1
|
Liu J, Sun Q, Tang L, Lv D, Chen Y, Ye F, Liu D, Liang H, Wang C, Zhang Q. Widely targeted plasma lipidomic analysis of Term Low Birth Weight Infants: unraveling signatures and implications for early growth patterns. J Pharm Biomed Anal 2025; 258:116732. [PMID: 39933396 DOI: 10.1016/j.jpba.2025.116732] [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: 11/17/2024] [Revised: 01/15/2025] [Accepted: 02/02/2025] [Indexed: 02/13/2025]
Abstract
Low birth weight is recognized as a risk factor for adult metabolic and cardiovascular diseases. This study investigates whether term low birth weight (TLBW) neonates, who have been exposed to unfavorable settings, demonstrate compromised lipid metabolism. A widely targeted lipidomic analysis was conducted using ultra performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) on 59 plasma samples (28 TLBW and 31 term normal birth weight (TNBW) neonates. We conducted univariate and multivariate analyses to identify differential lipids. Spearman's correlation coefficient assessed the association between lipid content at birth and Z-scores for the subsequent physical growth of enrolled children. A total of 1523 lipids in 36 subcategories across 6 major categories were detected. 269 differential lipids were discerned, with 114 up-regulated and 155 down-regulated. In the TLBW group, we observed higher levels of sphingomyelins (including SM(d18:1/16:1), SM(d18:2/23:1), SM(d18:1/22:0), and SM(d18:2/24:1)), Hex3Cer(d18:1/16:0), as well as phosphatidylcholines (PC(O-14:0_20:4) and PC(O-16:0_20:4)), and cholesterol esters (CE (20:4)). In contrast, phosphatidylethanolamines (PE) such as PE (18:2_22:1), PE (18:1_22:1), and triglyceride (TG(10:0_16:2_18:2)) were lower. The KEGG enrichment analysis revealed a consistent alteration in both sphingolipid metabolism and steroid biosynthesis. Moreover, PC(O-16:0_20:4), Hex3Cer(d18:1/16:0), and CE(20:4) exhibited positive correlations with the Z-score of height-for-age at follow-up, while PE(O-18:1_24:4) and PS(20:2_22:4) showed negative correlations with the Z-score of weight-for-age. Our findings reveal novel lipidomic differences between TLBW and TNBW neonates. The observed lipid variations at birth, including sphingomyelins and glycerophospholipids, could affect subsequent growth. Further studies are needed to validate these findings in diverse populations, address confounding factors, and investigate underlying mechanisms.
Collapse
Affiliation(s)
- Jing Liu
- Department of Pediatrics, China-Japan Friendship Hospital, Beijing, China; Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Qi Sun
- Department of Pediatrics, China-Japan Friendship Hospital, Beijing, China
| | - Lijuan Tang
- Department of Pediatrics, China-Japan Friendship Hospital, Beijing, China
| | - Di Lv
- Department of Pediatrics, China-Japan Friendship Hospital, Beijing, China; Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yuanmei Chen
- Department of Pediatrics, China-Japan Friendship Hospital, Beijing, China
| | - Fang Ye
- Department of Pediatrics, China-Japan Friendship Hospital, Beijing, China
| | - Die Liu
- Department of Pediatrics, China-Japan Friendship Hospital, Beijing, China
| | - Haixiao Liang
- Department of Pediatrics, China-Japan Friendship Hospital, Beijing, China
| | - Chao Wang
- Department of Pediatrics, China-Japan Friendship Hospital, Beijing, China
| | - Qi Zhang
- Department of Pediatrics, China-Japan Friendship Hospital, Beijing, China; Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
2
|
Li B, Liang C, Xu B, Song P, Liu D, Zhang J, Gu H, Jiang F, Gao H, Cai Z, Zhang T. Extreme winter environment dominates gut microbiota and metabolome of white-lipped deer. Microbiol Res 2025; 297:128182. [PMID: 40252261 DOI: 10.1016/j.micres.2025.128182] [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: 07/25/2024] [Revised: 03/23/2025] [Accepted: 04/10/2025] [Indexed: 04/21/2025]
Abstract
Qinghai-Tibet Plateau (QTP) is marked by harsh environments that drive the evolution of unique nutrient metabolism mechanism in indigenous animal gut microbiotas. Yet, responses of these microbiotas to different extreme environments remain poorly understood. White-lipped deer (Przewalskium albirostris), a native endangered species in the QTP, serves as an ideal model to study how gut microbiotas adapt to season and human disturbances. Here, a multi-omics integrated analysis of 16S rRNA, metagenomics, and untargeted metabolomics was performed to investigate the composition, function, and metabolic characteristics of gut microbiota in White-lipped deer across different seasons and living environments. Our results revealed that extreme winter environment dominated the composition, function, and metabolism of gut microbiota in white-lipped deer. The white-lipped deer exhibited an enriched gut microbiota associated with producing short-chain fatty acids in winter, with core feature genera including norank_o_Rhodospirillales, Rikenellaceae_RC9_gut_group, and unclassified_c_Clostridia. However, potential pathogenic bacteria and few short-chain fatty acid producers, with core feature genera including norank_f_p-2534-18B5_gut_group, Cellulosilyticum, and Paeniclostridium, showed enrichment in captivity. Pathways associated with carbohydrate metabolism, amino acid metabolism, and immune regulation showed enrichment in winter group as an adaptation to the cold and food scarcity. Among these, Rikenellaceae_RC9_gut_group and unclassified_c_Clostridia contributed significantly to these metabolic pathways. The gut microbiota of white-lipped deer exhibited enrichment in pathways related to intestinal inflammation and enhanced immune regulation to alleviate the stress of captivity. Among these, norank_f_p-2534-18B5_gut_group contributed the most to these pathways. Butyric, valeric, and valproic acids were significantly more abundant in the winter group, while 3-hydroxybutyric and (S)-beta-aminoisobutyric acids were higher in the captive group. Furthermore, enriched metabolites and associated pathways in both groups further supported the inferences on metagenomic functions. This study confirms the key role of specific gut microbiota in adapting to high-altitude winters and anthropogenic disturbances, emphasizing its importance for environmental resilience in wild, high-altitude mammals.
Collapse
Affiliation(s)
- Bin Li
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China; Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Xining, China
| | - Chengbo Liang
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China; Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Xining, China
| | - Bo Xu
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China; Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Xining, China
| | - Pengfei Song
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China; Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Xining, China
| | | | | | - Haifeng Gu
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, China; Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Xining, China
| | - Feng Jiang
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, China; Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Xining, China
| | - Hongmei Gao
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, China; Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Xining, China
| | - Zhenyuan Cai
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, China; Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Xining, China.
| | - Tongzuo Zhang
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, China; Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Xining, China.
| |
Collapse
|
3
|
Chen H, Li J, Wu Y, Li Y, Zheng S, Wu Y, Xuan R, Wu L, Miao J, Wang Y, Tan H, Zhou J, Huang J, Yan X. Structural characteristics of intestinal microbiota of domestic ducks with different body sizes. Poult Sci 2025; 104:104930. [PMID: 40056781 PMCID: PMC11930160 DOI: 10.1016/j.psj.2025.104930] [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: 11/17/2024] [Revised: 02/14/2025] [Accepted: 02/19/2025] [Indexed: 03/10/2025] Open
Abstract
Domestic ducks are economically important agricultural animals, and their body size is a crucial economic trait. The intestinal flora plays a pivotal role in influencing body metabolism, growth, and development. Currently, no literature is available on the potential effect of the intestinal flora of domestic ducks on body size. This study used 16S rRNA sequencing technology to investigate the fecal microbiota of 229 individuals reared under identical feeding conditions. The findings revealed that partridge ducks with large body sizes (LBS) exhibited a higher level of intestinal microbial diversity than ducks with small body sizes (SBS). Notably, the gut microbiota composition of SBS displayed significantly elevated proportions of Streptococcus, Rothia, and Psychrobacter compared to their counterparts with LBS. Conversely, Lactobacillus was significantly more abundant in LBS. Jeotgalibaca and Psychrobacter were identified as key biomarkers of SBS, whereas Lactobacillus and Bacteroides were predominant biomarkers of LBS. Functional predictions based on intestinal microbiota indicated discernible differences among different body types, particularly evident in non- partridge ducks. The present study investigated the correlation between the intestinal microbiota and body size of domestic ducks, aiming to provide practical insights for the production management of domestic duck farming.
Collapse
Affiliation(s)
- Hao Chen
- Key Laboratory of Natural Microbial Medicine Research of Jiangxi Province, College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Jiawei Li
- Key Laboratory of Natural Microbial Medicine Research of Jiangxi Province, College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Yongfei Wu
- Key Laboratory of Natural Microbial Medicine Research of Jiangxi Province, College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Yuhang Li
- Key Laboratory of Natural Microbial Medicine Research of Jiangxi Province, College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Sumei Zheng
- Key Laboratory of Natural Microbial Medicine Research of Jiangxi Province, College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Yan Wu
- Key Laboratory of Natural Microbial Medicine Research of Jiangxi Province, College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Rui Xuan
- Key Laboratory of Natural Microbial Medicine Research of Jiangxi Province, College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Liping Wu
- Key Laboratory of Natural Microbial Medicine Research of Jiangxi Province, College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Junjie Miao
- Key Laboratory of Natural Microbial Medicine Research of Jiangxi Province, College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Yanan Wang
- Key Laboratory of Natural Microbial Medicine Research of Jiangxi Province, College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Hongli Tan
- Key Laboratory of Natural Microbial Medicine Research of Jiangxi Province, College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Jing Zhou
- Key Laboratory of Natural Microbial Medicine Research of Jiangxi Province, College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Jianhua Huang
- Key Laboratory of Natural Microbial Medicine Research of Jiangxi Province, College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang 330013, China.
| | - Xueming Yan
- Key Laboratory of Natural Microbial Medicine Research of Jiangxi Province, College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang 330013, China.
| |
Collapse
|
4
|
He P, Lyu Y, Shen X, Liu W, Zhang Y, Li Y, Huang Y, Xu L, Zhang L, Guo S. Gut microbiome and short-chain fatty acids associated with the efficacy of growth hormone treatment in children with short stature. Front Pediatr 2025; 13:1557878. [PMID: 40230807 PMCID: PMC11994682 DOI: 10.3389/fped.2025.1557878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/14/2025] [Indexed: 04/16/2025] Open
Abstract
Objective To investigate associations between fecal microbiota, short-chain fatty acids (SCFAs), and the efficacy of recombinant human growth hormone (rhGH) treatment in children with growth hormone deficiency (GHD) or idiopathic short stature (ISS). Methods A 2-phase cohort study was conducted. Phase I included 102 participants (GHD: n = 33, ISS: n = 28, controls: n = 41) for cross-sectional analysis using 16S rRNA sequencing and targeted metabolomics to compare microbial diversity, predicted metabolic pathways, and SCFA levels. Phase II longitudinally monitored 61 rhGH-treated children (GHD = 33, ISS = 28) over 2 years, assessing growth velocity, IGF-1 levels, and fecal microbiota/SCFA dynamics. Statistical analyses included alpha/beta diversity metrics, LEfSe, PERMANOVA, and redundancy analysis (RDA) to link microbial/SCFA profiles with clinical outcomes. Results (1). Microbiota Dysbiosis: Untreated GHD/ISS children exhibited reduced beneficial taxa (e.g., Faecalibacterium, Akkermansia) and increased pathobionts (e.g., Streptococcus, Collinsella) compared to controls (PERMANOVA: R 2 = 0.114, P = 0.001). (2). Metabolic Pathways: GHD/ISS groups showed enrichment in xenobiotic degradation (e.g., atrazine) and deficits in nutrient-associated pathways (e.g., carotenoid biosynthesis). (3). rhGH Effects: Treatment increased beneficial taxa (e.g., Bifidobacterium, Faecalibacterium) and modulated amino acid/lipid metabolism pathways (e.g., glycine-serine-threonine metabolism, P = 0.035). (4). SCFAs and Growth Velocity: Higher growth velocity percentiles correlated with elevated acetic acid (GHD-treated: 1952 ± 962.4 vs. untreated: 1290 ± 886.0 μg/g, P = 0.037) and butyric acid levels. Conclusion GHD, ISS, and healthy children have different fecal microbiota compositions and SCFA metabolisms. rhGH therapy partially restores microbial balance and alters metabolic pathways, with SCFA levels associated with treatment efficacy. These findings highlight the gut microbiome as a potential modulator of rhGH response and provide insight into microbiota-targeted therapies to improve growth outcomes (e.g., "probiotic interventions").
Collapse
Affiliation(s)
- Pingsihua He
- Department of Endocrine and Metabolism, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yongfen Lyu
- Department of Endocrine and Metabolism, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xinyuan Shen
- Department of Endocrine and Metabolism, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wenxin Liu
- Department of Endocrine and Metabolism, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ying Zhang
- Department of Endocrine and Metabolism, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yan Li
- Department of Endocrine and Metabolism, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yuying Huang
- Department of Traditional Chinese Medicine, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Liya Xu
- Department of Endocrine and Metabolism, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Liang Zhang
- Department of Endocrine and Metabolism, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Sheng Guo
- Department of Endocrine and Metabolism, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
5
|
Ji P, Wang N, Yu Y, Zhu J, Zuo Z, Zhang B, Zhao F. Single-cell delineation of the microbiota-gut-brain axis: Probiotic intervention in Chd8 haploinsufficient mice. CELL GENOMICS 2025; 5:100768. [PMID: 39914389 PMCID: PMC11872533 DOI: 10.1016/j.xgen.2025.100768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 12/02/2024] [Accepted: 01/14/2025] [Indexed: 02/16/2025]
Abstract
Emerging research underscores the gut microbiome's impact on the nervous system via the microbiota-gut-brain axis, yet comprehensive insights remain limited. Using a CHD8-haploinsufficient model for autism spectrum disorder (ASD), we explored host-gut microbiota interactions by constructing a single-cell transcriptome atlas of brain and intestinal tissues in wild-type and mutant mice across three developmental stages. CHD8 haploinsufficiency caused delayed development of radial glial precursors and excitatory neural progenitors in the E14.5 brain, inflammation in the adult brain, immunodeficiency, and abnormal intestinal development. Selective CHD8 knockdown in intestinal epithelial cells generated Chd8ΔIEC mice, which exhibited normal sociability but impaired social novelty recognition. Probiotic intervention with Lactobacillus murinus selectively rescued social deficits in Chd8ΔIEC mice, with single-cell transcriptome analysis revealing underlying mechanisms. This study provides a detailed single-cell transcriptomic dataset of ASD-related neural and intestinal changes, advancing our understanding of the gut-brain axis and offering potential therapeutic strategies for ASD.
Collapse
Affiliation(s)
- Peifeng Ji
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Ning Wang
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - You Yu
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Junjie Zhu
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China; Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - Zhenqiang Zuo
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Bing Zhang
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Fangqing Zhao
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China; Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China.
| |
Collapse
|
6
|
Zha Z, Jia C, Zhou R, Yin Q, Hu Y, Huang Z, Peng L, Zhang Y, Qiu X, Chen Y, Zhong Y, Wang Y, Pang M, Lu S, Sheng C, Huang L. Clostridium difficile-derived membrane vesicles promote fetal growth restriction via inhibiting trophoblast motility through PPARγ/RXRα/ANGPTL4 axis. NPJ Biofilms Microbiomes 2024; 10:158. [PMID: 39741137 DOI: 10.1038/s41522-024-00630-5] [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: 07/30/2024] [Accepted: 12/07/2024] [Indexed: 01/02/2025] Open
Abstract
Fetal growth restriction (FGR) is a common complication of pregnancy, which seriously endangers fetal health and still lacks effective therapeutic targets. Clostridium difficile (C. difficile) is associated with fetal birth weight, and its membrane vesicles (MVs) are pathogenic vectors. However, the role of C. difficile and its MVs in FGR remains unclear. Here we found that supplementation with C. difficile altered the characteristics of gut microbiota and reduced the birth weight in mice. Interestingly, C. difficile MVs entered placenta, inhibited trophoblast motility, and induced fetal weight loss in mice. Mechanistically, C. difficile MVs activated the PPAR pathway via enhancing the transcriptional activity of PPARγ promoter, consequently inhibiting trophoblast motility. Moreover, PPARγ expression was significantly elevated in FGR placenta, and negatively correlated with fetal birth weight. Together, our findings reveal the significance of C. difficile and its MVs in FGR, providing new insights into the mechanisms of FGR development.
Collapse
Affiliation(s)
- Zhiqiang Zha
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Chunhong Jia
- Department of Neonatology, Guangzhou Key Laboratory of Neonatal Intestinal Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ruisi Zhou
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Qinlan Yin
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yu Hu
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhipeng Huang
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Linyu Peng
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yichi Zhang
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaowei Qiu
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Ying Chen
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yawen Zhong
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yu Wang
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Menglan Pang
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Shijing Lu
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Chao Sheng
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| | - Liping Huang
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
7
|
Kawikova I, Hakenova K, Lebedeva M, Kleteckova L, Jakob L, Spicka V, Wen L, Spaniel F, Vales K. Perinatal Hypoxia and Immune System Activation in Schizophrenia Pathogenesis: Critical Considerations During COVID-19 Pandemic. Physiol Res 2024; 73:S615-S639. [PMID: 39589306 PMCID: PMC11627263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 10/01/2024] [Indexed: 11/27/2024] Open
Abstract
Schizophrenia, a severe psychiatric, neurodevelopmental disorder affecting about 0.29-1 % of the global population, is characterized by hallucinations, delusions, cognitive impairments, disorganized thoughts and speech, leading to significant social withdrawal and emotional blunting. During the 1980s, considerations about diseases that result from complex interactions of genetic background and environmental factors started to appear. One of the critical times of vulnerability is the perinatal period. Concerning schizophrenia, obstetric complications that are associated with hypoxia of the fetus or neonate were identified as a risk. Also, maternal infections during pregnancy were linked to schizophrenia by epidemiological, serologic and genetic studies. Research efforts then led to the development of experimental models testing the impact of perinatal hypoxia or maternal immune activation on neurodevelopmental disorders. These perinatal factors are usually studied separately, but given that the models are now validated, it is feasible to investigate both factors together. Inclusion of additional factors, such as metabolic disturbances or chronic stress, may need to be considered also. Understanding the interplay of perinatal factors in schizophrenia's etiology is crucial for developing targeted prevention and therapeutic strategies.
Collapse
Affiliation(s)
- I Kawikova
- Department of Medicine, Yale University, New Haven, CT, USA,
| | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Łuszczki E, Wyszyńska J, Dymek A, Drożdż D, González-Ramos L, Hartgring I, García-Carbonell N, Mazur A, Erdine S, Parnarauskienė J, Alvarez-Pitti J. The Effect of Maternal Diet and Lifestyle on the Risk of Childhood Obesity. Metabolites 2024; 14:655. [PMID: 39728436 DOI: 10.3390/metabo14120655] [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: 10/31/2024] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 12/28/2024] Open
Abstract
Background/Objectives: Childhood obesity is a global health problem that affects at least 41 million children under the age of five. Increased BMI in children is associated with serious long-term health consequences, such as type 2 diabetes, cardiovascular disease, and psychological problems, including depression and low self-esteem. Although the etiology of obesity is complex, research suggests that the diet and lifestyle of pregnant women play a key role in shaping metabolic and epigenetic changes that can increase the risk of obesity in their children. Excessive gestational weight gain, unhealthy dietary patterns (including the Western diet), and pregnancy complications (such as gestational diabetes) are some of the modifiable factors that contribute to childhood obesity. The purpose of this narrative review is to summarize the most important and recent information on the impact of the diet and lifestyle of pregnant women on the risk of childhood obesity. Methods: This article is a narrative review that aims to summarize the available literature on the impact of pregnant women's diet and lifestyle on the risk of obesity in their offspring, with a focus on metabolic and epigenetic mechanisms. Results/Conclusions: Current evidence suggests that a pregnant woman's lifestyle and diet can significantly contribute to lowering the risk of obesity in their offspring. However, further high-quality research is needed to understand better the metabolic and epigenetic relationships concerning maternal factors that predispose offspring to obesity.
Collapse
Affiliation(s)
- Edyta Łuszczki
- Institute of Health Sciences, Medical College of Rzeszów University, 35-959 Rzeszów, Poland
| | - Justyna Wyszyńska
- Institute of Health Sciences, Medical College of Rzeszów University, 35-959 Rzeszów, Poland
| | - Agnieszka Dymek
- Institute of Health Sciences, Medical College of Rzeszów University, 35-959 Rzeszów, Poland
| | - Dorota Drożdż
- Department of Pediatric Nephrology and Hypertension, Pediatric Institute, Jagiellonian University Medical College, 31-007 Krakow, Poland
| | - Laura González-Ramos
- Innovation in Paediatrics and Technologies-iPEDITEC- Research Group, Fundación de Investigación, Consorcio Hospital General, University of Valencia, 46010 Valencia, Spain
| | - Isa Hartgring
- Innovation in Paediatrics and Technologies-iPEDITEC- Research Group, Fundación de Investigación, Consorcio Hospital General, University of Valencia, 46010 Valencia, Spain
| | - Nuria García-Carbonell
- Innovation in Paediatrics and Technologies-iPEDITEC- Research Group, Fundación de Investigación, Consorcio Hospital General, University of Valencia, 46010 Valencia, Spain
- Pediatric Department, Consorcio Hospital General, University of Valencia, 46014 Valencia, Spain
| | - Artur Mazur
- Institute of Medical Sciences, Medical College of Rzeszów University, 35-959 Rzeszów, Poland
| | - Serap Erdine
- Cerrahpasa Faculty of Medicine, Department of Cardiology, Istanbul University-Cerrahpasa, 34320 Istanbul, Turkey
| | - Justė Parnarauskienė
- Pediatric Department, Vilnius University Hospital Santaros Klinikos, 08661 Vilnius, Lithuania
| | - Julio Alvarez-Pitti
- Innovation in Paediatrics and Technologies-iPEDITEC- Research Group, Fundación de Investigación, Consorcio Hospital General, University of Valencia, 46010 Valencia, Spain
- Pediatric Department, Consorcio Hospital General, University of Valencia, 46014 Valencia, Spain
- CIBER Fisiopatología Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
9
|
Xiao L, Zhao F. Exploring the frontier of microbiome biomarker discovery with artificial intelligence. Natl Sci Rev 2024; 11:nwae325. [PMID: 39606148 PMCID: PMC11601805 DOI: 10.1093/nsr/nwae325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/23/2024] [Accepted: 09/12/2024] [Indexed: 11/29/2024] Open
Affiliation(s)
- Liwen Xiao
- Institute of Zoology, Chinese Academy of Sciences, China
| | - Fangqing Zhao
- Institute of Zoology, Chinese Academy of Sciences, China
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, China
| |
Collapse
|
10
|
Gilley SP, Ruebel ML, Chintapalli SV, Wright CJ, Rozance PJ, Shankar K. Calorie restriction during gestation impacts maternal and offspring fecal microbiome in mice. Front Endocrinol (Lausanne) 2024; 15:1423464. [PMID: 39429739 PMCID: PMC11487197 DOI: 10.3389/fendo.2024.1423464] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 08/28/2024] [Indexed: 10/22/2024] Open
Abstract
Background Maternal undernutrition is the most common cause of fetal growth restriction (FGR) worldwide. FGR increases morbidity and mortality during infancy, as well as contributes to adult-onset diseases including obesity and type 2 diabetes. The role of the maternal or offspring microbiome in growth outcomes following FGR is not well understood. Methods FGR was induced by 30% maternal calorie restriction (CR) during the second half of gestation in C57BL/6 mice. Pup weights were obtained on day of life 0, 1, and 7 and ages 3, 4 and 16 weeks. Fecal pellets were collected from pregnant dams at gestational day 18.5 and from offspring at ages 3 and 4 weeks of age. Bacterial genomic DNA was used for amplification of the V4 variable region of the 16S rRNA gene. Multivariable associations between maternal CR and taxonomic abundance were assessed using the MaAsLin2 package. Associations between microbial taxa and offspring outcomes were performed using distance-based redundancy analysis and Pearson correlations. Results FGR pups weighed about 20% less than controls. Beta but not alpha diversity differed between control and CR dam microbiomes. CR dams had lower relative abundance of Turicibacter, Flexispira, and Rikenella, and increased relative abundance of Parabacteroides and Prevotella. Control and FGR offspring microbiota differed by beta diversity at ages 3 and 4 weeks. At 3 weeks, FGR offspring had decreased relative abundance of Akkermansia and Sutterella and increased relative abundance of Anaerostipes and Paraprevotella. At 4 weeks, FGR animals had decreased relative abundance of Allobaculum, Sutterella, Bifidobacterium, and Lactobacillus, among others, and increased relative abundance of Turcibacter, Dorea, and Roseburia. Maternal Helicobacter abundance was positively associated with offspring weight. Akkermansia abundance at age 3 and 4 weeks was negatively associated with adult weight. Conclusions We demonstrate gut microbial dysbiosis in pregnant dams and offspring at two timepoints following maternal calorie restriction. Additional research is needed to test for functional roles of the microbiome in offspring growth outcomes.
Collapse
Affiliation(s)
- Stephanie P. Gilley
- Department of Pediatrics, Section of Nutrition, University of Colorado School of Medicine, Aurora, CO, United States
| | - Meghan L. Ruebel
- Microbiome and Metabolism Research Unit (MMRU), United States Department of Agriculture - Agricultural Research Service (USDA-ARS), Southeast Area, Little Rock, AR, United States
- Arkansas Children’s Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Sree V. Chintapalli
- Arkansas Children’s Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR, United States
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Clyde J. Wright
- Department of Pediatrics, Section of Neonatology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Paul J. Rozance
- Department of Pediatrics, Section of Neonatology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Kartik Shankar
- Department of Pediatrics, Section of Nutrition, University of Colorado School of Medicine, Aurora, CO, United States
| |
Collapse
|
11
|
Elwyn R, Williams M, Smith E, Smith S. Two identical twin pairs discordant for longstanding anorexia nervosa and OSFED: lived experience accounts of eating disorder and recovery processes. J Eat Disord 2024; 12:127. [PMID: 39223672 PMCID: PMC11367789 DOI: 10.1186/s40337-024-01078-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 08/02/2024] [Indexed: 09/04/2024] Open
Abstract
Research into the risk of anorexia nervosa (AN) has examined twin pairs to further the understanding of the contributions of genetics, trait inheritance, and environmental factors to eating disorder (ED) development. Investigations of twin experiences of EDs have been biologically-based and have not considered the qualitative, phenomenological aspects of twin experiences. A gap in the literature exists regarding understanding of discordant twins with EDs. This research was developed in response, with the aim to deepen understanding of AN in discordant twins and to create novel ideas for further research and testing. The case studies presented in this article provide lived experience insights of two identical discordant twin pairs: one twin pair discordant for longstanding AN and one twin pair discordant for 'atypical' AN (the twin with AN has recovered). The perspectives and experiences of each co-twin (one with AN and one without) explore a number of factors that may have contributed to twin discordance in these cases, and how each twin has responded to the impact of AN in their lives. Through use of first-person accounts in case study presentation, this article centres social justice values of lived experience leadership and involvement in research. This article aims to extend current knowledge and understanding of EDs in discordant twins, particularly regarding risk for ED development, ED duration, diagnosis and treatment, and recovery processes.
Collapse
Affiliation(s)
- Rosiel Elwyn
- Neuroscience and Psychiatry, Thompson Institute, University of the Sunshine Coast, Birtinya, QLD, Australia.
| | | | | | | |
Collapse
|
12
|
Qin Z, He X, Gao Q, Li Y, Zhang Y, Wang H, Qin N, Wang C, Huang B, Shi Y, Liu C, Wang S, Zhang H, Li Y, Shi H, Tian X, Song L. Postweaning sodium citrate exposure induces long-lasting and sex-dependent effects on social behaviours in mice. Pharmacol Biochem Behav 2024; 242:173807. [PMID: 38925482 DOI: 10.1016/j.pbb.2024.173807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND Postweaning is a pivotal period for brain development and individual growth. As an important chemical used in medicines, foods and beverages, sodium citrate (SC) is commonly available. Although some effects of SC exposure on individual physiology have been demonstrated, the potential long-lasting effects of postweaning dietary SC exposure on social behaviours are still elusive. METHODS Both postweaning male and female C57BL/6 mice were exposed to SC through drinking water for a total of 3 weeks. A series of behavioural tests, including social dominance test (SDT), social interaction test (SIT), bedding preference test (BPT) and sexual preference test (SPT), were performed in adolescence and adulthood. After these tests, serum oxytocin (OT) levels and gut microbiota were detected. RESULTS The behavioural results revealed that postweaning SC exposure decreased the social dominance of male mice in adulthood and female mice in both adolescence and adulthood. SC exposure also reduced the sexual preference rates of both males and females, while it had no effect on social interaction behaviour. ELISA results indicated that SC exposure decreased the serum OT levels of females but not males. 16S rRNA sequencing analysis revealed a significant difference in β-diversity after SC exposure in both males and females. The correlation coefficient indicated the correlation between social behaviours, OT levels and dominant genera of gut microbiota. CONCLUSION Our findings suggest that postweaning SC exposure may have enduring and sex-dependent effects on social behaviours, which may be correlated with altered serum OT levels and gut microbiota composition.
Collapse
Affiliation(s)
- Zihan Qin
- Neuroscience Research Center, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang 050017, China
| | - Xinyue He
- Neuroscience Research Center, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang 050017, China
| | - Qiang Gao
- Neuroscience Research Center, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Neurophysiology (SZX2020013), Hebei Medical University, 050017, China
| | - Yuxin Li
- Neuroscience Research Center, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang 050017, China
| | - Yue Zhang
- Neuroscience Research Center, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang 050017, China
| | - Huajian Wang
- Neuroscience Research Center, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang 050017, China
| | - Na Qin
- Neuroscience Research Center, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang 050017, China
| | - Chen Wang
- Neuroscience Research Center, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang 050017, China
| | - Boya Huang
- Neuroscience Research Center, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Neurophysiology (SZX2020013), Hebei Medical University, 050017, China
| | - Yun Shi
- Neuroscience Research Center, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Neurophysiology (SZX2020013), Hebei Medical University, 050017, China
| | - Congcong Liu
- Neuroscience Research Center, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang 050017, China; Early Life Health Research Center, Hebei Medical University, Shijiazhuang 050017, China
| | - Sheng Wang
- Neuroscience Research Center, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Neurophysiology (SZX2020013), Hebei Medical University, 050017, China
| | - Huifeng Zhang
- Neuroscience Research Center, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang 050017, China; Department of Pediatrics, The Second Hospital of Hebei Medical University, Shijiazhuang 050017, China; Early Life Health Research Center, Hebei Medical University, Shijiazhuang 050017, China
| | - Youdong Li
- Neuroscience Research Center, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang 050017, China; Early Life Health Research Center, Hebei Medical University, Shijiazhuang 050017, China
| | - Haishui Shi
- Neuroscience Research Center, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Neurophysiology (SZX2020013), Hebei Medical University, 050017, China; Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang 050017, China; Early Life Health Research Center, Hebei Medical University, Shijiazhuang 050017, China
| | - Xiaoyu Tian
- Neuroscience Research Center, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang 050017, China; Department of Pediatrics, The Second Hospital of Hebei Medical University, Shijiazhuang 050017, China.
| | - Li Song
- Neuroscience Research Center, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Neurophysiology (SZX2020013), Hebei Medical University, 050017, China.
| |
Collapse
|
13
|
Xiao L, Zhou T, Zuo Z, Sun N, Zhao F. Spatiotemporal patterns of the pregnancy microbiome and links to reproductive disorders. Sci Bull (Beijing) 2024; 69:1275-1285. [PMID: 38388298 DOI: 10.1016/j.scib.2024.02.001] [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: 09/25/2023] [Revised: 01/04/2024] [Accepted: 01/27/2024] [Indexed: 02/24/2024]
Abstract
The microbiome of females undergoes extensive remodeling during pregnancy, which is likely to have an impact on the health of both mothers and offspring. Nevertheless, large-scale integrated investigations characterizing microbiome dynamics across key body habitats are lacking. Here, we performed an extensive meta-analysis that compiles and analyzes microbiome profiles from >10,000 samples across the gut, vagina, and oral cavity of pregnant women from diverse geographical regions. We have unveiled unexpected variations in the taxonomic, functional, and ecological characteristics of microbial communities throughout the course of pregnancy. The gut microbiota showed distinct trajectories between Western and non-Western populations. The vagina microbiota exhibited fluctuating transitions at the genus level across gestation, while the oral microbiota remained relatively stable. We also identified distinctive microbial signatures associated with prevalent pregnancy-related disorders, including opposite variations in the oral and gut microbiota of patients with gestational diabetes and disrupted microbial networks in preterm birth. This study establishes a comprehensive atlas of the pregnancy microbiome by integrating multidimensional datasets and offers foundational insights into the intricate interplay between microbes and host factors that underlie reproductive health.
Collapse
Affiliation(s)
- Liwen Xiao
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China
| | - Tian Zhou
- Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhenqiang Zuo
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China
| | - Ningxia Sun
- Department of Reproductive Medicine, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China.
| | - Fangqing Zhao
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China; Department of Reproductive Medicine, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China; Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
14
|
Xiao L, Zuo Z, Zhao F. Microbiome in Female Reproductive Health: Implications for Fertility and Assisted Reproductive Technologies. GENOMICS, PROTEOMICS & BIOINFORMATICS 2024; 22:qzad005. [PMID: 38862423 PMCID: PMC11104452 DOI: 10.1093/gpbjnl/qzad005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 10/16/2023] [Accepted: 10/19/2023] [Indexed: 06/13/2024]
Abstract
The microbiome plays a critical role in the process of conception and the outcomes of pregnancy. Disruptions in microbiome homeostasis in women of reproductive age can lead to various pregnancy complications, which significantly impact maternal and fetal health. Recent studies have associated the microbiome in the female reproductive tract (FRT) with assisted reproductive technology (ART) outcomes, and restoring microbiome balance has been shown to improve fertility in infertile couples. This review provides an overview of the role of the microbiome in female reproductive health, including its implications for pregnancy outcomes and ARTs. Additionally, recent advances in the use of microbial biomarkers as indicators of pregnancy disorders are summarized. A comprehensive understanding of the characteristics of the microbiome before and during pregnancy and its impact on reproductive health will greatly promote maternal and fetal health. Such knowledge can also contribute to the development of ARTs and microbiome-based interventions.
Collapse
Affiliation(s)
- Liwen Xiao
- CAS Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- Beijing Institutes of Life Science/Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhenqiang Zuo
- Beijing Institutes of Life Science/Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Fangqing Zhao
- CAS Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- Beijing Institutes of Life Science/Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
15
|
Zheng X, Zheng Y, Chen T, Hou C, Zhou L, Liu C, Zheng J, Hu R. Effect of Laryngopharyngeal Reflux and Potassium-Competitive Acid Blocker (P-CAB) on the Microbiological Comprise of the Laryngopharynx. Otolaryngol Head Neck Surg 2024; 170:1380-1390. [PMID: 38385787 DOI: 10.1002/ohn.682] [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: 08/30/2023] [Revised: 12/27/2023] [Accepted: 01/21/2024] [Indexed: 02/23/2024]
Abstract
OBJECTIVE To probe the microbiota composition progressing from healthy individuals to those with laryngopharyngeal reflux disease (LPRD) and subsequently undergoing potassium-competitive acid inhibitor (P-CAB) therapy. STUDY DESIGN Prospective case-control study. SETTING Academic Medical Center. METHODS Forty patients with LPRD and 51 patients without LPRD were recruited. An 8-week P-CAB therapy was initiated (post-T-LPRD), and 39 had return visits. In total, 130 laryngopharyngeal saliva samples were collected and sequenced by targeting the V3-V4 region of the 16S ribosomal RNA (rRNA) gene using an Illumina MiSeq. Amplicon sequence variants (ASVs) and clinical indices were analyzed. RESULTS Alpha and beta diversities were compared among the non-LPRD, LPRD, and post-T-LPRD groups, and the Observed_ASVs were not significantly different. At the same time, the Shannon and Simpson indices, unweighted Unifrac, weighted Unifrac, and binary Jaccard distance were significantly different between non-LPRD and LPRD groups. In addition, significant differences were found in the abundance of Streptococcus, Prevotella, and Prevotellaceae in the LPRD versus non-LPRD groups, and Neisseria, Leptotrichia, and Allprevotella in the LPRD versus post-T-LPRD groups. The genera model was used to distinguish patients with LPRD from those without, and a better receiver operating characteristic curve was formed after combining the clinical indices of reflux symptom index, reflux finding score, and pepsin, with an area under the curve of 0.960. CONCLUSION Laryngopharyngeal microbial communities changed after laryngopharyngeal reflux and were modified further after P-CAB treatment, which provides a potential diagnostic value for LPRD, especially when combined with clinical indices.
Collapse
Affiliation(s)
- Xiaowei Zheng
- Department of Otorhinolaryngology-Head and Neck Surgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Yujin Zheng
- Department of Otorhinolaryngology-Head and Neck Surgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Ting Chen
- Department of Otorhinolaryngology-Head and Neck Surgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Chenjie Hou
- Department of Otorhinolaryngology-Head and Neck Surgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Liqun Zhou
- Department of Otorhinolaryngology-Head and Neck Surgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Chaofeng Liu
- Department of Otorhinolaryngology-Head and Neck Surgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Jingyi Zheng
- Department of Otorhinolaryngology-Head and Neck Surgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Renyou Hu
- Chongqing Jinshan Science & Technology (Group) Co. Ltd., Chongqing, China
| |
Collapse
|
16
|
Tang H, Li D, Peng J, Yang W, Zhang X, Li H. Potential Association of Gut Microbial Metabolism and Circulating mRNA Based on Multiomics Sequencing Analysis in Fetal Growth Restriction. Mediators Inflamm 2024; 2024:9986187. [PMID: 38716374 PMCID: PMC11074908 DOI: 10.1155/2024/9986187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/27/2024] [Accepted: 03/25/2024] [Indexed: 08/21/2024] Open
Abstract
Objective Fetal growth restriction (FGR) is a significant contributor to negative pregnancy and postnatal developmental outcomes. Currently, the exact pathological mechanism of FGR remains unknown. This study aims to utilize multiomics sequencing technology to investigate potential relationships among mRNA, gut microbiota, and metabolism in order to establish a theoretical foundation for diagnosing and understanding the molecular mechanisms underlying FGR. Methods In this study, 11 healthy pregnant women and nine pregnant women with FGR were divided into Control group and FGR group based on the health status. Umbilical cord blood, maternal serum, feces, and placental tissue samples were collected during delivery. RNA sequencing, 16S rRNA sequencing, and metabolomics methods were applied to analyze changes in umbilical cord blood circulating mRNA, fecal microbiota, and metabolites. RT-qPCR, ELISA, or western blot were used to detect the expression of top 5 differential circulating mRNA in neonatal cord blood, maternal serum, or placental tissue samples. Correlation between differential circulating mRNA, microbiota, and metabolites was analyzed by the Spearman coefficient. Results The top 5 mRNA genes in FGR were altered with the downregulation of TRIM34, DEFA3, DEFA1B, DEFA1, and QPC, and the upregulation of CHPT1, SMOX, FAM83A, GDF15, and NAPG in newborn umbilical cord blood, maternal serum, and placental tissue. The abundance of Bacteroides, Akkermansia, Eubacterium_coprostanoligenes_group, Phascolarctobacterium, Parasutterella, Odoribacter, Lachnospiraceae_UCG_010, and Dielma were significantly enriched in the FGR group. Metabolites such as aspartic acid, methionine, alanine, L-tryptophan, 3-methyl-2-oxovalerate, and ketoleucine showed notable functional alterations. Spearman correlation analysis indicated that metabolites like methionine and alanine, microbiota (Tyzzerella), and circulating mRNA (TRIM34, SMOX, FAM83A, NAPG) might play a role as mediators in the communication between the gut and circulatory system interaction in FGR. Conclusion Metabolites (METHIONINE, alanine) as well as microbiota (Tyzzerella) and circulating mRNA (TRIM34, SMOX, FAM83A, NAPG) were possible mediators that communicated the interaction between the gut and circulatory systems in FGR.
Collapse
Affiliation(s)
- Hui Tang
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defects Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, China
| | - Dan Li
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defects Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, China
| | - Jing Peng
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defects Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, China
| | - Weitao Yang
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defects Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, China
| | - Xian Zhang
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defects Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, China
| | - Hanmei Li
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defects Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, China
| |
Collapse
|
17
|
Beckers KF, Flanagan JP, Sones JL. Microbiome and pregnancy: focus on microbial dysbiosis coupled with maternal obesity. Int J Obes (Lond) 2024; 48:439-448. [PMID: 38145995 PMCID: PMC10978494 DOI: 10.1038/s41366-023-01438-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 11/22/2023] [Accepted: 12/01/2023] [Indexed: 12/27/2023]
Abstract
Obesity is becoming a worldwide pandemic with over one billion people affected. Of women in the United States, who are of childbearing age, two-thirds of them are considered overweight/obese. Offspring of women with obesity have a greater likelihood of developing cardiometabolic disease later in life, therefore making obesity a transgenerational issue. Emerging topics such as maternal microbial dysbiosis with altered levels of bacterial phyla and maternal obesity programming offspring cardiometabolic disease are a novel area of research discussed in this review. In the authors' opinion, beneficial therapeutics will be developed from knowledge of bacterial-host interactions at the most specific level possible. Although there is an abundance of obesity-related microbiome research, it is not concise, readily available, nor easy to interpret at this time. This review details the current knowledge regarding the relationship between obesity and the gut microbiome, with an emphasis on maternal obesity.
Collapse
Affiliation(s)
- Kalie F Beckers
- Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - Juliet P Flanagan
- Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - Jenny L Sones
- Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA.
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA.
- Clinical Sciences, Colorado State University College of Veterinary Medicine and Biomedical Sciences, Fort Collins, CO, USA.
| |
Collapse
|
18
|
Tian Z, Zhang X, Yao G, Jin J, Zhang T, Sun C, Wang Z, Zhang Q. Intestinal flora and pregnancy complications: Current insights and future prospects. IMETA 2024; 3:e167. [PMID: 38882493 PMCID: PMC11170975 DOI: 10.1002/imt2.167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/27/2023] [Accepted: 12/22/2023] [Indexed: 06/18/2024]
Abstract
Numerous studies have demonstrated the pivotal roles of intestinal microbiota in many physiopathological processes through complex interactions with the host. As a unique period in a woman's lifespan, pregnancy is characterized by changes in hormones, immunity, and metabolism. The gut microbiota also changes during this period and plays a crucial role in maintaining a healthy pregnancy. Consequently, anomalies in the composition and function of the gut microbiota, namely, gut microbiota dysbiosis, can predispose individuals to various pregnancy complications, posing substantial risks to both maternal and neonatal health. However, there are still many controversies in this field, such as "sterile womb" versus "in utero colonization." Therefore, a thorough understanding of the roles and mechanisms of gut microbiota in pregnancy and its complications is essential to safeguard the health of both mother and child. This review provides a comprehensive overview of the changes in gut microbiota during pregnancy, its abnormalities in common pregnancy complications, and potential etiological implications. It also explores the potential of gut microbiota in diagnosing and treating pregnancy complications and examines the possibility of gut-derived bacteria residing in the uterus/placenta. Our aim is to expand knowledge in maternal and infant health from the gut microbiota perspective, aiding in developing new preventive and therapeutic strategies for pregnancy complications based on intestinal microecology.
Collapse
Affiliation(s)
- Zhenyu Tian
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology Qilu Hospital of Shandong University Jinan China
| | - Xinjie Zhang
- Department of Biology University College London London UK
| | - Guixiang Yao
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology Qilu Hospital of Shandong University Jinan China
| | - Jiajia Jin
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology Qilu Hospital of Shandong University Jinan China
| | - Tongxue Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology Qilu Hospital of Shandong University Jinan China
| | - Chunhua Sun
- Department of Health Management Center, Qilu Hospital, Cheeloo College of Medicine Shandong University Jinan China
| | - Zhe Wang
- Department of Geriatrics Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan China
| | - Qunye Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology Qilu Hospital of Shandong University Jinan China
- Cardiovascular Disease Research Center of Shandong First Medical University Central Hospital Affiliated to Shandong First Medical University Jinan China
| |
Collapse
|
19
|
Ni Lochlainn M, Bowyer RCE, Moll JM, García MP, Wadge S, Baleanu AF, Nessa A, Sheedy A, Akdag G, Hart D, Raffaele G, Seed PT, Murphy C, Harridge SDR, Welch AA, Greig C, Whelan K, Steves CJ. Effect of gut microbiome modulation on muscle function and cognition: the PROMOTe randomised controlled trial. Nat Commun 2024; 15:1859. [PMID: 38424099 PMCID: PMC10904794 DOI: 10.1038/s41467-024-46116-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: 08/04/2023] [Accepted: 02/12/2024] [Indexed: 03/02/2024] Open
Abstract
Studies suggest that inducing gut microbiota changes may alter both muscle physiology and cognitive behaviour. Gut microbiota may play a role in both anabolic resistance of older muscle, and cognition. In this placebo controlled double blinded randomised controlled trial of 36 twin pairs (72 individuals), aged ≥60, each twin pair are block randomised to receive either placebo or prebiotic daily for 12 weeks. Resistance exercise and branched chain amino acid (BCAA) supplementation is prescribed to all participants. Outcomes are physical function and cognition. The trial is carried out remotely using video visits, online questionnaires and cognitive testing, and posting of equipment and biological samples. The prebiotic supplement is well tolerated and results in a changed gut microbiome [e.g., increased relative Bifidobacterium abundance]. There is no significant difference between prebiotic and placebo for the primary outcome of chair rise time (β = 0.579; 95% CI -1.080-2.239 p = 0.494). The prebiotic improves cognition (factor score versus placebo (β = -0.482; 95% CI,-0.813, -0.141; p = 0.014)). Our results demonstrate that cheap and readily available gut microbiome interventions may improve cognition in our ageing population. We illustrate the feasibility of remotely delivered trials for older people, which could reduce under-representation of older people in clinical trials. ClinicalTrials.gov registration: NCT04309292.
Collapse
Affiliation(s)
- Mary Ni Lochlainn
- King's College London, Department of Twin Research and Genetic Epidemiology, London, SE1 7EH, UK.
| | - Ruth C E Bowyer
- King's College London, Department of Twin Research and Genetic Epidemiology, London, SE1 7EH, UK
- The Alan Turing Institute, London, NW1 2DB, UK
| | | | - María Paz García
- King's College London, Department of Twin Research and Genetic Epidemiology, London, SE1 7EH, UK
| | - Samuel Wadge
- King's College London, Department of Twin Research and Genetic Epidemiology, London, SE1 7EH, UK
| | - Andrei-Florin Baleanu
- King's College London, Department of Twin Research and Genetic Epidemiology, London, SE1 7EH, UK
| | - Ayrun Nessa
- King's College London, Department of Twin Research and Genetic Epidemiology, London, SE1 7EH, UK
| | - Alyce Sheedy
- King's College London, Department of Twin Research and Genetic Epidemiology, London, SE1 7EH, UK
| | - Gulsah Akdag
- King's College London, Department of Twin Research and Genetic Epidemiology, London, SE1 7EH, UK
| | - Deborah Hart
- King's College London, Department of Twin Research and Genetic Epidemiology, London, SE1 7EH, UK
| | - Giulia Raffaele
- GKT School of Medical Education, King's College London, London, UK
| | - Paul T Seed
- Unit for Medical Statistics/Department for Women and Children's Health, School of Life Course and Population Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Caroline Murphy
- King's Clinical Trials Unit, Research Management and Innovation Directorate, King's College London, London, UK
| | - Stephen D R Harridge
- Centre for Human & Applied Physiological Sciences, King's College London, London, UK
| | - Ailsa A Welch
- Department of Epidemiology and Public Health, Norwich Medical School, University of East Anglia, Norwich, NR4 7TJ, UK
| | - Carolyn Greig
- School of Sport, Exercise, and Rehabilitation Sciences, University of Birmingham, Birmingham, UK
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham, UK
| | - Kevin Whelan
- King's College London, Department of Nutritional Sciences, Franklin Wilkins Building, SE1 9NH, London, UK
| | - Claire J Steves
- King's College London, Department of Twin Research and Genetic Epidemiology, London, SE1 7EH, UK.
| |
Collapse
|
20
|
Zhang H, Zha X, Zhang B, Zheng Y, Elsabagh M, Wang H, Wang M. Gut microbiota contributes to bisphenol A-induced maternal intestinal and placental apoptosis, oxidative stress, and fetal growth restriction in pregnant ewe model by regulating gut-placental axis. MICROBIOME 2024; 12:28. [PMID: 38365714 PMCID: PMC10874076 DOI: 10.1186/s40168-024-01749-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 01/02/2024] [Indexed: 02/18/2024]
Abstract
BACKGROUND Bisphenol A (BPA) is an environmental contaminant with endocrine-disrupting properties that induce fetal growth restriction (FGR). Previous studies on pregnant ewes revealed that BPA exposure causes placental apoptosis and oxidative stress (OS) and decreases placental efficiency, consequently leading to FGR. Nonetheless, the response of gut microbiota to BPA exposure and its role in aggravating BPA-mediated apoptosis, autophagy, mitochondrial dysfunction, endoplasmic reticulum stress (ERS), and OS of the maternal placenta and intestine are unclear in an ovine model of gestation. RESULTS Two pregnant ewe groups (n = 8/group) were given either a subcutaneous (sc) injection of corn oil (CON group) or BPA (5 mg/kg/day) dissolved in corn oil (BPA group) once daily, from day 40 to day 110 of gestation. The maternal colonic digesta and the ileum and placental tissue samples were collected to measure the biomarkers of autophagy, apoptosis, mitochondrial dysfunction, ERS, and OS. To investigate the link between gut microbiota and the BPA-induced FGR in pregnant ewes, gut microbiota transplantation (GMT) was conducted in two pregnant mice groups (n = 10/group) from day 0 to day 18 of gestation after removing their intestinal microbiota by antibiotics. The results indicated that BPA aggravates apoptosis, ERS and autophagy, mitochondrial function injury of the placenta and ileum, and gut microbiota dysbiosis in pregnant ewes. GMT indicated that BPA-induced ERS, autophagy, and apoptosis in the ileum and placenta are attributed to gut microbiota dysbiosis resulting from BPA exposure. CONCLUSIONS Our findings indicate the underlying role of gut microbiota dysbiosis and gut-placental axis behind the BPA-mediated maternal intestinal and placental apoptosis, OS, and FGR. The findings further provide novel insights into modulating the balance of gut microbiota through medication or probiotics, functioning via the gut-placental axis, to alleviate gut-derived placental impairment or FGR. Video Abstract.
Collapse
Affiliation(s)
- Hao Zhang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, P. R. China.
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, 225009, P. R. China.
| | - Xia Zha
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, P. R. China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, 225009, P. R. China
| | - Bei Zhang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, P. R. China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, 225009, P. R. China
| | - Yi Zheng
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, P. R. China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, 225009, P. R. China
| | - Mabrouk Elsabagh
- Department of Animal Production and Technology, Faculty of Agricultural Sciences and Technologies, Niğde Ömer Halisdemir University, Nigde, 51240, Turkey
- Department of Nutrition and Clinical Nutrition, Faculty of Veterinary Medicine, Kafrelsheikh University, KafrelSheikh, Egypt
| | - Hongrong Wang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, P. R. China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, 225009, P. R. China
| | - Mengzhi Wang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, P. R. China.
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, 225009, P. R. China.
- State Key Laboratory of Sheep Genetic Improvement and Healthy Production, Xinjiang Academy of Agricultural Reclamation Science, Shihezi, 832000, P. R. China.
| |
Collapse
|
21
|
Gilley SP, Zarate MA, Zheng L, Jambal P, Yazza DN, Chintapalli SV, MacLean PS, Wright CJ, Rozance PJ, Shankar K. Metabolic and fecal microbial changes in adult fetal growth restricted mice. Pediatr Res 2024; 95:647-659. [PMID: 37935884 PMCID: PMC10899111 DOI: 10.1038/s41390-023-02869-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 09/13/2023] [Accepted: 10/12/2023] [Indexed: 11/09/2023]
Abstract
BACKGROUND Fetal growth restriction (FGR) increases risk for development of obesity and type 2 diabetes. Using a mouse model of FGR, we tested whether metabolic outcomes were exacerbated by high-fat diet challenge or associated with fecal microbial taxa. METHODS FGR was induced by maternal calorie restriction from gestation day 9 to 19. Control and FGR offspring were weaned to control (CON) or 45% fat diet (HFD). At age 16 weeks, offspring underwent intraperitoneal glucose tolerance testing, quantitative MRI body composition assessment, and energy balance studies. Total microbial DNA was used for amplification of the V4 variable region of the 16 S rRNA gene. Multivariable associations between groups and genera abundance were assessed using MaAsLin2. RESULTS Adult male FGR mice fed HFD gained weight faster and had impaired glucose tolerance compared to control HFD males, without differences among females. Irrespective of weaning diet, adult FGR males had depletion of Akkermansia, a mucin-residing genus known to be associated with weight gain and glucose handling. FGR females had diminished Bifidobacterium. Metabolic changes in FGR offspring were associated with persistent gut microbial changes. CONCLUSION FGR results in persistent gut microbial dysbiosis that may be a therapeutic target to improve metabolic outcomes. IMPACT Fetal growth restriction increases risk for metabolic syndrome later in life, especially if followed by rapid postnatal weight gain. We report that a high fat diet impacts weight and glucose handling in a mouse model of fetal growth restriction in a sexually dimorphic manner. Adult growth-restricted offspring had persistent changes in fecal microbial taxa known to be associated with weight, glucose homeostasis, and bile acid metabolism, particularly Akkermansia, Bilophilia and Bifidobacteria. The gut microbiome may represent a therapeutic target to improve long-term metabolic outcomes related to fetal growth restriction.
Collapse
Affiliation(s)
- Stephanie P Gilley
- Department of Pediatrics, Section of Nutrition, University of Colorado School of Medicine, Aurora, CO, USA.
| | - Miguel A Zarate
- Department of Pediatrics, Section of Neonatology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Lijun Zheng
- Department of Pediatrics, Section of Neonatology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Purevsuren Jambal
- Department of Pediatrics, Section of Nutrition, University of Colorado School of Medicine, Aurora, CO, USA
| | - Deaunabah N Yazza
- Department of Pediatrics, Section of Nutrition, University of Colorado School of Medicine, Aurora, CO, USA
| | - Sree V Chintapalli
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Paul S MacLean
- Department of Medicine, Division of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Clyde J Wright
- Department of Pediatrics, Section of Neonatology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Paul J Rozance
- Department of Pediatrics, Section of Neonatology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Kartik Shankar
- Department of Pediatrics, Section of Nutrition, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
22
|
Zhou T, Xiao L, Zuo Z, Zhao F. MAMI: a comprehensive database of mother-infant microbiome and probiotic resources. Nucleic Acids Res 2024; 52:D738-D746. [PMID: 37819042 PMCID: PMC10767955 DOI: 10.1093/nar/gkad813] [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: 08/03/2023] [Revised: 09/04/2023] [Accepted: 09/19/2023] [Indexed: 10/13/2023] Open
Abstract
Extensive evidence has demonstrated that the human microbiome and probiotics confer great impacts on human health, particularly during critical developmental stages such as pregnancy and infancy when microbial communities undergo remarkable changes and maturation. However, a major challenge in understanding the microbial community structure and interactions between mothers and infants lies in the current lack of comprehensive microbiome databases specifically focused on maternal and infant health. To address this gap, we have developed an extensive database called MAMI (Microbiome Atlas of Mothers and Infants) that archives data on the maternal and neonatal microbiome, as well as abundant resources on edible probiotic strains. By leveraging this resource, we can gain profound insights into the dynamics of microbial communities, contributing to lifelong wellness for both mothers and infants through precise modulation of the developing microbiota. The functionalities incorporated into MAMI provide a unique perspective on the study of the mother-infant microbiome, which not only advance microbiome-based scientific research but also enhance clinical practice. MAMI is publicly available at https://bioinfo.biols.ac.cn/mami/.
Collapse
Affiliation(s)
- Tian Zhou
- Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Liwen Xiao
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhenqiang Zuo
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China
| | - Fangqing Zhao
- Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
23
|
Jiang H, Han TL, Yang J, Yang Y, Wang F, Chen Y, Huang N, Mansell T, Craig JM, Scurrah KJ, Novakovic B, Baker PN, Zhang H, Wei Y, Wang L, Saffery R. Evidence for ethnicity and location as regulators of the newborn blood metabolome: a monozygous twin study. Front Nutr 2024; 10:1259777. [PMID: 38239842 PMCID: PMC10794553 DOI: 10.3389/fnut.2023.1259777] [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: 07/16/2023] [Accepted: 12/07/2023] [Indexed: 01/22/2024] Open
Abstract
Introduction Monochorionic, diamniotic (MCDA) monozygotic twins share nearly all genetic variation and a common placenta in utero. Despite this, MCDA twins are often discordant for a range of common phenotypes, including early growth and birth weight. As such, MCDA twins represent a unique model to explore variation in early growth attributable primarily to in utero environmental factors. Methods MCDA twins with a range of within-pair birth weight discordance were sampled from the peri/postnatal epigenetic twin study (PETS, Melbourne; n = 26 pairs), Beijing twin study (BTS, Beijing; n = 25), and the Chongqing longitudinal twin study (LoTiS, Chongqing; n = 22). All PETS participants were of European-Australian ancestry, while all Chinese participants had Han ancestry. The average of the birth weight difference between the larger and smaller co-twins for all twin pairs was determined and metabolomic profiles of amino acids, TCA cycle intermediates, fatty acids, organic acids, and their derivatives generated from cord blood plasma by gas chromatograph mass spectrometry. Within and between co-twin pair analyses were performed to identify metabolites specifically associated with discordance in birth weight. Multivariable regression and pathway enrichment analyses between different regions were performed to evaluate the geographical effects on the metabolism of MCDA twin pairs. Results PETS twins showed a markedly different metabolic profile at birth compared to the two Chinese samples. Within-pair analysis revealed an association of glutathione, creatinine, and levulinic acid with birth weight discordance. Caffeine, phenylalanine, and several saturated fatty acid levels were uniquely elevated in PETS twins and were associated with maternal BMI and average within pair birth weight, in addition to birth weight discordance. LoTiS twins had higher levels of glutathione, tyrosine, and gamma-linolenic acid relative to PETS and BTS twins, potentially associated with eating habits. Conclusion This study highlights the potential role of underlying genetic variation (shared by MZ twins), in utero (non-shared by MZ twins) and location-specific (shared by MZ twins) environmental factors, in regulating the cord blood metabolome of uncomplicated MCDA twins. Future research is needed to unravel these complex relationships that may play a key role in phenotypic metabolic alterations of twins independent of genetic diversity.
Collapse
Affiliation(s)
- Huimin Jiang
- Department of Reproductive Medicine Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ting-Li Han
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jing Yang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Yang Yang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, China
- Mass Spectrometry Centre of Maternal-Fetal Medicine, Life Science Institution, Chongqing Medical University, Chongqing, China
| | - Fengdi Wang
- Department of Reproductive Medicine Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuelu Chen
- Department of Reproductive Medicine Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Nana Huang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Toby Mansell
- Murdoch Children’s Research Institute, and Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
| | - Jeffrey M. Craig
- Murdoch Children’s Research Institute, and Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), School of Medicine, Deakin University, Melbourne, VIC, Australia
| | - Katrina J. Scurrah
- Twins Research Australia and Centre for Mental Health, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Boris Novakovic
- Murdoch Children’s Research Institute, and Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
| | - Philip N. Baker
- College of Life Sciences, University of Leicester, Leicester, United Kingdom
| | - Hua Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, China
| | - Yuan Wei
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Lianlian Wang
- Department of Reproductive Medicine Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Richard Saffery
- Murdoch Children’s Research Institute, and Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
24
|
Huang N, Jiang H, Zhang Y, Sun X, Li Y, Wei Y, Yang J, Zhao Y. Amniotic fluid metabolic fingerprinting contributes to shaping the unfavourable intrauterine environment in monochorionic diamniotic twins. Clin Nutr 2024; 43:111-123. [PMID: 38035859 DOI: 10.1016/j.clnu.2023.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 10/07/2023] [Accepted: 11/02/2023] [Indexed: 12/02/2023]
Abstract
BACKGROUND & AIMS Amniotic fluid (AF) is the primary intrauterine environment for fetal growth throughout gestation. Selective fetal growth restriction (sFGR) is an adverse complication characterized by unequal growth in twins with nearly identical genetic makeup. However, the influence of AF-mediated intrauterine environment on the development and progression of sFGR remains unexplored. METHODS High-throughput targeted metabolomics analysis (G350) was performed on AF samples collected from sFGR (n = 18) and MCDA twins with birth weight concordance (MCDA-C, n = 20) cases. Weighted correlation network analysis (WGCNA) was used to identify clinical features that may influence the metabolite composition in AF. Subsequently, partial least-squares discriminant analysis (PLS-DA) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were performed to compare the different types of sFGR and MCDA-C twins. Receiver operating characteristic (ROC) and multivariate ROC curves were utilized to explore potential AF markers in twins with sFGR. RESULTS In our study, 182 metabolites were quantified in 76 AF samples. WGCNA indicated that the metabolite composition in late AF may not be influenced by gestational age. PLSDA demonstrated distinct variations between the metabolite profiles of AF in the sFGR and MCDA-C twins, with a significant emphasis on amino acids as the primary differential metabolite. The dissimilarities observed in sFGR twins were predominantly attributed to lipid metabolism-related metabolites. In particular, the KEGG enrichment metabolic pathway analysis revealed significant associations of both types of sFGR twins with central carbon metabolism in cancer. The multivariate ROC curves indicated that the combination of carnosine, sarcosine, l-alanine, beta-alanine, and alpha-n-phenylacetylglutamine significantly improved the AUC to 0.928. Notably, the ROC curves highlighted creatine (AUC:0.934) may be a potential biomarker for severe sFGR. CONCLUSION The data presented in this study offer a comprehensive metabolic map of the AF in cases of sFGR, shedding light on potential biomarkers associated with fetal growth and development in MCDA twins.
Collapse
Affiliation(s)
- Nana Huang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China; National Center for Healthcare Quality Management in Obstetrics, Beijing, China
| | - Hai Jiang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China; National Center for Healthcare Quality Management in Obstetrics, Beijing, China
| | - Youzhen Zhang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China; National Center for Healthcare Quality Management in Obstetrics, Beijing, China
| | - Xiya Sun
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China; National Center for Healthcare Quality Management in Obstetrics, Beijing, China
| | - Yixin Li
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China; National Center for Healthcare Quality Management in Obstetrics, Beijing, China
| | - Yuan Wei
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China; National Center for Healthcare Quality Management in Obstetrics, Beijing, China
| | - Jing Yang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China; National Center for Healthcare Quality Management in Obstetrics, Beijing, China.
| | - Yangyu Zhao
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China; National Center for Healthcare Quality Management in Obstetrics, Beijing, China.
| |
Collapse
|
25
|
Huang N, Chen W, Jiang H, Yang J, Zhang Y, Shi H, Wang Y, Yuan P, Qiao J, Wei Y, Zhao Y. Metabolic dynamics and prediction of sFGR and adverse fetal outcomes: a prospective longitudinal cohort study. BMC Med 2023; 21:455. [PMID: 37996847 PMCID: PMC10666385 DOI: 10.1186/s12916-023-03134-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 10/26/2023] [Indexed: 11/25/2023] Open
Abstract
BACKGROUND Selective fetal growth restriction (sFGR) is an extreme complication that significantly increases the risk of perinatal mortality and long-term adverse neurological outcomes in offspring, affecting approximately 15% of monochorionic diamniotic (MCDA) twin pregnancies. The lack of longitudinal cohort studies hinders the early prediction and intervention of sFGR. METHODS We constructed a prospective longitudinal cohort study of sFGR, and quantified 25 key metabolites in 337 samples from maternal plasma in the first, second, and third trimester and from cord plasma. In particular, our study examined fetal growth and brain injury data from ultrasonography and used the Ages and Stages Questionnaire-third edition subscale (ASQ-3) to evaluate the long-term neurocognitive behavioral development of infants aged 2-3 years. Furthermore, we correlated metabolite levels with ultrasound data, including physical development and brain injury indicators, and ASQ-3 data using Spearman's-based correlation tests. In addition, special combinations of differential metabolites were used to construct predictive models for the occurrence of sFGR and fetal brain injury. RESULTS Our findings revealed various dynamic patterns for these metabolites during pregnancy and a maximum of differential metabolites between sFGR and MCDA in the second trimester (n = 8). The combination of L-phenylalanine, L-leucine, and L-isoleucine in the second trimester, which were closely related to fetal growth indicators, was highly predictive of sFGR occurrence (area under the curve [AUC]: 0.878). The combination of L-serine, L-histidine, and L-arginine in the first trimester and creatinine in the second trimester was correlated with long-term neurocognitive behavioral development and showed the capacity to identify fetal brain injury with high accuracy (AUC: 0.94). CONCLUSIONS The performance of maternal plasma metabolites from the first and second trimester is superior to those from the third trimester and cord plasma in discerning sFGR and fetal brain injury. These metabolites may serve as useful biomarkers for early prediction and promising targets for early intervention in clinical settings.
Collapse
Affiliation(s)
- Nana Huang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, No. 49 Huayuan North Road, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China
- National Center for Healthcare Quality Management in Obstetrics, Beijing, China
| | - Wei Chen
- Department of Obstetrics and Gynecology, Peking University Third Hospital, No. 49 Huayuan North Road, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, No. 49 Huayuan North Road, Beijing, 100191, China
| | - Hai Jiang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, No. 49 Huayuan North Road, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China
- National Center for Healthcare Quality Management in Obstetrics, Beijing, China
| | - Jing Yang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, No. 49 Huayuan North Road, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China
- National Center for Healthcare Quality Management in Obstetrics, Beijing, China
| | - Youzhen Zhang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, No. 49 Huayuan North Road, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China
- National Center for Healthcare Quality Management in Obstetrics, Beijing, China
| | - Huifeng Shi
- Department of Obstetrics and Gynecology, Peking University Third Hospital, No. 49 Huayuan North Road, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China
- National Center for Healthcare Quality Management in Obstetrics, Beijing, China
| | - Ying Wang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, No. 49 Huayuan North Road, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China
- National Center for Healthcare Quality Management in Obstetrics, Beijing, China
| | - Pengbo Yuan
- Department of Obstetrics and Gynecology, Peking University Third Hospital, No. 49 Huayuan North Road, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China
- National Center for Healthcare Quality Management in Obstetrics, Beijing, China
| | - Jie Qiao
- Department of Obstetrics and Gynecology, Peking University Third Hospital, No. 49 Huayuan North Road, Beijing, 100191, China.
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China.
- National Center for Healthcare Quality Management in Obstetrics, Beijing, China.
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China.
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China.
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, No. 49 Huayuan North Road, Beijing, 100191, China.
- Beijing Advanced Innovation Center for Genomics, Beijing, China.
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China.
| | - Yuan Wei
- Department of Obstetrics and Gynecology, Peking University Third Hospital, No. 49 Huayuan North Road, Beijing, 100191, China.
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China.
- National Center for Healthcare Quality Management in Obstetrics, Beijing, China.
| | - Yangyu Zhao
- Department of Obstetrics and Gynecology, Peking University Third Hospital, No. 49 Huayuan North Road, Beijing, 100191, China.
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China.
- National Center for Healthcare Quality Management in Obstetrics, Beijing, China.
| |
Collapse
|
26
|
Alsharairi NA, Li L. Gut Microbiota, Inflammation, and Probiotic Supplementation in Fetal Growth Restriction-A Comprehensive Review of Human and Animal Studies. Life (Basel) 2023; 13:2239. [PMID: 38137841 PMCID: PMC10745050 DOI: 10.3390/life13122239] [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: 09/07/2023] [Revised: 10/30/2023] [Accepted: 11/17/2023] [Indexed: 12/24/2023] Open
Abstract
Fetal growth restriction (FGR) is a pathological state that represents a fetus's inability to achieve adequate growth during pregnancy. Several maternal, placental, and fetal factors are likely associated with FGR etiology. FGR is linked to severe fetal and neonatal complications, as well as adverse health consequences in adulthood. Numerous randomized controlled trials (RCTs) have demonstrated improved growth in FGR fetuses with promising treatment strategies such as maternal micronutrient, amino acid, and nitric oxide supplementation. Elevated inflammation in pregnant women diagnosed with FGR has been associated with an imbalance between pro- and anti-inflammatory cytokines. Gut microbiota dysbiosis may result in increased FGR-related inflammation. Probiotic treatment may relieve FGR-induced inflammation and improve fetal growth. The aim of this review is to provide an overview of the gut microbiota and inflammatory profiles associated with FGR and explore the potential of probiotics in treating FGR.
Collapse
Affiliation(s)
- Naser A. Alsharairi
- Heart, Mind and Body Research Group, Griffith University, Gold Coast, QLD 4222, Australia
| | - Li Li
- School of Science, Western Sydney University, Richmond, NSW 2753, Australia;
| |
Collapse
|
27
|
Wang N, Zhang J, Yu Z, Yan X, Zhang L, Peng H, Chen C, Li R. Oropharyngeal administration of colostrum targeting gut microbiota and metabolites in very preterm infants: protocol for a multicenter randomized controlled trial. BMC Pediatr 2023; 23:508. [PMID: 37845612 PMCID: PMC10577906 DOI: 10.1186/s12887-023-04346-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 10/03/2023] [Indexed: 10/18/2023] Open
Abstract
BACKGROUND Oropharyngeal administration of colostrum (OAC) has an immune-stimulating effect on oropharyngeal-associated lymphoid tissue, and can promote the maturation of the gastrointestinal tract. However, how OAC promotes intestinal maturation in preterm infants by altering gut microbiota remains unclear. We aim to assess changes in gut microbiota and metabolites after OAC in very preterm infants. METHODS A multicenter, double-blind, randomized controlled trial will be conducted in three large neonatal intensive care units in Shenzhen, China, with preterm infants with gestational age less than 32 weeks at birth and birth weight less than 1500 g. It is estimated that 320 preterm infants will be enrolled in this study within one year. The intervention group will receive oropharyngeal administration of 0.2 ml colostrum every 3 h, starting between the first 48 to 72 h and continued for 5 consecutive days. Following a similar administration scheme, the control group will receive oropharyngeal administration of sterile water. Stool samples will be collected at the first defecation, as well as on the 7th, 14th, 21st and 28th days after birth for analysis of effect of OAC on gut microbiota and metabolites through 16sRNA gene sequencing and liquid chromatography-mass spectrometry. DISCUSSION This proposal advocates for the promotion of OAC as a safe and relatively beneficial protocol in neonatal intensive care units, which may contribute to the establishment of a dominant intestinal flora. Findings of this study may help improve the health outcomes of preterm infants by establishment of targeted gut microbiota in future studies. TRIAL REGISTRATION NCT05481866 (registered July 30, 2022 on ClinicalTrials.gov).
Collapse
Affiliation(s)
- Na Wang
- Department of Neonatology, The Affiliated Suqian First People's Hospital of Nanjing Medical University, Jiangsu, China
| | - Jia Zhang
- Department of Neonatology, The Affiliated Suqian First People's Hospital of Nanjing Medical University, Jiangsu, China
| | - Zhangbin Yu
- Department of Neonatology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China.
| | - Xudong Yan
- Department of Neonatology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Lian Zhang
- Department of Neonatology, Bao'an Maternal and Child Health Hospital, Shenzhen, Guangdong, China
| | - Haibo Peng
- Department of Neonatology, Bao'an Maternal and Child Health Hospital, Shenzhen, Guangdong, China
| | - Cheng Chen
- Department of Neonatology, Longgang Maternal and Child Health Hospital, Shenzhen, Guangdong, China
| | - Rui Li
- Department of Neonatology, Longgang Maternal and Child Health Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
28
|
Yao J, Ai T, Zhang L, Tang W, Chen Z, Huang Y, Fan Y. Bacterial Colonization in the Airways and Intestines of Twin and Singleton Preterm Neonates: A Single-Center Study. THE CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY = JOURNAL CANADIEN DES MALADIES INFECTIEUSES ET DE LA MICROBIOLOGIE MEDICALE 2023; 2023:2973605. [PMID: 37560084 PMCID: PMC10409585 DOI: 10.1155/2023/2973605] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 07/14/2023] [Accepted: 07/21/2023] [Indexed: 08/11/2023]
Abstract
Limited studies have investigated the microbial colonization of the airways and intestines in preterm neonates. We studied the composition of intestinal and airway bacterial colonies in several preterm twin pairs and singletons to explore the dominant bacteria, assess their variability, and predict their phenotypic and metabolic functions. In this descriptive study, we collected sputum and fetal stool specimens from 10 twin pairs (20 cases) and 20 singleton preterm neonates. These specimens were analyzed using 16S rRNA deep sequencing to study the alpha and beta diversities and community structures of airway and intestinal bacteria and predict their metabolic functions. Specimens from twins and singleton neonates had distinct aggregations of intestinal and airway bacteria but showed similarities and high microbial diversities during initial colonization. The top five phyla were Proteobacteria, Firmicutes, Actinobacteriota, Bacteroidota, and Cyanobacteria. The top ten genera were Streptococcus, Acinetobacter, Ralstonia, Staphylococcus, Comamonas, Enterococcus, Stenotrophomonas, Dechlorosoma, Sphingopyxis, and Rothia. Potentially pathogenic and highly stress-tolerant Gram-negative bacteria were predominant in the intestinal flora. A considerable proportion of colonies recovered from the airway and intestines of preterm neonates were functional bacteria. The richness of the intestinal and airway flora was not significantly different between twins and singletons, and the flora clustered together. Both intestinal and airway bacteria of twins and singletons were similar. The species involved in initial colonization were similar but different in proportions; therefore, changes in microbial structure and richness may not be attributed to these species.
Collapse
Affiliation(s)
- Jiawei Yao
- Division of Pediatric Pulmonology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Tao Ai
- Division of Pediatric Pulmonology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Lei Zhang
- Division of Pediatric Pulmonology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Wei Tang
- Division of Pediatric Pulmonology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Zijin Chen
- Division of Pediatric Pulmonology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yuedong Huang
- Division of Pediatric Pulmonology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yinghong Fan
- Division of Pediatric Pulmonology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| |
Collapse
|
29
|
Stupak A, Gęca T, Kwaśniewska A, Mlak R, Piwowarczyk P, Nawrot R, Goździcka-Józefiak A, Kwaśniewski W. Comparative Analysis of the Placental Microbiome in Pregnancies with Late Fetal Growth Restriction versus Physiological Pregnancies. Int J Mol Sci 2023; 24:ijms24086922. [PMID: 37108086 PMCID: PMC10139004 DOI: 10.3390/ijms24086922] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/03/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
A comparative analysis of the placental microbiome in pregnancies with late fetal growth restriction (FGR) was performed with normal pregnancies to assess the impact of bacteria on placental development and function. The presence of microorganisms in the placenta, amniotic fluid, fetal membranes and umbilical cord blood throughout pregnancy disproves the theory of the "sterile uterus". FGR occurs when the fetus is unable to follow a biophysically determined growth path. Bacterial infections have been linked to maternal overproduction of pro-inflammatory cytokines, as well as various short- and long-term problems. Proteomics and bioinformatics studies of placental biomass allowed the development of new diagnostic options. In this study, the microbiome of normal and FGR placentas was analyzed by LC-ESI-MS/MS mass spectrometry, and the bacteria present in both placentas were identified by analysis of a set of bacterial proteins. Thirty-six pregnant Caucasian women participated in the study, including 18 women with normal pregnancy and eutrophic fetuses (EFW > 10th percentile) and 18 women with late FGR diagnosed after 32 weeks of gestation. Based on the analysis of the proteinogram, 166 bacterial proteins were detected in the material taken from the placentas in the study group. Of these, 21 proteins had an exponentially modified protein abundance index (emPAI) value of 0 and were not included in further analysis. Of the remaining 145 proteins, 52 were also present in the material from the control group. The remaining 93 proteins were present only in the material collected from the study group. Based on the proteinogram analysis, 732 bacterial proteins were detected in the material taken from the control group. Of these, 104 proteins had an emPAI value of 0 and were not included in further analysis. Of the remaining 628 proteins, 52 were also present in the material from the study group. The remaining 576 proteins were present only in the material taken from the control group. In both groups, we considered the result of ns prot ≥ 60 as the cut-off value for the agreement of the detected protein with its theoretical counterpart. Our study found significantly higher emPAI values of proteins representative of the following bacteria: Actinopolyspora erythraea, Listeria costaricensis, E. coli, Methylobacterium, Acidobacteria bacterium, Bacteroidetes bacterium, Paenisporsarcina sp., Thiodiazotropha endol oripes and Clostridiales bacterium. On the other hand, in the control group statistically more frequently, based on proteomic data, the following were found: Flavobacterial bacterium, Aureimonas sp. and Bacillus cereus. Our study showed that placental dysbiosis may be an important factor in the etiology of FGR. The presence of numerous bacterial proteins present in the control material may indicate their protective role, while the presence of bacterial proteins detected only in the material taken from the placentas of the study group may indicate their potentially pathogenic nature. This phenomenon is probably important in the development of the immune system in early life, and the placental microbiota and its metabolites may have great potential in the screening, prevention, diagnosis and treatment of FGR.
Collapse
Affiliation(s)
- Aleksandra Stupak
- Chair and Department of Obstetrics and Pathology of Pregnancy, Medical University of Lublin, 20-059 Lublin, Poland
| | - Tomasz Gęca
- Chair and Department of Obstetrics and Pathology of Pregnancy, Medical University of Lublin, 20-059 Lublin, Poland
| | - Anna Kwaśniewska
- Chair and Department of Obstetrics and Pathology of Pregnancy, Medical University of Lublin, 20-059 Lublin, Poland
| | - Radosław Mlak
- Body Composition Research Laboratory, Department of Preclinical Science, Medical University of Lublin, 20-059 Lublin, Poland
| | - Paweł Piwowarczyk
- 2nd Department of Anesthesiology and Intensive Care Unit, Medical University of Lublin, 20-059 Lublin, Poland
| | - Robert Nawrot
- Department of Molecular Virology, Institute of Experimental Biology, Adam Mickiewicz University in Poznan, 61-712 Poznań, Poland
| | - Anna Goździcka-Józefiak
- Department of Molecular Virology, Institute of Experimental Biology, Adam Mickiewicz University in Poznan, 61-712 Poznań, Poland
| | - Wojciech Kwaśniewski
- Department of Gynecologic Oncology and Gynecology, Medical University of Lublin, 20-059 Lublin, Poland
| |
Collapse
|
30
|
Xiao L, Zhao F. Microbial transmission, colonisation and succession: from pregnancy to infancy. Gut 2023; 72:772-786. [PMID: 36720630 PMCID: PMC10086306 DOI: 10.1136/gutjnl-2022-328970] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 01/10/2023] [Indexed: 02/02/2023]
Abstract
The microbiome has been proven to be associated with many diseases and has been used as a biomarker and target in disease prevention and intervention. Currently, the vital role of the microbiome in pregnant women and newborns is increasingly emphasised. In this review, we discuss the interplay of the microbiome and the corresponding immune mechanism between mothers and their offspring during the perinatal period. We aim to present a comprehensive picture of microbial transmission and potential immune imprinting before and after delivery. In addition, we discuss the possibility of in utero microbial colonisation during pregnancy, which has been highly debated in recent studies, and highlight the importance of the microbiome in infant development during the first 3 years of life. This holistic view of the role of the microbial interplay between mothers and infants will refine our current understanding of pregnancy complications as well as diseases in early life and will greatly facilitate the microbiome-based prenatal diagnosis and treatment of mother-infant-related diseases.
Collapse
Affiliation(s)
- Liwen Xiao
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Fangqing Zhao
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Key Laboratory of System Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| |
Collapse
|
31
|
Kaplina A, Kononova S, Zaikova E, Pervunina T, Petrova N, Sitkin S. Necrotizing Enterocolitis: The Role of Hypoxia, Gut Microbiome, and Microbial Metabolites. Int J Mol Sci 2023; 24:2471. [PMID: 36768793 PMCID: PMC9917134 DOI: 10.3390/ijms24032471] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 01/15/2023] [Accepted: 01/17/2023] [Indexed: 02/01/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is a life-threatening disease that predominantly affects very low birth weight preterm infants. Development of NEC in preterm infants is accompanied by high mortality. Surgical treatment of NEC can be complicated by short bowel syndrome, intestinal failure, parenteral nutrition-associated liver disease, and neurodevelopmental delay. Issues surrounding pathogenesis, prevention, and treatment of NEC remain unclear. This review summarizes data on prenatal risk factors for NEC, the role of pre-eclampsia, and intrauterine growth retardation in the pathogenesis of NEC. The role of hypoxia in NEC is discussed. Recent data on the role of the intestinal microbiome in the development of NEC, and features of the metabolome that can serve as potential biomarkers, are presented. The Pseudomonadota phylum is known to be associated with NEC in preterm neonates, and the role of other bacteria and their metabolites in NEC pathogenesis is also discussed. The most promising approaches for preventing and treating NEC are summarized.
Collapse
Affiliation(s)
- Aleksandra Kaplina
- Research Laboratory for Physiology and Diseases of Newborns, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
| | - Svetlana Kononova
- Group of Protein Synthesis Regulation, Institute of Protein Research, Russian Academy of Sciences, Pushchino 142290, Russia
| | - Ekaterina Zaikova
- Research Laboratory of Autoimmune and Autoinflammatory Diseases, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
| | - Tatiana Pervunina
- Institute of Perinatology and Pediatrics, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
| | - Natalia Petrova
- Research Laboratory for Physiology and Diseases of Newborns, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
| | - Stanislav Sitkin
- Epigenetics and Metagenomics Group, Institute of Perinatology and Pediatrics, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
- Department of Internal Diseases, Gastroenterology and Dietetics, North-Western State Medical University Named after I.I. Mechnikov, St. Petersburg 191015, Russia
| |
Collapse
|
32
|
Messman RD, Lemley CO. Bovine neonatal microbiome origins: a review of proposed microbial community presence from conception to colostrum. Transl Anim Sci 2023; 7:txad057. [PMID: 37334245 PMCID: PMC10276551 DOI: 10.1093/tas/txad057] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 05/24/2023] [Indexed: 06/20/2023] Open
Abstract
In recent years, there has been an influx of research evaluating the roles of the reproductive tract microbiota in modulating reproductive performance. These efforts have resulted in a breadth of research exploring the bovine reproductive tract microbiota. The female reproductive tract microbiota has been characterized during the estrus cycle, at timed artificial insemination, during gestation, and postpartum. Additionally, there are recently published studies investigating in-utero inoculation of the bovine fetus. However, critical review of the literature to understand how the microbial shifts during a dam's lifecycle could impact neonatal outcomes is limited. This review demonstrates a consistency at the phyla level throughout both the maternal, paternal, and neonatal microbiomes. Moreover, this review challenges the current gestational inoculation hypothesis and suggests instead a maturation of the resident uterine microbiota throughout gestation to parturition. Recent literature is indicative of microbial composition influencing metabolomic parameters that have developmental programming effects in feed utilization and metabolic performance later in life. Thus, this review enumerates the potential origins of neonatal microbial inoculation from conception, through gestation, parturition, and colostrum consumption while introducing clear paucities where future research is needed to better understand the ramifications of the reproductive microbiome on neonates.
Collapse
Affiliation(s)
- Riley D Messman
- Department of Animal and Dairy Sciences, Mississippi State University, Mississippi State, MS 39762, USA
| | | |
Collapse
|
33
|
Dilemmas in initiation of very preterm infant enteral feeds-when, what, how? J Perinatol 2023; 43:108-113. [PMID: 36447040 DOI: 10.1038/s41372-022-01564-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 11/04/2022] [Accepted: 11/08/2022] [Indexed: 12/03/2022]
Abstract
With limited clinical evidence available to guide common nutritional decisions, significant variation exists in approaches to enteral feeding for very preterm infants, specifically when feedings are initiated, what is fed, and the method used for feedings. Preclinical studies have highlighted the benefits associated with avoiding nil per os and providing early-stage mother's own milk or colostrum. However, these recommended approaches are often mutually exclusive due to the delays in lactation associated with very preterm delivery, resulting in uncertainty regarding which approach should be prioritized. Few studies have evaluated feeding frequency in preterm infants, with limited generalizability to extremely preterm infants. Therefore, even evidence-based approaches to very preterm infant feed initiation can differ. Future research is needed to identify optimal strategies for enteral nutrition in very preterm infants, but, until then, evidence-informed approaches may vary depending on each neonatal intensive care unit's assessment of risk and benefit.
Collapse
|
34
|
Zeng S, Wang S, Ross RP, Stanton C. The road not taken: host genetics in shaping intergenerational microbiomes. Trends Genet 2022; 38:1180-1192. [PMID: 35773025 DOI: 10.1016/j.tig.2022.05.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/31/2022] [Accepted: 05/31/2022] [Indexed: 02/09/2023]
Abstract
The early-life gut microbiome is linked to human phenotypes as an imbalanced microbiome of this period is implicated in diseases throughout life. Several determinants of early-life gut microbiome are explored, however, mechanisms of acquisition, colonization, and stability of early-life gut microbiome and their interindividual variability remain elusive. Host genetics play a vital role to shape the gut microbiome and interact with it to modulate individual phenotypes in human studies and animal models. Given the microbial linkage between host generations, we discuss the current state of roles of host genetics in forming intergenerational microbiomes associated with mothers, offspring, and those vertically transmitted, providing a basis for taking into account host genetics in future early-life microbiome research. We further expand our discussion to the bidirectional interactions between host gene expression and microbiome in human health.
Collapse
Affiliation(s)
- Shuqin Zeng
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, 610041, China; APC Microbiome Ireland, University College Cork, Cork, T12 YT20, Ireland; Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, P61 C996, Ireland
| | - Shaopu Wang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, 610041, China; APC Microbiome Ireland, University College Cork, Cork, T12 YT20, Ireland; Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, P61 C996, Ireland.
| | - R Paul Ross
- APC Microbiome Ireland, University College Cork, Cork, T12 YT20, Ireland
| | - Catherine Stanton
- APC Microbiome Ireland, University College Cork, Cork, T12 YT20, Ireland; Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, P61 C996, Ireland
| |
Collapse
|
35
|
Wang J, Xiao L, Xiao B, Zhang B, Zuo Z, Ji P, Zheng J, Li X, Zhao F. Maternal and neonatal viromes indicate the risk of offspring's gastrointestinal tract exposure to pathogenic viruses of vaginal origin during delivery. MLIFE 2022; 1:303-310. [PMID: 38818221 PMCID: PMC10989755 DOI: 10.1002/mlf2.12034] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 07/09/2022] [Indexed: 06/01/2024]
Abstract
A cumulative effect of enterovirus and gluten intake on the risk of celiac disease autoimmunity in infants highlights the significance of viral exposure in early life on the health of children. However, pathogenic viruses may be transmitted to the offspring in an earlier period, raising the possibility that women whose vaginas are inhabited by such viruses may have had their babies infected as early as the time of delivery. A high-resolution intergenerational virome atlas was obtained by metagenomic sequencing and virome analysis on 486 samples from six body sites of 99 mother-neonate pairs. We found that neonates had less diverse oral and enteric viruses than mothers. Vaginally delivered newborns seconds after birth had a more similar oral virome and more viruses of vaginal origin than cesarean-section (C-section) newborns (56.9% vs. 5.8%). Such viruses include both Lactobacillus phage and potentially pathogenic viruses, such as herpesvirus, vaccinia virus, and hepacivirus, illustrating a relatively high variety of the pioneer viral taxa at the time of delivery and a delivery-dependent mother-to-neonate transmission along the vaginal-oral-intestinal route. Neonates are exposed to vaginal viruses as they pass through the reproductive tract, and viruses of vaginal origin may threaten their health. These findings challenge the conventional notion that vaginal delivery is definitely better than cesarean delivery from the perspective of microbial transmission. Screening for vaginal virome before delivery is a worthwhile step to advocate in normal labor to eliminate the risk of intergenerational transmission of pathogenic viruses to offspring.
Collapse
Affiliation(s)
- Jinfeng Wang
- College of Food Science and Nutritional EngineeringChina Agricultural UniversityBeijingChina
- Beijing Institutes of Life ScienceChinese Academy of SciencesBeijingChina
| | - Liwen Xiao
- Beijing Institutes of Life ScienceChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Baichuan Xiao
- Beijing Institutes of Life ScienceChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Bing Zhang
- Beijing Institutes of Life ScienceChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Zhenqiang Zuo
- Beijing Institutes of Life ScienceChinese Academy of SciencesBeijingChina
| | - Peifeng Ji
- Beijing Institutes of Life ScienceChinese Academy of SciencesBeijingChina
| | - Jiayong Zheng
- Department of Gynecology and ObstetricsWenzhou People's HospitalWenzhouChina
| | - Xiaoqing Li
- Department of Gynecology and ObstetricsWenzhou People's HospitalWenzhouChina
| | - Fangqing Zhao
- Beijing Institutes of Life ScienceChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| |
Collapse
|
36
|
Gorczyca K, Obuchowska A, Kimber-Trojnar Ż, Wierzchowska-Opoka M, Leszczyńska-Gorzelak B. Changes in the Gut Microbiome and Pathologies in Pregnancy. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19169961. [PMID: 36011603 PMCID: PMC9408136 DOI: 10.3390/ijerph19169961] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/04/2022] [Accepted: 08/10/2022] [Indexed: 05/08/2023]
Abstract
Pregnancy is a special period in a woman's life when her organism undergoes multiple physiological changes so that the fetus has optimal conditions for growth and development. These include modifications in the composition of the microbiome that occur between the first and third trimesters of pregnancy. There is an increase in Akkermansia, Bifidobacterium, and Firmicutes, which have been associated with an increase in the need for energy storage. The growth in Proteobacteria and Actinobacteria levels has a protective effect on both the mother and the fetus via proinflammatory mechanisms. The aim of the study is to review the research on the relationship between the mother's intestinal microbiome and gestational pathologies. Changes in the maternal gut microbiome is probably one of the mechanisms that occurs in various pregnancy diseases such as preeclampsia, fetal growth restriction, gestational diabetes mellitus, excessive gestational weight gain, and premature birth. For this reason, it seems vital to pay attention to certain interventions that can benefit the affected patients both in the short term, by preventing complications during pregnancy, and in the long term, as one of the mechanisms occurring in various gestational diseases is dysbiosis of the maternal intestinal flora.
Collapse
|
37
|
Xu T, Yan L, Sun B, Xu Q, Zhang J, Zhu W, Zhang Q, Chen N, Liu G, Chen F. Impacts of Delivery Mode and Maternal Factors on Neonatal Oral Microbiota. Front Microbiol 2022; 13:915423. [PMID: 35832807 PMCID: PMC9271910 DOI: 10.3389/fmicb.2022.915423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 05/23/2022] [Indexed: 11/16/2022] Open
Abstract
Objectives Initial oral microbial colonization has complicatedly interacted with growth and development. The aim of our study was to discover links between oral microbiota community structure and mode of delivery, maternal factors, such as systemic diseases, abortion history, and pregnancy complications. Methods A total of 177 pregnant women and their neonates were enrolled at Peking university people's hospital. We collected oral samples, medical history, and development phenotype and used a 16S rRNA gene sequence to analyze microbial diversity at all taxonomic levels, network structure, and metabolic characteristics. Results Firmicutes, Proteobacteria, and Actinobacteriota were the most predominant bacteria of neonatal oral samples among these phyla. Alpha-diversity of pregnant women with gestational diabetes mellitus (GDM), abortion history, and without immune diseases was higher than in control groups, and no significant dissimilarity in beta-diversity was observed between different maternal factors. Obvious separation or trend failed to be seen in different development phenotype groups. Besides, Oscillospirales were significantly more abundant in a natural delivery group than in the cesarean section group. Conclusion Our study indicated that maternal factors and mode of delivery influenced the oral microbial structure, but longitudinal studies were indispensable for capturing the long-term effects on neonatal development phenotype and oral microbiota.
Collapse
Affiliation(s)
- Tiansong Xu
- Central Laboratory, Peking University School of Stomatology, Beijing, China
| | - Lihuang Yan
- Department of Obstetrics, Peking University People’s Hospital, Beijing, China
| | - Bohui Sun
- Central Laboratory, Peking University School of Stomatology, Beijing, China
| | - Qi Xu
- Department of Obstetrics, Peking University People’s Hospital, Beijing, China
| | - Jieni Zhang
- Central Laboratory, Peking University School of Stomatology, Beijing, China
| | - Wenhui Zhu
- Central Laboratory, Peking University School of Stomatology, Beijing, China
| | - Qian Zhang
- Central Laboratory, Peking University School of Stomatology, Beijing, China
| | - Ning Chen
- Department of Gastroenterology, Peking University People’s Hospital, Beijing, China
| | - Guoli Liu
- Department of Obstetrics, Peking University People’s Hospital, Beijing, China
| | - Feng Chen
- Central Laboratory, Peking University School of Stomatology, Beijing, China
| |
Collapse
|
38
|
He R, Li P, Wang J, Cui B, Zhang F, Zhao F. The interplay of gut microbiota between donors and recipients determines the efficacy of fecal microbiota transplantation. Gut Microbes 2022; 14:2100197. [PMID: 35854629 PMCID: PMC9302524 DOI: 10.1080/19490976.2022.2100197] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 07/01/2022] [Indexed: 02/04/2023] Open
Abstract
Fecal microbiota transplantation (FMT) is a promising treatment for microbiota dysbiosis associated diseases, such as Clostridioides difficile infection (CDI) and inflammatory bowel disease (IBD). The engraftment of donor bacteria is essential for the effectiveness of FMT, which to some extent depends on the matching of donors and recipients. However, how different types of donor-derived bacteria affect FMT efficacy has not been fully dissected. We recruited two longitudinal IBD cohorts of 103 FMT recipients and further analyzed 1,280 microbiota datasets from 14 public CDI and IBD studies to uncover the effect of donor-derived microbiota in recipients. We found that two enterotypes, RCPT/E and RCPT/B (dominated by Enterobacteriaceae and Bacteroides, respectively), consistently exist in both CDI and IBD patients. Based on a time-course-based multi-cohort analysis of FMT fecal samples, we observed the interplay between recipient and donor-derived microbiota during FMT, in which the FMT outcome was significantly associated with the enterotype and microbiota distance between donor and recipient after FMT. We proposed a new measurement, the ratio of colonizers to residents after FMT (C2R), to quantify the engraftment of donor-derived bacteria in the recipients, and then constructed an enterotype-based statistical model for donor-recipient matching, which was validated by both cross-validation and an additional IBD FMT cohort (n = 42). We believe that with the accumulation of FMT multi-omics datasets, machine learning-based methods will be helpful for rational donor selection for improving efficacy and precision FMT practices.
Collapse
Affiliation(s)
- Ruiqiao He
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Pan Li
- Medical Center for Digestive Diseases, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Key Laboratory of Holistic Integrative Enterology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jinfeng Wang
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China
| | - Bota Cui
- Medical Center for Digestive Diseases, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Key Laboratory of Holistic Integrative Enterology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Faming Zhang
- Medical Center for Digestive Diseases, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Key Laboratory of Holistic Integrative Enterology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Fangqing Zhao
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China
- Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| |
Collapse
|