1
|
Silveira MAD, Rodrigues RR, Trinchieri G. Intestinal Microbiome Modulation of Therapeutic Efficacy of Cancer Immunotherapy. Gastroenterol Clin North Am 2025; 54:295-315. [PMID: 40348489 PMCID: PMC12066836 DOI: 10.1016/j.gtc.2024.10.005] [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] [Indexed: 05/14/2025]
Abstract
Bacteria are associated with certain cancers and may induce genetic instability and cancer progression. The gut microbiome modulates the response to cancer therapy. Training machine learning models with response associated taxa or bacterial genes predict patients' response to immunotherapies with moderate accuracy. Clinical trials targeting the gut microbiome to improve immunotherapy efficacy have been conducted. While single bacterial strains or small consortia have not be reported yet to be successful, encouraging results have been reported in small single arm and randomized studies using transplant of fecal microbiome from cancer patients who successfully responded to therapy or from healthy volunteers.
Collapse
Affiliation(s)
- Maruhen A D Silveira
- Cancer Immunobiology Section, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Building 37, Room 4146, Bethesda, MD 20852, USA
| | - Richard R Rodrigues
- Microbiome and Genetics Core, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Building 37, Room 4140B, Bethesda, MD 20852, USA; Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA
| | - Giorgio Trinchieri
- Cancer Immunobiology Section, Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Building 37, Room 4146, Bethesda, MD 20852, USA.
| |
Collapse
|
2
|
Langer-Gould AM, Cepon-Robins TJ, Benn Torres J, Yeh EA, Gildner TE. Embodiment of structural racism and multiple sclerosis risk and outcomes in the USA. Nat Rev Neurol 2025:10.1038/s41582-025-01096-5. [PMID: 40425864 DOI: 10.1038/s41582-025-01096-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/28/2025] [Indexed: 05/29/2025]
Abstract
Disparities in the incidence, prevalence and outcomes of multiple sclerosis (MS) exist in the USA, often to the detriment of Black and Hispanic people. Despite the common misconception that MS is a disease of white people, the incidence is highest in Black people. Disability accumulates faster and at younger ages in Black and Hispanic people with MS than in their white counterparts, and MS-related mortality in early and mid-adulthood is highest in Black people. These differences are often erroneously interpreted as evidence of innate racial or ethnic variations. In this Perspective, we demonstrate how race and ethnicity - social constructs with a limited biological basis that are often assigned by systems of power - can influence biology through lived experiences, a phenomenon termed 'embodiment'. We review how downstream consequences of structural racism can lead to biological outcomes strongly associated with MS susceptibility, such as imbalanced immune system development, dysregulated immune responses to the Epstein-Barr virus and childhood obesity. We also consider how inequitable health-care access and quality, combined with the younger age of onset and higher comorbidity burdens, might explain racial and ethnic disparities in MS prognosis. Our proposed conceptual model offers a roadmap for generating knowledge and implementing interventions to narrow racial and ethnic disparities in MS susceptibility and outcomes.
Collapse
Affiliation(s)
- Annette M Langer-Gould
- Department of Neurology, Los Angeles Medical Center, Southern California Permanente Medical Group, Los Angeles, CA, USA.
- Department of Clinical Science, Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena, CA, USA.
| | - Tara J Cepon-Robins
- Department of Anthropology, University of Colorado Colorado Springs, Colorado Springs, CO, USA
| | - Jada Benn Torres
- Department of Anthropology, Genetic Anthropology and Biocultural Studies Laboratory, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - E Ann Yeh
- Department of Paediatrics Neurology, University of Toronto, Toronto, Ontario, Canada
- Paediatric MS and Neuroinflammatory Disorders Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Program in Neuroscience and Mental Health, SickKids Research Institute, Toronto, Ontario, Canada
| | - Theresa E Gildner
- Department of Anthropology, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
3
|
Laue HE, Willis AD, Wang F, MacDougall MC, Xu Y, Karagas MR, Madan JC, Fleisch AF, Lanphear BP, Cecil KM, Yolton K, Chen A, Buckley JP, Braun JM. Early-life and concurrent predictors of the healthy adolescent microbiome in a cohort study. Genome Med 2025; 17:50. [PMID: 40340756 PMCID: PMC12060534 DOI: 10.1186/s13073-025-01481-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 04/25/2025] [Indexed: 05/10/2025] Open
Abstract
BACKGROUND The microbiome of adolescents is poorly understood, as are factors influencing its composition. We aimed to describe the healthy adolescent microbiome and identify early-life and concurrent predictors of its composition. METHODS We performed metagenomic sequencing of 247 fecal specimens from 167 adolescents aged 11-14 years participating in the Health Outcomes and Measures of the Environment (HOME) Study, a longitudinal pregnancy and birth cohort (Cincinnati, OH). We described common features of the adolescent gut microbiome and applied self-organizing maps (SOMs)-a machine-learning approach-to identify distinct microbial profiles (n = 4). Using prospectively collected data on sociodemographic characteristics, lifestyle, diet, and sexual maturation, we identified early-life and concurrent factors associated with microbial diversity and phylum relative abundance with linear regression models and composition with Kruskal-Wallis and Fisher's exact tests. RESULTS We found that household income and other sociodemographic factors were consistent predictors of the microbiome, with higher income associated with lower diversity and differential relative abundances of Firmicutes (increased) and Actinobacteria (decreased). Sexual maturation, distinct from chronological age, was related to higher diversity in females and differences in phylum relative abundances and compositional profiles in both males and females. CONCLUSIONS Our study suggests that adolescence is a unique window for gut microbial composition and that it may be shaped by both early-life and concurrent exposures, highlighting its potential in future epidemiologic research.
Collapse
Grants
- K99/R00ES034086 , P01ES011261, R01ES0272244, R01ES025214 National Institute of Environmental Health Sciences,United States
- K99/R00ES034086 , P01ES011261, R01ES0272244, R01ES025214 National Institute of Environmental Health Sciences,United States
- K99/R00ES034086 , P01ES011261, R01ES0272244, R01ES025214 National Institute of Environmental Health Sciences,United States
- K99/R00ES034086 , P01ES011261, R01ES0272244, R01ES025214 National Institute of Environmental Health Sciences,United States
- K99/R00ES034086 , P01ES011261, R01ES0272244, R01ES025214 National Institute of Environmental Health Sciences,United States
- K99/R00ES034086 , P01ES011261, R01ES0272244, R01ES025214 National Institute of Environmental Health Sciences,United States
- K99/R00ES034086 , P01ES011261, R01ES0272244, R01ES025214 National Institute of Environmental Health Sciences,United States
- K99/R00ES034086 , P01ES011261, R01ES0272244, R01ES025214 National Institute of Environmental Health Sciences,United States
- R35GM133420 NIGMS NIH HHS
- UL1TR001425 NCATS NIH HHS
- UL1TR001425 NCATS NIH HHS
- UL1TR001425 NCATS NIH HHS
- UL1TR001425 NCATS NIH HHS
- UL1TR001425 NCATS NIH HHS
- UL1TR001425 NCATS NIH HHS
- UL1TR001425 NCATS NIH HHS
- National Institute of General Medical Sciences
- National Center for Advancing Translational Sciences
Collapse
Affiliation(s)
- Hannah E Laue
- Department of Biostatistics and Epidemiology, University of Massachusetts Amherst School of Public Health and Health Sciences, 715 N. Pleasant Street, Arnold House 429, Amherst, MA, 01003, USA.
| | - Amy D Willis
- Department of Biostatistics, University of Washington Hans Rosling Center for Population Health, 3980 15 Avenue NE, Box 351617, Seattle, WA, 98195-1617, USA
| | - Fang Wang
- Department of Environmental Health Sciences, Columbia Mailman School of Public Health, 630 W 168th St, P&S 16-416, New York, NY, 10032, USA
| | - Melinda C MacDougall
- Division of General and Community Pediatrics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
| | - Yingying Xu
- Division of General and Community Pediatrics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
| | - Margaret R Karagas
- Department of Epidemiology, Dartmouth Geisel School of Medicine, One Medical Center Dr Lebanon, Lebanon, NH, 03756, USA
| | - Juliette C Madan
- Department of Epidemiology, Dartmouth Geisel School of Medicine, One Medical Center Dr Lebanon, Lebanon, NH, 03756, USA
- Department of Psychiatry, Dartmouth Hitchcock Medical Center, Lebanon, NH, USA
| | - Abby F Fleisch
- Center for Interdisciplinary and Population Health Research, Maine Institute for Research, Westbrook, ME, USA
- Pediatric Endocrinology and Diabetes, Maine Medical Center, 887 Congress St, Portland, ME, USA
| | - Bruce P Lanphear
- Faculty of Health Sciences, Simon Fraser University, Blusson Hall, 8888 University Dr, Burnaby, BC, Canada
| | - Kim M Cecil
- Division of General and Community Pediatrics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
- Department of Radiology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
- Department of Environmental and Public Health Sciences, University of Cincinnati, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
| | - Kimberly Yolton
- Division of General and Community Pediatrics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
- Department of Environmental and Public Health Sciences, University of Cincinnati, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
| | - Aimin Chen
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, 423 Guardian Drive, Philadelphia, PA, 19104, USA
| | - Jessie P Buckley
- Department of Epidemiology, University of North Carolina at Chapel Hill, 2106-B McGavran-Greenberg Hall CB#7435, Chapel Hill, NC, 27599, USA
| | - Joseph M Braun
- Department of Epidemiology, Brown University, 121 S Main St, Providence, RI, USA
| |
Collapse
|
4
|
Origüela V, Lopez-Zaplana A. Gut Microbiota: An Immersion in Dysbiosis, Associated Pathologies, and Probiotics. Microorganisms 2025; 13:1084. [PMID: 40431257 PMCID: PMC12113704 DOI: 10.3390/microorganisms13051084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 04/29/2025] [Accepted: 05/05/2025] [Indexed: 05/29/2025] Open
Abstract
The importance of the microbiome, particularly the gut microbiota and its implications for health, is well established. However, an increasing number of studies further strengthen the link between an imbalanced gut microbiota and a greater predisposition to different diseases. The gut microbiota constitutes a fundamental ecosystem for maintaining human health. Its alteration, known as dysbiosis, is associated with a wide range of conditions, including intestinal, metabolic, immunological, or neurological pathologies, among others. In recent years, there has been a substantial increase in knowledge about probiotics-bacterial species that enhance health or address various diseases-with numerous studies reporting their benefits in preventing or improving these conditions. This review aims to analyze the most common pathologies resulting from an imbalance in the gut microbiota, as well as detail the most important and known gut probiotics, their functions, and mechanisms of action in relation to these conditions.
Collapse
Affiliation(s)
- Valentina Origüela
- Department of Physiology, Faculty of Biology, University of Murcia, 30100 Murcia, Spain;
| | | |
Collapse
|
5
|
Zaiou M, Joubert O. Racial and Ethnic Disparities in NAFLD: Harnessing Epigenetic and Gut Microbiota Pathways for Targeted Therapeutic Approaches. Biomolecules 2025; 15:669. [PMID: 40427561 PMCID: PMC12109303 DOI: 10.3390/biom15050669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 04/28/2025] [Accepted: 04/29/2025] [Indexed: 05/29/2025] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a growing global health concern, impacting approximately 32.4% of the worldwide population. As a disease linked to metabolic dysfunction, NAFLD continues to rise alongside global increases in obesity, type 2 diabetes mellitus (T2DM), and metabolic syndrome. There is considerable evidence indicating that NAFLD disproportionately affects racial, ethnic, and minority groups, although the exact reasons for these disparities remain elusive. Contributing factors to this disease may include socioeconomic status, cultural influences, stress, genetic factors, and lifestyle choices. Emerging evidence suggests that these causal factors could influence epigenetic mechanisms, particularly DNA methylation and histone modifications, as well as the composition and diversity of gut microbiota. Nevertheless, there is a scarcity of research that comprehensively examines the interplay between epigenetic changes and gut microbiome variations in relation to NAFLD disparities across different racial and ethnic populations globally. This paper intends to (i) explore the connections between NAFLD, ethnic disparities, gut microbiota composition, and epigenetic alterations, while reviewing pertinent studies that illustrate how these factors contribute to health inequities among various ethnic groups impacted by this disease; (ii) explore potential therapeutic targets and biomarkers to advance the management of NAFLD; and (iii) provide insights to enhance our understanding of the mechanisms associated with this disease, thereby promoting further research in this field. Advancements in this area are anticipated to enhance our understanding of disease susceptibilities in at-risk groups and to provide new therapeutic options for NAFLD and its associated complications.
Collapse
Affiliation(s)
- Mohamed Zaiou
- Université de Lorraine, CNRS, IJL, F-54000 Nancy, France;
| | | |
Collapse
|
6
|
Cole LD, Kuhn KA. It Takes a Village: Juvenile Idiopathic Arthritis and the Microbiome. Rheum Dis Clin North Am 2025; 51:233-282. [PMID: 40246440 DOI: 10.1016/j.rdc.2025.01.005] [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: 04/19/2025]
Abstract
Multiple risk factors for juvenile idiopathic arthritis (JIA) influence the microbiome, and various differences in the oral and fecal microbiome have been described to date in JIA. This review summarizes what is known and discusses potential implications for future research on the microbiome in JIA.
Collapse
Affiliation(s)
- Lyndsey D Cole
- Sections of Rheumatology & Infectious Diseases, Department of Pediatrics, University of Colorado Anschutz Medical Campus, 13123 East 16th Avenue, B311, Aurora, CO 80045, USA.
| | - Kristine A Kuhn
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
7
|
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
|
8
|
Ghemrawi M, Ramírez Torres A, Netherland M, Wang Y, Hasan NA, El-Fahmawi B, Duncan G, McCord B. Forensic insights from shotgun metagenomics: Tracing microbial exchange during sexual intercourse. Forensic Sci Int Genet 2025; 78:103266. [PMID: 40117915 DOI: 10.1016/j.fsigen.2025.103266] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 03/05/2025] [Accepted: 03/05/2025] [Indexed: 03/23/2025]
Abstract
The microbiome is becoming an emerging field of interest within forensic science with high potential for individualization; however, little is known about bacterial species specific to the genital area or their ability to transfer between individuals during sexual contact. In this proof-of-concept study, we investigated microbial transfer dynamics in seven monogamous, heterosexual couples by collecting pre- and post-sexual intercourse samples from their genital areas, including penile, vaginal, and labial locations. Utilizing Shotgun Metagenomic Sequencing, we sequenced the microbial profiles of these samples. Our findings reveal significant transfer from the vaginal microbiome onto the penile microbiome, predominantly originating from the labial genitalia. Moreover, strain analysis unveiled distinct differentiation between the same species of bacteria across individuals, underscoring the potential for microbial forensics to distinguish individuals. This study contributes to our understanding of microbial transfer during sexual contact and highlights the forensic implications of the genital microbiome.
Collapse
Affiliation(s)
- Mirna Ghemrawi
- The Center for Forensic Science Research and Education, Fredric Rieders Family Foundation, PA 19044, USA; Department of Chemistry and Biochemistry, Florida International University, FL 33199, USA
| | - Andrea Ramírez Torres
- Department of Chemistry and Biochemistry, Florida International University, FL 33199, USA
| | | | - Ying Wang
- MAWI DNA Technologies, CA 94566, USA
| | | | | | | | - Bruce McCord
- Department of Chemistry and Biochemistry, Florida International University, FL 33199, USA.
| |
Collapse
|
9
|
Golob J, Rao K, Berinstein JA, Singh P, Chey WD, Owyang C, Kamada N, Higgins PDR, Young V, Bishu S, Lee AA. Why Symptoms Linger in Quiescent Crohn's Disease: Investigating the Impact of Sulfidogenic Microbes and Sulfur Metabolic Pathways. Inflamm Bowel Dis 2025; 31:763-776. [PMID: 39541261 PMCID: PMC11879174 DOI: 10.1093/ibd/izae238] [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: 07/12/2024] [Indexed: 11/16/2024]
Abstract
INTRODUCTION Even in the absence of inflammation, persistent symptoms in patients with Crohn's disease (CD) are prevalent and worsen quality of life. We previously demonstrated enrichment in sulfidogenic microbes in quiescent Crohn's disease patients with (qCD + S) vs without persistent GI symptoms (qCD-S). Thus, we hypothesized that sulfur metabolic pathways would be enriched in stool while differentially abundant microbes would be associated with important sulfur metabolic pathways in qCD + S. METHODS We performed a multicenter observational study nested within SPARC IBD. Quiescent inflammation was defined by fecal calprotectin level < 150 mcg/g. Persistent symptoms were defined by CD-PRO2. Active CD (aCD) and non-IBD diarrhea-predominant irritable bowel syndrome (IBS-D) were included as controls. RESULTS Thirty-nine patients with qCD + S, 274 qCD-S, 21 aCD, and 40 IBS-D underwent paired shotgun metagenomic sequencing and untargeted metabolomic profiling. The fecal metabolome in qCD + S was significantly different relative to qCD-S and IBS-D but not aCD. Patients with qCD + S were enriched in sulfur-containing amino acid pathways, including cysteine and methionine, as well as serine, glycine, and threonine. Glutathione and nicotinate/nicotinamide pathways were also enriched in qCD + S relative to qCD-S, suggestive of mitochondrial dysfunction, a downstream target of H2S signaling. Multi-omic integration demonstrated that enriched microbes in qCD + S were associated with important sulfur metabolic pathways. Bacterial sulfur metabolic genes, including CTH, isfD, sarD, and asrC, were dysregulated in qCD + S. Finally, sulfur metabolites with and without sulfidogenic microbes showed good accuracy in predicting the presence of qCD + S. DISCUSSION Microbial-derived sulfur pathways and downstream mitochondrial function are perturbed in qCD + S, which implicate H2S signaling in the pathogenesis of this condition. Future studies will determine whether targeting H2S pathways results in improved quality of life in qCD + S.
Collapse
Affiliation(s)
- Jonathan Golob
- Division of Infectious Diseases, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Krishna Rao
- Division of Infectious Diseases, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Jeffrey A Berinstein
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Prashant Singh
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - William D Chey
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Chung Owyang
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Nobuhiko Kamada
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Peter D R Higgins
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Vincent Young
- Division of Infectious Diseases, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA
| | - Shrinivas Bishu
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Allen A Lee
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
10
|
Mikles B, Schmidt CJ, Benbow ME, Jordan HR, Pechal JL. Potential postmortem microbial biomarkers of infant and younger children death investigation. J Forensic Sci 2025; 70:607-618. [PMID: 39682072 PMCID: PMC11874197 DOI: 10.1111/1556-4029.15677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 08/08/2024] [Accepted: 11/13/2024] [Indexed: 12/18/2024]
Abstract
Microbial communities associated with the human body are highly dynamic and reflect the host environment and lifestyle over time. Studies show death is no exception, with data demonstrating similar antemortem and postmortem microbiomes up to 48 h following death. These predictable microbial biomarkers can inform death investigation by helping to estimate the postmortem interval and build models to identify cause and manner of death. However, no attempts have been made to model potential microbial biomarkers in pediatric (≤2 years) deaths. This study provided a cross-sectional survey of the microbiota of 53 pediatric cases (black, white, both sexes) seen in Wayne County, Michigan. Autopsy cases represented accidents, homicides, or natural causes. Postmortem microbiome were collected by swabbing the eyes, ears, nose, mouth, umbilicus, brain, rectum, trabecular space, and cardiac blood. 16S rRNA sequence analyses indicated that sex, race, age, body site, and manner of death (MOD) had significant effects on microbiome composition, with significant interactions among MOD, race, and age. Amplicon sequence variants identified intra- and interhost dispersion of the postmortem microbiome depending on death circumstance. Among manners of death, non-accidental deaths were significantly distinct from all other deaths, and among body sites the rectum was distinct in its microbial composition. There is a real need for robust postmortem microbiome before it can be standardized as a practical tool for use in forensic investigation or public health. These results inform postmortem microbial variability during pediatric death investigation that contributes to a larger effort to understand the postmortem microbiome.
Collapse
Affiliation(s)
- Bethany Mikles
- Department of EntomologyMichigan State UniversityEast LansingMichiganUSA
| | - Carl J. Schmidt
- Department of PathologyUniversity of MichiganAnn ArborMichiganUSA
| | - M. Eric Benbow
- Department of EntomologyMichigan State UniversityEast LansingMichiganUSA
- Department of Osteopathic Medical SpecialtiesMichigan State UniversityEast LansingMichiganUSA
- AgBioResearchMichigan State UniversityEast LansingMichiganUSA
- Ecology, Evolution and Behavior ProgramMichigan State UniversityEast LansingMichiganUSA
| | - Heather R. Jordan
- Department of Biological SciencesMississippi State UniversityStarkvilleMississippiUSA
| | - Jennifer L. Pechal
- Department of EntomologyMichigan State UniversityEast LansingMichiganUSA
| |
Collapse
|
11
|
Magalhães MI, Azevedo MJ, Castro F, Oliveira MJ, Costa ÂM, Sampaio Maia B. The link between obesity and the gut microbiota and immune system in early-life. Crit Rev Microbiol 2025; 51:264-284. [PMID: 38651972 DOI: 10.1080/1040841x.2024.2342427] [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: 12/23/2023] [Revised: 04/04/2024] [Accepted: 04/06/2024] [Indexed: 04/25/2024]
Abstract
In early-life, the gut microbiota is highly modifiable, being modulated by external factors such as maternal microbiota, mode of delivery, and feeding strategies. The composition of the child's gut microbiota will deeply impact the development and maturation of its immune system, with consequences for future health. As one of the main sources of microorganisms to the child, the mother represents a crucial factor in the establishment of early-life microbiota, impacting the infant's wellbeing. Recent studies have proposed that dysbiotic maternal gut microbiota could be transmitted to the offspring, influencing the development of its immunity, and leading to the development of diseases such as obesity. This paper aims to review recent findings in gut microbiota and immune system interaction in early-life, highlighting the benefits of a balanced gut microbiota in the regulation of the immune system.
Collapse
Affiliation(s)
- Maria Inês Magalhães
- Doctoral Program in Biomedical Sciences, ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
- Nephrology and Infectious Diseases R&D group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Tumor and Microenvironment Interactions group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- nBTT, NanoBiomaterials for Targeted Therapies group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- FMDUP - Faculdade de Medicina Dentária da Universidade do Porto, Porto, Portugal
| | - Maria João Azevedo
- Nephrology and Infectious Diseases R&D group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- nBTT, NanoBiomaterials for Targeted Therapies group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- FMDUP - Faculdade de Medicina Dentária da Universidade do Porto, Porto, Portugal
- Academic Center for Dentistry Amsterdam (ACTA), Universiteit van Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Flávia Castro
- Tumor and Microenvironment Interactions group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Maria José Oliveira
- Tumor and Microenvironment Interactions group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Ângela M Costa
- Tumor and Microenvironment Interactions group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Benedita Sampaio Maia
- Nephrology and Infectious Diseases R&D group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- nBTT, NanoBiomaterials for Targeted Therapies group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- FMDUP - Faculdade de Medicina Dentária da Universidade do Porto, Porto, Portugal
| |
Collapse
|
12
|
Chacon J, Faizuddin F, McKee JC, Sheikh A, Vasquez VM, Gadad SS, Mayer G, Siby S, McCabe M, Dhandayuthapani S. Unlocking the Microbial Symphony: The Interplay of Human Microbiota in Cancer Immunotherapy Response. Cancers (Basel) 2025; 17:813. [PMID: 40075661 PMCID: PMC11899421 DOI: 10.3390/cancers17050813] [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: 01/11/2025] [Revised: 02/13/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
INTRODUCTION The emergence of cancer immunotherapy has revolutionized cancer treatment, offering remarkable outcomes for patients across various malignancies. However, the heterogeneous response to immunotherapy underscores the necessity of understanding additional factors influencing treatment efficacy. Among these factors, the human microbiota has garnered significant attention for its potential role in modulating immune response. Body: This review explores the intricate relationship between the human microbiota and cancer immunotherapy, highlighting recent advances and potential mechanisms underlying microbial influence on treatment outcomes. CONCLUSION Insights into the microbiome's impact on immunotherapy response not only deepen our understanding of cancer pathogenesis but also hold promise for personalized therapeutic strategies aimed at optimizing patient outcomes.
Collapse
Affiliation(s)
- Jessica Chacon
- Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (F.F.); (J.C.M.); (A.S.); (S.S.G.); (G.M.); (S.S.); (M.M.); (S.D.)
| | - Farah Faizuddin
- Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (F.F.); (J.C.M.); (A.S.); (S.S.G.); (G.M.); (S.S.); (M.M.); (S.D.)
| | - Jack C. McKee
- Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (F.F.); (J.C.M.); (A.S.); (S.S.G.); (G.M.); (S.S.); (M.M.); (S.D.)
| | - Aadil Sheikh
- Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (F.F.); (J.C.M.); (A.S.); (S.S.G.); (G.M.); (S.S.); (M.M.); (S.D.)
| | - Victor M. Vasquez
- Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (F.F.); (J.C.M.); (A.S.); (S.S.G.); (G.M.); (S.S.); (M.M.); (S.D.)
| | - Shrikanth S. Gadad
- Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (F.F.); (J.C.M.); (A.S.); (S.S.G.); (G.M.); (S.S.); (M.M.); (S.D.)
- L. Frederick Francis Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
- Center of Emphasis in Cancer, Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
| | - Ghislaine Mayer
- Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (F.F.); (J.C.M.); (A.S.); (S.S.G.); (G.M.); (S.S.); (M.M.); (S.D.)
| | - Sharon Siby
- Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (F.F.); (J.C.M.); (A.S.); (S.S.G.); (G.M.); (S.S.); (M.M.); (S.D.)
| | - Molly McCabe
- Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (F.F.); (J.C.M.); (A.S.); (S.S.G.); (G.M.); (S.S.); (M.M.); (S.D.)
| | - Subramanian Dhandayuthapani
- Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (F.F.); (J.C.M.); (A.S.); (S.S.G.); (G.M.); (S.S.); (M.M.); (S.D.)
- L. Frederick Francis Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
- Center of Emphasis in Infectious Diseases, Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
| |
Collapse
|
13
|
Abdill RJ, Graham SP, Rubinetti V, Ahmadian M, Hicks P, Chetty A, McDonald D, Ferretti P, Gibbons E, Rossi M, Krishnan A, Albert FW, Greene CS, Davis S, Blekhman R. Integration of 168,000 samples reveals global patterns of the human gut microbiome. Cell 2025; 188:1100-1118.e17. [PMID: 39848248 PMCID: PMC11848717 DOI: 10.1016/j.cell.2024.12.017] [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: 10/02/2023] [Revised: 09/09/2024] [Accepted: 12/13/2024] [Indexed: 01/25/2025]
Abstract
The factors shaping human microbiome variation are a major focus of biomedical research. While other fields have used large sequencing compendia to extract insights requiring otherwise impractical sample sizes, the microbiome field has lacked a comparably sized resource for the 16S rRNA gene amplicon sequencing commonly used to quantify microbiome composition. To address this gap, we processed 168,464 publicly available human gut microbiome samples with a uniform pipeline. We use this compendium to evaluate geographic and technical effects on microbiome variation. We find that regions such as Central and Southern Asia differ significantly from the more thoroughly characterized microbiomes of Europe and Northern America and that composition alone can be used to predict a sample's region of origin. We also find strong associations between microbiome variation and technical factors such as primers and DNA extraction. We anticipate this growing work, the Human Microbiome Compendium, will enable advanced applied and methodological research.
Collapse
Affiliation(s)
- Richard J Abdill
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Samantha P Graham
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN, USA
| | - Vincent Rubinetti
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO, USA; Center for Health Artificial Intelligence (CHAI), University of Colorado School of Medicine, Aurora, CO, USA
| | - Mansooreh Ahmadian
- Department of Biostatistics and Informatics, University of Colorado Anschutz Medical Campus, School of Public Health, Aurora, CO, USA
| | - Parker Hicks
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Ashwin Chetty
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Daniel McDonald
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Pamela Ferretti
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Elizabeth Gibbons
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Marco Rossi
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Arjun Krishnan
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO, USA; Department of Biostatistics and Informatics, University of Colorado Anschutz Medical Campus, School of Public Health, Aurora, CO, USA
| | - Frank W Albert
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN, USA
| | - Casey S Greene
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO, USA; Center for Health Artificial Intelligence (CHAI), University of Colorado School of Medicine, Aurora, CO, USA
| | - Sean Davis
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO, USA; Center for Health Artificial Intelligence (CHAI), University of Colorado School of Medicine, Aurora, CO, USA
| | - Ran Blekhman
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
14
|
Guers JJ, Heffernan KS, Campbell SC. Getting to the Heart of the Matter: Exploring the Intersection of Cardiovascular Disease, Sex and Race and How Exercise, and Gut Microbiota Influence these Relationships. Rev Cardiovasc Med 2025; 26:26430. [PMID: 40026503 PMCID: PMC11868917 DOI: 10.31083/rcm26430] [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: 09/04/2024] [Revised: 11/16/2024] [Accepted: 11/28/2024] [Indexed: 03/05/2025] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death worldwide, with physical inactivity being a known contributor to the global rates of CVD incidence. CVD incidence, however, is not uniform with recognized sex differences as well and racial and ethnic differences. Furthermore, gut microbiota have been associated with CVD, sex, and race/ethnicity. Researchers have begun to examine the interplay of these complicated yet interrelated topics. This review will present evidence that CVD (risk and development), and gut microbiota are distinct between the sexes and racial/ethnic groups, which appear to be influenced by acculturation, discrimination, stress, and lifestyle factors like exercise. Furthermore, this review will address the beneficial impacts of exercise on the cardiovascular system and will provide recommendations for future research in the field.
Collapse
Affiliation(s)
- John J. Guers
- Department of Health Sciences and Nursing, Rider University, Lawrenceville, NJ 08648, USA
| | - Kevin S. Heffernan
- Department of Biobehavioral Sciences, Teachers College, Columbia University, New York, NY 10027, USA
| | - Sara C. Campbell
- Department of Kinesiology and Health, The State University of New Jersey, New Brunswick, NJ 08901, USA
- Centers for Human Nutrition, Exercise, and Metabolism, Nutrition, Microbiome, and Health, and Lipid Research, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| |
Collapse
|
15
|
Jones JA, Newton IG, Moczek AP. Microbiome composition and turnover in the face of complex lifecycles and bottlenecks: insights through the study of dung beetles. Appl Environ Microbiol 2025; 91:e0127824. [PMID: 39704535 PMCID: PMC11784073 DOI: 10.1128/aem.01278-24] [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: 06/28/2024] [Accepted: 11/04/2024] [Indexed: 12/21/2024] Open
Abstract
Microbiome composition and function often change throughout a host's life cycle, reflecting shifts in the ecological niche of the host. The mechanisms that establish these relationships are therefore important dimensions of host ecology and evolution; yet, their nature remains poorly understood. Here, we sought to investigate the microbial communities associated with the complex life cycle of the dung beetle Onthophagus taurus and the relative contributions of host life stage, sex, and environment in determining microbiome assembly. We find that O. taurus plays host to a diverse microbiota that undergo drastic community shifts throughout host development, influenced by host life stage, environmental microbiota, and, to a lesser degree, sex. Contrary to predictions, we found that egg and pupal stages-despite the absence of a digestive tract or defined microbe-storing organs-do not constrain microbial maintenance, while host-constructed environments, such as a maternally derived fecal pellet or the pupal chamber constructed by late larvae, may still serve as complementary microbial refugia for select taxa. Lastly, we identify a small community of putative core microbiota likely to shape host development and fitness. Our results provide important insights into mechanisms employed by solitary organisms to assemble, maintain, and adjust beneficial microbiota to confront life-stage-specific needs and challenges. IMPORTANCE As the influence of symbionts on host ecology, evolution, and development has become more apparent so has the importance of understanding how hosts facilitate the reliable maintenance of their interactions with these symbionts. A growing body of work has thus begun to identify diverse behaviors and physiological mechanisms underpinning the selective colonization of beneficial symbionts across a range of host taxa. Yet, how organisms with complex life cycles, such as holometabolous insects, establish and maintain key symbionts remains poorly understood. This is particularly interesting considering the drastic transformations of both internal and external host morphology, and the ecological niche shifts in diet and environment, that are the hallmark of metamorphosis. This work investigates the dynamic changes of the microbiota associated with the complex life cycle and host-constructed environments of the bull-headed dung beetle, Onthophagus taurus, a useful model for understanding how organisms may maintain and modulate their microbiota across development.
Collapse
Affiliation(s)
- Joshua A. Jones
- Department of Biology, Indiana University Bloomington, Bloomington, Indiana, USA
| | - Irene Garcia Newton
- Department of Biology, Indiana University Bloomington, Bloomington, Indiana, USA
| | - Armin P. Moczek
- Department of Biology, Indiana University Bloomington, Bloomington, Indiana, USA
| |
Collapse
|
16
|
Govender P, Ghai M. Population-specific differences in the human microbiome: Factors defining the diversity. Gene 2025; 933:148923. [PMID: 39244168 DOI: 10.1016/j.gene.2024.148923] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 08/15/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
Differences in microbial communities at different body habitats define the microbiome composition of the human body. The gut, oral, skin vaginal fluid and tissue microbiome, are pivotal for human development and immune response and cross talk between these microbiomes is evident. Population studies reveal that various factors, such as host genetics, diet, lifestyle, aging, and geographical location are strongly associated with population-specific microbiome differences. The present review discusses the factors that shape microbiome diversity in humans, and microbiome differences in African, Asian and Caucasian populations. Gut microbiome studies show that microbial species Bacteroides is commonly found in individuals living in Western countries (Caucasian populations), while Prevotella is prevalent in non-Western countries (African and Asian populations). This association is mainly due to the high carbohydrate, high fat diet in western countries in contrast to high fibre, low fat diets in African/ Asian regions. Majority of the microbiome studies focus on the bacteriome component; however, interesting findings reveal that increased bacteriophage richness, which makes up the virome component, correlates with decreased bacterial diversity, and causes microbiome dysbiosis. An increase of Caudovirales (bacteriophages) is associated with a decrease in enteric bacteria in inflammatory bowel diseases. Future microbiome studies should evaluate the interrelation between bacteriome and virome to fully understand their significance in the pathogenesis and progression of human diseases. With ethnic health disparities becoming increasingly apparent, studies need to emphasize on the association of population-specific microbiome differences and human diseases, to develop microbiome-based therapeutics. Additionally, targeted phage therapy is emerging as an attractive alternative to antibiotics for bacterial infections. With rapid rise in microbiome research, focus should be on standardizing protocols, advanced bioinformatics tools, and reducing sequencing platform related biases. Ultimately, integration of multi-omics data (genomics, transcriptomics, proteomics and metabolomics) will lead to precision models for personalized microbiome therapeutics advancement.
Collapse
Affiliation(s)
- Priyanka Govender
- Discipline of Genetics, School of Life Sciences, University of KwaZulu-Natal, Westville, South Africa
| | - Meenu Ghai
- Discipline of Genetics, School of Life Sciences, University of KwaZulu-Natal, Westville, South Africa.
| |
Collapse
|
17
|
Kreouzi M, Theodorakis N, Nikolaou M, Feretzakis G, Anastasiou A, Kalodanis K, Sakagianni A. Skin Microbiota: Mediator of Interactions Between Metabolic Disorders and Cutaneous Health and Disease. Microorganisms 2025; 13:161. [PMID: 39858932 PMCID: PMC11767725 DOI: 10.3390/microorganisms13010161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 01/05/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Metabolic disorders, including type 2 diabetes mellitus (T2DM), obesity, and metabolic syndrome, are systemic conditions that profoundly impact the skin microbiota, a dynamic community of bacteria, fungi, viruses, and mites essential for cutaneous health. Dysbiosis caused by metabolic dysfunction contributes to skin barrier disruption, immune dysregulation, and increased susceptibility to inflammatory skin diseases, including psoriasis, atopic dermatitis, and acne. For instance, hyperglycemia in T2DM leads to the formation of advanced glycation end products (AGEs), which bind to the receptor for AGEs (RAGE) on keratinocytes and immune cells, promoting oxidative stress and inflammation while facilitating Staphylococcus aureus colonization in atopic dermatitis. Similarly, obesity-induced dysregulation of sebaceous lipid composition increases saturated fatty acids, favoring pathogenic strains of Cutibacterium acnes, which produce inflammatory metabolites that exacerbate acne. Advances in metabolomics and microbiome sequencing have unveiled critical biomarkers, such as short-chain fatty acids and microbial signatures, predictive of therapeutic outcomes. For example, elevated butyrate levels in psoriasis have been associated with reduced Th17-mediated inflammation, while the presence of specific Lactobacillus strains has shown potential to modulate immune tolerance in atopic dermatitis. Furthermore, machine learning models are increasingly used to integrate multi-omics data, enabling personalized interventions. Emerging therapies, such as probiotics and postbiotics, aim to restore microbial diversity, while phage therapy selectively targets pathogenic bacteria like Staphylococcus aureus without disrupting beneficial flora. Clinical trials have demonstrated significant reductions in inflammatory lesions and improved quality-of-life metrics in patients receiving these microbiota-targeted treatments. This review synthesizes current evidence on the bidirectional interplay between metabolic disorders and skin microbiota, highlighting therapeutic implications and future directions. By addressing systemic metabolic dysfunction and microbiota-mediated pathways, precision strategies are paving the way for improved patient outcomes in dermatologic care.
Collapse
Affiliation(s)
- Magdalini Kreouzi
- Department of Internal Medicine, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Athens, Greece;
| | - Nikolaos Theodorakis
- NT-CardioMetabolics, Clinic for Metabolism and Athletic Performance, 47 Tirteou Str., 17564 Palaio Faliro, Greece;
- Department of Cardiology & Preventive Cardiology Outpatient Clinic, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece;
- School of Medicine, National and Kapodistrian University of Athens, 75 Mikras Asias, 11527 Athens, Greece
- School of Science and Technology, Hellenic Open University, 18 Aristotelous Str., 26335 Patras, Greece
| | - Maria Nikolaou
- Department of Cardiology & Preventive Cardiology Outpatient Clinic, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece;
| | - Georgios Feretzakis
- School of Science and Technology, Hellenic Open University, 18 Aristotelous Str., 26335 Patras, Greece
| | - Athanasios Anastasiou
- Biomedical Engineering Laboratory, National Technical University of Athens, 15780 Athens, Greece;
| | - Konstantinos Kalodanis
- Department of Informatics & Telematics, Harokopio University of Athens, 17676 Kallithea, Greece;
| | - Aikaterini Sakagianni
- Intensive Care Unit, Sismanogleio General Hospital, 37 Sismanogleiou Str., 15126 Marousi, Greece
| |
Collapse
|
18
|
Westerbeke FHM, Rios-Morales M, Attaye I, Nieuwdorp M. Fructose catabolism and its metabolic effects: Exploring host-microbiota interactions and the impact of ethnicity. J Physiol 2025. [PMID: 39805044 DOI: 10.1113/jp287316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 12/16/2024] [Indexed: 01/16/2025] Open
Abstract
Important health disparities are observed in the prevalence of obesity and associated non-communicable diseases (NCDs), including type 2 diabetes (T2D) and metabolic dysfunction-associated steatotic liver disease (MASLD) among ethnic groups. Yet, the underlying factors accounting for these disparities remain poorly understood. Fructose has been widely proposed as a potential mediator of these NCDs, given that hepatic fructose catabolism can result in deleterious metabolic effects, including insulin resistance and hepatic steatosis. Moreover, the fermentation of fructose by the gut microbiota can produce metabolites such as ethanol and acetate, both which serve as potential substrates for de novo lipogenesis (DNL) and could therefore contribute to the development of these metabolic conditions. Significant inter-ethnic differences in gut microbiota composition have been observed. Moreover, fructose consumption varies across ethnic groups, and fructose intake has been demonstrated to significantly alter gut microbiota composition, which can influence its fermenting properties and metabolic effects. Therefore, ethnic differences in gut microbiota composition, which may be influenced by variations in fructose consumption, could contribute to the observed health disparities. This review provides an overview of the complex interactions between host and microbial fructose catabolism, the role of ethnicity in shaping these metabolic processes and their impact on host health. Understanding these interactions could provide insights into the mechanisms driving ethnic health disparities to improve personalized nutrition strategies. KEY POINTS: Dietary fructose consumption has increased substantially over recent decades, which has been associated with the rising prevalence of obesity and non-communicable diseases (NCDs) such as type 2 diabetes and metabolic dysfunction-associated steatotic liver disease. Pronounced disparities among different ethnic groups in NCD prevalence and dietary fructose consumption underscore the need to elucidate the underlying mechanisms of fructose catabolism and its health effects. Together with the well-known toxic effects of hepatic fructose catabolism, emerging evidence highlights a role for the small intestinal microbiota in fermenting sugars like fructose into various bacterial products with potential deleterious metabolic effects. There are significant ethnic differences in gut microbiota composition that, combined with varying fructose consumption, could mediate the observed health disparities. To comprehensively understand the role of the gut microbiota in mediating fructose-induced adverse metabolic effects, future research should focus on the small intestinal microbiota. Future research on fructose - microbiota - host interactions should account for ethnic differences in dietary habits and microbial composition to elucidate the potential role of the gut microbiota in driving the mentioned health disparities.
Collapse
Affiliation(s)
- Florine H M Westerbeke
- Department of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Melany Rios-Morales
- Department of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Ilias Attaye
- Department of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Max Nieuwdorp
- Department of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| |
Collapse
|
19
|
Janssens-Böcker C, Doberenz C, Monteiro M, de Oliveira Ferreira M. Influence of Cosmetic Skincare Products with pH < 5 on the Skin Microbiome: A Randomized Clinical Evaluation. Dermatol Ther (Heidelb) 2025; 15:141-159. [PMID: 39709312 PMCID: PMC11785879 DOI: 10.1007/s13555-024-01321-x] [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: 09/10/2024] [Accepted: 11/27/2024] [Indexed: 12/23/2024] Open
Abstract
INTRODUCTION The human skin acts as a protective barrier against external pathogens and hosts a diverse microbiome consisting of bacteria, fungi, viruses, and archaea. Disruptions to the skin microbiome can impact immune function, leading to inflammatory and autoimmune conditions. The importance of pH for the microbiome is paramount. Cosmetic skincare products interact with the skin microbiome and skin pH, playing a key role in maintaining microbial balance. Research suggests that products with non-physiological pH levels may disrupt the skin microbiota. Our clinical study aimed to evaluate the effects of low-pH cosmetic products (pH < 5) on the skin microbiome, contributing to improved skin health. METHODS The clinical study focused on evaluating the skin microbiome diversity following the application for 28 days of four different low-pH cosmetic products (vitamin C, resveratrol, a collagen mask, and a native algae mask) on the forearms of post-menopausal women with skin pH > 5.5. RESULTS The diversity of the natural skin microbiome increased consistently throughout the study, evident in both the untreated area and after the application of the Vitamin C Concentrate, Resveratrol Concentrate, Collagen Mask, and Native Algae Mask, as indicated by Shannon's diversity index. The native algae mask notably reduced the Corynebacterium genus and significantly lowered the pH. The skin pH changes corresponded with microbiota stability. CONCLUSIONS In conclusion, enhanced diversity of the natural skin microbiome was observed over the study duration. None of the investigational products caused significant disruption to the skin microbiome diversity, as evidenced by the stable Shannon's diversity index and relative abundance of specific genera. Notably, the native algae mask significantly decreased the presence of the opportunistic pathogenic Corynebacterium genus, which is likely attributable to a minor reduction in skin pH following extended product use. The findings suggest that the use of low-pH skincare products, like the native algae mask, do not disrupt skin microbiome diversity and may have the potential to positively impact skin microbiome diversity and health by reducing certain pathogenic microbial populations.
Collapse
Affiliation(s)
| | | | - Marta Monteiro
- Inovapotek, Pharmaceutical Research & Development, Porto, Portugal
| | | |
Collapse
|
20
|
Liu X, Shi L, Zhang S, Zhou A. Exploring potential plasma drug targets for cholelithiasis through multiancestry Mendelian randomization. Int J Surg 2025; 111:302-310. [PMID: 38976909 PMCID: PMC11745627 DOI: 10.1097/js9.0000000000001925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 06/25/2024] [Indexed: 07/10/2024]
Abstract
BACKGROUND Cholelithiasis poses significant health and economic burdens, necessitating novel pharmacological targets to enhance treatment efficacy. METHOD Based on genome-wide association analysis studies, the authors performed a two-sample Mendelian randomization (MR) analysis based on plasma proteomics to explore potential drug targets in European (n Case =40 191 and n Control =361 641) and Asian (n Case =9305 and n Control =168 253) populations. The authors confirmed the directionality and robust correlation of the drug targets with the results through reverse MR analysis, Steiger filtering, Bayesian colocalization, phenotype scanning, and replication in multiple databases. Further exploration of the safety and possible mechanisms of action of phenome-wide MR analysis and protein-protein interactions (PPIs) as individual drug targets was performed. RESULTS Our proteomics-based MR analyses suggested that FUT3 (OR=0.87; 95% CI: 0.84-0.89; P =4.70×10 -32 ), NOE1 (OR=0.58; 95% CI: 0.52-0.66; P =4.21×10 -23 ), UGT1A6 (OR=0.68; 95% CI: 0.64-0.73; P =9.58×10 -30 ), and FKBP52 (OR=1.75; 95% CI: 1.37-2.24; P =8.61×10 -6 ) were potential drug targets in Europeans, whereas KLB (OR=1.11; 95% CI: 1.07-1.16; P =7.59×10 -7 ) and FGFR4 (OR=0.94; 95% CI: 0.91-0.96; P =4.07×10 -6 ) were valid targets in East Asians. There was no reverse causality for these drug targets. Evidence from Bayesian colocalization analyses supported that exposure and outcome shared consistent genetic variables. Phenome-wide MR analysis suggested the potential deleterious effects of NOE1 and FGFR4. PPI analysis confirmed the pathways associated with the potential targets involved in bile acid metabolism. CONCLUSIONS Genetically predicted levels of the plasma proteins FUT3, NOE1, UGT1A6, and FKBP52 have the potential as prospective targets in Europeans. Moreover, the plasma levels of KLB and FGFR4 may serve as potential targets for the treatment of cholelithiasis in East Asians.
Collapse
Affiliation(s)
- Xiaoduo Liu
- Department of Neurology and Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, National Center for Neurological Disorders
| | - Lubo Shi
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing Digestive Disease Center, National Clinical Research Center for Digestive Diseases, Beijing, People’s Republic of China
| | - Shutian Zhang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing Digestive Disease Center, National Clinical Research Center for Digestive Diseases, Beijing, People’s Republic of China
| | - Anni Zhou
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing Digestive Disease Center, National Clinical Research Center for Digestive Diseases, Beijing, People’s Republic of China
| |
Collapse
|
21
|
Hernandez J, Rodriguez JB, Trak-Fellermeier MA, Galvan R, Macchi A, Martinez-Motta P, Palacios C. Suboptimal vitamin D status and overweight/obesity are associated with gut integrity and inflammation in minority children and adolescents: A cross-sectional analysis from the MetA-bone trial. Nutr Res 2025; 133:13-21. [PMID: 39662375 DOI: 10.1016/j.nutres.2024.11.006] [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: 07/10/2024] [Revised: 11/18/2024] [Accepted: 11/18/2024] [Indexed: 12/13/2024]
Abstract
Preserving gut integrity is essential to preventing the development of chronic diseases. Several factors are associated with gut integrity and inflammation in adults. However, there is limited evidence in healthy children. This study evaluated the factors associated with gut integrity and inflammation in healthy children participating in the MetA-Bone trial. We hypothesized that age, sex, race/ethnicity, diet, vitamin D, and body composition will be associated with gut integrity and inflammation. Socio-demographic variables were collected with a questionnaire. Measures included markers of gut integrity (Intestinal Fatty Acid Binding Protein; I-FABP), and inflammation (IL-17 and calprotectin) determined by ELISA in 24-h urine and serum; serum 25(OH)D concentration (commercial lab), BMI percentile, and diet (24-h recalls). Analyses included descriptive statistics, chi-square, and adjusted logistic regressions. Participants (n=138) median age was 12.4 (11.1-13.3), 53.6% were male, 9.4% were Black/African American, and 71.1% were Hispanic/Latino. Children with suboptimal vitamin D were 3.35 times more likely to present gut integrity damage (elevated I-FABP) than those with optimal status (P = .005). Overweight/obesity and fructose intake were associated with inflammation (elevated calprotectin) (P < .05). Those with lower gut integrity damage had lower odds of having higher inflammation (P = .021). Other factors were not associated with inflammation. Suboptimal vitamin D status, overweight/obesity and inflammation may compromise the gut integrity in healthy children, suggesting an impairment on the intestinal barrier repair system. More research with a longitudinal design is needed to gain a deeper understanding of the role of additional factors linked to gut integrity and inflammation in healthy children.
Collapse
Affiliation(s)
- Jacqueline Hernandez
- Department of Dietetics and Nutrition, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, USA.
| | - Jose Bastida Rodriguez
- Department of Dietetics and Nutrition, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, USA
| | - Maria Angelica Trak-Fellermeier
- Department of Dietetics and Nutrition, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, USA
| | - Rodolfo Galvan
- Department of Dietetics and Nutrition, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, USA
| | - Alison Macchi
- Department of Dietetics and Nutrition, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, USA
| | - Preciosa Martinez-Motta
- Department of Dietetics and Nutrition, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, USA
| | - Cristina Palacios
- Department of Dietetics and Nutrition, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, USA
| |
Collapse
|
22
|
Wang Q, Wang BY, Williams S, Xie H. Diversity and Characteristics of the Oral Microbiome Associated with Self-Reported Ancestral/Ethnic Groups. Int J Mol Sci 2024; 25:13303. [PMID: 39769067 PMCID: PMC11677810 DOI: 10.3390/ijms252413303] [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: 10/14/2024] [Revised: 12/06/2024] [Accepted: 12/07/2024] [Indexed: 01/11/2025] Open
Abstract
Periodontitis disproportionately affects genetic ancestral/ethnic groups. To characterize the oral microbiome from different genetic ancestral/ethnic groups, we collected 161 dental plaque samples from self-identified African Americans (AAs), Caucasian Americans (CAs), and Hispanic Americans (HAs) with clinical gingival health or biofilm-induced gingivitis on an intact periodontium. DNA was extracted from these samples, and then DNA libraries were prepared and sequenced using an Illumina NovaSeq high-throughput sequencer. We found significant differences in the diversity and abundance of microbial taxa among dental plaque samples of the AA, CA, and HA groups. We also identified unique microbial species in a self-reported ancestral/ethnic group. Moreover, we revealed variations in functional potentials of the oral microbiome among the three ancestral/ethnic groups, with greater diversity and abundance of antibiotic-resistant genes in the oral microbiome and significantly more genes involved in the modification of glycoconjugates and oligo- and polysaccharides in AAs than in CAs and HAs. Our observations suggest that the variations in the oral microbiome associated with ancestral/ethnic backgrounds may directly relate to their virulence potential including their abilities to induce host immune responses and to resist antibiotic treatment. These finding can be a steppingstone for developing precision medicine and personalized periodontal prevention/treatment and for reducing oral health disparities.
Collapse
Affiliation(s)
- Qingguo Wang
- School of Medicine, Meharry Medical College, Nashville, TN 37208, USA
| | - Bing-Yan Wang
- School of Dentistry, University of Texas Health Science Center at Houston, Houston, TX 77504, USA;
| | - She’Neka Williams
- School of Dentistry, Meharry Medical College, Nashville, TN 37208, USA;
| | - Hua Xie
- School of Dentistry, Meharry Medical College, Nashville, TN 37208, USA;
| |
Collapse
|
23
|
Mehlferber EC, Arnault G, Joshi B, Partida-Martinez LP, Patras KA, Simonin M, Koskella B. A cross-systems primer for synthetic microbial communities. Nat Microbiol 2024; 9:2765-2773. [PMID: 39478083 PMCID: PMC11660114 DOI: 10.1038/s41564-024-01827-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 09/11/2024] [Indexed: 11/02/2024]
Abstract
The design and use of synthetic communities, or SynComs, is one of the most promising strategies for disentangling the complex interactions within microbial communities, and between these communities and their hosts. Compared to natural communities, these simplified consortia provide the opportunity to study ecological interactions at tractable scales, as well as facilitating reproducibility and fostering interdisciplinary science. However, the effective implementation of the SynCom approach requires several important considerations regarding the development and application of these model systems. There are also emerging ethical considerations when both designing and deploying SynComs in clinical, agricultural or environmental settings. Here we outline current best practices in developing, implementing and evaluating SynComs across different systems, including a focus on important ethical considerations for SynCom research.
Collapse
Affiliation(s)
- Elijah C Mehlferber
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA.
- Center for Microbial Dynamics and Infection, Georgia Institute of Technology, Atlanta, GA, USA.
| | - Gontran Arnault
- Univ Angers, Institut Agro, INRAE, IRHS, SFR QUASAV, Angers, France
| | - Bishnu Joshi
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Laila P Partida-Martinez
- Departamento de Ingeniería Genética, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav), Irapuato, México
| | - Kathryn A Patras
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
- Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, TX, USA
| | - Marie Simonin
- Univ Angers, Institut Agro, INRAE, IRHS, SFR QUASAV, Angers, France
| | - Britt Koskella
- Department of Integrative Biology, University of California, Berkeley, CA, USA.
- San Francisco Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
24
|
Wang Q, Wang BY, Williams S, Xie H. Diversity and characteristics of the oral microbiome influenced by race and ethnicity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.07.617037. [PMID: 39416080 PMCID: PMC11482769 DOI: 10.1101/2024.10.07.617037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Periodontitis disproportionately affects racial/ethnic populations. Besides social determinants contributing to disparities in periodontal health, variations of oral microbial communities may also be a key factor influencing oral immune responses. To characterize the oral microbiome from different racial/ethnic populations, we collected 161 dental plaque samples from African Americans (AAs), Caucasian Americans (CAs), and Hispanic Americans (HAs) with clinical gingival health or biofilm-induced gingivitis on an intact periodontium. Using metagenomic sequencing, we found significant difference in diversity and abundance of microbial taxa in the dental plaque samples from AA, CA, and HA groups and unique microbial species that can only be detected in a particular racial/ethnic group. Moreover, we revealed racial/ethnic associated variations in functional potential of the oral microbiome, showing that diversity and abundance of antibiotic resistant genes were greater in the oral microbiome of the AAs than those in CAs or HAs, and that the AAs exhibited higher levels of genes involving in modification of glycoconjugates, oligo- and polysaccharides. These findings indicate more complex and higher virulence potential oral microbiome in AA and HA populations, which likely contributes to higher prevalence of periodontitis in AAs and HAs.
Collapse
Affiliation(s)
- Qingguo Wang
- School of Medicine, Meharry Medical College, Nashville, TN, USA
| | - Bing-Yan Wang
- School of Dentistry, University of Texas Health Science Center at Houston, Houston, TX, USA
| | | | - Hua Xie
- School of Dentistry, Meharry Medical College, Nashville, TN, USA
| |
Collapse
|
25
|
Golob J, Rao K, Berinstein JA, Singh P, Chey WD, Owyang C, Kamada N, Higgins PDR, Young V, Bishu S, Lee AA. Why Symptoms Linger in Quiescent Crohn's Disease: Investigating the Impact of Sulfidogenic Microbes and Sulfur Metabolic Pathways. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.08.24313266. [PMID: 39314983 PMCID: PMC11419226 DOI: 10.1101/2024.09.08.24313266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Introduction Even in the absence of inflammation, persistent symptoms in patients with Crohn's disease (CD) are prevalent and worsen quality of life. We previously demonstrated enrichment in sulfidogenic microbes in quiescent Crohn's disease patients with ( qCD+S ) vs. without persistent GI symptoms ( qCD-S ). Thus, we hypothesized that sulfur metabolic pathways would be enriched in stool while differentially abundant microbes would be associated with important sulfur-metabolic pathways in qCD+S. Methods We performed a multi-center observational study nested within SPARC IBD. Quiescent inflammation was defined by fecal calprotectin level <150 mcg/g. Persistent symptoms were defined by CD-PRO2. Active CD ( aCD ) and non-IBD diarrhea-predominant irritable bowel syndrome ( IBS-D ) were included as controls. Results Thirty-nine patients with qCD+S, 274 qCD-S, 21 aCD, and 40 IBS-D underwent paired shotgun metagenomic sequencing and untargeted metabolomic profiling. The fecal metabolome in qCD+S was significantly different relative to qCD-S and IBS-D but not aCD. Patients with qCD+S were enriched in sulfur-containing amino acid pathways, including cysteine and methionine, as well as serine, glycine, and threonine. Glutathione and nicotinate/nicotinamide pathways were also enriched in qCD+S relative to qCD-S, suggestive of mitochondrial dysfunction, a downstream target of H 2 S signaling. Multi-omic integration demonstrated that enriched microbes in qCD+S were associated with important sulfur-metabolic pathways. Bacterial sulfur-metabolic genes, including CTH , isfD , sarD , and asrC , were dysregulated in qCD+S. Finally, sulfur metabolites with and without sulfidogenic microbes showed good accuracy in predicting presence of qCD+S. Discussion Microbial-derived sulfur pathways and downstream mitochondrial function are perturbed in qCD+S, which implicate H 2 S signaling in the pathogenesis of this condition. Future studies will determine whether targeting H 2 S pathways results in improved quality of life in qCD+S. Key Messages What is Already Known Even in the absence of inflammation, persistent gastrointestinal symptoms are common in Crohn's disease.The microbiome is altered in quiescent Crohn's disease patients with persistent symptoms, but the functional significance of these changes is unknown. What is New Here Sulfur metabolites and sulfur metabolic pathways were enriched in stool in quiescent Crohn's disease patients with persistent symptoms.Multi-omic integration showed enriched microbes were associated with important sulfur metabolic pathways in quiescent Crohn's disease patients with persistent symptoms. How Can This Study Help Patient Care Strategies to decrease sulfidogenic microbes and associated sulfur metabolic pathways could represent a novel strategy to improve quality of life in quiescent Crohn's disease with persistent GI symptoms.
Collapse
|
26
|
Dickstein DR, Edwards CR, Rowan CR, Avanessian B, Chubak BM, Wheldon CW, Simoes PK, Buckstein MH, Keefer LA, Safer JD, Sigel K, Goodman KA, Rosser BRS, Goldstone SE, Wong SY, Marshall DC. Pleasurable and problematic receptive anal intercourse and diseases of the colon, rectum and anus. Nat Rev Gastroenterol Hepatol 2024; 21:377-405. [PMID: 38763974 DOI: 10.1038/s41575-024-00932-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/12/2024] [Indexed: 05/21/2024]
Abstract
The ability to experience pleasurable sexual activity is important for human health. Receptive anal intercourse (RAI) is a common, though frequently stigmatized, pleasurable sexual activity. Little is known about how diseases of the colon, rectum, and anus and their treatments affect RAI. Engaging in RAI with gastrointestinal disease can be difficult due to the unpredictability of symptoms and treatment-related toxic effects. Patients might experience sphincter hypertonicity, gastrointestinal symptom-specific anxiety, altered pelvic blood flow from structural disorders, decreased sensation from cancer-directed therapies or body image issues from stoma creation. These can result in problematic RAI - encompassing anodyspareunia (painful RAI), arousal dysfunction, orgasm dysfunction and decreased sexual desire. Therapeutic strategies for problematic RAI in patients living with gastrointestinal diseases and/or treatment-related dysfunction include pelvic floor muscle strengthening and stretching, psychological interventions, and restorative devices. Providing health-care professionals with a framework to discuss pleasurable RAI and diagnose problematic RAI can help improve patient outcomes. Normalizing RAI, affirming pleasure from RAI and acknowledging that the gastrointestinal system is involved in sexual pleasure, sexual function and sexual health will help transform the scientific paradigm of sexual health to one that is more just and equitable.
Collapse
Affiliation(s)
- Daniel R Dickstein
- Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Collin R Edwards
- Department of Radiology, Vagelos College of Physicians and Surgeons of Columbia University, New York, NY, USA
| | - Catherine R Rowan
- Inflammatory Bowel Disease Unit, Division of Gastroenterology and Hepatology, University of Calgary, Calgary, Alberta, Canada
| | - Bella Avanessian
- Center for Transgender Medicine and Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Plastic and Reconstructive Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Barbara M Chubak
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christopher W Wheldon
- Department of Social and Behavioral Sciences, College of Public Health at Temple University, Philadelphia, PA, USA
| | - Priya K Simoes
- The Dr. Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Michael H Buckstein
- Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Laurie A Keefer
- The Dr. Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joshua D Safer
- Center for Transgender Medicine and Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Endocrinology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Keith Sigel
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Karyn A Goodman
- Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - B R Simon Rosser
- Division of Epidemiology and Community Health, School of Public Health at University of Minnesota, Minneapolis, MN, USA
| | - Stephen E Goldstone
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Serre-Yu Wong
- The Dr. Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Deborah C Marshall
- Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
27
|
Sidiropoulos T, Dovrolis N, Katifelis H, Michalopoulos NV, Kokoropoulos P, Arkadopoulos N, Gazouli M. Dysbiosis Signature of Fecal Microbiota in Patients with Pancreatic Adenocarcinoma and Pancreatic Intraductal Papillary Mucinous Neoplasms. Biomedicines 2024; 12:1040. [PMID: 38791002 PMCID: PMC11117863 DOI: 10.3390/biomedicines12051040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 05/05/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Pancreatic cancer (PC) ranks as the seventh leading cause of cancer-related deaths, with approximately 500,000 new cases reported in 2020. Existing strategies for early PC detection primarily target individuals at high risk of developing the disease. Nevertheless, there is a pressing need to identify innovative clinical approaches and personalized treatments for effective PC management. This study aimed to explore the dysbiosis signature of the fecal microbiota in PC and potential distinctions between its Intraductal papillary mucinous neoplasm (IPMN) and pancreatic ductal adenocarcinoma (PDAC) phenotypes, which could carry diagnostic significance. The study enrolled 33 participants, including 22 diagnosed with PDAC, 11 with IPMN, and 24 healthy controls. Fecal samples were collected and subjected to microbial diversity analysis across various taxonomic levels. The findings revealed elevated abundances of Firmicutes and Proteobacteria in PC patients, whereas healthy controls exhibited higher proportions of Bacteroidota. Both LEfSe and Random Forest analyses indicated the microbiome's potential to effectively distinguish between PC and healthy control samples but fell short of differentiating between IPMN and PDAC samples. These results contribute to the current understanding of this challenging cancer type and highlight the applications of microbiome research. In essence, the study provides clear evidence of the gut microbiome's capability to serve as a biomarker for PC detection, emphasizing the steps required for further differentiation among its diverse phenotypes.
Collapse
Affiliation(s)
- Theodoros Sidiropoulos
- 4th Department of Surgery, Attikon University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece; (T.S.); (N.V.M.); (P.K.); (N.A.)
| | - Nikolas Dovrolis
- Laboratory of Biology, Department of Basic Medical Sciences, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (N.D.); (H.K.)
| | - Hector Katifelis
- Laboratory of Biology, Department of Basic Medical Sciences, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (N.D.); (H.K.)
| | - Nikolaos V. Michalopoulos
- 4th Department of Surgery, Attikon University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece; (T.S.); (N.V.M.); (P.K.); (N.A.)
| | - Panagiotis Kokoropoulos
- 4th Department of Surgery, Attikon University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece; (T.S.); (N.V.M.); (P.K.); (N.A.)
| | - Nikolaos Arkadopoulos
- 4th Department of Surgery, Attikon University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece; (T.S.); (N.V.M.); (P.K.); (N.A.)
| | - Maria Gazouli
- Laboratory of Biology, Department of Basic Medical Sciences, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (N.D.); (H.K.)
| |
Collapse
|
28
|
Cadena-Ullauri S, Guevara-Ramírez P, Ruiz-Pozo VA, Tamayo-Trujillo R, Paz-Cruz E, Zambrano-Villacres R, Simancas-Racines D, Zambrano AK. The effect of intermittent fasting on microbiota as a therapeutic approach in obesity. Front Nutr 2024; 11:1393292. [PMID: 38725575 PMCID: PMC11079193 DOI: 10.3389/fnut.2024.1393292] [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: 02/28/2024] [Accepted: 04/15/2024] [Indexed: 05/12/2024] Open
Abstract
Obesity, a public health challenge, arises from a complex interplay of factors such as dietary habits and genetic predisposition. Alterations in gut microbiota, characterized by an imbalance between Firmicutes and Bacteroidetes, further exacerbate metabolic dysregulation, promoting inflammation and metabolic disturbances. Intermittent fasting (IF) emerges as a promising dietary strategy showing efficacy in weight management and favoring fat utilization. Studies have used mice as animal models to demonstrate the impact of IF on gut microbiota composition, highlighting enhanced metabolism and reduced inflammation. In humans, preliminary evidence suggests that IF promotes a healthy microbiota profile, with increased richness and abundance of beneficial bacterial strains like Lactobacillus and Akkermansia. However, further clinical trials are necessary to validate these findings and elucidate the long-term effects of IF on microbiota and obesity. Future research should focus on specific tissues and cells, the use of advanced -omics techniques, and exploring the interaction of IF with other dietary patterns, to analyze microbiota composition, gene expression, and potential synergistic effects for enhanced metabolic health. While preliminary evidence supports the potential benefits of IF in obesity management and microbiota regulation, further research with diverse populations and robust methodologies is necessary to understand its implications and optimize personalized dietary interventions. This review explores the potential impact of IF on gut microbiota and its intricate relationship with obesity. Specifically, we will focus on elucidating the underlying mechanisms through which IF affects microbiota composition, as well as its subsequent effects on obesity.
Collapse
Affiliation(s)
- Santiago Cadena-Ullauri
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| | - Patricia Guevara-Ramírez
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| | - Viviana A. Ruiz-Pozo
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| | - Rafael Tamayo-Trujillo
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| | - Elius Paz-Cruz
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| | | | - Daniel Simancas-Racines
- Centro de Investigación de Salud Pública y Epidemiología Clínica (CISPEC), Universidad UTE, Quito, Ecuador
| | - Ana Karina Zambrano
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| |
Collapse
|
29
|
Tilves C, Mueller NT, Zmuda JM, Kuipers AL, Methé B, Li K, Carr JJ, Terry JG, Wheeler V, Nair S, Miljkovic I. Associations of Fecal Microbiota with Ectopic Fat in African Caribbean Men. Microorganisms 2024; 12:812. [PMID: 38674756 PMCID: PMC11052294 DOI: 10.3390/microorganisms12040812] [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: 03/25/2024] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
OBJECTIVE The gut microbiome has been associated with visceral fat (VAT) in European and Asian populations; however, associations with VAT and with ectopic fats among African-ancestry individuals are not known. Our objective was to investigate cross-sectional associations of fecal microbiota diversity and composition with VAT and ectopic fat, as well as body mass index (BMI), among middle-aged and older African Caribbean men. METHODS We included in our analysis n = 193 men (mean age = 62.2 ± 7.6 years; mean BMI = 28.3 ± 4.9 kg/m2) from the Tobago Health Study. We assessed fecal microbiota using V4 16s rRNA gene sequencing. We evaluated multivariable-adjusted associations of microbiota features (alpha diversity, beta diversity, microbiota differential abundance) with BMI and with computed tomography-measured VAT and ectopic fats (pericardial and intermuscular fat; muscle and liver attenuation). RESULTS Lower alpha diversity was associated with higher VAT and BMI, and somewhat with higher pericardial and liver fat. VAT, BMI, and pericardial fat each explained similar levels of variance in beta diversity. Gram-negative Prevotellaceae and Negativicutes microbiota showed positive associations, while gram-positive Ruminococcaceae microbiota showed inverse associations, with ectopic fats. CONCLUSIONS Fecal microbiota features associated with measures of general adiposity also extend to metabolically pernicious VAT and ectopic fat accumulation in older African-ancestry men.
Collapse
Affiliation(s)
- Curtis Tilves
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO 80045, USA;
- LEAD Center, Colorado School of Public Health, Aurora, CO 80045, USA
| | - Noel T. Mueller
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO 80045, USA;
- LEAD Center, Colorado School of Public Health, Aurora, CO 80045, USA
- Department of Pediatrics, Colorado School of Medicine, Aurora, CO 80045, USA
| | - Joseph M. Zmuda
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA 15261, USA; (J.M.Z.); (A.L.K.); (I.M.)
| | - Allison L. Kuipers
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA 15261, USA; (J.M.Z.); (A.L.K.); (I.M.)
| | - Barbara Methé
- Center for Medicine and the Microbiome, University of Pittsburgh, Pittsburgh, PA 15213, USA; (B.M.); (K.L.)
| | - Kelvin Li
- Center for Medicine and the Microbiome, University of Pittsburgh, Pittsburgh, PA 15213, USA; (B.M.); (K.L.)
| | - John Jeffrey Carr
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (J.J.C.); (J.G.T.); (S.N.)
| | - James G. Terry
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (J.J.C.); (J.G.T.); (S.N.)
| | - Victor Wheeler
- Tobago Health Studies Office, TTMF Jerningham Court, James Park Upper Scarborough, Scarborough, Trinidad and Tobago;
| | - Sangeeta Nair
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (J.J.C.); (J.G.T.); (S.N.)
| | - Iva Miljkovic
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA 15261, USA; (J.M.Z.); (A.L.K.); (I.M.)
| |
Collapse
|
30
|
Ferenc K, Sokal-Dembowska A, Helma K, Motyka E, Jarmakiewicz-Czaja S, Filip R. Modulation of the Gut Microbiota by Nutrition and Its Relationship to Epigenetics. Int J Mol Sci 2024; 25:1228. [PMID: 38279228 PMCID: PMC10816208 DOI: 10.3390/ijms25021228] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/12/2024] [Accepted: 01/16/2024] [Indexed: 01/28/2024] Open
Abstract
The intestinal microbiota is a community of microorganisms inhabiting the human intestines, potentially influencing both physiological and pathophysiological processes in the human body. Existing evidence suggests that nutrients can influence the modulation of the gut microbiota. However, there is still limited evidence regarding the effects of vitamin and mineral supplementation on the human gut microbiota through epigenetic modification. It is plausible that maintaining an adequate dietary intake of vitamin D, iron, fibre, zinc and magnesium may have a beneficial effect on alleviating inflammation in the body, reducing oxidative stress, and improving the condition of the intestinal microbiota through various epigenetic mechanisms. Moreover, epigenetics involves alterations in the phenotype of a cell without changing its fundamental DNA sequence. It appears that the modulation of the microbiota by various nutrients may lead to epigenetic regulation. The correlations between microbiota and epigenetics are potentially interdependent. Therefore, the primary objective of this review is to identify the complex relationships between diet, gut microbiota, and epigenetic regulation. These interactions could play a crucial role in systemic health.
Collapse
Affiliation(s)
- Katarzyna Ferenc
- Institute of Medicine, Medical College of Rzeszow University, 35-959 Rzeszow, Poland
| | - Aneta Sokal-Dembowska
- Institute of Health Sciences, Medical College of Rzeszow University, 35-959 Rzeszow, Poland
| | - Kacper Helma
- Institute of Health Sciences, Medical College of Rzeszow University, 35-959 Rzeszow, Poland
| | - Elżbieta Motyka
- Centre for Innovative Research in Medical and Natural Sciences, Medical College of Rzeszow University, 35-959 Rzeszow, Poland
| | | | - Rafał Filip
- Institute of Medicine, Medical College of Rzeszow University, 35-959 Rzeszow, Poland
- Department of Gastroenterology with IBD Unit, Clinical Hospital No. 2, 35-301 Rzeszow, Poland
| |
Collapse
|
31
|
Bell V, Varzakas T, Psaltopoulou T, Fernandes T. Sickle Cell Disease Update: New Treatments and Challenging Nutritional Interventions. Nutrients 2024; 16:258. [PMID: 38257151 PMCID: PMC10820494 DOI: 10.3390/nu16020258] [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: 12/19/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
Sickle cell disease (SCD), a distinctive and often overlooked illness in the 21st century, is a congenital blood disorder characterized by considerable phenotypic diversity. It comprises a group of disorders, with sickle cell anemia (SCA) being the most prevalent and serious genotype. Although there have been some systematic reviews of global data, worldwide statistics regarding SCD prevalence, morbidity, and mortality remain scarce. In developed countries with a lower number of sickle cell patients, cutting-edge technologies have led to the development of new treatments. However, in developing settings where sickle cell disease (SCD) is more prevalent, medical management, rather than a cure, still relies on the use of hydroxyurea, blood transfusions, and analgesics. This is a disease that affects red blood cells, consequently affecting most organs in diverse manners. We discuss its etiology and the advent of new technologies, but the aim of this study is to understand the various types of nutrition-related studies involving individuals suffering from SCD, particularly in Africa. The interplay of the environment, food, gut microbiota, along with their respective genomes collectively known as the gut microbiome, and host metabolism is responsible for mediating host metabolic phenotypes and modulating gut microbiota. In addition, it serves the purpose of providing essential nutrients. Moreover, it engages in direct interactions with host homeostasis and the immune system, as well as indirect interactions via metabolites. Nutrition interventions and nutritional care are mechanisms for addressing increased nutrient expenditures and are important aspects of supportive management for patients with SCD. Underprivileged areas in Sub-Saharan Africa should be accompanied by efforts to define and promote of the nutritional aspects of SCD. Their importance is key to maintaining well-being and quality of life, especially because new technologies and products remain limited, while the use of native medicinal plant resources is acknowledged.
Collapse
Affiliation(s)
- Victoria Bell
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal;
| | - Theodoros Varzakas
- Department of Food Science and Technology, University of the Peloponnese, 24100 Kalamata, Greece
| | - Theodora Psaltopoulou
- Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Tito Fernandes
- CIISA, Faculty of Veterinary Medicine, University of Lisbon, 1649-004 Lisbon, Portugal
| |
Collapse
|
32
|
Javan GT, Singh K, Finley SJ, Green RL, Sen CK. Complexity of human death: its physiological, transcriptomic, and microbiological implications. Front Microbiol 2024; 14:1345633. [PMID: 38282739 PMCID: PMC10822681 DOI: 10.3389/fmicb.2023.1345633] [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: 11/28/2023] [Accepted: 12/28/2023] [Indexed: 01/30/2024] Open
Abstract
Human death is a complex, time-governed phenomenon that leads to the irreversible cessation of all bodily functions. Recent molecular and genetic studies have revealed remarkable experimental evidence of genetically programmed cellular death characterized by several physiological processes; however, the basic physiological function that occurs during the immediate postmortem period remains inadequately described. There is a paucity of knowledge connecting necrotic pathologies occurring in human organ tissues to complete functional loss of the human organism. Cells, tissues, organs, and organ systems show a range of differential resilience and endurance responses that occur during organismal death. Intriguingly, a persistent ambiguity in the study of postmortem physiological systems is the determination of the trajectory of a complex multicellular human body, far from life-sustaining homeostasis, following the gradual or sudden expiry of its regulatory systems. Recent groundbreaking investigations have resulted in a paradigm shift in understanding the cell biology and physiology of death. Two significant findings are that (i) most cells in the human body are microbial, and (ii) microbial cell abundance significantly increases after death. By addressing the physiological as well as the microbiological aspects of death, future investigations are poised to reveal innovative insights into the enigmatic biological activities associated with death and human decomposition. Understanding the elaborate crosstalk of abiotic and biotic factors in the context of death has implications for scientific discoveries important to informing translational knowledge regarding the transition from living to the non-living. There are important and practical needs for a transformative reestablishment of accepted models of biological death (i.e., artificial intelligence, AI) for more precise determinations of when the regulatory mechanisms for homeostasis of a living individual have ceased. In this review, we summarize mechanisms of physiological, genetic, and microbiological processes that define the biological changes and pathways associated with human organismal death and decomposition.
Collapse
Affiliation(s)
- Gulnaz T. Javan
- Department of Physical and Forensic Sciences, Alabama State University, Montgomery, AL, United States
| | - Kanhaiya Singh
- Department of Surgery, School of Medicine, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Sheree J. Finley
- Department of Physical and Forensic Sciences, Alabama State University, Montgomery, AL, United States
| | - Robert L. Green
- Department of Physical and Forensic Sciences, Alabama State University, Montgomery, AL, United States
| | - Chandan K. Sen
- Department of Surgery, School of Medicine, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
33
|
Ahmad AF, Caparrós-Martin JA, Gray N, Lodge S, Wist J, Lee S, O'Gara F, Shah A, Ward NC, Dwivedi G. Insights into the associations between the gut microbiome, its metabolites, and heart failure. Am J Physiol Heart Circ Physiol 2023; 325:H1325-H1336. [PMID: 37737730 DOI: 10.1152/ajpheart.00436.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/05/2023] [Accepted: 09/17/2023] [Indexed: 09/23/2023]
Abstract
Heart failure (HF) is the end stage of most cardiovascular diseases and remains a significant health problem globally. We aimed to assess whether patients with left ventricular ejection fraction ≤45% had alterations in both the gut microbiome profile and production of associated metabolites when compared with a healthy cohort. We also examined the associated inflammatory, metabolomic, and lipidomic profiles of patients with HF. This single center, observational study, recruited 73 patients with HF and 59 healthy volunteers. Blood and stool samples were collected at baseline and 6-mo follow-up, along with anthropometric and clinical data. When compared with healthy controls, patients with HF had reduced gut bacterial alpha diversity at follow-up (P = 0.004) but not at baseline. The stool microbiota of patients with HF was characterized by a depletion of operational taxonomic units representing commensal Clostridia at both baseline and follow-up. Patients with HF also had significantly elevated baseline plasma acetate (P = 0.007), plasma trimethylamine-N-oxide (TMAO) (P = 0.003), serum soluble CD14 (sCD14; P = 0.005), and soluble CD163 (sCD163; P = 0.004) levels compared with healthy controls. Furthermore, patients with HF had a distinct metabolomic and lipidomic profile at baseline when compared with healthy controls. Differences in the composition of the gut microbiome and the levels of associated metabolites were observed in patients with HF when compared with a healthy cohort. This was also associated with an altered metabolomic and lipidomic profile. Our study identifies microorganisms and metabolites that could represent new therapeutic targets and diagnostic tools in the pathogenesis of HF.NEW & NOTEWORTHY We found a reduction in gut bacterial alpha diversity in patients with heart failure (HF) and that the stool microbiota of patients with HF was characterized by depletion of operational taxonomic units representing commensal Clostridia at both baseline and follow-up. Patients with HF also had altered bacterial metabolites and increased inflammatory profiles compared with healthy controls. A distinct metabolomic and lipidomic profile was present in patients with HF at baseline when compared with healthy controls.
Collapse
Affiliation(s)
- Adilah F Ahmad
- Department of Advanced Clinical and Translational Cardiovascular Imaging, Harry Perkins Institute of Medial Research, Perth, Western Australia, Australia
- Medical School, The University of Western Australia, Perth, Western Australia, Australia
| | - Jose A Caparrós-Martin
- Wal-Yan Respiratory Research Centre, Telethon Kids Institute, Perth, Western Australia, Australia
| | - Nicola Gray
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
| | - Samantha Lodge
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
| | - Julien Wist
- Australian National Phenome Centre and Computational and Systems Medicine, Health Futures Institute, Murdoch University, Perth, Western Australia, Australia
| | - Silvia Lee
- Department of Advanced Clinical and Translational Cardiovascular Imaging, Harry Perkins Institute of Medial Research, Perth, Western Australia, Australia
- Medical School, The University of Western Australia, Perth, Western Australia, Australia
- Department of Microbiology, PathWest Laboratory Medicine, Perth, Western Australia, Australia
| | - Fergal O'Gara
- Wal-Yan Respiratory Research Centre, Telethon Kids Institute, Perth, Western Australia, Australia
- BIOMERIT Research Centre, School of Microbiology, University College Cork, Cork, Ireland
| | - Amit Shah
- Department of Cardiology, Fiona Stanley Hospital, Perth, Western Australia, Australia
| | - Natalie C Ward
- Dobney Hypertension Centre, Medical School, The University of Western Australia, Perth, Western Australia, Australia
| | - Girish Dwivedi
- Department of Advanced Clinical and Translational Cardiovascular Imaging, Harry Perkins Institute of Medial Research, Perth, Western Australia, Australia
- Medical School, The University of Western Australia, Perth, Western Australia, Australia
- Department of Cardiology, Fiona Stanley Hospital, Perth, Western Australia, Australia
| |
Collapse
|
34
|
Abdill RJ, Graham SP, Rubinetti V, Albert FW, Greene CS, Davis S, Blekhman R. Integration of 168,000 samples reveals global patterns of the human gut microbiome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.11.560955. [PMID: 37873416 PMCID: PMC10592789 DOI: 10.1101/2023.10.11.560955] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Understanding the factors that shape variation in the human microbiome is a major goal of research in biology. While other genomics fields have used large, pre-compiled compendia to extract systematic insights requiring otherwise impractical sample sizes, there has been no comparable resource for the 16S rRNA sequencing data commonly used to quantify microbiome composition. To help close this gap, we have assembled a set of 168,484 publicly available human gut microbiome samples, processed with a single pipeline and combined into the largest unified microbiome dataset to date. We use this resource, which is freely available at microbiomap.org, to shed light on global variation in the human gut microbiome. We find that Firmicutes, particularly Bacilli and Clostridia, are almost universally present in the human gut. At the same time, the relative abundance of the 65 most common microbial genera differ between at least two world regions. We also show that gut microbiomes in undersampled world regions, such as Central and Southern Asia, differ significantly from the more thoroughly characterized microbiomes of Europe and Northern America. Moreover, humans in these overlooked regions likely harbor hundreds of taxa that have not yet been discovered due to this undersampling, highlighting the need for diversity in microbiome studies. We anticipate that this new compendium can serve the community and enable advanced applied and methodological research.
Collapse
Affiliation(s)
- Richard J. Abdill
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Samantha P. Graham
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota, USA
| | - Vincent Rubinetti
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO, USA
- Center for Health Artificial Intelligence (CHAI), University of Colorado School of Medicine, Aurora, CO, USA
| | - Frank W. Albert
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota, USA
| | - Casey S. Greene
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO, USA
- Center for Health Artificial Intelligence (CHAI), University of Colorado School of Medicine, Aurora, CO, USA
| | - Sean Davis
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO, USA
- Center for Health Artificial Intelligence (CHAI), University of Colorado School of Medicine, Aurora, CO, USA
| | - Ran Blekhman
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|