1
|
Xiao HL, Zhu H, Zeng TA, Xu F, Yu SH, Yang CJ. Potential similarities in gut microbiota composition between autism spectrum disorder and neurotypical siblings: Insights from a comprehensive meta-analysis. Neuroscience 2025; 567:172-181. [PMID: 39788315 DOI: 10.1016/j.neuroscience.2025.01.010] [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: 10/23/2024] [Revised: 12/22/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
BACKGROUND Previous studies have explored the differences in gut microbiota (GM) between individuals with autism spectrum disorder (ASD) and neurotypical controls. However, factors such as diet, lifestyle, and environmental exposure influence GM, leading to significant variability, even among neurotypical individuals. Comparing the GM of ASD individuals with neurotypical siblings, who share similar genes and living conditions, may offer better insights into the GM mechanisms associated with ASD. Therefore, this study aims to analyze the GM composition in ASD by comparing it to that of neurotypical siblings, potentially identifying microbiota that influence ASD. METHODS We explored electronic databases up to July 2024, including EBSCOhost, PubMed, ScienceDirect, Web of Science, and Scopus. Meta-analysis using RevMan 5.4 assessed the relative abundance (RA) of gut bacteria from 8 phyla and 4 genera in ASD individuals and neurotypical siblings. RESULTS Eight studies were included, involving 248 people with ASD and 197 neurotypical siblings. Significant but unstable differences were observed in the RA of Bacteroidetes, Firmicutes, and Fusobacteria. No significant differences were found in the RA of Proteobacteria, Cyanobacteria, Actinobacteria, Verrucomicrobia, Tenericutes, or Bacteroides, Roseburia, Sutterella, Bifidobacterium. CONCLUSIONS GM composition in ASD individuals closely resembles that of neurotypical siblings, with only a few unstable differences. This suggests that other crucial bacteria or certain interacting environmental factors play a role. Further studies are needed to gather stronger evidence to uncover the differences in GM and their mechanisms in ASD people.
Collapse
Affiliation(s)
- Hong-Li Xiao
- Faculty of Education, East China Normal University, Shanghai, China
| | - Han Zhu
- Faculty of Education, East China Normal University, Shanghai, China
| | - Tong-Ao Zeng
- Faculty of Education, East China Normal University, Shanghai, China
| | - Fang Xu
- Faculty of Education, East China Normal University, Shanghai, China; Hangzhou Health Experimental School, Zhejiang, China
| | - Su-Hong Yu
- Faculty of Education, East China Normal University, Shanghai, China.
| | - Chang-Jiang Yang
- Faculty of Education, East China Normal University, Shanghai, China; China Research Institute of Care and Education of Infants and Young, ECNU, Shanghai, China.
| |
Collapse
|
2
|
Vicente-Valor J, Tesolato S, Paz-Cabezas M, Gómez-Garre D, Ortega-Hernández A, de la Serna S, Domínguez-Serrano I, Dziakova J, Rivera D, Jarabo JR, Gómez-Martínez AM, Hernando F, Torres A, Iniesta P. Fecal Microbiota Strongly Correlates with Tissue Microbiota Composition in Colorectal Cancer but Not in Non-Small Cell Lung Cancer. Int J Mol Sci 2025; 26:717. [PMID: 39859429 PMCID: PMC11766298 DOI: 10.3390/ijms26020717] [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: 12/12/2024] [Revised: 01/13/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Microbiota could be of interest in the diagnosis of colorectal and non-small cell lung cancer (CRC and NSCLC). However, how the microbial components of tissues and feces reflect each other remains unknown. In this work, our main objective is to discover the degree of correlation between the composition of the tissue microbiota and that of the feces of patients affected by CRC and NSCLC. Specifically, we investigated tumor and non-tumor tissues from 38 recruited patients with CRC and 19 with NSCLC. DNA from samples was submitted for 16S rDNA metagenomic sequencing, followed by data analysis through the QIIME2 pipeline and further statistical processing with STATA IC16. Tumor and non-tumor tissue selected genera were highly correlated in both CRC and NSCLC (100% and 81.25%). Following this, we established tissue-feces correlations, using selected genera from a LEfSe analysis previously published. In CRC, we found a strong correlation between the taxa detected in feces and those from colorectal tissues. However, our data do not demonstrate this correlation in NSCLC. In conclusion, our findings strongly reinforce the utility of fecal microbiota as a non-invasive biomarker for CRC diagnosis, while highlighting critical distinctions for NSCLC. Furthermore, our data demonstrate that the microbiota components of tumor and non-tumor tissues are similar, with only minor differences being detected.
Collapse
Affiliation(s)
- Juan Vicente-Valor
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University, 28040 Madrid, Spain; (J.V.-V.); (S.T.)
- San Carlos Health Research Institute (IdISSC), 28040 Madrid, Spain; (M.P.-C.); (D.G.-G.); (A.O.-H.); (S.d.l.S.); (I.D.-S.); (J.D.); (D.R.); (J.-R.J.); (A.-M.G.-M.); (F.H.); (A.T.)
| | - Sofía Tesolato
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University, 28040 Madrid, Spain; (J.V.-V.); (S.T.)
- San Carlos Health Research Institute (IdISSC), 28040 Madrid, Spain; (M.P.-C.); (D.G.-G.); (A.O.-H.); (S.d.l.S.); (I.D.-S.); (J.D.); (D.R.); (J.-R.J.); (A.-M.G.-M.); (F.H.); (A.T.)
| | - Mateo Paz-Cabezas
- San Carlos Health Research Institute (IdISSC), 28040 Madrid, Spain; (M.P.-C.); (D.G.-G.); (A.O.-H.); (S.d.l.S.); (I.D.-S.); (J.D.); (D.R.); (J.-R.J.); (A.-M.G.-M.); (F.H.); (A.T.)
- Biomedical Research Networking Center in Cancer (CIBERONC), Carlos III Health Institute, 28029 Madrid, Spain
| | - Dulcenombre Gómez-Garre
- San Carlos Health Research Institute (IdISSC), 28040 Madrid, Spain; (M.P.-C.); (D.G.-G.); (A.O.-H.); (S.d.l.S.); (I.D.-S.); (J.D.); (D.R.); (J.-R.J.); (A.-M.G.-M.); (F.H.); (A.T.)
- Cardiovascular Risk Group, Microbiota Laboratory, San Carlos Hospital, 28040 Madrid, Spain
- Department of Physiology, Faculty of Medicine, Complutense University, 28040 Madrid, Spain
- Biomedical Research Networking Center in Cardiovascular Diseases (CIBERCV), Carlos III Health Institute, 28029 Madrid, Spain
| | - Adriana Ortega-Hernández
- San Carlos Health Research Institute (IdISSC), 28040 Madrid, Spain; (M.P.-C.); (D.G.-G.); (A.O.-H.); (S.d.l.S.); (I.D.-S.); (J.D.); (D.R.); (J.-R.J.); (A.-M.G.-M.); (F.H.); (A.T.)
- Cardiovascular Risk Group, Microbiota Laboratory, San Carlos Hospital, 28040 Madrid, Spain
| | - Sofía de la Serna
- San Carlos Health Research Institute (IdISSC), 28040 Madrid, Spain; (M.P.-C.); (D.G.-G.); (A.O.-H.); (S.d.l.S.); (I.D.-S.); (J.D.); (D.R.); (J.-R.J.); (A.-M.G.-M.); (F.H.); (A.T.)
- Digestive Surgery Service, San Carlos Hospital, 28040 Madrid, Spain
- Department of Surgery, Faculty of Medicine, Complutense University, 28040 Madrid, Spain
| | - Inmaculada Domínguez-Serrano
- San Carlos Health Research Institute (IdISSC), 28040 Madrid, Spain; (M.P.-C.); (D.G.-G.); (A.O.-H.); (S.d.l.S.); (I.D.-S.); (J.D.); (D.R.); (J.-R.J.); (A.-M.G.-M.); (F.H.); (A.T.)
- Digestive Surgery Service, San Carlos Hospital, 28040 Madrid, Spain
- Department of Surgery, Faculty of Medicine, Complutense University, 28040 Madrid, Spain
| | - Jana Dziakova
- San Carlos Health Research Institute (IdISSC), 28040 Madrid, Spain; (M.P.-C.); (D.G.-G.); (A.O.-H.); (S.d.l.S.); (I.D.-S.); (J.D.); (D.R.); (J.-R.J.); (A.-M.G.-M.); (F.H.); (A.T.)
- Digestive Surgery Service, San Carlos Hospital, 28040 Madrid, Spain
- Department of Surgery, Faculty of Medicine, Complutense University, 28040 Madrid, Spain
| | - Daniel Rivera
- San Carlos Health Research Institute (IdISSC), 28040 Madrid, Spain; (M.P.-C.); (D.G.-G.); (A.O.-H.); (S.d.l.S.); (I.D.-S.); (J.D.); (D.R.); (J.-R.J.); (A.-M.G.-M.); (F.H.); (A.T.)
- Digestive Surgery Service, San Carlos Hospital, 28040 Madrid, Spain
| | - Jose-Ramón Jarabo
- San Carlos Health Research Institute (IdISSC), 28040 Madrid, Spain; (M.P.-C.); (D.G.-G.); (A.O.-H.); (S.d.l.S.); (I.D.-S.); (J.D.); (D.R.); (J.-R.J.); (A.-M.G.-M.); (F.H.); (A.T.)
- Department of Surgery, Faculty of Medicine, Complutense University, 28040 Madrid, Spain
- Thoracic Surgery Service, San Carlos Hospital, 28040 Madrid, Spain
| | - Ana-María Gómez-Martínez
- San Carlos Health Research Institute (IdISSC), 28040 Madrid, Spain; (M.P.-C.); (D.G.-G.); (A.O.-H.); (S.d.l.S.); (I.D.-S.); (J.D.); (D.R.); (J.-R.J.); (A.-M.G.-M.); (F.H.); (A.T.)
- Department of Surgery, Faculty of Medicine, Complutense University, 28040 Madrid, Spain
- Thoracic Surgery Service, San Carlos Hospital, 28040 Madrid, Spain
| | - Florentino Hernando
- San Carlos Health Research Institute (IdISSC), 28040 Madrid, Spain; (M.P.-C.); (D.G.-G.); (A.O.-H.); (S.d.l.S.); (I.D.-S.); (J.D.); (D.R.); (J.-R.J.); (A.-M.G.-M.); (F.H.); (A.T.)
- Department of Surgery, Faculty of Medicine, Complutense University, 28040 Madrid, Spain
- Thoracic Surgery Service, San Carlos Hospital, 28040 Madrid, Spain
| | - Antonio Torres
- San Carlos Health Research Institute (IdISSC), 28040 Madrid, Spain; (M.P.-C.); (D.G.-G.); (A.O.-H.); (S.d.l.S.); (I.D.-S.); (J.D.); (D.R.); (J.-R.J.); (A.-M.G.-M.); (F.H.); (A.T.)
- Digestive Surgery Service, San Carlos Hospital, 28040 Madrid, Spain
- Department of Surgery, Faculty of Medicine, Complutense University, 28040 Madrid, Spain
| | - Pilar Iniesta
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University, 28040 Madrid, Spain; (J.V.-V.); (S.T.)
- San Carlos Health Research Institute (IdISSC), 28040 Madrid, Spain; (M.P.-C.); (D.G.-G.); (A.O.-H.); (S.d.l.S.); (I.D.-S.); (J.D.); (D.R.); (J.-R.J.); (A.-M.G.-M.); (F.H.); (A.T.)
| |
Collapse
|
3
|
Ding T, Liu C, Li Z. The mycobiome in human cancer: analytical challenges, molecular mechanisms, and therapeutic implications. Mol Cancer 2025; 24:18. [PMID: 39815314 PMCID: PMC11734361 DOI: 10.1186/s12943-025-02227-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 01/06/2025] [Indexed: 01/18/2025] Open
Abstract
The polymorphic microbiome is considered a new hallmark of cancer. Advances in High-Throughput Sequencing have fostered rapid developments in microbiome research. The interaction between cancer cells, immune cells, and microbiota is defined as the immuno-oncology microbiome (IOM) axis. Fungal microbes (the mycobiome), although representing only ∼ 0.1-1% of the microbiome, are a critical immunologically active component of the tumor microbiome. Accumulating evidence suggests a possible involvement of commensal and pathogenic fungi in cancer initiation, progression, and treatment responsiveness. The tumor-associated mycobiome mainly consists of the gut mycobiome, the oral mycobiome, and the intratumoral mycobiome. However, the role of fungi in cancer remains poorly understood, and the diversity and complexity of analytical methods make it challenging to access this field. This review aims to elucidate the causal and complicit roles of mycobiome in cancer development and progression while highlighting the issues that need to be addressed in executing such research. We systematically summarize the advantages and limitations of current fungal detection and analysis methods. We enumerate and integrate these recent findings into our current understanding of the tumor mycobiome, accompanied by the prospect of novel and exhilarating clinical implications.
Collapse
Affiliation(s)
- Ting Ding
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, No. 20, Section 3, Renmin South Road, Chengdu, Sichuan Province, 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Chang Liu
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, No. 20, Section 3, Renmin South Road, Chengdu, Sichuan Province, 610041, China
| | - Zhengyu Li
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, No. 20, Section 3, Renmin South Road, Chengdu, Sichuan Province, 610041, China.
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China.
| |
Collapse
|
4
|
Luo Y, Li M, Luo D, Tang B. Gut Microbiota: An Important Participant in Childhood Obesity. Adv Nutr 2024; 16:100362. [PMID: 39733798 DOI: 10.1016/j.advnut.2024.100362] [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: 09/04/2024] [Revised: 12/19/2024] [Accepted: 12/23/2024] [Indexed: 12/31/2024] Open
Abstract
Increasing prevalence of childhood obesity has emerged as a critical global public health concern. Recent studies have challenged the previous belief that obesity was solely a result of excessive caloric intake. Alterations in early-life gut microbiota can contribute to childhood obesity through their influence on nutrient absorption and metabolism, initiation of inflammatory responses, and regulation of gut-brain communication. The gut microbiota is increasingly acknowledged to play a crucial role in human health, as certain beneficial bacteria have been scientifically proven to possess the capacity to reduce body fat content and enhance intestinal barrier function and their metabolic products to exhibit anti-inflammatory effect. Examples of such microbes include bifidobacteria, Akkermansia muciniphila, and Lactobacillus reuteri. In contrast, an increase in Enterobacteriaceae and propionate-producing bacteria (Prevotellaceae and Veillonellaceae) has been implicated in the induction of low-grade systemic inflammation and disturbances in lipid metabolism, which can predispose individuals to obesity. Studies have demonstrated that modulating the gut microbiota through diet, lifestyle changes, prebiotics, probiotics, or fecal microbiota transplantation may contribute to gut homeostasis and the management of obesity and its associated comorbidities. This review aimed to elucidate the impact of alterations in gut microbiota composition during early life on childhood obesity and explores the mechanisms by which gut microbiota contributes to the pathogenesis of obesity and specifically focused on recent advances in using short-chain fatty acids for regulating gut microbiota and ameliorating obesity. Additionally, it aimed to discuss the therapeutic strategies for childhood obesity from the perspective of gut microbiota, aiming to provide a theoretical foundation for interventions targeting pediatric obesity based on gut microbiota.
Collapse
Affiliation(s)
- Yu Luo
- Department of Pediatrics, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Maojun Li
- Department of Pediatrics, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Dan Luo
- Department of Pediatrics, School of Medicine and Life Science of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Binzhi Tang
- Department of Pediatrics, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China; Department of Pediatrics, School of Medicine and Life Science of Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
5
|
Ostrowska M, Nowosad K, Mikoluc B, Szczerba H, Komon-Janczara E. Changes in the Gut and Oral Microbiome in Children with Phenylketonuria in the Context of Dietary Restrictions-A Preliminary Study. Nutrients 2024; 16:3915. [PMID: 39599702 PMCID: PMC11597790 DOI: 10.3390/nu16223915] [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: 10/22/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Phenylketonuria (PKU) is a metabolic disorder that necessitates dietary restrictions, potentially impacting the composition of gut and oral microbiota. This study aimed to compare the microbiota composition between children with PKU and healthy controls. METHODS Using 16S rRNA gene sequencing, we analysed microbial communities at six phylogenetic levels. RESULTS Our findings revealed significant differences in the gut microbiota: Euryarchaeota was more abundant in controls (p = 0.01), while Bacilli and Lactobacillales were higher in PKU children (p = 0.019). Methanobacteriales were significantly elevated in controls (p = 0.01). At the genus and species levels, PKU children had higher Streptococcus and Eubacterium dolichum (p = 0.019, p = 0.015), whereas controls had more Barnesiella, Coprococcus, and Faecalibacterium prausnitzii (p = 0.014, p = 0.019, p = 0.014). In the oral microbiota, control children exhibited significantly higher Bacteroidetes (p = 0.032), while PKU children had increased Bacilli and Betaproteobacteria (p = 0.0079, p = 0.016). Streptococcus and Neisseria were more prevalent in PKU (p = 0.0079, p = 0.016). CONCLUSIONS These results suggest that PKU and its dietary management significantly alter the gut and oral microbiota composition. Understanding these microbial shifts could have implications for managing PKU and improving patient outcomes.
Collapse
Affiliation(s)
- Malgorzata Ostrowska
- Department of Biotechnology, Microbiology and Human Nutrition, University of Life Sciences in Lublin, 8 Skromna St., 20-704 Lublin, Poland; (M.O.)
| | - Karolina Nowosad
- Department of Biotechnology, Microbiology and Human Nutrition, University of Life Sciences in Lublin, 8 Skromna St., 20-704 Lublin, Poland; (M.O.)
| | - Bozena Mikoluc
- Department of Pediatrics, Rheumatology, Immunology and Metabolic Bone Diseases, Medical University of Bialystok, 15-274 Bialystok, Poland
| | - Hubert Szczerba
- Department of Biotechnology, Microbiology and Human Nutrition, University of Life Sciences in Lublin, 8 Skromna St., 20-704 Lublin, Poland; (M.O.)
| | - Elwira Komon-Janczara
- Department of Biotechnology, Microbiology and Human Nutrition, University of Life Sciences in Lublin, 8 Skromna St., 20-704 Lublin, Poland; (M.O.)
| |
Collapse
|
6
|
Zumkhawala-Cook A, Gallagher P, Raymann K. Diet affects reproductive development and microbiota composition in honey bees. Anim Microbiome 2024; 6:64. [PMID: 39501371 PMCID: PMC11539837 DOI: 10.1186/s42523-024-00350-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 10/23/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND Gut microbes are important to the health and fitness of many animals. Many factors have been shown to affect gut microbial communities including diet, lifestyle, and age. Most animals have very complex physiologies, lifestyles, and microbiomes, making it virtually impossible to disentangle what factors have the largest impact on microbiota composition. Honeybees are an excellent model to study host-microbe interactions due to their relatively simple gut microbiota, experimental tractability, and eusociality. Worker honey bees have distinct gut microbiota from their queen mothers despite being close genetic relatives and living in the same environment. Queens and workers differ in numerous ways including development, physiology, pheromone production, diet, and behavior. In the prolonged absence of a queen or Queen Mandibular Pheromones (QMP), some but not all workers will develop ovaries and become "queen-like". Using this inducible developmental change, we aimed to determine if diet and/or reproductive development impacts the gut microbiota of honey bee workers. RESULTS Microbiota-depleted newly emerged workers were inoculated with a mixture of queen and worker gut homogenates and reared under four conditions varying in diet and pheromone exposure. Three weeks post-emergence, workers were evaluated for ovary development and their gut microbiota communities were characterized. The proportion of workers with developed ovaries was increased in the absence of QMP but also when fed a queen diet (royal jelly). Overall, we found that diet, rather than reproductive development or pheromone exposure, led to more "queen-like" microbiota in workers. However, we revealed that diet alone cannot explain the microbiota composition of workers. CONCLUSION The hypothesis that reproductive development explains microbiota differences between queens and workers was rejected. We found evidence that diet is one of the main drivers of differences between the gut microbial community compositions of queens and workers but cannot fully explain the distinct microbiota of queens. Thus, we predict that behavioral and other physiological differences dictate microbiota composition in workers and queens. Our findings not only contribute to our understanding of the factors affecting the honey bee microbiota, which is important for bee health, but also illustrate the versatility and benefits of utilizing honeybees as a model system to study host-microbe interactions.
Collapse
Affiliation(s)
- Anjali Zumkhawala-Cook
- Department of Plant and Microbial Biology, North Carolina State University, Raleigh, North Carolina, USA
- Department of Biochemistry and Molecular Biology, Kenyon College, Gambier, Ohio, USA
| | - Patrick Gallagher
- Department of Plant and Microbial Biology, North Carolina State University, Raleigh, North Carolina, USA
| | - Kasie Raymann
- Department of Plant and Microbial Biology, North Carolina State University, Raleigh, North Carolina, USA.
| |
Collapse
|
7
|
Probul N, Huang Z, Saak CC, Baumbach J, List M. AI in microbiome-related healthcare. Microb Biotechnol 2024; 17:e70027. [PMID: 39487766 PMCID: PMC11530995 DOI: 10.1111/1751-7915.70027] [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: 04/30/2024] [Accepted: 09/23/2024] [Indexed: 11/04/2024] Open
Abstract
Artificial intelligence (AI) has the potential to transform clinical practice and healthcare. Following impressive advancements in fields such as computer vision and medical imaging, AI is poised to drive changes in microbiome-based healthcare while facing challenges specific to the field. This review describes the state-of-the-art use of AI in microbiome-related healthcare. It points out limitations across topics such as data handling, AI modelling and safeguarding patient privacy. Furthermore, we indicate how these current shortcomings could be overcome in the future and discuss the influence and opportunities of increasingly complex data on microbiome-based healthcare.
Collapse
Affiliation(s)
- Niklas Probul
- Institute for Computational Systems BiologyUniversity of HamburgHamburgGermany
| | - Zihua Huang
- Data Science in Systems Biology, TUM School of Life SciencesTechnical University of MunichFreisingGermany
| | | | - Jan Baumbach
- Institute for Computational Systems BiologyUniversity of HamburgHamburgGermany
- Computational Biomedicine Lab, Department of Mathematics and Computer ScienceUniversity of Southern DenmarkOdenseDenmark
| | - Markus List
- Data Science in Systems Biology, TUM School of Life SciencesTechnical University of MunichFreisingGermany
- Munich Data Science InstituteTechnical University of MunichGarchingGermany
| |
Collapse
|
8
|
Matsuki T, Nakamura S, Nishiyama M, Narimatsu H. Holistic Evaluation of the Gut Microbiota through Data Envelopment Analysis: A Cross-Sectional Study. Curr Dev Nutr 2024; 8:104469. [PMID: 39524216 PMCID: PMC11550754 DOI: 10.1016/j.cdnut.2024.104469] [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: 09/19/2024] [Accepted: 09/20/2024] [Indexed: 11/16/2024] Open
Abstract
Background The gut microbiome plays a crucial role in human health, but maintaining a healthy gut microbiome remains challenging. Current approaches often focus on individual components rather than providing a holistic assessment. Objectives To introduce and evaluate a novel approach using data envelopment analysis (DEA) for assessing gut microbiota efficiency and identifying potential targets for personalized interventions. Methods We conducted a cross-sectional analysis of 577 participants from the Kanagawa "ME-BYO" Prospective Cohort Study. Lifestyle factors and gut microbiota composition were assessed. DEA was employed to calculate an efficiency score for each participant, incorporating multiple inputs (lifestyle factors) and outputs (gut microbiotas). This score represents how efficiently an individual's lifestyle factors contribute to their gut microbiota composition. Tobit regression analysis was used to assess associations between efficiency scores and demographic and health-related factors. Results The mean efficiency score was 0.86, with 14.2% of participants classified as efficient. Efficiency scores showed positive correlations with alcohol intake and Faith's phylogenetic diversity. Tobit regression analysis revealed significant associations between efficiency scores and sex, fat intake, and yogurt consumption. DEA identified specific targets for improving gut microbiota composition in inefficient individuals. Conclusions This study demonstrates the potential of DEA as a tool for evaluating gut microbiota efficiency and providing personalized recommendations for microbiota optimization. This approach could lead to more effective strategies for optimizing gut health across diverse populations.
Collapse
Affiliation(s)
- Taizo Matsuki
- Graduate School of Health Innovation, Kanagawa University of Human Services, Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, Japan
- Cancer Prevention and Control Division, Kanagawa Cancer Center Research Institute, Asahi-ku, Yokohama, Kanagawa, Japan
| | - Sho Nakamura
- Graduate School of Health Innovation, Kanagawa University of Human Services, Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, Japan
- Cancer Prevention and Control Division, Kanagawa Cancer Center Research Institute, Asahi-ku, Yokohama, Kanagawa, Japan
- Department of Medical Genetics, Kanagawa Cancer Center, Nakao, Asahi-ku, Yokohama, Kanagawa, Japan
| | - Minami Nishiyama
- Graduate School of Health Innovation, Kanagawa University of Human Services, Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, Japan
- Cancer Prevention and Control Division, Kanagawa Cancer Center Research Institute, Asahi-ku, Yokohama, Kanagawa, Japan
| | - Hiroto Narimatsu
- Graduate School of Health Innovation, Kanagawa University of Human Services, Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, Japan
- Cancer Prevention and Control Division, Kanagawa Cancer Center Research Institute, Asahi-ku, Yokohama, Kanagawa, Japan
- Department of Medical Genetics, Kanagawa Cancer Center, Nakao, Asahi-ku, Yokohama, Kanagawa, Japan
| |
Collapse
|
9
|
Wang L, Gan D, Guo W, Zhu Y, Zhang M, Zhou A, Chen G, Wu W. Alteration of the gut microbiota changes during the one-year Antarctic deployment in a group of Chinese Han population. BMC Microbiol 2024; 24:427. [PMID: 39443844 PMCID: PMC11515739 DOI: 10.1186/s12866-024-03569-x] [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: 04/06/2024] [Accepted: 10/07/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND It is common knowledge that people's intestinal microbiota is significantly influenced by the external environment. Although the Antarctic continent has been discovered for nearly 200 years, it is still unclear how this environment affects the human intestinal microbiota, especially that of the Chinese Han population. METHODS Twelve explorers underwent a one-year Antarctic deployment from December 2017 to December 2018. The gut microbiota and clinical indexes at five time points, including two months (T1), five months (T2), eight months (T3), 11 months (T4) of residence in Antarctica and 7 months after returning to China (T5), were investigated. RESULTS The intestinal microbiota of the participants was changed after one-year Antarctic deployment even after they left Antarctica. For the microbiota tested after returning to China (T5), the amount of Citrobacter, Akkermansia and conditional pathogens such as Escherichia-Shigella increased significantly (P < 0.05). The concentration of the major biochemical indicators in the serum exhibited an increasing trend before T3, and decreased significantly at T4. When tested again at T5, most of the serum concentrations increased, only 5-hydroxytryptamine was significantly decreased. Spearman correlation analysis showed the change in the relative abundance of Anaerotruncus was negatively associated with the changes in the concentration of total thyroxine, alanine transaminase, γ-glutamyl transpeptidase, serum cystatin C and apolipoprotein A1. The relative abundance change in Citrobacter was positively associated with the change in the concentration of uric acid. CONCLUSION By objectively analyzing the influence of the Antarctic environment on the change of intestinal microbiota, we were able to provide theoretical support for subsequent Antarctic related research.
Collapse
Affiliation(s)
- Lili Wang
- Department of Laboratory Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Laboratory Medicine, Zhoushan Women and Children Hospital, Zhoushan, China
| | - Di Gan
- Department of Emergency Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wenzheng Guo
- Department of Laboratory Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yunxia Zhu
- Department of Laboratory Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Min Zhang
- Department of Laboratory Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Aiping Zhou
- Department of Laboratory Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Guoting Chen
- Department of Emergency Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Wenjuan Wu
- Department of Laboratory Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
10
|
Muraglia M, Faienza MF, Tardugno R, Clodoveo ML, Matias De la Cruz C, Bermúdez FG, Munizaga MG, Valencia L, Corbo F, Orellana-Manzano A. Breastfeeding: science and knowledge in pediatric obesity prevention. Front Med (Lausanne) 2024; 11:1430395. [PMID: 39399112 PMCID: PMC11466875 DOI: 10.3389/fmed.2024.1430395] [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: 05/09/2024] [Accepted: 09/16/2024] [Indexed: 10/15/2024] Open
Abstract
The increasing prevalence of childhood obesity worldwide is a significant concern due to its link to severe health issues in adulthood, such as non-communicable diseases (NCDs). To address this issue, this review evaluates the effectiveness of various preventive measures for childhood obesity, focusing on maternal nutrition and breastfeeding. The study underscores the criticality of the periconceptional period, where the diets of both parents can influence epigenetic modifications that impact the child's metabolic pathways and obesity risks. Breastfeeding is a potent protective mechanism against early-onset obesity, significantly enhancing the infant's metabolic and immune health by modifying DNA methylation and gene expression. Furthermore, the perspective underscores the significance of the Mediterranean diet during the periconceptional period and lactation. This diet can effectively prevent gestational complications and improve breast milk quality, fostering optimal infant development. Recognizing that obesity results from genetic, epigenetic, environmental, and social factors, the paper advocates for a comprehensive, multidisciplinary approach from the earliest stages of life. This approach champions a balanced maternal diet, exclusive breastfeeding, and timely introduction to complementary foods. In conclusion, addressing pediatric obesity requires a multifaceted strategy emphasizing improving prenatal and postnatal nutrition. Further research is necessary to understand the epigenetic mechanisms influenced by nutrition and their long-term effects on children's health. This will help refine interventions that curb the obesity epidemic among future generations.
Collapse
Affiliation(s)
- Marilena Muraglia
- Department of Pharmacy - Drug Science, University of Bari ‘Aldo Moro’, Bari, Italy
| | - Maria Felicia Faienza
- Pediatric Unit, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari “A. Moro”, Bari, Italy
| | - Roberta Tardugno
- Department of Pharmacy - Drug Science, University of Bari ‘Aldo Moro’, Bari, Italy
| | - Maria Lisa Clodoveo
- Interdisciplinary Department of Medicine, School of Medicine, University of Bari-Aldo Moro, Bari, Italy
| | - Carmen Matias De la Cruz
- Laboratorio Para Investigaciones Biomédicas, Facultad de Ciencias de la Vida, Escuela Superior Politécnica del Litoral, ESPOL, Guayaquil, Ecuador
| | - Fátima German Bermúdez
- Laboratorio Para Investigaciones Biomédicas, Facultad de Ciencias de la Vida, Escuela Superior Politécnica del Litoral, ESPOL, Guayaquil, Ecuador
| | - María Gabriela Munizaga
- Laboratorio Para Investigaciones Biomédicas, Facultad de Ciencias de la Vida, Escuela Superior Politécnica del Litoral, ESPOL, Guayaquil, Ecuador
| | - Luz Valencia
- Licenciatura en Nutrición y Dietética, Facultad de Ciencias de la Vida, Escuela Superior Politécnica del Litoral, ESPOL, Guayaquil, Ecuador
| | - Filomena Corbo
- Department of Pharmacy - Drug Science, University of Bari ‘Aldo Moro’, Bari, Italy
| | - Andrea Orellana-Manzano
- Laboratorio Para Investigaciones Biomédicas, Facultad de Ciencias de la Vida, Escuela Superior Politécnica del Litoral, ESPOL, Guayaquil, Ecuador
| |
Collapse
|
11
|
Erb C, Erb C, Kazakov A, Kapanova G, Weisser B. Lifestyle Changes in Aging and their Potential Impact on POAG. Klin Monbl Augenheilkd 2024. [PMID: 39191386 DOI: 10.1055/a-2372-3505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
Primary open angle glaucoma is a primary mitochondrial disease with oxidative stress triggering neuroinflammation, eventually resulting in neurodegeneration. This affects many other areas of the brain in addition to the visual system. Aging also leads to inflammaging - a low-grade chronic inflammatory reaction in mitochondrial dysfunction, so these inflammatory processes overlap in the aging process and intensify pathophysiological processes associated with glaucoma. Actively counteracting these inflammatory events involves optimising treatment for any manifest systemic diseases while maintaining chronobiology and improving the microbiome. Physical and mental activity also provides support. This requires a holistic approach towards optimising neurodegeneration treatment in primary open angle glaucoma in addition to reducing intraocular pressure according personalised patient targets.
Collapse
Affiliation(s)
- Carl Erb
- Augenklinik am Wittenbergplatz, Berlin, Deutschland
| | | | - Avaz Kazakov
- External Relations and Development, Salymbekov University, Bishkek, Kyrgyzstan
| | - Gulnara Kapanova
- Medical Faculty of Medicine, Al-Farabi Kazakh National University, Almaty, Kazakhstan
| | | |
Collapse
|
12
|
Maitin-Shepard M, O'Tierney-Ginn P, Kraneveld AD, Lyall K, Fallin D, Arora M, Fasano A, Mueller NT, Wang X, Caulfield LE, Dickerson AS, Diaz Heijtz R, Tarui T, Blumberg JB, Holingue C, Schmidt RJ, Garssen J, Almendinger K, Lin PID, Mozaffarian D. Food, nutrition, and autism: from soil to fork. Am J Clin Nutr 2024; 120:240-256. [PMID: 38677518 DOI: 10.1016/j.ajcnut.2024.04.020] [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: 05/16/2023] [Revised: 04/18/2024] [Accepted: 04/22/2024] [Indexed: 04/29/2024] Open
Abstract
Food and nutrition-related factors have the potential to impact development of autism spectrum disorder (ASD) and quality of life for people with ASD, but gaps in evidence exist. On 10 November 2022, Tufts University's Friedman School of Nutrition Science and Policy and Food and Nutrition Innovation Institute hosted a 1-d meeting to explore the evidence and evidence gaps regarding the relationships of food and nutrition with ASD. This meeting report summarizes the presentations and deliberations from the meeting. Topics addressed included prenatal and child dietary intake, the microbiome, obesity, food-related environmental exposures, mechanisms and biological processes linking these factors and ASD, food-related social factors, and data sources for future research. Presentations highlighted evidence for protective associations with prenatal folic acid supplementation and ASD development, increases in risk of ASD with maternal gestational obesity, and the potential for exposure to environmental contaminants in foods and food packaging to influence ASD development. The importance of the maternal and child microbiome in ASD development or ASD-related behaviors in the child was reviewed, as was the role of discrimination in leading to disparities in environmental exposures and psychosocial factors that may influence ASD. The role of child diet and high prevalence of food selectivity in children with ASD and its association with adverse outcomes were also discussed. Priority evidence gaps identified by participants include further clarifying ASD development, including biomarkers and key mechanisms; interactions among psychosocial, social, and biological determinants; interventions addressing diet, supplementation, and the microbiome to prevent and improve quality of life for people with ASD; and mechanisms of action of diet-related factors associated with ASD. Participants developed research proposals to address the priority evidence gaps. The workshop findings serve as a foundation for future prioritization of scientific research to address evidence gaps related to food, nutrition, and ASD.
Collapse
Affiliation(s)
| | | | - Aletta D Kraneveld
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, the Netherlands; Center for Neurogenomics and Cognitive Research, VU University, Amsterdam, the Netherlands
| | - Kristen Lyall
- AJ Drexel Autism Institute, Drexel University, Philadelphia, PA, United States
| | - Daniele Fallin
- Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Manish Arora
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Alessio Fasano
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA, United States; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Noel T Mueller
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Xiaobin Wang
- Department of Population, Family and Reproductive Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Laura E Caulfield
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Aisha S Dickerson
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | | | - Tomo Tarui
- Department of Pediatrics, Hasbro Children's Hospital, Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Jeffrey B Blumberg
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, United States
| | - Calliope Holingue
- Center for Autism Services, Science and Innovation, Kennedy Krieger Institute and Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Rebecca J Schmidt
- Department of Public Health Sciences, the MIND Institute, University of California Davis, Davis, CA, United States
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | - Katherine Almendinger
- Department of Health Policy and Management, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Pi-I Debby Lin
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, United States
| | - Dariush Mozaffarian
- Food is Medicine Institute, Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, United States.
| |
Collapse
|
13
|
Hu Y, Zhang Y, Cui X, Wang D, Hu Y, Wang C. Structure-function relationship and biological activity of polysaccharides from mulberry leaves: A review. Int J Biol Macromol 2024; 268:131701. [PMID: 38643920 DOI: 10.1016/j.ijbiomac.2024.131701] [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: 08/28/2023] [Revised: 03/12/2024] [Accepted: 04/18/2024] [Indexed: 04/23/2024]
Abstract
Mulberry (Latin name "Morus alba L.") is a perennial deciduous tree in the family of Moraceae, widely distributed around the world. In China, mulberry is mainly distributed in the south and the Yangtze River basin. Its leaves can be harvested 3-6 times a year, which has a great resource advantage. Mulberry leaves are regarded as the homology of medicine and food traditional Chinese medicine (TCM). Polysaccharides, as its main active ingredients, have various effects, such as antioxidant, hypoglycemic, hepatoprotective, and immunomodulatory. This review summarizes the research progress in the extraction, purification, structural characterization, and structure-function relationship of polysaccharides from mulberry leaves in the last decade, hoping to provide a reference for the subsequent development and market application of polysaccharides from mulberry leaves.
Collapse
Affiliation(s)
- Yexian Hu
- College of Biology, Food & Environment, Hefei University, Hefei 230601, PR China
| | - Yan Zhang
- College of Biology, Food & Environment, Hefei University, Hefei 230601, PR China
| | - Xiaoao Cui
- College of Biology, Food & Environment, Hefei University, Hefei 230601, PR China
| | - Dongsheng Wang
- College of Biology, Food & Environment, Hefei University, Hefei 230601, PR China
| | - Yong Hu
- Agricultural Products Processing Institute, Anhui Academy of Agricultural Sciences, Hefei 230031, PR China
| | - Chuyan Wang
- College of Biology, Food & Environment, Hefei University, Hefei 230601, PR China; Key Laboratory of Berry Processing and Resource Comprehensive Utilization, Hefei University, Hefei 230601, PR China.
| |
Collapse
|
14
|
Ravella S. Association between oral nutrition and inflammation after intestinal transplantation. Hum Immunol 2024; 85:110809. [PMID: 38724327 DOI: 10.1016/j.humimm.2024.110809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 04/27/2024] [Accepted: 05/01/2024] [Indexed: 06/04/2024]
Abstract
Intestinal transplantation (Itx) can be a life-saving treatment for certain patient populations, including those patients with intestinal failure (IF) who develop life-threatening complications due to the use of parenteral nutrition (PN). Most patients who have undergone Itx are eventually able to tolerate a full oral diet. However, little guidance or consensus exists regarding optimizing the specific components of an oral diet for Itx patients, including macronutrients, micronutrients and dietary patterns. While oral dietary prescriptions have moved to the forefront of primary and preventive care, this movement has yet to occur across the field of organ transplantation. Evidence to date points to the role of systemic chronic inflammation (SCI) in a wide variety of chronic diseases as well as post-transplant graft dysfunction. This review will discuss current trends in oral nutrition for Itx patients and also offer novel insights into nutritional management techniques that may help to decrease SCI and chronic disease risk as well as optimize graft function.
Collapse
|
15
|
Zakiudin DP, Thyssen JP, Zachariae C, Videm V, Øien T, Simpson MR. Filaggrin Mutation Status and Prevention of Atopic Dermatitis with Maternal Probiotic Supplementation. Acta Derm Venereol 2024; 104:adv24360. [PMID: 38655655 PMCID: PMC11064679 DOI: 10.2340/actadv.v104.24360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 03/22/2024] [Indexed: 04/26/2024] Open
Abstract
The World Allergy Organization recommends probiotics in the prevention of atopic dermatitis in high-risk populations. Mutations in the filaggrin gene (FLG) result in an increased risk of atopic dermatitis through disruption of the skin keratin layer. This exploratory study investigated whether the preventive effect of maternal probiotics was evident in children with and without FLG mutations. DNA was collected from children (n = 228) from the Probiotic in the Prevention of Allergy among Children in Trondheim (ProPACT) study. Samples were analysed for 3 common FLG mutations (R501X, R2447X, and 2282del4). Overall, 7% of children had heterozygous FLG mutations; each child had only one of the 3 mutations. Mutation status had no association with atopic dermatitis (RR = 1.1; 95% CI 0.5 to 2.3). The risk ratio (RR) for having atopic dermatitis following maternal probiotics was 0.6 (95% CI 0.4 to 0.9) and RR was similar if the child expressed an FLG mutation (RR = 0.6; 95% CI 0.1 to 4.1) or wildtype FLG (RR = 0.6; 95% CI 0.4 to 0.9). The preventive effect of probiotics for atopic dermatitis was also evident in children without FLG mutation. Larger confirmatory studies are needed.
Collapse
Affiliation(s)
- Dinastry Pramadita Zakiudin
- Department of Public Health and Nursing, NTNU - Norwegian University of Science and Technology, Trondheim, Norway; Clinic for Laboratory Medicine, St Olavs Hospital, Trondheim, Norway.
| | - Jacob P Thyssen
- Department of Dermatology and Venereology, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Claus Zachariae
- Department of Clinical Medicine, University Hospital of Copenhagen Gentofte, Hellerup, Denmark; Department of Dermatology and Allergy, University Hospital of Copenhagen Gentofte, Hellerup, Denmark
| | - Vibeke Videm
- Department of Clinical and Molecular Medicine, NTNU - Norwegian University of Science and Technology, Trondheim, Norway; St. Olavs Hospital, Trondheim University Hospital, Department of Immunology and Transfusion Medicine, Trondheim, Norway
| | - Torbjørn Øien
- Department of Public Health and Nursing, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
| | - Melanie Rae Simpson
- Department of Public Health and Nursing, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
16
|
Watai K, Suda W, Kurokawa R, Sekiya K, Hayashi H, Iwata M, Nagayama K, Nakamura Y, Hamada Y, Kamide Y, Fukutomi Y, Nakabayashi T, Tanaka K, Kamita M, Taniguchi M, Hattori M. Metagenomic gut microbiome analysis of Japanese patients with multiple chemical sensitivity/idiopathic environmental intolerance. BMC Microbiol 2024; 24:84. [PMID: 38468206 PMCID: PMC10926566 DOI: 10.1186/s12866-024-03239-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 02/26/2024] [Indexed: 03/13/2024] Open
Abstract
BACKGROUND Although the pathology of multiple chemical sensitivity (MCS) is unknown, the central nervous system is reportedly involved. The gut microbiota is important in modifying central nervous system diseases. However, the relationship between the gut microbiota and MCS remains unclear. This study aimed to identify gut microbiota variations associated with MCS using shotgun metagenomic sequencing of fecal samples. METHODS We prospectively recruited 30 consecutive Japanese female patients with MCS and analyzed their gut microbiomes using shotgun metagenomic sequencing. The data were compared with metagenomic data obtained from 24 age- and sex-matched Japanese healthy controls (HC). RESULTS We observed no significant difference in alpha and beta diversity of the gut microbiota between the MCS patients and HC. Focusing on the important changes in the literatures, at the genus level, Streptococcus, Veillonella, and Akkermansia were significantly more abundant in MCS patients than in HC (p < 0.01, p < 0.01, p = 0.01, respectively, fold change = 4.03, 1.53, 2.86, respectively). At the species level, Akkermansia muciniphila was significantly more abundant (p = 0.02, fold change = 3.3) and Faecalibacterium prausnitzii significantly less abundant in MCS patients than in HC (p = 0.03, fold change = 0.53). Functional analysis revealed that xylene and dioxin degradation pathways were significantly enriched (p < 0.01, p = 0.01, respectively, fold change = 1.54, 1.46, respectively), whereas pathways involved in amino acid metabolism and synthesis were significantly depleted in MCS (p < 0.01, fold change = 0.96). Pathways related to antimicrobial resistance, including the two-component system and cationic antimicrobial peptide resistance, were also significantly enriched in MCS (p < 0.01, p < 0.01, respectively, fold change = 1.1, 1.2, respectively). CONCLUSIONS The gut microbiota of patients with MCS shows dysbiosis and alterations in bacterial functions related to exogenous chemicals and amino acid metabolism and synthesis. These findings may contribute to the further development of treatment for MCS. TRIAL REGISTRATION This study was registered with the University Hospital Medical Information Clinical Trials Registry as UMIN000031031. The date of first trial registration: 28/01/2018.
Collapse
Affiliation(s)
- Kentaro Watai
- Center for Immunology and Allergy, Shonan Kamakura General Hospital, 1370-1 Okamoto, Kamakura, Kanagawa, 247-8533, Japan.
- Clinical Research Center for Allergy and Rheumatology, NHO Sagamihara National Hospital, Sagamihara, Kanagawa, Japan.
| | - Wataru Suda
- Laboratory for Microbiome Sciences, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Rina Kurokawa
- Laboratory for Microbiome Sciences, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Kiyoshi Sekiya
- Clinical Research Center for Allergy and Rheumatology, NHO Sagamihara National Hospital, Sagamihara, Kanagawa, Japan
| | - Hiroaki Hayashi
- Clinical Research Center for Allergy and Rheumatology, NHO Sagamihara National Hospital, Sagamihara, Kanagawa, Japan
| | - Maki Iwata
- Clinical Research Center for Allergy and Rheumatology, NHO Sagamihara National Hospital, Sagamihara, Kanagawa, Japan
| | - Kisako Nagayama
- Clinical Research Center for Allergy and Rheumatology, NHO Sagamihara National Hospital, Sagamihara, Kanagawa, Japan
| | - Yuto Nakamura
- Clinical Research Center for Allergy and Rheumatology, NHO Sagamihara National Hospital, Sagamihara, Kanagawa, Japan
| | - Yuto Hamada
- Clinical Research Center for Allergy and Rheumatology, NHO Sagamihara National Hospital, Sagamihara, Kanagawa, Japan
| | - Yosuke Kamide
- Clinical Research Center for Allergy and Rheumatology, NHO Sagamihara National Hospital, Sagamihara, Kanagawa, Japan
| | - Yuma Fukutomi
- Clinical Research Center for Allergy and Rheumatology, NHO Sagamihara National Hospital, Sagamihara, Kanagawa, Japan
| | | | - Kosei Tanaka
- H.U. Group Research Institute G.K., Akiruno, Tokyo, Japan
| | | | - Masami Taniguchi
- Clinical Research Center for Allergy and Rheumatology, NHO Sagamihara National Hospital, Sagamihara, Kanagawa, Japan
| | - Masahira Hattori
- Laboratory for Microbiome Sciences, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
- Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| |
Collapse
|
17
|
Lenover MB, Shenk MK. Evolutionary medicine approaches to chronic disease: The case of irritable bowel syndrome. Evol Anthropol 2024; 33:e22010. [PMID: 37909359 DOI: 10.1002/evan.22010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 10/13/2023] [Accepted: 10/16/2023] [Indexed: 11/03/2023]
Abstract
Irritable bowel syndrome (IBS), a gastrointestinal disease, is a global phenomenon correlated with industrialization. We propose that an evolutionary medicine approach is useful to understand this disease from an ultimate perspective and conducted a scoping literature review to synthesize the IBS literature within this framework. Our review suggests five potential evolutionary hypotheses for the cause of IBS, including (a) a dietary mismatch accompanying a nutritional transition, (b) an early hygienic life environment leading to the immune system and microbiotic changes, (c) an outcome of decreased physical activity, (d) a response to changes in environmental light-dark cycles, and (e) an artifact of an evolved fight or flight response. We find key limitations in the available data needed to understand early life, nutritional, and socioeconomic experiences that would allow us to understand evolutionarily relevant risk factors and identify a need for further empirical research to distinguish potential causes and test evolutionary hypotheses.
Collapse
Affiliation(s)
- Makenna B Lenover
- Department of Anthropology, The Pennsylvania State University, State College, Pennsylvania, USA
| | - Mary K Shenk
- Department of Anthropology, The Pennsylvania State University, State College, Pennsylvania, USA
| |
Collapse
|
18
|
Mohanan A, Harilal SL, Plakkot B, Pottakkat B, Kanakkaparambil R. Nutritional Epigenetics and Gut Microbiome. EPIGENETICS AND HUMAN HEALTH 2024:121-159. [DOI: 10.1007/978-3-031-54215-2_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
19
|
Yan H, Liang X, Luo H, Tang X, Xiao X. Association between gut microbiota, microbial network, and immunity in pregnancy with a focus on specific bacterial clusters. Front Microbiol 2023; 14:1314257. [PMID: 38156011 PMCID: PMC10753819 DOI: 10.3389/fmicb.2023.1314257] [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: 10/10/2023] [Accepted: 11/28/2023] [Indexed: 12/30/2023] Open
Abstract
Background The community characteristics of the gut microbiota are not well defined and are not as widely studied as the functions of individual bacteria. This study aims to investigate the community composition of intestinal flora in women of childbearing age by conducting cluster analysis of gut microbiota and analyzing the relationship between different clusters and immune status. Methods A total of 45 women of childbearing age were recruited in the study, including 15 non-pregnant women and 30 women in late pregnancy, and stool samples were collected twice during the third trimester, specifically at 32 weeks and at full term. The gut microbiota data was analyzed using 16S rRNA amplicon sequencing. Partitioning Around Medoids algorithm was employed to assess microbial clustering patterns. Microbial network for each cluster was performed and plasm cytokines were measured to analyze the relationship between specific genera and immune state in clusters. Results There were three distinct clusters of intestinal community composition in women of childbearing age. Cluster 1 (PAM_1) was characterized by a high abundance of Bacteroides, while cluster 2 (PAM_2) showed higher levels of Bifidobacterium and Blautia, along with a significantly increased Firmicutes to Bacteroidota ratio. Cluster 3 (PAM_3) displayed a high abundance of Escherichia-shigella. PAM_1 was the most dominant cluster in non-pregnant women, and this dominant cluster was also one of the main in late pregnancy. At full term, the majority of subjects retained the same cluster as at 32 weeks, while a few experienced a shift. The microbial correlation networks differed across the three clusters, with PAM_1 exhibiting higher modularity and fewer connections. Analysis of the correlation between genera and plasma cytokines showed significant differences in their associations with cytokines between pregnancy and nonpregnancy within the same cluster, and the same genera had different effects in different clusters. Conclusion Women of childbearing age exhibit three distribution patterns of gut microbiota, and the intestinal clusters reshaped during late pregnancy in a small population. Different clusters may have diverse immunomodulatory effects in different physiological states. When studying the gut microbiome during pregnancy, it is crucial to consider the cluster differences within healthy women.
Collapse
Affiliation(s)
- Hao Yan
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xinyuan Liang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Department of Obstetrics, The Second Clinical Medical College (Shenzhen People's Hospital), Jinan University, Shenzhen, China
| | - Huijuan Luo
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xiaomei Tang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xiaomin Xiao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
20
|
Ubaldi F, Frangella C, Volpini V, Fortugno P, Valeriani F, Romano Spica V. Systematic Review and Meta-Analysis of Dietary Interventions and Microbiome in Phenylketonuria. Int J Mol Sci 2023; 24:17428. [PMID: 38139256 PMCID: PMC10744015 DOI: 10.3390/ijms242417428] [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: 10/22/2023] [Revised: 11/25/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023] Open
Abstract
Inborn errors of metabolism (IEMs) comprise a diverse group of monogenic disorders caused by enzyme deficiencies that result either in a toxic accumulation of metabolic intermediates or a shortage of essential end-products. Certain IEMs, like phenylketonuria (PKU), necessitate stringent dietary intervention that could lead to microbiome dysbiosis, thereby exacerbating the clinical phenotype. The objective of this systematic review was to examine the impact of PKU therapies on the intestinal microbiota. This research was conducted following the PRISMA Statement, with data from PubMed, Scopus, ScienceDirect, and Web of Science. A total of 18 articles meeting the inclusion criteria were published from 2011 to 2022. Significant reductions in several taxonomic groups in individuals with PKU when compared to the control group were detected in a quantitative analysis conducted across seven studies. The meta-analysis synthesis indicates a contrast in biodiversity between PKU subjects and the control population. Additionally, the meta-regression results, derived from the Bacillota/Bacteroidota ratio data, suggest a potential influence of diet in adult PKU populations (p = 0.004). It is worth noting that the limited number of studies calls for further research and analysis in this area. Our findings indicate the necessity of enhancing understanding of microbiota variability in reaction to treatments among PKU subjects to design tailored therapeutic and nutritional interventions to prevent complications resulting from microbiota disruption.
Collapse
Affiliation(s)
- Francesca Ubaldi
- Department of Movement, Human, and Health Sciences, University of Rome “Foro Italico”, 00135 Rome, Italy; (F.U.); (C.F.); (V.V.); (V.R.S.)
| | - Claudia Frangella
- Department of Movement, Human, and Health Sciences, University of Rome “Foro Italico”, 00135 Rome, Italy; (F.U.); (C.F.); (V.V.); (V.R.S.)
| | - Veronica Volpini
- Department of Movement, Human, and Health Sciences, University of Rome “Foro Italico”, 00135 Rome, Italy; (F.U.); (C.F.); (V.V.); (V.R.S.)
| | - Paola Fortugno
- Department of Human Sciences for the Promotion of Quality of Life, University San Raffaele, Via di Val Cannuta 247, 00166 Rome, Italy;
- Human Functional Genomics Laboratory, IRCCS San Raffaele Roma, Via di Val Cannuta 247, 00166 Rome, Italy
| | - Federica Valeriani
- Department of Movement, Human, and Health Sciences, University of Rome “Foro Italico”, 00135 Rome, Italy; (F.U.); (C.F.); (V.V.); (V.R.S.)
| | - Vincenzo Romano Spica
- Department of Movement, Human, and Health Sciences, University of Rome “Foro Italico”, 00135 Rome, Italy; (F.U.); (C.F.); (V.V.); (V.R.S.)
| |
Collapse
|
21
|
Galeana-Cadena D, Gómez-García IA, Lopez-Salinas KG, Irineo-Moreno V, Jiménez-Juárez F, Tapia-García AR, Boyzo-Cortes CA, Matías-Martínez MB, Jiménez-Alvarez L, Zúñiga J, Camarena A. Winds of change a tale of: asthma and microbiome. Front Microbiol 2023; 14:1295215. [PMID: 38146448 PMCID: PMC10749662 DOI: 10.3389/fmicb.2023.1295215] [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: 09/15/2023] [Accepted: 11/15/2023] [Indexed: 12/27/2023] Open
Abstract
The role of the microbiome in asthma is highlighted, considering its influence on immune responses and its connection to alterations in asthmatic patients. In this context, we review the variables influencing asthma phenotypes from a microbiome perspective and provide insights into the microbiome's role in asthma pathogenesis. Previous cohort studies in patients with asthma have shown that the presence of genera such as Bifidobacterium, Lactobacillus, Faecalibacterium, and Bacteroides in the gut microbiome has been associated with protection against the disease. While, the presence of other genera such as Haemophilus, Streptococcus, Staphylococcus, and Moraxella in the respiratory microbiome has been implicated in asthma pathogenesis, indicating a potential link between microbial dysbiosis and the development of asthma. Furthermore, respiratory infections have been demonstrated to impact the composition of the upper respiratory tract microbiota, increasing susceptibility to bacterial diseases and potentially triggering asthma exacerbations. By understanding the interplay between the microbiome and asthma, valuable insights into disease mechanisms can be gained, potentially leading to the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- David Galeana-Cadena
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
| | - Itzel Alejandra Gómez-García
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Karen Gabriel Lopez-Salinas
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Valeria Irineo-Moreno
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Fabiola Jiménez-Juárez
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Alan Rodrigo Tapia-García
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
- Red de Medicina para la Educación, el Desarrollo y la Investigación Científica de Iztacala, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Carlos Alberto Boyzo-Cortes
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
| | - Melvin Barish Matías-Martínez
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Luis Jiménez-Alvarez
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
| | - Joaquín Zúñiga
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Angel Camarena
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
| |
Collapse
|
22
|
Zhao WL, Xu D, Wang JS. Torachrysone-8-O-β-d-glucoside mediates anti-inflammatory effects by blocking aldose reductase-catalyzed metabolism of lipid peroxidation products. Biochem Pharmacol 2023; 218:115931. [PMID: 37981172 DOI: 10.1016/j.bcp.2023.115931] [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: 10/12/2023] [Revised: 11/14/2023] [Accepted: 11/15/2023] [Indexed: 11/21/2023]
Abstract
Aldose reductase (AR) is an important enzyme involved in the reduction of various aldehyde and carbonyl compounds, including the highly reactive and toxic 4-hydroxynonenal (4-HNE), which has been linked to the progression of various pathologies such as atherosclerosis, hyperglycemia, inflammation, and tumors. AR inhibitors have potential therapeutic benefits for these diseases by reducing lipid peroxidation and mitigating the harmful effects of reactive aldehydes. In this study, we found that torachrysone-8-O-β-d-glucoside (TG), a natural product isolated from Polygonum multiflorum Thunb., functions as an effective inhibitor of AR, exhibiting potent effects in clearing reactive aldehydes and reducing inflammation. TG up-regulated the mRNA levels of several antioxidant factors downstream of NRF2, especially glutathione S-transferase (GST), which is significantly increased, thus detoxifying 4-HNE by facilitating the conjugation of 4-HNE to glutathione, forming glutathione-4-hydroxynonenal (GS-HNE). By employing a combination of molecular docking, cellular thermal shift assay, and enzyme activity experiments, we demonstrated that TG exhibited strong binding affinity with AR and inhibited its activity and blocked the conversion of GS-HNE to glutathionyl-1,4-dihydroxynonene (GS-DHN), thereby preventing the formation of protein adducts and inducing severe cellular damage. This study provides novel insights into the anti-inflammatory mechanisms of AR inhibitors and offers potential avenues for developing therapeutic strategies for AR-related pathologies. Our findings suggest that TG, as an AR inhibitor, may hold promise as a therapeutic agent for treating conditions characterized by excessive lipid peroxidation and inflammation. Further investigations are needed to fully explore the clinical potential of TG and evaluate its efficacy in the treatment and management of these complex diseases.
Collapse
Affiliation(s)
- Wen-Long Zhao
- Center for Molecular Metabolism, School of Environmental and Biological Engineering, Nanjing University of Science and Technology, 200 Xiao Ling Wei Street, Nanjing 210094, People's Republic of China
| | - Di Xu
- Center for Molecular Metabolism, School of Environmental and Biological Engineering, Nanjing University of Science and Technology, 200 Xiao Ling Wei Street, Nanjing 210094, People's Republic of China
| | - Jun-Song Wang
- Center for Molecular Metabolism, School of Environmental and Biological Engineering, Nanjing University of Science and Technology, 200 Xiao Ling Wei Street, Nanjing 210094, People's Republic of China.
| |
Collapse
|
23
|
Wicaksono WA, Cernava T, Wassermann B, Abdelfattah A, Soto-Giron MJ, Toledo GV, Virtanen SM, Knip M, Hyöty H, Berg G. The edible plant microbiome: evidence for the occurrence of fruit and vegetable bacteria in the human gut. Gut Microbes 2023; 15:2258565. [PMID: 37741805 PMCID: PMC10519362 DOI: 10.1080/19490976.2023.2258565] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 09/08/2023] [Indexed: 09/25/2023] Open
Abstract
Diversity of the gut microbiota is crucial for human health. However, whether fruit and vegetable associated bacteria contribute to overall gut bacterial diversity is still unknown. We reconstructed metagenome-assembled genomes from 156 fruit and vegetable metagenomes to investigate the prevalence of associated bacteria in 2,426 publicly available gut metagenomes. The microbiomes of fresh fruits and vegetables and the human gut are represented by members in common such as Enterobacterales, Burkholderiales, and Lactobacillales. Exposure to bacteria via fruit and vegetable consumption potentially has a beneficial impact on the functional diversity of gut microbiota particularly due to the presence of putative health-promoting genes for the production of vitamin and short-chain fatty acids. In the human gut, they were consistently present, although at a low abundance, approx. 2.2%. Host age, vegetable consumption frequency, and the diversity of plants consumed were drivers favoring a higher proportion. Overall, these results provide one of the primary links between the human microbiome and the environmental microbiome. This study revealed evidence that fruit and vegetable-derived microbes could be found in the human gut and contribute to gut microbiome diversity.
Collapse
Affiliation(s)
- Wisnu Adi Wicaksono
- Institute of Environmental Biotechnology, Graz University of Technology, Graz, Austria
| | - Tomislav Cernava
- Institute of Environmental Biotechnology, Graz University of Technology, Graz, Austria
- School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, UK
| | - Birgit Wassermann
- Institute of Environmental Biotechnology, Graz University of Technology, Graz, Austria
| | - Ahmed Abdelfattah
- Institute of Environmental Biotechnology, Graz University of Technology, Graz, Austria
- Leibniz Institute for Agricultural Engineering and Bioeconomy (ATB), Potsdam, Germany
| | | | | | - Suvi M. Virtanen
- Finnish Institute for Health and Welfare, Helsinki, Finland
- Center for Child Health Research, Tampere University and Tampere University Hospital, Tampere, Finland
- Faculty of Social Sciences, Tampere University, Tampere, Finland
- Research, Development and Innovation Center, Tampere University Hospital, Tampere, Finland
| | - Mikael Knip
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Pediatric Research Center, Children’s Hospital, University of Helsinki, Helsinki, Finland
| | - Heikki Hyöty
- Faculty of Medicine and Health Technology, Tampere University, and Fimlab Laboratories, Tampere, Finland
| | - Gabriele Berg
- Institute of Environmental Biotechnology, Graz University of Technology, Graz, Austria
- Leibniz Institute for Agricultural Engineering and Bioeconomy (ATB), Potsdam, Germany
- Institute for Biochemistry and Biology, University of Potsdam, Potsdam, Germany
| |
Collapse
|
24
|
Yang P, Yang J, Long H, Huang K, Ji L, Lin H, Jiang X, Wang AK, Tian G, Ning K. MicroEXPERT: Microbiome profiling platform with cross-study metagenome-wide association analysis functionality. IMETA 2023; 2:e131. [PMID: 38868224 PMCID: PMC10989818 DOI: 10.1002/imt2.131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/06/2023] [Accepted: 07/10/2023] [Indexed: 06/14/2024]
Abstract
The framework of the MicroEXPERT platform. Our Platform was composed of five modules. Data management module: Users upload raw data and metadata to the system using a guided workflow. Data processing module: Uploaded data is processed to generate taxonomical distribution and functional composition results. Metagenome-wide association studies module (MWAS): Various methods, including biomarker analysis, PCA, co-occurrence networks, and sample classification, are employed using metadata. Data search module: Users can query nucleotide sequences to retrieve information in the MicroEXPERT database. Data visualization module: Visualization tools are used to illustrate the metagenome analysis results.
Collapse
Affiliation(s)
- Pengshuo Yang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular‐imaging, Center of AI Biology, Department of Bioinformatics and Systems BiologyCollege of Life Science and Technology, Huazhong University of Science and TechnologyWuhanHubeiChina
- Institute of Medical GenomicsBiomedical Sciences College, Shandong First Medical UniversityJinanShandongChina
| | - Jialiang Yang
- Department of SciencesGeneis Beijing Co., Ltd.BeijingChina
- Department of SciencesQingdao Geneis Institute of Big Data Mining and Precision MedicineQingdaoChina
- Department of SciencesAcademician Workstation, Changsha Medical UniversityChangshaChina
| | - Haixia Long
- Department of Information Science TechnologyHainan Normal UniversityHaikouChina
| | - Kaimei Huang
- Department of MathematicsZhejiang Normal UniversityJinhuaChina
| | - Lei Ji
- Department of SciencesGeneis Beijing Co., Ltd.BeijingChina
- Department of SciencesQingdao Geneis Institute of Big Data Mining and Precision MedicineQingdaoChina
| | - Hanyang Lin
- Department of SciencesSequenxe Biological Technology Co., Ltd.XiamenChina
| | - Xiuli Jiang
- Department of SciencesSequenxe Biological Technology Co., Ltd.XiamenChina
| | | | - Geng Tian
- Department of SciencesGeneis Beijing Co., Ltd.BeijingChina
- Department of SciencesQingdao Geneis Institute of Big Data Mining and Precision MedicineQingdaoChina
| | - Kang Ning
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular‐imaging, Center of AI Biology, Department of Bioinformatics and Systems BiologyCollege of Life Science and Technology, Huazhong University of Science and TechnologyWuhanHubeiChina
| |
Collapse
|
25
|
Baltazar P, de Melo Junior AF, Fonseca NM, Lança MB, Faria A, Sequeira CO, Teixeira-Santos L, Monteiro EC, Campos Pinheiro L, Calado J, Sousa C, Morello J, Pereira SA. Oxalate (dys)Metabolism: Person-to-Person Variability, Kidney and Cardiometabolic Toxicity. Genes (Basel) 2023; 14:1719. [PMID: 37761859 PMCID: PMC10530622 DOI: 10.3390/genes14091719] [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: 06/25/2023] [Revised: 08/11/2023] [Accepted: 08/16/2023] [Indexed: 09/29/2023] Open
Abstract
Oxalate is a metabolic end-product whose systemic concentrations are highly variable among individuals. Genetic (primary hyperoxaluria) and non-genetic (e.g., diet, microbiota, renal and metabolic disease) reasons underlie elevated plasma concentrations and tissue accumulation of oxalate, which is toxic to the body. A classic example is the triad of primary hyperoxaluria, nephrolithiasis, and kidney injury. Lessons learned from this example suggest further investigation of other putative factors associated with oxalate dysmetabolism, namely the identification of precursors (glyoxylate, aromatic amino acids, glyoxal and vitamin C), the regulation of the endogenous pathways that produce oxalate, or the microbiota's contribution to oxalate systemic availability. The association between secondary nephrolithiasis and cardiovascular and metabolic diseases (hypertension, type 2 diabetes, and obesity) inspired the authors to perform this comprehensive review about oxalate dysmetabolism and its relation to cardiometabolic toxicity. This perspective may offer something substantial that helps advance understanding of effective management and draws attention to the novel class of treatments available in clinical practice.
Collapse
Affiliation(s)
- Pedro Baltazar
- Centro Hospitalar Universitário de Lisboa Central, E.P.E, 1150-199 Lisboa, Portugal; (P.B.); (N.M.F.); (M.B.L.); (L.C.P.); (J.C.)
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, 1150-082 Lisboa, Portugal; (A.F.d.M.J.); (C.O.S.); (L.T.-S.); (E.C.M.); (C.S.); (J.M.)
- Centro Clínico Académico de Lisboa, 1159-056 Lisboa, Portugal
| | - Antonio Ferreira de Melo Junior
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, 1150-082 Lisboa, Portugal; (A.F.d.M.J.); (C.O.S.); (L.T.-S.); (E.C.M.); (C.S.); (J.M.)
- Centro Clínico Académico de Lisboa, 1159-056 Lisboa, Portugal
| | - Nuno Moreira Fonseca
- Centro Hospitalar Universitário de Lisboa Central, E.P.E, 1150-199 Lisboa, Portugal; (P.B.); (N.M.F.); (M.B.L.); (L.C.P.); (J.C.)
- Centro Clínico Académico de Lisboa, 1159-056 Lisboa, Portugal
| | - Miguel Brito Lança
- Centro Hospitalar Universitário de Lisboa Central, E.P.E, 1150-199 Lisboa, Portugal; (P.B.); (N.M.F.); (M.B.L.); (L.C.P.); (J.C.)
| | - Ana Faria
- CHRC, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, 1150-082 Lisboa, Portugal;
| | - Catarina O. Sequeira
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, 1150-082 Lisboa, Portugal; (A.F.d.M.J.); (C.O.S.); (L.T.-S.); (E.C.M.); (C.S.); (J.M.)
| | - Luísa Teixeira-Santos
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, 1150-082 Lisboa, Portugal; (A.F.d.M.J.); (C.O.S.); (L.T.-S.); (E.C.M.); (C.S.); (J.M.)
- Centro Clínico Académico de Lisboa, 1159-056 Lisboa, Portugal
| | - Emilia C. Monteiro
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, 1150-082 Lisboa, Portugal; (A.F.d.M.J.); (C.O.S.); (L.T.-S.); (E.C.M.); (C.S.); (J.M.)
- Centro Clínico Académico de Lisboa, 1159-056 Lisboa, Portugal
| | - Luís Campos Pinheiro
- Centro Hospitalar Universitário de Lisboa Central, E.P.E, 1150-199 Lisboa, Portugal; (P.B.); (N.M.F.); (M.B.L.); (L.C.P.); (J.C.)
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, 1150-082 Lisboa, Portugal; (A.F.d.M.J.); (C.O.S.); (L.T.-S.); (E.C.M.); (C.S.); (J.M.)
- Centro Clínico Académico de Lisboa, 1159-056 Lisboa, Portugal
| | - Joaquim Calado
- Centro Hospitalar Universitário de Lisboa Central, E.P.E, 1150-199 Lisboa, Portugal; (P.B.); (N.M.F.); (M.B.L.); (L.C.P.); (J.C.)
- Centro Clínico Académico de Lisboa, 1159-056 Lisboa, Portugal
- ToxOmics, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, 1150-082 Lisboa, Portugal
| | - Cátia Sousa
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, 1150-082 Lisboa, Portugal; (A.F.d.M.J.); (C.O.S.); (L.T.-S.); (E.C.M.); (C.S.); (J.M.)
- Centro Clínico Académico de Lisboa, 1159-056 Lisboa, Portugal
| | - Judit Morello
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, 1150-082 Lisboa, Portugal; (A.F.d.M.J.); (C.O.S.); (L.T.-S.); (E.C.M.); (C.S.); (J.M.)
| | - Sofia A. Pereira
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, 1150-082 Lisboa, Portugal; (A.F.d.M.J.); (C.O.S.); (L.T.-S.); (E.C.M.); (C.S.); (J.M.)
- Centro Clínico Académico de Lisboa, 1159-056 Lisboa, Portugal
| |
Collapse
|
26
|
Querdasi FR, Vogel SC, Thomason ME, Callaghan BL, Brito NH. A comparison of the infant gut microbiome before versus after the start of the covid-19 pandemic. Sci Rep 2023; 13:13289. [PMID: 37587195 PMCID: PMC10432475 DOI: 10.1038/s41598-023-40102-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 08/04/2023] [Indexed: 08/18/2023] Open
Abstract
The COVID-19 pandemic and resulting public health directives led to many changes in families' social and material environments. Prior research suggests that these changes are likely to impact composition of the gut microbiome, particularly during early childhood when the gut microbiome is developing most rapidly. Importantly, disruption to the gut microbiome during this sensitive period can have potentially long-lasting impacts on health and development. In the current study, we compare gut microbiome composition among a socioeconomically and racially diverse group of 12-month old infants living in New York City who provided stool samples before the pandemic (N = 34) to a group who provided samples during the first 9-months of the pandemic (March-December 2020; N = 20). We found that infants sampled during the pandemic had lower alpha diversity of the microbiome, lower abundance of Pasteurellaceae and Haemophilus, and significantly different beta diversity based on unweighted Unifrac distance than infants sampled before the pandemic. Exploratory analyses suggest that gut microbiome changes due to the pandemic occurred relatively quickly after the start of the pandemic and were sustained. Our results provide evidence that pandemic-related environmental disruptions had an impact on community-level taxonomic diversity of the developing gut microbiome, as well as abundance of specific members of the gut bacterial community.
Collapse
|
27
|
Ma H, Zhao W, Song T, Baijiu Z, Zhang Z. Comparative Analysis of the Pre-Parturition and Post-Parturition Genital Tract Microbiota in Plateau Bangor Sewa Sheep. Vet Sci 2023; 10:523. [PMID: 37624310 PMCID: PMC10459245 DOI: 10.3390/vetsci10080523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/09/2023] [Accepted: 08/09/2023] [Indexed: 08/26/2023] Open
Abstract
(1) Background: Bangor Sewa sheep are an economically significant livestock species on the plateau. The roles of microbiota in reproduction are complex and critical for animal health. But little is known currently about the microbiome of plateau Bangor Sewa sheep. The purpose of this study was to discover the changes in the genital tract microbiota of pre- and post-partum Bangor Sewa sheep. (2) Methods: Samples from the birth canal were obtained for 16S rRNA sequencing, three days before and after delivery, respectively. (3) Results: The results showed that there was a noticeable difference in three phyla and 74 genera between the pre- and post-parturition groups in the microbiota of Bangor Sewa sheep. The changes included a decrease in the abundance of genera related to health (unclassified_Cellulomonadaceae, Cellulomonas, Fibrobacti, Flavobacterium, Eubacterium_ventriosum_group, Acetitomaculum, Aeromicrobium, Dietzia, Romboutsia, Ruminococcus, etc.) and an increased abundance of negatively related genera (Nocardioides, unclassified_Clostridia, Sphingobacteriaceae, unclassified_Ruminococcaceae, Prevotellaceae_UCG_004, Micromonospora, Streptococcus, Facklamia, Bosea, etc.) spp. (4) Conclusions: Microbes can serve as indicators of the physical state of Bangor Sewa sheep. These findings laid the foundation for deciphering the effects of microbial changes during birth on the reproductive health of plateau Bangor Sewa sheep.
Collapse
Affiliation(s)
- Hongcai Ma
- Institute of Animal Husbandry and Veterinary Medicine, Tibet Autonomous Region Academy of Agriculture and Animal Science, Lhasa 850009, China; (H.M.); (T.S.)
| | - Wangsheng Zhao
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang 621010, China;
| | - Tianzeng Song
- Institute of Animal Husbandry and Veterinary Medicine, Tibet Autonomous Region Academy of Agriculture and Animal Science, Lhasa 850009, China; (H.M.); (T.S.)
| | - Zhaxi Baijiu
- Cultural Service Center of Maqian Township, Nagqu 852599, China;
| | - Zhenzhen Zhang
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang 621010, China;
| |
Collapse
|
28
|
Lai S, Yan Y, Pu Y, Lin S, Qiu JG, Jiang BH, Keller MI, Wang M, Bork P, Chen WH, Zheng Y, Zhao XM. Enterotypes of the human gut mycobiome. MICROBIOME 2023; 11:179. [PMID: 37563687 PMCID: PMC10416509 DOI: 10.1186/s40168-023-01586-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 05/31/2023] [Indexed: 08/12/2023]
Abstract
BACKGROUND The fungal component of the human gut microbiome, also known as the mycobiome, plays a vital role in intestinal ecology and human health. However, the overall structure of the gut mycobiome as well as the inter-individual variations in fungal composition remains largely unknown. In this study, we collected a total of 3363 fungal sequencing samples from 16 cohorts across three continents, including 572 newly profiled samples from China. RESULTS We identify and characterize four mycobiome enterotypes using ITS profiling of 3363 samples from 16 cohorts. These enterotypes exhibit stability across populations and geographical locations and significant correlation with bacterial enterotypes. Particularly, we notice that fungal enterotypes have a strong age preference, where the enterotype dominated by Candida (i.e., Can_type enterotype) is enriched in the elderly population and confers an increased risk of multiple diseases associated with a compromised intestinal barrier. In addition, bidirectional mediation analysis reveals that the fungi-contributed aerobic respiration pathway associated with the Can_type enterotype might mediate the association between the compromised intestinal barrier and aging. CONCLUSIONS We show that the human gut mycobiome has stable compositional patterns across individuals and significantly correlates with multiple host factors, such as diseases and host age. Video Abstract.
Collapse
Affiliation(s)
- Senying Lai
- Department of Neurology, Zhongshan Hospital and Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Yan Yan
- CAS Key Laboratory of Molecular Virology and Immunology, Chinese Academy of Sciences (CAS), Shanghai, China
| | - Yanni Pu
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, School of Life Sciences, Fudan University, Shanghai, China
| | - Shuchun Lin
- The Academy of Medical Science, Zhengzhou University, Zhengzhou, China
| | - Jian-Ge Qiu
- The Academy of Medical Science, Zhengzhou University, Zhengzhou, China
| | - Bing-Hua Jiang
- The Academy of Medical Science, Zhengzhou University, Zhengzhou, China
| | - Marisa Isabell Keller
- European Molecular Biology Laboratory, Structural and Computational Biology Unit, Heidelberg, Germany
| | - Mingyu Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular Imaging, Center for Artificial Intelligence Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Peer Bork
- European Molecular Biology Laboratory, Structural and Computational Biology Unit, Heidelberg, Germany.
- Max Delbrück Centre for Molecular Medicine, Berlin, Germany.
- Department of Bioinformatics, Biocenter, University of Würzburg, Würzburg, Germany.
| | - Wei-Hua Chen
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular Imaging, Center for Artificial Intelligence Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China.
- College of Life Science, Henan Normal University, Xinxiang, Henan, China.
| | - Yan Zheng
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, School of Life Sciences, Fudan University, Shanghai, China.
| | - Xing-Ming Zhao
- Department of Neurology, Zhongshan Hospital and Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China.
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Zhejiang Province, China.
- MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Fudan University, Shanghai, China.
- International Human Phenome Institutes (Shanghai), Shanghai, China.
| |
Collapse
|
29
|
Dong TS, Gee GC, Beltran-Sanchez H, Wang M, Osadchiy V, Kilpatrick LA, Chen Z, Subramanyam V, Zhang Y, Guo Y, Labus JS, Naliboff B, Cole S, Zhang X, Mayer EA, Gupta A. How Discrimination Gets Under the Skin: Biological Determinants of Discrimination Associated With Dysregulation of the Brain-Gut Microbiome System and Psychological Symptoms. Biol Psychiatry 2023; 94:203-214. [PMID: 36754687 PMCID: PMC10684253 DOI: 10.1016/j.biopsych.2022.10.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 10/11/2022] [Accepted: 10/12/2022] [Indexed: 11/02/2022]
Abstract
BACKGROUND Discrimination is associated with negative health outcomes as mediated in part by chronic stress, but a full understanding of the biological pathways is lacking. Here we investigate the effects of discrimination involved in dysregulating the brain-gut microbiome (BGM) system. METHODS A total of 154 participants underwent brain magnetic resonance imaging to measure functional connectivity. Fecal samples were obtained for 16S ribosomal RNA profiling and fecal metabolites and serum for inflammatory markers, along with questionnaires. The Everyday Discrimination Scale was administered to measure chronic and routine experiences of unfair treatment. A sparse partial least squares-discriminant analysis was conducted to predict BGM alterations as a function of discrimination, controlling for sex, age, body mass index, and diet. Associations between discrimination-related BGM alterations and psychological variables were assessed using a tripartite analysis. RESULTS Discrimination was associated with anxiety, depression, and visceral sensitivity. Discrimination was associated with alterations of brain networks related to emotion, cognition and self-perception, and structural and functional changes in the gut microbiome. BGM discrimination-related associations varied by race/ethnicity. Among Black and Hispanic individuals, discrimination led to brain network changes consistent with psychological coping and increased systemic inflammation. For White individuals, discrimination was related to anxiety but not inflammation, while for Asian individuals, the patterns suggest possible somatization and behavioral (e.g., dietary) responses to discrimination. CONCLUSIONS Discrimination is attributed to changes in the BGM system more skewed toward inflammation, threat response, emotional arousal, and psychological symptoms. By integrating diverse lines of research, our results demonstrate evidence that may explain how discrimination contributes to health inequalities.
Collapse
Affiliation(s)
- Tien S Dong
- Vatche and Tamar Manoukian Division of Digestive Diseases, University of California, Los Angeles, Los Angeles, California; David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California; UCLA Microbiome Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California; G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California, Los Angeles, Los Angeles, California; Division of Gastroenterology, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, California.
| | - Gilbert C Gee
- Department of Community Health Sciences Fielding School of Public Health, Los Angeles, California; California Center for Population Research, University of California, Los Angeles, Los Angeles, California
| | - Hiram Beltran-Sanchez
- Department of Community Health Sciences Fielding School of Public Health, Los Angeles, California; California Center for Population Research, University of California, Los Angeles, Los Angeles, California
| | - May Wang
- Department of Community Health Sciences Fielding School of Public Health, Los Angeles, California
| | - Vadim Osadchiy
- Department of Urology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Lisa A Kilpatrick
- Vatche and Tamar Manoukian Division of Digestive Diseases, University of California, Los Angeles, Los Angeles, California; David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California; G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California, Los Angeles, Los Angeles, California
| | - Zixi Chen
- Vatche and Tamar Manoukian Division of Digestive Diseases, University of California, Los Angeles, Los Angeles, California
| | - Vishvak Subramanyam
- Vatche and Tamar Manoukian Division of Digestive Diseases, University of California, Los Angeles, Los Angeles, California
| | - Yurui Zhang
- Vatche and Tamar Manoukian Division of Digestive Diseases, University of California, Los Angeles, Los Angeles, California
| | - Yinming Guo
- Vatche and Tamar Manoukian Division of Digestive Diseases, University of California, Los Angeles, Los Angeles, California
| | - Jennifer S Labus
- Vatche and Tamar Manoukian Division of Digestive Diseases, University of California, Los Angeles, Los Angeles, California; David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California; UCLA Microbiome Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California; G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California, Los Angeles, Los Angeles, California
| | - Bruce Naliboff
- Vatche and Tamar Manoukian Division of Digestive Diseases, University of California, Los Angeles, Los Angeles, California; David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California; UCLA Microbiome Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California; G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California, Los Angeles, Los Angeles, California
| | - Steve Cole
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California; Department of Psychiatry & Biobehavioral Sciences and Medicine, University of California, Los Angeles, Los Angeles, California
| | - Xiaobei Zhang
- Vatche and Tamar Manoukian Division of Digestive Diseases, University of California, Los Angeles, Los Angeles, California; David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California; G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California, Los Angeles, Los Angeles, California
| | - Emeran A Mayer
- Vatche and Tamar Manoukian Division of Digestive Diseases, University of California, Los Angeles, Los Angeles, California; David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California; UCLA Microbiome Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California; G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California, Los Angeles, Los Angeles, California
| | - Arpana Gupta
- Vatche and Tamar Manoukian Division of Digestive Diseases, University of California, Los Angeles, Los Angeles, California; David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California; UCLA Microbiome Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California; G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California, Los Angeles, Los Angeles, California.
| |
Collapse
|
30
|
Summer M, Ali S, Fiaz U, Tahir HM, Ijaz M, Mumtaz S, Mushtaq R, Khan R, Shahzad H, Fiaz H. Therapeutic and immunomodulatory role of probiotics in breast cancer: A mechanistic review. Arch Microbiol 2023; 205:296. [PMID: 37486419 DOI: 10.1007/s00203-023-03632-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 07/03/2023] [Accepted: 07/09/2023] [Indexed: 07/25/2023]
Abstract
Breast cancer has become the most prevalent and noxious type of malignancy around the globe (Giaquinto et al., 2022). Multiple clinical strategies including chemotherapy, radiotherapy, and immunotherapy have been in practice to manage breast cancer. Besides the protective roles of conventional remedial approaches, and non-reversible and deteriorative impacts like healthy cell damage, organ failure, etc., the world scientific community is in a continuous struggle to find some alternative biocompatible and comparatively safe solutions. Among novel breast cancer management/treatment options, the role of probiotics has become immensely important. The current review encompasses the prevalence statistics of breast cancer across the globe concerning developed and undeveloped counties, intestinal microbiota linkage with breast cancer, and association of breast microbiome with breast carcinoma. Furthermore, this review also narrates the role of probiotics against breast cancer and their mode of action. In Vivo and In Vitro studies under breast cancer research regarding probiotics are mechanistically explained. The current review systematically explains the immunomodulatory role of probiotics to prevent breast cancer. Last, but not the least, current review concludes the use of probiotics in the treatment of breast cancer through various mechanisms and future recommendations for molecular basis studies.
Collapse
Affiliation(s)
- Muhammad Summer
- Department of Zoology, Government College University Lahore, Lahore, 54000, Pakistan
| | - Shaukat Ali
- Department of Zoology, Government College University Lahore, Lahore, 54000, Pakistan.
| | - Umaima Fiaz
- Department of Zoology, Government College University Lahore, Lahore, 54000, Pakistan
| | - Hafiz Muhammad Tahir
- Department of Zoology, Government College University Lahore, Lahore, 54000, Pakistan
| | - Muhammad Ijaz
- Department of Veterinary Medicine, University of Veterinary and Animal Sciences Lahore, Lahore, Pakistan
| | - Shumaila Mumtaz
- Department of Zoology, Government College University Lahore, Lahore, 54000, Pakistan
| | - Rabia Mushtaq
- Department of Zoology, Government College University Lahore, Lahore, 54000, Pakistan
| | - Rida Khan
- Department of Zoology, Government College University Lahore, Lahore, 54000, Pakistan
| | - Hafsa Shahzad
- Department of Zoology, Government College University Lahore, Lahore, 54000, Pakistan
| | - Hashim Fiaz
- Department of Medicine and Surgery, Ammer-ul-din Medical College Lahore, Lahore, Pakistan
| |
Collapse
|
31
|
Casto-Rebollo C, Argente MJ, García ML, Pena RN, Blasco A, Ibáñez-Escriche N. Selection for environmental variance shifted the gut microbiome composition driving animal resilience. MICROBIOME 2023; 11:147. [PMID: 37400907 DOI: 10.1186/s40168-023-01580-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 05/23/2023] [Indexed: 07/05/2023]
Abstract
BACKGROUND Understanding how the host's microbiome shapes phenotypes and participates in the host response to selection is fundamental for evolutionists and animal and plant breeders. Currently, selection for resilience is considered a critical step in improving the sustainability of livestock systems. Environmental variance (V E), the within-individual variance of a trait, has been successfully used as a proxy for animal resilience. Selection for reduced V E could effectively shift gut microbiome composition; reshape the inflammatory response, triglyceride, and cholesterol levels; and drive animal resilience. This study aimed to determine the gut microbiome composition underlying the V E of litter size (LS), for which we performed a metagenomic analysis in two rabbit populations divergently selected for low (n = 36) and high (n = 34) V E of LS. Partial least square-discriminant analysis and alpha- and beta-diversity were computed to determine the differences in gut microbiome composition among the rabbit populations. RESULTS We identified 116 KEGG IDs, 164 COG IDs, and 32 species with differences in abundance between the two rabbit populations studied. These variables achieved a classification performance of the V E rabbit populations of over than 80%. Compared to the high V E population, the low V E (resilient) population was characterized by an underrepresentation of Megasphaera sp., Acetatifactor muris, Bacteroidetes rodentium, Ruminococcus bromii, Bacteroidetes togonis, and Eggerthella sp. and greater abundances of Alistipes shahii, Alistipes putredinis, Odoribacter splanchnicus, Limosilactobacillus fermentum, and Sutterella, among others. Differences in abundance were also found in pathways related to biofilm formation, quorum sensing, glutamate, and amino acid aromatic metabolism. All these results suggest differences in gut immunity modulation, closely related to resilience. CONCLUSIONS This is the first study to show that selection for V E of LS can shift the gut microbiome composition. The results revealed differences in microbiome composition related to gut immunity modulation, which could contribute to the differences in resilience among rabbit populations. The selection-driven shifts in gut microbiome composition should make a substantial contribution to the remarkable genetic response observed in the V E rabbit populations. Video Abstract.
Collapse
Affiliation(s)
- Cristina Casto-Rebollo
- Institute for Animal Science and Technology, Universitat Politècnica de València, València, Spain
| | - María José Argente
- Centro de Investigación e Innovación Agroalimentaria Y Agroambiental (CIAGRO_UMH), Miguel Hernández University, Orihuela, 03312, Spain
| | - María Luz García
- Centro de Investigación e Innovación Agroalimentaria Y Agroambiental (CIAGRO_UMH), Miguel Hernández University, Orihuela, 03312, Spain
| | - Ramona Natacha Pena
- Departament de Ciència Animal, Universitat de Lleida-AGROTECNIO Center, Lleida, Catalonia, Spain
| | - Agustín Blasco
- Institute for Animal Science and Technology, Universitat Politècnica de València, València, Spain
| | - Noelia Ibáñez-Escriche
- Institute for Animal Science and Technology, Universitat Politècnica de València, València, Spain.
| |
Collapse
|
32
|
Agarwal M, Hoffman J, Ngo Tenlep SY, Santarossa S, Pearson KJ, Sitarik AR, Cassidy-Bushrow AE, Petriello MC. Maternal polychlorinated biphenyl 126 (PCB 126) exposure modulates offspring gut microbiota irrespective of diet and exercise. Reprod Toxicol 2023; 118:108384. [PMID: 37061048 PMCID: PMC10257154 DOI: 10.1016/j.reprotox.2023.108384] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 04/07/2023] [Accepted: 04/12/2023] [Indexed: 04/17/2023]
Abstract
The gut microbiota plays an important role throughout the lifespan in maintaining host health, and several factors can modulate microbiota composition including diet, exercise, and environmental exposures. Maternal microbiota is transferred to offspring during early life; thus, environmental exposures before gestation may also modulate offspring microbiota. Here we aimed to investigate the effects of maternal exposure to dioxin-like polychlorinated biphenyls (PCBs) on the microbiota of aged offspring and to determine if lifestyle factors, including maternal exercise or offspring high-fat feeding alter these associations. To test this, dams were exposed to PCB 126 (0.5 μmole/kg body weight) or vehicle oil by oral gavage during preconception, gestation, and during lactation. Half of each group was allowed access to running wheels for ≥ 7 days before and during pregnancy and up through day 14 of lactation. Female offspring born from the 4 maternal groups (PCB exposure or not, with/without exercise) were subsequently placed either on regular diet or switched to a high-fat diet during adulthood. Microbiota composition was quantified in female offspring at 49 weeks of age by 16 S rRNA sequencing. Maternal exposure to PCB 126 resulted in significantly reduced richness and diversity in offspring microbiota regardless of diet or exercise. Overall compositional differences were largely driven by offspring diet, but alterations in specific taxa due to maternal PCB 126 exposure, included the depletion of Verrucomicrobiaceae and Akkermansia muciniphila, and an increase in Anaeroplasma. Perturbation of microbiota due to PCB 126 may predispose offspring to a variety of chronic diseases later in adulthood.
Collapse
Affiliation(s)
- Manisha Agarwal
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI, 48202, USA
| | - Jessie Hoffman
- Department of Human Nutrition, Winthrop University, Rock Hill, SC 29733, USA
| | - Sara Y Ngo Tenlep
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, USA
| | - Sara Santarossa
- Department of Public Health Sciences, Henry Ford Health, Detroit, MI 48202, USA
| | - Kevin J Pearson
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, USA
| | - Alexandra R Sitarik
- Department of Public Health Sciences, Henry Ford Health, Detroit, MI 48202, USA
| | | | - Michael C Petriello
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI, 48202, USA; Institute of Environmental Health Sciences, Wayne State University, Detroit, MI 48202, USA.
| |
Collapse
|
33
|
Wollmuth EM, Angert ER. Microbial circadian clocks: host-microbe interplay in diel cycles. BMC Microbiol 2023; 23:124. [PMID: 37161348 PMCID: PMC10173096 DOI: 10.1186/s12866-023-02839-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 03/28/2023] [Indexed: 05/11/2023] Open
Abstract
BACKGROUND Circadian rhythms, observed across all domains of life, enable organisms to anticipate and prepare for diel changes in environmental conditions. In bacteria, a circadian clock mechanism has only been characterized in cyanobacteria to date. These clocks regulate cyclical patterns of gene expression and metabolism which contribute to the success of cyanobacteria in their natural environments. The potential impact of self-generated circadian rhythms in other bacterial and microbial populations has motivated extensive research to identify novel circadian clocks. MAIN TEXT Daily oscillations in microbial community composition and function have been observed in ocean ecosystems and in symbioses. These oscillations are influenced by abiotic factors such as light and the availability of nutrients. In the ocean ecosystems and in some marine symbioses, oscillations are largely controlled by light-dark cycles. In gut systems, the influx of nutrients after host feeding drastically alters the composition and function of the gut microbiota. Conversely, the gut microbiota can influence the host circadian rhythm by a variety of mechanisms including through interacting with the host immune system. The intricate and complex relationship between the microbiota and their host makes it challenging to disentangle host behaviors from bacterial circadian rhythms and clock mechanisms that might govern the daily oscillations observed in these microbial populations. CONCLUSIONS While the ability to anticipate the cyclical behaviors of their host would likely be enhanced by a self-sustained circadian rhythm, more evidence and further studies are needed to confirm whether host-associated heterotrophic bacteria possess such systems. In addition, the mechanisms by which heterotrophic bacteria might respond to diel cycles in environmental conditions has yet to be uncovered.
Collapse
Affiliation(s)
- Emily M Wollmuth
- Department of Microbiology, Cornell University, 123 Wing Drive, Ithaca, NY, 14853, USA
| | - Esther R Angert
- Department of Microbiology, Cornell University, 123 Wing Drive, Ithaca, NY, 14853, USA.
| |
Collapse
|
34
|
Yue R, Chen H, Xu X, Xia Y, Sun Y, Xia M, Xia D, Sun B. Maternal and infantile gut mycobiome during the weaning period in free ranging Tibetan macaques ( Macaca thibetana). Ecol Evol 2023; 13:e10108. [PMID: 37214608 PMCID: PMC10196218 DOI: 10.1002/ece3.10108] [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: 02/09/2023] [Revised: 05/02/2023] [Accepted: 05/05/2023] [Indexed: 05/24/2023] Open
Abstract
Gut microbiome is critical to the health of mammals. Many previous studies have revealed the gut bacterial microbiomes of mother and infant changed significantly during the weaning period. However, little is known concerning the gut mycobiome of wild primates. Here, we examined the variations on gut mycobiome between weaning and post-weaning for both mother and infant in wild-living Tibetan macaques (Macaca thibetana). Our results showed that the gut mycobiomes of mother and infant were dominated by two phyla Ascomycota and Basidiomycota. For both mother and infant, the ASV richness of gut mycobiome remained relatively steady from weaning to post-weaning periods, while the Shannon indexes increased significant in weaning compared to post-weaning periods. However, no significant difference between mother and infant ASV richness and Shannon indexes during weaning and post-weaning periods respectively. Compared to mothers, we found that much more known taxa of gut fungi were enriched in weaning or post-weaning periods of infants. In particular, we found that the dominant genus Aspergillus was enriched in infants during weaning period. Furthermore, we found that the relative abundance of plant pathogens were significantly higher in the post-weaning period than in the weaning period for infants. Our results indicated that weaning events could affect the gut mycobiome significantly for both mothers and infant in Tibetan macaques, which had a stronger effect on the gut mycobiome of infant monkeys than on their mothers.
Collapse
Affiliation(s)
- Ran Yue
- School of Resource and Environmental EngineeringAnhui UniversityHefeiChina
- International Collaborative Research Center for Huangshan Biodiversity and Tibetan Macaque Behavioral EcologyAnhui UniversityHefeiChina
| | - Huijuan Chen
- School of Resource and Environmental EngineeringAnhui UniversityHefeiChina
- International Collaborative Research Center for Huangshan Biodiversity and Tibetan Macaque Behavioral EcologyAnhui UniversityHefeiChina
| | - Xiaojuan Xu
- School of Life SciencesHefei Normal UniversityHefeiChina
| | - Yingna Xia
- School of Resource and Environmental EngineeringAnhui UniversityHefeiChina
- International Collaborative Research Center for Huangshan Biodiversity and Tibetan Macaque Behavioral EcologyAnhui UniversityHefeiChina
| | - Yu Sun
- School of Resource and Environmental EngineeringAnhui UniversityHefeiChina
- International Collaborative Research Center for Huangshan Biodiversity and Tibetan Macaque Behavioral EcologyAnhui UniversityHefeiChina
| | - Mengyi Xia
- School of Resource and Environmental EngineeringAnhui UniversityHefeiChina
- International Collaborative Research Center for Huangshan Biodiversity and Tibetan Macaque Behavioral EcologyAnhui UniversityHefeiChina
| | - Dongpo Xia
- School of Life SciencesAnhui UniversityHefeiChina
| | - Binghua Sun
- School of Resource and Environmental EngineeringAnhui UniversityHefeiChina
- International Collaborative Research Center for Huangshan Biodiversity and Tibetan Macaque Behavioral EcologyAnhui UniversityHefeiChina
| |
Collapse
|
35
|
Balmant BD, Fonseca DC, Prudêncio APA, Rocha IM, Callado L, Alves JTM, Torrinhas RSMDM, Borba EF, Waitzberg DL. Megamonas funiformis, Plasma Zonulin, and Sodium Intake Affect C3 Complement Levels in Inactive Systemic Lupus Erythematosus. Nutrients 2023; 15:nu15081999. [PMID: 37111218 PMCID: PMC10144636 DOI: 10.3390/nu15081999] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/11/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023] Open
Abstract
The etiology of systemic lupus erythematosus (SLE) remains unclear, with both genetic and environmental factors potentially contributing. This study aimed to explore the relationship among gut microbiota (GM), intestinal permeability, and food intake with inflammatory markers in inactive SLE patients. A total of 22 women with inactive SLE and 20 healthy volunteers were enrolled, and dietary intake was assessed through 24-h dietary recalls. Plasma zonulin was used to evaluate intestinal permeability, while GM was determined by 16S rRNA sequencing. Regression models were used to analyze laboratory markers of lupus disease (C3 and C4 complement and C-reactive protein). Our results showed that the genus Megamonas was significantly enriched in the iSLE group (p < 0.001), with Megamonas funiformis associated with all evaluated laboratory tests (p < 0.05). Plasma zonulin was associated with C3 levels (p = 0.016), and sodium intake was negatively associated with C3 and C4 levels (p < 0.05). A combined model incorporating variables from each group (GM, intestinal permeability, and food intake) demonstrated a significant association with C3 complement levels (p < 0.01). These findings suggest that increased Megamonas funiformis abundance, elevated plasma zonulin, and higher sodium intake may contribute to reduced C3 complement levels in women with inactive SLE.
Collapse
Affiliation(s)
- Bianca Depieri Balmant
- Laboratory of Nutrition and Metabolic Surgery of the Digestive System, LIM 35, Department of Gastroenterology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo 01246-903, Brazil
| | - Danielle Cristina Fonseca
- Laboratory of Nutrition and Metabolic Surgery of the Digestive System, LIM 35, Department of Gastroenterology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo 01246-903, Brazil
| | - Ana Paula Aguiar Prudêncio
- Laboratory of Nutrition and Metabolic Surgery of the Digestive System, LIM 35, Department of Gastroenterology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo 01246-903, Brazil
| | - Ilanna Marques Rocha
- Laboratory of Nutrition and Metabolic Surgery of the Digestive System, LIM 35, Department of Gastroenterology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo 01246-903, Brazil
| | - Letícia Callado
- Laboratory of Nutrition and Metabolic Surgery of the Digestive System, LIM 35, Department of Gastroenterology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo 01246-903, Brazil
| | | | - Raquel Susana Matos de Miranda Torrinhas
- Laboratory of Nutrition and Metabolic Surgery of the Digestive System, LIM 35, Department of Gastroenterology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo 01246-903, Brazil
| | - Eduardo Ferreira Borba
- Rheumatology Division, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo 01246-903, Brazil
| | - Dan Linetzky Waitzberg
- Laboratory of Nutrition and Metabolic Surgery of the Digestive System, LIM 35, Department of Gastroenterology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo 01246-903, Brazil
| |
Collapse
|
36
|
Zhao F, Hu X, Ying C. Advances in Research on the Relationship between Vaginal Microbiota and Adverse Pregnancy Outcomes and Gynecological Diseases. Microorganisms 2023; 11:microorganisms11040991. [PMID: 37110417 PMCID: PMC10146011 DOI: 10.3390/microorganisms11040991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/01/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
The human microbiota inhabiting different parts of the body has been shown to have a significant impact on human health, with the gut microbiota being the most extensively studied in relation to disease. However, the vaginal microbiota is also an essential commensal microbiota in the female body that plays a crucial role in female health. Despite receiving less attention than gut microbiota, its importance in regulating reproductive immunity and its complex dynamic properties have been increasingly recognized in recent years. Advances in research on the relationship between vaginal microbiota and pregnancy outcomes & gynecological diseases in women have shed light on the importance of maintaining a healthy vaginal microbiota. In this review, we aim to compile recent developments in the study of the vaginal microbial ecosystem and its role in female health and reproductive outcomes. We provide a comprehensive account of the normal vaginal microbiota, the association between the vaginal microbiota and pregnancy outcomes, and the impact of the vaginal microbiota on gynecological diseases in women. By reviewing recent research, we hope to contribute to the advancement of academic medicine's understanding of the vaginal microbiota's importance in female health. We also aim to raise awareness among healthcare professionals and the general public of the significance of maintaining a healthy vaginal microbiota for better reproductive health and the prevention of gynecological diseases.
Collapse
Affiliation(s)
- Fuju Zhao
- Clinical Laboratory, Huadong Hospital, Fudan University, Shanghai 200031, China
- Clinical Laboratory, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
| | - Xianyang Hu
- Huadong Hospital, Fudan University, Shanghai 200031, China
| | - Chunmei Ying
- Clinical Laboratory, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
| |
Collapse
|
37
|
Korobkova L, Morin EL, Aoued H, Sannigrahi S, Garza KM, Siebert ER, Walum H, Cabeen RP, Sanchez MM, Dias BG. RNA in extracellular vesicles during adolescence reveal immune, energetic and microbial imprints of early life adversity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.23.529808. [PMID: 36865138 PMCID: PMC9980043 DOI: 10.1101/2023.02.23.529808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
Exposure to early life adversity (ELA), including childhood maltreatment, is one of the most significant risk factors for the emergence of neuropsychiatric disorders in adolescence and adulthood. Despite this relationship being well established, the underlying mechanisms remain unclear. One way to achieve this understanding is to identify molecular pathways and processes that are perturbed as a consequence of childhood maltreatment. Ideally, these perturbations would be evident as changes in DNA, RNA or protein profiles in easily accessible biological samples collected in the shadow of childhood maltreatment. In this study, we isolated circulating extracellular vesicles (EVs) from plasma collected from adolescent rhesus macaques that had either experienced nurturing maternal care (CONT) or maternal maltreatment (MALT) in infancy. RNA sequencing of RNA in plasma EVs and gene enrichment analysis revealed that genes related to translation, ATP synthesis, mitochondrial function and immune response were downregulated in MALT samples, while genes involved in ion transport, metabolism and cell differentiation were upregulated. Interestingly, we found that a significant proportion of EV RNA aligned to the microbiome and that MALT altered the diversity of microbiome-associated RNA signatures found in EVs. Part of this altered diversity suggested differences in prevalence of bacterial species in CONT and MALT animals noted in the RNA signatures of the circulating EVs. Our findings provide evidence that immune function, cellular energetics and the microbiome may be important conduits via which infant maltreatment exerts effects on physiology and behavior in adolescence and adulthood. As a corollary, perturbations of RNA profiles related to immune function, cellular energetics and the microbiome may serve as biomarkers of responsiveness to ELA. Our results demonstrate that RNA profiles in EVs can serve as a powerful proxy to identify biological processes that might be perturbed by ELA and that may contribute to the etiology of neuropsychiatric disorders in the aftermath of ELA.
Collapse
|
38
|
Kuraji R, Shiba T, Dong TS, Numabe Y, Kapila YL. Periodontal treatment and microbiome-targeted therapy in management of periodontitis-related nonalcoholic fatty liver disease with oral and gut dysbiosis. World J Gastroenterol 2023; 29:967-996. [PMID: 36844143 PMCID: PMC9950865 DOI: 10.3748/wjg.v29.i6.967] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 11/14/2022] [Accepted: 01/30/2023] [Indexed: 02/10/2023] Open
Abstract
A growing body of evidence from multiple areas proposes that periodontal disease, accompanied by oral inflammation and pathological changes in the microbiome, induces gut dysbiosis and is involved in the pathogenesis of nonalcoholic fatty liver disease (NAFLD). A subgroup of NAFLD patients have a severely progressive form, namely nonalcoholic steatohepatitis (NASH), which is characterized by histological findings that include inflammatory cell infiltration and fibrosis. NASH has a high risk of further progression to cirrhosis and hepatocellular carcinoma. The oral microbiota may serve as an endogenous reservoir for gut microbiota, and transport of oral bacteria through the gastro-intestinal tract can set up a gut microbiome dysbiosis. Gut dysbiosis increases the production of potential hepatotoxins, including lipopolysaccharide, ethanol, and other volatile organic compounds such as acetone, phenol and cyclopentane. Moreover, gut dysbiosis increases intestinal permeability by disrupting tight junctions in the intestinal wall, leading to enhanced translocation of these hepatotoxins and enteric bacteria into the liver through the portal circulation. In particular, many animal studies support that oral administration of Porphyromonas gingivalis, a typical periodontopathic bacterium, induces disturbances in glycolipid metabolism and inflammation in the liver with gut dysbiosis. NAFLD, also known as the hepatic phenotype of metabolic syndrome, is strongly associated with metabolic complications, such as obesity and diabetes. Periodontal disease also has a bidirectional relationship with metabolic syndrome, and both diseases may induce oral and gut microbiome dysbiosis with insulin resistance and systemic chronic inflammation cooperatively. In this review, we will describe the link between periodontal disease and NAFLD with a focus on basic, epidemiological, and clinical studies, and discuss potential mechanisms linking the two diseases and possible therapeutic approaches focused on the microbiome. In conclusion, it is presumed that the pathogenesis of NAFLD involves a complex crosstalk between periodontal disease, gut microbiota, and metabolic syndrome. Thus, the conventional periodontal treatment and novel microbiome-targeted therapies that include probiotics, prebiotics and bacteriocins would hold great promise for preventing the onset and progression of NAFLD and subsequent complications in patients with periodontal disease.
Collapse
Affiliation(s)
- Ryutaro Kuraji
- Department of Periodontology, The Nippon Dental University School of Life Dentistry at Tokyo, Tokyo 102-0071, Japan
- Department of Orofacial Sciences, University of California San Francisco, San Francisco, CA 94143, United States
| | - Takahiko Shiba
- Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Boston, MA 02115, United States
- Department of Periodontology, Tokyo Medical and Dental University, Tokyo 113-8549, Japan
| | - Tien S Dong
- The Vatche and Tamar Manoukian Division of Digestive Diseases, University of California Los Angeles, Department of Medicine, University of California David Geffen School of Medicine, Los Angeles, CA 90095, United States
| | - Yukihiro Numabe
- Department of Periodontology, The Nippon Dental University School of Life Dentistry at Tokyo, Tokyo 102-8159, Japan
| | - Yvonne L Kapila
- Department of Orofacial Sciences, University of California San Francisco, San Francisco, CA 94143, United States
- Sections of Biosystems and Function and Periodontics, Professor and Associate Dean of Research, Felix and Mildred Yip Endowed Chair in Dentistry, University of California Los Angeles, Los Angeles, CA 90095, United States
| |
Collapse
|
39
|
Flynn JK, Ortiz AM, Herbert R, Brenchley JM. Host Genetics and Environment Shape the Composition of the Gastrointestinal Microbiome in Nonhuman Primates. Microbiol Spectr 2023; 11:e0213922. [PMID: 36475838 PMCID: PMC9927375 DOI: 10.1128/spectrum.02139-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 11/16/2022] [Indexed: 12/13/2022] Open
Abstract
The bacterial component of the gastrointestinal tract microbiome is comprised of hundreds of species, the majority of which live in symbiosis with the host. The bacterial microbiome is influenced by host diet and disease history, and host genetics may additionally play a role. To understand the degree to which host genetics shapes the gastrointestinal tract microbiome, we studied fecal microbiomes in 4 species of nonhuman primates (NHPs) held in separate facilities but fed the same base diet. These animals include Chlorocebus pygerythrus, Chlorocebus sabaeus, Macaca mulatta, and Macaca nemestrina. We also followed gastrointestinal tract microbiome composition in 20 Macaca mulatta (rhesus macaques [RMs]) as they transitioned from an outdoor to indoor environment and compared 6 Chlorocebus pygerythrus monkeys that made the outdoor to indoor transition to their 9 captive-born offspring. We found that genetics can influence microbiome composition, with animals of different genera (Chlorocebus versus Macaca) having significantly different gastrointestinal (GI) microbiomes despite controlled diets. Animals within the same genera have more similar microbiomes, although still significantly different, and animals within the same species have even more similar compositions that are not significantly different. Significant differences were also not observed between wild-born and captive-born Chlorocebus pygerythrus, while there were significant changes in RMs as they transitioned into captivity. Together, these results suggest that the effects of captivity have a larger impact on the microbiome than other factors we examined within a single NHP species, although host genetics does significantly influence microbiome composition between NHP genera and species. IMPORTANCE Our data point to the degree to which host genetics can influence GI microbiome composition and suggest, within primate species, that individual host genetics is unlikely to significantly alter the microbiome. These data are important for the development of therapeutics aimed at altering the microbiome within populations of genetically disparate members of primate species.
Collapse
Affiliation(s)
- Jacob K. Flynn
- Barrier Immunity Section, Lab of Viral Diseases, NIAID, NIH, Bethesda, Maryland, USA
| | - Alexandra M. Ortiz
- Barrier Immunity Section, Lab of Viral Diseases, NIAID, NIH, Bethesda, Maryland, USA
| | - Richard Herbert
- Comparative Medicine Branch, NIAID, NIH, Bethesda, Maryland, USA
| | - Jason M. Brenchley
- Barrier Immunity Section, Lab of Viral Diseases, NIAID, NIH, Bethesda, Maryland, USA
| |
Collapse
|
40
|
Animal Age Affects the Gut Microbiota and Immune System in Captive Koalas ( Phascolarctos cinereus). Microbiol Spectr 2023; 11:e0410122. [PMID: 36602319 PMCID: PMC9927321 DOI: 10.1128/spectrum.04101-22] [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] [Indexed: 01/06/2023] Open
Abstract
Gut microbiota is one of the major elements in the control of host health. However, the composition of gut microbiota in koalas has rarely been investigated. Here, we performed 16S rRNA gene sequencing to determine the individual and environmental determinants of gut microbiota diversity and function in 35 fecal samples collected from captive koalas. Meanwhile, blood immune-related cytokine levels were examined by quantitative reverse transcription-PCR to initially explore the relationship between the gut microbiota and the immune system in koalas. The relative abundance of many bacteria, such as Lonepinella koalarum, varies at different ages in koalas and decreases with age. Conversely, Ruminococcus flavefaciens increases with age. Moreover, bacterial pathways involved in lipid metabolism, the biosynthesis of other secondary metabolites, and infectious disease show a significant correlation with age. Age affects the relationship between the microbiota and the host immune system. Among them, the gut microbiota of subadult and aged koalas was closely correlated with CD8β and CD4, whereas adult koalas were correlated with CLEC4E. We also found that sex, reproductive status, and living environment have little impact on the koala gut microbiota and immune system. These results shed suggest age is a key factor affecting gut microbiota and immunity in captive koalas and thus provide new insight into its role in host development and the host immune system. IMPORTANCE Although we have a preliminary understanding of the gut microbiota of koalas, we lack insight into which factors potentially impact captive koalas. This study creates the largest koala gut microbiota data set in China to date and describes several factors that may affect gut microbiota and the immune system in captive koalas, highlighting that age may be a key factor affecting captive koalas. Moreover, this study is the first to characterize the correlation between gut microbiota and cytokines in koalas. Better treatment strategies for infectious disorders may be possible if we can better understand the interactions between the immune system and the microbiota.
Collapse
|
41
|
Dietary Intake and Systemic Inflammation: Can We Use Food as Medicine? Curr Nutr Rep 2023:10.1007/s13668-023-00458-z. [PMID: 36662358 DOI: 10.1007/s13668-023-00458-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2022] [Indexed: 01/21/2023]
Abstract
PURPOSE OF REVIEW This review summarizes the current literature regarding the association between diet and systemic inflammation. RECENT FINDINGS Data in humans suggests that consumption of plant-based nutrients is associated with a reduction in systemic inflammation, while consumption of red meat and excessive dairy has the opposite effect and may increase risk of acute flares in those that suffer from certain chronic diseases like inflammatory bowel disease or psoriasis and certain cancers. There is a known association between diet and systemic inflammation; thus, we recommend that clinicians discuss plant-based, whole food diets with patients, particularly those that suffer from chronic inflammatory diseases as an adjunct treatment for these conditions. Future research should evaluate whether adherence to these types of diets is sustainable in the long term and how these changes affect important quality of life concerns.
Collapse
|
42
|
Thriene K, Michels KB. Human Gut Microbiota Plasticity throughout the Life Course. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:1463. [PMID: 36674218 PMCID: PMC9860808 DOI: 10.3390/ijerph20021463] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/06/2023] [Accepted: 01/09/2023] [Indexed: 06/02/2023]
Abstract
The role of the gut microbiota in human health and disease has garnered heightened attention over the past decade. A thorough understanding of microbial variation over the life course and possible ways to influence and optimize the microbial pattern is essential to capitalize on the microbiota's potential to influence human health. Here, we review our current understanding of the concept of plasticity of the human gut microbiota throughout the life course. Characterization of the plasticity of the microbiota has emerged through recent research and suggests that the plasticity in the microbiota signature is largest at birth when the microbial colonization of the gut is initiated and mode of birth imprints its mark, then decreases postnatally continuously and becomes less malleable and largely stabilized with advancing age. This continuing loss of plasticity has important implication for the impact of the exposome on the microbiota and health throughout the life course and the identification of susceptible 'windows of opportunity' and methods for interventions.
Collapse
Affiliation(s)
- Kerstin Thriene
- Institute for Prevention and Cancer Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, 79110 Freiburg, Germany
| | - Karin B. Michels
- Institute for Prevention and Cancer Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, 79110 Freiburg, Germany
- Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
43
|
Strobel KM, Del Vecchio G, Devaskar SU, Calkins KL. Gut Microbes and Circulating Cytokines in Preterm Infants with Growth Failure. J Nutr 2023; 153:120-130. [PMID: 36913445 PMCID: PMC10196572 DOI: 10.1016/j.tjnut.2022.10.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 10/12/2022] [Accepted: 10/23/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Growth failure (GF) is a multifactorial problem in preterm infants. The intestinal microbiome and inflammation may contribute to GF. OBJECTIVES This study's objective was to compare the gut microbiome and plasma cytokines in preterm infants with and without GF. METHODS This was a prospective cohort study of infants with birth weights of <1750 g. Infants with a weight or length z-score change from birth to discharge or death that was less than or equal to -0.8 (GF group) were compared with infants without GF [control (CON) group]. The primary outcome was the gut microbiome (at weeks 1-4 of age), assessed by 16S rRNA gene sequencing using Deseq2. Secondary outcomes included inferred metagenomic function and plasma cytokines. Phylogenetic Investigation of Communities by Reconstruction of Unobserved States determined metagenomic function, which was compared using ANOVA. Cytokines were measured by 2-multiplexed immunometric assays and compared using Wilcoxon tests and linear mixed models. RESULTS GF (n = 14) and CON group (n = 13) had similar median (IQR) birth weight (1380 [780-1578] g vs. 1275 [1013-1580] g) and gestational age (29 [25-31] weeks vs. 30 [29-32] weeks). Compared with the CON group, the GF group had a greater abundance of Escherichia/Shigella in weeks 2 and 3, Staphylococcus in week 4, and Veillonella in weeks 3 and 4 (P-adjusted < 0.001 for all). Plasma cytokine concentrations did not differ significantly between the cohorts. When all time points are combined, fewer microbes were involved in TCA cycle activity in the GF group compared with the CON group (P = 0.023). CONCLUSIONS In this study, when compared with CON infants, GF infants had a distinct microbial signature with increased Escherichia/Shigella and Firmicutes and fewer microbes associated with energy production at later weeks of hospitalization. These findings may suggest a mechanism for aberrant growth.
Collapse
Affiliation(s)
- Katie M Strobel
- Department of Pediatrics, Division of Neonatology and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA; The University of California Los Angeles Children's Discovery and Innovation Institute, University of California Los Angeles, Los Angeles, CA, USA
| | - Giorgia Del Vecchio
- Department of Pediatrics, Division of Neonatology and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA; The University of California Los Angeles Children's Discovery and Innovation Institute, University of California Los Angeles, Los Angeles, CA, USA
| | - Sherin U Devaskar
- Department of Pediatrics, Division of Neonatology and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA; The University of California Los Angeles Children's Discovery and Innovation Institute, University of California Los Angeles, Los Angeles, CA, USA
| | - Kara L Calkins
- Department of Pediatrics, Division of Neonatology and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA; The University of California Los Angeles Children's Discovery and Innovation Institute, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
44
|
Watson MM, van der Giezen M, Søreide K. Gut Microbiome Influence on Human Epigenetics, Health, and Disease. HANDBOOK OF EPIGENETICS 2023:669-686. [DOI: 10.1016/b978-0-323-91909-8.00012-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
45
|
Tian M, Li Q, Zheng T, Yang S, Chen F, Guan W, Zhang S. Maternal microbe-specific modulation of the offspring microbiome and development during pregnancy and lactation. Gut Microbes 2023; 15:2206505. [PMID: 37184203 DOI: 10.1080/19490976.2023.2206505] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/16/2023] Open
Abstract
The maternal microbiome is essential for the healthy growth and development of offspring and has long-term effects later in life. Recent advances indicate that the maternal microbiome begins to regulate fetal health and development during pregnancy. Furthermore, the maternal microbiome continues to affect early microbial colonization via birth and breastfeeding. Compelling evidence indicates that the maternal microbiome is involved in the regulation of immune and brain development and affects the risk of related diseases. Modulating offspring development by maternal diet and probiotic intervention during pregnancy and breastfeeding could be a promising therapy in the future. In this review, we summarize and discuss the current understanding of maternal microbiota development, perinatal microbial metabolite transfer, mother-to-infant microbial transmission during/after birth and its association with immune and brain development as well as corresponding diseases.
Collapse
Affiliation(s)
- Min Tian
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Qihui Li
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Tenghui Zheng
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Siwang Yang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Fang Chen
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, China
| | - Wutai Guan
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, China
| | - Shihai Zhang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, China
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
46
|
Liu Z, Gonzalez-Nahm S, Zhang G, Dorsey A, Sela DA. Nutrition and the Gut Microbiome: Insights into New Dietary Strategies for Health. Nutr Health 2023:307-322. [DOI: 10.1007/978-3-031-24663-0_25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
47
|
Wang S, Cui J, Jiang S, Zheng C, Zhao J, Zhang H, Zhai Q. Early life gut microbiota: Consequences for health and opportunities for prevention. Crit Rev Food Sci Nutr 2022; 64:5793-5817. [PMID: 36537331 DOI: 10.1080/10408398.2022.2158451] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The gut microbiota influences many aspects of the host, including immune system maturation, nutrient absorption and metabolism, and protection from pathogens. Increasing evidences from cohort and animal studies indicate that changes in the gut microbiota early in life increases the risk of developing specific diseases early and later in life. Therefore, it is becoming increasingly important to identify specific disease prevention or therapeutic solutions targeting the gut microbiota, especially during infancy, which is the window of the human gut microbiota establishment process. In this review, we provide an overview of current knowledge concerning the relationship between disturbances in the gut microbiota early in life and health consequences later in life (e.g., necrotizing enterocolitis, celiac disease, asthma, allergies, autism spectrum disorders, overweight/obesity, diabetes and growth retardation), with a focus on changes in the gut microbiota prior to disease onset. In addition, we summarize and discuss potential microbiota-based interventions early in life (e.g., diet adjustments, probiotics, prebiotics, fecal microbiota transplantation, environmental changes) to promote health or prevent the development of specific diseases. This knowledge should aid the understanding of early life microbiology and inform the development of prediction and prevention measures for short- and long-term health disorders based on the gut microbiota.
Collapse
Affiliation(s)
- Shumin Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Jingjing Cui
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Shilong Jiang
- Nutrition and Metabolism Research Division, Innovation Center, Heilongjiang Feihe Dairy Co., Ltd, Beijing, China
- PKUHSC-China Feihe Joint Research Institute of Nutrition and Healthy Lifespan Development, Beijing, China
| | - Chengdong Zheng
- Nutrition and Metabolism Research Division, Innovation Center, Heilongjiang Feihe Dairy Co., Ltd, Beijing, China
- PKUHSC-China Feihe Joint Research Institute of Nutrition and Healthy Lifespan Development, Beijing, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Heng Zhang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- Department of Child Health Care, Wuxi Maternity and Child Health Care Hospital, Wuxi, Jiangsu, China
| | - Qixiao Zhai
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| |
Collapse
|
48
|
Loganathan T, Priya Doss C G. The influence of machine learning technologies in gut microbiome research and cancer studies - A review. Life Sci 2022; 311:121118. [DOI: 10.1016/j.lfs.2022.121118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 10/19/2022] [Accepted: 10/19/2022] [Indexed: 11/18/2022]
|
49
|
Li H, Wang Y, Shao S, Yu H, Wang D, Li C, Yuan Q, Liu W, Cao J, Wang X, Guo H, Wu X, Wang S. Rabdosia serra alleviates dextran sulfate sodium salt-induced colitis in mice through anti-inflammation, regulating Th17/Treg balance, maintaining intestinal barrier integrity, and modulating gut microbiota. J Pharm Anal 2022; 12:824-838. [PMID: 36605573 PMCID: PMC9805946 DOI: 10.1016/j.jpha.2022.08.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 07/27/2022] [Accepted: 08/02/2022] [Indexed: 02/01/2023] Open
Abstract
Rabdosia serra (R. serra), an important component of Chinese herbal tea, has traditionally been used to treat hepatitis, jaundice, cholecystitis, and colitis. However, the chemical composition of R. serra and its effect against colitis remain unclear. In this study, the chemical composition of the water extract of R. serra was analyzed using ultra performance liquid chromatography coupled with a hybrid linear ion trap quadrupole-orbitrap mass spectrometer (UPLC-LTQ-Orbitrap-MS). A total of 46 compounds, comprising ent-kaurane diterpenoids, flavonoids, phenolic acids, and steroids, were identified in the water extract of R. serra, and the extract could significantly alleviate dextran sulfate sodium salt-induced colitis by improving colon length, upregulating anti-inflammatory factors, downregulating proinflammatory factors, and restoring the balance of T helper 17/T regulatory cells. R. serra also preserved intestinal barrier function by increasing the level of tight junction proteins (zonula occludens 1 and occludin) in mouse colonic tissue. In addition, R. serra modulated the gut microbiota composition by increasing bacterial richness and diversity, increasing the abundance of beneficial bacteria (Muribaculaceae, Bacteroides, Lactobacillus, and Prevotellaceae_UCG-001), and decreasing the abundance of pathogenic bacteria (Turicibacter, Eubacterium_fissicatena_group, and Eubacterium_xylanophilum_group). Gut microbiota depletion by antibiotics further confirmed that R. serra alleviated colitis in a microbiota-dependent manner. Overall, our findings provide chemical and biological evidence for the potential application of R. serra in the management of colitis.
Collapse
Affiliation(s)
- Hongyi Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078, China
| | - Yi Wang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Shumin Shao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078, China
| | - Hui Yu
- Hutchison Whampoa Guangzhou Baiyunshan Chinese Medicine Co., Ltd., Guangzhou, 510000, China
| | - Deqin Wang
- Hutchison Whampoa Guangzhou Baiyunshan Chinese Medicine Co., Ltd., Guangzhou, 510000, China
| | - Chuyuan Li
- Hutchison Whampoa Guangzhou Baiyunshan Chinese Medicine Co., Ltd., Guangzhou, 510000, China
| | - Qin Yuan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078, China
| | - Wen Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078, China
| | - Jiliang Cao
- College of Pharmacy, Shenzhen Technology University, Shenzhen, Guangdong, 518118, China
| | - Xiaojuan Wang
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China
| | - Haibiao Guo
- Hutchison Whampoa Guangzhou Baiyunshan Chinese Medicine Co., Ltd., Guangzhou, 510000, China
| | - Xu Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
- Corresponding author.
| | - Shengpeng Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078, China
- Macau Centre for Research and Development in Chinese Medicine, University of Macau, Macao, 999078, China
- Corresponding author. State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078, China.
| |
Collapse
|
50
|
Abomoelak B, Saps M, Sudakaran S, Deb C, Mehta D. Gut Microbiome Remains Static in Functional Abdominal Pain Disorders Patients Compared to Controls: Potential for Diagnostic Tools. BIOTECH 2022; 11:biotech11040050. [PMID: 36412751 PMCID: PMC9680443 DOI: 10.3390/biotech11040050] [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: 09/15/2022] [Revised: 10/25/2022] [Accepted: 10/26/2022] [Indexed: 11/07/2022] Open
Abstract
Background: Functional Abdominal Pain disorders (FAPDs) are a group of heterogeneous gastrointestinal disorders with unclear pathophysiology. In children, FAPDs are more common in the winter months than summer months. The possible influence of school stressors has been proposed. Previously, our group showed differences in bacterial relative abundances and alpha diversity in the gut microbiome and its relationship with stressors in a cross-sectional evaluation of children suffering from FAPDs compared to a healthy control group. We present longitudinal data to assess whether the gut microbiome changes over school terms in the control and FAPDs groups. Methods: The longitudinal study included children with FAPDs (n = 28) and healthy controls (n = 54). Gastrointestinal symptoms, as well as stool microbiome, were assessed in both groups. Stool samples were serially collected from all participants during both the school term and summer vacation. The stool samples were subjected to total genomic extraction, 16S rRNA amplicon sequencing, and bioinformatics analysis. The gut microbiome was compared at school and during vacation. Other metrics, alpha diversity, and beta diversity, were also compared between the two school terms in every group. Results: In the healthy group, there were differences in microbiome composition between school terms and summer vacation. Conversely, we found no differences in the FAPDs group between the two terms. The healthy control group revealed differences (p-value < 0.05) in 55 bacterial species between the school term and vacation. Several of the differentially abundant identified bacteria were involved in short-chain fatty acids production (SCFAs), inflammation reduction, and gut homeostasis. Alpha diversity metrics, such as the Shannon index, were different in the control group and remained unchanged in the FAPDs group. Conclusion: Although preliminary, our findings suggest that the gut microbiome is static in FAPDs. This compares with a more dynamic healthy gut microbiome. Further studies are warranted to corroborate this and understand the interplay between stress, symptoms, and a less diverse and static microbiome. Future studies will also account for different variables such as diet and other patient demographic criteria that were missing in the current study.
Collapse
Affiliation(s)
- Bassam Abomoelak
- Arnold Palmer Pediatric Gastroenterology Clinic, Orlando Health, Orlando, FL 32806, USA
| | - Miguel Saps
- Pediatric Gastroenterology, Hepatology and Nutrition Division, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | | | - Chirajyoti Deb
- Arnold Palmer Pediatric Gastroenterology Clinic, Orlando Health, Orlando, FL 32806, USA
| | - Devendra Mehta
- Arnold Palmer Pediatric Gastroenterology Clinic, Orlando Health, Orlando, FL 32806, USA
- Correspondence:
| |
Collapse
|