods? Please take care of your bugs if you want to improve endurance performance: a narrative review. Eur J Appl Physiol 2024; 124:383-402. [PMID: 37839038 DOI: 10.1007/s00421-023-05331-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 09/20/2023] [Indexed: 10/17/2023]
Abstract
To improve performance and recovery faster, athletes are advised to eat more often than usual and consume higher doses of simple carbohydrates, during and after exercise. Sports energetic supplements contain food additives, such as artificial sweeteners, emulsifiers, acidity regulators, preservatives, and salts, which could be harmful to the gut microbiota and impair the intestinal barrier function. The intestinal barrier plays a critical function in bidirectionally regulation of the selective transfer of nutrients, water, and electrolytes, while preventing at the same time, the entrance of harmful substances (selective permeability). The gut microbiota helps to the host to regulate intestinal homeostasis through metabolic, protective, and immune functions. Globally, the gut health is essential to maintain systemic homeostasis in athletes, and to ensure proper digestion, metabolization, and substrate absorption. Gastrointestinal complaints are an important cause of underperformance and dropout during endurance events. These complications are directly related to the loss of gut equilibrium, mainly linked to microbiota dysbiosis and leaky gut. In summary, athletes must be cautious with the elevated intake of ultra-processed foods and specifically those contained on sports nutrition supplements. This review points out the specific nutritional interventions that should be implemented and/or discontinued depending on individual gut functionality.
Collapse
Affiliation(s)
- Jesús Álvarez-Herms
- Phymolab (Physiology and Molecular Laboratory), Collado Hermoso, Segovia, Spain.
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Bilbao, Spain.
| | - A González-Benito
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Bilbao, Spain
| | - F Corbi
- Institut Nacional d'Educació Física de Catalunya (INEFC), University of Lleida (UdL), Lleida, Spain
| | - A Odriozola
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Bilbao, Spain
| |
Collapse
|
38
|
Ruiz-Valdepeñas Montiel V, Vargas E, Ben Hassine A, Simon I, Duvvuri A, Chang AY, Nandhakumar P, Bulbarello A, Düsterloh A, Mak T, Wang J. Decentralized ORP Measurements for Gut Redox Status Monitoring: Toward Personalized Gut Microbiota Balance. Anal Chem 2024; 96:480-487. [PMID: 38150379 DOI: 10.1021/acs.analchem.3c04570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
Gut microbiome targeting has emerged as a new generation of personalized medicine and a potential wellness and disease driver. Specifically, the gut redox balance plays a key role in shaping the gut microbiota and its link with the host, immune system, and disease evolution. In this sense, precise and personalized nutrition has proven synergy and capability to modulate the gut microbiome environment through the formulation of dietary interventions, such as vitamin support. Accordingly, there are urgent demands for simple and effective analytical platforms for understanding the relationship between the tailored vitamin administration and the gut microbiota balance by rapid noninvasive on-the-spot oxidation/reduction potential monitoring for frequent and close surveillance of the gut redox status and targeting by personalized nutrition interventions. Herein, we present a disposable potentiometric sensor chip and a homemade multiwell potentiometric array to address the interplay of vitamin levels with the oxidation/reduction potential in human feces and saliva. The potentiometric ORP sensing platforms have been successfully validated and scaled up for the setup of a multiapplication prototype for cross-talk-free simple screening of many specimens. The interpersonal variability of the gut microbiota environment illustrates the potential of feces and saliva samples for noninvasive, frequent, and decentralized monitoring of the gut redox status to support timely human microbiota surveillance and guide precise dietary intervention toward restoring and promoting personalized gut redox balance.
Collapse
Affiliation(s)
- Víctor Ruiz-Valdepeñas Montiel
- Department of Nanoengineering, University of California San Diego, La Jolla, California 92093, United States
- Department of Analytical Chemistry, Chemistry Faculty, University Complutense of Madrid, E-28040 Madrid, Spain
| | - Eva Vargas
- Department of Nanoengineering, University of California San Diego, La Jolla, California 92093, United States
| | - Amira Ben Hassine
- Department of Nanoengineering, University of California San Diego, La Jolla, California 92093, United States
| | - Ignasi Simon
- Department of Nanoengineering, University of California San Diego, La Jolla, California 92093, United States
| | - Andres Duvvuri
- Department of Nanoengineering, University of California San Diego, La Jolla, California 92093, United States
| | - An-Yi Chang
- Department of Nanoengineering, University of California San Diego, La Jolla, California 92093, United States
| | - Ponnusamy Nandhakumar
- Department of Nanoengineering, University of California San Diego, La Jolla, California 92093, United States
| | | | | | - Tim Mak
- DSM-Firmenich AG, Kaiseraugst 4303, Switzerland
| | - Joseph Wang
- Department of Nanoengineering, University of California San Diego, La Jolla, California 92093, United States
| |
Collapse
|
39
|
Niederhuber JE. The microbiome and cancer immunotherapy. CLINICAL IMMUNO-ONCOLOGY 2024:223-236.e4. [DOI: 10.1016/b978-0-323-87763-3.00020-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
40
|
Fanfan D, Mulligan CJ, Groer M, Mai V, Weaver M, Huffman F, Lyon DE. The intersection of social determinants of health, the microbiome, and health outcomes in immigrants: A scoping review. AMERICAN JOURNAL OF BIOLOGICAL ANTHROPOLOGY 2024; 183:3-19. [PMID: 37737631 PMCID: PMC11185843 DOI: 10.1002/ajpa.24850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 08/23/2023] [Accepted: 09/03/2023] [Indexed: 09/23/2023]
Abstract
In the present scoping review, we explore whether existing evidence supports the premise that social determinants of health (SDoH) affect immigrant health outcomes through their effects on the microbiome. We adapt the National Institute on Minority Health and Health Disparities' research framework to propose a conceptual model that considers the intersection of SDoH, the microbiome, and health outcomes in immigrants. We use this conceptual model as a lens through which to explore recent research about SDoH, biological factors associated with changes to immigrants' microbiomes, and long-term health outcomes. In the 17 articles reviewed, dietary acculturation, physical activity, ethnicity, birthplace, age at migration and length of time in the host country, socioeconomic status, and social/linguistic acculturation were important determinants of postmigration microbiome-related transformations. These factors are associated with progressive shifts in microbiome profile with time in host country, increasing the risks for cardiometabolic, mental, immune, and inflammatory disorders and antibiotic resistance. The evidence thus supports the premise that SDoH influence immigrants' health postmigration, at least in part, through their effects on the microbiome. Omission of important postmigration social-ecological variables (e.g., stress, racism, social/family relationships, and environment), limited research among minoritized subgroups of immigrants, complexity and inter- and intra-individual differences in the microbiome, and limited interdisciplinary and biosocial collaboration restrict our understanding of this area of study. To identify potential microbiome-based interventions and promote immigrants' well-being, more research is necessary to understand the intersections of immigrant health with factors from the biological, behavioral/psychosocial, physical/built environment, and sociocultural environment domains at all social-ecological levels.
Collapse
Affiliation(s)
- Dany Fanfan
- College of Nursing, University of Florida, Gainesville, Florida, USA
| | - Connie J. Mulligan
- Department of Anthropology, University of Florida, Gainesville, Florida, USA
- Genetics Institute, University of Florida, Gainesville, Florida, USA
| | - Maureen Groer
- College of Nursing, University of Tennessee, Knoxville, Tennessee, USA
| | - Volker Mai
- College of Public Health and Health Professions and College of Medicine, Emerging Pathogens Institute, University of Florida, Gainesville, Florida, USA
| | - Michael Weaver
- College of Nursing, University of Florida, Gainesville, Florida, USA
| | - Fatma Huffman
- College of Public Health and Social Work, Florida International University, Miami, Florida, USA
| | - Debra E. Lyon
- College of Nursing, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
41
|
Mohanan A, Harilal SL, Plakkot B, Pottakkat B, Kanakkaparambil R. Nutritional Epigenetics and Gut Microbiome. EPIGENETICS AND HUMAN HEALTH 2024:121-159. [DOI: 10.1007/978-3-031-54215-2_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
42
|
DI Pierro F, Zerbinati N, Guasti L, Bertuccioli A, Cazzaniga M, Gerardi V, Piccirelli S, Salvi D, Pugliano CL, Cesaro P, Spada C. Can the analysis of the gut microbiota have a clinical application in real life? Minerva Gastroenterol (Torino) 2023; 69:576-580. [PMID: 37439694 DOI: 10.23736/s2724-5985.23.03499-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Affiliation(s)
- Francesco DI Pierro
- Scientific Department, Velleja Research, Milan, Italy -
- Department of Medicine and Surgery, University of Insubria, Varese, Italy -
| | - Nicola Zerbinati
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Luigina Guasti
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | | | | | - Viviana Gerardi
- Digestive Endoscopy and Gastroenterology, Poliambulanza Foundation, Brescia, Italy
| | - Stefania Piccirelli
- Digestive Endoscopy and Gastroenterology, Poliambulanza Foundation, Brescia, Italy
| | - Daniele Salvi
- Digestive Endoscopy and Gastroenterology, Poliambulanza Foundation, Brescia, Italy
| | - Cecilia L Pugliano
- Digestive Endoscopy and Gastroenterology, Poliambulanza Foundation, Brescia, Italy
| | - Paola Cesaro
- Digestive Endoscopy and Gastroenterology, Poliambulanza Foundation, Brescia, Italy
| | - Cristiano Spada
- Unit of Digestive Endoscopy, IRCCS A. Gemelli University Polyclinic Foundation, Rome, Italy
| |
Collapse
|
43
|
Farooqi T, Bhuyan DJ, Low M, Sinclair J, Leonardi M, Armour M. Cannabis and Endometriosis: The Roles of the Gut Microbiota and the Endocannabinoid System. J Clin Med 2023; 12:7071. [PMID: 38002684 PMCID: PMC10671947 DOI: 10.3390/jcm12227071] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 10/31/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
Endometriosis, a chronic condition affecting around 10-14% of women, is challenging to manage, due to its complex pathogenesis and limited treatment options. Research has suggested a potential role of the gut microbiota and the endocannabinoid system in the development and progression of endometriosis. This narrative review aims to explore the role of, and any potential interactions between, the endocannabinoid system (ECS) and the gut microbiota in endometriosis. This review found that both the ECS and microbiota influence endometriosis, with the former regulating inflammation and pain perception and the latter influencing immune responses and hormonal balance. There is evidence that a dysregulation of the endocannabinoid system and the gut microbiota influence endometriosis symptoms and progression via changes in CB1 receptor expression and increased circulating levels of endocannabinoids. Microbial imbalances in the gut, such as increases in Prevotella, have been directly correlated to increased bloating, a common endometriosis symptom, while increases in E. coli have supported the bacterial contamination hypothesis as a potential pathway for endometriosis pathogenesis. These microbial imbalances have been correlated with increases in inflammatory markers such as TNF-α and IL-6, both often raised in those with endometriosis. Protective effects of the ECS on the gut were observed by increases in endocannabinoids, including 2-AG, resulting in decreased inflammation and improved gut permeability. Given these findings, both the ECS and the gut microbiota may be targets for therapeutic interventions for endometriosis; however, clinical studies are required to determine effectiveness.
Collapse
Affiliation(s)
- Toobah Farooqi
- NICM Health Research Institute, Western Sydney University, Sydney 2751, Australia; (T.F.); (D.J.B.); (M.L.); (J.S.)
| | - Deep Jyoti Bhuyan
- NICM Health Research Institute, Western Sydney University, Sydney 2751, Australia; (T.F.); (D.J.B.); (M.L.); (J.S.)
- School of Science, Western Sydney University, Sydney 2751, Australia
| | - Mitchell Low
- NICM Health Research Institute, Western Sydney University, Sydney 2751, Australia; (T.F.); (D.J.B.); (M.L.); (J.S.)
| | - Justin Sinclair
- NICM Health Research Institute, Western Sydney University, Sydney 2751, Australia; (T.F.); (D.J.B.); (M.L.); (J.S.)
| | - Mathew Leonardi
- Department of Obstetrics and Gynecology, McMaster University, Hamilton, ON L8V 5C2, Canada;
- Robinson Research Institute, University of Adelaide, Adelaide 5006, Australia
| | - Mike Armour
- NICM Health Research Institute, Western Sydney University, Sydney 2751, Australia; (T.F.); (D.J.B.); (M.L.); (J.S.)
- Translational Health Research Institute, Western Sydney University, Sydney 2751, Australia
- Medical Research Institute of New Zealand, P.O. Box 7902, Wellington 6242, New Zealand
| |
Collapse
|
44
|
Russell MW, Muste JC, Kuo BL, Wu AK, Singh RP. Clinical trials targeting the gut-microbiome to effect ocular health: a systematic review. Eye (Lond) 2023; 37:2877-2885. [PMID: 36918627 PMCID: PMC10516887 DOI: 10.1038/s41433-023-02462-7] [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: 03/25/2022] [Revised: 11/21/2022] [Accepted: 02/21/2023] [Indexed: 03/16/2023] Open
Abstract
Clinical trials targeting the gut microbiome to mitigate ocular disease are now on the horizon. A review of clinical data thus far is essential to determine future directions in this novel promising field. This review examines recent clinical trials that support the plausibility of a gut-eye axis, and may form the basis of novel clinical interventions. PubMed was queried for English language clinical studies examining the relationships between gut microbiota and ocular pathology. 25 studies were extracted from 828 candidate publications, which suggest that gut imbalance is associated with ocular pathology. Of these, only four interventional studies exist which suggest probiotic supplementation or fecal microbiota transplant can reduce symptoms of chalazion or uveitis. The gut-eye axis appears to hold clinical relevance, but current data is limited in sample size and design. Further investigation via longitudinal clinical trials may be warranted.
Collapse
Affiliation(s)
- Matthew W Russell
- Center for Ophthalmic Bioinformatics, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH, USA
| | - Justin C Muste
- Center for Ophthalmic Bioinformatics, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Blanche L Kuo
- Center for Ophthalmic Bioinformatics, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA
- Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Anna K Wu
- Center for Ophthalmic Bioinformatics, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA
- Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Rishi P Singh
- Center for Ophthalmic Bioinformatics, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
45
|
Nayman EI, Schwartz BA, Polanco FC, Firek AK, Gumabong AC, Hofstee NJ, Narasimhan G, Cickovski T, Mathee K. Microbiome depiction through user-adapted bioinformatic pipelines and parameters. J Med Microbiol 2023; 72. [PMID: 37823280 DOI: 10.1099/jmm.0.001756] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2023] Open
Abstract
Introduction. The role of the microbiome in health and disease continues to be increasingly recognized. However, there is significant variability in the bioinformatic protocols for analysing genomic data. This, in part, has impeded the potential incorporation of microbiomics into the clinical setting and has challenged interstudy reproducibility. In microbial compositional analysis, there is a growing recognition for the need to move away from a one-size-fits-all approach to data processing.Gap Statement. Few evidence-based recommendations exist for setting parameters of programs that infer microbiota community profiles despite these parameters significantly impacting the accuracy of taxonomic inference.Aim. To compare three commonly used programs (DADA2, QIIME2, and mothur) and optimize them into four user-adapted pipelines for processing paired-end amplicon reads. We aim to increase the accuracy of compositional inference and help standardize microbiomic protocol.Methods. Two key parameters were isolated across four pipelines: filtering sequence reads based on a whole-number error threshold (maxEE) and truncating read ends based on a quality score threshold (QTrim). Closeness of sample inference was then evaluated using a mock community of known composition.Results. We observed that raw genomic data lost were proportionate to how stringently parameters were set. Exactly how much data were lost varied by pipeline. Accuracy of sample inference correlated with increased sequence read retention. Falsely detected taxa and unaccounted for microbial constituents were unique to pipeline and parameter. Implementation of optimized parameter values led to better approximation of the known mock community.Conclusions. Microbial compositions generated based on the 16S rRNA marker gene should be interpreted with caution. To improve microbial community profiling, bioinformatic protocols must be user-adapted. Analysis should be performed with consideration for the select target amplicon, pipelines and parameters used, and taxa of interest.
Collapse
Affiliation(s)
- Eric I Nayman
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
- Bioinformatics Research Group (BioRG), Knight Foundation School of Computing and Information Sciences, Florida International University, Miami, FL, USA
| | - Brooke A Schwartz
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
- Bioinformatics Research Group (BioRG), Knight Foundation School of Computing and Information Sciences, Florida International University, Miami, FL, USA
| | - Fantaysia C Polanco
- Bioinformatics Research Group (BioRG), Knight Foundation School of Computing and Information Sciences, Florida International University, Miami, FL, USA
| | - Alexandra K Firek
- Translational Glycobiology Institute, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Alayna C Gumabong
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Nolan J Hofstee
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Giri Narasimhan
- Bioinformatics Research Group (BioRG), Knight Foundation School of Computing and Information Sciences, Florida International University, Miami, FL, USA
- Biomolecular Sciences Institute, Florida International University, Miami, FL, USA
| | - Trevor Cickovski
- Bioinformatics Research Group (BioRG), Knight Foundation School of Computing and Information Sciences, Florida International University, Miami, FL, USA
| | - Kalai Mathee
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
- Biomolecular Sciences Institute, Florida International University, Miami, FL, USA
| |
Collapse
|
46
|
Rieg T, Xue J, Stevens M, Thomas L, White J, Dominguez Rieg J. Intravenous ferric carboxymaltose and ferric derisomaltose alter the intestinal microbiome in female iron-deficient anemic mice. Biosci Rep 2023; 43:BSR20231217. [PMID: 37671923 PMCID: PMC10520285 DOI: 10.1042/bsr20231217] [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: 07/08/2023] [Revised: 08/31/2023] [Accepted: 09/06/2023] [Indexed: 09/07/2023] Open
Abstract
Iron deficiency anemia (IDA) is a leading global health concern affecting approximately 30% of the population. Treatment for IDA consists of replenishment of iron stores, either by oral or intravenous (IV) supplementation. There is a complex bidirectional interplay between the gut microbiota, the host's iron status, and dietary iron availability. Dietary iron deficiency and supplementation can influence the gut microbiome; however, the effect of IV iron on the gut microbiome is unknown. We studied how commonly used IV iron preparations, ferric carboxymaltose (FCM) and ferric derisomaltose (FDI), affected the gut microbiome in female iron-deficient anemic mice. At the phylum level, vehicle-treated mice showed an expansion in Verrucomicrobia, mostly because of the increased abundance of Akkermansia muciniphila, along with contraction in Firmicutes, resulting in a lower Firmicutes/Bacteroidetes ratio (indicator of dysbiosis). Treatment with either FCM or FDI restored the microbiome such that Firmicutes and Bacteroidetes were the dominant phyla. Interestingly, the phyla Proteobacteria and several members of Bacteroidetes (e.g., Alistipes) were expanded in mice treated with FCM compared with those treated with FDI. In contrast, several Clostridia class members were expanded in mice treated with FDI compared with FCM (e.g., Dorea spp., Eubacterium). Our data demonstrate that IV iron increases gut microbiome diversity independently of the iron preparation used; however, differences exist between FCM and FDI treatments. In conclusion, replenishing iron stores with IV iron preparations in clinical conditions, such as inflammatory bowel disease or chronic kidney disease, could affect gut microbiome composition and consequently contribute to an altered disease outcome.
Collapse
Affiliation(s)
- Timo Rieg
- Department of Molecular Pharmacology and Physiology, Hypertension and Kidney Research Center, University of South Florida, Tampa, FL 33612, U.S.A
- James A. Haley Veterans’ Hospital, Tampa, FL 33612, U.S.A
| | - Jianxiang Xue
- Department of Molecular Pharmacology and Physiology, Hypertension and Kidney Research Center, University of South Florida, Tampa, FL 33612, U.S.A
| | - Monica Stevens
- Department of Molecular Pharmacology and Physiology, Hypertension and Kidney Research Center, University of South Florida, Tampa, FL 33612, U.S.A
| | - Linto Thomas
- Department of Molecular Pharmacology and Physiology, Hypertension and Kidney Research Center, University of South Florida, Tampa, FL 33612, U.S.A
| | | | - Jessica A. Dominguez Rieg
- Department of Molecular Pharmacology and Physiology, Hypertension and Kidney Research Center, University of South Florida, Tampa, FL 33612, U.S.A
- James A. Haley Veterans’ Hospital, Tampa, FL 33612, U.S.A
| |
Collapse
|
47
|
Arıkan M, Muth T. Integrated multi-omics analyses of microbial communities: a review of the current state and future directions. Mol Omics 2023; 19:607-623. [PMID: 37417894 DOI: 10.1039/d3mo00089c] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/08/2023]
Abstract
Integrated multi-omics analyses of microbiomes have become increasingly common in recent years as the emerging omics technologies provide an unprecedented opportunity to better understand the structural and functional properties of microbial communities. Consequently, there is a growing need for and interest in the concepts, approaches, considerations, and available tools for investigating diverse environmental and host-associated microbial communities in an integrative manner. In this review, we first provide a general overview of each omics analysis type, including a brief history, typical workflow, primary applications, strengths, and limitations. Then, we inform on both experimental design and bioinformatics analysis considerations in integrated multi-omics analyses, elaborate on the current approaches and commonly used tools, and highlight the current challenges. Finally, we discuss the expected key advances, emerging trends, potential implications on various fields from human health to biotechnology, and future directions.
Collapse
Affiliation(s)
- Muzaffer Arıkan
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey.
- Department of Medical Biology, Faculty of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Thilo Muth
- Section eScience (S.3), Federal Institute for Materials Research and Testing (BAM), Berlin, Germany.
| |
Collapse
|
48
|
Horseman TS, Frank AM, Shupp JW, Burmeister DM. Meta-Analysis of Publicly Available Clinical and Preclinical Microbiome Data From Studies of Burn Injury. J Burn Care Res 2023; 44:1041-1050. [PMID: 37352011 DOI: 10.1093/jbcr/irad098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Indexed: 06/25/2023]
Abstract
Following burn injury, alterations in host commensal microbiota across body spaces may leave patients susceptible to opportunistic pathogens and serious sequelae such as sepsis. Generally, studies examining the microbiome postburn have had a limited sample size and lack of longitudinal data, which coupled with experimental and analytic variation, impacts overall interpretation. We performed a meta-analysis of publicly available sequencing data from preclinical and clinical burn studies to determine if there were consistent alterations in the microbiome across various anatomical sites and hosts. Ten human and animal 16S rRNA sequencing studies spanning respiratory, urinary, cutaneous, and gastrointestinal microbiomes were included. Taxonomic classification and alpha and beta diversity metrics were analyzed using QIIME2 v2021.8. Alpha diversity was consistently higher in control samples compared to burn-injured samples which were also different based on host and anatomical location; however, phylogenetic evaluation (ie, Faith PD) elucidated more significant differences compared to taxonomic metrics (ie, Shannon entropy). Beta diversity analysis based on weighted UniFrac showed that rodent specimens clustered less closely to humans than pig samples for both rectal and skin sources. Host species and performing institute were found to have a significant impact on community structure. In rectal samples, bacterial composition in pig and human burn samples included Bacteroidetes, Firmicutes, and Proteobacteria, while rodent samples were dominated by Firmicutes. Proteobacteria and Firmicutes increased on burned skin in each host species. Our results suggest that host species and the performing institute strongly influence microbiome structure. Burn-induced alterations in microbiome diversity and taxa exist across hosts, with phylogenetic metrics more valuable than others. Coordinated, multicenter studies, both clinical and preclinical, within the burn community are needed to more completely realize the diagnostic and therapeutic potential of the microbiome for improving outcomes postburn.
Collapse
Affiliation(s)
- Timothy S Horseman
- School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Andrew M Frank
- School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Jeffrey W Shupp
- The Burn Center, MedStar Washington Hospital Center, Washington, DC, USA
- Department of Surgery, Georgetown University School of Medicine, Washington, DC, USA
| | - David M Burmeister
- School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| |
Collapse
|
49
|
Damianos J, Perumareddi P. Gut Microbiome and Dietar Considerations. Prim Care 2023; 50:493-505. [PMID: 37516516 DOI: 10.1016/j.pop.2023.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/31/2023]
Abstract
The gut microbiome represents a complex microbial ecosystem that exerts direct and indirect effects on other organ systems and contributes to both health and disease. It is sensitive to various stimuli such as childhood immunity, medications, diet, stressors, and sleep. Modulating the gut microbiome can prevent and even treat certain disease states. Although no definitive guidelines exist to support a healthy microbiome, there are several evidence-based interventions proved to improve gut health and reduce the risk for numerous chronic diseases.
Collapse
Affiliation(s)
- John Damianos
- Division of Gastroenterology and Hepatology, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA
| | - Parvathi Perumareddi
- Department of Medicine, Florida Atlantic University, Boca Raton, FL, USA; Charles E Schmidt College of Medicine- Florida Atlantic University, 777 Glades Road, Boca Raton, FL 33431, USA.
| |
Collapse
|
50
|
Metafuni E, Di Marino L, Giammarco S, Bellesi S, Limongiello MA, Sorà F, Frioni F, Maggi R, Chiusolo P, Sica S. The Role of Fecal Microbiota Transplantation in the Allogeneic Stem Cell Transplant Setting. Microorganisms 2023; 11:2182. [PMID: 37764025 PMCID: PMC10536954 DOI: 10.3390/microorganisms11092182] [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: 07/30/2023] [Revised: 08/16/2023] [Accepted: 08/23/2023] [Indexed: 09/29/2023] Open
Abstract
Microbiota changes during allogeneic hematopoietic stem cell transplantation has several known causes: conditioning chemotherapy and radiation, broad-spectrum antibiotic administration, modification in nutrition status and diet, and graft-versus-host disease. This article aims to review the current knowledge about the close link between microbiota and allogeneic stem cell transplantation setting. The PubMed search engine was used to perform this review. We analyzed data on microbiota dysbiosis related to the above-mentioned affecting factors. We also looked at treatments aimed at modifying gut dysbiosis and applications of fecal microbiota transplantation in the allogeneic stem cell transplant field, with particular interest in fecal microbiota transplantation for graft-versus-host disease (GvHD), multidrug-resistant and clostridium difficile infections, and microbiota restoration after chemotherapy and antibiotic therapy.
Collapse
Affiliation(s)
- Elisabetta Metafuni
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica e Ematologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (S.G.); (S.B.); (M.A.L.); (F.S.); (P.C.); (S.S.)
| | - Luca Di Marino
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (L.D.M.); (F.F.); (R.M.)
| | - Sabrina Giammarco
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica e Ematologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (S.G.); (S.B.); (M.A.L.); (F.S.); (P.C.); (S.S.)
| | - Silvia Bellesi
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica e Ematologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (S.G.); (S.B.); (M.A.L.); (F.S.); (P.C.); (S.S.)
| | - Maria Assunta Limongiello
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica e Ematologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (S.G.); (S.B.); (M.A.L.); (F.S.); (P.C.); (S.S.)
| | - Federica Sorà
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica e Ematologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (S.G.); (S.B.); (M.A.L.); (F.S.); (P.C.); (S.S.)
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (L.D.M.); (F.F.); (R.M.)
| | - Filippo Frioni
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (L.D.M.); (F.F.); (R.M.)
| | - Roberto Maggi
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (L.D.M.); (F.F.); (R.M.)
| | - Patrizia Chiusolo
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica e Ematologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (S.G.); (S.B.); (M.A.L.); (F.S.); (P.C.); (S.S.)
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (L.D.M.); (F.F.); (R.M.)
| | - Simona Sica
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica e Ematologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (S.G.); (S.B.); (M.A.L.); (F.S.); (P.C.); (S.S.)
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (L.D.M.); (F.F.); (R.M.)
| |
Collapse
|