1
|
Qi C, Li Z, Tu H, Sun F, Guo W, Di C, He R, Ze X, Zhang L, Gao R, Hu P, Yang W, Li K, Liu J, Pan X, Jin Z, Sun J. 2'-FL and cross-feeding bifidobacteria reshaped the gut microbiota of infants with atopic dermatitis ex vivo and prevented dermatitis in mice post-microbiota transplantation through retinol metabolism activation. Gut Microbes 2025; 17:2474148. [PMID: 40025650 PMCID: PMC11881859 DOI: 10.1080/19490976.2025.2474148] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 02/21/2025] [Accepted: 02/25/2025] [Indexed: 03/04/2025] Open
Abstract
2'-Fucosyllactose (2'-FL), a predominant human milk oligosaccharide, plays a crucial role in the development of the infant gut microbiota and immune system. However, the microbiota of infants with atopic dermatitis (AD) often has difficulty utilizing 2'-FL. Here, we found that strains from human milk, Bifidobacterium bifidum FN120 and Bifidobacterium longum subsp. longum FN103, utilized 2'-FL for growth by cross-feeding. Through an ex vivo continuous fermentation system, we found that 2'-FL and cross-feeding bifidobacteria synergistically enhanced the production of short-chain fatty acids (SCFAs), particularly acetate and propionate, while reshaping the gut microbiota in infants with AD. The reshaped microbiota was then transplanted into oxazolone-induced mice. We observed that AD symptoms in mice were effectively prevented, with significant changes in the ileum microbiota and increased intestinal SCFA levels. RNA sequencing analysis of Peyer's patches in the small intestine revealed activation of the retinol metabolic pathway. Nontargeted metabolomics analysis revealed a significant increase in plasma retinoate levels, which correlated markedly with AD-related markers. Collectively, our study demonstrated that supplementation with cross-feeding bifidobacteria and 2'-FL reshaped the gut microbiota, activated retinol metabolic pathways, promoted immune tolerance, and thereby prevented AD. Our findings provide novel insights into the therapeutic potential of combining prebiotics and probiotics to modulate the gut - skin axis and support immune tolerance in early life, offering a promising strategy for infantile AD management and prevention.
Collapse
Affiliation(s)
- Ce Qi
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Zhongxia Li
- BYHEALTH Institute of Nutrition & Health, Guangzhou, China
| | - Huayu Tu
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Fang Sun
- Pediatrics, Jiaozhou Maternal and Child Health and Family Planning Service Center, Qingdao, China
| | - Wenbo Guo
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Can Di
- BYHEALTH Institute of Nutrition & Health, Guangzhou, China
| | - Ruikun He
- BYHEALTH Institute of Nutrition & Health, Guangzhou, China
| | - Xiaolei Ze
- BYHEALTH Institute of Nutrition & Health, Guangzhou, China
| | - Lintao Zhang
- Pediatrics, Jiaozhou Maternal and Child Health and Family Planning Service Center, Qingdao, China
| | - Ruijuan Gao
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Pengyue Hu
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Wenjing Yang
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Kexin Li
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Jiayi Liu
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Xiaonan Pan
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Zilu Jin
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Jin Sun
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| |
Collapse
|
2
|
Dongre DS, Saha UB, Saroj SD. Exploring the role of gut microbiota in antibiotic resistance and prevention. Ann Med 2025; 57:2478317. [PMID: 40096354 PMCID: PMC11915737 DOI: 10.1080/07853890.2025.2478317] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 02/24/2025] [Accepted: 02/27/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND/INTRODUCTION Antimicrobial resistance (AMR) and the evolution of multiple drug-resistant (MDR) bacteria is of grave public health concern. To combat the pandemic of AMR, it is necessary to focus on novel alternatives for drug development. Within the host, the interaction of the pathogen with the microbiome plays a pivotal role in determining the outcome of pathogenesis. Therefore, microbiome-pathogen interaction is one of the potential targets to be explored for novel antimicrobials. MAIN BODY This review focuses on how the gut microbiome has evolved as a significant component of the resistome as a source of antibiotic resistance genes (ARGs). Antibiotics alter the composition of the native microbiota of the host by favouring resistant bacteria that can manifest as opportunistic infections. Furthermore, gut dysbiosis has also been linked to low-dosage antibiotic ingestion or subtherapeutic antibiotic treatment (STAT) from food and the environment. DISCUSSION Colonization by MDR bacteria is potentially acquired and maintained in the gut microbiota. Therefore, it is pivotal to understand microbial diversity and its role in adapting pathogens to AMR. Implementing several strategies to prevent or treat dysbiosis is necessary, including faecal microbiota transplantation, probiotics and prebiotics, phage therapy, drug delivery models, and antimicrobial stewardship regulation.
Collapse
Affiliation(s)
- Devyani S. Dongre
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University) (SIU), Lavale, Pune, Maharashtra, India
| | - Ujjayni B. Saha
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University) (SIU), Lavale, Pune, Maharashtra, India
| | - Sunil D. Saroj
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University) (SIU), Lavale, Pune, Maharashtra, India
| |
Collapse
|
3
|
Lau RI, Su Q, Ng SC. Long COVID and gut microbiome: insights into pathogenesis and therapeutics. Gut Microbes 2025; 17:2457495. [PMID: 39854158 PMCID: PMC11776476 DOI: 10.1080/19490976.2025.2457495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 01/15/2025] [Accepted: 01/17/2025] [Indexed: 01/26/2025] Open
Abstract
Post-acute coronavirus disease 2019 syndrome (PACS), following severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection or coronavirus disease 2019 (COVID-19), is typically characterized by long-term debilitating symptoms affecting multiple organs and systems. Unfortunately, there is currently a lack of effective treatment strategies. Altered gut microbiome has been proposed as one of the plausible mechanisms involved in the pathogenesis of PACS; extensive studies have emerged to bridge the gap between the persistent symptoms and the dysbiosis of gut microbiome. Recent clinical trials have indicated that gut microbiome modulation using probiotics, prebiotics, and fecal microbiota transplantation (FMT) led to improvements in multiple symptoms related to PACS, including fatigue, memory loss, difficulty in concentration, gastrointestinal upset, and disturbances in sleep and mood. In this review, we highlight the latest evidence on the key microbial alterations observed in PACS, as well as the use of microbiome-based therapeutics in managing PACS symptoms. These novel findings altogether shed light on the treatment of PACS and other chronic conditions.
Collapse
Affiliation(s)
- Raphaela I. Lau
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong KongSAR, China
- Microbiota I-Center (MagIC), Hong KongSAR, China
| | - Qi Su
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong KongSAR, China
- Microbiota I-Center (MagIC), Hong KongSAR, China
| | - Siew C. Ng
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong KongSAR, China
- Microbiota I-Center (MagIC), Hong KongSAR, China
- Li Ka Shing Institute of Health Sciences, State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong KongSAR, China
| |
Collapse
|
4
|
Tancredi A, Matthijs T, Cox E, Van Immerseel F, Goossens E. From mother to piglet: the lasting influence of the maternal microbiome. Anim Microbiome 2025; 7:52. [PMID: 40420287 DOI: 10.1186/s42523-025-00420-0] [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: 09/28/2024] [Accepted: 05/06/2025] [Indexed: 05/28/2025] Open
Abstract
BACKGROUND Given their crucial roles in agriculture and biomedical research, promoting pig health is essential. A balanced gut microbiota is vital for immune development, metabolism and pathogen resistance, and requires optimal initial colonization by beneficial bacteria. This becomes particularly evident during early life stages, like suckling and weaning, where disruptions can lead to long-term health issues. Understanding the factors influencing microbiome development during these phases is fundamental for enhancing pig health. On these basis, rectal swab samples from eighteen sow-piglet pairs were collected at multiple time points from 7 days after birth to 10 days post-weaning, and analyzed through 16S rRNA gene sequencing. This study aims to understand the maternal influence on piglet microbiota development during the suckling-weaning period, exploring microbial diversity, composition and additional influencing factors such as age, piglet and weaning. RESULTS α diversity significantly increased with piglet age (p < 0.001) and stabilized upon weaning, with maternal influence and differences between individual piglet affecting variability before weaning. Post-weaning α diversity was influenced by the pen environment (contributing to 14.5-16% of the variability between piglets) rather than age. Both the sow (~ 9.6%) and age of the piglets (20-30%) had a significant impact on the microbial β diversity over the entire timeframe. Moreover, at 10 days post-weaning a significant influence of the cage mates on piglets microbial β diversity was observed (~ 24.6%). Source-tracking analysis revealed a significant maternal contribution to piglet microbiome at 7 days (31.68%), which decreased over time but remained at 13.33% post-weaning. Piglet microbiome exhibited consistency across time, with 22.55-61.23% of bacteria retained from previous stages. Cage mates contributed 53.54% to the microbiome at 10 days post-weaning. Additionally, 68.32% of piglets microbiome at 7 days was derived from sources not included in the study, decreasing to 37.6% by 10 days post-weaning. ASV-level analysis showed that the majority of maternally transmitted ASVs pre-weaning persisted until the last time point, with both beneficial bacteria and pathobionts being transmitted. CONCLUSIONS This study highlights the significant influence of maternal microbiota on piglet gut microbiome development, affecting both diversity and composition. Beneficial bacteria are transmitted from mothers to offspring and persist through early developmental stages, thereby emphasizing the long-lasting impact of maternal microbiome and the importance of early microbial colonization for piglet health.
Collapse
Affiliation(s)
- Alessandra Tancredi
- Livestock Gut Health Team (LiGHT) Ghent, Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
- Laboratory of Immunology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Thomas Matthijs
- Laboratory of Immunology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Eric Cox
- Laboratory of Immunology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Filip Van Immerseel
- Livestock Gut Health Team (LiGHT) Ghent, Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Evy Goossens
- Livestock Gut Health Team (LiGHT) Ghent, Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium.
| |
Collapse
|
5
|
Izadifar Z, Stejskalova A, Gulati A, Gutzeit O, Ingber DE. Human Cervix Chip: A Preclinical Model for Studying the Role of the Cervical Mucosa and Microbiome in Female Reproductive Health. Bioessays 2025:e70014. [PMID: 40401380 DOI: 10.1002/bies.70014] [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: 11/25/2024] [Revised: 04/04/2025] [Accepted: 04/08/2025] [Indexed: 05/23/2025]
Abstract
Advancements in women's reproductive health have been hindered by insufficient knowledge and the underrepresentation of women in research, leading to symptom-focused care with poor outcomes. Modeling female reproductive biology and disease pathophysiology has been challenging due to the complexity and dynamic nature of the female organs. Here, we briefly review recent advancements made with a new in vitro microfluidic organ-on-a-chip model of the human cervix (Cervix Chip) that faithfully mimics key features of the cervix, including mucus production and physiological responses to hormonal, environmental, and microbial stimuli. We also discuss how this preclinical platform can provide a way to obtain unique insights into the role of mucosal immunity, genetic and risk factors, as well as microbiome and pathogen interactions in human cervix health and disease, while bridging knowledge gaps in fertility and pregnancy-related conditions. By enabling preclinical drug screening and accelerating translational research, the Cervix Chip holds the potential to improve the development of therapeutics, diagnostics, and ultimately, the sexual and reproductive health of millions of women globally.
Collapse
Affiliation(s)
- Zohreh Izadifar
- Urology Department, Boston Children's Hospital and Department of Surgery, Harvard Medical School, Boston, Massachusetts, USA
| | - Anna Stejskalova
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts, USA
| | - Aakanksha Gulati
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts, USA
| | - Ola Gutzeit
- IVF Unit, Department of Obstetrics and Gynecology, Rambam Medical Center, Haifa, Israel
- Clinical Research Institute at Rambam (CRIR), Rambam Health Care Campus, Haifa, Israel
| | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts, USA
- Vascular Biology Program, Boston Children's Hospital and Department of Surgery, Harvard Medical School, Boston, Massachusetts, USA
- Harvard John A. Paulson School of Engineering and Applied Sciences, Cambridge, Massachusetts, USA
| |
Collapse
|
6
|
Liaquat M, Minihane AM, Vauzour D, Pontifex MG. The gut microbiota in menopause: Is there a role for prebiotic and probiotic solutions? Post Reprod Health 2025:20533691251340491. [PMID: 40335047 DOI: 10.1177/20533691251340491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
The gut microbiota, comprising a diverse array of microorganisms in the gastrointestinal tract, has emerged as a key player in human health. Emerging research indicates that this gut microbial composition is influenced by sex. These sex differences are not necessarily static and likely alter across the life course in response to several factors including changing hormone profile. As such, the menopause transition-a pivotal phase in female ageing in which the hormone profile changes dramatically is receiving increasing attention. Declining estrogen which occurs during menopause appears to influence the microbiota, which may in turn contribute to menopause-related conditions such as weight gain, bone health, cancer risk and cognitive health. The modulation of estrogen through the gut's 'estrobolome', a collection of bacterial genes involved in estrogen metabolism, may offer explanation for some of the interindividual differences observed during menopause (e.g. length, symptoms and disease risk). Therapeutic modulation of the gut microbiota therefore represents a potential approach towards managing menopausal symptoms. Indeed, prebiotics and probiotics such as Lactobacillus have been shown to increase bacterial diversity and improve metabolic and overall health in menopausal women. However, evidence remains limited regarding the specific underlying mechanisms, highlighting an urgent need for a research focus in the area. This review summarizes the current understanding of the gut microbiota's role in menopausal health and the potential of prebiotics and probiotics as therapeutic interventions. Further research into gut microbiota modulation may enable more effective, personalised treatments for menopause-associated health challenges, and supporting women's health into older ages.
Collapse
Affiliation(s)
- Marrium Liaquat
- Norwich Medical School, University of East Anglia, Norwich, UK
| | | | - David Vauzour
- Norwich Medical School, University of East Anglia, Norwich, UK
| | | |
Collapse
|
7
|
Milhouse W, Organski AC, Sun X, Ai D, Zhou B, Cross TL, Ren H. Microbiome affects mice metabolic homeostasis via differential regulation of gene expression in the brain and gut. Physiol Rep 2025; 13:e70373. [PMID: 40387487 PMCID: PMC12087290 DOI: 10.14814/phy2.70373] [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: 02/03/2025] [Revised: 04/25/2025] [Accepted: 05/01/2025] [Indexed: 05/20/2025] Open
Abstract
The gut microbiome (GMB) regulates digestion, metabolism, immunity, and energy homeostasis. This study investigates how gut microbiota integrate the regulation in the neuroendocrine and enteroendocrine systems, with a focus on G protein-coupled receptors (GPCRs) in the brain-gut axis and sex differences. Germ-free (GF) mice exhibited increased hypothalamic expression of the anorexigenic neuropeptide and decreased expression of the negative regulator of leptin signaling. GF males had significantly lower serum leptin levels compared to conventional (CON) males, highlighting a potential link between the microbiome and leptin resistance. In the gut, GF mice demonstrated heightened expression of anorexigenic gut hormones, including peptide YY (Pyy) and cholecystokinin (Cck), in addition to increased levels of G protein-coupled receptors (GPCRs) involved in gut hormone secretion and nutrient metabolism, particularly in females. While carbohydrate metabolism genes were upregulated in CON mice, lipid metabolism genes were predominantly higher in GF mice. These findings suggest that the gut microbiota downregulates genes involved in appetite suppression, modulates GPCRs linked to gut hormone secretion, and contributes to leptin resistance, particularly in males. This research underscores the importance of the gut microbiome in host metabolism and reveals potential molecular targets for novel treatments of metabolic diseases.
Collapse
Affiliation(s)
- Wynne Milhouse
- Department of PediatricsHerman B. Wells Center for Pediatric Research, Indiana University School of MedicineIndianapolisIndianaUSA
- Center for Diabetes and Metabolic DiseaseIndiana University School of MedicineIndianapolisIndianaUSA
- Stark Neurosciences Research Institute, Indiana University School of MedicineIndianapolisIndianaUSA
| | | | - Xun Sun
- Department of PediatricsHerman B. Wells Center for Pediatric Research, Indiana University School of MedicineIndianapolisIndianaUSA
- Center for Diabetes and Metabolic DiseaseIndiana University School of MedicineIndianapolisIndianaUSA
- Stark Neurosciences Research Institute, Indiana University School of MedicineIndianapolisIndianaUSA
| | - Derek Ai
- Center for Diabetes and Metabolic DiseaseIndiana University School of MedicineIndianapolisIndianaUSA
| | - Baohua Zhou
- Department of PediatricsHerman B. Wells Center for Pediatric Research, Indiana University School of MedicineIndianapolisIndianaUSA
| | - Tzu‐Wen L. Cross
- Department of Nutrition SciencePurdue UniversityWest LafayetteIndianaUSA
| | - Hongxia Ren
- Department of PediatricsHerman B. Wells Center for Pediatric Research, Indiana University School of MedicineIndianapolisIndianaUSA
- Center for Diabetes and Metabolic DiseaseIndiana University School of MedicineIndianapolisIndianaUSA
- Stark Neurosciences Research Institute, Indiana University School of MedicineIndianapolisIndianaUSA
| |
Collapse
|
8
|
Molloy MM, McLennan EA, Fox S, Belov K, Hogg CJ. Range-Wide Assessment of the Tasmanian Devil Gut Microbiome. Ecol Evol 2025; 15:e71196. [PMID: 40330104 PMCID: PMC12050263 DOI: 10.1002/ece3.71196] [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: 10/28/2024] [Revised: 02/20/2025] [Accepted: 03/18/2025] [Indexed: 05/08/2025] Open
Abstract
The gut microbiome is an important component of host health and function and is influenced by internal and external factors such as host phylogeny, age, diet, and environment. Monitoring the gut microbiome has become an increasingly important management tool for wild populations of threatened species. The Tasmanian devil (Sarcophilus harrisii) is the largest extant carnivorous marsupial from the island state of Tasmania, Australia. Devils are currently endangered due to devil facial tumor disease. Previous assessments have shown differences between captive and wild devil gut microbiomes and changes during translocations. However, wild gut microbiome variability across Tasmania and the drivers of these differences are not well understood. We conducted a range-wide assessment of gut microbiomes at 10 locations across Tasmania, via 16S rRNA sequencing, and tested the influence of diet (12S vertebrate sequencing), location, sex, and cohort. We show that the five most abundant phyla and genera were consistent across all 10 locations. Location, cohort, and sex impacted bacterial richness, but location did not impact diversity. While there were differences in diet across the state, there was no strong evidence of differences between juveniles and adults, nor between males and females. Contrary to our hypothesis, the vertebrate diet explained a small amount of variation in microbial communities. We suspect that other variables, such as environmental factors and immune system development, may have a stronger influence on gut microbiome variability. Dietary components missed by our 12S primer, including invertebrates and plants, may also contribute to these patterns. Adjustments to dietary supplementation are not recommended when preparing devils for translocation to different sites. Future research should prioritize collecting environmental samples for microbial analysis and integrating metabolomics to elucidate functional differences associated with Tasmanian devil gut microbiome variability.
Collapse
Affiliation(s)
- Meadhbh M. Molloy
- School of Life and Environmental SciencesThe University of SydneySydneyNew South WalesAustralia
- Department of Environmental Science and PolicyGeorge Mason UniversityFairfaxVirginiaUSA
| | - Elspeth A. McLennan
- School of Life and Environmental SciencesThe University of SydneySydneyNew South WalesAustralia
| | - Samantha Fox
- Save the Tasmanian Devil ProgramDepartment of Natural Resources and EnvironmentHobartTasmaniaAustralia
- Toledo Zoo and AquariumToledoOhioUSA
| | - Katherine Belov
- School of Life and Environmental SciencesThe University of SydneySydneyNew South WalesAustralia
| | - Carolyn J. Hogg
- School of Life and Environmental SciencesThe University of SydneySydneyNew South WalesAustralia
| |
Collapse
|
9
|
Freeman LR. Interactions between sex hormones and the gut microbiome. Brain Behav Immun 2025; 126:313-314. [PMID: 40010547 DOI: 10.1016/j.bbi.2025.02.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 02/22/2025] [Indexed: 02/28/2025] Open
Affiliation(s)
- Linnea R Freeman
- Furman University, Department of Biology and Neurosciences Program, Greenville, South Carolina, 29613, United States
| |
Collapse
|
10
|
Zhang R, Zhang X, Lau HCH, Yu J. Gut microbiota in cancer initiation, development and therapy. SCIENCE CHINA. LIFE SCIENCES 2025; 68:1283-1308. [PMID: 39821827 DOI: 10.1007/s11427-024-2831-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 12/12/2024] [Indexed: 01/19/2025]
Abstract
Cancer has long been associated with genetic and environmental factors, but recent studies reveal the important role of gut microbiota in its initiation and progression. Around 13% of cancers are linked to infectious agents, highlighting the need to identify the specific microorganisms involved. Gut microbiota can either promote or inhibit cancer growth by influencing oncogenic signaling pathways and altering immune responses. Dysbiosis can lead to cancer, while certain probiotics and their metabolites may help reestablish micro-ecological balance and improve anti-tumor immune responses. Research into targeted approaches that enhance therapy with probiotics is promising. However, the effects of probiotics in humans are complex and not yet fully understood. Additionally, methods to counteract harmful bacteria are still in development. Early clinical trials also indicate that modifying gut microbiota may help manage side effects of cancer treatments. Ongoing research is crucial to understand better how gut microbiota can be used to improve cancer prevention and treatment outcomes.
Collapse
Affiliation(s)
- Ruyi Zhang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiang Zhang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Harry Cheuk Hay Lau
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Jun Yu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
11
|
Koller AM, Săsăran MO, Mărginean CO. Small Intestinal Bacterial Overgrowth and Pediatric Obesity-A Systematic Review. Nutrients 2025; 17:1499. [PMID: 40362809 PMCID: PMC12073544 DOI: 10.3390/nu17091499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Revised: 04/26/2025] [Accepted: 04/26/2025] [Indexed: 05/15/2025] Open
Abstract
Background/Objectives: Childhood obesity is a growing global concern linked to metabolic disorders such as nonalcoholic fatty liver disease (NAFLD). Small intestinal bacterial overgrowth (SIBO) may exacerbate these conditions by promoting systemic inflammation and metabolic dysfunction. This review evaluates the prevalence of SIBO in obese children, its association with inflammatory and metabolic markers, and the efficacy of diagnostic and therapeutic strategies. Methods: A systematic search of PubMed, Scopus, and Web of Science (2010-present) was conducted using Boolean operators: ('small intestinal bacterial overgrowth' OR 'SIBO') AND 'prevalence' AND ('low-grade inflammatory markers' OR 'metabolic status') AND 'gut microbiome' AND 'dysbiosis' AND 'obese children'. Results: The data show that SIBO is frequently observed in obese pediatric populations and is associated with gut dysbiosis, impaired nutrient absorption, and reduced production of short-chain fatty acids. These changes contribute to increased intestinal permeability, endotoxemia, and chronic low-grade inflammation. Several microbial taxa have been proposed as biomarkers and therapeutic targets. Diagnostic inconsistencies persist, but treatments such as probiotics, prebiotics, dietary interventions, and selective antibiotics show potential, pending further validation. Conclusions: Early identification and treatment of SIBO with tailored strategies may help reduce metabolic complications and improve outcomes in children with obesity.
Collapse
Affiliation(s)
- Ana Maria Koller
- Doctoral School, “George Emil Palade” University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Gheorghe Marinescu Street No 38, 540136 Targu Mures, Romania;
| | - Maria Oana Săsăran
- Department of Pediatrics 3, “George Emil Palade” University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Gheorghe Marinescu Street No 38, 540136 Targu Mures, Romania
| | - Cristina Oana Mărginean
- Department of Pediatrics 1, “George Emil Palade” University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Gheorghe Marinescu Street No 38, 540136 Targu Mures, Romania;
| |
Collapse
|
12
|
Lázaro Á, Gómez-Ramírez P, Vila-Donat P, Cimbalo A, Manyes L. Effects of pumpkin and fermented whey on fecal microbiota profile against AFB1 and OTA exposure in Wistar rats. Toxicol Mech Methods 2025:1-13. [PMID: 40129370 DOI: 10.1080/15376516.2025.2484636] [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: 01/16/2025] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 03/26/2025]
Abstract
Mycotoxins perturb the gut microbiota performance. Bioactive compounds have been recently used as a new food strategy to diminish mycotoxins bioaccessibility and prevent their toxic effects on human and animal health. Male and female Wistar rats were exposed orally to twelve different diets containing aflatoxin B1 (AFB1) and/or ochratoxin A (OTA) with or without fermented whey (FW) and pumpkin (P) for 28 days. Fecal microbiota using 16S rRNA gene sequencing and subsequent metagenomics analysis were analyzed to study the effect of 28-day exposure through diet of contaminated and enriched feed. QIIME 2 microbiome analysis package (version 2024.5) was used to analyze the demultiplexed data. Mycotoxins-functional ingredients combination contributed more to microbial phylogenetic faith α-diversity rather than the functional ingredients alone, while the same combination reported a microbial α-diversity enhancement in comparison to the mycotoxins alone. Proteobacteria phylum was reduced in rat samples fed with contaminated diets (AFB1, OTA, and AFB1+OTA), while there was an increase-although not in all groups-when adding the functional ingredients. The main difference between the sexes was found in FW+AFB1+OTA group, with males (25%) showing higher % of Proteobacteria than females (1.86%). Phylogenetic diversity faith only focuses on microbial genetic (dis)similarity, not considering the biological function. Morganella morganii, a Proteobacteria found in some groups presents anticancer activity, but it is also related to inflammatory bowel disease and colorectal cancer. To sum up, both mycotoxins and functional ingredients trigger changes in the microbiota profile of Wistar rats in a sex-specific manner.
Collapse
Affiliation(s)
- Álvaro Lázaro
- Biotech Agrifood, University of Valencia, Burjassot, Spain
| | - Pilar Gómez-Ramírez
- Area of Toxicology, Department of Health Sciences, University of Murcia, Murcia, Spain
| | | | | | - Lara Manyes
- Biotech Agrifood, University of Valencia, Burjassot, Spain
| |
Collapse
|
13
|
Zhang M, Zhou C, Li X, Li H, Han Q, Chen Z, Tang W, Yin J. Interactions between Gut Microbiota, Host Circadian Rhythms, and Metabolic Diseases. Adv Nutr 2025; 16:100416. [PMID: 40139315 DOI: 10.1016/j.advnut.2025.100416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 03/17/2025] [Accepted: 03/21/2025] [Indexed: 03/29/2025] Open
Abstract
The circadian rhythm arises endogenously from genetically encoded molecular clocks, wherein the components collaborate to induce cyclic fluctuations, occurring approximately every 24 h. The rhythms synchronize biological processes with regular and predictable environmental patterns to guarantee the host metabolism and energy homeostasis function and well-being. Disruptions to circadian rhythms are widely associated with metabolic disorders. Notably, microbial rhythms are influenced by both the host's intrinsic circadian clock and external rhythmic factors (i.e., light-dark cycle, diet patterns, and diet composition), which affect the structure of microbial communities and metabolic functions. Moreover, microbiota and the metabolites also reciprocally influence host rhythms, potentially impacting host metabolic function. This review aimed to explore the bidirectional interactions between the circadian clock, factors influencing host-microbial circadian rhythms, and the effects on lipid metabolism and energy homeostasis.
Collapse
Affiliation(s)
- Mingliang Zhang
- College of Animal Science and Technology, Hunan Agriculture University, Changsha, China
| | | | - Xinguo Li
- Hunan Institute of Animal and Veterinary Science, Changsha, China
| | - Hui Li
- Xiangxi Vocational and Technical College for Nationalities, Jishou, China
| | - Qi Han
- College of Animal Science and Technology, Hunan Agriculture University, Changsha, China
| | - Zhong Chen
- College of Animal Science and Technology, Hunan Agriculture University, Changsha, China
| | - Wenjie Tang
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu, China; Livestock and Poultry Biological Products Key Laboratory of Sichuan Province, Sichuan Animtche Group, Co Ltd, Chengdu, China.
| | - Jie Yin
- College of Animal Science and Technology, Hunan Agriculture University, Changsha, China.
| |
Collapse
|
14
|
Beyoğlu D, Idle JR. The Microbiome and Metabolic Dysfunction-Associated Steatotic Liver Disease. Int J Mol Sci 2025; 26:2882. [PMID: 40243472 PMCID: PMC11988851 DOI: 10.3390/ijms26072882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/17/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a condition wherein excessive fat accumulates in the liver, leading to inflammation and potential liver damage. In this narrative review, we evaluate the tissue microbiota, how they arise and their constituent microbes, and the role of the intestinal and hepatic microbiota in MASLD. The history of bacteriophages (phages) and their occurrence in the microbiota, their part in the potential causation of MASLD, and conversely, "phage therapy" for antibiotic resistance, obesity, and MASLD, are all described. The microbiota metabolism of bile acids and dietary tryptophan and histidine is defined, together with the impacts of their individual metabolites on MASLD pathogenesis. Both periodontitis and intestinal microbiota dysbiosis may cause MASLD, and how individual microorganisms and their metabolites are involved in these processes is discussed. Novel treatment opportunities for MASLD involving the microbiota exist and include fecal microbiota transplantation, probiotics, prebiotics, synbiotics, tryptophan dietary supplements, intermittent fasting, and phages or their holins and endolysins. Although FDA is yet to approve phage therapy in clinical use, there are multiple FDA-approved clinical trials, and this may represent a new horizon for the future treatment of MASLD.
Collapse
Affiliation(s)
- Diren Beyoğlu
- Department of Pharmaceutical and Administrative Sciences, College of Pharmacy and Health Sciences, Western New England University, Springfield, MA 01119, USA;
| | - Jeffrey R. Idle
- Department of Pharmaceutical and Administrative Sciences, College of Pharmacy and Health Sciences, Western New England University, Springfield, MA 01119, USA;
- Department of Biomedical Research, University of Bern, 3008 Bern, Switzerland
| |
Collapse
|
15
|
Lin YC, Wu CC, Li YE, Chen CL, Lin CR, Ni YH. Full-length 16S rRNA Sequencing Reveals Gut Microbiome Signatures Predictive of MASLD in children with obesity. BMC Microbiol 2025; 25:146. [PMID: 40091070 PMCID: PMC11912586 DOI: 10.1186/s12866-025-03849-0] [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: 05/23/2024] [Accepted: 02/26/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND The gut microbiota plays a crucial role in metabolic dysfunction-associated steatotic liver disease (MASLD). Next-generation sequencing technologies are essential for exploring the gut microbiome. While recent advancements in full-length 16S (FL16S) rRNA sequencing offer better taxonomic resolution, whether they establish stronger associations with the risk of MASLD remains to be determined. METHOD This study utilized long-read FL16S and short-read V3-V4 16S rRNA sequencing to profile gut microbiome compositions in age-, sex-, and BMI-matched case-control pairs of obese children with and without MASLD. A random forest predictive model was employed, using gut-microbiota features selected based on the top 35 most abundant taxa or a linear discriminant analysis score greater than 3. The model's performance was evaluated by comparing the area under the receiver operating characteristic curve (AUC) through a tenfold cross-validation method. RESULTS Subjects with MASLD exhibited significantly elevated serum alanine aminotransferase, triglycerides, and homeostasis model assessment of insulin resistance levels compared to controls. At the genus level, the gut microbiome compositions detected by both FL16S and V3-V4 sequencing were similar, predominantly comprising Phocaeicola and Bacteroides, followed by Prevotella, Bifidobacterium, Parabacteroides, and Blautia. The AUC for the model based on FL16S sequencing data (86.98%) was significantly higher than that based on V3-V4 sequencing data (70.27%), as determined by DeLong's test (p = 0.008). CONCLUSION FL16S rRNA sequencing data demonstrates stronger associations with the risk of MASLD in obese children, highlighting its potential for real-world clinical applications.
Collapse
Affiliation(s)
- Yu-Cheng Lin
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei City, Taiwan.
- Department of Healthcare Administration, Asia Eastern University of Science and Technology, New Taipei City, Taiwan.
- Department of Pediatrics, Far Eastern Memorial Hospital, New Taipei City, Taiwan.
| | - Chi-Chien Wu
- Department of Pediatrics, Far Eastern Memorial Hospital, New Taipei City, Taiwan
| | - Yun-Er Li
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei City, Taiwan
| | - Chun-Liang Chen
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei City, Taiwan
| | - Chia-Ray Lin
- Departments of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | - Yen-Hsuan Ni
- Departments of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
16
|
Wagner CA, Frey-Wagner I, Ortiz A, Unwin R, Liabeuf S, Suzumoto Y, Iervolino A, Stasi A, Di Marzo V, Gesualdo L, Massy ZA. The role of the intestinal microbiome in cognitive decline in patients with kidney disease. Nephrol Dial Transplant 2025; 40:ii4-ii17. [PMID: 40080091 PMCID: PMC11905753 DOI: 10.1093/ndt/gfae253] [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: 08/15/2024] [Indexed: 03/15/2025] Open
Abstract
Cognitive decline is frequently seen in patients with chronic kidney disease (CKD). The causes of cognitive decline in these patients are likely to be multifactorial, including vascular disease, uraemic toxins, blood-brain barrier leakage, and metabolic and endocrine changes. Gut dysbiosis is common in patients with CKD and contributes to the increase in uraemic toxins. However, the gut microbiome modulates local and systemic levels of several metabolites such as short-chain fatty acids or derivatives of tryptophan metabolism, neurotransmitters, endocannabinoid-like mediators, bile acids, hormones such as glucagon-like peptide 1 (GLP1) or cholecystokinin (CCK). These factors can affect gut function, immunity, autonomic nervous system activity and various aspects of brain function. Key areas include blood-brain barrier integrity, nerve myelination and survival/proliferation, appetite, metabolism and thermoregulation, mood, anxiety and depression, stress and local inflammation. Alterations in the composition of the gut microbiota and the production of biologically active metabolites in patients with CKD are well documented and are favoured by low-fiber diets, elevated urea levels, sedentary lifestyles, slow stool transit times and polypharmacy. In turn, dysbiosis can modulate brain function and cognitive processes, as discussed in this review. Thus, the gut microbiome may contribute to alterations in cognition in patients with CKD and may be a target for therapeutic interventions using diet, prebiotics and probiotics.
Collapse
Affiliation(s)
- Carsten A Wagner
- Institute of Physiology and Zurich Kidney Center, University of Zurich, Switzerland
| | | | - Alberto Ortiz
- Department of Nephrology and Hypertension, IIS-Fundacion Jimenez Diaz UAM, RICORS2040, Madrid, Spain
| | - Robert Unwin
- Department of Renal Medicine, University College London, London, UK
| | - Sophie Liabeuf
- Pharmacoepidemiology Unit, Department of Clinical Pharmacology, Amiens-Picardie University Medical Center, Amiens, France
- MP3CV Laboratory, Jules Verne University of Picardie, Amiens, France
| | - Yoko Suzumoto
- Biogem, Molecular Biology and Genetics Research Institute, Ariano Irpino, Italy
- Institute of Biochemistry and Cell Biology, National Research Council of Italy, Naples, Italy
| | - Anna Iervolino
- Biogem, Molecular Biology and Genetics Research Institute, Ariano Irpino, Italy
- University of Campania “L. Vanvitelli”, Naples, Italy
| | - Alessandra Stasi
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J) Nephrology, Dialysis and Transplantation Unit, University of Bari Aldo Moro, Bari, Italy
| | - Vincenzo Di Marzo
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, CRIUCPQ and INAF, Centre NUTRISS, Faculties of Medicine and Agriculture and Food Sciences, Université Laval, Québec City, Canada
- Joint International Research Unit for Chemical and Biomolecular Research on the Microbiome and its impact on Metabolic Health and Nutrition (JIRU-MicroMeNu) between Université Laval Québec, Canada and Consiglio Nazionale delle Ricerche, Institute of Biomolecular Chemistry, Pozzuoli, Italy
| | - Loreto Gesualdo
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J) Nephrology, Dialysis and Transplantation Unit, University of Bari Aldo Moro, Bari, Italy
| | - Ziad A Massy
- INSERM Unit 1018, Team 5, CESP, Hôpital Paul Brousse, Paris-Saclay University and Versailles Saint-Quentin-en-Yvelines University (UVSQ), Villejuif, France
- Association pour l'Utilisation du Rein Artificiel dans la région parisienne (AURA) Paris, France and Ambroise Paré University Hospital, APHP, Department of Nephrology Boulogne-Billancourt, Paris, France
| |
Collapse
|
17
|
Winsor NJ, Bayer G, Singh O, Chan JK, Li LY, Lieng BY, Foerster E, Popovic A, Tsankov BK, Maughan H, Lemire P, Tam E, Streutker C, Chen L, Heaver SL, Ley RE, Parkinson J, Montenegro-Burke JR, Birchenough GMH, Philpott DJ, Girardin SE. Cross-kingdom-mediated detection of intestinal protozoa through NLRP6. Cell Host Microbe 2025; 33:388-407.e9. [PMID: 40043701 DOI: 10.1016/j.chom.2025.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 12/13/2024] [Accepted: 02/10/2025] [Indexed: 03/15/2025]
Abstract
Intestinal protists are detected by the host innate immune system through mechanisms that remain poorly understood. Here, we demonstrate that Tritrichomonas protozoa induce thickening of the colonic mucus in an NLRP6-, ASC-, and caspase-11-dependent manner, consistent with the activation of sentinel goblet cells. Mucus growth is recapitulated with cecal extracts from Tritrichomonas-infected mice but not purified protozoa, suggesting that NLRP6 may detect infection-induced microbial dysbiosis. In agreement, Tritrichomonas infection causes a shift in the microbiota with the expansion of Bacteroides and Prevotella, and untargeted metabolomics reveals a dramatic increase in several classes of metabolites, including sphingolipids. Finally, using a combination of gnotobiotic mice and ex vivo mucus analysis, we demonstrate that wild-type, but not sphingolipid-deficient, B. thetaiotaomicron is sufficient to induce NLRP6-dependent sentinel goblet cell function, with the greatest effect observed in female mice. Thus, we propose that NLRP6 is a sensor of intestinal protozoa infection through monitoring microbial sphingolipids.
Collapse
Affiliation(s)
- Nathaniel J Winsor
- Department of Immunology, University of Toronto, Toronto, ON, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Giuliano Bayer
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Ojas Singh
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Jeremy K Chan
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Lu Yi Li
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Brandon Y Lieng
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | | | - Ana Popovic
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada; Molecular Medicine, Hospital for Sick Children, Toronto, ON, Canada
| | - Boyan K Tsankov
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | | | - Paul Lemire
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Elaine Tam
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | | | - Lina Chen
- Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Stacey L Heaver
- Department of Microbiome Science, Max Planck Institute for Biology, Tübingen, Germany
| | - Ruth E Ley
- Department of Microbiome Science, Max Planck Institute for Biology, Tübingen, Germany
| | - John Parkinson
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada; Department of Biochemistry, University of Toronto, Toronto, ON, Canada; Molecular Medicine, Hospital for Sick Children, Toronto, ON, Canada
| | - J Rafael Montenegro-Burke
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada; Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - George M H Birchenough
- Department of Medical Biochemistry, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Dana J Philpott
- Department of Immunology, University of Toronto, Toronto, ON, Canada.
| | - Stephen E Girardin
- Department of Immunology, University of Toronto, Toronto, ON, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
18
|
Conforti F, Pala L, Di Mitri D, Catania C, Cocorocchio E, Laszlo D, Ceresoli G, Locatelli M, Facella F, De Pas T, Rambaldi B, Rambaldi A, Viale G, Bagnardi V, Giaccone G, Mantovani A. Sex hormones, the anticancer immune response, and therapeutic opportunities. Cancer Cell 2025; 43:343-360. [PMID: 40068594 DOI: 10.1016/j.ccell.2025.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 01/11/2025] [Accepted: 02/11/2025] [Indexed: 05/13/2025]
Abstract
Sex-based differences have been observed in the incidence and prognosis of various cancers, as well as in the response to immune check point inhibitors (ICIs). These disparities are partially attributed to sex-based differences in the molecular characteristics of the anticancer immune response, which are largely influenced by sex hormones. Here, we provide a comprehensive overview on how sex hormones affect innate and adaptive immunity and contribute to shaping the features of tumor immune microenvironment and response to anticancer immunotherapy. We also discuss the promising potential and challenges of combining sex hormone manipulation with anticancer immunotherapy as new therapeutic strategy. We surmise that a sex-based perspective should be part of precision medicine approaches, and sex hormones manipulation provides opportunities for innovative immune therapeutic approaches.
Collapse
Affiliation(s)
- Fabio Conforti
- Division of Medical Oncology, Humanitas Gavazzeni, Bergamo, Italy; Humanitas University, Milan, Italy.
| | - Laura Pala
- Division of Medical Oncology, Humanitas Gavazzeni, Bergamo, Italy; Tumor Microenviroment Unit, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Diletta Di Mitri
- Humanitas University, Milan, Italy; Tumor Microenviroment Unit, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Chiara Catania
- Division of Medical Oncology, Humanitas Gavazzeni, Bergamo, Italy
| | | | - Daniele Laszlo
- Division of Medical Oncology, Humanitas Gavazzeni, Bergamo, Italy
| | | | - Marzia Locatelli
- Division of Medical Oncology, Humanitas Gavazzeni, Bergamo, Italy
| | - Flaminia Facella
- Division of Medical Oncology, Humanitas Gavazzeni, Bergamo, Italy
| | - Tommaso De Pas
- Division of Medical Oncology, Humanitas Gavazzeni, Bergamo, Italy
| | - Benedetta Rambaldi
- Department of Oncology and Hematology, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Alessandro Rambaldi
- Department of Oncology and Hematology, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Giuseppe Viale
- Department of Pathology, European Institute of Oncology, IRCCS, Milan, Italy
| | - Vincenzo Bagnardi
- Department of Statistics and Quantitative Methods, University of Milan-Bicocca, Milan, Italy
| | | | - Alberto Mantovani
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; IRCCS Humanitas Research Hospital, Rozzano, Italy; William Harvey Research Institute, Queen Mary University, London, UK
| |
Collapse
|
19
|
Özdemir BC, Bill R, Okyar A, Scheiermann C, Hayoz S, Olivier T. Chrono-immunotherapy as a low-hanging fruit for cancer treatment? A call for pragmatic randomized clinical trials. J Immunother Cancer 2025; 13:e010644. [PMID: 40032603 PMCID: PMC11877229 DOI: 10.1136/jitc-2024-010644] [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: 09/22/2024] [Accepted: 02/12/2025] [Indexed: 03/05/2025] Open
Abstract
The share of immune checkpoint inhibitors (ICIs) used in cancer treatment has rapidly increased in recent years. Although ICIs have the potential to provide a durable survival benefit in a subset of patients, many patients do not respond to these costly and often toxic therapies.Recent retrospective clinical data indicate that the time of day of ICI infusion may be a powerful modulator of their efficacy. These observational studies suggest an enhanced efficacy of morning over evening infusion. However, randomized trials have not confirmed in other fields findings obtained by observational studies, possibly because of selection bias and residual confounding factors. Thus, while the data are intriguing, the time dependence of the efficacy of immunotherapy needs to be confirmed in pragmatic randomized clinical trials. Here, we provide an overview of the modulation of ICI efficacy by the timing of immunotherapy infusion and critically discuss the biological rationale for chrono-immunotherapy, the circadian regulation of the immune system, and the need for pragmatic randomized clinical trials to confirm an effect of the timing of immunotherapy infusions on patient outcomes.
Collapse
Affiliation(s)
- Berna C Özdemir
- Department of Medical Oncology, Bern University Hospital, Bern, Switzerland
| | - Ruben Bill
- Department of Medical Oncology, Bern University Hospital, Bern, Switzerland
| | - Alper Okyar
- Department of Pharmacology, Istanbul University Faculty of Pharmacy, Istanbul, Türkiye
| | - Christoph Scheiermann
- Geneva Centre for Inflammation Research (GCIR), Geneva, Switzerland
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Biomedical Center (BMC), Institute for Cardiovascular Physiology and Pathophysiology, Walter Brendel Center for Experimental Medicine (WBex), Faculty of Medicine, Ludwig-Maximilians-Universität (LMU), Munich, Planegg-Martinsried, Germany
- Translational Research Centre in Onco-Hematology (CRTOH), Geneva, Switzerland
| | - Stefanie Hayoz
- Swiss Group for Clinical Cancer Research (SAKK) Competence Center, Bern, Switzerland
| | - Timothée Olivier
- Department of Oncology, Geneva University Hospital, Geneva, Switzerland
| |
Collapse
|
20
|
Zhao Y, Lai Y, Darweesh SK, Bloem BR, Forsgren L, Hansen J, Katzke VA, Masala G, Sieri S, Sacerdote C, Panico S, Zamora‐Ros R, Sánchez M, Huerta JM, Guevara M, Vinagre‐Aragon A, Vineis P, Lill CM, Miller GW, Peters S, Vermeulen R. Gut Microbial Metabolites and Future Risk of Parkinson's Disease: A Metabolome-Wide Association Study. Mov Disord 2025; 40:556-560. [PMID: 39530417 PMCID: PMC11926468 DOI: 10.1002/mds.30054] [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: 05/28/2024] [Revised: 10/10/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Alterations in gut microbiota are observed in Parkinson's disease (PD). Previous studies on microbiota-derived metabolites in PD were small-scale and post-diagnosis, raising concerns about reverse causality. OBJECTIVES Our goal was to prospectively investigate the association between plasma microbial metabolites and PD risk within a metabolomics framework. METHODS A nested case-control study within the prospective EPIC4PD cohort, measured pre-diagnostic plasma microbial metabolites using untargeted metabolomics. RESULTS Thirteen microbial metabolites were identified nominally associated with PD risk (P-value < 0.05), including amino acids, bile acid, indoles, and hydroxy acid, although none remained significant after multiple testing correction. Three pathways were implicated in PD risk: valine, leucine, and isoleucine degradation, butanoate metabolism, and propanoate metabolism. PD-associated microbial pathways were more pronounced in men, smokers, and overweight/obese individuals. CONCLUSION Changes in microbial metabolites may represent a pre-diagnostic feature of PD. We observed biologically plausible associations between microbial pathways and PD, potentially influenced by individual characteristics. © 2024 The Author(s). Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Yujia Zhao
- Institute for Risk Assessment SciencesUtrecht UniversityUtrechtThe Netherlands
| | - Yunjia Lai
- Department of Environmental Health Sciences, Mailman School of Public HealthColumbia UniversityNew York CityUSA
| | - Sirwan K.L. Darweesh
- Radboud University Medical Center; Donders Institute for Brain, Cognition and Behavior; Department of NeurologyCenter of Expertise for Parkinson and Movement DisordersNijmegenThe Netherlands
| | - Bastiaan R. Bloem
- Radboud University Medical Center; Donders Institute for Brain, Cognition and Behavior; Department of NeurologyCenter of Expertise for Parkinson and Movement DisordersNijmegenThe Netherlands
| | - Lars Forsgren
- Department of Clinical Science, NeurosciencesUmeå UniversityUmeåSweden
| | - Johnni Hansen
- Danish Cancer InstituteDanish Cancer SocietyCopenhagenDenmark
| | - Verena A. Katzke
- Division of Cancer EpidemiologyGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Giovanna Masala
- Institute for Cancer Research, Prevention and Clinical Network (ISPRO)FlorenceItaly
| | - Sabina Sieri
- Epidemiology and Prevention Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di MilanoMilanItaly
| | - Carlotta Sacerdote
- Unit of Cancer EpidemiologyCittà della Salute e della Scienza University‐HospitalTurinItaly
| | | | - Raul Zamora‐Ros
- Unit of Nutrition and Cancer, Cancer Epidemiology Research Program, Catalan Institute of Oncology (ICO), Bellvitge Biomedical Research Institute (IDIBELL)BarcelonaSpain
| | - Maria‐Jose Sánchez
- Escuela Andaluza de Salud Pública (EASP)GranadaSpain
- Instituto de Investigación Biosanitaria ibs.GRANADAGranadaSpain
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP)MadridSpain
| | - José María Huerta
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP)MadridSpain
- Department of EpidemiologyMurcia Regional Health Council‐IMIBMurciaSpain
| | - Marcela Guevara
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP)MadridSpain
- Instituto de Salud Pública y Laboral de NavarraPamplonaSpain
- Navarra Institute for Health Research (IdiSNA)PamplonaSpain
| | - Ana Vinagre‐Aragon
- Movement Disorders Unit, Department of NeurologyUniversity Hospital DonostiaSan SebastianSpain
- BioDonostia Health Research Institute, Neurodegenerative Diseases AreaSan SebastiánSpain
| | - Paolo Vineis
- Ageing Epidemiology Research Unit (AGE), School of Public Health, Imperial College LondonLondonUnited Kingdom
| | - Christina M. Lill
- Ageing Epidemiology Research Unit (AGE), School of Public Health, Imperial College LondonLondonUnited Kingdom
- Institute of Epidemiology and Social MedicineUniversity of MünsterMünsterGermany
| | - Gary W. Miller
- Department of Environmental Health Sciences, Mailman School of Public HealthColumbia UniversityNew York CityUSA
| | - Susan Peters
- Institute for Risk Assessment SciencesUtrecht UniversityUtrechtThe Netherlands
| | - Roel Vermeulen
- Institute for Risk Assessment SciencesUtrecht UniversityUtrechtThe Netherlands
- University Medical Centre UtrechtUtrechtThe Netherlands
| |
Collapse
|
21
|
Brigagão Pacheco da Silva C, Nascimento-Silva EA, Zaramela LS, da Costa BRB, Rodrigues VF, De Martinis BS, Carlos D, Tostes RC. Drinking pattern and sex modulate the impact of ethanol consumption on the mouse gut microbiome. Physiol Genomics 2025; 57:179-194. [PMID: 39918827 DOI: 10.1152/physiolgenomics.00031.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 04/17/2024] [Accepted: 01/28/2025] [Indexed: 03/04/2025] Open
Abstract
Gut microbiota impacts host homeostasis and diseases. Chronic plus binge ethanol consumption has been linked to increased injuries than chronic or binge ethanol intake alone. We hypothesized that distinct shapes in gut microbiota composition are induced by chronic, binge, and the association of these treatments, thereby affecting host functions and contributing to sex-based differences in alcohol use disorders. Male and female C57BL/6J mice were submitted to chronic, binge, or chronic plus binge ethanol feeding. DNA was extracted from fecal microbiota, followed by analysis of the V3-V4 region of the 16S rRNA gene and sequencing on an Illumina platform. Gut microbiome analysis was performed using QIIME v2022.2.0. Functional profiling of the gut microbiome was performed using PICRUSt2. Ethanol differentially affected the gut microbiota of female and male mice. Decreased α diversity was observed in male and female mice from the chronic plus binge and chronic groups, respectively. The genera Faecalibaculum, Lachnospiraceae, and Alistipes were identified as major potential biomarkers for gut dysbiosis induced by ethanol consumption. In addition, ethanol-induced gut dysbiosis altered several metabolic pathways. Ethanol consumption modifies the mouse gut microbiome in a drinking pattern- and sex-dependent manner, potentially leading to different susceptibility to ethanol-related diseases. Chronic plus binge ethanol intake induces a more pronounced gut dysbiosis in male mice. Conversely, chronic ethanol is linked to a greater degree of gut dysbiosis in female mice. The changed gut microbiome may be potentially targeted to prevent, mitigate, or treat alcohol use disorders.NEW & NOTEWORTHY Ethanol alters the mouse gut microbiome in a drinking pattern- and sex-dependent manner. Chronic plus binge ethanol intake induces a more severe gut dysbiosis in male mice, whereas chronic ethanol consumption appears to be a more potent inductor of gut dysbiosis in female mice. Ethanol-induced gut dysbiosis alters several pathways linked to metabolism, genetic and environmental information processing, cellular processes, organism systems, and neurological human diseases.
Collapse
Affiliation(s)
| | | | - Lívia Soares Zaramela
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Bruno Ruiz Brandão da Costa
- Department of Clinical, Toxicological and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Vanessa Fernandes Rodrigues
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Bruno Spinosa De Martinis
- Department of Chemistry, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Daniela Carlos
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Rita C Tostes
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
22
|
Arakelyan NA, Kupriyanova DA, Vasilevska J, Rogaev EI. Sexual dimorphism in immunity and longevity among the oldest old. Front Immunol 2025; 16:1525948. [PMID: 40034689 PMCID: PMC11872714 DOI: 10.3389/fimmu.2025.1525948] [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: 11/10/2024] [Accepted: 01/28/2025] [Indexed: 03/05/2025] Open
Abstract
Human longevity is a sex-biased process in which sex chromosomes and sex-specific immunity may play a crucial role in the health and lifespan disparities between men and women. Generally, women have a higher life expectancy than men, exhibiting lower infection rates for a broad range of pathogens, which results in a higher prevalence of female centenarians compared to males. Investigation of the immunological changes that occur during the process of healthy aging, while taking into account the differences between sexes, can significantly enhance our understanding of the mechanisms that underlie longevity. In this review, we aim to summarize the current knowledge on sexual dimorphism in the human immune system and gut microbiome during aging, with a particular focus on centenarians, based exclusively on human data.
Collapse
Affiliation(s)
- Nelli A. Arakelyan
- Center for Genetics and Life Science, Sirius University of Science and Technology, Sochi, Russia
| | - Daria A. Kupriyanova
- Center for Genetics and Life Science, Sirius University of Science and Technology, Sochi, Russia
| | - Jelena Vasilevska
- Center for Genetics and Life Science, Sirius University of Science and Technology, Sochi, Russia
| | - Evgeny I. Rogaev
- Center for Genetics and Life Science, Sirius University of Science and Technology, Sochi, Russia
- Department of Psychiatry, University of Massachusetts Medical School, Worcester, MA, United States
| |
Collapse
|
23
|
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
|
24
|
Asri AK, Liu T, Tsai HJ, Wang JY, Wu CD. Environmental exposures related to gut microbiota among children with asthma: a pioneer study in Taiwan. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 291:117793. [PMID: 39892323 DOI: 10.1016/j.ecoenv.2025.117793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 01/01/2025] [Accepted: 01/21/2025] [Indexed: 02/03/2025]
Abstract
Gut microbiota plays a crucial role in human health and can be influenced by environmental factors. While past studies have examined the impact of the environment on gut microbiota, vulnerable populations have often been overlooked. This study aimed to investigate the association between environmental exposures, air pollution and greenspace, and gut microbiota in asthmatic children. Data were collected during the recovery period for 41 eligible children. Air pollution was estimated using an ensemble learning model that combined regression and machine-learning algorithms, while greenspace was quantified using the normalized difference vegetation index (NDVI) and green land-cover data. The lag effects of exposures were assessed within defined buffer zones surrounding each child's residence. A generalized additive model was applied to examine associations. Results revealed a marginally significant negative association between 1-day lag exposure to NO₂ and gut microbiota indices, such as observed bacteria (Coef.: -1.130; 95 %CI -2.287, 0.027) and bacterial richness (Coef.: -2.420; 95 %CI -4.987, 0.146). The 8-day lagged average exposure to PM2.5 and O₃ also showed negative impacts on bacterial diversity. In contrast, the 1-month lagged average exposure to greenspace was positively associated with microbiota indices. Air pollution and greenspace were also linked to specific bacterial abundances, such as Streptococcus. This study underscores the need for further research on how environmental factors may influence immunity in asthmatic children by altering gut microbiota.
Collapse
Affiliation(s)
- Aji Kusumaning Asri
- Department of Geomatics, National Cheng Kung University, Tainan 701, Taiwan, ROC.
| | - Tsunglin Liu
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan 701, Taiwan, ROC.
| | - Hui-Ju Tsai
- Institute of Population Health Sciences, National Health Research Institutes, Miaoli 350, Taiwan, ROC.
| | - Jiu-Yao Wang
- Department of Allergy and Immunology, China Medical University Children's Hospital, Taichung 404, Taiwan, ROC; Allergy, Immunology, and Microbiome (A.I.M.) Research Center, China Medical University, Taichung 404, Taiwan, ROC.
| | - Chih-Da Wu
- Department of Geomatics, National Cheng Kung University, Tainan 701, Taiwan, ROC; National Institute of Environmental Health Sciences, National Health Research Institutes, Miaoli 35053, Taiwan, ROC; Innovation and Development Center of Sustainable Agriculture, National Chung Hsing University, Taichung 402, Taiwan, ROC; Research Center for Precision Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC.
| |
Collapse
|
25
|
Kaltsas A, Giannakodimos I, Markou E, Adamos K, Stavropoulos M, Kratiras Z, Zachariou A, Dimitriadis F, Sofikitis N, Chrisofos M. The Role of Gut Microbiota Dysbiosis in Erectile Dysfunction: From Pathophysiology to Treatment Strategies. Microorganisms 2025; 13:250. [PMID: 40005617 PMCID: PMC11857656 DOI: 10.3390/microorganisms13020250] [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: 12/10/2024] [Revised: 01/16/2025] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
Erectile dysfunction (ED) is a prevalent male sexual disorder characterized by the persistent inability to achieve or maintain an erection sufficient for satisfactory sexual performance. While its etiology is multifactorial, encompassing vascular, neurological, hormonal, and psychological components, emerging evidence suggests a significant role for gut microbiota dysbiosis in its development. The gut microbiota influences various metabolic, inflammatory, and neuropsychological processes critical to erectile function. Dysbiosis can lead to systemic inflammation, endothelial dysfunction, hormonal imbalances, and altered neurotransmitter production, all of which are key factors in ED pathogenesis. This narrative review synthesizes current research on the association between gut microbiota alterations and ED, highlighting specific bacterial taxa implicated in ED through mechanisms involving inflammation, metabolic disturbances, and hormonal regulation. This review explores potential mechanisms linking gut microbiota and ED, including pro-inflammatory cytokines, gut barrier integrity disruption, metabolic disorders, psychological factors via the gut-brain axis, and hormonal regulation. Furthermore, the gut microbiota offers promising avenues for developing non-invasive biomarkers and therapeutic interventions such as probiotics, prebiotics, dietary modifications, and fecal microbiota transplantation. Future research should focus on longitudinal studies, mechanistic explorations, and clinical trials to validate these findings and translate them into clinical practice. Understanding the interplay between the gut microbiota and erectile function could unveil novel diagnostic biomarkers and pave the way for innovative treatments targeting the microbiota, ultimately improving men's sexual and overall health.
Collapse
Affiliation(s)
- Aris Kaltsas
- Third Department of Urology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (A.K.); (I.G.); (K.A.); (M.S.); (Z.K.)
| | - Ilias Giannakodimos
- Third Department of Urology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (A.K.); (I.G.); (K.A.); (M.S.); (Z.K.)
| | - Eleftheria Markou
- Department of Microbiology, University Hospital of Ioannina, 45500 Ioannina, Greece;
| | - Konstantinos Adamos
- Third Department of Urology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (A.K.); (I.G.); (K.A.); (M.S.); (Z.K.)
| | - Marios Stavropoulos
- Third Department of Urology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (A.K.); (I.G.); (K.A.); (M.S.); (Z.K.)
| | - Zisis Kratiras
- Third Department of Urology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (A.K.); (I.G.); (K.A.); (M.S.); (Z.K.)
| | - Athanasios Zachariou
- Laboratory of Spermatology, Department of Urology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (A.Z.); (N.S.)
| | - Fotios Dimitriadis
- Department of Urology, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Nikolaos Sofikitis
- Laboratory of Spermatology, Department of Urology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (A.Z.); (N.S.)
| | - Michael Chrisofos
- Third Department of Urology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (A.K.); (I.G.); (K.A.); (M.S.); (Z.K.)
| |
Collapse
|
26
|
Luna-Guzmán CE, Zarzoza-Mendoza IC, Cervantes-Monroy E, Villa-Morales J, Carmona-Sierra FV, Maldonado-Hernández J, Domínguez-Calderón I, Rodriguez-Cruz M. Composition of linear and branched short-chain fatty acids in human milk and newborn feces: influence of perinatal and maternal factors. Food Funct 2025; 16:499-509. [PMID: 39679783 DOI: 10.1039/d4fo03568b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
This research aimed to analyze the percentage of short-chain fatty acids (SCFAs) in human milk (HM) and newborn feces and to explore potential associations with factors such as maternal nutrition, age, biological sex, delivery mode, diet, and the type of HM. Gas chromatography was used to measure the percentage of SCFAs in colostrum (n = 23), transitional HM (n = 23), and mature HM (n = 92) and feces of newborn (n = 36) at day 30 postpartum. Anthropometry was also evaluated in the mother and the infant. The results showed that acetic acid was the most abundant in HM. The percentage of butyric acid and isovaleric acid was higher (p < 0.05) in the feces of newborns whose mothers were overweight/obese or were male, respectively, compared to newborns whose mothers were of normal weight or were female. The percentage of valeric acid was higher in the feces of newborns whose mothers were over 30 years old and who were delivered by C-section, compared to newborns whose mothers were 30 years old or younger and who were delivered vaginally. Inadequate intake of proteins, carbohydrates, and fiber was associated (p < 0.05) with lower acetic acid and higher butyric acid, higher propionic acid and lower butyric acid, and higher isovaleric acid percentage, respectively, in mature HM. The percentage of acetic acid was higher (p < 0.01) and that of propionic acid, butyric acid, isobutyric acid, and isovaleric acid was lower (p < 0.01) in colostrum compared to mature HM. The intake of lipids was associated with the percentage of butyric acid (β = -0.32, p = 0.01), and the percentage of propionic acid (β = 0.43, p < 0.01) was associated with carbohydrate intake. Overall, this study concluded that factors such as maternal nutritional status, diet, age, biological sex, and delivery mode were related to the composition of specific SCFAs in mature HM and newborn feces. Additionally, the percentage of SCFAs gradually decreased from colostrum to mature HM.
Collapse
Affiliation(s)
- Cristian Emmanuel Luna-Guzmán
- Laboratorio de Nutrición Molecular, Unidad de Investigación Médica en Nutrición, Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México (CDMX), Mexico.
| | - Imelda Cecilia Zarzoza-Mendoza
- Laboratorio de Nutrición Molecular, Unidad de Investigación Médica en Nutrición, Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México (CDMX), Mexico.
- Laboratorio de Biotecnología y Bioinformática Genómica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Emmanuel Cervantes-Monroy
- Laboratorio de Nutrición Molecular, Unidad de Investigación Médica en Nutrición, Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México (CDMX), Mexico.
| | - Judith Villa-Morales
- Laboratorio de Nutrición Molecular, Unidad de Investigación Médica en Nutrición, Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México (CDMX), Mexico.
| | | | - Jorge Maldonado-Hernández
- Laboratorio de Nutrición Molecular, Unidad de Investigación Médica en Nutrición, Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México (CDMX), Mexico.
| | - Israel Domínguez-Calderón
- Laboratorio de Nutrición Molecular, Unidad de Investigación Médica en Nutrición, Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México (CDMX), Mexico.
| | - Maricela Rodriguez-Cruz
- Laboratorio de Nutrición Molecular, Unidad de Investigación Médica en Nutrición, Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México (CDMX), Mexico.
| |
Collapse
|
27
|
Bhusri B, Sutheeworapong S, Kittichotirat W, Kusonmano K, Thammarongtham C, Lertampaiporn S, Prommeenate P, Praphanphoj V, Kittitharaphan W, Dulsawat S, Paenkaew P, Cheevadhanarak S. Characterization of gut microbiota on gender and age groups bias in Thai patients with autism spectrum disorder. Sci Rep 2025; 15:2587. [PMID: 39833480 PMCID: PMC11747245 DOI: 10.1038/s41598-025-86740-2] [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: 08/03/2024] [Accepted: 01/13/2025] [Indexed: 01/22/2025] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by social communication and interaction problems. The prevalence of ASD is increasing globally, with a higher ratio of males to females. Gastrointestinal symptoms are common in individuals with ASD, and gut microbiota has been implicated in the disorder's development. This study aimed to investigate the gut microbiota alteration in Thai individuals with ASD compared to healthy controls using 16S rRNA gene sequencing. The influence of gender and age on gut microbiota composition and function was also examined. A total of 65 ASD individuals and 30 neurotypical (NT) individuals were included in the analysis. The results revealed notable differences in gut microbiota composition between the ASD and NT groups, with variations observed in microbial richness and the presence of enriched microbial taxa. These differences were influenced by both gender and age. Fusobacteriota, Fusobacteriaceae, and Fusobacterium were found to be enriched in individuals with ASD. Furthermore, the study identified gender-related taxa, such as Bacteroides plebeius, enriched in ASD females. Age-related taxa, including Veillonella, known to be associated with poor oral hygiene, were also observed in ASD children. The analysis of differentially abundant pathways highlighted the enrichment of various metabolic pathways in individuals with ASD, including those related to endocrine-disrupting chemicals. These findings underscore the importance of considering gender and age when studying gut microbiota in ASD. They provide valuable insights into the potential role of gut microbiota dysbiosis in ASD pathogenesis and highlight the influence of environmental factors.
Collapse
Affiliation(s)
- Benjaporn Bhusri
- Bioinformatics and Systems Biology Program, School of Bioresources and Technology, and School of Information Technology, King Mongkut's University of Technology Thonburi, Bangkok, 10150, Thailand
| | - Sawannee Sutheeworapong
- Systems Biology and Bioinformatics Unit, Pilot Plant Development and Training Institute, King Mongkut's University of Technology Thonburi, Bangkok, 10150, Thailand
| | - Weerayuth Kittichotirat
- Bioinformatics and Systems Biology Program, School of Bioresources and Technology, and School of Information Technology, King Mongkut's University of Technology Thonburi, Bangkok, 10150, Thailand
- Systems Biology and Bioinformatics Unit, Pilot Plant Development and Training Institute, King Mongkut's University of Technology Thonburi, Bangkok, 10150, Thailand
| | - Kanthida Kusonmano
- Bioinformatics and Systems Biology Program, School of Bioresources and Technology, King Mongkut's University of Technology Thonburi, Bangkok, 10150, Thailand
| | - Chinae Thammarongtham
- Biochemical Engineering and Systems Biology Research Group, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency at King Mongkut's University of Technology Thonburi, Bangkok, 10150, Thailand
| | - Supatcha Lertampaiporn
- Biochemical Engineering and Systems Biology Research Group, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency at King Mongkut's University of Technology Thonburi, Bangkok, 10150, Thailand
| | - Peerada Prommeenate
- Biochemical Engineering and Systems Biology Research Group, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency at King Mongkut's University of Technology Thonburi, Bangkok, 10150, Thailand
| | | | - Wiranpat Kittitharaphan
- Yuwaprasart Waithayopathum Child and Adolescent Psychiatric Hospital, Ministry of Public Health, Samut Prakan, 10270, Thailand
| | - Sudarat Dulsawat
- Fungal Biotechnology Unit, Pilot Plant Development and Training Institute, King Mongkut's University of Technology Thonburi, Bangkok, 10150, Thailand
| | - Prasobsook Paenkaew
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Supapon Cheevadhanarak
- Systems Biology and Bioinformatics Unit, Pilot Plant Development and Training Institute, King Mongkut's University of Technology Thonburi, Bangkok, 10150, Thailand.
- Fungal Biotechnology Unit, Pilot Plant Development and Training Institute, King Mongkut's University of Technology Thonburi, Bangkok, 10150, Thailand.
- School of Bioresources and Technology, King Mongkut's University of Technology Thonburi, Bangkok, 10150, Thailand.
| |
Collapse
|
28
|
Levine BH, Hoffman JM. Microbiome transplants may not improve health and longevity in Drosophila melanogaster. Biol Open 2025; 14:bio061745. [PMID: 39835966 PMCID: PMC11789278 DOI: 10.1242/bio.061745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 11/25/2024] [Indexed: 01/22/2025] Open
Abstract
The gut microbiome, which is composed of bacteria, viruses, and fungi, and is involved in multiple essential physiological processes, changes measurably as a person ages, and can be associated with negative health outcomes. Microbiome transplants have been proposed as a method to improve gut function and reduce or reverse multiple disorders, including age-related diseases. Here, we take advantage of the laboratory model organism, Drosophila melanogaster, to test the effects of transplanting the microbiome of a young fly into middle-aged flies, across multiple genetic backgrounds and both sexes, to test whether age-related lifespan could be increased, and late-life physical health declines mitigated. Our results suggest that, overall, microbiome transplants do not improve longevity and may even be detrimental in flies, and the health effects of microbiome transplants were minor, but sex- and genotype-dependent. This discovery supports previous evidence that axenic flies, those with no gut microbiome, live healthier and longer lives than their non-axenic counterparts. The results of this study suggest that, at least for fruit flies, microbiome transplants may not be a viable intervention to improve health and longevity, though more research is still warranted.
Collapse
Affiliation(s)
- Benjamin H. Levine
- Department of Biological Sciences, Augusta University, Augusta, GA 30912, USA
| | - Jessica M. Hoffman
- Department of Biological Sciences, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
29
|
Perez V, Boulo V, De Lorgeril J, Pham D, Ansquer D, Plougoulen G, Ballan V, Lam JS, Romatif O, Le Luyer J, Falchetto C, Basset C, Flohr S, Maamaatuaiahutapu M, Lafille MA, Lau C, Saulnier D, Wabete N, Callac N. Hemolymph microbiota and immune effectors' expressions driven by geographical rearing acclimation of the aquacultured Penaeus stylirostris. Anim Microbiome 2025; 7:5. [PMID: 39799372 PMCID: PMC11725212 DOI: 10.1186/s42523-025-00376-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 01/02/2025] [Indexed: 01/15/2025] Open
Abstract
BACKGROUND In holobiont, microbiota is known to play a central role on the health and immunity of its host. Then, understanding the microbiota, its dynamic according to the environmental conditions and its link to the immunity would help to react to potential dysbiosis of aquacultured species. While the gut microbiota is highly studied, in marine invertebrates the hemolymph microbiota is often set aside even if it remains an important actor of the hemolymph homeostasis. Indeed, the hemolymph harbors the factors involved in the animal homeostasis that interacts with the microbiota, the immunity. In the Southwest Pacific, the high economical valued shrimp Penaeus stylirostris is reared in two contrasted sites, in New Caledonia (NC) and in French Polynesia (FP). RESULTS We characterized the active microbiota inhabiting the hemolymph of shrimps while considering its stability during two seasons and at a one-month interval and evidenced an important microbial variability between the shrimps according to the rearing conditions and the sites. We highlighted specific biomarkers along with a common core microbiota composed of 6 ASVs. Putative microbial functions were mostly associated with bacterial competition, infections and metabolism in NC, while they were highly associated with the cell metabolism in FP suggesting a rearing site discrimination. Differential relative expression of immune effectors measured in the hemolymph of two shrimp populations from NC and FP, exhibited higher level of expression in NC compared to FP. In addition, differential relative expression of immune effectors was correlated to bacterial biomarkers based on their geographical location. CONCLUSIONS Our data suggest that, in Pacific shrimps, both the microbiota and the expression of the immune effectors could have undergone differential immunostimulation according to the rearing site as well as a geographical adaptative divergence of the shrimps as an holobiont, to their rearing sites. Further, the identification of proxies such as the core microbiota and site biomarkers, could be used to guide future actions to monitor the bacterial microbiota and thus preserve the productions.
Collapse
Affiliation(s)
- Valérie Perez
- Ifremer, IRD, Université de la Nouvelle-Calédonie, Université de La Réunion, CNRS, UMR 9220 ENTROPIE, Nouméa, 98800, New Caledonia
- Ifremer, MASAE Microbiologie Aliment Santé Environnement, Nantes, 44000, France
| | - Viviane Boulo
- Ifremer, IRD, Université de la Nouvelle-Calédonie, Université de La Réunion, CNRS, UMR 9220 ENTROPIE, Nouméa, 98800, New Caledonia
- Interactions Hôtes Pathogènes Environnements (IHPE), Université de Montpellier, CNRS, Ifremer, Université de Perpignan Via Domitia, Montpellier, 34090, France
| | - Julien De Lorgeril
- Ifremer, IRD, Université de la Nouvelle-Calédonie, Université de La Réunion, CNRS, UMR 9220 ENTROPIE, Nouméa, 98800, New Caledonia
| | - Dominique Pham
- Ifremer, IRD, Université de la Nouvelle-Calédonie, Université de La Réunion, CNRS, UMR 9220 ENTROPIE, Nouméa, 98800, New Caledonia
| | - Dominique Ansquer
- Ifremer, IRD, Université de la Nouvelle-Calédonie, Université de La Réunion, CNRS, UMR 9220 ENTROPIE, Nouméa, 98800, New Caledonia
| | - Gwenola Plougoulen
- Ifremer, IRD, Université de la Nouvelle-Calédonie, Université de La Réunion, CNRS, UMR 9220 ENTROPIE, Nouméa, 98800, New Caledonia
| | - Valentine Ballan
- Ifremer, IRD, Université de la Nouvelle-Calédonie, Université de La Réunion, CNRS, UMR 9220 ENTROPIE, Nouméa, 98800, New Caledonia
| | - Jean-Sébastien Lam
- Ifremer, IRD, Université de la Nouvelle-Calédonie, Université de La Réunion, CNRS, UMR 9220 ENTROPIE, Nouméa, 98800, New Caledonia
| | - Océane Romatif
- Interactions Hôtes Pathogènes Environnements (IHPE), Université de Montpellier, CNRS, Ifremer, Université de Perpignan Via Domitia, Montpellier, 34090, France
| | - Jeremy Le Luyer
- Ifremer, Université de la Polynésie Française, ILM, IRD, UMR 241 SECOPOL, Tahiti, French Polynesia, F-98719, France
| | - Corinne Falchetto
- Ifremer, Université de la Polynésie Française, ILM, IRD, UMR 241 SECOPOL, Tahiti, French Polynesia, F-98719, France
| | - Caline Basset
- Ifremer, Université de la Polynésie Française, ILM, IRD, UMR 241 SECOPOL, Tahiti, French Polynesia, F-98719, France
| | - Stanley Flohr
- DRM, Direction des Ressources Marines, Papeete, 98713, French Polynesia
| | | | | | - Christophe Lau
- DRM, Direction des Ressources Marines, Papeete, 98713, French Polynesia
| | - Denis Saulnier
- Ifremer, Université de la Polynésie Française, ILM, IRD, UMR 241 SECOPOL, Tahiti, French Polynesia, F-98719, France
| | - Nelly Wabete
- Ifremer, IRD, Université de la Nouvelle-Calédonie, Université de La Réunion, CNRS, UMR 9220 ENTROPIE, Nouméa, 98800, New Caledonia
| | - Nolwenn Callac
- Ifremer, IRD, Université de la Nouvelle-Calédonie, Université de La Réunion, CNRS, UMR 9220 ENTROPIE, Nouméa, 98800, New Caledonia.
| |
Collapse
|
30
|
Leao L, Miri S, Hammami R. Gut feeling: Exploring the intertwined trilateral nexus of gut microbiota, sex hormones, and mental health. Front Neuroendocrinol 2025; 76:101173. [PMID: 39710079 DOI: 10.1016/j.yfrne.2024.101173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 11/25/2024] [Accepted: 12/18/2024] [Indexed: 12/24/2024]
Abstract
The complex interplay between the gut microbiota, sex hormones, and mental health is emerging as a pivotal factor in understanding and managing psychiatric disorders. Beyond their traditional roles, sex hormones exert profound effects on various physiological systems including the gut microbiota. Fluctuations in sex hormone levels, notably during the menstrual cycle, influence gut physiology and barrier function, shaping gut microbiota composition and immune responses. Conversely, the gut microbiota actively modulates sex hormone levels via enzymatic processes. This bidirectional relationship underscores the significance of the gut-brain axis in maintaining mental well-being. This review explores the multifaceted interactions between sex hormones, the gut microbiota, and mental health outcomes. We highlight the potential of personalized interventions in treating psychiatric disorders, particularly in vulnerable populations such as premenopausal women and individuals with depressive disorders. By elucidating these complex interactions, we aim to provide insights for future research into targeted interventions, enhancing mental health outcomes.
Collapse
Affiliation(s)
- Luana Leao
- NuGut Research Platform, School of Nutrition Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Saba Miri
- NuGut Research Platform, School of Nutrition Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Riadh Hammami
- NuGut Research Platform, School of Nutrition Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada; Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada.
| |
Collapse
|
31
|
Mak PHW, Yin X, Clairmont L, Bean-Hodgins L, Kiarie EG, Tang J, Lepp D, Diarra MS. Cecal microbiome in broiler chicken related to antimicrobial feeding and bird's sex. Can J Microbiol 2025; 71:1-19. [PMID: 39993268 DOI: 10.1139/cjm-2024-0190] [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: 02/26/2025]
Abstract
This study investigated the cecal microbiome of broilers raised under specific antimicrobial feeding programs (AFPs). A total of 2304 day-old Ross-708 male (M, n = 1152) and female (F, n = 1152) chicks were distributed into 48 floor pens which were allocated to one of three AFPs: Conventional, raised without medically important antibiotics (RWMIA), and raised without antibiotics (RWA). At 28 (D28) and 41 (D41) days of age, cecal contents were collected for culture dependent and independent analyses. At both 28 and 41 days, Enterococcus was more abundant in RWA-raised broilers than other groups with the most abundance of this bacterium being found in female birds (P < 0.05). At D41, the most abundant Eimeria tenella counts was observed in RWA-raised broiler ceca (P < 0.05). Sex effects were observed on the abundances of four of the 248 identified antimicrobial resistance genes while abundances of 10 were modulated by AFPs (P < 0.05). Ceca of females birds showed more tssB than males, and ceca of RWMIA-raised birds contained the highest abundance of chuY genes regardless of sex. This study showed that in a specific feeding program, cecal resistome can be affected by chicken's sex contributing to understand the AMR related to the AMU.
Collapse
Affiliation(s)
- Philip H W Mak
- Guelph Research and Development Centre, Agriculture and Agri-Food Canada (AAFC), Guelph, ON, Canada
- Department of Animal Biosciences, University of Guelph, Guelph, ON, Canada
| | - Xianhua Yin
- Guelph Research and Development Centre, Agriculture and Agri-Food Canada (AAFC), Guelph, ON, Canada
| | - Lindsey Clairmont
- Guelph Research and Development Centre, Agriculture and Agri-Food Canada (AAFC), Guelph, ON, Canada
| | - Lisa Bean-Hodgins
- Department of Animal Biosciences, University of Guelph, Guelph, ON, Canada
| | - Elijah G Kiarie
- Department of Animal Biosciences, University of Guelph, Guelph, ON, Canada
| | - Joshua Tang
- Guelph Research and Development Centre, Agriculture and Agri-Food Canada (AAFC), Guelph, ON, Canada
| | - Dion Lepp
- Guelph Research and Development Centre, Agriculture and Agri-Food Canada (AAFC), Guelph, ON, Canada
| | - Moussa S Diarra
- Guelph Research and Development Centre, Agriculture and Agri-Food Canada (AAFC), Guelph, ON, Canada
| |
Collapse
|
32
|
Salia S, Burke FF, Hinks ME, Randell AM, Matheson MA, Walling SG, Swift-Gallant A. Gut microbiota transfer from the preclinical maternal immune activation model of autism is sufficient to induce sex-specific alterations in immune response and behavioural outcomes. Brain Behav Immun 2025; 123:813-823. [PMID: 39471905 DOI: 10.1016/j.bbi.2024.10.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/22/2024] [Accepted: 10/26/2024] [Indexed: 11/01/2024] Open
Abstract
The gut microbiome plays a vital role in health and disease, including neurodevelopmental disorders like autism spectrum disorder (ASD). ASD affects 4:1 males-to-females, and sex differences are apparent in gut microbiota composition among ASD individuals and in animal models of this condition, such as the maternal immune activation (MIA) mouse model. However, few studies have included sex as a biological variable when assessing the role of gut microbiota in mediating ASD symptoms. Using the MIA model of ASD, we assessed whether gut microbiota contributes to the sex differences in the presentation of ASD-like behaviors. Gut microbiota transplantation from MIA or vehicle/control male and female mice into healthy, otherwise unmanipulated, 4-week-old C57Bl/6 mice was performed for 6 treatments over 12 days. Colonization with male, but not female, MIA microbiota was sufficient to reduce sociability, decrease microbiota diversity and increase neuroinflammation with more pronounced deficits in male recipients. Colonization with both male and female donor microbiota altered juvenile ultrasonic vocalizations and anxiety-like behavior in recipients of both sexes, and there was an accompanied change in the gut microbiota and serum cytokine IL-4 and IL-7 levels of all recipients of MIA gut microbiota. In addition to the increases in gut microbes associated with pathological states, the female donor microbiota profile also had increases in gut microbes with known neural protective effects (e.g., Lactobacillus and Rikenella). These results suggest that gut reactivity to environmental insults, such as in the MIA model, may play a role in shaping the sex disparity in ASD development.
Collapse
Affiliation(s)
- Stephanie Salia
- Department of Psychology, Memorial University of Newfoundland, 232 Elizabeth Avenue, St. John's, NL A1B 3X9, Canada.
| | - Francine F Burke
- Department of Psychology, Memorial University of Newfoundland, 232 Elizabeth Avenue, St. John's, NL A1B 3X9, Canada
| | - Meagan E Hinks
- Department of Psychology, Memorial University of Newfoundland, 232 Elizabeth Avenue, St. John's, NL A1B 3X9, Canada
| | - Alison M Randell
- Department of Psychology, Memorial University of Newfoundland, 232 Elizabeth Avenue, St. John's, NL A1B 3X9, Canada
| | - Mairead Anna Matheson
- Department of Psychology, Memorial University of Newfoundland, 232 Elizabeth Avenue, St. John's, NL A1B 3X9, Canada
| | - Susan G Walling
- Department of Psychology, Memorial University of Newfoundland, 232 Elizabeth Avenue, St. John's, NL A1B 3X9, Canada
| | - Ashlyn Swift-Gallant
- Department of Psychology, Memorial University of Newfoundland, 232 Elizabeth Avenue, St. John's, NL A1B 3X9, Canada.
| |
Collapse
|
33
|
Lattanzi G, Perillo F, Díaz-Basabe A, Caridi B, Amoroso C, Baeri A, Cirrincione E, Ghidini M, Galassi B, Cassinotti E, Baldari L, Boni L, Vecchi M, Caprioli F, Facciotti F, Strati F. Estrogen-related differences in antitumor immunity and gut microbiome contribute to sexual dimorphism of colorectal cancer. Oncoimmunology 2024; 13:2425125. [PMID: 39548749 PMCID: PMC11572150 DOI: 10.1080/2162402x.2024.2425125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 10/18/2024] [Accepted: 10/30/2024] [Indexed: 11/18/2024] Open
Abstract
Colorectal cancer (CRC) is a multifaceted disease whose development and progression varies depending on tumor location, age of patients, infiltration of immune cells within cancer lesions, and the tumor microenvironment. These pathophysiological characteristics are additionally influenced by sex-related differences. The gut microbiome plays a role in initiation and progression of CRC, and shapes anti-tumor immune responses but how responsiveness of the immune system to the intestinal microbiota may contribute to sexual dimorphism of CRC is largely unknown. We studied survival, tumor-infiltrating immune cell populations and tumor-associated microbiome of a cohort of n = 184 male and female CRC patients through high-dimensional single-cell flow cytometry and 16S rRNA gene sequencing. We functionally tested the immune system-microbiome interactions in in-vivo and in-vitro models of the disease. High-dimensional single-cell flow cytometry showed that female patients are enriched by tumor-infiltrating invariant Natural Killer T (iNKT) cells but depleted by cytotoxic T lymphocytes. The enrichment of oral pathobionts and a reduction of β-glucuronidase activity are distinctive traits characterizing the gut microbiome of female patients affected by CRC. Functional assays using a collection of human primary iNKT cell lines demonstrated that the gut microbiota of female patients functionally impairs iNKT cell anti-tumor functions interfering with the granzyme-perforin cytotoxic pathway. Our results highlight a sex-dependent functional relationship between the gut microbiome, estrogen metabolism, and the decline of cytotoxic T cell responses, contributing to the sexual dimorphism observed in CRC patients with relevant implications for precision medicine and the design of targeted therapeutic approaches addressing sex bias in cancer.
Collapse
Affiliation(s)
- Georgia Lattanzi
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milan, Italy
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Federica Perillo
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Angélica Díaz-Basabe
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Bruna Caridi
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Chiara Amoroso
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Alberto Baeri
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Elisa Cirrincione
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Michele Ghidini
- Medical Oncology, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Barbara Galassi
- Medical Oncology, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Elisa Cassinotti
- Department of General & Minimally Invasive Surgery, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Ludovica Baldari
- Department of General & Minimally Invasive Surgery, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Luigi Boni
- Department of General & Minimally Invasive Surgery, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Maurizio Vecchi
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Flavio Caprioli
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Federica Facciotti
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Francesco Strati
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| |
Collapse
|
34
|
Salvado R, Lugones-Sánchez C, Santos-Minguez S, González-Sánchez S, Quesada JA, Benito R, Rodríguez-Sánchez E, Gómez-Marcos MA, Guimarães-Cunha P, Hernandez-Rivas JM, Mira A, García-Ortiz L, MIVAS Investigators. Sex Differences in Gut Microbiota and Their Relation to Arterial Stiffness (MIVAS Study). Nutrients 2024; 17:53. [PMID: 39796488 PMCID: PMC11723250 DOI: 10.3390/nu17010053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 12/18/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND Recent research highlights the potential role of sex-specific variations in cardiovascular disease. The gut microbiome has been shown to differ between the sexes in patients with cardiovascular risk factors. OBJECTIVES The main objective of this study is to analyze the differences between women and men in the relationship between gut microbiota and measures of arterial stiffness. METHODS We conducted a cross-sectional study in Spain, selecting 180 subjects (122 women, 58 men) aged between 45 and 74. Subjects with arterial stiffness were identified by the presence of at least one of the following: carotid-femoral pulse wave velocity (cf-PWV) above 12 mm/s, cardio-ankle vascular index (CAVI) above nine, or brachial-ankle pulse wave velocity (ba-PWV) above 17.5 m/s. All other cases were considered subjects without arterial stiffness. The composition of the gut microbiome in fecal samples was determined by 16S rRNA sequencing. RESULTS We found that women have a more diverse microbiome than men (Shannon, p < 0.05). There is also a significant difference in gut microbiota composition between sexes (Bray-Curtis, p < 0.01). Dorea, Roseburia, and Agathobacter, all of them short-chain fatty-acid producers, were more abundant in women's microbiota (log values > 1, p-value and FDR < 0.05). Additionally, Blautia was more abundant in women when only the subjects with arterial stiffness were considered. According to logistic regression, Roseburia was negatively associated with arterial stiffness in men, while Bifidobacterium and Subdoligranulum were positively related to arterial stiffness. CONCLUSIONS In the Spanish population under study, women had higher microbiome diversity and potentially protective genera. The host's gender determines the influence of the same bacteria on arterial stiffness. TRIAL REGISTRATION NUMBER NCT03900338.
Collapse
Affiliation(s)
- Rita Salvado
- Primary Care Research Unit of Salamanca (APISAL), Salamanca Primary Healthcare Management, Castilla y León Regional Health Authority (SACyL), Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (C.L.-S.); (S.G.-S.); (M.A.G.-M.); (L.G.-O.)
| | - Cristina Lugones-Sánchez
- Primary Care Research Unit of Salamanca (APISAL), Salamanca Primary Healthcare Management, Castilla y León Regional Health Authority (SACyL), Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (C.L.-S.); (S.G.-S.); (M.A.G.-M.); (L.G.-O.)
- Research Network on Chronicity, Primary Care and Health Promotion (RICAPPS), 37005 Salamanca, Spain
| | - Sandra Santos-Minguez
- Cancer Research Centre, Institute of Molecular and Cellular Biology of Cancer (IBMCC), Institute of Biomedical Research of Salamanca (IBSAL), University of Salamanca—CSIC, 37007 Salamanca, Spain; (S.S.-M.); (J.M.H.-R.)
| | - Susana González-Sánchez
- Primary Care Research Unit of Salamanca (APISAL), Salamanca Primary Healthcare Management, Castilla y León Regional Health Authority (SACyL), Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (C.L.-S.); (S.G.-S.); (M.A.G.-M.); (L.G.-O.)
- Research Network on Chronicity, Primary Care and Health Promotion (RICAPPS), 37005 Salamanca, Spain
| | - José A. Quesada
- Clinical Medicine Department, Miguel Hernandez University, 03550 Alicante, Spain;
- Network of Research on Chronicity, Primary Care and Health Promotion (RICAPPS), 03550 Alicante, Spain
| | - Rocío Benito
- Cancer Research Centre, Institute of Molecular and Cellular Biology of Cancer (IBMCC), Institute of Biomedical Research of Salamanca (IBSAL), University of Salamanca—CSIC, 37007 Salamanca, Spain; (S.S.-M.); (J.M.H.-R.)
| | - Emiliano Rodríguez-Sánchez
- Primary Care Research Unit of Salamanca (APISAL), Salamanca Primary Healthcare Management, Castilla y León Regional Health Authority (SACyL), Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (C.L.-S.); (S.G.-S.); (M.A.G.-M.); (L.G.-O.)
- Research Network on Chronicity, Primary Care and Health Promotion (RICAPPS), 37005 Salamanca, Spain
- Department of Medicine, University of Salamanca (USAL), 37007 Salamanca, Spain
| | - Manuel A. Gómez-Marcos
- Primary Care Research Unit of Salamanca (APISAL), Salamanca Primary Healthcare Management, Castilla y León Regional Health Authority (SACyL), Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (C.L.-S.); (S.G.-S.); (M.A.G.-M.); (L.G.-O.)
- Research Network on Chronicity, Primary Care and Health Promotion (RICAPPS), 37005 Salamanca, Spain
- Department of Medicine, University of Salamanca (USAL), 37007 Salamanca, Spain
| | - Pedro Guimarães-Cunha
- Unidade Local de Saúde do Alto Ave, Center for the Research and Treatment of Arterial Hypertension and Cardiovascular Risk, 4835-044 Guimarâes, Portugal
- Life and Health Sciences Research Institute (IICVS), School of Medicine, Minho University, 4704-553 Braga, Portugal
| | - Jesús M. Hernandez-Rivas
- Cancer Research Centre, Institute of Molecular and Cellular Biology of Cancer (IBMCC), Institute of Biomedical Research of Salamanca (IBSAL), University of Salamanca—CSIC, 37007 Salamanca, Spain; (S.S.-M.); (J.M.H.-R.)
- Department of Medicine, University of Salamanca (USAL), 37007 Salamanca, Spain
- Haematology Department, Institute of Biomedical Research of Salamanca (IBSAL), University Hospital of Salamanca (USAL), 37007 Salamanca, Spain
| | - Alex Mira
- Department of Health and Genomics, FISABIO Foundation, 46020 Valencia, Spain;
- CIBER Center for Epidemiology and Public Health, 28029 Madrid, Spain
| | - Luis García-Ortiz
- Primary Care Research Unit of Salamanca (APISAL), Salamanca Primary Healthcare Management, Castilla y León Regional Health Authority (SACyL), Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (C.L.-S.); (S.G.-S.); (M.A.G.-M.); (L.G.-O.)
- Research Network on Chronicity, Primary Care and Health Promotion (RICAPPS), 37005 Salamanca, Spain
- Department of Biomedical and Diagnostic Sciences, University of Salamanca, 37007 Salamanca, Spain
| | - MIVAS Investigators
- Research Network on Chronicity, Primary Care and Health Promotion (RICAPPS), 37005 Salamanca, Spain
| |
Collapse
|
35
|
Vriend EMC, Galenkamp H, Herrema H, Nieuwdorp M, van den Born BJH, Verhaar BJH. Machine learning analysis of sex and menopausal differences in the gut microbiome in the HELIUS study. NPJ Biofilms Microbiomes 2024; 10:152. [PMID: 39702650 DOI: 10.1038/s41522-024-00628-z] [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: 10/09/2024] [Accepted: 12/03/2024] [Indexed: 12/21/2024] Open
Abstract
Sex differences in the gut microbiome have been examined previously, but results are inconsistent, often due to small sample sizes. We investigated sex and menopausal differences in the gut microbiome in a large multi-ethnic population cohort study, including 5166 participants. Using machine learning models, we revealed modest associations between sex and menopausal status, and gut microbiota composition (AUC 0.61-0.63). After adjustments for age, cardiovascular risk factors, and diet, a part of the associations of the highest-ranked gut microbes with sex were attenuated, but most associations remained significant. In contrast, most associations with menopausal status were driven by age and lost significance after adjustment. Using pathway analyses on metagenomic data, we identified sex differences in vitamin B6 synthesis and stachyose degradation pathways. Since some of sex differences in gut microbiome composition and function could not be explained by covariates, we recommend sex stratification in future microbiome studies.
Collapse
Affiliation(s)
- Esther M C Vriend
- Amsterdam UMC, University of Amsterdam, Department of Internal Medicine, Section Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
- Amsterdam UMC, University of Amsterdam, Department of Public and Occupational Health, Amsterdam Public Health Research institute, Amsterdam, The Netherlands
| | - Henrike Galenkamp
- Amsterdam UMC, University of Amsterdam, Department of Public and Occupational Health, Amsterdam Public Health Research institute, Amsterdam, The Netherlands
| | - Hilde Herrema
- Department of Experimental Vascular Medicine, Amsterdam UMC, Amsterdam, The Netherlands
| | - Max Nieuwdorp
- Amsterdam UMC, University of Amsterdam, Department of Internal Medicine, Section Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Bert-Jan H van den Born
- Amsterdam UMC, University of Amsterdam, Department of Internal Medicine, Section Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
- Amsterdam UMC, University of Amsterdam, Department of Public and Occupational Health, Amsterdam Public Health Research institute, Amsterdam, The Netherlands
| | - Barbara J H Verhaar
- Amsterdam UMC, University of Amsterdam, Department of Internal Medicine, Section Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands.
- Amsterdam UMC, University of Amsterdam, Department of Public and Occupational Health, Amsterdam Public Health Research institute, Amsterdam, The Netherlands.
| |
Collapse
|
36
|
Girdhar K, Mine K, DaCosta JM, Atkinson MA, Ludvigsson J, Altindis E. Sex-specific cytokine, chemokine, and growth factor signatures in T1D patients and progressors. FASEB J 2024; 38:e70270. [PMID: 39704278 DOI: 10.1096/fj.202402354r] [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: 10/02/2024] [Revised: 11/26/2024] [Accepted: 12/11/2024] [Indexed: 12/21/2024]
Abstract
Numerous studies have reported altered cytokine levels in type 1 diabetes (T1D) patients, yet findings remain inconsistent. In this pilot study, we tested the hypothesis that circulating immune markers exhibit sex-based differences in T1D, both prior to and after disease onset. We analyzed 47-48 cytokine, chemokine, and growth factor levels in two cohorts. To assess post-disease differences, we analyzed serum samples from 25 controls and 25 T1D patients. To examine pre-disease progression, we utilized samples from 21 control children and 16 T1D progressors, collected at age 5 years before disease onset. Across all T1D patients and controls, only macrophage colony-stimulating factor and interleukin (IL)-6 showed significant differences. However, we identified notable alterations when comparing sex-age-matched controls and T1D samples. Female T1D patients exhibited lower levels of inflammatory cytokines (tumor necrosis factor-α, IL-6, IL-1a), Th2 cytokines (IL-4, IL-13), and chemokines (macrophage inflammatory protein (MIP)-1α, regulated upon activation, normal T cell expressed and secreted, MIP-3) compared to female controls, differences that were not observed in males. Notably, IL-22 was lower in female T1D patients compared to female controls, whereas it was higher in male T1D patients compared to male controls. Male T1D patients showed elevated levels of growth factors (epidermal growth factor, platelet-derived growth factor-AB/BB) compared to male controls. In T1D progressors, growth-regulated alpha was lower compared to controls in both sexes. Multiple regression analysis further revealed associations between cytokine levels and factors such as age, BMI, and breastfeeding duration. Overall, our findings serve as a proof of concept, highlighting the importance of sex-specific differences in T1D pathogenesis. However, follow-up studies with larger sample sizes are needed to validate and generalize these results.
Collapse
Affiliation(s)
- Khyati Girdhar
- Biology Department, Boston College, Chestnut Hill, Massachusetts, USA
| | - Keiichiro Mine
- Biology Department, Boston College, Chestnut Hill, Massachusetts, USA
| | - Jeffrey M DaCosta
- Biology Department, Boston College, Chestnut Hill, Massachusetts, USA
| | - Mark A Atkinson
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, Florida, USA
- Department of Pediatrics, Diabetes Institute, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Johnny Ludvigsson
- Division of Pediatrics, Department of Biomedical and Clinical Sciences, Crown Princess Victoria Children's Hospital, Linköping University, Linköping, Sweden
| | - Emrah Altindis
- Biology Department, Boston College, Chestnut Hill, Massachusetts, USA
| |
Collapse
|
37
|
Monshizadeh M, Hong Y, Ye Y. Multitask knowledge-primed neural network for predicting missing metadata and host phenotype based on human microbiome. BIOINFORMATICS ADVANCES 2024; 5:vbae203. [PMID: 39735577 PMCID: PMC11676323 DOI: 10.1093/bioadv/vbae203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/27/2024] [Accepted: 12/11/2024] [Indexed: 12/31/2024]
Abstract
Motivation Microbial signatures in the human microbiome are closely associated with various human diseases, driving the development of machine learning models for microbiome-based disease prediction. Despite progress, challenges remain in enhancing prediction accuracy, generalizability, and interpretability. Confounding factors, such as host's gender, age, and body mass index, significantly influence the human microbiome, complicating microbiome-based predictions. Results To address these challenges, we developed MicroKPNN-MT, a unified model for predicting human phenotype based on microbiome data, as well as additional metadata like age and gender. This model builds on our earlier MicroKPNN framework, which incorporates prior knowledge of microbial species into neural networks to enhance prediction accuracy and interpretability. In MicroKPNN-MT, metadata, when available, serves as additional input features for prediction. Otherwise, the model predicts metadata from microbiome data using additional decoders. We applied MicroKPNN-MT to microbiome data collected in mBodyMap, covering healthy individuals and 25 different diseases, and demonstrated its potential as a predictive tool for multiple diseases, which at the same time provided predictions for the missing metadata. Our results showed that incorporating real or predicted metadata helped improve the accuracy of disease predictions, and more importantly, helped improve the generalizability of the predictive models. Availability and implementation https://github.com/mgtools/MicroKPNN-MT.
Collapse
Affiliation(s)
- Mahsa Monshizadeh
- Computer Science Department, Indiana University, Bloomington, IN 47408, United States
| | - Yuhui Hong
- Computer Science Department, Indiana University, Bloomington, IN 47408, United States
| | - Yuzhen Ye
- Computer Science Department, Indiana University, Bloomington, IN 47408, United States
| |
Collapse
|
38
|
Muruganandam A, Migliorini F, Jeyaraman N, Vaishya R, Balaji S, Ramasubramanian S, Maffulli N, Jeyaraman M. Molecular Mimicry Between Gut Microbiome and Rheumatoid Arthritis: Current Concepts. Med Sci (Basel) 2024; 12:72. [PMID: 39728421 PMCID: PMC11677576 DOI: 10.3390/medsci12040072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 12/02/2024] [Accepted: 12/09/2024] [Indexed: 12/28/2024] Open
Abstract
Rheumatoid arthritis (RA) represents an autoimmune condition impacted by a combination of genetic and environmental factors, with the gut microbiome (GMB) being one of the influential environmental factors. Patients with RA display notable modifications in the composition of their GMB, characterised by decreased diversity and distinct bacterial alterations. The GMB, comprising an extensive array of approximately 35,000 bacterial species residing within the gastrointestinal tract, has garnered considerable attention as a pivotal contributor to both human health and the pathogenesis of diseases. This article provides an in-depth exploration of the intricate involvement of the GMB in the context of RA. The oral-GMB axis highlights the complex role of bacteria in RA pathogenesis by producing antibodies to citrullinated proteins (ACPAs) through molecular mimicry. Dysbiosis affects Tregs, cytokine levels, and RA disease activity, suggesting that regulating cytokines could be a strategy for managing inflammation in RA. The GMB also has significant implications for drug responses and toxicity, giving rise to the field of pharmacomicrobiomics. The composition of the microbiota can impact the efficacy and toxicity of drugs, while the microbiota's metabolites can influence drug response. Recent research has identified specific bacteria, metabolites, and immune responses associated with RA, offering potential targets for personalised management. However, several challenges, including the variation in microbial composition, establishing causality, accounting for confounding factors, and translating findings into clinical practice, need to be addressed. Microbiome-targeted therapy is still in its early stages and requires further research and standardisation for effective implementation.
Collapse
Affiliation(s)
- Anandanarayan Muruganandam
- Department of Orthopaedics, Faculty of Medicine—Sri Lalithambigai Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai 600095, India;
| | - Filippo Migliorini
- Department of Orthopedics and Trauma Surgery, Academic Hospital of Bolzano (SABES-ASDAA), 39100 Bolzano, Italy
- Department of Life Sciences, Health, and Health Professions, Link Campus University, 00165 Rome, Italy
| | - Naveen Jeyaraman
- Department of Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai 600077, India;
| | - Raju Vaishya
- Department of Orthopaedics and Joint Replacement Surgery, Indraprastha Apollo Hospital, New Delhi 110076, India;
| | - Sangeetha Balaji
- Department of Orthopaedics, Government Medical College, Omandurar Government Estate, Chennai 600002, India; (S.B.); (S.R.)
| | - Swaminathan Ramasubramanian
- Department of Orthopaedics, Government Medical College, Omandurar Government Estate, Chennai 600002, India; (S.B.); (S.R.)
| | - Nicola Maffulli
- Department of Trauma and Orthopaedic Surgery, Faculty of Medicine and Psychology, University La Sapienza, 00185 Roma, Italy;
- School of Pharmacy and Bioengineering, Keele University Faculty of Medicine, Stoke on Trent ST4 7QB, UK
- Centre for Sports and Exercise Medicine, Barts and the London School of Medicine and Dentistry, Mile End Hospital, Queen Mary University of London, London E1 4DG, UK
| | - Madhan Jeyaraman
- Department of Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai 600077, India;
| |
Collapse
|
39
|
Bardhan P, Mei X, Lai NK, Mell B, Tummala R, Aryal S, Manandhar I, Hwang H, Jhuma TA, Atluri RR, Kyoung J, Li Y, Joe B, Li HB, Yang T. Salt-Responsive Gut Microbiota Induces Sex-Specific Blood Pressure Changes. Circ Res 2024; 135:1122-1137. [PMID: 39440438 PMCID: PMC11905770 DOI: 10.1161/circresaha.124.325056] [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: 06/10/2024] [Revised: 10/02/2024] [Accepted: 10/11/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND Tryptophan metabolism is important in blood pressure regulation. The tryptophan-indole pathway is exclusively mediated by the gut microbiota. ACE2 (angiotensin-converting enzyme 2) participates in tryptophan absorption, and a lack of ACE2 leads to changes in the gut microbiota. The gut microbiota has been recognized as a regulator of blood pressure. Furthermore, there is ample evidence for sex differences in the gut microbiota. However, it is unclear whether such sex differences impact blood pressure differentially through the tryptophan-indole pathway. METHODS To study the sex-specific mechanisms of gut microbiota-mediated tryptophan-indole pathway in hypertension, we generated a novel rat model with Clustered Regularly Interspaced Short Palindromic Repeats/Cas9 (Clustered Regularly Interspaced Short Palindromic Repeats-associated protein 9)-targeted deletion of Ace2 in the Dahl salt-sensitive rat. Cecal microbiota transfers from donors of both sexes to female S recipients were performed. Also, Dahl salt-sensitive rats of both sexes were orally gavaged with indole to investigate blood pressure response. RESULTS The female gut microbiota and its tryptophan-indole pathway exhibited greater buffering capacity when exposed to tryptophan, due to Ace2 deficiency, and salt. In contrast, the male gut microbiota and its tryptophan-indole pathway were more vulnerable. Female rats with male cecal microbiota responded to salt with a higher blood pressure increase compared with those with female cecal microbiota. Indole, a tryptophan-derived metabolite produced by gut bacteria, increased blood pressure in male but not in female rats. Moreover, salt altered host-mediated tryptophan metabolism, characterized by reduced serum serotonin of both sexes and higher levels of kynurenine derivatives in the females. CONCLUSIONS We uncovered a novel sex-specific mechanism in the gut microbiota-mediated tryptophan-indole pathway in blood pressure regulation. Salt tipped the tryptophan metabolism between the host and gut microbiota in a sex-dependent manner. Our study provides evidence for a novel concept that gut microbiota and its metabolism play sex-specific roles in the development of salt-sensitive hypertension.
Collapse
Affiliation(s)
- Pritam Bardhan
- Department of Physiology and Pharmacology, Microbiome Consortium, Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, OH (P.B., X.M., N.K.L., B.M., R.T., S.A., I.M., H.H., T.A.J., R.R.A., J.K., B.J., T.Y.)
| | - Xue Mei
- Department of Physiology and Pharmacology, Microbiome Consortium, Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, OH (P.B., X.M., N.K.L., B.M., R.T., S.A., I.M., H.H., T.A.J., R.R.A., J.K., B.J., T.Y.)
- Now with Department of Pharmacy, North Sichuan Medical College, Nanchong, China (X.M.)
| | - Ngoc Khanh Lai
- Department of Physiology and Pharmacology, Microbiome Consortium, Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, OH (P.B., X.M., N.K.L., B.M., R.T., S.A., I.M., H.H., T.A.J., R.R.A., J.K., B.J., T.Y.)
| | - Blair Mell
- Department of Physiology and Pharmacology, Microbiome Consortium, Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, OH (P.B., X.M., N.K.L., B.M., R.T., S.A., I.M., H.H., T.A.J., R.R.A., J.K., B.J., T.Y.)
| | - Ramakumar Tummala
- Department of Physiology and Pharmacology, Microbiome Consortium, Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, OH (P.B., X.M., N.K.L., B.M., R.T., S.A., I.M., H.H., T.A.J., R.R.A., J.K., B.J., T.Y.)
| | - Sachin Aryal
- Department of Physiology and Pharmacology, Microbiome Consortium, Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, OH (P.B., X.M., N.K.L., B.M., R.T., S.A., I.M., H.H., T.A.J., R.R.A., J.K., B.J., T.Y.)
| | - Ishan Manandhar
- Department of Physiology and Pharmacology, Microbiome Consortium, Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, OH (P.B., X.M., N.K.L., B.M., R.T., S.A., I.M., H.H., T.A.J., R.R.A., J.K., B.J., T.Y.)
| | - Hyeongu Hwang
- Department of Physiology and Pharmacology, Microbiome Consortium, Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, OH (P.B., X.M., N.K.L., B.M., R.T., S.A., I.M., H.H., T.A.J., R.R.A., J.K., B.J., T.Y.)
| | - Tania Akter Jhuma
- Department of Physiology and Pharmacology, Microbiome Consortium, Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, OH (P.B., X.M., N.K.L., B.M., R.T., S.A., I.M., H.H., T.A.J., R.R.A., J.K., B.J., T.Y.)
| | - Rohit Reddy Atluri
- Department of Physiology and Pharmacology, Microbiome Consortium, Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, OH (P.B., X.M., N.K.L., B.M., R.T., S.A., I.M., H.H., T.A.J., R.R.A., J.K., B.J., T.Y.)
| | - Jun Kyoung
- Department of Physiology and Pharmacology, Microbiome Consortium, Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, OH (P.B., X.M., N.K.L., B.M., R.T., S.A., I.M., H.H., T.A.J., R.R.A., J.K., B.J., T.Y.)
| | - Ying Li
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, China (Y.L., H.-B.L.)
| | - Bina Joe
- Department of Physiology and Pharmacology, Microbiome Consortium, Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, OH (P.B., X.M., N.K.L., B.M., R.T., S.A., I.M., H.H., T.A.J., R.R.A., J.K., B.J., T.Y.)
| | - Hong-Bao Li
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, China (Y.L., H.-B.L.)
| | - Tao Yang
- Department of Physiology and Pharmacology, Microbiome Consortium, Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, OH (P.B., X.M., N.K.L., B.M., R.T., S.A., I.M., H.H., T.A.J., R.R.A., J.K., B.J., T.Y.)
| |
Collapse
|
40
|
Pfau M, Degregori S, Barber PH, Blumstein DT, Philson CS. Differences in Gut Microbes Across Age and Sex Linked to Metabolism and Microbial Stability in a Hibernating Mammal. Ecol Evol 2024; 14:e70519. [PMID: 39524311 PMCID: PMC11550910 DOI: 10.1002/ece3.70519] [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: 06/17/2024] [Revised: 10/01/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024] Open
Abstract
The gut microbiome has a well-documented relationship with host fitness, physiology, and behavior. However, most of what is known comes from captive animals where diets and environments are more homogeneous or controlled. Studies in wild populations that experience dynamic environments and have natural life history variation are less common but are key to understanding the drivers of variation in the gut microbiome. Here we examine a wild population of yellow-bellied marmots (Marmota flaviventer), an obligate winter hibernator, to quantify multivariate associations between host-associated factors (e.g., age, sex, environmental harshness, and social behavior) and gut microbial composition. Across 5 years and 143 individuals, we found that males had a higher relative abundance of microbes associated with mass gain and cellulose digestion, which suggests a metabolic investment in mass gain (such as phylum Firmicutes and family Lachnospiraceae). By contrast, females had higher relative abundances of microbes associated with inflammation and metabolism (from microbial groups such as Tenericutes and Ruminococcus), possibly reflecting the importance of lactation and offspring investment. Post hoc analyses of lactating females showed a negative relationship with the abundance of microbes associated with mass gain but a positive relationship with microbes associated with metabolic energy, suggesting a trade-off between investment in pups and maternal mass gain. Older animals also had reduced Proteobacteria relative abundance, a phylum associated with reduced inflammation. Results demonstrate that sex and age-based traits, not sociality or environmental harshness, are associated with microbe-mediated metabolism and inflammation in a wild, hibernating mammal.
Collapse
Affiliation(s)
- Madison Pfau
- Department of Ecology and Evolutionary BiologyUCLALos AngelesCaliforniaUSA
- Department of Environmental Science, Policy, and ManagementUniversity of California, BerkeleyBerkeleyCaliforniaUSA
| | - Samuel Degregori
- Department of AnthropologyNorthwestern UniversityEvanstonIllinoisUSA
| | - Paul H. Barber
- Department of Ecology and Evolutionary BiologyUCLALos AngelesCaliforniaUSA
| | - Daniel T. Blumstein
- Department of Ecology and Evolutionary BiologyUCLALos AngelesCaliforniaUSA
- Rocky Mountain Biological LaboratoryCrested ButteColoradoUSA
| | - Conner S. Philson
- Department of Ecology and Evolutionary BiologyUCLALos AngelesCaliforniaUSA
- Rocky Mountain Biological LaboratoryCrested ButteColoradoUSA
- Centre for Research in Animal BehaviourUniversity of ExeterExeterUK
| |
Collapse
|
41
|
Paz HA, Buddha L, Zhong Y, Sikes JD, Wankhade UD. Impact of maternal high-fat diet on offspring gut microbiota during short-term high-fat diet exposure in mice. Physiol Rep 2024; 12:e70111. [PMID: 39489538 PMCID: PMC11531878 DOI: 10.14814/phy2.70111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/16/2024] [Accepted: 10/16/2024] [Indexed: 11/05/2024] Open
Abstract
Alterations in the gut microbiome have been linked to obesity, with maternal high-fat diet (HF) playing a role in shaping offspring microbiome composition. However, the sex-specific responses to maternal HF diet and the impact of subsequent dietary challenges remain unclear. This study investigated the effects of maternal HF diet on offspring gut microbiota structure and predicted functional profile in response to short-term postnatal HF diet exposure with a focus on sex-specific responses. Female and male offspring of maternal control (C) diet or maternal HF diet were weaned onto C diet or HF diet. Offspring were euthanized at 13 weeks of age and cecal contents were collected for bacterial taxonomic profiling. Maternal HF diet reduced α-diversity, notably in male offspring weaned onto HF diet. Sex-specific differences were observed in the gut microbial composition and predicted functional potential. Furthermore, the influence of maternal diet on bacterial community structure and functional potential varied depending on postnatal diet. Maternal HF diet led to increased relative abundance of Corynebacterium in female offspring and decreased abundance of Akkermansia and Roseburia in male offspring. These findings underscore the sexually dimorphic nature of maternal HF diet effects on gut microbiota composition and function, with implications for developmental programming and metabolic health.
Collapse
Affiliation(s)
- Henry A. Paz
- Department of Pediatrics, College of MedicineUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
- Arkansas Children's Nutrition CenterLittle RockArkansasUSA
| | - Lasya Buddha
- Department of Pediatrics, College of MedicineUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
| | - Ying Zhong
- Department of Pediatrics, College of MedicineUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
- Arkansas Children's Nutrition CenterLittle RockArkansasUSA
| | - James D. Sikes
- Arkansas Children's Nutrition CenterLittle RockArkansasUSA
| | - Umesh D. Wankhade
- Department of Pediatrics, College of MedicineUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
- Arkansas Children's Nutrition CenterLittle RockArkansasUSA
| |
Collapse
|
42
|
Dowden RA, Kerkhof LJ, Wisniewski PJ, Häggblom MM, Campbell SC. Temporal changes in the mouse gut bacteriota influenced by host sex, diet, and exercise. J Appl Physiol (1985) 2024; 137:1374-1388. [PMID: 39298618 DOI: 10.1152/japplphysiol.00487.2023] [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: 07/18/2023] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 09/22/2024] Open
Abstract
The gut microbiota plays an important role in host physiology. However, the effects of host sex, lifestyle, and temporal influences on the bacterial community within the gut remain ill-defined. To address this gap, we evaluated 56 male and female mice over a 10-wk study to assess the effects of sex, diet, and exercise on gut community dynamics. Mice were randomly assigned to high-fat or control diet feeding and had free access to running wheels or remained sedentary throughout the study period. The fecal bacterial community was characterized by rRNA operon profiling via nanopore sequencing. Differential abundance testing indicated that ∼200 bacterial taxa were significantly influenced by sex, diet, or exercise (4.2% of total community), which also changed over time (82 taxa, 1.7% of total community). Phylogenetic analysis of taxa closely related to Dysosmobacter welbionis, and several members of the family Muribaculaceae were examined more closely and demonstrated distinct species/strain level subclustering by host sex, diet, and exercise. Collectively, these data suggest that sex and lifestyle can alter the gut bacteriota at the species/strain level that may play a role in host health. These results also highlight the need for improved characterization methods to survey microbial communities at finer taxonomic resolution.NEW & NOTEWORTHY This study demonstrates that host sex, diet, and exercise can each modulate gut bacterial community structure, which may have consequences to host physiology. Our analysis shows selection of novel strains and genera for some members of the Oscillospiraceae and Muribaculaceae by host sex, diet, and activity status. Overall, these findings provide a framework for detecting the next generation of beneficial bacteria targeting obesity and associated metabolic diseases in a sex-specific manner.
Collapse
Affiliation(s)
- Robert A Dowden
- Department of Kinesiology and Health, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, United States
- 10x Genomics, Pleasanton, California, United States
| | - Lee J Kerkhof
- Department of Marine and Coastal Sciences, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, United States
| | | | - Max M Häggblom
- Department of Biochemistry and Microbiology, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, United States
| | - Sara C Campbell
- Department of Kinesiology and Health, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, United States
| |
Collapse
|
43
|
Costa CFFA, Correia-de-Sá T, Araujo R, Barbosa F, Burnet PWJ, Ferreira-Gomes J, Sampaio-Maia B. The oral-gut microbiota relationship in healthy humans: identifying shared bacteria between environments and age groups. Front Microbiol 2024; 15:1475159. [PMID: 39512939 PMCID: PMC11540997 DOI: 10.3389/fmicb.2024.1475159] [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: 08/02/2024] [Accepted: 09/25/2024] [Indexed: 11/15/2024] Open
Abstract
Introduction Although the oral cavity and the gut are anatomically continuous regions of the gastrointestinal tract, research on the relationship between oral and gut microbiota remains sparse. Oral-gut bacterial translocation is mostly studied in pathological contexts, thus evidence of translocation in healthy conditions is still scarce. Studying the oral-gut microbiota relationship in humans in different life stages is necessary in order to understand how these microbial communities might relate throughout life. Methods In this study, saliva and fecal samples were collected from healthy participants (39 children, 97 adults). Microbiota analysis was carried out by sequencing the V4 region of the 16S ribosomal RNA gene, followed by amplicon sequence variant (ASV) analysis. Results and discussion Although the oral and gut microbiota are vastly different, a subset of 61 ASVs were present in both the oral cavity and gut of the same individual, and represented 1.6% of all ASVs detected. From these, 26 ASVs (classified into 18 genera: Actinomyces, Rothia, Bacteroides, Porphyromonas, Prevotella, Alistipes, Fusobacterium, Neisseria, Haemophilus, Akkermansia, Solobacterium, Granulicatella, Streptococcus, Gemella, Mogibacterium, Dialister, Veillonella, Christensenellaceae R-7 group) were present in both children and adults, suggesting the possibility of persistent colonization of both habitats by these microorganisms, initiating in childhood. Additionally, 62% of shared ASVs were more abundant in the oral cavity, indicating that oral-to-gut translocation may be the main route of translocation between environments, and highlighting that this phenomenon might be more common than previously thought in healthy individuals of all ages.
Collapse
Affiliation(s)
- Carolina F. F. A. Costa
- ICBAS – Instituto de Ciências Biomédicas Abel Salazar da Universidade do Porto, Porto, Portugal
- NanoBiomaterials for Targeted Therapies, INEB – Instituto Nacional de Engenharia Biomédica, i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Teresa Correia-de-Sá
- NanoBiomaterials for Targeted Therapies, INEB – Instituto Nacional de Engenharia Biomédica, i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Departamento de Biomedicina, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
| | - Ricardo Araujo
- NanoBiomaterials for Targeted Therapies, INEB – Instituto Nacional de Engenharia Biomédica, i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Fernando Barbosa
- Laboratório de Neuropsicofisiologia, Faculdade de Psicologia e de Ciências da Educação, Universidade do Porto, Porto, Portugal
| | | | - Joana Ferreira-Gomes
- Departamento de Biomedicina, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
- IBMC – Instituto de Biologia Molecular e Celular, i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Benedita Sampaio-Maia
- NanoBiomaterials for Targeted Therapies, INEB – Instituto Nacional de Engenharia Biomédica, i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Faculdade de Medicina Dentária, Universidade do Porto, Porto, Portugal
| |
Collapse
|
44
|
Mitchell LK, Heussler HS, Burgess CJ, Rehman A, Steinert RE, Davies PSW. Gastrointestinal, Behaviour and Anxiety Outcomes in Autistic Children Following an Open Label, Randomised Pilot Study of Synbiotics vs Synbiotics and Gut-Directed Hypnotherapy. J Autism Dev Disord 2024:10.1007/s10803-024-06588-9. [PMID: 39417900 DOI: 10.1007/s10803-024-06588-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2024] [Indexed: 10/19/2024]
Abstract
Alterations of the microbiome-gut-brain (MGB) axis have been associated with autism spectrum disorder (ASD) and disorders of gut-brain interaction (DGBI). DGBI are highly prevalent in autistic children and are associated with worsening behaviour and anxiety. Treatments such as probiotics, prebiotics and gut-directed hypnotherapy (GDH) have shown efficacy in improving gut symptoms in children. The primary objective of the study was to compare changes in gastrointestinal (GI) scores following a 12-week intervention of synbiotics (prebiotic + probiotic) +/- GDH with a follow-up at 24 weeks. Secondary objectives included changes in behavioural and anxiety symptoms, while changes in gut microbiome composition were assessed as an exploratory objective. Children diagnosed with ASD aged 5.00-10.99 years (n = 40) were recruited and randomised (1:1) to a 12-week intervention of either synbiotics (SYN group) or synbiotics + GDH (COM group). Both the SYN and COM group experienced significant reductions in total GI scores post-intervention and at follow-up (p < 0.001), with no superiority of the COM treatment over the SYN treatment. The COM group showed beneficial reductions in anxiety scores (p = 0.002) and irritability behaviours (p < 0.001) which were not present in the SYN group. At follow-up, only those in the COM group maintained significant reductions in GI pain scores (p < 0.001). There were significant changes in gut microbiota such as increases in Bifidobacterium animalis and Dialister in both groups over time. In conclusion, synbiotics with or without GDH may help support standard care for autistic children who suffer comorbid DGBI. The trial was prospectively registered at clinicialtrials.gov on 16 November 2020 (NCTO4639141).
Collapse
Affiliation(s)
- Leanne K Mitchell
- Child Health Research Centre, Faculty of Medicine, The University of Queensland, South Brisbane, QLD, Australia.
| | - Helen S Heussler
- Child Health Research Centre, Faculty of Medicine, The University of Queensland, South Brisbane, QLD, Australia
- Child Development Program, Children's Health Queensland, Brisbane, QLD, Australia
- Centre for Clinical Trials in Rare Neuro Developmental Disorders, Children's Health Queensland, Brisbane, QLD, Australia
| | - Christopher J Burgess
- Child Health Research Centre, Faculty of Medicine, The University of Queensland, South Brisbane, QLD, Australia
- Department of Gastroenterology, Hepatology and Liver Transplant, Queensland Children's Hospital, Brisbane, QLD, Australia
| | - Ateequr Rehman
- DSM-Firmenich, Health, Nutrition & Care (HNC), Kaiseraugst, Switzerland
| | - Robert E Steinert
- DSM-Firmenich, Health, Nutrition & Care (HNC), Kaiseraugst, Switzerland
- Department of Surgery, Division of Visceral and Transplantation Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Peter S W Davies
- Child Health Research Centre, Faculty of Medicine, The University of Queensland, South Brisbane, QLD, Australia
| |
Collapse
|
45
|
Shoer S, Reicher L, Zhao C, Pollard KS, Pilpel Y, Segal E. Pangenomes of human gut microbiota uncover links between genetic diversity and stress response. Cell Host Microbe 2024; 32:1744-1757.e2. [PMID: 39353429 PMCID: PMC12060796 DOI: 10.1016/j.chom.2024.08.017] [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: 04/22/2024] [Revised: 07/11/2024] [Accepted: 08/23/2024] [Indexed: 10/04/2024]
Abstract
The genetic diversity of the gut microbiota has a central role in host health. Here, we created pangenomes for 728 human gut prokaryotic species, quadrupling the genes of strain-specific genomes. Each of these species has a core set of a thousand genes, differing even between closely related species, and an accessory set of genes unique to the different strains. Functional analysis shows high strain variability associates with sporulation, whereas low variability is linked with antibiotic resistance. We further map the antibiotic resistome across the human gut population and find 237 cases of extreme resistance even to last-resort antibiotics, with a predominance among Enterobacteriaceae. Lastly, the presence of specific genes in the microbiota relates to host age and sex. Our study underscores the genetic complexity of the human gut microbiota, emphasizing its significant implications for host health. The pangenomes and antibiotic resistance map constitute a valuable resource for further research.
Collapse
Affiliation(s)
- Saar Shoer
- Department of Computer Science and Applied Mathematics, The Weizmann Institute of Science, Rehovot, Israel; Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot, Israel
| | - Lee Reicher
- Department of Computer Science and Applied Mathematics, The Weizmann Institute of Science, Rehovot, Israel; Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot, Israel; Lis Maternity and Women's Hospital, Sourasky Medical Center, Tel Aviv, Israel
| | - Chunyu Zhao
- Gladstone Institute for Data Science and Biotechnology, San Francisco, CA, USA; Chan Zuckerberg Biohub San Francisco, San Francisco, CA, USA
| | - Katherine S Pollard
- Gladstone Institute for Data Science and Biotechnology, San Francisco, CA, USA; Chan Zuckerberg Biohub San Francisco, San Francisco, CA, USA; Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| | - Yitzhak Pilpel
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| | - Eran Segal
- Department of Computer Science and Applied Mathematics, The Weizmann Institute of Science, Rehovot, Israel; Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
46
|
Srivastava A, Anand S. Profiling the dynamic pediatric urobiome: Missing links and future directions! J Pediatr Urol 2024; 20:894-895. [PMID: 38981782 DOI: 10.1016/j.jpurol.2024.06.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 07/11/2024]
Affiliation(s)
- Anjali Srivastava
- All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Sachit Anand
- All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India.
| |
Collapse
|
47
|
Hayase E, Hayase T, Mukherjee A, Stinson SC, Jamal MA, Ortega MR, Sanchez CA, Ahmed SS, Karmouch JL, Chang CC, Flores II, McDaniel LK, Brown AN, El-Himri RK, Chapa VA, Tan L, Tran BQ, Xiao Y, Fan C, Pham D, Halsey TM, Jin Y, Tsai WB, Prasad R, Glover IK, Enkhbayar A, Mohammed A, Schmiester M, King KY, Britton RA, Reddy P, Wong MC, Ajami NJ, Wargo JA, Shelburne S, Okhuysen PC, Liu C, Fowler SW, Conner ME, Katsamakis Z, Smith N, Burgos da Silva M, Ponce DM, Peled JU, van den Brink MRM, Peterson CB, Rondon G, Molldrem JJ, Champlin RE, Shpall EJ, Lorenzi PL, Mehta RS, Martens EC, Alousi AM, Jenq RR. Bacteroides ovatus alleviates dysbiotic microbiota-induced graft-versus-host disease. Cell Host Microbe 2024; 32:1621-1636.e6. [PMID: 39214085 PMCID: PMC11441101 DOI: 10.1016/j.chom.2024.08.004] [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: 12/19/2023] [Revised: 06/12/2024] [Accepted: 08/02/2024] [Indexed: 09/04/2024]
Abstract
Acute lower gastrointestinal GVHD (aLGI-GVHD) is a serious complication of allogeneic hematopoietic stem cell transplantation. Although the intestinal microbiota is associated with the incidence of aLGI-GVHD, how the intestinal microbiota impacts treatment responses in aLGI-GVHD has not been thoroughly studied. In a cohort of patients with aLGI-GVHD (n = 37), we found that non-response to standard therapy with corticosteroids was associated with prior treatment with carbapenem antibiotics and a disrupted fecal microbiome characterized by reduced abundances of Bacteroides ovatus. In a murine GVHD model aggravated by carbapenem antibiotics, introducing B. ovatus reduced GVHD severity and improved survival. These beneficial effects of Bacteroides ovatus were linked to its ability to metabolize dietary polysaccharides into monosaccharides, which suppressed the mucus-degrading capabilities of colonic mucus degraders such as Bacteroides thetaiotaomicron and Akkermansia muciniphila, thus reducing GVHD-related mortality. Collectively, these findings reveal the importance of microbiota in aLGI-GVHD and therapeutic potential of B. ovatus.
Collapse
Affiliation(s)
- Eiko Hayase
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA.
| | - Tomo Hayase
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Akash Mukherjee
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Stuart C Stinson
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mohamed A Jamal
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Miriam R Ortega
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Christopher A Sanchez
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Saira S Ahmed
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Jennifer L Karmouch
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Chia-Chi Chang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Ivonne I Flores
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Lauren K McDaniel
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Alexandria N Brown
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Rawan K El-Himri
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Valerie A Chapa
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Lin Tan
- Metabolomics Core Facility, Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77230, USA
| | - Bao Q Tran
- Metabolomics Core Facility, Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77230, USA
| | - Yao Xiao
- Department of Microbiology & Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Christopher Fan
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Dung Pham
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Taylor M Halsey
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Yimei Jin
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Wen-Bin Tsai
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Rishika Prasad
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Israel K Glover
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Altai Enkhbayar
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Aqsa Mohammed
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Maren Schmiester
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Katherine Y King
- Center for Cell and Gene Therapy and Department of Pediatrics, Division of Infectious Diseases, Baylor College of Medicine, Houston, TX 77030, USA
| | - Robert A Britton
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Pavan Reddy
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Matthew C Wong
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Nadim J Ajami
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Jennifer A Wargo
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Samuel Shelburne
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA; Department of Infectious Diseases, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Pablo C Okhuysen
- Department of Infectious Diseases, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Chen Liu
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Stephanie W Fowler
- Department of Molecular Virology and Microbiology and Department of Education, Innovation, and Technology, Baylor College of Medicine, Houston, TX 77030, USA; Center for Comparative Medicine and Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Margaret E Conner
- Department of Molecular Virology and Microbiology and Department of Education, Innovation, and Technology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Zoe Katsamakis
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Natalie Smith
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Marina Burgos da Silva
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Doris M Ponce
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jonathan U Peled
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Weill Cornell Medical College, New York, NY 10021, USA
| | - Marcel R M van den Brink
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Weill Cornell Medical College, New York, NY 10021, USA
| | - Christine B Peterson
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Gabriela Rondon
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jeffrey J Molldrem
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Hematopoietic Biology & Malignancy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Richard E Champlin
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Elizabeth J Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Philip L Lorenzi
- Metabolomics Core Facility, Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77230, USA
| | - Rohtesh S Mehta
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Eric C Martens
- Department of Microbiology & Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Amin M Alousi
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Robert R Jenq
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA; Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; CPRIT Scholar in Cancer Research, Houston, TX, USA.
| |
Collapse
|
48
|
Girdhar K, Mine K, DaCosta JM, Atkinson MA, Ludvigsson J, Altindis E. Sex-Specific Cytokine, Chemokine, and Growth Factor Signatures in T1D Patients and Progressors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.05.611513. [PMID: 39282401 PMCID: PMC11398455 DOI: 10.1101/2024.09.05.611513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2024]
Abstract
While studies have reported altered levels of cytokines in type 1 diabetes (T1D) patients, the results are inconsistent, likely because of variable factors. This study tests the hypothesis that there are sex-based differences in cytokine levels in T1D, prior to and after disease onset. We analyzed 48 blood cytokine, chemokine, and growth factor levels using a multiplex assay. We found only two cytokines, M-CSF and IL-6, with significant differences between T1D patients (n=25) versus controls overall (n=25). However, we identified notable alterations when comparing sex-age-matched controls and T1D samples. Inflammatory cytokines (TNF-α, IL-6, IL-1a), Th2 cytokines (IL-4, IL-13), and chemokines (MIP-1α, RANTES, MIP-3) were lower in female T1D patients compared to female controls, but not in males. IL-22 was lower in female T1D patients compared to female controls, while it was higher in male T1D patients compared to male controls. In contrast, growth factors (EGF, PDGF-AB/BB) were higher in male T1D patients compared to male controls. In T1D progressors (children who developed the disease years after the sample collection, n=16-21), GROa was lower compared to controls in both sexes. Our findings underscore the importance of understanding sex-specific differences in T1D pathogenesis and their implications for developing personalized treatments.
Collapse
|
49
|
James D, Poveda C, Walton GE, Elmore JS, Linden B, Gibson J, Griffin BA, Robertson MD, Lewis MC. Do high-protein diets have the potential to reduce gut barrier function in a sex-dependent manner? Eur J Nutr 2024; 63:2035-2054. [PMID: 38662018 PMCID: PMC11377480 DOI: 10.1007/s00394-024-03407-w] [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: 12/05/2023] [Accepted: 04/16/2024] [Indexed: 04/26/2024]
Abstract
PURPOSE Impaired gut barrier function is associated with systemic inflammation and many chronic diseases. Undigested dietary proteins are fermented in the colon by the gut microbiota which produces nitrogenous metabolites shown to reduce barrier function in vitro. With growing evidence of sex-based differences in gut microbiotas, we determined whether there were sex by dietary protein interactions which could differentially impact barrier function via microbiota modification. METHODS Fermentation systems were inoculated with faeces from healthy males (n = 5) and females (n = 5) and supplemented with 0.9 g of non-hydrolysed proteins sourced from whey, fish, milk, soya, egg, pea, or mycoprotein. Microbial populations were quantified using fluorescence in situ hybridisation with flow cytometry. Metabolite concentrations were analysed using gas chromatography, solid phase microextraction coupled with gas chromatography-mass spectrometry and ELISA. RESULTS Increased protein availability resulted in increased proteolytic Bacteroides spp (p < 0.01) and Clostridium coccoides (p < 0.01), along with increased phenol (p < 0.01), p-cresol (p < 0.01), indole (p = 0.018) and ammonia (p < 0.01), varying by protein type. Counts of Clostridium cluster IX (p = 0.03) and concentration of p-cresol (p = 0.025) increased in males, while females produced more ammonia (p = 0.02), irrespective of protein type. Further, we observed significant sex-protein interactions affecting bacterial populations and metabolites (p < 0.005). CONCLUSIONS Our findings suggest that protein fermentation by the gut microbiota in vitro is influenced by both protein source and the donor's sex. Should these results be confirmed through human studies, they could have major implications for developing dietary recommendations tailored by sex to prevent chronic illnesses.
Collapse
Affiliation(s)
- Daniel James
- Department of Food and Nutritional Sciences, University of Reading, Whiteknights Campus, Reading, RG6 6DZ, UK.
| | - Carlos Poveda
- Department of Food and Nutritional Sciences, University of Reading, Whiteknights Campus, Reading, RG6 6DZ, UK
| | - Gemma E Walton
- Department of Food and Nutritional Sciences, University of Reading, Whiteknights Campus, Reading, RG6 6DZ, UK
| | - J Stephen Elmore
- Department of Food and Nutritional Sciences, University of Reading, Whiteknights Campus, Reading, RG6 6DZ, UK
| | - Brandon Linden
- Department of Nutrition, Food & Exercise Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - John Gibson
- Food and Feed Innovations, Woodstock, Newcastle Rd, Woore, N Shropshire, CW3 95N, UK
| | - Bruce A Griffin
- Department of Nutrition, Food & Exercise Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - M Denise Robertson
- Department of Nutrition, Food & Exercise Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - Marie C Lewis
- Department of Food and Nutritional Sciences, University of Reading, Whiteknights Campus, Reading, RG6 6DZ, UK
| |
Collapse
|
50
|
Wang Z, Li S, Zhang S, Zhang T, Wu Y, Liu A, Wang K, Ji X, Cao H, Zhang Y, Tan EK, Wang Y, Wang Y, Liu W. Hosts manipulate lifestyle switch and pathogenicity heterogeneity of opportunistic pathogens in the single-cell resolution. eLife 2024; 13:RP96789. [PMID: 39190452 PMCID: PMC11349298 DOI: 10.7554/elife.96789] [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: 08/28/2024] Open
Abstract
Host-microbe interactions are virtually bidirectional, but how the host affects their microbiome is poorly understood. Here, we report that the host is a critical modulator to regulate the lifestyle switch and pathogenicity heterogeneity of the opportunistic pathogens Serratia marcescens utilizing the Drosophila and bacterium model system. First, we find that Drosophila larvae efficiently outcompete S. marcescens and typically drive a bacterial switch from pathogenicity to commensalism toward the fly. Furthermore, Drosophila larvae reshape the transcriptomic and metabolic profiles of S. marcescens characterized by a lifestyle switch. More importantly, the host alters pathogenicity and heterogeneity of S. marcescens in the single-cell resolution. Finally, we find that larvae-derived AMPs are required to recapitulate the response of S. marcescens to larvae. Altogether, our findings provide an insight into the pivotal roles of the host in harnessing the life history and heterogeneity of symbiotic bacterial cells, advancing knowledge of the reciprocal relationships between the host and pathogen.
Collapse
Affiliation(s)
- Ziguang Wang
- School of Plant Protection; Anhui Province Key Laboratory of Crop Integrated Pest Management; Anhui Province Key Laboratory of Resource Insect Biology and Innovative Utilization, Anhui Agricultural UniversityHefeiChina
- College of Life Sciences, Nankai UniversityTianjinChina
- First Clinical Medical College, Mudanjiang Medical CollegeMudanjiangChina
| | - Shuai Li
- Bioinformatics Center, College of Biology, Hunan UniversityChangshaChina
| | - Sheng Zhang
- School of Plant Protection; Anhui Province Key Laboratory of Crop Integrated Pest Management; Anhui Province Key Laboratory of Resource Insect Biology and Innovative Utilization, Anhui Agricultural UniversityHefeiChina
| | - Tianyu Zhang
- Liangzhu Laboratory, Zhejiang UniversityHangzhouChina
| | - Yujie Wu
- School of Plant Protection; Anhui Province Key Laboratory of Crop Integrated Pest Management; Anhui Province Key Laboratory of Resource Insect Biology and Innovative Utilization, Anhui Agricultural UniversityHefeiChina
| | - Anqi Liu
- School of Plant Protection; Anhui Province Key Laboratory of Crop Integrated Pest Management; Anhui Province Key Laboratory of Resource Insect Biology and Innovative Utilization, Anhui Agricultural UniversityHefeiChina
| | - Kui Wang
- School of Plant Protection; Anhui Province Key Laboratory of Crop Integrated Pest Management; Anhui Province Key Laboratory of Resource Insect Biology and Innovative Utilization, Anhui Agricultural UniversityHefeiChina
| | - Xiaowen Ji
- School of Plant Protection; Anhui Province Key Laboratory of Crop Integrated Pest Management; Anhui Province Key Laboratory of Resource Insect Biology and Innovative Utilization, Anhui Agricultural UniversityHefeiChina
| | - Haiqun Cao
- School of Plant Protection; Anhui Province Key Laboratory of Crop Integrated Pest Management; Anhui Province Key Laboratory of Resource Insect Biology and Innovative Utilization, Anhui Agricultural UniversityHefeiChina
| | - Yinglao Zhang
- School of Plant Protection; Anhui Province Key Laboratory of Crop Integrated Pest Management; Anhui Province Key Laboratory of Resource Insect Biology and Innovative Utilization, Anhui Agricultural UniversityHefeiChina
| | - Eng King Tan
- Department of Neurology, National Neuroscience Institute, Singapore General Hospital CampusSingaporeSingapore
| | | | - Yirong Wang
- Bioinformatics Center, College of Biology, Hunan UniversityChangshaChina
| | - Wei Liu
- School of Plant Protection; Anhui Province Key Laboratory of Crop Integrated Pest Management; Anhui Province Key Laboratory of Resource Insect Biology and Innovative Utilization, Anhui Agricultural UniversityHefeiChina
| |
Collapse
|