1
|
Askew LC, Gacasan CA, Barbian ME, Weinberg J, Luo L, Robinson BS, Jones DP, Scharer CD, Jones RM. The Microbial Metabolite δ-Valerobetaine Strengthens the Gut Epithelial Barrier. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:1109-1123. [PMID: 40122460 DOI: 10.1016/j.ajpath.2025.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 02/14/2025] [Accepted: 02/28/2025] [Indexed: 03/25/2025]
Abstract
Metabolic processes within gut microbes generate bioactive metabolites that impact intestinal epithelial barrier function. Herein, gnotobiotic mice and mass spectrometry-based metabolomics were used to identify novel metabolites in host tissues of microbial origin. Of those detected, the gut microbe-generated metabolite δ-valerobetaine (δ-VB) is a potent inhibitor of l-carnitine biosynthesis and a modulator of fatty acid oxidation by mitochondria in liver cells. The bioactivity of δ-VB toward gut epithelial barrier function was assessed. Germ-free mice are devoid of δ-VB, and administration of δ-VB to germ-free mice induced the enrichment of transcript sets associated with gut mitochondrial respiration and fatty acid oxidation in colonic tissue. Furthermore, δ-VB induced the differential expression of genes that function in barrier function in germ-free and conventionally raised mice. Functionally, δ-VB decreased gut barrier permeability and augmented wound healing in cultured gut epithelial cells and elicited cytoprotective and prorestitutive effects in a mouse model of colonic injury. These data indicate that the microbial-derived metabolite δ-VB is a modulator of gut epithelium function, and thus is a molecular target to potentially manage microbiome-host dysbiosis in intestinal health and disease.
Collapse
Affiliation(s)
- Lauren C Askew
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| | - C Anthony Gacasan
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| | - Maria E Barbian
- Division of Neonatology, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| | - Jaclyn Weinberg
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Liping Luo
- Department of Pathology, Emory University School of Medicine, Atlanta, Georgia
| | - Brian S Robinson
- Department of Pathology, Emory University School of Medicine, Atlanta, Georgia
| | - Dean P Jones
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Christopher D Scharer
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia
| | - Rheinallt M Jones
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia.
| |
Collapse
|
2
|
Xu L, He R, Ye X, Wang Y, Hui S, Li H, Chen H, Huang P. Leveraging transcriptome-wide association studies identifies the relationship between upper respiratory flora and cell type-specific gene expression in severe respiratory disease. PLoS One 2025; 20:e0322864. [PMID: 40343915 PMCID: PMC12063895 DOI: 10.1371/journal.pone.0322864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 03/30/2025] [Indexed: 05/11/2025] Open
Abstract
ObjectivesThe upper respiratory tract flora may influence host immunity and modulate susceptibility to viral respiratory infections. This study aimed to investigate the associations between upper respiratory tract flora and immune cells in severe ILI, identify specific microbial taxa and immune response pathways contributing to disease severity, and elucidate how flora influences ILI progression by modulating immune cell functions.MethodsHeritability of GWAS summary data was estimated using LDSC (v1.0.1). Gene-level genetic associations were analyzed with MAGMA. scRNA-seq data were integrated with genetic association data using scDRS. FUSION was used to construct cell type-specific expression quantitative trait locus models based on genotypes and scRNA-seq data from the onek1k project, which were combined with flora abundance-related GWAS data for a transcriptome-wide association study.ResultsFrom the LDSC analysis, data from 1195 severe ILI-associated GWASs with upper respiratory flora(h2 > 0.1) were included in subsequent analysis. TWAS identified 19 significant association pairs (Padj < 0.05), and 1226 differentially expressed genes between mild and severe ILI patients (Padj < 0.05 and | log2FC|>0.25). Functional enrichment analyses using GO, KEGG, and Reactome databases revealed that immune cells,such as CD4 + T effector memory cells, cDCs, NK cells, were enriched in multiple biological processes or pathways.ConclusionsThis study identified associations between severe ILI-related upper respiratory tract flora and cell type-specific gene expression, potentially explaining how differential flora influences ILI progression. CD16 + monocytes exhibited the most differentially expressed genes, followed by proliferating cells and cDCs, highlighting the significant role of immune cell-enriched pathways in ILI progression.
Collapse
Affiliation(s)
- Lei Xu
- Depatment of Epidemiology, Center for Global Health, School of Public Health, National Vaccine Innovation Platiorm, Nanjing Medical University, Nanjing, China
| | - Ran He
- Depatment of Epidemiology, Center for Global Health, School of Public Health, National Vaccine Innovation Platiorm, Nanjing Medical University, Nanjing, China
| | - Xiangyu Ye
- Depatment of Epidemiology, Center for Global Health, School of Public Health, National Vaccine Innovation Platiorm, Nanjing Medical University, Nanjing, China
| | - Yifan Wang
- Department of Infectious Disease, Jurong Hospital Affiliated to Jiangsu University, Jurong, Jiangsu, China
| | - Shirong Hui
- Depatment of Epidemiology, Center for Global Health, School of Public Health, National Vaccine Innovation Platiorm, Nanjing Medical University, Nanjing, China
| | - Haochang Li
- Depatment of Epidemiology, Center for Global Health, School of Public Health, National Vaccine Innovation Platiorm, Nanjing Medical University, Nanjing, China
| | - Hongbo Chen
- Department of Infectious Disease, Jurong Hospital Affiliated to Jiangsu University, Jurong, Jiangsu, China
| | - Peng Huang
- Depatment of Epidemiology, Center for Global Health, School of Public Health, National Vaccine Innovation Platiorm, Nanjing Medical University, Nanjing, China
| |
Collapse
|
3
|
Zhao JX, Elsheikha HM, Shang KM, Su JW, Wei YJ, Qin Y, Zhao ZY, Ma H, Zhang XX. Investigation of the genetic diversity of gut mycobiota of the wild and laboratory mice. Microbiol Spectr 2025; 13:e0284024. [PMID: 40162766 PMCID: PMC12054021 DOI: 10.1128/spectrum.02840-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 03/05/2025] [Indexed: 04/02/2025] Open
Abstract
Mice are colonized by diverse gut fungi, known as the mycobiota, which have received much less attention than bacterial microbiota. Here, we studied the diversities and structures of cecal fungal communities in wild (Lasiopodomys brandtii, Apodemus agrarius, and Microtus fortis) vs laboratory C57BL/6J mice to disentangle the contributions of gut fungi to the adaptation of mice to genetic diversity. Using ITS1 gene sequencing, we obtained 2,912 amplicon sequence variants (ASVs) and characterized the composition and diversity of cecal mycobiota in mice. There were significant differences in the composition of cecal fungal communities between wild and C57BL/6J mice, with more species diversity and richness of fungi in wild mice than C57BL/6J mice. We cultured 428 fungal strains from the cecal mycobiota, sequenced the whole genome of 48 selected strains, and identified 500,849 genes. Functional annotation analysis revealed multiple pathways related to energy metabolism, carbohydrate metabolism, fatty acid metabolism, and enzymes involved in the degradation of polysaccharides, lipids, and proteins, and secondary metabolite biosynthesis. The functions and abundance of Hypocreales and Pleosporales, which included the majority of the crucial metabolic pathways, were significantly higher in wild mice than in C57BL/6J mice. The results suggest that variations in the fungal community composition may relate to the adaptability of mice to their environmental habitats. IMPORTANCE In this study, we analyzed the fungal microbiota of three wild mouse species alongside laboratory mice using ITS1 amplicon sequencing. By integrating whole-genome sequencing with culturomics, we sequenced the genomes of 48 fungi isolated from cultured strains and investigated their biological functions to understand the role of intestinal fungi in the environmental adaptability of wild mice. This investigation has expanded the functional gene repository of gut fungi and shed new light on the intricate interplay between mice and their gut fungal communities. The data offer valuable insight into the ecological adaptation in wild mice, emphasizing the complex and dynamic relationship between the murine hosts and their mycobiota.
Collapse
Affiliation(s)
- Ji-Xin Zhao
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong, China
| | - Hany M. Elsheikha
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, University of Nottingham, Loughborough, United Kingdom
| | - Kai-Meng Shang
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong, China
| | - Jin-Wen Su
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong, China
| | - Yong-Jie Wei
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong, China
| | - Ya Qin
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong, China
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, Jilin, China
| | - Zi-Yu Zhao
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, Jilin, China
| | - He Ma
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong, China
| | - Xiao-Xuan Zhang
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong, China
| |
Collapse
|
4
|
Song J, Fu J. Association between dietary index for gut microbiota and osteoarthritis in the US population: the mediating role of systemic immune-inflammation index. Front Nutr 2025; 12:1543674. [PMID: 40357032 PMCID: PMC12066459 DOI: 10.3389/fnut.2025.1543674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 04/02/2025] [Indexed: 05/15/2025] Open
Abstract
Objective Osteoarthritis (OA) is one of the most prevalent chronic conditions among the elderly. The dietary index for gut microbiota (DI-GM) is a novel proposed indicator reflecting gut microbiome diversity. However, the role of DI-GM in OA remains unclear. This study thus aims to explore the association between DI-GM and the risk of OA and analyze the mediating roles of systemic immune-inflammation index (SII). Methods We utilized data from the National Health and Nutrition Examination Survey (NHANES) spanning 2007-2018. OA was assessed through self-reported questionnaires, and dietary recall data were used to calculate the DI-GM. Univariate and weighted multivariate logistic regression analyses were employed to evaluate the association between DI-GM and OA, the weighted linear regression analyses were employed to investigate the association of DI-GM with SII, while restricted cubic splines (RCS) curves were used to assess the non-linear relationship between these variables. Subgroup analyses were subsequently conducted to validate the robustness of the findings. Mediation analysis evaluated the role of SII. Results This study included 15,875 participants, revealing a significant inverse association between the DI-GM and OA risk (p < 0.001), higher DI-GM demonstrated a substantially reduced OA risk (adjusted model OR: 0.83; 95% CI: 0.79-0.86) and were negatively associated with the SII [β (95% CI): -9.2 (-13.0, -2.0)]. The RCS curve indicated a non-linear relationship between DI-GM and OA risk. Subgroup analysis showed that various demographic and clinical factors did not significantly alter the association between DI-GM and OA risk (interaction p-value > 0.05). The mediating effect of SII accounted for 12.69% of association between DI-GM and OA. Conclusion This study found a significant negatively association between DI-GM and OA prevalence in the US population. Mediation analyses demonstrated a significant mediating effect of SII.
Collapse
Affiliation(s)
| | - Jian Fu
- College of Physical Education, Yangzhou University, Yangzhou, Jiangsu, China
| |
Collapse
|
5
|
Valencia S, Zuluaga M, Florian Pérez MC, Montoya-Quintero KF, Candamil-Cortés MS, Robledo S. Human Gut Microbiome: A Connecting Organ Between Nutrition, Metabolism, and Health. Int J Mol Sci 2025; 26:4112. [PMID: 40362352 PMCID: PMC12071897 DOI: 10.3390/ijms26094112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Revised: 03/21/2025] [Accepted: 03/22/2025] [Indexed: 05/15/2025] Open
Abstract
The gut microbiome plays a vital role in human health, functioning as a metabolic organ that influences nutrient absorption and overall well-being. With growing evidence that dietary interventions can modulate the microbiome and improve health, this review examines whether healthcare systems should prioritize personalized microbiome-targeted therapies, such as probiotics, prebiotics, and microbiota transplants, over traditional pharmaceutical treatments for chronic diseases like obesity, diabetes, cardiovascular risk, and inflammatory conditions. A systematic review using Web of Science and Scopus databases was conducted, followed by a scientometric analysis. Key metabolic pathways, such as dietary fiber fermentation and short-chain fatty acid production, were explored, focusing on their impact on lipid and glucose metabolism. The interactions between microbial metabolites and the immune system were also investigated. Dietary interventions, including increased fiber and probiotic intake, show potential for addressing dysbiosis linked to conditions, such as type 2 diabetes, obesity, and autoimmune diseases. The review emphasizes the need to incorporate microbiome modulation strategies into clinical practice and research, calling for a multidisciplinary approach that integrates nutrition, microbiology, and biochemistry to better understand the gut microbiome's complex role in health.
Collapse
Affiliation(s)
- Sandra Valencia
- Centro de Bioinformática y Biología Computacional de Colombia—BIOS, Grupo de Investigación—BIOS, Parque los Yarumos, Manizales 170002, Colombia;
- Departamento de Ciencias Básicas de la Salud, Facultad de Ciencias para la Salud, Universidad de Caldas, Calle 65 # 26-10, Manizales 170004, Colombia; (M.C.F.P.); (K.F.M.-Q.)
| | - Martha Zuluaga
- Dirección Académica, Universidad Nacional de Colombia, Sede De La Paz, Km 9 Valledupar—La Paz, Cesar 202010, Colombia;
| | - María Cristina Florian Pérez
- Departamento de Ciencias Básicas de la Salud, Facultad de Ciencias para la Salud, Universidad de Caldas, Calle 65 # 26-10, Manizales 170004, Colombia; (M.C.F.P.); (K.F.M.-Q.)
| | - Kevin Fernando Montoya-Quintero
- Departamento de Ciencias Básicas de la Salud, Facultad de Ciencias para la Salud, Universidad de Caldas, Calle 65 # 26-10, Manizales 170004, Colombia; (M.C.F.P.); (K.F.M.-Q.)
| | - Mariana S. Candamil-Cortés
- Centro de Bioinformática y Biología Computacional de Colombia—BIOS, Grupo de Investigación—BIOS, Parque los Yarumos, Manizales 170002, Colombia;
| | - Sebastian Robledo
- Dirección Académica, Universidad Nacional de Colombia, Sede De La Paz, Km 9 Valledupar—La Paz, Cesar 202010, Colombia;
| |
Collapse
|
6
|
Liu Z, Wang P, Wei J, Li J, Luo X, Huang X, Zhang X, Li W, Qin Q. Effect of intestinal microbiota on adaptation to overcrowding stress in grouper (Epinephelus fuscoguttatus♀×E. lanceolatus♂). FISH & SHELLFISH IMMUNOLOGY 2025; 159:110165. [PMID: 39922247 DOI: 10.1016/j.fsi.2025.110165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 01/27/2025] [Accepted: 01/27/2025] [Indexed: 02/10/2025]
Abstract
Density is an important aquaculture parameter. When the pearl gentian grouper (Epinephelus fuscoguttatus♀ × E. lanceolatus♂) is farmed intensively, it could lead to a degradation in genetic resources and an increase in disease outbreaks. The composition of the intestinal microbiota plays a key role in creating a specific intestinal microecosystem, which is essential for the survival, growth, and immune response of the host under environmental stress like overcrowding. This study utilized 16S rRNA sequencing and metabolomics analysis techniques to investigate the differences in intestinal microbial community stability of grouper under different stocking time and density pressure conditions. The research results showed that compared to the low-density group, the high-density group of groupers experienced an increase in mortality rate and feed coefficient in the early stages of culture, while the weight gain rate decreased. Differential analysis of intestinal microbial communities revealed significant differences in the gut microbiota of grouper between different density groups after 10 days of culture, but no significant differences were observed after 20 days of culture. At the same time, intestinal histopathology showed that the high-density group of groupers exhibited a reduction in intestinal villi length and thickness of the intestinal wall after 10 days of culture. However, the intergroup differences had reduced after 20 days of culture. Furthermore, high density cultivation upregulated the expression of inflammatory factors like IL-1β, TNF-α, IL-8, and IL-6 in the intestinal tract of groupers after 10 days of culture. However, after 20 days of culture, the expression levels of intestinal inflammatory factors in both the high-density and low-density groups of groupers were significantly reduced, and the differences between the intergroup diminished. Through correlation analysis of differential metabolites and species in the intestine, multiple metabolites significantly upregulated and associated with the upregulation of the Staphylococcus genus were identified in the intestinal tract of groupers after 20 days of high-density cultivation. The selected four associated metabolites (including creatine, fosinopril, 4-aminobutyric acid, and guanidinopropanoic acid) were validated to significantly reduce the expression of cellular inflammatory factors using the self-established grouper head kidney (HK) cell line. In conclusion, density pressure in the early culture period could affect the stability of the intestinal microbial environment of grouper. As aquaculture time increases, the intestinal microbial community of grouper drives the body's anti-inflammatory response and enhanced its adaptation to density pressure by regulating own structure and secretion of metabolites.
Collapse
Affiliation(s)
- Zetian Liu
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - Pandeng Wang
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Stress Biology, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Jingguang Wei
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - Jialing Li
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Stress Biology, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Xiaoqing Luo
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Stress Biology, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Xiaohong Huang
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - Xin Zhang
- School of Marine Biology and Fisheries, Hainan University, Haikou, 570228, Hainan Province, China
| | - Wenjun Li
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Stress Biology, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China.
| | - Qiwei Qin
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, 528478, China.
| |
Collapse
|
7
|
Kaden T, Alonso‐Román R, Stallhofer J, Gresnigt MS, Hube B, Mosig AS. Leveraging Organ-on-Chip Models to Investigate Host-Microbiota Dynamics and Targeted Therapies for Inflammatory Bowel Disease. Adv Healthc Mater 2025; 14:e2402756. [PMID: 39491534 PMCID: PMC12004439 DOI: 10.1002/adhm.202402756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/29/2024] [Indexed: 11/05/2024]
Abstract
Inflammatory bowel disease (IBD) is an idiopathic gastrointestinal disease with drastically increasing incidence rates. Due to its multifactorial etiology, a precise investigation of the pathogenesis is extremely difficult. Although reductionist cell culture models and more complex disease models in animals have clarified the understanding of individual disease mechanisms and contributing factors of IBD in the past, it remains challenging to bridge research and clinical practice. Conventional 2D cell culture models cannot replicate complex host-microbiota interactions and stable long-term microbial culture. Further, extrapolating data from animal models to patients remains challenging due to genetic and environmental diversity leading to differences in immune responses. Human intestine organ-on-chip (OoC) models have emerged as an alternative in vitro model approach to investigate IBD. OoC models not only recapitulate the human intestinal microenvironment more accurately than 2D cultures yet may also be advantageous for the identification of important disease-driving factors and pharmacological interventions targets due to the possibility of emulating different complexities. The predispositions and biological hallmarks of IBD focusing on host-microbiota interactions at the intestinal mucosal barrier are elucidated here. Additionally, the potential of OoCs to explore microbiota-related therapies and personalized medicine for IBD treatment is discussed.
Collapse
Affiliation(s)
- Tim Kaden
- Dynamic42 GmbH07745JenaGermany
- Institute of Biochemistry IICenter for Sepsis Control and CareJena University Hospital07747JenaGermany
| | - Raquel Alonso‐Román
- Department of Microbial Pathogenicity MechanismsLeibniz Institute for Natural Product Research and Infection Biology – Hans‐Knöll‐Institute07745JenaGermany
- Cluster of Excellence Balance of the MicroverseFriedrich Schiller University Jena07745JenaGermany
- Junior Research Group Adaptive Pathogenicity StrategiesLeibniz Institute for Natural Product Research and Infection Biology – Hans‐Knöll‐Institute07745JenaGermany
| | | | - Mark S. Gresnigt
- Cluster of Excellence Balance of the MicroverseFriedrich Schiller University Jena07745JenaGermany
- Junior Research Group Adaptive Pathogenicity StrategiesLeibniz Institute for Natural Product Research and Infection Biology – Hans‐Knöll‐Institute07745JenaGermany
| | - Bernhard Hube
- Department of Microbial Pathogenicity MechanismsLeibniz Institute for Natural Product Research and Infection Biology – Hans‐Knöll‐Institute07745JenaGermany
- Cluster of Excellence Balance of the MicroverseFriedrich Schiller University Jena07745JenaGermany
- Institute of MicrobiologyFaculty of Biological SciencesFriedrich Schiller University07743JenaGermany
| | - Alexander S. Mosig
- Institute of Biochemistry IICenter for Sepsis Control and CareJena University Hospital07747JenaGermany
- Cluster of Excellence Balance of the MicroverseFriedrich Schiller University Jena07745JenaGermany
| |
Collapse
|
8
|
Weng Q, Hu M, Peng G, Zhu J. DMoVGPE: predicting gut microbial associated metabolites profiles with deep mixture of variational Gaussian Process experts. BMC Bioinformatics 2025; 26:93. [PMID: 40148806 PMCID: PMC11951675 DOI: 10.1186/s12859-025-06110-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 03/10/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Understanding the metabolic activities of the gut microbiome is vital for deciphering its impact on human health. While direct measurement of these metabolites through metabolomics is effective, it is often expensive and time-consuming. In contrast, microbial composition data obtained through sequencing is more accessible, making it a promising resource for predicting metabolite profiles. However, current computational models frequently face challenges related to limited prediction accuracy, generalizability, and interpretability. METHOD Here, we present the Deep Mixture of Variational Gaussian Process Experts (DMoVGPE) model, designed to overcome these issues. DMoVGPE utilizes a dynamic gating mechanism, implemented through a neural network with fully connected layers and dropout for regularization, to select the most relevant Gaussian Process experts. During training, the gating network refines expert selection, dynamically adjusting their contribution based on the input features. The model also incorporates an Automatic Relevance Determination (ARD) mechanism, which assigns relevance scores to microbial features by evaluating their predictive power. Features linked to metabolite profiles are given smaller length scales to increase their influence, while irrelevant features are down-weighted through larger length scales, improving both prediction accuracy and interpretability. CONCLUSIONS Through extensive evaluations on various datasets, DMoVGPE consistently achieves higher prediction performance than existing models. Furthermore, our model reveals significant associations between specific microbial taxa and metabolites, aligning well with findings from existing studies. These results highlight DMoVGPE's potential to provide accurate predictions and to uncover biologically meaningful relationships, paving the way for its application in disease research and personalized healthcare strategies.
Collapse
Affiliation(s)
- Qinghui Weng
- The State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, Jiangsu, People's Republic of China
- The School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi, 214122, Jiangsu, People's Republic of China
| | - Mingyi Hu
- The State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, Jiangsu, People's Republic of China
- The School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi, 214122, Jiangsu, People's Republic of China
| | - Guohao Peng
- The School of Electrical and Electronic Engineering, Nanyang Technological University, Singapore, 639798, Singapore
| | - Jinlin Zhu
- The State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, Jiangsu, People's Republic of China.
- The School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi, 214122, Jiangsu, People's Republic of China.
| |
Collapse
|
9
|
Ageitos L, Boaro A, Cesaro A, Torres MDT, Broset E, de la Fuente-Nunez C. Frog-derived synthetic peptides display anti-infective activity against Gram-negative pathogens. Trends Biotechnol 2025:S0167-7799(25)00044-7. [PMID: 40140310 DOI: 10.1016/j.tibtech.2025.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 02/03/2025] [Accepted: 02/05/2025] [Indexed: 03/28/2025]
Abstract
Novel antibiotics are urgently needed since bacteria are becoming increasingly resistant to existing antimicrobial drugs. Furthermore, available antibiotics are broad spectrum, often causing off-target effects on host cells and the beneficial microbiome. To overcome these limitations, we used structure-guided design to generate synthetic peptides derived from Andersonin-D1, an antimicrobial peptide (AMP) produced by the odorous frog Odorrana andersonii. We found that both hydrophobicity and net charge were critical for its bioactivity, enabling the design of novel, optimized synthetic peptides. These peptides selectively targeted Gram-negative pathogens in single cultures and complex microbial consortia, showed no off-target effects on human cells or beneficial gut microbes, and did not select for bacterial resistance. Notably, they also exhibited in vivo activity in two preclinical murine models. Overall, we present synthetic peptides that selectively target pathogenic infections and offer promising preclinical antibiotic candidates.
Collapse
Affiliation(s)
- Lucía Ageitos
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA; Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA; Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Andreia Boaro
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA; Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA; Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Angela Cesaro
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA; Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA; Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Marcelo D T Torres
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA; Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA; Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Esther Broset
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA; Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA; Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Cesar de la Fuente-Nunez
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA; Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA; Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
10
|
Zhra M, Elahi MA, Tariq A, Abu-Zaid A, Yaqinuddin A. Sirtuins and Gut Microbiota: Dynamics in Health and a Journey from Metabolic Dysfunction to Hepatocellular Carcinoma. Cells 2025; 14:466. [PMID: 40136715 PMCID: PMC11941559 DOI: 10.3390/cells14060466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 03/27/2025] Open
Abstract
Metabolic dysfunction leading to non-alcoholic fatty liver disease (NAFLD) exhibits distinct molecular and immune signatures that are influenced by factors like gut microbiota. The gut microbiome interacts with the liver via a bidirectional relationship with the gut-liver axis. Microbial metabolites, sirtuins, and immune responses are pivotal in different metabolic diseases. This extensive review explores the complex and multifaceted interrelationship between sirtuins and gut microbiota, highlighting their importance in health and disease, particularly metabolic dysfunction and hepatocellular carcinoma (HCC). Sirtuins (SIRTs), classified as a group of NAD+-dependent deacetylases, serve as crucial modulators of a wide spectrum of cellular functions, including metabolic pathways, the inflammatory response, and the process of senescence. Their subcellular localization and diverse functions link them to various health conditions, including NAFLD and cancer. Concurrently, the gut microbiota, comprising diverse microorganisms, significantly influences host metabolism and immune responses. Recent findings indicate that sirtuins modulate gut microbiota composition and function, while the microbiota can affect sirtuin activity. This bidirectional relationship is particularly relevant in metabolic disorders, where dysbiosis contributes to disease progression. The review highlights recent findings on the roles of specific sirtuins in maintaining gut health and their implications in metabolic dysfunction and HCC development. Understanding these interactions offers potential therapeutic avenues for managing diseases linked to metabolic dysregulation and liver pathology.
Collapse
Affiliation(s)
- Mahmoud Zhra
- Department of Anatomy and Genetics, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia;
| | - Muhammad Affan Elahi
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (M.A.E.); (A.A.-Z.)
| | - Aamira Tariq
- Department of Biosciences, COMSATS University Islamabad, Islamabad Campus, Islamabad 45550, Pakistan
| | - Ahmed Abu-Zaid
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (M.A.E.); (A.A.-Z.)
| | - Ahmed Yaqinuddin
- Department of Anatomy and Genetics, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia;
| |
Collapse
|
11
|
Chavez-Arroyo A, Radlinski LC, Bäumler AJ. Principles of gut microbiota assembly. Trends Microbiol 2025:S0966-842X(25)00071-X. [PMID: 40089422 DOI: 10.1016/j.tim.2025.02.014] [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: 02/02/2025] [Revised: 02/20/2025] [Accepted: 02/24/2025] [Indexed: 03/17/2025]
Abstract
The gut microbiota plays a critical role in human health, yet its taxonomic complexity, interpersonal variability, and resistance to change in adulthood present challenges for understanding the factors driving shifts in its composition and function. Here, we propose a hierarchy of ecological factors governing gut microbiota assembly, stability, and resilience. At the apex of this hierarchy is habitat filtering by host-derived electron acceptors, which dictates the ecological guilds that dominate distinct gut regions. Host dietary behavior shapes niche availability within these ecological guilds by regulating nutrient availability. Priority effects preserve taxonomic stability whereas microbial antagonism governs competition for open ecological positions. This framework highlights how host control over microbial energy metabolism directs microbiota self-assembly and maintains gut homeostasis.
Collapse
Affiliation(s)
- Alfredo Chavez-Arroyo
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Ave, Davis, CA 95616, USA
| | - Lauren C Radlinski
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Ave, Davis, CA 95616, USA
| | - Andreas J Bäumler
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Ave, Davis, CA 95616, USA.
| |
Collapse
|
12
|
Fan H, Zhan Y, Cheng X, Tan M, Li Y, Xiong Y, Li Q, Liu W. Lacidophilin tablets relieve irritable bowel syndrome in rats by regulating gut microbiota dysbiosis and intestinal inflammation. Sci Rep 2025; 15:8151. [PMID: 40059226 PMCID: PMC11891319 DOI: 10.1038/s41598-025-91883-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 02/24/2025] [Indexed: 05/13/2025] Open
Abstract
Irritable bowel syndrome (IBS) is a common clinical functional gastrointestinal disease. It has a complex pathophysiological mechanism, in which the imbalance of gut microbiota might play an important role. Lacidophilin tablets (LH) can regulate gut microbiota, but their effect on IBS is unknown. In this study, the IBS model was established by acetic acid enema combined with the constrained stress method, and rats were fed LH for 2 weeks. LH significantly reduced visceral sensitivity and intestinal propulsion rate and improved IBS-induced anxiety and depressive behavior in IBS rats. LH elevated the expression levels of mucin 2, claudin1, and occludin, and ameliorated IBS-induced structural damage to colonic tissues. The gut microbiota analysis revealed that LH altered the structure and composition of the gut microbiota in IBS rats. In addition, LH reduced the expression levels of inflammatory factor-related genes. These results suggest that LH could significantly improve the visceral sensitivity and intestinal motility disorders of IBS rats, relieve anxiety and depression levels, and alleviate the symptoms of IBS rats by regulating gut microbiota and reducing intestinal inflammation.
Collapse
Affiliation(s)
- Huiqun Fan
- State Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Nanchang, 330096, Jiangxi, China
- Research and Development Department, Jiangzhong Pharmaceutical Co., Ltd, Nanchang, 330103, Jiangxi, China
| | - Yang Zhan
- State Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Nanchang, 330096, Jiangxi, China
- Research and Development Department, Jiangzhong Pharmaceutical Co., Ltd, Nanchang, 330103, Jiangxi, China
| | - Xiaoying Cheng
- State Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Nanchang, 330096, Jiangxi, China
- Research and Development Department, Jiangzhong Pharmaceutical Co., Ltd, Nanchang, 330103, Jiangxi, China
| | - Mintao Tan
- State Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Nanchang, 330096, Jiangxi, China
- Research and Development Department, Jiangzhong Pharmaceutical Co., Ltd, Nanchang, 330103, Jiangxi, China
| | - Yingmeng Li
- State Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Nanchang, 330096, Jiangxi, China
- Research and Development Department, Jiangzhong Pharmaceutical Co., Ltd, Nanchang, 330103, Jiangxi, China
| | - Yanxia Xiong
- State Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Nanchang, 330096, Jiangxi, China
- Research and Development Department, Jiangzhong Pharmaceutical Co., Ltd, Nanchang, 330103, Jiangxi, China
| | - Qiong Li
- State Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Nanchang, 330096, Jiangxi, China.
- Department of Food Nutrition and Safety, College of Pharmacy, Jiangxi University of Chinese Medicine, No. 1899 Meiling Road, Nanchang, 330004, Jiangxi, China.
| | - Wenjun Liu
- State Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Nanchang, 330096, Jiangxi, China.
- Research and Development Department, Jiangzhong Pharmaceutical Co., Ltd, Nanchang, 330103, Jiangxi, China.
| |
Collapse
|
13
|
Haridevamuthu B, Sudhakaran G, Rajagopal R, Alfarhan A, Arshad A, Arockiaraj J. Host-Parasite Interactions and Integrated Management Strategies for Ecytonucleospora Hepatopenaei Infection in Shrimp. Acta Parasitol 2025; 70:67. [PMID: 40050501 DOI: 10.1007/s11686-025-01007-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 02/25/2025] [Indexed: 04/26/2025]
Abstract
BACKGROUND Ecytonucleospora hepatopenaei (EHP) is a major parasitic pathogen in shrimp causing hepatopancreatic microsporidiosis, which leads to significant growth retardation and global economic losses. This pathogen employs various immune evasion strategies that complicate treatment and management. PURPOSE This review examines the complex host-parasite interactions, focusing on the immune evasion mechanisms used by EHP. The study explores how EHP manipulates host immune pathways, including NF-κB, JAK/STAT, Toll, and IMD, to suppress immune responses, inhibit antimicrobial peptide production, and avoid detection, thus ensuring its persistence in the host. METHODS The authors reviewed recent research from databases like PubMed, Scopus, and Web of Science, including studies up to 2024. The keywords Ecytonucleospora hepatopenaei, immune evasion, EHP treatment, and associated words with topics were used in this search. RESULTS EHP induces oxidative stress, which weakens the host immune system while simultaneously upregulating antioxidant responses to favor its survival. The parasite also alters the gut microbiota and disrupts key cellular processes, such as cell cycle regulation, further enhancing its ability to sustain infection. CONCLUSION This review highlights the need for integrated management strategies, including disease-resistant breeding, microbiota modulation, and advanced diagnostics, to combat EHP. By providing an overview of EHP's immune evasion tactics, this study aims to advance knowledge in the field and support efforts to improve shrimp health and aquaculture sustainability.
Collapse
Affiliation(s)
- Balasubramanian Haridevamuthu
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulatur, Chengalpattu District, Tamil Nadu, 603203, India
| | - Gokul Sudhakaran
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Thandalam, Chennai, Tamil Nadu, 602105, India
| | - Rajakrishnan Rajagopal
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Ahmed Alfarhan
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Aziz Arshad
- Department of Aquaculture, Faculty of Agriculture, Universiti Putra Malaysia, Serdang, Selangor, 43400, Malaysia.
| | - Jesu Arockiaraj
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulatur, Chengalpattu District, Tamil Nadu, 603203, India.
| |
Collapse
|
14
|
Zhu TF, Guo HP, Nie L, Chen J. Oral administration of LEAP2 enhances immunity against Edwardsiella tarda through regulation of gut bacterial community and metabolite in mudskipper. FISH & SHELLFISH IMMUNOLOGY 2025; 158:110128. [PMID: 39824300 DOI: 10.1016/j.fsi.2025.110128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 01/07/2025] [Accepted: 01/14/2025] [Indexed: 01/20/2025]
Abstract
The liver-expressed antimicrobial peptide 2 (LEAP2) is gaining recognition for its immune regulatory functions beyond direct antimicrobial activity. In this study, we investigated the role of mudskipper (Boleophthalmus pectinirostris) LEAP2 (BpLEAP2) in enhancing the survival, gut health, and immune resilience against Edwardsiella tarda infection. Pre-oral delivery of BpLEAP2 significantly improved survival rates and mitigated infection-induced damage to the gut, as evidenced by preserved villus length and goblet cell count. Analysis of gut microbial communities using 16S rRNA sequencing revealed that pre-oral delivery of BpLEAP2 increased microbial diversity, evenness, and the abundance of beneficial genera such as Pseudoalteromonas and Shewanella, while reducing pathogenic genera like Pseudorhodobacter. Metabolomic profiling showed that BpLEAP2 altered the gut metabolite composition, significantly increasing levels of bile acids and amino acids, which are known to support gut health and immune responses. Correlation analysis demonstrated strong positive associations between BpLEAP2-induced microbial shifts and increased metabolites involved in amino acid metabolism. These findings suggest that BpLEAP2 promotes intestinal homeostasis by modulating gut microbiota composition and enhancing beneficial metabolite production, ultimately improving gut barrier integrity and conferring resistance against E. tarda infection. This study highlights the potential application of BpLEAP2 in enhancing disease resilience in aquaculture species, offering a promising strategy for sustainable aquaculture practices.
Collapse
Affiliation(s)
- Ting-Fang Zhu
- State Key Laboratory for Quality and Safety of Agro-Products, Ningbo University, Ningbo 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo, 315832, China; Key Laboratory of Aquacultural Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China
| | - Hai-Peng Guo
- State Key Laboratory for Quality and Safety of Agro-Products, Ningbo University, Ningbo 315211, China
| | - Li Nie
- State Key Laboratory for Quality and Safety of Agro-Products, Ningbo University, Ningbo 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo, 315832, China; Key Laboratory of Aquacultural Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China.
| | - Jiong Chen
- State Key Laboratory for Quality and Safety of Agro-Products, Ningbo University, Ningbo 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo, 315832, China; Key Laboratory of Aquacultural Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China.
| |
Collapse
|
15
|
Medriano CA, Kim S, Kim LH, Bae S. Chronic Exposure of Adult Zebrafish to Polyethylene and Polyester-based Microplastics: Metabolomic and Gut Microbiome Alterations Reflecting Dysbiosis and Resilience. JOURNAL OF HAZARDOUS MATERIALS 2025; 484:136691. [PMID: 39642737 DOI: 10.1016/j.jhazmat.2024.136691] [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: 08/24/2024] [Revised: 11/19/2024] [Accepted: 11/25/2024] [Indexed: 12/09/2024]
Abstract
The study explored the ecotoxicological effects of chronic exposure to microplastic (MP) on adult zebrafish, focusing on environmentally relevant concentrations of polyethylene (PE) beads and polyester (PES). High-throughput untargeted metabolomics via UPLC-QToF-MS and 16S metagenomics for gut microbiota analysis were used to assess ecotoxicity in zebrafish exposed to varying concentrations of PE and PES. The VIP (Variable Importance in Projection) scores indicated PE exposure primarily impacted phospholipids, ceramides, and nucleotide-related compounds, while PES exposure led to alterations in lipid-related compounds, chitin, and amino acid derivatives. From MSEA (Metabolite Set Enrichment Analysis) and Mummichog analyses, PE and PES significantly disrupted key metabolomic pathways associated with inflammation, immune responses, and apoptosis, including leukotriene and arachidonic acid metabolism and the formation of putative anti-inflammatory metabolites from EPA. PE caused physical disruption and inflammation of the epithelial barrier, whereas PES affected gut microbiota interactions, impairing digestion and metabolism. Although alpha diversity within the gut microbiome remained stable, beta diversity analysis revealed significant shifts in microbial composition and structure, suggesting a disruption of functional balance and an increased susceptibility to pathogens. Chronic PE and PES exposures induced shifts in the gut microbial community and interaction network with potential increases in pathogenic bacteria and alteration in commensal bacteria, demonstrating the microbiome's resilience and adaptability to stressors of MPs exposure. High-throughput metabolomics and 16S metagenomics revealed potential chronic diseases associated with inflammation, immune system disorders, metabolic dysfunction, and gut dysbiosis, highlighting the complex relationship between gut microbiome resilience and metabolic disruption under MP-induced stress, with significant ecological implications.
Collapse
Affiliation(s)
- Carl Angelo Medriano
- Department of Civil and Environmental Engineering, National University of Singapore, Singapore
| | - Sungpyo Kim
- Research Institute for Advanced Industrial Technology, Korea University, Republic of Korea
| | - Lan Hee Kim
- Research Institute for Advanced Industrial Technology, Korea University, Republic of Korea; Department of Environmental System Engineering, Korea University, Republic of Korea
| | - Sungwoo Bae
- Department of Environmental System Engineering, Korea University, Republic of Korea.
| |
Collapse
|
16
|
Afzal H, Shaukat A, Ul Haq MZ, Khaliq N, Zahid M, Shakeel L, Wasay Zuberi MA, Akilimali A. Serum metabolic profiling analysis of chronic gastritis and gastric cancer by untargeted metabolomics. Ann Med Surg (Lond) 2025; 87:583-597. [PMID: 40110261 PMCID: PMC11918594 DOI: 10.1097/ms9.0000000000002977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 01/12/2025] [Indexed: 03/22/2025] Open
Abstract
Chronic gastritis (CG), particularly when associated with Helicobacter pylori (H. pylori) infection, is a significant precursor to gastric cancer (GC), a leading cause of cancer-related deaths worldwide. The persistent inflammation in CG, driven by factors such as H. pylori, induces oxidative stress and DNA damage in gastric epithelial cells, which can lead to malignant transformation. Atrophic gastritis, a form of CG, can be categorized into autoimmune and H. pylori-associated types, both of which increase the risk of GC development, particularly when compounded by external factors like smoking and dietary habits. This manuscript explores the pathophysiological mechanisms underlying CG and its progression to GC, highlighting the critical role of metabolomics in advancing our understanding of these processes. Metabolomics, the comprehensive study of metabolites, offers a novel approach to identifying biomarkers that could facilitate early detection and improve the accuracy of GC diagnosis and prognosis. The analysis of metabolic alterations, particularly in glucose, lipid, and amino acid metabolism, reveals distinct biochemical pathways associated with the progression from benign gastritis to malignancy. Integrating metabolomic profiling with traditional diagnostic methods can revolutionize GC management, enabling more personalized treatment strategies and improving clinical outcomes. However, significant challenges remain, including the need to validate biomarkers across diverse populations and standardize metabolomic techniques. Future research should address these challenges to fully realize the potential of metabolomics in early GC detection and treatment, ultimately aiming to reduce the global burden of this deadly disease.
Collapse
Affiliation(s)
- Hadiya Afzal
- Department of Internal Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | - Ayesha Shaukat
- Department of Internal Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | - Muhammad Zain Ul Haq
- Department of Internal Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | - Nawal Khaliq
- Department of Internal Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | - Maha Zahid
- Department of Internal Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | - Laiba Shakeel
- Department of Internal Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | | | - Aymar Akilimali
- Department of Research, Medical Research Circle (MedReC), Goma, Democratic Republic of the Congo
| |
Collapse
|
17
|
Gregory CL, Bradford EL, Fell RD, Haak DC, Belden LK. Utilizing a novel fecal sampling method to examine resistance of the honey bee (Apis mellifera) gut microbiome to a low dose of tetracycline. PLoS One 2025; 20:e0317129. [PMID: 39820943 PMCID: PMC11737664 DOI: 10.1371/journal.pone.0317129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 12/21/2024] [Indexed: 01/19/2025] Open
Abstract
Disruption of host-associated microbial communities can have detrimental impacts on host health. However, the capacity of individual host-associated microbial communities to resist disturbance has not been well defined. Using a novel fecal sampling method for honey bees (Apis mellifera), we examined the resistance of the honey bee gut microbiome to disruption from a low dose of the antibiotic, tetracycline (4.5 μg). Prior to the experiment, bacterial communities from fecal samples were compared to communities from dissected whole guts of the same individuals to ensure fecal samples accurately represented the gut microbiome. Fecal samples were collected from lab-caged honey bees prior to, and five days after, tetracycline exposure to assess how antibiotic disturbance affected the communities of individuals. We used metrics of alpha and beta diversity calculated from 16S rRNA gene amplicon sequences to compare gut community structure. Low dose tetracycline exposure did not consistently change honey bee gut microbiome structure, but there was individual variation in response to exposure and specific taxa (one ASV assigned to Lactobacillus kunkeei and one ASV in the genus Bombella) were differentially abundant following tetracycline treatment. To assess whether individual variation could be influenced by the presence of tetracycline resistance genes, we quantified the abundance of tet(B) and tet(M) with qPCR. The abundance of tet(M) prior to tetracycline treatment was negatively correlated with change in community membership, assessed by difference in Jaccard dissimilarity over the five-day experiment. Our results suggest that the honey bee gut microbiome has some ability to resist or recover from antibiotic-induced change, specific taxa may vary in their susceptibility to tetracycline exposure, and antibiotic resistance genes may contribute to gut microbiome resistance.
Collapse
Affiliation(s)
- Casey L. Gregory
- Department of Biological Sciences, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Emma L. Bradford
- Department of Biological Sciences, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Richard D. Fell
- Department of Entomology, Virginia Tech, Blacksburg, Virginia, United States of America
| | - David C. Haak
- School of Plant and Environmental Sciences, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Lisa K. Belden
- Department of Biological Sciences, Virginia Tech, Blacksburg, Virginia, United States of America
| |
Collapse
|
18
|
Ali Z, Khan I, Iqbal MS, Shi H, Ding L, Hong M. Impact of copper stress in the intestinal barriers and gut microbiota of Chinese stripe-necked turtle (Mauremys sinensis). ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 290:117723. [PMID: 39827614 DOI: 10.1016/j.ecoenv.2025.117723] [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: 09/16/2024] [Revised: 01/07/2025] [Accepted: 01/10/2025] [Indexed: 01/22/2025]
Abstract
Copper is used to treat algal blooms, macrophyte infestations and other environmental issues, but its rising ambient levels harm aquatic animals, especially their intestines. However, its impact on turtles' digestive health is not well understood, and the risks are unclear. This study investigates the effects of copper on the intestinal health of Chinese stripe-necked turtle, focusing on histomorphology, mucosal barrier function, gene expression, and gut microbiota. Copper stress caused intestinal damage, characterized by shortened villi, inflammatory cell infiltration, and reduced epithelial layer thickness, as well as decreased acidic mucins, increased villi edema and inflammation. The mRNA expression level of bacteriostatic enzymes significantly reduced. Furthermore, This study found that copper exposure increases gut permeability by suppressing tight junction genes and triggers an inflammatory response in the gut, as indicated by elevated inflammatory cytokines. At the phylum level, Firmicutes exhibited a significant decrease, whereas Bacteroidota displayed a notable increase, and Fusobacteriota showed a substantial reduction in relative abundance in copper-treated groups. Similarly, at genus level Romboutsia, Cetobacterium decreased, while Turicibacter and Sarcina significantly increases in copper-treated groups compared to the control. This indicating the unique properties of copper including its essentiality, reactivity, and accumulation enables it to profoundly impact gut bacteria, altering both their composition and function. Copper's dual role as a nutrient and toxicant uniquely impacts gut microbes. Our findings suggest that copper stress compromises the intestinal physical, immune, chemical, and microbial barrier in M. sinensis, all of which contribute to the turtle's poor health.
Collapse
Affiliation(s)
- Zeeshan Ali
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, Key Laboratory of Tropical Animal and Plant Ecology of Hainan Province, College of Life Sciences, Hainan Normal University, Haikou 571158, China
| | - Ijaz Khan
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, Key Laboratory of Tropical Animal and Plant Ecology of Hainan Province, College of Life Sciences, Hainan Normal University, Haikou 571158, China
| | - Muhammad Shahid Iqbal
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, Key Laboratory of Tropical Animal and Plant Ecology of Hainan Province, College of Life Sciences, Hainan Normal University, Haikou 571158, China
| | - Haitao Shi
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, Key Laboratory of Tropical Animal and Plant Ecology of Hainan Province, College of Life Sciences, Hainan Normal University, Haikou 571158, China
| | - Li Ding
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, Key Laboratory of Tropical Animal and Plant Ecology of Hainan Province, College of Life Sciences, Hainan Normal University, Haikou 571158, China.
| | - Meiling Hong
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, Key Laboratory of Tropical Animal and Plant Ecology of Hainan Province, College of Life Sciences, Hainan Normal University, Haikou 571158, China.
| |
Collapse
|
19
|
Perez V, Boulo V, De Lorgeril J, Pham D, Ansquer D, Plougoulen G, Ballan V, Lam JS, Romatif O, Le Luyer J, Falchetto C, Basset C, Flohr S, Maamaatuaiahutapu M, Lafille MA, Lau C, Saulnier D, Wabete N, Callac N. Hemolymph microbiota and immune effectors' expressions driven by geographical rearing acclimation of the aquacultured Penaeus stylirostris. Anim Microbiome 2025; 7:5. [PMID: 39799372 PMCID: PMC11725212 DOI: 10.1186/s42523-025-00376-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 01/02/2025] [Indexed: 01/15/2025] Open
Abstract
BACKGROUND In holobiont, microbiota is known to play a central role on the health and immunity of its host. Then, understanding the microbiota, its dynamic according to the environmental conditions and its link to the immunity would help to react to potential dysbiosis of aquacultured species. While the gut microbiota is highly studied, in marine invertebrates the hemolymph microbiota is often set aside even if it remains an important actor of the hemolymph homeostasis. Indeed, the hemolymph harbors the factors involved in the animal homeostasis that interacts with the microbiota, the immunity. In the Southwest Pacific, the high economical valued shrimp Penaeus stylirostris is reared in two contrasted sites, in New Caledonia (NC) and in French Polynesia (FP). RESULTS We characterized the active microbiota inhabiting the hemolymph of shrimps while considering its stability during two seasons and at a one-month interval and evidenced an important microbial variability between the shrimps according to the rearing conditions and the sites. We highlighted specific biomarkers along with a common core microbiota composed of 6 ASVs. Putative microbial functions were mostly associated with bacterial competition, infections and metabolism in NC, while they were highly associated with the cell metabolism in FP suggesting a rearing site discrimination. Differential relative expression of immune effectors measured in the hemolymph of two shrimp populations from NC and FP, exhibited higher level of expression in NC compared to FP. In addition, differential relative expression of immune effectors was correlated to bacterial biomarkers based on their geographical location. CONCLUSIONS Our data suggest that, in Pacific shrimps, both the microbiota and the expression of the immune effectors could have undergone differential immunostimulation according to the rearing site as well as a geographical adaptative divergence of the shrimps as an holobiont, to their rearing sites. Further, the identification of proxies such as the core microbiota and site biomarkers, could be used to guide future actions to monitor the bacterial microbiota and thus preserve the productions.
Collapse
Affiliation(s)
- Valérie Perez
- Ifremer, IRD, Université de la Nouvelle-Calédonie, Université de La Réunion, CNRS, UMR 9220 ENTROPIE, Nouméa, 98800, New Caledonia
- Ifremer, MASAE Microbiologie Aliment Santé Environnement, Nantes, 44000, France
| | - Viviane Boulo
- Ifremer, IRD, Université de la Nouvelle-Calédonie, Université de La Réunion, CNRS, UMR 9220 ENTROPIE, Nouméa, 98800, New Caledonia
- Interactions Hôtes Pathogènes Environnements (IHPE), Université de Montpellier, CNRS, Ifremer, Université de Perpignan Via Domitia, Montpellier, 34090, France
| | - Julien De Lorgeril
- Ifremer, IRD, Université de la Nouvelle-Calédonie, Université de La Réunion, CNRS, UMR 9220 ENTROPIE, Nouméa, 98800, New Caledonia
| | - Dominique Pham
- Ifremer, IRD, Université de la Nouvelle-Calédonie, Université de La Réunion, CNRS, UMR 9220 ENTROPIE, Nouméa, 98800, New Caledonia
| | - Dominique Ansquer
- Ifremer, IRD, Université de la Nouvelle-Calédonie, Université de La Réunion, CNRS, UMR 9220 ENTROPIE, Nouméa, 98800, New Caledonia
| | - Gwenola Plougoulen
- Ifremer, IRD, Université de la Nouvelle-Calédonie, Université de La Réunion, CNRS, UMR 9220 ENTROPIE, Nouméa, 98800, New Caledonia
| | - Valentine Ballan
- Ifremer, IRD, Université de la Nouvelle-Calédonie, Université de La Réunion, CNRS, UMR 9220 ENTROPIE, Nouméa, 98800, New Caledonia
| | - Jean-Sébastien Lam
- Ifremer, IRD, Université de la Nouvelle-Calédonie, Université de La Réunion, CNRS, UMR 9220 ENTROPIE, Nouméa, 98800, New Caledonia
| | - Océane Romatif
- Interactions Hôtes Pathogènes Environnements (IHPE), Université de Montpellier, CNRS, Ifremer, Université de Perpignan Via Domitia, Montpellier, 34090, France
| | - Jeremy Le Luyer
- Ifremer, Université de la Polynésie Française, ILM, IRD, UMR 241 SECOPOL, Tahiti, French Polynesia, F-98719, France
| | - Corinne Falchetto
- Ifremer, Université de la Polynésie Française, ILM, IRD, UMR 241 SECOPOL, Tahiti, French Polynesia, F-98719, France
| | - Caline Basset
- Ifremer, Université de la Polynésie Française, ILM, IRD, UMR 241 SECOPOL, Tahiti, French Polynesia, F-98719, France
| | - Stanley Flohr
- DRM, Direction des Ressources Marines, Papeete, 98713, French Polynesia
| | | | | | - Christophe Lau
- DRM, Direction des Ressources Marines, Papeete, 98713, French Polynesia
| | - Denis Saulnier
- Ifremer, Université de la Polynésie Française, ILM, IRD, UMR 241 SECOPOL, Tahiti, French Polynesia, F-98719, France
| | - Nelly Wabete
- Ifremer, IRD, Université de la Nouvelle-Calédonie, Université de La Réunion, CNRS, UMR 9220 ENTROPIE, Nouméa, 98800, New Caledonia
| | - Nolwenn Callac
- Ifremer, IRD, Université de la Nouvelle-Calédonie, Université de La Réunion, CNRS, UMR 9220 ENTROPIE, Nouméa, 98800, New Caledonia.
| |
Collapse
|
20
|
Kim MJ, Song MH, Ji YS, Park JW, Shin YK, Kim SC, Kim G, Cho B, Park H, Ku JL, Jeong SY. Cell free supernatants of Bifidobacterium adolescentis and Bifidobacterium longum suppress the tumor growth in colorectal cancer organoid model. Sci Rep 2025; 15:935. [PMID: 39762302 PMCID: PMC11704243 DOI: 10.1038/s41598-024-83048-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
The probiotic gut microbiome and its metabolites are pivotal in regulating host metabolism, inflammation, and immunity. Host genetics, colonization at birth, the host lifestyle, and exposure to diseases and drugs determine microbial composition. Dysbiosis and disruption of homeostasis in the beneficial microbiome have been reported to be involved in the tumorigenesis and progression of colorectal cancer (CRC). However, the influence of bacteria-secreted metabolites on CRC growth is yet to be fully elucidated. In this study, we compared the microbial composition of CRC patients to healthy controls to identify distinct patterns of microbiota-derived metabolites in CRC patients. Metagenomic analysis demonstrated that beneficial bacteria strains; Blautia producta, Bifidobacterium adolescentis, and Bifidobacterium longum decreased, while Parabacteroides distasonis and Bacteroides ovatus were more prevalent in the CRC patient group. Treatment of cancer organoid lines with microbial culture supernatants from Blautia producta, Bifidobacterium adolescentis, and Bifidobacterium longum showed remarkable inhibition of cancer growth. This study demonstrates that the bacterial metabolites depleted in CRC patients may inhibit cancer growth and highlights the effects of microbiome-derived metabolites on CRC growth.
Collapse
Affiliation(s)
- Min Jung Kim
- Department of Surgery, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Cancer Research Institute, Seoul National University, Seoul, 03080, Republic of Korea
| | - Myoung-Hyun Song
- Cancer Research Institute, Seoul National University, Seoul, 03080, Republic of Korea
- Korean Cell Line Bank, Laboratory of Cell Biology, Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Yo-Sep Ji
- Holzapfel Effective Microbes (HEM) Pharma, Handong Global University, Pohang, Gyungbuk, Republic of Korea
| | - Ji Won Park
- Department of Surgery, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Cancer Research Institute, Seoul National University, Seoul, 03080, Republic of Korea
| | - Young-Kyoung Shin
- Cancer Research Institute, Seoul National University, Seoul, 03080, Republic of Korea
- Korean Cell Line Bank, Laboratory of Cell Biology, Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Soon-Chan Kim
- Cancer Research Institute, Seoul National University, Seoul, 03080, Republic of Korea
- Korean Cell Line Bank, Laboratory of Cell Biology, Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Gihyeon Kim
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Beomki Cho
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Hansoo Park
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Ja-Lok Ku
- Cancer Research Institute, Seoul National University, Seoul, 03080, Republic of Korea.
- Korean Cell Line Bank, Laboratory of Cell Biology, Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
- Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
| | - Seung-Yong Jeong
- Department of Surgery, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
- Cancer Research Institute, Seoul National University, Seoul, 03080, Republic of Korea.
| |
Collapse
|
21
|
Shang Y, Wu X, Wang X, Chen Y, Dong Y, Wei Q, Pang B, Wang Q, Liu G, Dou H, Zhang H. Microcystin exposure alters gut microbiota composition in fish: An in-Situ analysis of post-bloom effects in Hulun Lake, China. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 363:125174. [PMID: 39461610 DOI: 10.1016/j.envpol.2024.125174] [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: 07/02/2024] [Revised: 09/23/2024] [Accepted: 10/20/2024] [Indexed: 10/29/2024]
Abstract
Cyanobacterial blooms are one of the most common stressors aquatic plants and animals encounter in freshwater ecosystems such as rivers and lakes. Following such outbreaks, some cyanobacteria release toxins, notably microcystins, which are highly toxic. Although numerous studies have explored the effects of microcystins on fish, their in-situ effects on the fish gut microbiome remain unexamined. Our objectives were to examine the fish gut microbiome before (Ju) and after (Au) cyanobacterial blooms and to monitor water quality. We collected Ju and Au fish gut and water samples from Hulun Lake. Using 16S rRNA full-length sequencing, we analyzed the composition, structure, and function of the fish gut microbiome. Results revealed significant disparities in microcystin concentrations between the Ju and Au water samples. In addition, the microcystin concentration in the carp gut was significantly higher than that in its muscle after the cyanobacterial bloom outbreak. Notably, the Au group demonstrated an increase in the microcystin level in water and a marked reduction in fish gut microbiota diversity compared with the Ju group. The cyanobacterial bloom decreased gut microbiome diversity in fish, with the Au group exhibiting a significantly reduced abundance of bacteria related to gut stability and microcystin degradation compared to the Ju group. Furthermore, we observed an upregulation of disease-associated bacterial metabolic functions in the Au group. In conclusion, these findings suggest that microcystins influence the composition and function of fish gut microbiota, improving our understanding of the interaction between fish gut microbiome and their environment. This study offers new perspectives on the adaptive mechanisms of aquatic organisms to cyanobacterial blooms.
Collapse
Affiliation(s)
- Yongquan Shang
- College of Life Sciences, Qufu Normal University, Qufu, Shandong Province, China
| | - Xiaoyang Wu
- College of Life Sciences, Qufu Normal University, Qufu, Shandong Province, China
| | - Xibao Wang
- College of Life Sciences, Qufu Normal University, Qufu, Shandong Province, China
| | - Yao Chen
- College of Life Sciences, Qufu Normal University, Qufu, Shandong Province, China
| | - Yuehuan Dong
- College of Life Sciences, Qufu Normal University, Qufu, Shandong Province, China; College of Wildlife and Protected Area, Northeast Forestry University, Harbin, China
| | - Qinguo Wei
- College of Life Sciences, Qufu Normal University, Qufu, Shandong Province, China
| | - Bo Pang
- Hulunbuir Academy of Inland Lakes in Northern Cold & Arid Areas, Hulunbuir, China
| | - Qi Wang
- Hulunbuir Academy of Inland Lakes in Northern Cold & Arid Areas, Hulunbuir, China
| | - Gang Liu
- College of Life Sciences, Qufu Normal University, Qufu, Shandong Province, China
| | - Huashan Dou
- Hulunbuir Academy of Inland Lakes in Northern Cold & Arid Areas, Hulunbuir, China
| | - Honghai Zhang
- College of Life Sciences, Qufu Normal University, Qufu, Shandong Province, China.
| |
Collapse
|
22
|
Dawkins JJ, Gerber GK. MMETHANE: interpretable AI for predicting host status from microbial composition and metabolomics data. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.13.628441. [PMID: 39713330 PMCID: PMC11661223 DOI: 10.1101/2024.12.13.628441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Metabolite production, consumption, and exchange are intimately involved with host health and disease, as well as being key drivers of host-microbiome interactions. Despite the increasing prevalence of datasets that jointly measure microbiome composition and metabolites, computational tools for linking these data to the status of the host remain limited. To address these limitations, we developed MMETHANE, an open-source software package that implements a purpose-built deep learning model for predicting host status from paired microbial sequencing and metabolomic data. MMETHANE incorporates prior biological knowledge, including phylogenetic and chemical relationships, and is intrinsically interpretable, outputting an English-language set of rules that explains its decisions. Using a compendium of six datasets with paired microbial composition and metabolomics measurements, we showed that MMETHANE always performed at least on par with existing methods, including blackbox machine learning techniques, and outperformed other methods on >80% of the datasets evaluated. We additionally demonstrated through two cases studies analyzing inflammatory bowel disease gut microbiome datasets that MMETHANE uncovers biologically meaningful links between microbes, metabolites, and disease status.
Collapse
|
23
|
Gopal RK, Ganesh PS, Pathoor NN. Synergistic Interplay of Diet, Gut Microbiota, and Insulin Resistance: Unraveling the Molecular Nexus. Mol Nutr Food Res 2024; 68:e2400677. [PMID: 39548908 DOI: 10.1002/mnfr.202400677] [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: 08/21/2024] [Revised: 10/04/2024] [Indexed: 11/18/2024]
Abstract
This comprehensive review explores the intricate relationship between gut microbiota, diet, and insulin resistance, emphasizing the novel roles of diet-induced microbial changes in influencing metabolic health. It highlights how diet significantly influences gut microbiota composition, with different dietary patterns fostering diverse microbial communities. These diet-induced changes in the microbiome impact human metabolism by affecting inflammation, energy balance, and insulin sensitivity, particularly through microbial metabolites like short-chain fatty acids (SCFAs). Focusing the key mediators like endotoxemia and systemic inflammation, and introduces personalized microbiome-based therapeutic strategies, it also investigates the effects of dietary components-fiber, polyphenols, and lipids-on microbiota and insulin sensitivity, along with the roles of protein intake and amino acid metabolism. The study compares the effects of Western and Mediterranean diets on the microbiota-insulin resistance axis. Therapeutic implications, including probiotics, fecal microbiota transplantation (FMT), and personalized diets, are discussed. Key findings reveal that high-fat diets, especially those rich in saturated fats, contribute to dysbiosis and increased intestinal permeability, while high-fiber diets promote beneficial bacteria and SCFAs. The review underscores the future potential of food and microbiota interventions for preventing or managing insulin resistance.
Collapse
Affiliation(s)
- Rajesh Kanna Gopal
- Department of Microbiology, Centre for Infectious Diseases, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University (Deemed to be University), Chennai, Tamil Nadu, 600077, India
| | - Pitchaipillai Sankar Ganesh
- Department of Microbiology, Centre for Infectious Diseases, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University (Deemed to be University), Chennai, Tamil Nadu, 600077, India
| | - Naji Naseef Pathoor
- Department of Microbiology, Centre for Infectious Diseases, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University (Deemed to be University), Chennai, Tamil Nadu, 600077, India
| |
Collapse
|
24
|
Liu W, Cheng H, Zhang H, Liu G, Yin X, Zhang C, Jiang R, Wang Z, Ding X. Effect of Lactobacillus paracasei LK01 on Growth Performance, Antioxidant Capacity, Immunity, Intestinal Health, and Serum Biochemical Indices in Broilers. Animals (Basel) 2024; 14:3474. [PMID: 39682439 DOI: 10.3390/ani14233474] [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: 10/19/2024] [Revised: 11/11/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
This study aimed to investigate the effects of adding L. paracasei LK01 to the diet on the growth performance, antioxidant capacity, immunity, intestinal health, and serum biochemical indicators of broilers. This study selected 1080 one-day-old broiler chickens with similar body weight, and randomly divided them into six groups, with six replicates in each group and 30 chicks in each replicate. The chicks were fed (1) the basal diet (CON), (2) the basal diet with 106 CFU/kg L. paracasei LK01(T1), (3) the basal diet with 107 CFU/kg L. paracasei LK01(T2), (4) the basal diet with 108 CFU/kg L. paracasei LK01(T3), (5) the basal diet with 109 CFU/kg L. paracasei LK01(T4), and (6) the basal diet with 1010 CFU/kg L. paracasei LK01(T5). The experiment lasted for 42 days. In this study, compared with the CON group, the diet supplemented with L. paracasei significantly increased body weight from 1 to 21 days (p < 0.05). In addition, the 106 CFU/kg L. paracasei LK01 group significantly reduced the activity of glutamic oxaloacetic transaminase and triglyceride levels; the 107 CFU/kg,108 CFU/kg, and 109 CFU/kg L. paracasei LK01 groups also reduced serum uric acid and total cholesterol levels (p < 0.05). The experimental groups all had lower serum levels of malondialdehyde and interleukin-1β (p < 0.01). Except for the 106 CFU/kg group, all experimental groups had significantly lower tumor necrosis factor-α, and the 106 and 107 CFU/kg groups had higher immunoglobulin M levels (p < 0.05). In addition, the 106 CFU/kg group significantly reduced the depth of the ileocecal crypts and increased the villus-to-crypt ratio (V/C) of the jejunum and ileum. In addition, dietary supplementation with L. paracasei LK01 did not change the α diversity of the microbial community in the cecum, but significantly increased the proportion of Bacteroides (phylum) (p < 0.05). The 106 CFU/kg group also significantly increased the abundance of beneficial bacteria such as Ruminococcaceae (genus), Lachnospiraceae (genus), and Faecalibacterium (genus) (p < 0.05). In summary, this study revealed that adding 106 CFU/kg of L. paracasei LKO1 to broiler diets can improve their production performance, serum biochemical indicators, antioxidant, and immune capabilities, as well as cecal flora.
Collapse
Affiliation(s)
- Weixin Liu
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230031, China
| | - Hong Cheng
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230031, China
| | - Hao Zhang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230031, China
| | - Guozhen Liu
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230031, China
| | - Xinyu Yin
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230031, China
| | - Cheng Zhang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230031, China
| | - Runsheng Jiang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230031, China
| | - Zaigui Wang
- College of Life Science and Technology, Anhui Agricultural University, Hefei 230031, China
| | - Xiaoling Ding
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230031, China
| |
Collapse
|
25
|
Patloka O, Komprda T, Franke G. Review of the Relationships Between Human Gut Microbiome, Diet, and Obesity. Nutrients 2024; 16:3996. [PMID: 39683390 DOI: 10.3390/nu16233996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 11/15/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
Obesity is a complex disease that increases the risk of other pathologies. Its prevention and long-term weight loss maintenance are problematic. Gut microbiome is considered a potential obesity modulator. The objective of the present study was to summarize recent findings regarding the relationships between obesity, gut microbiota, and diet (vegetable/animal proteins, high-fat diets, restriction of carbohydrates), with an emphasis on dietary fiber and resistant starch. The composition of the human gut microbiome and the methods of its quantification are described. Products of the gut microbiome metabolism, such as short-chain fatty acids and secondary bile acids, and their effects on the gut microbiota, intestinal barrier function and immune homeostasis are discussed in the context of obesity. The importance of dietary fiber and resistant starch is emphasized as far as effects of the host diet on the composition and function of the gut microbiome are concerned. The complex relationships between human gut microbiome and obesity are finally summarized.
Collapse
Affiliation(s)
- Ondřej Patloka
- Department of Food Technology, Mendel University in Brno, 61300 Brno, Czech Republic
| | - Tomáš Komprda
- Department of Food Technology, Mendel University in Brno, 61300 Brno, Czech Republic
| | - Gabriela Franke
- Department of Food Technology, Mendel University in Brno, 61300 Brno, Czech Republic
| |
Collapse
|
26
|
Lee JY, Bays DJ, Savage HP, Bäumler AJ. The human gut microbiome in health and disease: time for a new chapter? Infect Immun 2024; 92:e0030224. [PMID: 39347570 PMCID: PMC11556149 DOI: 10.1128/iai.00302-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2024] Open
Abstract
The gut microbiome, composed of the colonic microbiota and their host environment, is important for many aspects of human health. A gut microbiome imbalance (gut dysbiosis) is associated with major causes of human morbidity and mortality. Despite the central part our gut microbiome plays in health and disease, mechanisms that maintain homeostasis and properties that demarcate dysbiosis remain largely undefined. Here we discuss that sorting taxa into meaningful ecological units reveals that the availability of respiratory electron acceptors, such as oxygen, in the host environment has a dominant influence on gut microbiome health. During homeostasis, host functions that limit the diffusion of oxygen into the colonic lumen shelter a microbial community dominated by primary fermenters from atmospheric oxygen. In turn, primary fermenters break down unabsorbed nutrients into fermentation products that support host nutrition. This symbiotic relationship is disrupted when host functions that limit the luminal availability of host-derived electron acceptors become weakened. The resulting changes in the host environment drive alterations in the microbiota composition, which feature an elevated abundance of facultatively anaerobic microbes. Thus, the part of the gut microbiome that becomes imbalanced during dysbiosis is the host environment, whereas changes in the microbiota composition are secondary to this underlying cause. This shift in our understanding of dysbiosis provides a novel starting point for therapeutic strategies to restore microbiome health. Such strategies can either target the microbes through metabolism-based editing or strengthen the host functions that control their environment.
Collapse
Affiliation(s)
- Jee-Yon Lee
- Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, Davis, California, USA
| | - Derek J. Bays
- Department of Internal Medicine, Division of Infectious Diseases, School of Medicine, University of California Davis, Sacramento, California, USA
| | - Hannah P. Savage
- Department of Pathology Microbiology and Immunology, School of Veterinary Medicine, University of California Davis, Davis, California, USA
| | - Andreas J. Bäumler
- Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, Davis, California, USA
| |
Collapse
|
27
|
Yilmaz G, Chan M, Lau CHF, Capitani S, Kang M, Charron P, Hoover E, Topp E, Guan J. How Gut Microbiome Perturbation Caused by Antibiotic Pre-Treatments Affected the Conjugative Transfer of Antimicrobial Resistance Genes. Microorganisms 2024; 12:2148. [PMID: 39597538 PMCID: PMC11596856 DOI: 10.3390/microorganisms12112148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 10/10/2024] [Accepted: 10/22/2024] [Indexed: 11/29/2024] Open
Abstract
The global spread of antimicrobial resistance genes (ARGs) poses a significant threat to public health. While antibiotics effectively treat bacterial infections, they can also induce gut dysbiosis, the severity of which varies depending on the specific antibiotic treatment used. However, it remains unclear how gut dysbiosis affects the mobility and dynamics of ARGs. To address this, mice were pre-treated with streptomycin, ampicillin, or sulfamethazine, and then orally inoculated with Salmonella enterica serovar Typhimurium and S. Heidelberg carrying a multi-drug resistance IncA/C plasmid. The streptomycin pre-treatment caused severe microbiome perturbation, promoting the high-density colonization of S. Heidelberg and S. Typhimurium, and enabling an IncA/C transfer from S. Heidelberg to S. Typhimurium and a commensal Escherichia coli. The ampicillin pre-treatment induced moderate microbiome perturbation, supporting only S. Heidelberg colonization and the IncA/C transfer to commensal E. coli. The sulfamethazine pre-treatment led to mild microbiome perturbation, favoring neither Salmonella spp. colonization nor a conjugative plasmid transfer. The degree of gut dysbiosis also influenced the enrichment or depletion of the ARGs associated with mobile plasmids or core commensal bacteria, respectively. These findings underscore the significance of pre-existing gut dysbiosis induced by various antibiotic treatments on ARG dissemination and may inform prudent antibiotic use practices.
Collapse
Affiliation(s)
- Gokhan Yilmaz
- Ottawa Laboratory-Fallowfield, Canadian Food Inspection Agency, Ottawa, ON K2J 4S1, Canada (M.K.); (E.H.)
| | - Maria Chan
- Ottawa Laboratory-Fallowfield, Canadian Food Inspection Agency, Ottawa, ON K2J 4S1, Canada (M.K.); (E.H.)
| | - Calvin Ho-Fung Lau
- Ottawa Laboratory-Carling, Canadian Food Inspection Agency, Ottawa, ON K1A 0Z, Canada; (C.H.-F.L.)
| | - Sabrina Capitani
- Ottawa Laboratory-Carling, Canadian Food Inspection Agency, Ottawa, ON K1A 0Z, Canada; (C.H.-F.L.)
| | - Mingsong Kang
- Ottawa Laboratory-Fallowfield, Canadian Food Inspection Agency, Ottawa, ON K2J 4S1, Canada (M.K.); (E.H.)
| | - Philippe Charron
- Ottawa Laboratory-Fallowfield, Canadian Food Inspection Agency, Ottawa, ON K2J 4S1, Canada (M.K.); (E.H.)
| | - Emily Hoover
- Ottawa Laboratory-Fallowfield, Canadian Food Inspection Agency, Ottawa, ON K2J 4S1, Canada (M.K.); (E.H.)
| | - Edward Topp
- Agroecology Research Unit, INRAE, University of Burgundy, 21065 Dijon, France;
| | - Jiewen Guan
- Ottawa Laboratory-Fallowfield, Canadian Food Inspection Agency, Ottawa, ON K2J 4S1, Canada (M.K.); (E.H.)
| |
Collapse
|
28
|
Russo MA, Puccetti M, Costantini C, Giovagnoli S, Ricci M, Garaci E, Romani L. Human and gut microbiota synergy in a metabolically active superorganism: a cardiovascular perspective. Front Cardiovasc Med 2024; 11:1411306. [PMID: 39465131 PMCID: PMC11502352 DOI: 10.3389/fcvm.2024.1411306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 09/26/2024] [Indexed: 10/29/2024] Open
Abstract
Despite significant advances in diagnosis and treatment over recent decades, cardiovascular disease (CVD) remains one of the leading causes of morbidity and mortality in Western countries. This persistent burden is partly due to the incomplete understanding of fundamental pathogenic mechanisms, which limits the effectiveness of current therapeutic interventions. In this context, recent evidence highlights the pivotal role of immuno-inflammatory activation by the gut microbiome in influencing cardiovascular disorders, potentially opening new therapeutic avenues. Indeed, while atherosclerosis has been established as a chronic inflammatory disease of the arterial wall, accumulating data suggest that immune system regulation and anti-inflammatory pathways mediated by gut microbiota metabolites play a crucial role in a range of CVDs, including heart failure, pericardial disease, arrhythmias, and cardiomyopathies. Of particular interest is the emerging understanding of how tryptophan metabolism-by both host and microbiota-converges on the Aryl hydrocarbon Receptor (AhR), a key regulator of immune homeostasis. This review seeks to enhance our understanding of the role of the immune system and inflammation in CVD, with a focus on how gut microbiome-derived tryptophan metabolites, such as indoles and their derivatives, contribute to cardioimmunopathology. By exploring these mechanisms, we aim to facilitate the development of novel, microbiome-centered strategies for combating CVD.
Collapse
Affiliation(s)
| | - Matteo Puccetti
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Claudio Costantini
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Stefano Giovagnoli
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Maurizio Ricci
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Enrico Garaci
- San Raffaele Research Center, Sulmona, L’Aquila, Italy
| | - Luigina Romani
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
- San Raffaele Research Center, Sulmona, L’Aquila, Italy
| |
Collapse
|
29
|
Lu P, Li D, Tian Q, Zhang J, Zhao Z, Wang H, Zhao H. Effect of mixed probiotics on pulmonary flora in patients with mechanical ventilation: an exploratory randomized intervention study. Eur J Med Res 2024; 29:473. [PMID: 39343939 PMCID: PMC11440949 DOI: 10.1186/s40001-024-02059-0] [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: 04/25/2023] [Accepted: 09/11/2024] [Indexed: 10/01/2024] Open
Abstract
OBJECTIVE The study objective was to investigate the effect of mixed probiotics on the diversity of the pulmonary flora in critically ill patients requiring mechanical ventilation by analysing the changes in lung microbes. METHODS 24 adult critically ill patients who needed mechanical ventilation in our hospital were randomly divided into a probiotic group and a control group. Then, the probiotic group was given Live Combined Bifidobacterium, Lactobacillus and Enterococcus Capsules, Oral (Bifico) by nasal feeding within 24 h after mechanical ventilation. Bronchoalveolar lavage fluid (BALF) and venous blood were collected within 24 h after mechanical ventilation and on the 5th day after mechanical ventilation, and the treatment status of patients (mechanical ventilation time, 28-day survival), measured cytokine levels (IL-1 β, IL-6, IL-8, IL-17A) and changes in pulmonary microorganisms were observed. RESULTS The microbial diversity of BALF samples decreased in the control group, and there was no significant difference in the probiotic group. Species difference analysis showed that among the three probiotics (Bifidobacterium, Lactobacillus, Enterococcus) used for intervention, Lactobacillus caused significant differences in BALF in the control group. Clinical factor association analysis displayed significant associations with IL-17A levels in both blood and BALF. CONCLUSION Mechanical ventilation can cause a decline in pulmonary microbial diversity, which can be improved by administering mixed probiotics.
Collapse
Affiliation(s)
- Peng Lu
- Department of Emergency Medicine, Hebei Medical University, Shijiazhuang, Hebei, China
- Department of Intensive Care Unit, Hebei General Hospital, Shijiazhuang, Hebei, China
- Department of Intensive Care Unit I, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Dongliang Li
- Department of Intensive Care Unit I, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Qing Tian
- Department of Chest Surgery, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jie Zhang
- Department of Intensive Care Unit I, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zhitao Zhao
- Department of Intensive Care Unit I, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Huawei Wang
- Department of Intensive Care Unit, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Heling Zhao
- Department of Intensive Care Unit, Hebei General Hospital, Shijiazhuang, Hebei, China.
| |
Collapse
|
30
|
Bian DD, Liu X, Zhang X, Zhang GY, Wu RC, Shi YX, Zhu XR, Zhang DZ, Liu QN, Tang BP, Zhu BJ. Correlative analysis of transcriptome and 16S rDNA in Procambarus clarkii reveals key signaling pathways are involved in Chlorantraniliprole stress response by phosphoinositide 3-kinase (PI3K). Int J Biol Macromol 2024; 280:135966. [PMID: 39326603 DOI: 10.1016/j.ijbiomac.2024.135966] [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: 07/05/2024] [Revised: 09/02/2024] [Accepted: 09/21/2024] [Indexed: 09/28/2024]
Abstract
Chlorantraniliprole (CAP), a diamide insecticide, is extensively used in agricultural production. With the increasing adoption of the rice-crayfish integrated farming model, pesticide application has become more frequent. However, the potential risk of CAP to crayfish (Procambarus clarkii) remains unclear. In this study, crayfish were exposed to 30, 60, 90 mg/L CAP for 96 h. As CAP exposure time and concentration increased, crayfish survival rates and total hemocyte counts (THC) decreased. Biochemical indicators revealed that CAP exposure induced oxidative stress and immunosuppression in crayfish, leading to metabolic disorders and reduced ATP content. Additionally, pathological analysis and 16S rDNA sequencing demonstrated that CAP exposure compromised the intestinal barrier of crayfish, altered the intestinal microbial community structure, and caused apoptosis. Differential gene expression analysis showed that CAP exposure significantly suppressed the expression of genes related to immune and energy metabolism pathways, resulting in immune dysfunction and insufficient energy supply, while activating the PI3K/AKT/mTOR signaling pathway. PI3K knockdown reduced antioxidant and digestive activities, increased the expression of proinflammatory and apoptosis genes, and exacerbated CAP-induced intestinal toxicity. This study is the first to explore the characterization and function of PI3K in crustaceans, providing new insights for further research on crustacean antioxidants and defense mechanisms.
Collapse
Affiliation(s)
- Dan-Dan Bian
- Anhui Key Laboratory of Resource Insect Biology and Innovative Utilization, College of Life Sciences, Anhui Agricultural University, Hefei 230036, People's Republic of China; Jiangsu Key Laboratory for Bioresources of Saline Soils, Jiangsu Synthetic Innovation Center for Coastal Bio-agriculture, Jiangsu Provincial Key Laboratory of Coastal Wetland Bioresources and Environmental Protection, School of Wetlands, Yancheng Teachers University, Yancheng 224007, People's Republic of China
| | - Xin Liu
- Jiangsu Key Laboratory for Bioresources of Saline Soils, Jiangsu Synthetic Innovation Center for Coastal Bio-agriculture, Jiangsu Provincial Key Laboratory of Coastal Wetland Bioresources and Environmental Protection, School of Wetlands, Yancheng Teachers University, Yancheng 224007, People's Republic of China; College of Fisheries and Life Science, Shanghai Ocean University, Shanghai 201306, People's Republic of China
| | - Xue Zhang
- Jiangsu Key Laboratory for Bioresources of Saline Soils, Jiangsu Synthetic Innovation Center for Coastal Bio-agriculture, Jiangsu Provincial Key Laboratory of Coastal Wetland Bioresources and Environmental Protection, School of Wetlands, Yancheng Teachers University, Yancheng 224007, People's Republic of China; College of Fisheries and Life Science, Shanghai Ocean University, Shanghai 201306, People's Republic of China
| | - Geng-Yu Zhang
- Anhui Key Laboratory of Resource Insect Biology and Innovative Utilization, College of Life Sciences, Anhui Agricultural University, Hefei 230036, People's Republic of China; Jiangsu Key Laboratory for Bioresources of Saline Soils, Jiangsu Synthetic Innovation Center for Coastal Bio-agriculture, Jiangsu Provincial Key Laboratory of Coastal Wetland Bioresources and Environmental Protection, School of Wetlands, Yancheng Teachers University, Yancheng 224007, People's Republic of China
| | - Ren-Chao Wu
- Anhui Key Laboratory of Resource Insect Biology and Innovative Utilization, College of Life Sciences, Anhui Agricultural University, Hefei 230036, People's Republic of China; Jiangsu Key Laboratory for Bioresources of Saline Soils, Jiangsu Synthetic Innovation Center for Coastal Bio-agriculture, Jiangsu Provincial Key Laboratory of Coastal Wetland Bioresources and Environmental Protection, School of Wetlands, Yancheng Teachers University, Yancheng 224007, People's Republic of China
| | - Yan-Xia Shi
- Jiangsu Key Laboratory for Bioresources of Saline Soils, Jiangsu Synthetic Innovation Center for Coastal Bio-agriculture, Jiangsu Provincial Key Laboratory of Coastal Wetland Bioresources and Environmental Protection, School of Wetlands, Yancheng Teachers University, Yancheng 224007, People's Republic of China; College of Biotechnology and Pharmaceutical Engineering, Nanjing University of Technology, Nanjing 210009, People's Republic of China
| | - Xi-Rong Zhu
- Jiangsu Key Laboratory for Bioresources of Saline Soils, Jiangsu Synthetic Innovation Center for Coastal Bio-agriculture, Jiangsu Provincial Key Laboratory of Coastal Wetland Bioresources and Environmental Protection, School of Wetlands, Yancheng Teachers University, Yancheng 224007, People's Republic of China; College of Biotechnology and Pharmaceutical Engineering, Nanjing University of Technology, Nanjing 210009, People's Republic of China
| | - Dai-Zhen Zhang
- Jiangsu Key Laboratory for Bioresources of Saline Soils, Jiangsu Synthetic Innovation Center for Coastal Bio-agriculture, Jiangsu Provincial Key Laboratory of Coastal Wetland Bioresources and Environmental Protection, School of Wetlands, Yancheng Teachers University, Yancheng 224007, People's Republic of China
| | - Qiu-Ning Liu
- Jiangsu Key Laboratory for Bioresources of Saline Soils, Jiangsu Synthetic Innovation Center for Coastal Bio-agriculture, Jiangsu Provincial Key Laboratory of Coastal Wetland Bioresources and Environmental Protection, School of Wetlands, Yancheng Teachers University, Yancheng 224007, People's Republic of China.
| | - Bo-Ping Tang
- Jiangsu Key Laboratory for Bioresources of Saline Soils, Jiangsu Synthetic Innovation Center for Coastal Bio-agriculture, Jiangsu Provincial Key Laboratory of Coastal Wetland Bioresources and Environmental Protection, School of Wetlands, Yancheng Teachers University, Yancheng 224007, People's Republic of China.
| | - Bao-Jian Zhu
- Anhui Key Laboratory of Resource Insect Biology and Innovative Utilization, College of Life Sciences, Anhui Agricultural University, Hefei 230036, People's Republic of China.
| |
Collapse
|
31
|
Ci Y, Ku T, Su Y, He Z, Zhang Y, Ji J, Ning X, Yin S, Zhang K. Response signatures of intestinal microbiota and gene transcription of yellow catfish (Pelteobagrus fulvidraco) to Aeromonas hydrophila infection. FISH & SHELLFISH IMMUNOLOGY 2024; 152:109797. [PMID: 39084276 DOI: 10.1016/j.fsi.2024.109797] [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: 04/29/2024] [Revised: 07/15/2024] [Accepted: 07/28/2024] [Indexed: 08/02/2024]
Abstract
Bacterial intestinal inflammation is a common disease of yellow catfish (Pelteobagrus fulvidraco) in high-density aquaculture. Understanding the interactions between host and intestinal bacteria is helpful to intestinal inflammatory disease control. Here, we constructed a model of intestinal inflammation after Aeromonas hydrophila infection in yellow catfish, and characterized variations in gene expression and microbiome in the gut through high-throughput sequencing. Furthermore, host gene-microbiome interactions were identified. Histology observation showed disordered distribution of columnar epithelial cells and decrease of goblet cells in intestine. A total of 4741 genes showed differentially expression, mostly in comparisons between 12 hpi group with each other groups respectively, including control, 24 hpi and 48 hpi groups. These genes were enriched in immune-related pathways including the IL-17 signaling pathway, triggering strong inflammatory response at the invading stage within 12 h. Subsequently, the host strengthened energy consumption by activating carbohydrate and lipid metabolism pathways to repair the intestinal mucosal immune defense line. In addition, fish with A. hydrophila infection show decreased richness of gut microbial, reduced relative abundance of probiotics including Akkermansia, and elevated pathogenic bacteria such as Plesimonas. An integrative analysis identified A. hydrophila-related genes, such as il22 and stat3, for which expression level is close associated with the shift of A. hydrophila-related bacteria relative abundance, such as Akkermansia and Cetobacterium. Aside from picturing the variations of intestine gene expression and mucosal microbiome of yellow catfish coping with A. hydrophila infection, our study probed the underlying host-microbe interactions in A. hydrophila infection induced intestinal inflammatory, providing new insights for disease control in aquaculture.
Collapse
Affiliation(s)
- Yuting Ci
- College of Marine Science and Engineering, Nanjing Normal University, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing, 210023, China
| | - Tinglan Ku
- College of Marine Science and Engineering, Nanjing Normal University, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing, 210023, China
| | - Yiting Su
- College of Marine Science and Engineering, Nanjing Normal University, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing, 210023, China
| | - Zhimin He
- College of Marine Science and Engineering, Nanjing Normal University, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing, 210023, China
| | - Yufei Zhang
- College of Marine Science and Engineering, Nanjing Normal University, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing, 210023, China
| | - Jie Ji
- College of Marine Science and Engineering, Nanjing Normal University, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing, 210023, China; Co-Innovation Center for Marine Bio-Industry Technology, Lian Yungang, Jiangsu, 222005, China
| | - Xianhui Ning
- College of Marine Science and Engineering, Nanjing Normal University, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing, 210023, China; Co-Innovation Center for Marine Bio-Industry Technology, Lian Yungang, Jiangsu, 222005, China
| | - Shaowu Yin
- College of Marine Science and Engineering, Nanjing Normal University, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing, 210023, China; Co-Innovation Center for Marine Bio-Industry Technology, Lian Yungang, Jiangsu, 222005, China
| | - Kai Zhang
- College of Marine Science and Engineering, Nanjing Normal University, Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, Nanjing, 210023, China; Co-Innovation Center for Marine Bio-Industry Technology, Lian Yungang, Jiangsu, 222005, China.
| |
Collapse
|
32
|
Hu M, Du Y, Li W, Zong X, Du W, Sun H, Liu H, Zhao K, Li J, Farooq MZ, Wu J, Xu Q. Interplay of Food-Derived Bioactive Peptides with Gut Microbiota: Implications for Health and Disease Management. Mol Nutr Food Res 2024; 68:e2400251. [PMID: 39097954 DOI: 10.1002/mnfr.202400251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 06/19/2024] [Indexed: 08/06/2024]
Abstract
Bioactive peptides (BPs) are protein fragments with beneficial effects on metabolism, physiology, and diseases. This review focuses on proteolytic BPs, which are produced by the action of gut microbiota on proteins in food and have demonstrated to influence the composition of gut microbes. And gut microbiota are candidate targets of BPs to alleviate oxidative stress, enhance immunity, and control diseases, including diabetes, hypertension, obesity, cancer, and immune and neurodegenerative diseases. Despite promising results, further research is needed to understand the mechanisms underlying the interactions between BPs and gut microbes, and to identify and screen more BPs for industrial applications. Overall, BPs offer potential as therapeutic agents for various diseases through their interactions with gut microbes, highlighting the importance of continued research in this area.
Collapse
Affiliation(s)
- Mingyang Hu
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yufeng Du
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Wenyue Li
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Xiaomei Zong
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Wenjuan Du
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Huizeng Sun
- Institute of Dairy Science, MoE Key Laboratory of Molecular Animal Nutrition, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Hongyun Liu
- Institute of Dairy Science, MoE Key Laboratory of Molecular Animal Nutrition, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Ke Zhao
- Institute of Food Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310058, China
| | - Jianxiong Li
- Wuhan Jason Biotech Co., Ltd., Wuhan, 430070, China
| | - Muhammad Zahid Farooq
- Department of Animal Science, University of Veterinary and Animal Science, Lahore, 54000, Pakistan
| | - Jianping Wu
- Department of Agricultural, Food and Nutritional Science, University of Alberta, 4-10 Ag/For Building, Edmonton, Alberta, T6G 2P5, Canada
| | - Qingbiao Xu
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| |
Collapse
|
33
|
Qian Z, Hou D, Gao S, Wang X, Yu J, Dong J, Sun C. Toxic effects and mechanisms of chronic cadmium exposure on Litopenaeus vannamei growth performance based on combined microbiome and metabolome analysis. CHEMOSPHERE 2024; 361:142578. [PMID: 38857631 DOI: 10.1016/j.chemosphere.2024.142578] [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: 03/20/2024] [Revised: 05/28/2024] [Accepted: 06/08/2024] [Indexed: 06/12/2024]
Abstract
Cadmium (Cd) pollution seriously affects marine organisms' health and poses a threat to food safety. Although Cd pollution has attracted widespread attention in aquaculture, little is known about the toxic mechanisms of chronic Cd exposure on shrimp growth performance. The study investigated the combined effects of chronic exposure to Cd of different concentrations including 0, 75, 150, and 300 μg/L for 30 days on the growth performance, tissue bioaccumulation, intestinal microbiology, and metabolic responses of Litopenaeus vannamei. The results revealed that the growth was significantly inhibited under exposure to 150 and 300 μg/L Cd2+. The bioaccumulation in gills and intestines respectively showed an increasing and inverted "U" shaped trend with increasing Cd2+ concentration. Chronic Cd altered the intestinal microflora with a significant decrease in microbial richness and increasing trends in the abundances of the potentially pathogenic bacteria Vibrio and Maribacter at exposure to 75 and 150 μg/L Cd2+, and Maribacter at 300 μg/L. In addition, chronic Cd interfered with intestinal metabolic processes. The expressions of certain metabolites associated with growth promotion and enhanced antioxidant power, including N-methyl-D-aspartic acid, L-malic acid, guanidoacetic acid, betaine, and gluconic acid were significantly down-regulated, especially at exposure to 150 and 300 μg/L Cd2+, and were negatively correlated with Vibrio and Maribacter abundance levels. In summary, chronic Cd exposure resulted in severe growth inhibition and increased Cd accumulation in shrimp tissues. Increased levels of intestinal pathogenic bacteria and decreased levels of growth-promoting metabolites may be the key causes of growth inhibition. Harmful bacteria Vibrio and Maribacter may be associated with the inhibition of growth-promoting metabolite expression and may be involved in disrupting intestinal metabolic functions, ultimately impairing shrimp growth potential. This study sheds light on the potential toxicological mechanisms of chronic Cd inhibition on shrimp growth performance, offering new insights into Cd toxicity studies in aquaculture.
Collapse
Affiliation(s)
- Zhaoying Qian
- School of Economics, Guizhou University of Finance and Economics, Guiyang, 550025, Guizhou, China
| | - Danqing Hou
- College of Fisheries, Guangdong Ocean University, Zhanjiang, 524000, Guangdong, China
| | - Shan Gao
- College of Fisheries, Guangdong Ocean University, Zhanjiang, 524000, Guangdong, China
| | - Xuejie Wang
- College of Fisheries, Guangdong Ocean University, Zhanjiang, 524000, Guangdong, China
| | - Jianbo Yu
- College of Fisheries, Guangdong Ocean University, Zhanjiang, 524000, Guangdong, China
| | - Jiaxin Dong
- College of Fisheries, Guangdong Ocean University, Zhanjiang, 524000, Guangdong, China
| | - Chengbo Sun
- College of Fisheries, Guangdong Ocean University, Zhanjiang, 524000, Guangdong, China.
| |
Collapse
|
34
|
Rajamanickam A, Babu S. Helminth Infections and Diabetes: Mechanisms Accounting for Risk Amelioration. Annu Rev Nutr 2024; 44:339-355. [PMID: 38724017 DOI: 10.1146/annurev-nutr-061121-100742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2024]
Abstract
The global prevalence of type 2 diabetes mellitus (T2D) is increasing rapidly, with an anticipated 600 million cases by 2035. While infectious diseases such as helminth infections have decreased due to improved sanitation and health care, recent research suggests a link between helminth infections and T2D, with helminths such as Schistosoma, Nippostrongylus, Strongyloides, and Heligmosomoides potentially mitigating or slowing down T2D progression in human and animal models. Helminth infections enhance host immunity by promoting interactions between innate and adaptive immune systems. In T2D, type 1 immune responses are suppressed and type 2 responses are augmented, expanding regulatory T cells and innate immune cells, particularly type 2 immune cells and macrophages. This article reviews recent research shedding light on the favorable effects of helminth infections on T2D. The potential defense mechanisms identified include heightened insulin sensitivity and reduced inflammation. The synthesis of findings from studies investigating parasitic helminths and their derivatives underscores promising avenues for defense against T2D.
Collapse
Affiliation(s)
- Anuradha Rajamanickam
- National Institutes of Health-National Institute of Allergy and Infectious Diseases International Center for Excellence in Research, Chennai, India;
| | - Subash Babu
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
- National Institutes of Health-National Institute of Allergy and Infectious Diseases International Center for Excellence in Research, Chennai, India;
| |
Collapse
|
35
|
Zhang Y, Wan Y, Xin X, Qiao Y, Qiao W, Ping J, Su J. Signals from intestinal microbiota mediate the crosstalk between the lung-gut axis in an influenza infection mouse model. Front Immunol 2024; 15:1435180. [PMID: 39114658 PMCID: PMC11304505 DOI: 10.3389/fimmu.2024.1435180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 06/25/2024] [Indexed: 08/10/2024] Open
Abstract
Introduction Introduction: The influenza virus primarily targets the respiratory tract, yet both the respiratory and intestinal systems suffer damage during infection. The connection between lung and intestinal damage remains unclear. Methods Our experiment employs 16S rRNA technology and Liquid Chromatography-Mass Spectrometry (LC-MS) to detect the impact of influenza virus infection on the fecal content and metabolites in mice. Additionally, it investigates the effect of influenza virus infection on intestinal damage and its underlying mechanisms through HE staining, Western blot, Q-PCR, and flow cytometry. Results Our study found that influenza virus infection caused significant damage to both the lungs and intestines, with the virus detected exclusively in the lungs. Antibiotic treatment worsened the severity of lung and intestinal damage. Moreover, mRNA levels of Toll-like receptor 7 (TLR7) and Interferon-b (IFN-b) significantly increased in the lungs post-infection. Analysis of intestinal microbiota revealed notable shifts in composition after influenza infection, including increased Enterobacteriaceae and decreased Lactobacillaceae. Conversely, antibiotic treatment reduced microbial diversity, notably affecting Firmicutes, Proteobacteria, and Bacteroidetes. Metabolomics showed altered amino acid metabolism pathways due to influenza infection and antibiotics. Abnormal expression of indoleamine 2,3-dioxygenase 1 (IDO1) in the colon disrupted the balance between helper T17 cells (Th17) and regulatory T cells (Treg cells) in the intestine. Mice infected with the influenza virus and supplemented with tryptophan and Lactobacillus showed reduced lung and intestinal damage, decreased Enterobacteriaceae levels in the intestine, and decreased IDO1 activity. Discussion Overall, influenza infection caused damage to lung and intestinal tissues, disrupted intestinal microbiota and metabolites, and affected Th17/Treg balance. Antibiotic treatment exacerbated these effects. Supplementation with tryptophan and Lactobacillus improved lung and intestinal health, highlighting a new understanding of the lung-intestine connection in influenza-induced intestinal disease.
Collapse
Affiliation(s)
- Yijia Zhang
- Laboratory of Animal Neurobiology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Youdi Wan
- Laboratory of Animal Neurobiology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xin Xin
- Laboratory of Animal Neurobiology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yixuan Qiao
- Laboratory of Animal Neurobiology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Wenna Qiao
- Laboratory of Animal Neurobiology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Jihui Ping
- Ministry of Education (MOE) Joint International Research Laboratory of Animal Health and Food Safety, Engineering Laboratory of Animal Immunity of Jiangsu Province, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Juan Su
- Laboratory of Animal Neurobiology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
36
|
Buchanan CE, Galla SJ, Muscarella ME, Forbey JS, Reinking AK, Beck JL. Relating gut microbiome composition and life history metrics for pronghorn (Antilocapra americana) in the Red Desert, Wyoming. PLoS One 2024; 19:e0306722. [PMID: 38985706 PMCID: PMC11236126 DOI: 10.1371/journal.pone.0306722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 06/21/2024] [Indexed: 07/12/2024] Open
Abstract
Host microbial communities (hereafter, the 'microbiome') are recognized as an important aspect of host health and are gaining attention as a useful biomarker to understand the ecology and demographics of wildlife populations. Several studies indicate that the microbiome may contribute to the adaptive capacity of animals to changing environments associated with increasing habitat fragmentation and rapid climate change. To this end, we investigated the gut microbiome of pronghorn (Antilocapra americana), an iconic species in an environment that is undergoing both climatic and anthropogenic change. The bacterial composition of the pronghorn gut microbiome has yet to be described in the literature, and thus our study provides important baseline information about this species. We used 16S rRNA amplicon sequencing of fecal samples to characterize the gut microbiome of pronghorn-a facultative sagebrush (Artemisia spp.) specialist in many regions where they occur in western North America. We collected fecal pellets from 159 captured female pronghorn from four herds in the Red Desert of Wyoming during winters of 2013 and 2014. We found small, but significant differences in diversity of the gut microbiome relative to study area, capture period, and body fat measurements. In addition, we found a difference in gut microbiome composition in pronghorn across two regions separated by Interstate 80. Results indicated that the fecal microbiome may be a potential biomarker for the spatial ecology of free-ranging ungulates. The core gut microbiome of these animals-including bacteria in the phyla Firmicutes (now Bacillota) and Bacteroidota-remained relatively stable across populations and biological metrics. These findings provide a baseline for the gut microbiome of pronghorn that could potentially be used as a target in monitoring health and population structure of pronghorn relative to habitat fragmentation, climate change, and management practices.
Collapse
Affiliation(s)
- Courtney E Buchanan
- Department of Ecosystem Science and Management, University of Wyoming, Laramie, Wyoming, United States of America
| | - Stephanie J Galla
- Department of Biological Sciences, Boise State University, Boise, Idaho, United States of America
| | - Mario E Muscarella
- Institute of Arctic Biology and Department of Biology & Wildlife, University of Alaska Fairbanks, Fairbanks, Alaska, United States of America
| | - Jennifer S Forbey
- Department of Biological Sciences, Boise State University, Boise, Idaho, United States of America
| | - Adele K Reinking
- Department of Ecosystem Science and Management, University of Wyoming, Laramie, Wyoming, United States of America
- Cooperative Institute for Research in the Atmosphere, Colorado State University, Fort Collins, Colorado, United States of America
- Graduate Degree Program in Ecology, Colorado State University, Fort Collins, Colorado, United States of America
- Department of Fish, Wildlife, and Conservation Biology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Jeffrey L Beck
- Department of Ecosystem Science and Management, University of Wyoming, Laramie, Wyoming, United States of America
| |
Collapse
|
37
|
Yuan J, Yu Y, Li S, Zhang X, Zhang C, Li R, Hu J, Si S, Zhang C, Xiang J, Li F. Shrimp shapes a resistance trait against vibriosis by memorizing the colonization resistance of intestinal microbiota. PLoS Pathog 2024; 20:e1012321. [PMID: 38990823 PMCID: PMC11239079 DOI: 10.1371/journal.ppat.1012321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 06/06/2024] [Indexed: 07/13/2024] Open
Abstract
Vibriosis is one of the most serious diseases that commonly occurs in aquatic animals, thus, shaping a steady inherited resistance trait in organisms has received the highest priority in aquaculture. Whereas, the mechanisms underlying the development of such a resistance trait are mostly elusive. In this study, we constructed vibriosis-resistant and susceptible families of the Pacific white shrimp Litopenaeus vannamei after four generations of artificial selection. Microbiome sequencing indicated that shrimp can successfully develop a colonization resistance trait against Vibrio infections. This trait was characterized by a microbial community structure with specific enrichment of a single probiotic species (namely Shewanella algae), and notably, its formation was inheritable and might be memorized by host epigenetic remodeling. Regardless of the infection status, a group of genes was specifically activated in the resistant family through disruption of complete methylation. Specifically, hypo-methylation and hyper-expression of genes related to lactate dehydrogenase (LDH) and iron homeostasis might provide rich sources of specific carbon (lactate) and ions for the colonization of S. algae, which directly results in the reduction of Vibrio load in shrimp. Lactate feeding increased the survival of shrimp, while knockdown of LDH gene decreased the survival when shrimp was infected by Vibrio pathogens. In addition, treatment of shrimp with the methyltransferase inhibitor 5-azacytidine resulted in upregulations of LDH and some protein processing genes, significant enrichment of S. algae, and simultaneous reduction of Vibrio in shrimp. Our results suggest that the colonization resistance can be memorized as epigenetic information by the host, which has played a pivotal role in vibriosis resistance. The findings of this study will aid in disease control and the selection of superior lines of shrimp with high disease resistance.
Collapse
Affiliation(s)
- Jianbo Yuan
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Chinese Academy of Sciences, Wuhan, China
| | - Yang Yu
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Chinese Academy of Sciences, Wuhan, China
| | - Shihao Li
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Chinese Academy of Sciences, Wuhan, China
| | - Xiaojun Zhang
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Chinese Academy of Sciences, Wuhan, China
| | - Chuntao Zhang
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Roujing Li
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
| | - Jie Hu
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
| | - Shuqing Si
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Chengyi Zhang
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jianhai Xiang
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Chinese Academy of Sciences, Wuhan, China
| | - Fuhua Li
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Chinese Academy of Sciences, Wuhan, China
| |
Collapse
|
38
|
Ghani MU, Yang Z, Feng T, Chen J, Khosravi Z, Wu Q, Cui H. Comprehensive review on glucose 6 phosphate dehydrogenase: A critical immunometabolic and redox switch in insects. Int J Biol Macromol 2024; 273:132867. [PMID: 38838892 DOI: 10.1016/j.ijbiomac.2024.132867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/14/2024] [Accepted: 06/01/2024] [Indexed: 06/07/2024]
Abstract
Mounting an active immune response is energy intensive and demands the reallocation of nutrients to maintain the body's resistance and tolerance against infections. Central to this metabolic adaptation is Glucose-6-phosphate dehydrogenase (G6PDH), a housekeeping enzyme involve in pentose phosphate pathway (PPP). PPP play an essential role in generating ribose, which is critical for nicotinamide adenine dinucleotide phosphate (NADPH). It is vital for physiological and cellular processes such as generating nucleotides, fatty acids and reducing oxidative stress. The G6PDH is extremely conserved enzyme across species in PP shunt. The deficiency of enzymes leads to serious consequences on organism, particularly on adaptation and development. Acute deficiency can lead to impaired cell development, halted embryonic growth, reduce sensitivity to insulin, hypertension and increase inflammation. Historically, research focusing on G6PDH and PPP have primarily targeted diseases on mammalian. However, our review has investigated the unique functions of the G6PDH enzyme in insects and greatly improved mechanistic understanding of its operations. This review explore how G6PDH in insects plays a crucial role in managing the redox balance and immune related metabolism. This study aims to investigate the enzyme's role in different metabolic adaptations.
Collapse
Affiliation(s)
- Muhammad Usman Ghani
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, China; Medical Research Institute, Southwest University, Chongqing 400715, China
| | - Zihan Yang
- Medical Research Institute, Southwest University, Chongqing 400715, China
| | - Tianxiang Feng
- Medical Research Institute, Southwest University, Chongqing 400715, China
| | - Junfan Chen
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, China
| | - Zahra Khosravi
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, China
| | - Qishu Wu
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, China
| | - Hongjuan Cui
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, China; Medical Research Institute, Southwest University, Chongqing 400715, China; Jinfeng Laboratory, Chongqing, 401329, China.
| |
Collapse
|
39
|
Shishani R, Wang A, Lyo V, Nandakumar R, Cummings BP. Vertical Sleeve Gastrectomy Reduces Gut Luminal Deoxycholic Acid Concentrations in Mice. Obes Surg 2024; 34:2483-2491. [PMID: 38777944 PMCID: PMC11217124 DOI: 10.1007/s11695-024-07288-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 05/13/2024] [Accepted: 05/13/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND Bariatric surgery alters bile acid metabolism, which contributes to post-operative improvements in metabolic health. However, the mechanisms by which bariatric surgery alters bile acid metabolism are incompletely defined. In particular, the role of the gut microbiome in the effects of bariatric surgery on bile acid metabolism is incompletely understood. Therefore, we sought to define the changes in gut luminal bile acid composition after vertical sleeve gastrectomy (VSG). METHODS Bile acid profile was determined by UPLC-MS/MS in serum and gut luminal samples from VSG and sham-operated mice. Sham-operated mice were divided into two groups: one was fed ad libitum, while the other was food-restricted to match their body weight to the VSG-operated mice. RESULTS VSG decreased gut luminal secondary bile acids, which was driven by a decrease in gut luminal deoxycholic acid concentrations and abundance. However, gut luminal cholic acid (precursor for deoxycholic acid) concentration and abundance did not differ between groups. Therefore, the observed decrease in gut luminal deoxycholic acid abundance after VSG was not due to a reduction in substrate availability. CONCLUSION VSG decreased gut luminal deoxycholic acid abundance independently of body weight, which may be driven by a decrease in gut bacterial bile acid metabolism.
Collapse
Affiliation(s)
- Rahaf Shishani
- Department of Surgery, Division of Foregut, Metabolic, and General Surgery, Center for Alimentary and Metabolic Sciences, School of Medicine, University of California - Davis, Sacramento, CA, 95817, USA
- Department of Molecular Biosciences, School of Veterinary Medicine, University of CA - Davis, Davis, CA, 95616, USA
| | - Annie Wang
- Department of Surgery, Division of Foregut, Metabolic, and General Surgery, Center for Alimentary and Metabolic Sciences, School of Medicine, University of California - Davis, Sacramento, CA, 95817, USA
| | - Victoria Lyo
- Department of Surgery, Division of Foregut, Metabolic, and General Surgery, Center for Alimentary and Metabolic Sciences, School of Medicine, University of California - Davis, Sacramento, CA, 95817, USA
| | - Renu Nandakumar
- Biomarkers Core Laboratory, Irving Institute for Clinical and Translational Research, Columbia University Irving Medical Center, Columbia University, New York, NY, 10032, USA
| | - Bethany P Cummings
- Department of Surgery, Division of Foregut, Metabolic, and General Surgery, Center for Alimentary and Metabolic Sciences, School of Medicine, University of California - Davis, Sacramento, CA, 95817, USA.
- Department of Molecular Biosciences, School of Veterinary Medicine, University of CA - Davis, Davis, CA, 95616, USA.
| |
Collapse
|
40
|
Onyango MG, Payne AF, Stout J, Dieme C, Kuo L, Kramer LD, Ciota AT. Aedes albopictus saliva contains a richer microbial community than the midgut. Parasit Vectors 2024; 17:267. [PMID: 38918848 PMCID: PMC11197185 DOI: 10.1186/s13071-024-06334-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 05/28/2024] [Indexed: 06/27/2024] Open
Abstract
BACKGROUND Past findings demonstrate that arthropods can egest midgut microbiota into the host skin leading to dual colonization of the vertebrate host with pathogens and saliva microbiome. A knowledge gap exists on how the saliva microbiome interacts with the pathogen in the saliva. To fill this gap, we need to first define the microbial composition of mosquito saliva. METHODS The current study aimed at analyzing and comparing the microbial profile of Aedes albopictus saliva and midgut as well as assessing the impact of Zika virus (ZIKV) infection on the midgut and saliva microbial composition. Colony-reared Ae. albopictus strains were either exposed to ZIKV infectious or noninfectious bloodmeal. At 14 ays postinfection, the 16S V3-V4 hypervariable rRNA region was amplified from midgut and saliva samples and sequenced on an Illumina MiSeq platform. The relative abundance and diversity of midgut and saliva microbial taxa were assessed. RESULTS We observed a richer microbial community in the saliva compared with the midgut, yet some of the microbial taxa were common in the midgut and saliva. ZIKV infection did not impact the microbial diversity of midgut or saliva. Further, we identified Elizabethkingia spp. in the Ae. albopictus saliva. CONCLUSIONS This study provides insights into the microbial community of the Ae. albopictus saliva as well as the influence of ZIKV infection on the microbial composition of its midgut and saliva. The identification of Elizabethkingia spp., an emerging pathogen of global health significance, in Ae. albopictus saliva is of medical importance. Future studies to assess the interactions between Ae. albopictus saliva microbiome and ZIKV could lead to novel strategies for developing transmission barrier tools.
Collapse
Affiliation(s)
- Maria G Onyango
- Department of Biological Sciences, Texas Tech University, 2901 Main St, Lubbock, Texas, 79409-3131, USA.
| | - Anne F Payne
- New York State Department of Health, Wadsworth Center, 5668 State Farm Road, Slingerlands, NY, 12159, USA
| | - Jessica Stout
- New York State Department of Health, Wadsworth Center, 5668 State Farm Road, Slingerlands, NY, 12159, USA
| | - Constentin Dieme
- New York State Department of Health, Wadsworth Center, 5668 State Farm Road, Slingerlands, NY, 12159, USA
| | - Lili Kuo
- New York State Department of Health, Wadsworth Center, 5668 State Farm Road, Slingerlands, NY, 12159, USA
| | - Laura D Kramer
- School of Public Health, State University of New York Albany, 1400 Washington Avenue, Albany, NY, 12222, USA
| | - Alexander T Ciota
- New York State Department of Health, Wadsworth Center, 5668 State Farm Road, Slingerlands, NY, 12159, USA
- School of Public Health, State University of New York Albany, 1400 Washington Avenue, Albany, NY, 12222, USA
| |
Collapse
|
41
|
Li H, Dai J, Zhao C, Hu T, Zhao G, Wang Q, Zhang L. Gut Subdoligranulum variabile ameliorates rheumatoid arthritis by promoting TSG-6 synthesis from joint cells. Front Immunol 2024; 15:1418717. [PMID: 38979426 PMCID: PMC11229780 DOI: 10.3389/fimmu.2024.1418717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 05/27/2024] [Indexed: 07/10/2024] Open
Abstract
Background A burgeoning body of evidence has substantiated the association between alterations in the composition of the gut microbiota and rheumatoid arthritis (RA). Nevertheless, our understanding of the intricate mechanisms underpinning this association is limited. Methods To investigate whether the gut microbiota influences the pathogenesis of RA through metabolism or immunity, we performed rigorous synthesis analyses using aggregated statistics from published genome-wide association studies (GWAS) using two-sample Mendelian randomization (MR) and mediated MR techniques, including two-step MR and multivariate MR analyses. Subsequently, we conducted in vitro cellular validation of the analyzed Microbial-Cytokine-RA pathway. We determined the optimal culture conditions through co-culture experiments involving concentration and time. Cell Counting Kit-8 (CCK-8) assays were employed to assess cellular viability, and enzyme-linked immunosorbent assays (ELISA) were performed to assess tumor necrosis factor-inducible gene 6 protein (TSG-6) and tumor necrosis factor-α (TNF-α) levels. Results Our univariable MR results confirmed 15 microbial traits, 7 metabolites and 2 cytokines that may be causally associated with RA (P FDR < 0.05). Mediation analysis revealed that microbial traits influence the risk of RA through metabolite or cytokine (proportion mediated: 7.75% - 58.22%). In vitro experiments demonstrated that TSG-6 was highly expressed in the Subdoligranulum variabile treatment group and was correlated with decreased RA severity (reduced TNF-α expression). Silencing the TSG-6 gene significantly increased TNF-α expression, regardless of treatment with S. variabile. Additionally, S. variabile-secreted exosomes exhibited the same effect. Conclusion The results of this study suggest that S. variabile has the potential to promote TSG-6 secretion, thereby reducing RA inflammation.
Collapse
Affiliation(s)
- Hongfeng Li
- Microbiome-X, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Health Inspection and Quarantine, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Junhui Dai
- Microbiome-X, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Changying Zhao
- Microbiome-X, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Tianqi Hu
- Microbiome-X, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Guoping Zhao
- Microbiome-X, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
- Chinese Academy of Sciences Key Laboratory of Computational Biology, Bio-Med Big Data Center, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qinghua Wang
- School of Biological Science and Technology, University of Jinan, Jinan, China
| | - Lei Zhang
- Microbiome-X, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| |
Collapse
|
42
|
Zhao X, Pang J, Zhang W, Peng X, Yang Z, Bai G, Xia Y. Tryptophan metabolism and piglet diarrhea: Where we stand and the challenges ahead. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2024; 17:123-133. [PMID: 38766516 PMCID: PMC11101943 DOI: 10.1016/j.aninu.2024.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 02/13/2024] [Accepted: 03/20/2024] [Indexed: 05/22/2024]
Abstract
The intestinal architecture of piglets is vulnerable to disruption during weaning transition and leads to diarrhea, frequently accompanied by inflammation and metabolic disturbances (including amino acid metabolism). Tryptophan (Trp) plays an essential role in orchestrating intestinal immune tolerance through its metabolism via the kynurenine, 5-hydroxytryptamine, or indole pathways, which could be dictated by the gut microbiota either directly or indirectly. Emerging evidence suggests a strong association between piglet diarrhea and Trp metabolism. Here we aim to summarize the intricate balance of microbiota-host crosstalk by analyzing alterations in both the host and microbial pathways of Trp and discuss how Trp metabolism may affect piglet diarrhea. Overall, this review could provide valuable insights to explore effective strategies for managing piglet diarrhea and the related challenges.
Collapse
Affiliation(s)
- Xuan Zhao
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Jiaman Pang
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Wanghong Zhang
- Yunnan Vocational College of Agriculture, Kunming 650211, China
| | - Xie Peng
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Zhenguo Yang
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Guangdong Bai
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Yaoyao Xia
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| |
Collapse
|
43
|
Ma XQ, Wang B, Wei W, Tan FC, Su H, Zhang JZ, Zhao CY, Zheng HJ, Feng YQ, Shen W, Yang JB, Li FL. Alginate oligosaccharide assimilation by gut microorganisms and the potential role in gut inflammation alleviation. Appl Environ Microbiol 2024; 90:e0004624. [PMID: 38563787 PMCID: PMC11107165 DOI: 10.1128/aem.00046-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 03/10/2024] [Indexed: 04/04/2024] Open
Abstract
Dietary fiber metabolism by gut microorganisms plays important roles in host physiology and health. Alginate, the major dietary fiber of daily diet seaweeds, is drawing more attention because of multiple biological activities. To advance the understanding of alginate assimilation mechanism in the gut, we show the presence of unsaturated alginate oligosaccharides (uAOS)-specific alginate utilization loci (AUL) in human gut microbiome. As a representative example, a working model of the AUL from the gut microorganism Bacteroides clarus was reconstructed from biochemistry and transcriptome data. The fermentation of resulting monosaccharides through Entner-Doudoroff pathway tunes the metabolism of short-chain fatty acids and amino acids. Furthermore, we show that uAOS feeding protects the mice against dextran sulfate sodium-induced acute colitis probably by remodeling gut microbiota and metabolome. IMPORTANCE Alginate has been included in traditional Chinese medicine and daily diet for centuries. Recently discovered biological activities suggested that alginate-derived alginate oligosaccharides (AOS) might be an active ingredient in traditional Chinese medicine, but how these AOS are metabolized in the gut and how it affects health need more information. The study on the working mechanism of alginate utilization loci (AUL) by the gut microorganism uncovers the role of unsaturated alginate oligosaccharides (uAOS) assimilation in tuning short-chain fatty acids and amino acids metabolism and demonstrates that uAOS metabolism by gut microorganisms results in a variation of cell metabolites, which potentially contributes to the physiology and health of gut.
Collapse
Affiliation(s)
- Xiao-Qing Ma
- Qingdao C1 Refinery Engineering Research Center, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, China
| | - Bing Wang
- Qingdao C1 Refinery Engineering Research Center, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, China
| | - Wei Wei
- Innovation Center of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Fang-Cheng Tan
- Qingdao C1 Refinery Engineering Research Center, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, China
| | - Hang Su
- Qingdao C1 Refinery Engineering Research Center, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, China
| | - Jun-Zhe Zhang
- Qingdao C1 Refinery Engineering Research Center, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, China
| | - Chen-Yang Zhao
- Innovation Center of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Hua-Jun Zheng
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center at Shanghai, Shanghai, China
| | - Yan-Qin Feng
- College of Life Sciences, Qingdao Agricultural University, Qingdao, China
| | - Wei Shen
- College of Life Sciences, Qingdao Agricultural University, Qingdao, China
| | - Jin-Bo Yang
- Innovation Center of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Fu-Li Li
- Qingdao C1 Refinery Engineering Research Center, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, China
- Shandong Energy Institute, Qingdao, China
- Qingdao New Energy Shandong Laboratory, Qingdao, China
| |
Collapse
|
44
|
Chen J, Zhou M, Chen L, Yang C, Deng Y, Li J, Sun S. Evaluation of Physicochemical Properties and Prebiotics Function of a Bioactive Pleurotus eryngii Aqueous Extract Powder Obtained by Spray Drying. Nutrients 2024; 16:1555. [PMID: 38892489 PMCID: PMC11173815 DOI: 10.3390/nu16111555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 05/11/2024] [Accepted: 05/17/2024] [Indexed: 06/21/2024] Open
Abstract
A bioactive Pleurotus eryngii aqueous extract powder (SPAE) was obtained by spray drying and its performance in terms of physicochemical properties, in vitro digestion, inflammatory factors, and modulation of the intestinal microbiota was explored. The results indicated that the SPAE exhibited a more uniform particle size distribution than P. eryngii polysaccharide (PEP). Meanwhile, a typical absorption peak observed at 843 cm-1 in the SPAE FTIR spectra indicated the existence of α-glycosidic bonds. SPAE exhibited higher antioxidant abilities and superior resistance to digestion in vitro. In addition, SPAE supplementation to mice significantly reduced the release of factors that promote inflammation, enhanced the secretion of anti-inflammatory factors, and sustained maximum production of short-chain fatty acids (SCFAs). Additionally, it significantly enhanced the relative abundance of SCFAs-producing Akkermansia and reduced the abundance of Ruminococcus and Clostridiides in intestines of mice. These results show the potential of SPAE as a novel material with prebiotic effects for the food and pharmaceutical industries.
Collapse
Affiliation(s)
- Jianqiu Chen
- College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (J.C.); (M.Z.); (L.C.); (Y.D.)
| | - Mengling Zhou
- College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (J.C.); (M.Z.); (L.C.); (Y.D.)
| | - Liding Chen
- College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (J.C.); (M.Z.); (L.C.); (Y.D.)
- Gutian Edible Fungi Research Institute, Fujian Agriculture and Forestry University, Ningde 352200, China
| | - Chengfeng Yang
- Sanya Institute, China Agricultural University, Sanya 572025, China;
| | - Yating Deng
- College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (J.C.); (M.Z.); (L.C.); (Y.D.)
| | - Jiahuan Li
- College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (J.C.); (M.Z.); (L.C.); (Y.D.)
- Gutian Edible Fungi Research Institute, Fujian Agriculture and Forestry University, Ningde 352200, China
| | - Shujing Sun
- College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (J.C.); (M.Z.); (L.C.); (Y.D.)
- Gutian Edible Fungi Research Institute, Fujian Agriculture and Forestry University, Ningde 352200, China
| |
Collapse
|
45
|
Lu YP, Liu JH, Zhang XX, Xu C, Zheng PH, Li JT, Li JJ, Wang DM, Xian JA, Zhang ZL. Integration of transcriptome, gut microbiota, and physiology reveals toxic responses of the red claw crayfish (Cherax quadricarinatus) to imidacloprid. JOURNAL OF HAZARDOUS MATERIALS 2024; 470:134293. [PMID: 38615646 DOI: 10.1016/j.jhazmat.2024.134293] [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: 01/19/2024] [Revised: 03/29/2024] [Accepted: 04/10/2024] [Indexed: 04/16/2024]
Abstract
Imidacloprid enters the water environment through rainfall and causes harm to aquatic crustaceans. However, the potential chronic toxicity mechanism of imidacloprid in crayfish has not been comprehensively studied. In this study, red claw crayfish (Cherax quadricarinatus) were exposed to 11.76, 35.27, or 88.17 μg/L imidacloprid for 30 days, and changes in the physiology and biochemistry, gut microbiota, and transcriptome of C. quadricarinatus and the interaction between imidacloprid, gut microbiota, and genes were studied. Imidacloprid induced oxidative stress and decreased growth performance in crayfish. Imidacloprid exposure caused hepatopancreas damage and decreased serum immune enzyme activity. Hepatopancreatic and plasma acetylcholine decreased significantly in the 88.17 μg/L group. Imidacloprid reduced the diversity of the intestinal flora, increased the abundance of harmful flora, and disrupted the microbiota function. Transcriptomic analysis showed that the number of up-and-down-regulated differentially expressed genes (DEGs) increased significantly with increasing concentrations of imidacloprid. DEG enrichment analyses indicated that imidacloprid inhibits neurotransmitter transduction and immune responses and disrupts energy metabolic processes. Crayfish could alleviate imidacloprid stress by regulating antioxidant and detoxification-related genes. A high correlation was revealed between GST, HSPA1s, and HSP90 and the composition of gut microorganisms in crayfish under imidacloprid stress. This study highlights the negative effects and provides detailed sequencing data from transcriptome and gut microbiota to enhance our understanding of the molecular toxicity of imidacloprid in crustaceans.
Collapse
Affiliation(s)
- Yao-Peng Lu
- Hainan Provincial Key Laboratory for Functional Components Research and Utilization of Marine Bio-resources, Institute of Tropical Biosciences and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China
| | - Jia-Han Liu
- Hainan Provincial Key Laboratory for Functional Components Research and Utilization of Marine Bio-resources, Institute of Tropical Biosciences and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China
| | - Xiu-Xia Zhang
- Hainan Provincial Key Laboratory for Functional Components Research and Utilization of Marine Bio-resources, Institute of Tropical Biosciences and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China
| | - Chi Xu
- Hainan Provincial Key Laboratory for Functional Components Research and Utilization of Marine Bio-resources, Institute of Tropical Biosciences and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China
| | - Pei-Hua Zheng
- Hainan Provincial Key Laboratory for Functional Components Research and Utilization of Marine Bio-resources, Institute of Tropical Biosciences and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China
| | - Jun-Tao Li
- Hainan Provincial Key Laboratory for Functional Components Research and Utilization of Marine Bio-resources, Institute of Tropical Biosciences and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China
| | - Jia-Jun Li
- Hainan Provincial Key Laboratory for Functional Components Research and Utilization of Marine Bio-resources, Institute of Tropical Biosciences and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China
| | - Dong-Mei Wang
- Hainan Provincial Key Laboratory for Functional Components Research and Utilization of Marine Bio-resources, Institute of Tropical Biosciences and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China
| | - Jian-An Xian
- Hainan Provincial Key Laboratory for Functional Components Research and Utilization of Marine Bio-resources, Institute of Tropical Biosciences and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China.
| | - Ze-Long Zhang
- Hainan Provincial Key Laboratory for Functional Components Research and Utilization of Marine Bio-resources, Institute of Tropical Biosciences and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China.
| |
Collapse
|
46
|
Kim YS, Unno T, Park SY, Chung JO, Choi YD, Lee SM, Cho SH, Kim DH, Kim HS, Jung YD. Effect of bile reflux on gastric juice microbiota in patients with different histology phenotypes. Gut Pathog 2024; 16:26. [PMID: 38715101 PMCID: PMC11077708 DOI: 10.1186/s13099-024-00619-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 04/24/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND/AIMS Bile reflux (BR) can influence the gastric environment by altering gastric acidity and possibly the gastric microbiota composition. This study investigated the correlation between bile acids and microbial compositions in the gastric juice of 50 subjects with differing gastric pathologies. METHODS This study included 50 subjects, which were categorized into three groups based on the endoscopic BR grading system. The primary and secondary bile acid concentrations in gastric juice samples were measured, and microbiota profiling was conducted using 16 S rRNA gene sequencing. RESULTS Significant differences were observed in each bile acid level in the three endoscopic BR groups (P < 0.05). The Shannon index demonstrated a significant decrease in the higher BR groups (P < 0.05). Analysis of the β-diversity revealed that BR significantly altered the gastric microbiota composition. The presence of neoplastic lesions and the presence of H. pylori infection impacted the β-diversity of the gastric juice microbiota. The abundance of the Streptococcus and Lancefielfdella genera exhibited positive correlations for almost all bile acid components(P < 0.05). In addition, the abundance of Slobacterium, Veillonella, and Schaalia showed positive correlations with primary unconjugated bile acids (P < 0.05). CONCLUSION Changes in microbial diversity in the gastric juice were associated with BR presence in the stomach. This result suggests that the degree of BR should be considered when studying the gastric juice microbiome.
Collapse
Affiliation(s)
- Yong Sung Kim
- Digestive Disease Research Institute, Wonkwang University School of Medicine, Iksan, South Korea
| | - Tatsuya Unno
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Seowon-Gu, Cheongju, 28644, South Korea
| | - Seon-Young Park
- Division of Gastroenterology, Department of Internal Medicine, Chonnam National University Medical School, 42 Jaebong-ro, Donggu, Gwangju, 61572, South Korea.
| | - Jin Ook Chung
- Division of Endocrinology, Department of Internal Medicine, Chonnam National University Medical School, Gwangju, South Korea
| | - Yoo-Duk Choi
- Department of Pathology, Chonnam National University Medical School, Gwangju, South Korea
| | - Su-Mi Lee
- Division of Gastroenterology, Department of Internal Medicine, Chonnam National University Medical School, 42 Jaebong-ro, Donggu, Gwangju, 61572, South Korea
| | - Seong Hyun Cho
- Division of Gastroenterology, Department of Internal Medicine, Chonnam National University Medical School, 42 Jaebong-ro, Donggu, Gwangju, 61572, South Korea
| | - Dong Hyun Kim
- Division of Gastroenterology, Department of Internal Medicine, Chonnam National University Medical School, 42 Jaebong-ro, Donggu, Gwangju, 61572, South Korea
| | - Hyun-Soo Kim
- Division of Gastroenterology, Department of Internal Medicine, Chonnam National University Medical School, 42 Jaebong-ro, Donggu, Gwangju, 61572, South Korea
| | - Young Do Jung
- Department of Biochemistry, Chonnam National University Medical School, Gwangju, South Korea
| |
Collapse
|
47
|
O'Brien JW, Merali N, Pring C, Rockall T, Robertson D, Bartlett D, Frampton A. Gastrointestinal Permeability After Bariatric Surgery: A Systematic Review. Cureus 2024; 16:e60480. [PMID: 38883053 PMCID: PMC11180380 DOI: 10.7759/cureus.60480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2024] [Indexed: 06/18/2024] Open
Abstract
Gastrointestinal permeability refers to the movement of substances across the gut wall. This is mediated by endotoxemia (bacterial products entering the systemic circulation), and is associated with metabolic disease. The effect of bariatric surgery on permeability remains uncertain; the associated dietary, metabolic and weight changes are suggested to influence, or trigger, altered permeability. The primary aim of this study is to synthesize evidence and analyze the effect of bariatric surgery on permeability. A systematic review was performed, searching MEDLINE, EMBASE, and Scopus until February 2023, using MESH terms "intestinal permeability", "bariatric", for studies reporting in vivo assessment of permeability. Three cohort studies and two case series were identified (n=96). Data was heterogeneous; methodology and controls preclude meta-analysis. Gastroduodenal permeability reduced post-sleeve gastrectomy (SG). Two studies showed an increase in small intestinal permeability after biliopancreatic diversion. Two studies revealed a decrease in post-Roux-en-Y gastric bypass. One study identified increased colonic permeability six months post-SG. Evidence regarding permeability change after bariatric surgery is conflicting, notably for the small intestine. Impaired colonic permeability post-SG raises concerns regarding colonic protein fermentation and harmful dietary sequelae. There are multiple interacting variables confounding gastrointestinal permeability change; procedure type, altered microbiota and metabolic response to surgery. Further understanding of this important aspect of obesity is required, both before and after bariatric surgery.
Collapse
Affiliation(s)
- James W O'Brien
- Department of Surgery, School of Biosciences and Medicine, University of Surrey, Guildford, GBR
- Department of Minimal Access Therapy Training Unit, Royal Surrey NHS Foundation Trust, Guildford, GBR
| | - Nabeel Merali
- Department of Surgery, School of Biosciences and Medicine, University of Surrey, Guildford, GBR
- Department of Minimal Access Therapy Training Unit, Royal Surrey NHS Foundation Trust, Guildford, GBR
| | - Chris Pring
- Department of Bariatric Surgery, University Hospitals Sussex NHS Foundation Trust, Chichester, GBR
| | - Tim Rockall
- Department of Minimal Access Therapy Training Unit, Royal Surrey NHS Foundation Trust, Guildford, GBR
| | - Denise Robertson
- Department of Nutrition, School of Biosciences and Medicine, University of Surrey, Guildford, GBR
| | - David Bartlett
- Department of Nutrition, School of Biosciences and Medicine, University of Surrey, Guildford, GBR
| | - Adam Frampton
- Department of Surgery, School of Biosciences and Medicine, University of Surrey, Guildford, GBR
| |
Collapse
|
48
|
Huangfu W, Cao S, Li S, Zhang S, Liu M, Liu B, Zhu X, Cui Y, Wang Z, Zhao J, Shi Y. In vitro and in vivo fermentation models to study the function of dietary fiber in pig nutrition. Appl Microbiol Biotechnol 2024; 108:314. [PMID: 38683435 PMCID: PMC11058960 DOI: 10.1007/s00253-024-13148-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/08/2024] [Accepted: 04/15/2024] [Indexed: 05/01/2024]
Abstract
The importance of dietary fiber (DF) in animal diets is increasing with the advancement of nutritional research. DF is fermented by gut microbiota to produce metabolites, which are important in improving intestinal health. This review is a systematic review of DF in pig nutrition using in vitro and in vivo models. The fermentation characteristics of DF and the metabolic mechanisms of its metabolites were summarized in an in vitro model, and it was pointed out that SCFAs and gases are the important metabolites connecting DF, gut microbiota, and intestinal health, and they play a key role in intestinal health. At the same time, some information about host-microbe interactions could have been improved through traditional animal in vivo models, and the most direct feedback on nutrients was generated, confirming the beneficial effects of DF on sow reproductive performance, piglet intestinal health, and growing pork quality. Finally, the advantages and disadvantages of different fermentation models were compared. In future studies, it is necessary to flexibly combine in vivo and in vitro fermentation models to profoundly investigate the mechanism of DF on the organism in order to promote the development of precision nutrition tools and to provide a scientific basis for the in-depth and rational utilization of DF in animal husbandry. KEY POINTS: • The fermentation characteristics of dietary fiber in vitro models were reviewed. • Metabolic pathways of metabolites and their roles in the intestine were reviewed. • The role of dietary fiber in pigs at different stages was reviewed.
Collapse
Affiliation(s)
- Weikang Huangfu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, No.15 Longzihu University Area, Zhengdong New District, Zhengzhou, 450046, China
| | - Shixi Cao
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, No.15 Longzihu University Area, Zhengdong New District, Zhengzhou, 450046, China
| | - Shouren Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, No.15 Longzihu University Area, Zhengdong New District, Zhengzhou, 450046, China
| | - Shuhang Zhang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, No.15 Longzihu University Area, Zhengdong New District, Zhengzhou, 450046, China
| | - Mengqi Liu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, No.15 Longzihu University Area, Zhengdong New District, Zhengzhou, 450046, China
| | - Boshuai Liu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, No.15 Longzihu University Area, Zhengdong New District, Zhengzhou, 450046, China
- Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou, China
- Henan Forage Engineering Technology Research Center, Zhengzhou, 450002, Henan, China
| | - Xiaoyan Zhu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, No.15 Longzihu University Area, Zhengdong New District, Zhengzhou, 450046, China
- Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou, China
- Henan Forage Engineering Technology Research Center, Zhengzhou, 450002, Henan, China
| | - Yalei Cui
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, No.15 Longzihu University Area, Zhengdong New District, Zhengzhou, 450046, China
- Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou, China
- Henan Forage Engineering Technology Research Center, Zhengzhou, 450002, Henan, China
| | - Zhichang Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, No.15 Longzihu University Area, Zhengdong New District, Zhengzhou, 450046, China
- Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou, China
- Henan Forage Engineering Technology Research Center, Zhengzhou, 450002, Henan, China
| | - Jiangchao Zhao
- Department of Animal Science, Division of Agriculture, University of Arkansas, Fayetteville, AR, USA
| | - Yinghua Shi
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, No.15 Longzihu University Area, Zhengdong New District, Zhengzhou, 450046, China.
- Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou, China.
- Henan Forage Engineering Technology Research Center, Zhengzhou, 450002, Henan, China.
| |
Collapse
|
49
|
Stevens J, Culberson E, Kinder J, Ramiriqui A, Gray J, Bonfield M, Shao TY, Al Gharabieh F, Peterson L, Steinmeyer S, Zacharias W, Pryhuber G, Paul O, Sengupta S, Alenghat T, Way SS, Deshmukh H. Microbiota-derived inosine programs protective CD8 + T cell responses against influenza in newborns. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.09.588427. [PMID: 38645130 PMCID: PMC11030415 DOI: 10.1101/2024.04.09.588427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
The immunological defects causing susceptibility to severe viral respiratory infections due to early-life dysbiosis remain ill-defined. Here, we show that influenza virus susceptibility in dysbiotic infant mice is caused by CD8+ T cell hyporesponsiveness and diminished persistence as tissue-resident memory cells. We describe a previously unknown role for nuclear factor interleukin 3 (NFIL3) in repression of memory differentiation of CD8+ T cells in dysbiotic mice involving epigenetic regulation of T cell factor 1 (TCF 1) expression. Pulmonary CD8+ T cells from dysbiotic human infants share these transcriptional signatures and functional phenotypes. Mechanistically, intestinal inosine was reduced in dysbiotic human infants and newborn mice, and inosine replacement reversed epigenetic dysregulation of Tcf7 and increased memory differentiation and responsiveness of pulmonary CD8+ T cells. Our data unveils new developmental layers controlling immune cell activation and identifies microbial metabolites that may be used therapeutically in the future to protect at-risk newborns.
Collapse
Affiliation(s)
- Joseph Stevens
- Department of Pediatrics, University of Cincinnati College of Medicine
- Division of Neonatology, Cincinnati Children’s Hospital Medical Center
- Medical Scientist Training Program, University of Cincinnati College of Medicine
- Immunology Graduate Program, Cincinnati Children’s Hospital Medical Center
| | - Erica Culberson
- Department of Pediatrics, University of Cincinnati College of Medicine
- Division of Neonatology, Cincinnati Children’s Hospital Medical Center
- Medical Scientist Training Program, University of Cincinnati College of Medicine
- Immunology Graduate Program, Cincinnati Children’s Hospital Medical Center
| | - Jeremy Kinder
- Department of Pediatrics, University of Cincinnati College of Medicine
- Division of Infectious Disease, Cincinnati Children’s Hospital Medical Center
- Center for Inflammation and Tolerance, Cincinnati Children’s Hospital Medical Center
| | - Alicia Ramiriqui
- Division of Neonatology, Cincinnati Children’s Hospital Medical Center
| | - Jerilyn Gray
- Division of Neonatology, Cincinnati Children’s Hospital Medical Center
| | - Madeline Bonfield
- Department of Pediatrics, University of Cincinnati College of Medicine
- Division of Neonatology, Cincinnati Children’s Hospital Medical Center
- Immunology Graduate Program, Cincinnati Children’s Hospital Medical Center
| | - Tzu-Yu Shao
- Department of Pediatrics, University of Cincinnati College of Medicine
- Division of Infectious Disease, Cincinnati Children’s Hospital Medical Center
- Center for Inflammation and Tolerance, Cincinnati Children’s Hospital Medical Center
| | - Faris Al Gharabieh
- Department of Pediatrics, University of Cincinnati College of Medicine
- Division of Neonatology, Cincinnati Children’s Hospital Medical Center
| | - Laura Peterson
- Department of Pediatrics, University of Cincinnati College of Medicine
- Division of Neonatology, Cincinnati Children’s Hospital Medical Center
| | - Shelby Steinmeyer
- Department of Pediatrics, University of Cincinnati College of Medicine
- Division of Neonatology, Cincinnati Children’s Hospital Medical Center
| | - William Zacharias
- Department of Pediatrics, University of Cincinnati College of Medicine
- Medical Scientist Training Program, University of Cincinnati College of Medicine
| | - Gloria Pryhuber
- Department of Pediatrics, University of Rochester, School of Medicine
| | - Oindrila Paul
- Division of Neonatology, Children’s Hospital of Philadelphia; Perelman School of Medicine, University of Pennsylvania
| | - Shaon Sengupta
- Division of Neonatology, Children’s Hospital of Philadelphia; Perelman School of Medicine, University of Pennsylvania
| | - Theresa Alenghat
- Department of Pediatrics, University of Cincinnati College of Medicine
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center
- Center for Inflammation and Tolerance, Cincinnati Children’s Hospital Medical Center
| | - Sing Sing Way
- Department of Pediatrics, University of Cincinnati College of Medicine
- Division of Infectious Disease, Cincinnati Children’s Hospital Medical Center
- Center for Inflammation and Tolerance, Cincinnati Children’s Hospital Medical Center
| | - Hitesh Deshmukh
- Department of Pediatrics, University of Cincinnati College of Medicine
- Division of Neonatology, Cincinnati Children’s Hospital Medical Center
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center
- Center for Inflammation and Tolerance, Cincinnati Children’s Hospital Medical Center
| |
Collapse
|
50
|
Zhu H, Yang X, Zhao Y. Recent Advances in Current Uptake Situation, Metabolic and Nutritional Characteristics, Health, and Safety of Dietary Tryptophan. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:6787-6802. [PMID: 38512048 DOI: 10.1021/acs.jafc.3c06419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
Tryptophan (Trp) is an essential amino acid which is unable to be synthesized in the body. Main sources of Trp are uptake of foods such as oats and bananas. In this review, we describe the status of current dietary consumption, metabolic pathways and nutritional characteristics of Trp, as well as its ingestion and downstream metabolites for maintaining body health and safety. This review also summarizes the recent advances in Trp metabolism, particularly the 5-HT, KYN, and AhR activation pathways, revealing that its endogenous host metabolites are not only differentially affected in the body but also are closely linked to health. More attention should be paid to targeting its specific metabolic pathways and utilizing food molecules and probiotics for manipulating Trp metabolism. However, the complexity of microbiota-host interactions requires further exploration to precisely refine targets for innovating the gut microbiota-targeted diagnostic approaches and informing subsequent studies and targeted treatments of diseases.
Collapse
Affiliation(s)
- Haoyan Zhu
- Key Laboratory of the Ministry of Education for Medicinal Resource and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China
| | - Xingbin Yang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Yan Zhao
- Key Laboratory of the Ministry of Education for Medicinal Resource and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China
| |
Collapse
|