1
|
Thibaut MM, Roumain M, Piron E, Gillard J, Loriot A, Neyrinck AM, Rodriguez J, Massart I, Thissen JP, Huot JR, Pin F, Bonetto A, Delzenne NM, Muccioli GG, Bindels LB. The microbiota-derived bile acid taurodeoxycholic acid improves hepatic cholesterol levels in mice with cancer cachexia. Gut Microbes 2025; 17:2449586. [PMID: 39780051 PMCID: PMC11730681 DOI: 10.1080/19490976.2025.2449586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 11/20/2024] [Accepted: 12/30/2024] [Indexed: 01/11/2025] Open
Abstract
Alterations in bile acid profile and pathways contribute to hepatic inflammation in cancer cachexia, a syndrome worsening the prognosis of cancer patients. As the gut microbiota impinges on host metabolism through bile acids, the current study aimed to explore the functional contribution of gut microbial dysbiosis to bile acid dysmetabolism and associated disorders in cancer cachexia. Using three mouse models of cancer cachexia (the C26, MC38 and HCT116 models), we evidenced a reduction in the hepatic levels of several secondary bile acids, mainly taurodeoxycholic (TDCA). This reduction in hepatic TDCA occurred before the appearance of cachexia. Longitudinal analysis of the gut microbiota pinpointed an ASV, identified as Xylanibacter rodentium, as a bacterium potentially involved in the reduced production of TDCA. Coherently, stable isotope-based experiments highlighted a robust decrease in the microbial 7α-dehydroxylation (7α-DH) activity with no changes in the bile salt hydrolase (BSH) activity in cachectic mice. This approach also highlighted a reduced microbial 7α-hydroxysteroid dehydrogenase (7α-HSDH) and 12α-hydroxysteroid dehydrogenase (12α-HSDH) activities in these mice. The contribution of the lower production of TDCA to cancer cachexia was explored in vitro and in vivo. In vitro, TDCA prevented myotube atrophy, whereas in vivo hepatic whole transcriptome analysis revealed that TDCA administration to cachectic mice improved the unfolded protein response and cholesterol homeostasis pathways. Coherently, TDCA administration reversed hepatic cholesterol accumulation in these mice. Altogether, this work highlights the contribution of the gut microbiota to bile acid dysmetabolism and the therapeutic interest of the secondary bile acid TDCA for hepatic cholesterol homeostasis in the context of cancer cachexia. Such discovery may prove instrumental in the understanding of other metabolic diseases characterized by microbial dysbiosis. More broadly, our work demonstrates the interest and relevance of microbial activity measurements using stable isotopes, an approach currently underused in the microbiome field.
Collapse
Affiliation(s)
- Morgane M. Thibaut
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Martin Roumain
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Edwige Piron
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Justine Gillard
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
- Laboratory of Hepato-Gastroenterology, Institut de Recherche Expérimentale et Clinique, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Axelle Loriot
- Computational Biology and Bioinformatics Unit (CBIO), de Duve Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Audrey M. Neyrinck
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Julie Rodriguez
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Isabelle Massart
- Endocrinology, Diabetology and Nutrition Department, Institut de Recherches Expérimentales et Cliniques, UCLouvain, Université catholique de Louvain, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Jean-Paul Thissen
- Endocrinology, Diabetology and Nutrition Department, Institut de Recherches Expérimentales et Cliniques, UCLouvain, Université catholique de Louvain, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Joshua R. Huot
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Fabrizio Pin
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Andrea Bonetto
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Nathalie M. Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Giulio G. Muccioli
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Laure B. Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
- Welbio Department, WEL Research Institute, Wavre, Belgium
| |
Collapse
|
2
|
Ren X, Qu Y, Shari A, Li G. Transcriptome-wide study of mRNAs modified by m 6A RNA methylation in the testis development of dairy goats. Anim Biotechnol 2025; 36:2496641. [PMID: 40306318 DOI: 10.1080/10495398.2025.2496641] [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: 10/14/2024] [Accepted: 04/14/2025] [Indexed: 05/02/2025]
Abstract
N6-methyladenosine (m6A) is an important epigenetic modification in RNA, playing a crucial role in regulating the production and aging of animal testicular sperm. This study extracted mRNA from the testicular tissue of male goats before and after sexual maturity, generating a methylation map through preliminary experiments and methylation immunoprecipitation sequencing. The results showed that during the development of dairy goats, the expression levels of marker genes related to testicular development and methylation-related enzymes changed significantly. A total of 36,602 peaks and 11,223 genes were identified in the two groups, including 2989 differential peaks (427 upregulated and 2562 downregulated) and 1457 differentially expressed genes (833 upregulated and 624 downregulated). The abundance of m6A was positively correlated with gene expression levels. This study reports for the first time the mRNA profiles of m6A modifications across the entire transcriptome during testicular development in Guanzhong dairy goats, providing a new perspective for genetic improvement in goats.
Collapse
Affiliation(s)
- Xinyang Ren
- College of Animal Science and Technology, Northwest A&F University, Yangling, PR China
| | - Yingxin Qu
- College of Animal Science and Technology, Northwest A&F University, Yangling, PR China
| | - Akang Shari
- College of Animal Science and Technology, Northwest A&F University, Yangling, PR China
| | - Guang Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, PR China
| |
Collapse
|
3
|
O’Sullivan J, Patel S, Leventhal GE, Fitzgerald RS, Laserna-Mendieta EJ, Huseyin CE, Konstantinidou N, Rutherford E, Lavelle A, Dabbagh K, DeSantis TZ, Shanahan F, Temko A, Iwai S, Claesson MJ. Host-microbe multi-omics and succinotype profiling have prognostic value for future relapse in patients with inflammatory bowel disease. Gut Microbes 2025; 17:2450207. [PMID: 39812341 PMCID: PMC11740686 DOI: 10.1080/19490976.2025.2450207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 11/07/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
Crohn's disease (CD) and ulcerative colitis (UC) are chronic relapsing inflammatory bowel disorders (IBD), the pathogenesis of which is uncertain but includes genetic susceptibility factors, immune-mediated tissue injury and environmental influences, most of which appear to act via the gut microbiome. We hypothesized that host-microbe alterations could be used to prognostically stratify patients experiencing relapses up to four years after endoscopy. We therefore examined multiple omics data, including published and new datasets, generated from paired inflamed and non-inflamed mucosal biopsies from 142 patients with IBD (54 CD; 88 UC) and from 34 control (non-diseased) biopsies. The relapse-predictive potential of 16S rRNA gene and transcript amplicons (standing and active microbiota) were investigated along with host transcriptomics, epigenomics and genetics. While standard single-omics analysis could not distinguish between patients who relapsed and those that remained in remission within four years of colonoscopy, we did find an association between the number of flares and a patient's succinotype. Our multi-omics machine learning approach was also able to predict relapse when combining features from the microbiome and human host. Therefore multi-omics, rather than single omics, better predicts relapse within 4 years of colonoscopy, while a patient's succinotype is associated with a higher frequency of relapses.
Collapse
Affiliation(s)
- Jill O’Sullivan
- School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- SFI Centre for Research Training in Genomics Data Science, University of Galway, Galway, Ireland
| | - Shriram Patel
- School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- SeqBiome Ltd, Cork, Ireland
| | | | - Rachel S. Fitzgerald
- School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Emilio J. Laserna-Mendieta
- School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Chloe E. Huseyin
- School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Nina Konstantinidou
- School of Microbiology, University College Cork, Cork, Ireland
- Department of Informatics, Second Genome Inc, South San Francisco, California, USA
| | - Erica Rutherford
- Department of Informatics, Second Genome Inc, South San Francisco, California, USA
| | - Aonghus Lavelle
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, County Cork, Ireland
| | - Karim Dabbagh
- Department of Informatics, Second Genome Inc, South San Francisco, California, USA
| | - Todd Z. DeSantis
- Department of Informatics, Second Genome Inc, South San Francisco, California, USA
| | - Fergus Shanahan
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Medicine, University College Cork, Cork, Ireland
| | - Andriy Temko
- Department of Electrical and Electronic Engineering, University College Cork, Cork, Ireland
| | - Shoko Iwai
- Department of Informatics, Second Genome Inc, South San Francisco, California, USA
| | - Marcus J. Claesson
- School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| |
Collapse
|
4
|
Duan X, Liu W, Xiao Y, Rao M, Ji L, Wan X, Han S, Lin Z, Liu H, Chen P, Qiao K, Zheng M, Shen J, Zhou Y, Asakawa T, Xiao M, Lu H. Exploration of the feasibility of clinical application of phage treatment for multidrug-resistant Serratia marcescens-induced pulmonary infection. Emerg Microbes Infect 2025; 14:2451048. [PMID: 39764739 PMCID: PMC11740298 DOI: 10.1080/22221751.2025.2451048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 12/22/2024] [Accepted: 01/05/2025] [Indexed: 01/18/2025]
Abstract
Serratia marcescens (S. marcescens) commonly induces refractory infection due to its multidrug-resistant nature. To date, there have been no reports on the application of phage treatment for S. marcescens infection. This study was conducted to explore the feasibility of phage application in treating refractory S. marcescens infection by collaborating with a 59-year-old male patient with a pulmonary infection of multidrug-resistant S. marcescens. Our experiments included three domains: i) selection of the appropriate phage, ii) verification of the efficacy and safety of the selected phage, iii) confirmation of phage-bacteria interactions. Our results showed that phage Spe5P4 is appropriate for S. marcescens infection. Treatment with phage Spe5P4 showed good efficacy, manifested as amelioration of symptoms, hydrothorax examinations, and chest computed tomography findings. Phage treatment did not worsen hepatic and renal function, immunity-related indices, or indices of routine blood examination. It did not induce or deteriorate drug resistance of the involved antibiotics. Importantly, no adverse events were reported during the treatment or follow-up periods. Thus, phage treatment showed satisfactory safety. Finally, we found that phage treatment did not increase the bacterial load, cytotoxicity, virulence, or phage resistance of S. marcescens, indicating satisfactory phage-bacteria interactions between Spe5P4 and S. marcescens, which are useful for the future application of phage Spe5P4 against S. marcescens. This work provides evidence and a working basis for further application of phage Spe5P4 in treating refractory S. marcescens infections. We also provided a methodological basis for investigating clinical application of phage treatment against multidrug-resistant bacterial infections in the future.
Collapse
Affiliation(s)
- Xiangke Duan
- Department of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Shenzhen Third People’s Hospital, Shenzhen, People’s Republic of China
| | - Wenfeng Liu
- BGI Research, Shenzhen, People’s Republic of China
| | - Yanyu Xiao
- Department of Clinical Laboratory, National Clinical Research Center for Infectious Diseases, Shenzhen Third People’s Hospital, Shenzhen, People’s Republic of China
| | - Man Rao
- Department of Infection and Immunology, National Clinical Research Center for Infectious Diseases, Shenzhen Third People’s Hospital, Shenzhen, People’s Republic of China
| | - Liyin Ji
- Department of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Shenzhen Third People’s Hospital, Shenzhen, People’s Republic of China
| | - Xiaofu Wan
- Department of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Shenzhen Third People’s Hospital, Shenzhen, People’s Republic of China
| | - Shuhong Han
- Department of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Shenzhen Third People’s Hospital, Shenzhen, People’s Republic of China
- Department of Infection and Immunology, National Clinical Research Center for Infectious Diseases, Shenzhen Third People’s Hospital, Shenzhen, People’s Republic of China
| | - Zixun Lin
- Department of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Shenzhen Third People’s Hospital, Shenzhen, People’s Republic of China
- School of Medicine, Southern University of Science and Technology, Shenzhen, People’s Republic of China
| | - Haichen Liu
- BGI Research, Shenzhen, People’s Republic of China
| | - Peifen Chen
- Department of Respiratory Medicine, National Clinical Research Center for Infectious Diseases, Shenzhen Third People’s Hospital, Shenzhen, People’s Republic of China
| | - Kun Qiao
- Department of Thoracic Surgery, National Clinical Research Center for Infectious Diseases, Shenzhen Third People’s Hospital, Shenzhen, People’s Republic of China
| | - Mingbin Zheng
- Department of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Shenzhen Third People’s Hospital, Shenzhen, People’s Republic of China
- Institute of Neurology, National Clinical Research Center for Infectious Diseases, Shenzhen Third People’s Hospital, Shenzhen, People’s Republic of China
| | - Jiayin Shen
- Department of Science and Education, National Clinical Research Center for Infectious Diseases, Shenzhen Third People’s Hospital, Shenzhen, People’s Republic of China
| | - Yang Zhou
- Department of Infection and Immunology, National Clinical Research Center for Infectious Diseases, Shenzhen Third People’s Hospital, Shenzhen, People’s Republic of China
| | - Tetsuya Asakawa
- Institute of Neurology, National Clinical Research Center for Infectious Diseases, Shenzhen Third People’s Hospital, Shenzhen, People’s Republic of China
| | - Minfeng Xiao
- BGI Research, Shenzhen, People’s Republic of China
| | - Hongzhou Lu
- Department of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Shenzhen Third People’s Hospital, Shenzhen, People’s Republic of China
- Institute of Neurology, National Clinical Research Center for Infectious Diseases, Shenzhen Third People’s Hospital, Shenzhen, People’s Republic of China
| |
Collapse
|
5
|
Ma Y, Zhang M, Wang Z, Cao L, Li Y, Wan Z, Kane Y, Wang G, Li X, Zhang C. Short-term antiretroviral therapy may not correct the dysregulations of plasma virome and cytokines induced by HIV-1 infection. Virulence 2025; 16:2467168. [PMID: 39950859 PMCID: PMC11866967 DOI: 10.1080/21505594.2025.2467168] [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: 10/14/2024] [Revised: 12/09/2024] [Accepted: 02/09/2025] [Indexed: 02/28/2025] Open
Abstract
An expansion of plasma anelloviruses and dysregulation of inflammation was associated with HIV-1 infection. However, how antiretroviral therapy (ART) affects the dynamics of plasma virome and cytokine profile remains largely unknown. To characterize the dynamics of plasma virome and cytokines in HIV-1-infected individuals before and during the first year of ART, a cohort of 26 HIV-1-infected individuals and 19 healthy controls was recruited. Blood samples were collected and subjected to metagenomic analysis and the measurement of 27 cytokines. Metagenomic analysis revealed an increased abundance and prevalence of human pegivirus type 1 (HPgV-1) and a slightly decreased diversity and abundance of anellovirus in plasma of HIV-1-infected individuals after ART. No obvious impact was observed on other plasma commensal viruses. Increased abundance and prevalence of HPgV-1 were further confirmed by RT-qPCR assay in a larger cohort of 114 HIV-1-infected individuals. Notably, most dysregulated cytokines were not fully restored by ART, with extremely abnormal levels of IL-10, GM-CSF, VEGF, and eotaxin, and a significantly increased level of plasma I-FABP. Anelloviruses showed significantly negative correlations with other commensal viruses except HPgV-1 but had positive correlations with several anti-inflammatory and Th1 cytokines. These results suggest that short-term ART may not significantly correct the virome and cytokine dysregulations induced by HIV-1 infection. The results highlight a need for further investigation into the long-term effects of ART on virome and cytokine profiles in HIV-1-infected individuals.
Collapse
Affiliation(s)
- Yingying Ma
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Min Zhang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Zhenyan Wang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Le Cao
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Yanpeng Li
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Zhenzhou Wan
- Medical Laboratory,Taizhou Fourth People’s Hospital, Taizhou, China
| | - Yakhouba Kane
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Gang Wang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Xin Li
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Chiyu Zhang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| |
Collapse
|
6
|
Carasso S, Keshet-David R, Zhang J, Hajjo H, Kadosh-Kariti D, Gefen T, Geva-Zatorsky N. Bacteriophage-driven DNA inversions shape bacterial functionality and long-term co-existence in Bacteroides fragilis. Gut Microbes 2025; 17:2501492. [PMID: 40350564 PMCID: PMC12068327 DOI: 10.1080/19490976.2025.2501492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 04/21/2025] [Accepted: 04/28/2025] [Indexed: 05/14/2025] Open
Abstract
Bacterial genomic DNA inversions, which govern molecular phase-variations, provide the bacteria with functional plasticity and phenotypic diversity. These targeted rearrangements enable bacteria to respond to environmental challenges, such as bacteriophage predation, evading immune detection or gut colonization. This study investigated the short- and long-term effects of the lytic phage Barc2635 on the functional plasticity of Bacteroides fragilis, a gut commensal. Germ-free mice were colonized with B. fragilis and exposed to Barc2635 to identify genomic alterations driving phenotypic changes. Phage exposure triggered dynamic and prolonged bacterial responses, including significant shifts in phase-variable regions (PVRs), particularly in promoter orientations of polysaccharide biosynthesis loci. These shifts coincided with increased entropy in PVR inversion ratios, reflecting heightened genomic variability. In contrast, B. fragilis in control mice exhibited stable genomic configurations after gut adaptation. The phase-variable Type 1 restriction-modification system, which affects broad gene expression patterns, showed variability in both groups. However, phage-exposed bacteria displayed more restrained variability, suggesting phage-derived selection pressures. Our findings reveal that B. fragilis employs DNA inversions to adapt rapidly to phage exposure and colonization, highlighting a potential mechanism by which genomic variability contributes to its response to phage. This study demonstrates gut bacterial genomic and phenotypic plasticity upon exposure to the mammalian host and to bacteriophages.
Collapse
Affiliation(s)
- Shaqed Carasso
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion – Israel Institute of Technology, Rappaport Technion Integrated Cancer Center (RTICC), Haifa, Israel
| | - Roni Keshet-David
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion – Israel Institute of Technology, Rappaport Technion Integrated Cancer Center (RTICC), Haifa, Israel
| | - Jia Zhang
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion – Israel Institute of Technology, Rappaport Technion Integrated Cancer Center (RTICC), Haifa, Israel
| | - Haitham Hajjo
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion – Israel Institute of Technology, Rappaport Technion Integrated Cancer Center (RTICC), Haifa, Israel
| | - Dana Kadosh-Kariti
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion – Israel Institute of Technology, Rappaport Technion Integrated Cancer Center (RTICC), Haifa, Israel
| | - Tal Gefen
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion – Israel Institute of Technology, Rappaport Technion Integrated Cancer Center (RTICC), Haifa, Israel
| | - Naama Geva-Zatorsky
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion – Israel Institute of Technology, Rappaport Technion Integrated Cancer Center (RTICC), Haifa, Israel
- CIFAR Humans & the Microbiome Program, CIFAR, Toronto, ON, Canada
| |
Collapse
|
7
|
Hoops SL, Moutsoglou D, Vaughn BP, Khoruts A, Knights D. Metagenomic source tracking after microbiota transplant therapy. Gut Microbes 2025; 17:2487840. [PMID: 40229213 PMCID: PMC12005403 DOI: 10.1080/19490976.2025.2487840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 03/07/2025] [Accepted: 03/28/2025] [Indexed: 04/16/2025] Open
Abstract
Reliable engraftment assessment of donor microbial communities and individual strains is an essential component of characterizing the pharmacokinetics of microbiota transplant therapies (MTTs). Recent methods for measuring donor engraftment use whole-genome sequencing and reference databases or metagenome-assembled genomes (MAGs) to track individual bacterial strains but lack the ability to disambiguate DNA that matches both donor and patient microbiota. Here, we describe a new, cost-efficient analytic pipeline, MAGEnTa, which compares post-MTT samples to a database comprised MAGs derived directly from donor and pre-treatment metagenomic data, without relying on an external database. The pipeline uses Bayesian statistics to determine the likely sources of ambiguous reads that align with both the donor and pre-treatment samples. MAGEnTa recovers engrafted strains with minimal type II error in a simulated dataset and is robust to shallow sequencing depths in a downsampled dataset. Applying MAGEnTa to a dataset from a recent MTT clinical trial for ulcerative colitis, we found the results to be consistent with 16S rRNA gene SourceTracker analysis but with added MAG-level specificity. MAGEnTa is a powerful tool to study community and strain engraftment dynamics in the development of MTT-based treatments that can be integrated into frameworks for functional and taxonomic analysis.
Collapse
Affiliation(s)
- Susan L. Hoops
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, MN, USA
- Biotechnology Institute, University of Minnesota, Minneapolis, MN, USA
| | - Daphne Moutsoglou
- Gastroenterology Section, Minneapolis VA Health Care System, Minneapolis, MN, USA
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Byron P. Vaughn
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
- Division of Gastroenterology, University of Minnesota, Minneapolis, MN, USA
| | - Alexander Khoruts
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
- Division of Gastroenterology, University of Minnesota, Minneapolis, MN, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Dan Knights
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, MN, USA
- Biotechnology Institute, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
8
|
Serna C, Calderón Bernal JM, Torre-Fuentes L, García Muñoz Á, Díez Guerrier A, Hernández M, Fernández-Garayzábal JF, Vela AI, Cid D, Alvarez J. Integrative and conjugative elements associated with antimicrobial resistance in multidrug resistant Pasteurella multocida isolates from bovine respiratory disease (BRD)-affected animals in Spanish feedlots. Vet Q 2025; 45:1-15. [PMID: 40055923 PMCID: PMC11892046 DOI: 10.1080/01652176.2025.2474220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 12/16/2024] [Accepted: 02/24/2025] [Indexed: 05/13/2025] Open
Abstract
The emergence of multidrug-resistance (MDR) in Pasteurella multocida, a major contributor to bovine respiratory disease (BRD) is being increasingly reported, often linked to the carriage of antimicrobial resistance genes (ARGs) on integrative and conjugative elements (ICEs). The resistance phenotype for 19 antimicrobials was determined using broth microdilution in 75 Pasteurella multocida isolates from healthy and BRD-affected cattle from five feedlots. The genomes of 32 isolates were sequenced to identify ARG) and mobile genetic elements (MGEs) and assess their genetic diversity. MDR isolates (with phenotypic resistance to aminoglycosides, macrolides, fluoroquinolones and/or tetracyclines) were primarily found among BRD-affected compared to healthy animals. Non-susceptible isolates, belonging to ST79 and ST13, harbored point mutations and four to nine ARGs, including rarely reported mechanisms in Europe (mph(E), msr(E) and aadA31 ARGs and newly described mutations in the gyrA/parC genes). All ARGs were linked to the presence of MGEs including two ICEs, Tn7407 and the novel Tn7809, a prophage and a putative composite transposon. Clonally related isolates were found in different batches from the same feedlot, suggesting maintenance of MDR strains. Our findings demonstrate the diverse genetic basis of AMR in P. multocida from BRD-affected cattle in Spain, emphasizing the role of MGEs in the ARG dissemination.
Collapse
Affiliation(s)
- Carlos Serna
- Departamento de Sanidad Animal, Facultad de Veterinaria, Universidad Complutense, Madrid, Spain
| | | | - Laura Torre-Fuentes
- Centro de Vigilancia Sanitaria Veterinaria (VISAVET,), Universidad Complutense, Madrid, Spain
| | - Ángel García Muñoz
- Departamento de Producción y Sanidad Animal, Salud Pública Veterinaria y Ciencia y Tecnología de los Alimentos, Facultad de Veterinaria, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
| | - Alberto Díez Guerrier
- Departamento de Sanidad Animal, Facultad de Veterinaria, Universidad Complutense, Madrid, Spain
- Centro de Vigilancia Sanitaria Veterinaria (VISAVET,), Universidad Complutense, Madrid, Spain
| | - Marta Hernández
- Laboratorio de Biología Molecular y Microbiología, Instituto Tecnológico Agrario de Castilla y León, Valladolid, Spain
| | - José Francisco Fernández-Garayzábal
- Departamento de Sanidad Animal, Facultad de Veterinaria, Universidad Complutense, Madrid, Spain
- Centro de Vigilancia Sanitaria Veterinaria (VISAVET,), Universidad Complutense, Madrid, Spain
| | - Ana Isabel Vela
- Departamento de Sanidad Animal, Facultad de Veterinaria, Universidad Complutense, Madrid, Spain
- Centro de Vigilancia Sanitaria Veterinaria (VISAVET,), Universidad Complutense, Madrid, Spain
| | - Dolores Cid
- Departamento de Sanidad Animal, Facultad de Veterinaria, Universidad Complutense, Madrid, Spain
| | - Julio Alvarez
- Departamento de Sanidad Animal, Facultad de Veterinaria, Universidad Complutense, Madrid, Spain
- Centro de Vigilancia Sanitaria Veterinaria (VISAVET,), Universidad Complutense, Madrid, Spain
| |
Collapse
|
9
|
Aboulalazm FA, Kazen AB, deLeon O, Müller S, Saravia FL, Lozada-Fernandez V, Hadiono MA, Keyes RF, Smith BC, Kellogg SL, Grobe JL, Kindel TL, Kirby JR. Reutericyclin, a specialized metabolite of Limosilactobacillus reuteri, mitigates risperidone-induced weight gain in mice. Gut Microbes 2025; 17:2477819. [PMID: 40190120 PMCID: PMC11980487 DOI: 10.1080/19490976.2025.2477819] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 01/14/2025] [Accepted: 03/05/2025] [Indexed: 04/11/2025] Open
Abstract
The role of xenobiotic disruption of microbiota, corresponding dysbiosis, and potential links to host metabolic diseases are of critical importance. In this study, we used a widely prescribed antipsychotic drug, risperidone, known to influence weight gain in humans, to induce weight gain in C57BL/6J female mice. We hypothesized that microbes essential for maintaining gut homeostasis and energy balance would be depleted following treatment with risperidone, leading to enhanced weight gain relative to controls. Thus, we performed metagenomic analyses on stool samples to identify microbes that were excluded in risperidone-treated animals but remained present in controls. We identified multiple taxa including Limosilactobacillus reuteri as a candidate for further study. Oral supplementation with L. reuteri protected against risperidone-induced weight gain (RIWG) and was dependent on cellular production of a specialized metabolite, reutericyclin. Further, synthetic reutericyclin was sufficient to mitigate RIWG. Both synthetic reutericyclin and L. reuteri restored energy balance in the presence of risperidone to mitigate excess weight gain and induce shifts in the microbiome associated with leanness. In total, our results identify reutericyclin production by L. reuteri as a potential probiotic to restore energy balance induced by risperidone and to promote leanness.
Collapse
Affiliation(s)
- Fatima A. Aboulalazm
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Alexis B. Kazen
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Orlando deLeon
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Susanne Müller
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Fatima L. Saravia
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | - Matthew A. Hadiono
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Robert F. Keyes
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA
- Program in Chemical Biology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Brian C. Smith
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA
- Program in Chemical Biology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Stephanie L. Kellogg
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Justin L. Grobe
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI, USA
- Comprehensive Rodent Metabolic Phenotyping Core, Medical College of Wisconsin, Milwaukee, WI, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Tammy L. Kindel
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | - John R. Kirby
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA
- Center for Microbiome Research, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
10
|
Spiegelhauer MR, Offersen SM, Mao X, Gambino M, Sandris Nielsen D, Nguyen DN, Brunse A. Protection against experimental necrotizing enterocolitis by fecal filtrate transfer requires an active donor virome. Gut Microbes 2025; 17:2486517. [PMID: 40207909 PMCID: PMC11988273 DOI: 10.1080/19490976.2025.2486517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 03/10/2025] [Accepted: 03/25/2025] [Indexed: 04/11/2025] Open
Abstract
Necrotizing enterocolitis (NEC) remains a frequent catastrophic disease in preterm infants, and fecal filtrate transfer (FFT) has emerged as a promising prophylactic therapy. This study explored the role of virome viability for the protective effect of FFT. Using ultraviolet (UV) irradiation, we established a viral inactivation protocol and administered FFT, UV-inactivated FFT (iFFT) or sterile saline orally to preterm piglets at risk for experimental NEC. The gut pathology and barrier properties were assessed, while the microbiome was explored by 16S rRNA amplicon and metavirome sequencing. Like in prior studies, FFT reduced NEC severity and intestinal inflammation, while these effects were absent in the iFFT group. Unexpectedly, piglets receiving FFT exhibited mild side effects in the form of early-onset diarrhea. The FFT also converged the gut colonization by increased viral heterogeneity and a reduced abundance of pathobionts like Clostridium perfringens and Escherichia. In contrast, the gut microbiome of iFFT recipients diverged from both FFT and the controls. These findings highlight the clear distinction between the ability of active and inactivate viromes to modulate gut microbiota and decrease pathology. The efficacy of FFT may be driven by active bacteriophages, and loss of virome activity could have consequences for the treatment efficacy.
Collapse
Affiliation(s)
- Malene Roed Spiegelhauer
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Simone Margaard Offersen
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Xiaotian Mao
- Section for Food Microbiology, Gut Health and Fermentation, Department of Food Science, University of Copenhagen, Copenhagen, Denmark
| | - Michela Gambino
- Institute for Conservation, Royal Danish Academy, Copenhagen, Denmark
| | - Dennis Sandris Nielsen
- Section for Food Microbiology, Gut Health and Fermentation, Department of Food Science, University of Copenhagen, Copenhagen, Denmark
| | - Duc Ninh Nguyen
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anders Brunse
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
11
|
Jang YJ, Oh SD, Hong JK, Park JC, Lee SK, Chang A, Yun DW, Lee B. Impact of genetically modified herbicide-resistant maize on rhizosphere bacterial communities. GM CROPS & FOOD 2025; 16:186-198. [PMID: 39950610 PMCID: PMC11834531 DOI: 10.1080/21645698.2025.2466915] [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: 09/26/2024] [Revised: 02/05/2025] [Accepted: 02/06/2025] [Indexed: 02/20/2025]
Abstract
Rhizosphere bacterial community studies offer valuable insights into the environmental implications of genetically modified (GM) crops. This study compared the effects of a non-GM maize cultivar, namely Hi-IIA, with those of a herbicide-resistant maize cultivar containing the phosphinothricin N-acetyltransferase gene on the rhizosphere bacterial community across growth stages. 16s rRNA amplicon sequencing and data analysis tools revealed no significant differences in bacterial community composition or diversity between the cultivars. Principal component analysis revealed that differences in community structure were driven by plant growth stages rather than plant type. Polymerase chain reaction analysis was conducted to examine the potential horizontal transfer of the introduced gene from the GM maize to rhizosphere microorganisms; however, the introduced gene was not detected in the soil genomic DNA. Overall, the environmental impact of GM maize, particularly on soil microorganisms, is negligible, and the cultivation of GM maize does not alter significantly the rhizosphere bacterial community.
Collapse
Affiliation(s)
- Ye-Jin Jang
- Department of Agricultural Biotechnology, National Institute of Agricultural Sciences, Rural Development Administration, Jeonju, Korea
| | - Sung-Dug Oh
- Department of Agricultural Biotechnology, National Institute of Agricultural Sciences, Rural Development Administration, Jeonju, Korea
| | - Joon Ki Hong
- Department of Agricultural Biotechnology, National Institute of Agricultural Sciences, Rural Development Administration, Jeonju, Korea
| | - Jong-Chan Park
- Department of Agricultural Biotechnology, National Institute of Agricultural Sciences, Rural Development Administration, Jeonju, Korea
| | - Seong-Kon Lee
- Department of Agricultural Biotechnology, National Institute of Agricultural Sciences, Rural Development Administration, Jeonju, Korea
| | - Ancheol Chang
- Department of Agricultural Biotechnology, National Institute of Agricultural Sciences, Rural Development Administration, Jeonju, Korea
| | - Doh-Won Yun
- Department of Agricultural Biotechnology, National Institute of Agricultural Sciences, Rural Development Administration, Jeonju, Korea
| | - Bumkyu Lee
- Department of Environment Science & Biotechnology, Jeonju University, Jeonju, Korea
| |
Collapse
|
12
|
Nakamura K, Okamoto M, Mada T, Harada M, Okumura K, Takamatsu D. Honey-derived Paenibacillus spp. with potential to affect bee brood development in Apis mellifera: Are they a new threat to honey bees? Virulence 2025; 16:2451170. [PMID: 39954288 PMCID: PMC11834430 DOI: 10.1080/21505594.2025.2451170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/20/2024] [Accepted: 01/05/2025] [Indexed: 02/17/2025] Open
Abstract
Honey bees are important pollinators in both agriculture and ecosystems, and their health is essential for sustainable human development. Although only two bacteria, Paenibacillus larvae and Melissococcus plutonius, have been identified as bacterial pathogens in honey bee brood for over 100 years, we found three additional Paenibacillus strains (Paenibacillus sp. J27TS7, Paenibacillus azoreducens J34TS1, and Paenibacillus melissococcoides J46TS7) in honey that harmed honey bee brood development. In particular, Paenibacillus sp. J27TS7 was highly virulent in bee larvae (the median lethal dose [LD50] = 12.7 spores/larva) and was comparable to P. larvae (LD50 = 2.3-11.5 spores/larva). Paenibacillus azoreducens J34TS1 showed the second-highest virulence (LD50 = 45.9 spores/larva), and P. melissococcoides J46TS7 was the least virulent (LD50 = 469.0 spores/larva). However, P. melissococcoides was most frequently detected in Japanese honey among the three species, with the highest concentration being 1.8 × 106 spores/mL honey, suggesting its wide distribution in Japanese apiaries. The novel pathogenic Paenibacillus species were categorized into the fast killer (Paenibacillus sp. J27TS7), medium-fast killer (P. melissococcoides), and slow killer (P. azoreducens) like P. larvae strains in terms of the time to kill infected brood; however, histopathological and genome analyses indicated that their pathogenic mechanisms were different from those of P. larvae strains. Moreover, P. melissococcoides showed differences in virulence depending on the lineage of the strain. These findings represent the first discovery of honey bee brood pathogens in more than 100 years and indicate the need to look beyond known pathogens for a comprehensive understanding of honey bee diseases.
Collapse
Affiliation(s)
- Keiko Nakamura
- Research and Business Promotion Division, Research Institute for Animal Science in Biochemistry and Toxicology, Sagamihara, Kanagawa, Japan
| | - Mariko Okamoto
- Division of Infectious Animal Disease Research, National Institute of Animal Health, National Agriculture and Food Research Organization, Tsukuba, Ibaraki, Japan
| | - Takashi Mada
- Division of Infectious Animal Disease Research, National Institute of Animal Health, National Agriculture and Food Research Organization, Tsukuba, Ibaraki, Japan
| | - Mariko Harada
- Research and Business Promotion Division, Research Institute for Animal Science in Biochemistry and Toxicology, Sagamihara, Kanagawa, Japan
| | - Kayo Okumura
- Research Center for Biosafety, Laboratory Animal and Pathogen Bank, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
| | - Daisuke Takamatsu
- Division of Infectious Animal Disease Research, National Institute of Animal Health, National Agriculture and Food Research Organization, Tsukuba, Ibaraki, Japan
- The United Graduate School of Veterinary Sciences, Gifu University, Gifu, Japan
- Joint Graduate School of Veterinary Sciences, Gifu University, Gifu, Japan
| |
Collapse
|
13
|
Kempf F, Drumo R, Chaussé AM, Menanteau P, Kubasova T, Roche S, Lalmanach AC, Guabiraba R, Chaumeil T, Larivière-Gauthier G, Caballero-Posadas I, Laroche B, Rychlík I, Virlogeux-Payant I, Velge P. The immune response modulated by inoculation of commensal bacteria at birth impacts the gut microbiota and prevents Salmonella colonization. Gut Microbes 2025; 17:2474151. [PMID: 40079593 PMCID: PMC11913379 DOI: 10.1080/19490976.2025.2474151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/17/2025] [Accepted: 02/25/2025] [Indexed: 03/15/2025] Open
Abstract
Super- and low-shedding phenomena have been observed in genetically homogeneous hosts infected by a single bacterial strain. To decipher the mechanisms underlying these phenotypes, we conducted an experiment with chicks infected with Salmonella Enteritidis in a non-sterile isolator, which prevents bacterial transmission between animals while allowing the development of the gut microbiota. We investigated the impact of four commensal bacteria called Mix4, inoculated at hatching, on chicken systemic immune response and intestinal microbiota composition and functions, before and after Salmonella infection. Our results revealed that these phenotypes were not linked to changes in cell invasion capacity of bacteria during infection. Mix4 inoculation had both short- and long-term effects on immune response and microbiota and promoted the low-shedder phenotype. Kinetic analysis revealed that Mix4 activated immune response from day 4, which modified the microbiota on day 6. This change promotes a more fermentative microbiota, using the aromatic compounds degradation pathway, which inhibited Salmonella colonization by day 11 and beyond. In contrast, control animals exhibited a delayed TNF-driven pro-inflammatory response and developed a microbiota using anaerobic respiration, which facilitates Salmonella colonization and growth. This strategy offers promising opportunities to strengthen the barrier effect against Salmonella and possibly other pathogens.
Collapse
Affiliation(s)
- Florent Kempf
- ISP, INRAE, Université François Rabelais de Tours, Nouzilly, France
| | - Rosanna Drumo
- ISP, INRAE, Université François Rabelais de Tours, Nouzilly, France
| | | | | | - Tereza Kubasova
- Department of Microbiology and antimicrobial resistance, Veterinary Research Institute, Brno, Czech Republic
| | - Sylvie Roche
- ISP, INRAE, Université François Rabelais de Tours, Nouzilly, France
| | | | | | - Thierry Chaumeil
- Plate-Forme d’Infectiologie Expérimentale, INRAE, Nouzilly, France
| | | | | | | | - Ivan Rychlík
- Department of Microbiology and antimicrobial resistance, Veterinary Research Institute, Brno, Czech Republic
| | | | - Philippe Velge
- ISP, INRAE, Université François Rabelais de Tours, Nouzilly, France
| |
Collapse
|
14
|
Zhou F, Tajamul Mumtaz P, Dogan H, Madadjim R, Cui J, Zempleni J. Divergence of gut bacteria through the selection of genomic variants implicated in the metabolism of sugars, amino acids, and purines by small extracellular vesicles in milk. Gut Microbes 2025; 17:2449704. [PMID: 39762216 DOI: 10.1080/19490976.2025.2449704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 12/20/2024] [Accepted: 12/31/2024] [Indexed: 03/08/2025] Open
Abstract
Here, we report that small extracellular vesicles (sEVs) in milk mediate the communication between bacteria and animal kingdoms, increase the divergence of bacteria in the intestine, and alter metabolite production by bacteria. We show that bovine milk sEVs select approximately 55,000 genomic variants in 19 species of bacteria from the murine cecum ex vivo. The genomic variants are transcribed into mRNA. The selection of genomic variants by milk sEVs alters bacterial metabolism, leading to an up to 12-fold difference in the abundance of more than 1000 metabolites in bacteria cultured in milk sEV-free media compared to sEV-containing media. Evidence is particularly strong that selection of genomic variants by milk sEV changes the metabolism of sugars, amino acids, and purines which might contribute to the development of spatial learning and memory deficiencies and seizure phenotypes reported for milk sEV-depleted infants and mice. Human milk is a rich source of sEVs, whereas formula contains only trace amounts of milk sEVs. This report implicates nutritional sEVs in altered microbial metabolism beyond the mere selection of bacterial communities.
Collapse
Affiliation(s)
- Fang Zhou
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Peerzada Tajamul Mumtaz
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Haluk Dogan
- School of Computing, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Roland Madadjim
- School of Computing, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Juan Cui
- School of Computing, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Janos Zempleni
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA
| |
Collapse
|
15
|
Firth IJ, Sim MA, Fitzgerald BG, Moore AE, Pittao CR, Gianetto-Hill C, Hess S, Sweeney AR, Allen-Vercoe E, Sorbara MT. Urease in acetogenic Lachnospiraceae drives urea carbon salvage in SCFA pools. Gut Microbes 2025; 17:2492376. [PMID: 40231625 PMCID: PMC12001548 DOI: 10.1080/19490976.2025.2492376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 02/03/2025] [Accepted: 04/07/2025] [Indexed: 04/16/2025] Open
Abstract
The gut microbiota produces short-chain fatty acids (SCFA) and acidifies the proximal colon which inhibits enteric pathogens. However, for many microbiota constituents, how they themselves resist these stresses is unknown. The anaerobic Lachnospiraceae family, which includes the acetogenic genus Blautia, produce SCFA, are genomically diverse, and vary in their capacity to acidify culture media. Here, we investigated how Lachnospiraceae tolerate pH stress and found that subunits of urease were associated with acidification in a random forest model. Urease cleaves urea into ammonia and carbon dioxide, however the role of urease in the physiology of Lachnospiraceae is unknown. We demonstrate that urease-encoding Blautia show urea-dependent changes in SCFA production, acidification, growth, and, strikingly, urease encoding Blautia directly incorporate the carbon from urea into SCFAs. In contrast, ureolytic Klebsiella pneumoniae or Proteus mirabilis do not show the same urea-dependency or carbon salvage. In agreement, the combination of urease and acetogenesis functions is rare in gut taxa. We find that Lachnospiraceae urease and acetogenesis genes can be co-expressed in healthy individuals and colonization of mice with a ureolytic Blautia reduces urea availability in colon contents demonstrating Blautia urease activity in vivo. In human and mouse microbial communities, the acetogenic recycling of urea carbon into acetate by Blautia leads to the incorporation of urea carbon into butyrate indicating carbon salvage into broader metabolite pools. Altogether, this shows that urea plays a central role in the physiology of health-associated Lachnospiraceae which use urea in a distinct manner that is different from that of ureolytic pathogens.
Collapse
Affiliation(s)
- Isaac J. Firth
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Canada
| | - Marissa A.R. Sim
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Canada
| | | | - Ailish E. Moore
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Canada
| | - Christian R. Pittao
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Canada
| | - Connor Gianetto-Hill
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Canada
| | - Samantha Hess
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Canada
| | - Autumn R. Sweeney
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Canada
| | - Emma Allen-Vercoe
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Canada
| | - Matthew T. Sorbara
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Canada
| |
Collapse
|
16
|
Budzinski L, Kang GU, Riedel R, Sempert T, Lietz L, Maier R, Büttner J, Bochow B, Tordai MT, Shah A, Abbas A, Momtaz T, Krause JL, Kempkens R, Lehman K, Heinz GA, Benken AE, Bartsch S, Necke K, Hoffmann U, Mashreghi MF, Biesen R, Kallinich T, Alexander T, Jessen B, Weidinger C, Siegmund B, Radbruch A, Schirbel A, Moser B, Chang HD. Single-cell microbiota phenotyping reveals distinct disease and therapy-associated signatures in Crohn's disease. Gut Microbes 2025; 17:2452250. [PMID: 39815413 PMCID: PMC11740678 DOI: 10.1080/19490976.2025.2452250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 12/20/2024] [Accepted: 01/07/2025] [Indexed: 01/18/2025] Open
Abstract
IgA-coated fractions of the intestinal microbiota of Crohn's disease (CD) patients have been shown to contain taxa that hallmark the compositional dysbiosis in CD microbiomes. However, the correlation between other cellular properties of intestinal bacteria and disease has not been explored further, especially for features that are not directly driven by the host immune-system, e.g. the expression of surface sugars by bacteria. By sorting and sequencing IgA-coated and lectin-stained fractions from CD patients microbiota and healthy controls, we found that lectin-stained bacteria were distinct from IgA-coated bacteria, but still displayed specific differences between CD and healthy controls. To exploit the discriminatory potential of both, immunoglobulin coated bacteria and the altered surface sugar expression of bacteria in CD, we developed a multiplexed single cell-based analysis approach for intestinal microbiota. By multi-parameter microbiota flow cytometry (mMFC) we characterized the intestinal microbiota of 55 CD patients and 44 healthy controls for 11-parameters in total, comprising host-immunoglobulin coating and the presence of distinct surface sugar moieties. The data were analyzed by machine-learning to assess disease-specific marker patterns in the microbiota phenotype. mMFC captured detailed characteristics of CD microbiota and identified patterns to classify CD patients. In addition, we identified phenotypic signatures in the CD microbiota which not only reflected remission after 6 weeks of anti-TNF treatment, but were also able to predict remission before the start of an adalimumab treatment course in a pilot study. We here present the proof-of-concept demonstrating that multi-parameter single-cell bacterial phenotyping by mMFC could be a novel tool with high translational potential to expand current microbiome investigations by phenotyping of bacteria to identify disease- and therapy-associated cellular alterations and to reveal novel target properties of bacteria for functional assays and therapeutic approaches.
Collapse
Affiliation(s)
- Lisa Budzinski
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
- Department for Cytometry, Institute of Biotechnology, Technische Universität Berlin, Berlin, Germany
| | - Gi-Ung Kang
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
| | - René Riedel
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
- Bioinformatics and Computational Biology, Department of Cardiology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Toni Sempert
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
| | - Leonie Lietz
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
- Department for Cytometry, Institute of Biotechnology, Technische Universität Berlin, Berlin, Germany
| | - René Maier
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
| | - Janine Büttner
- Department of Hepatology and Gastroenterology, Campus Charité Mitte, Charité, Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Bettina Bochow
- Department of Hepatology and Gastroenterology, Campus Charité Mitte, Charité, Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Marcell T. Tordai
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Campus Benjamin Franklin, Charité – Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Aayushi Shah
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
- Department for Cytometry, Institute of Biotechnology, Technische Universität Berlin, Berlin, Germany
| | - Amro Abbas
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
| | - Tanisha Momtaz
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
- School of Pharmacy, BRAC University, Dhaka, Bangladesh
| | - Jannike L. Krause
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
| | - Robin Kempkens
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
| | - Katrin Lehman
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
| | - Gitta A. Heinz
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
| | - Anne E. Benken
- Department of Rheumatology, Campus Charité Mitte, Charité – Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Stefanie Bartsch
- Department of Paediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité Campus Virchow, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Kathleen Necke
- Department of Paediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité Campus Virchow, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Ute Hoffmann
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
| | - Mir-Farzin Mashreghi
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
| | - Robert Biesen
- Department of Rheumatology, Campus Charité Mitte, Charité – Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Tilmann Kallinich
- Department of Paediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité Campus Virchow, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Tobias Alexander
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
- Department of Rheumatology, Campus Charité Mitte, Charité – Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Bosse Jessen
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Campus Benjamin Franklin, Charité – Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Carl Weidinger
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Campus Benjamin Franklin, Charité – Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Britta Siegmund
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Campus Benjamin Franklin, Charité – Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- BIH Charité Clinician Scientist Program
| | - Andreas Radbruch
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
| | - Anja Schirbel
- Department of Hepatology and Gastroenterology, Campus Charité Mitte, Charité, Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Benjamin Moser
- Department of Hepatology and Gastroenterology, Campus Charité Mitte, Charité, Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- DRK Kliniken Berlin, Clinic for internal medicine – Gastroenterology, Haematology and Oncology, Nephrology, Centre for chronic gastrointestinal inflammations, Berlin, Germany
| | - Hyun-Dong Chang
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
- Department for Cytometry, Institute of Biotechnology, Technische Universität Berlin, Berlin, Germany
| |
Collapse
|
17
|
Chowdhury SR, Shilpi A, Felsenfeld G. RNA Pol-II transcripts in nucleolar associated domains of cancer cell nucleoli. Nucleus 2025; 16:2468597. [PMID: 39987497 PMCID: PMC11849958 DOI: 10.1080/19491034.2025.2468597] [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: 04/14/2023] [Revised: 01/03/2025] [Accepted: 02/13/2025] [Indexed: 02/25/2025] Open
Abstract
We performed a comparative study of the non-ribosomal gene content of nucleoli from seven cancer cell lines, using identical methods of purification and analysis. We identified unique chromosomal domains associated with the nucleolus (NADs) and genes within these domains (NAGs). Four cell lines have relatively few NAGs, which appears mostly transcriptionally inactive, consistent with literature. The remaining three lines formed a separate group with nucleoli with unique features and NADS. They constitute larger number of common NAGs, marked by ATAC-seq and having accessible promoters, with histone markers for transcriptional activity and detectable RNA Pol II bound at their promoters. The transcripts of these genes are almost entirely exported from the nucleolus. These results indicate that RNA Pol II dependent transcription in NADs can vary widely in different cell types, presumably dependent on the cell's developmental stage. Nucleolus-associated genes are likely to be distinguished marks reflecting the cell's metabolism.
Collapse
Affiliation(s)
- Soumya Roy Chowdhury
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases
| | - Arunima Shilpi
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases
| | | |
Collapse
|
18
|
Samarra A, Renwick S, Arzamasov AA, Rodionov DA, Spann K, Cabrera-Rubio R, Acuna-Gonzalez A, Martínez-Costa C, Hall L, Segata N, Osterman AL, Bode L, Collado MC. Human milk oligosaccharide metabolism and antibiotic resistance in early gut colonizers: insights from bifidobacteria and lactobacilli in the maternal-infant microbiome. Gut Microbes 2025; 17:2501192. [PMID: 40340669 PMCID: PMC12068340 DOI: 10.1080/19490976.2025.2501192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 03/22/2025] [Accepted: 04/28/2025] [Indexed: 05/10/2025] Open
Abstract
Breast milk, rich in human milk oligosaccharides (HMOs), supports the early-life colonization of beneficial bacteria such as bifidobacteria and lactobacilli, potentially reducing early-life antibiotic resistance. However, antibiotic treatment may interfere with the beneficial functions of HMO-degrading bacteria. This study investigated the metabolism of HMOs by bifidobacteria and lactobacilli isolated from human milk and mother-infant paired fecal samples, along with their antibiotic resistance profiles. Understanding these species- and sample-type-specific interactions will provide valuable insights into how bioactive components in human milk may shape the infant resistome during early life. A total of 39 Bifidobacterium and 14 Lactobacillaceae strains were isolated from paired mother-infant fecal and breast milk samples. Whole genome sequencing (WGS) allowed functional predictions on the HMO metabolism abilities and the resistance genotype of each strain. In vitro HMO utilization was assessed using growth kinetics assays combined with HMO glycoprofiling in culture supernatant. The minimum inhibitory concentration (MIC) was also determined for each strain. HMO metabolism by the bifidobacteria was species-specific. Bifidobacterium bifidum (B. bifidum) and Bifidobacterium longum subsp. infantis (B. infantis) exhibited the highest capacity for HMO degradation, consistent with genomic predictions. In contrast, lactobacilli were unable to degrade HMOs in vitro but were predicted to metabolize the by-products of HMO degradation. Phenotypic analysis revealed that B. bifidum strains had the lowest levels of antibiotic resistance, while Bifidobacterium animalis subsp. lactis (B. lactis) strains were resistant to most tested antibiotics. Overall, B. bifidum demonstrated the strongest HMO-degrading ability while remaining the most antibiotic-susceptible species. Early-life colonizing bifidobacterial species possess the essential machinery required to degrade HMOs and are highly susceptible to antibiotics. A better understanding of these dynamics could inform clinical strategies to protect and restore the infant gut microbiome, particularly in neonates exposed to antibiotics.
Collapse
Affiliation(s)
- Anna Samarra
- Department of Biotechnology, Institute of Agrochemistry and Food Technology- National Spanish Research Council (IATA-CSIC), Valencia, Spain
| | - Simone Renwick
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA, USA
- Mother-Milk-Infant Center of Research Excellence, University of California San Diego, La Jolla, CA, USA
| | - Aleksandr A. Arzamasov
- Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Dmitry A. Rodionov
- Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Kennedy Spann
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Raul Cabrera-Rubio
- Department of Biotechnology, Institute of Agrochemistry and Food Technology- National Spanish Research Council (IATA-CSIC), Valencia, Spain
| | - Antia Acuna-Gonzalez
- Food, Microbiome and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Cecilia Martínez-Costa
- Department of Pediatrics, School of Medicine, University of Valencia, Valencia, Spain
- Pediatric Gastroenterology and Nutrition Section, Hospital Clínico Universitario Valencia, Valencia, Spain
| | - Lindsay Hall
- Food, Microbiome and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
- Department of Microbes, Infection and Microbiomes, School of Infection, Inflammation and Immunology, College of Medicine and Health, University of Birmingham, Birmingham, UK
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Nicola Segata
- Department CIBIO, University of Trento, Trento, Italy
| | - Andrei L. Osterman
- Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Lars Bode
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA, USA
- Mother-Milk-Infant Center of Research Excellence, University of California San Diego, La Jolla, CA, USA
- Human Milk Institute, University of California San Diego, La Jolla, CA, USA
| | - MCarmen Collado
- Department of Biotechnology, Institute of Agrochemistry and Food Technology- National Spanish Research Council (IATA-CSIC), Valencia, Spain
| |
Collapse
|
19
|
Li P, Li M, Chen WH. Best practices for developing microbiome-based disease diagnostic classifiers through machine learning. Gut Microbes 2025; 17:2489074. [PMID: 40186338 PMCID: PMC11980492 DOI: 10.1080/19490976.2025.2489074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 03/13/2025] [Accepted: 03/28/2025] [Indexed: 04/07/2025] Open
Abstract
The human gut microbiome, crucial in various diseases, can be utilized to develop diagnostic models through machine learning (ML). The specific tools and parameters used in model construction such as data preprocessing, batch effect removal and modeling algorithms can impact model performance and generalizability. To establish an generally applicable workflow, we divided the ML process into three above-mentioned steps and optimized each sequentially using 83 gut microbiome cohorts across 20 diseases. We tested a total of 156 tool-parameter-algorithm combinations and benchmarked them according to internal- and external- AUCs. At the data preprocessing step, we identified four data preprocessing methods that performed well for regression-type algorithms and one method that excelled for non-regression-type algorithms. At the batch effect removal step, we identified the "ComBat" function from the sva R package as an effective batch effect removal method and compared the performance of various algorithms. Finally, at the ML algorithm selection step, we found that Ridge and Random Forest ranked the best. Our optimized work flow performed similarly comparing with previous exhaustive methods for disease-specific optimizations, thus is generally applicable and can provide a comprehensive guideline for constructing diagnostic models for a range of diseases, potentially serving as a powerful tool for future medical diagnostics.
Collapse
Affiliation(s)
- Peikun Li
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular Imaging, Center for Artificial Intelligence Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Min Li
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular Imaging, Center for Artificial Intelligence Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wei-Hua Chen
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular Imaging, Center for Artificial Intelligence Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
- School of Biological Science, Jining Medical University, Rizhao, China
| |
Collapse
|
20
|
Li Q, Tan D, Xiong S, Yu K, Su Y, Zhu W. Time-restricted feeding promotes glucagon-like peptide-1 secretion and regulates appetite via tryptophan metabolism of gut Lactobacillus in pigs. Gut Microbes 2025; 17:2467185. [PMID: 39951352 PMCID: PMC11834429 DOI: 10.1080/19490976.2025.2467185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 01/08/2025] [Accepted: 02/10/2025] [Indexed: 02/16/2025] Open
Abstract
Previous clinical trials have shown that time-restricted feeding can be involved in regulating the metabolic health of humans and animals. However, the underlying mechanism has not been fully explored. In this study, the pig model was employed to simulate four prevalent human eating habits, with the aim of investigating the impact of gut microbiota and microbial metabolites on gut hormone secretion and appetite regulation. Compared to the ad libitum feeding (ALF) pattern, three time-restricted feeding patterns reduced total food intake and eating time. Meanwhile, three time-restricted feeding patterns induced elevated levels of serum and hypothalamic glucagon-like peptide-1 (GLP-1), while suppressing reward-related circuits in the hypothalamus. It is noteworthy that the early time-restricted feeding (eTRF) pattern increased the number of intestinal enteroendocrine cells (EECs) compared to ALF. Metagenomic and metabonomic analyses revealed that three time-restricted feeding patterns induced colonization of Lactobacillus and significantly increased the levels of its metabolite, indole-3-lactic acid (ILA). Dietary supplementation with ILA exhibited an increasing trend in fasting serum GLP-1 level of piglets. In vitro studies with pig intestinal organoids showed the Lactobacillus metabolite ILA enhanced GLP-1 secretion through the promotion of intestinal stem cell differentiation into EECs, rather than activating the ability of EECs to secrete GLP-1. Overall, time-restricted feeding promoted GLP-1 secretion and affected long-term appetite regulation by promoting the colonization of Lactobacillus and modulating microbial tryptophan metabolism.
Collapse
Affiliation(s)
- Qiuke Li
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
| | - Ding Tan
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
| | - Shijie Xiong
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
| | - Kaifan Yu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
| | - Yong Su
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
| | - Weiyun Zhu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
21
|
Hamo CE, Muller M, Rosenfeld E, Xia Y, Akinlonu A, Luttrell-Williams E, Barrett TJ, Berger JS. Cardiometabolic risk factor burden associates with an immature platelet profile. Platelets 2025; 36:2459800. [PMID: 39882733 PMCID: PMC11801799 DOI: 10.1080/09537104.2025.2459800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 01/06/2025] [Accepted: 01/21/2025] [Indexed: 01/31/2025]
Abstract
Cardiometabolic risk factors, obesity, diabetes and hyperlipidemia contribute to cardiovascular disease (CVD). While platelets are involved in CVD pathogenesis, the relationship between risk factor burden on platelet indices and the platelet transcriptome remains uncertain. Blood was collected from CVD-free adults, measuring platelet count, mean platelet volume (MPV), immature platelet fraction (IPF), and absolute immature platelet fraction (AIPF) by hemogram. Platelets were isolated and analyzed via RNA sequencing. Participants were stratified by number of cardiometabolic risk factors (diabetes, obesity, hyperlipidemia). We calculated median (IQR) values of platelet indices and p-for-trend via linear regression across risk factor burden. To evaluate the association between risk factor burden and platelet transcripts, we performed multivariable linear regression adjusting for age, sex, and race/ethnicity. Among 141 participants, (50.5 ± 14.8 years, 42% male, 26% Black) risk factor burden was associated with increasing platelet size, IPF, and AIPF but not platelet count. Platelet RNA sequencing identified 100 differentially expressed transcripts (p < .01; 66 upregulated, 34 downregulated). Gene ontology enrichment analysis demonstrated upregulated pathways of secondary metabolic processes (NES = 1.96, p < .01), and hematopoietic stem cell proliferation (NES = 1.95, p < .01). Greater cardiometabolic risk factor burden is associated with increased platelet size and immaturity and suggesting novel platelet-mediated mechanisms linking risk factor burden with CVD.
Collapse
Affiliation(s)
- Carine E. Hamo
- Department of Medicine, Center for the Prevention of Cardiovascular Disease, New York University School of Medicine, New York City, New York, USA
- Leon H. Charney Division of Cardiology, Department of Medicine, Cardiovascular Research Center, New York University School of Medicine, New York City, New York, USA
| | - Matthew Muller
- Leon H. Charney Division of Cardiology, Department of Medicine, Cardiovascular Research Center, New York University School of Medicine, New York City, New York, USA
| | - Emily Rosenfeld
- Leon H. Charney Division of Cardiology, Department of Medicine, Cardiovascular Research Center, New York University School of Medicine, New York City, New York, USA
| | - Yuhe Xia
- Leon H. Charney Division of Cardiology, Department of Medicine, Cardiovascular Research Center, New York University School of Medicine, New York City, New York, USA
| | - Adedoyin Akinlonu
- Leon H. Charney Division of Cardiology, Department of Medicine, Cardiovascular Research Center, New York University School of Medicine, New York City, New York, USA
| | - Elliot Luttrell-Williams
- Leon H. Charney Division of Cardiology, Department of Medicine, Cardiovascular Research Center, New York University School of Medicine, New York City, New York, USA
| | - Tessa J. Barrett
- Leon H. Charney Division of Cardiology, Department of Medicine, Cardiovascular Research Center, New York University School of Medicine, New York City, New York, USA
| | - Jeffrey S. Berger
- Department of Medicine, Center for the Prevention of Cardiovascular Disease, New York University School of Medicine, New York City, New York, USA
- Leon H. Charney Division of Cardiology, Department of Medicine, Cardiovascular Research Center, New York University School of Medicine, New York City, New York, USA
| |
Collapse
|
22
|
Jansen D, Deleu S, Caenepeel C, Marcelis T, Simsek C, Falony G, Machiels K, Sabino J, Raes J, Vermeire S, Matthijnssens J. Virome drift in ulcerative colitis patients: faecal microbiota transplantation results in minimal phage engraftment dominated by microviruses. Gut Microbes 2025; 17:2499575. [PMID: 40371968 DOI: 10.1080/19490976.2025.2499575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 04/02/2025] [Accepted: 04/24/2025] [Indexed: 05/16/2025] Open
Abstract
Ulcerative colitis (UC) is an inflammatory bowel disease characterized by recurrent colonic inflammation. Standard treatments focus on controlling inflammation but remain ineffective for one-third of patients. This underscores the need for alternative approaches, such as fecal microbiota transplantation (FMT), which transfers healthy donor microbiota to patients. The role of viruses in this process, however, remains underexplored. To address this, we analyzed the gut virome using metagenomic sequencing of enriched viral particles from 320 longitudinal fecal samples of 44 patients enrolled in the RESTORE-UC FMT trial. Patients were treated with FMTs from healthy donors (allogenic, treatment) or themselves (autologous, control). We found that colonic inflammation, both its presence and location, had a greater impact on the gut virome than FMT itself. In autologous FMT patients, the virome was unstable and showed rapid divergence over time, a phenomenon we termed virome drift. In allogenic FMT patients, the virome temporarily shifted toward the healthy donor, lasting up to 5 weeks and primarily driven by microviruses. Notably, two distinct virome configurations were identified and linked to either healthy donors or patients. In conclusion, inflammation strongly affects the gut virome in UC patients, which may lead to instability and obstruct the engraftment of allogeneic FMT.
Collapse
Affiliation(s)
- Daan Jansen
- Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Viral Metagenomics, KU Leuven, Leuven, Belgium
| | - Sara Deleu
- Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, University Hospitals Leuven, Leuven, Belgium
| | - Clara Caenepeel
- Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, University Hospitals Leuven, Leuven, Belgium
| | - Tine Marcelis
- Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Viral Metagenomics, KU Leuven, Leuven, Belgium
| | - Ceren Simsek
- Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Viral Metagenomics, KU Leuven, Leuven, Belgium
| | - Gwen Falony
- Department of Microbiology Immunology and Transplantation, Rega Institute, Laboratory of Molecular Bacteriology, KU Leuven, Leuven, Belgium
- Center for Microbiology, VIB, Leuven, Belgium
- Institute of Medical Microbiology and Hygiene and Research Centre for Immunotherapy (FZI), University Medical Centre of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Kathleen Machiels
- Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, University Hospitals Leuven, Leuven, Belgium
| | - João Sabino
- Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, University Hospitals Leuven, Leuven, Belgium
| | - Jeroen Raes
- Department of Microbiology Immunology and Transplantation, Rega Institute, Laboratory of Molecular Bacteriology, KU Leuven, Leuven, Belgium
- Center for Microbiology, VIB, Leuven, Belgium
| | - Séverine Vermeire
- Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, University Hospitals Leuven, Leuven, Belgium
| | - Jelle Matthijnssens
- Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Viral Metagenomics, KU Leuven, Leuven, Belgium
| |
Collapse
|
23
|
Brugnoni R, Salvi E, Moresco E, Gallone A, Tufano L, Garibaldi M, Filosto M, Grandis M, Maggi L. Pharmacogenetic pilot study of CYP2D6 and CYP1A2 genes in Italian patients with non-dystrophic myotonia and myotonic dystrophy treated with mexiletine. Gene 2025; 960:149536. [PMID: 40324568 DOI: 10.1016/j.gene.2025.149536] [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: 10/28/2024] [Revised: 04/16/2025] [Accepted: 04/28/2025] [Indexed: 05/07/2025]
Abstract
Mexiletine (MXT) is the first-line anti-myotonic drug for myotonic dystrophies (MD) and non-dystrophic myotonias (NDM), metabolized by CYP2D6 and CYP1A2 enzymes. We investigated genetic variants in these genes and their influence on MXT response in Italian MD and NDM patients. Fifty patients (MD: 34, NDM: 16) were treated with MXT (200-600 mg daily) for at least two months. Based on the Myotonic Behaviour Scale (MBS) and neurological examination, 37 patients (74 %) were classified as responders (R), while 13 (26 %) were non-responders (NR). Comparison between R and NR revealed associations with 14 genetic variants (12 in CYP2D6, 2 in CYP1A2). In silico analysis suggested eQTL effects on liver and skeletal muscle gene expression. Functional annotation indicated the regulatory roles of these variants. The CYP2D6*2/*41 diplotype showed a nominal association with non-response (OR = 10.8, p = 0.049), being 11 times more frequent in NR (23 %) than in R (3 %). Most common diplotypes (CYP2D6*1/*2, *1/*1, *1/*10, *2/10) corresponded to Normal Metabolizers, while CYP2D6*10/*10, *10/*41, and *41/41 indicated intermediate metabolism, suggesting a higher risk of adverse reactions with concurrent drugs. The CYP2D6*41 allele was more frequent in NR patients than in the European population, supporting its role in MXT response variability. Our findings suggest CYP2D6 and CYP1A2 variants as potential predictors of MXT treatment response in MD and NDM patients.
Collapse
Affiliation(s)
- Raffaella Brugnoni
- Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy.
| | - Erika Salvi
- Computational multi-Omics of Neurological Disorders (MIND) Lab, Joint Research Platform, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy.
| | - Elisa Moresco
- Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy; Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy.
| | - Annamaria Gallone
- Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy; Neurophysiology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy.
| | - Laura Tufano
- Neuromuscular and Rare Disease Centre, Department of Neuroscience, Mental Health and Sensory Organs (NESMOS), SAPIENZA University of Rome, Sant'Andrea Hospital, Rome, Italy.
| | - Matteo Garibaldi
- Neuromuscular and Rare Disease Centre, Department of Neuroscience, Mental Health and Sensory Organs (NESMOS), SAPIENZA University of Rome, Sant'Andrea Hospital, Rome, Italy.
| | - Massimiliano Filosto
- Department of Clinical and Experimental Sciences, University of Brescia, NeMO-Brescia Clinical Center for Neuromuscular Diseases, Brescia, Italy.
| | - Marina Grandis
- Department of Neuroscience, Ophthalmology, Rehabilitation, Genetics and Maternal and Child Sciences, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genova, Italy.
| | - Lorenzo Maggi
- Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy.
| |
Collapse
|
24
|
Zhang X, Amradi NK, Moore M, Hassanein A, Mickol RL, McCoy EL, Eddie BJ, Shepard JS, Wang J, Lansing S, Yates MD, Kim YT, Wang ZW. Enhancement of 3-hydroxyvalerate fraction in poly(3-hydroxybutyrate-co-3-hydroxyvalerate) produced by Haloferax mediterranei fed with food waste pretreated via arrested anaerobic digestion integrated with microbial electrolysis cells. BIORESOURCE TECHNOLOGY 2025; 430:132536. [PMID: 40228720 DOI: 10.1016/j.biortech.2025.132536] [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: 01/08/2025] [Revised: 04/10/2025] [Accepted: 04/11/2025] [Indexed: 04/16/2025]
Abstract
Bioplastics made of poly(3-hydroxybutyrate-co-3-hydroxyvalerate) (PHBV) with a 20 mol% HV fraction are highly desirable in the market for 3-Hydroxyvalerate (HV)-conferred superior thermal, biological, and mechanical properties. Although Haloferax mediterranei (HM) is capable of producing PHBV from food waste, its HV fraction is generally lower than 10 mol%. This study for the first time investigated the engineering approach to increasing HV fraction through elevating the propionic and valeric acid fractions in volatile fatty acids (VFAs) produced from food waste via arrested anaerobic digestion with and without microbial electrolysis cells (MECs) incorporation. Results showed that HV fraction in PHBV produced by HM is proportional to the fractions of propionic and valeric acids in VFAs. A 20 mol% HV fraction can be achieved by MECs incorporation, which might be attributable to pH regulation by the MECs. These findings lay a foundation for developing waste-processing technologies that enable the production of high-value, microbially-derived materials.
Collapse
Affiliation(s)
- Xueyao Zhang
- Department of Biological System Engineering, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Naresh Kumar Amradi
- Department of Environmental Science and Technology, University of Maryland, College Park, MD 20742, USA
| | - Martin Moore
- Center for Bio/Molecular Science and Engineering, Naval Research Laboratory, Washington, DC 20375, USA
| | - Amro Hassanein
- Department of Environmental Science and Technology, University of Maryland, College Park, MD 20742, USA; Yokogawa Corporation of America, Sugar Land, TX 77478, USA
| | - Rebecca L Mickol
- Center for Bio/Molecular Science and Engineering, Naval Research Laboratory, Washington, DC 20375, USA
| | - Emily L McCoy
- Department of Environmental Science and Technology, University of Maryland, College Park, MD 20742, USA
| | - Brian J Eddie
- Center for Bio/Molecular Science and Engineering, Naval Research Laboratory, Washington, DC 20375, USA
| | - Jamia S Shepard
- Department of Environmental Science and Technology, University of Maryland, College Park, MD 20742, USA
| | - Jiefu Wang
- Department of Biological System Engineering, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Stephanie Lansing
- Department of Environmental Science and Technology, University of Maryland, College Park, MD 20742, USA
| | - Matthew D Yates
- Center for Bio/Molecular Science and Engineering, Naval Research Laboratory, Washington, DC 20375, USA
| | - Young-Teck Kim
- Department of Sustainable Biomaterials, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Zhi-Wu Wang
- Department of Biological System Engineering, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA.
| |
Collapse
|
25
|
Wang S, Wang Q, Xu J, Yu J, Wang S. In vitro fecal fermentation characteristics of starch-chitosan composite prepared by screw extrusion. Carbohydr Polym 2025; 361:123657. [PMID: 40368569 DOI: 10.1016/j.carbpol.2025.123657] [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: 10/16/2024] [Revised: 04/08/2025] [Accepted: 04/24/2025] [Indexed: 05/16/2025]
Abstract
In this study, we prepared a series of composites with various chitosan-to-starch ratios by means of screw extrusion. The morphology and structural order of the composite were examined, and the fermentability was investigated through an in vitro human fecal fermentation model. Fourier transform infrared (FTIR) spectroscopy demonstrated the formation of hydrogen bonds between starch and chitosan in the composite, and X-ray diffraction (XRD) analysis indicated the structural order of the composite was enhanced with increasing chitosan proportions. The addition of chitosan slowed down the pH drop rate during the fermentation, and high chitosan proportion decreased the production of short-chain fatty acids (SCFAs), especially butyrate. The final butyrate concentrations of the composite with chitosan to starch ratio of 1:6 (CS:S = 1:6, 0.97 mmol/g) was almost twice that of the composite with chitosan to starch ratio of 2:1 (CS:S = 2:1, 0.51 mmol/g). Health-promoting microbes including Agathobacter, Prevotella_9, Ruminococcus, and Bifidobacterium were obviously promoted by extruded starch. Moreover, the composite sample with chitosan to starch ratio of 1:6 (CS:S = 1:6) exhibited a good synergistic effect in regulating the microbiota composition, reflected by the increase in the relative abundance of Agathobacter and Prevotella_9. This finding demonstrated that the ratio of starch to chitosan might be a controlling factor in determining the fermentation properties of screw extruded composite.
Collapse
Affiliation(s)
- Shaokang Wang
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin 300457, China; College of Food Science and Engineering, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Qian Wang
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin 300457, China; College of Food Science and Engineering, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Jinchuan Xu
- Key Laboratory of Smart Breeding (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Tianjin Agricultural University, Tianjin 300392, China
| | - Jinglin Yu
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Shujun Wang
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin 300457, China; College of Food Science and Engineering, Tianjin University of Science & Technology, Tianjin 300457, China.
| |
Collapse
|
26
|
Dettleff P, Palomino J, González-Coppia F, De Los Reyes M. Transcriptomic profiling of miRNA-mRNA interactions in canine oocytes and cumulus cells during in vitro maturation: a sequencing analysis. Theriogenology 2025; 242:117454. [PMID: 40288148 DOI: 10.1016/j.theriogenology.2025.117454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 04/19/2025] [Accepted: 04/20/2025] [Indexed: 04/29/2025]
Abstract
Oocyte maturation is a critical process for successful fertilization and early embryonic development. In this study, we investigated the molecular mechanisms underlying oocyte maturation in dogs by analyzing the transcriptomic profiles of miRNAs and mRNAs in canine cumulus cells (CCs) and oocytes during in vitro maturation (IVM). RNA sequencing identified 285 miRNAs expressed in oocytes and 310 in CCs, with 282 miRNAs shared between the two cell types, highlighting the role of intercellular communication in maintaining miRNA expression equilibrium. Differential expression analysis revealed 222 mRNAs with significant differences between CCs and oocytes, including genes involved in transcriptional regulation and nuclear structure. Enrichment analyses identified pathways such as actin cytoskeleton regulation, mTOR signaling, cAMP signaling, and calcium signaling, all critical to oocyte maturation. Network analysis revealed 643 significant miRNA-mRNA coexpression relationships, suggesting miRNAs play pivotal roles in regulating mRNA expression during oocyte maturation. Notably, key miRNAs such as miR-30b, miR-375, and miR-503 were implicated in regulating genes involved in oocyte maturation pathways, while others like miR-378 and miR-21 aligned with known roles in suppressing cumulus expansion and influencing maturation. The absence of differential miRNA expression between CCs and oocytes suggests the miRNA transfer through gap junctions. These findings provide new insights into the transcriptional and post-transcriptional regulation of oocyte maturation in dogs, offering valuable knowledge to improve reproductive biotechnologies such as in vitro fertilization and embryo development in this species.
Collapse
Affiliation(s)
- Phillip Dettleff
- School of Veterinary Medicine, Faculty of Agronomy and Natural Systems, Faculty of Biological Sciences and Faculty of Medicine, Pontifical Catholic University of Chile. Santiago, Chile
| | - Jaime Palomino
- School of Veterinary Medicine, Center for Health and Society Research Studies, Bernardo O'Higgins University, Santiago, Chile; Laboratory of Animal Reproduction, Faculty of Veterinary Sciences, University of Chile, Santiago, Chile
| | - Fabiola González-Coppia
- Laboratory of Animal Reproduction, Faculty of Veterinary Sciences, University of Chile, Santiago, Chile
| | - Monica De Los Reyes
- Laboratory of Animal Reproduction, Faculty of Veterinary Sciences, University of Chile, Santiago, Chile.
| |
Collapse
|
27
|
Ma F, Zou Q, Zhao X, Liu H, Du H, Xing K, Ding X, Wang C. Multi-omics integration reveals the regulatory mechanisms of APC and CREB5 genes in lipid biosynthesis and fatty acid composition in pigs. Food Chem 2025; 482:143999. [PMID: 40187300 DOI: 10.1016/j.foodchem.2025.143999] [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: 10/03/2024] [Revised: 03/13/2025] [Accepted: 03/20/2025] [Indexed: 04/07/2025]
Abstract
The intramuscular fat (IMF), fatty acid and amino acid compositions of pork are intricately linked to meat quality, flavor profile, and nutritional composition, and have potential implications for human health. Lipid accumulation in pork is initiated by the biosynthesis of fatty acids and regulated by a complex network of genes. In this study, the IMF content and genotyping of large-scale slaughtered Yorkshire pigs were assessed. Transcriptome sequencing of muscles from 17 individuals and fatty and amino acid analyses of muscles from 28 individuals according to IMF content were conducted. Phenotypic analysis showed a high correlation between IMF and most fatty acids, and the composition ratio of different types of fatty acids varied with IMF content. A negative correlation between the n-6/n-3 polyunsaturated fatty acid (PUFA) ratio and increase in IMF content significantly enhanced the levels of essential fatty acids and ameliorated the n-6/n-3 PUFA ratio in pork, thereby elevating its nutritional value to better align with contemporary health standards. A comprehensive analysis that integrated a genome-wide association study, differential gene expression analysis, and weighted gene co-expression network analysis was employed to identify the regulatory mechanisms of lipids. PRLR, SEC11C, ALPK2, CPLX4, APC, and CREB5 were identified as key candidate genes that affect intramuscular lipids and fatty acids. Through molecular and cellular experiments, our results indicated that high APC and CREB5 gene expression significantly promotes lipogenesis in cells, where these genes play an important role in regulatory pathways related to lipid synthesis in animals, which may affect fat deposition and fatty acid composition in pork. Overall, these results lay the foundation for an in-depth analysis of the genetic regulation of pork lipids and nutrition, and also provide molecular regulatory markers for the primary selection of pigs with better meat quality.
Collapse
Affiliation(s)
- Fuping Ma
- State Key Laboratory of Animal Biotech Breeding; National Engineering Laboratory for Animal Breeding; Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs; College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Quan Zou
- State Key Laboratory of Animal Biotech Breeding; National Engineering Laboratory for Animal Breeding; Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs; College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Xitong Zhao
- Beijing Shunxin Agriculture Co., Ltd., Beijing 101300, China
| | - Huatao Liu
- State Key Laboratory of Animal Biotech Breeding; National Engineering Laboratory for Animal Breeding; Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs; College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Hehe Du
- State Key Laboratory of Animal Biotech Breeding; National Engineering Laboratory for Animal Breeding; Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs; College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Kai Xing
- State Key Laboratory of Animal Biotech Breeding; National Engineering Laboratory for Animal Breeding; Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs; College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Xiangdong Ding
- State Key Laboratory of Animal Biotech Breeding; National Engineering Laboratory for Animal Breeding; Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs; College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Chuduan Wang
- State Key Laboratory of Animal Biotech Breeding; National Engineering Laboratory for Animal Breeding; Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs; College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
28
|
Kakar FL, Aqeel H, Okoye F, Elbeshbishy E, Liss SN. Microbial shifts and VFA production in the optimization of anaerobic digestion by thermal hydrolysis coupled with vacuum fermentation. BIORESOURCE TECHNOLOGY 2025; 429:132481. [PMID: 40187500 DOI: 10.1016/j.biortech.2025.132481] [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: 03/28/2024] [Revised: 03/31/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
This study investigated a novel thermal-hydrolysis combined with a vacuum fermentation system for high-grade volatile fatty acids (VFA) recovery, and the corresponding changes in the microbial community. Four systems with and without hydrothermal pre-treatment (HTP) and vacuum were mobilized; results revealed that integration of HTP with vacuum has the highest potential in terms of VFA recovery, sludge disintegration, and solid reduction. HTP and vacuum fermentation systems were associated with the highest COD solubilization (45 %), and VFA yield (0.32 g COD/g VSS added). Vacuum fermenters with and without pre-treatment have the highest specific denitrification rates of 7.6 and 7.2 mg NO3-N/g VSS.h, respectively, compared to all other samples and control (acetate). Changes brought about by vacuum fermentation included a shift in the microbial community toward enriching fermenters, mainly Caprothermobacteria and Thermotagea, responsible for VFA production.
Collapse
Affiliation(s)
- Farokh Laqa Kakar
- Environmental Research Group for Resource Recovery, Department of Civil Engineering, Faculty of Engineering, Architecture and Science, Toronto Metropolitan University, 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
| | - Hussain Aqeel
- Department of Chemistry and Biology, Faculty of Science, Toronto Metropolitan University 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
| | - Frances Okoye
- Environmental Research Group for Resource Recovery, Department of Civil Engineering, Faculty of Engineering, Architecture and Science, Toronto Metropolitan University, 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
| | - Elsayed Elbeshbishy
- Environmental Research Group for Resource Recovery, Department of Civil Engineering, Faculty of Engineering, Architecture and Science, Toronto Metropolitan University, 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
| | - Steven N Liss
- Department of Chemistry and Biology, Faculty of Science, Toronto Metropolitan University 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada; School of Environmental Studies, Queen's University, Kingston, ON K7L 3N6, Canada; Department of Microbiology, Stellenbosch University, Private Bag, XI, Matieland, 7602 Stellenbosch, South Africa.
| |
Collapse
|
29
|
Contreras P, Fica-León V, Navarrete J, Oviedo C. De novo transcriptome assembly and functional annotation supports potential biotechnological applications for the non-model thraustochytrid Ulkenia visurgensis Lng2. Gene 2025; 958:149492. [PMID: 40228758 DOI: 10.1016/j.gene.2025.149492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/19/2025] [Accepted: 04/10/2025] [Indexed: 04/16/2025]
Abstract
Thraustochytrids are heterotrophic marine protists known for their ability to produce valuable lipids such as docosahexaenoic acid (DHA). However, like many non-model organisms, they present challenges for transcriptomic studies due to the limited reliable reference genomes and compatibility with curated protein databases, complicating the respective assembly and annotation. This study presents a de novo transcriptome assembly and functional annotation for the recently isolated thraustochytrid strain Ulkenia visurgensis Lng2 using solely free access software and code available in public domain repositories. The assembled transcriptome presented 45,867 unique gene models, with a total of 66,623 transcripts and a contig N50 of 3162. Functional annotations highlighted high amounts of transcripts related to the biosynthesis of relevant lipidic molecules, as well as stress-adaptive features including catalytic and xenobiotic degrading activity. Likewise, 2381 transcripts were linked to enzymes with potential biotechnological applications. Notably, several transcripts corresponding to uncommon enzymatic activities in thraustochytrids, including laccases, cellulases, and chitinases, were identified. Additionally, evidence for a potential lactamase activity was found, marking the first report of such activity in thraustochytrids. Overall, this study offers a simple free-access procedural strategy for a de novo transcriptome assembly and functional annotation in non-model organisms. These results provide valuable insights into the biotechnological potential of thraustochytrids, while also expanding the limited transcriptomic data available for these protists. Notably, it represents the first transcriptomic analysis of the Ulkenia genus.
Collapse
Affiliation(s)
- Pedro Contreras
- Departamento de Ingeniería en Maderas, Facultad de Ingeniería, Universidad del Bio-Bio, Concepción, Chile.
| | - Víctor Fica-León
- Facultad de Ingeniería, Arquitectura y Diseño, Universidad San Sebastián, Concepción, Chile.
| | - José Navarrete
- Departamento de Ingeniería en Maderas, Facultad de Ingeniería, Universidad del Bio-Bio, Concepción, Chile.
| | - Claudia Oviedo
- Departamento de Química, Facultad de Ciencias, Universidad del Bío-Bío, Concepción, Chile.
| |
Collapse
|
30
|
Weinrauch AM, Kwan GT, Giacomin M, Bouyoucos IA, Tresguerres M, Goss GG. Evolutionary insights into gut acidification: invertebrate-like mechanisms in the basal vertebrate hagfish. J Exp Biol 2025; 228:jeb249641. [PMID: 39882670 DOI: 10.1242/jeb.249641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 01/21/2025] [Indexed: 01/31/2025]
Abstract
Acidification is a key component of digestion throughout metazoans. The gut digestive fluid of many invertebrates is acidified by the vesicular-type H+-ATPase (VHA). In contrast, vertebrates generate acidic gut fluids using the gastric H+/K+-ATPase (HKA), an evolutionary innovation linked with the appearance of a true stomach that greatly improves digestion, absorption and immune function. Hagfishes are the most basal extant vertebrates, and their mechanism of digestive acidification remains unclear. Herein, we report that the stomachless Pacific hagfish (Eptatretus stoutii) acidify their gut using the VHA, and searches of E. stoutii gut transcriptomes and the genome of a closely related hagfish species (E. burgerii) indicate they lack HKA, consistent with its emergence following the 2R whole-genome duplication. Immunostaining revealed prominent VHA presence in the apical membrane of enterocytes and sub-apical expression of both VHA and soluble adenylyl cyclase. Interestingly, akin to vertebrates, VHA was also observed in immature pancreatic-like zymogen granules and was noticeably absent from the mature granules. Furthermore, isolated gut sacs from fed hagfish demonstrate increased VHA-dependent luminal H+ secretion that is stimulated by the cAMP pathway. Overall, these results suggest that the hagfish gut shares the trait of VHA-dependent acidification with invertebrates, while simultaneously performing some roles of the pancreas and intestine of gnathostomes.
Collapse
Affiliation(s)
- Alyssa M Weinrauch
- Department of Biological Sciences, University of Alberta, 116 St and 85 Ave, Edmonton, AB, Canada, T6G 2R3
- Bamfield Marine Sciences Centre, 100 Pachena Rd, Bamfield, BC, Canada, V0R 1B0
| | - Garfield T Kwan
- Marine Biology Research Division, Scripps Institution of Oceanography, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Marina Giacomin
- Department of Biological Sciences, University of Alberta, 116 St and 85 Ave, Edmonton, AB, Canada, T6G 2R3
- Bamfield Marine Sciences Centre, 100 Pachena Rd, Bamfield, BC, Canada, V0R 1B0
| | - Ian A Bouyoucos
- Department of Biological Sciences, University of Manitoba, Winnipeg, MB, Canada, R3T 2N2
| | - Martin Tresguerres
- Marine Biology Research Division, Scripps Institution of Oceanography, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Greg G Goss
- Department of Biological Sciences, University of Alberta, 116 St and 85 Ave, Edmonton, AB, Canada, T6G 2R3
- Bamfield Marine Sciences Centre, 100 Pachena Rd, Bamfield, BC, Canada, V0R 1B0
| |
Collapse
|
31
|
Hayashi M, Koga Y, Kozono Y, Kobayashi S. Maternal ovo represses the expression of transposable elements in adult ovaries. Dev Biol 2025; 523:111-114. [PMID: 40280385 DOI: 10.1016/j.ydbio.2025.04.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 04/20/2025] [Accepted: 04/23/2025] [Indexed: 04/29/2025]
Abstract
In germ cells, repressing transposable elements (TEs) is important to maintain genomic integrity. TE expression and transposition are repressed by PIWI-interacting RNAs (piRNAs). Although many genes for piRNA synthesis have been described, few transcription factors activating their expression have been identified. We previously reported that a transcription factor, maternal Ovo (Ovo-B) protein activates germline-specific gene expression in progenitors of germ cells. In this study, we found that maternal Ovo also activates several genes, including aubergine (aub), for TE silencing. Knocking down maternal Ovo de-repressed TEs in adult ovaries. In addition, embryonic knockdown of aub caused de-repression of TEs in adult Drosophila ovaries. Surprisingly, embryonic knockdown of maternal Ovo affected neither expression of ovo nor its downstream TE-silencing genes in adult ovaries after growth. These results strongly suggest that maternal Ovo is required for TE silencing in ovaries, via transcriptional activation of genes for piRNA synthesis in embryos.
Collapse
Affiliation(s)
- Makoto Hayashi
- Institute for Aquaculture Biotechnology (IAB), Tokyo University of Marine Science and Technology, 4-5-7, Konan, Minato-ku, Tokyo, 108-8477, Japan; Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki, 305-8577, Japan; Degree Programs in Life and Earth Sciences, Graduate School of Science and Technology, University of Tsukuba, Tsukuba, Ibaraki, Japan.
| | - Yuica Koga
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, 113-0032, Japan
| | - Yasuhiro Kozono
- Degree Programs in Life and Earth Sciences, Graduate School of Science and Technology, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Satoru Kobayashi
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki, 305-8577, Japan; Degree Programs in Life and Earth Sciences, Graduate School of Science and Technology, University of Tsukuba, Tsukuba, Ibaraki, Japan.
| |
Collapse
|
32
|
Lee HK, Chen J, Philips RL, Lee SG, Feng X, Wu Z, Liu C, Schultz AB, Dalzell M, Meggendorfer M, Haferlach C, Birnbaum F, Sexton JA, Keating AE, O'Shea JJ, Young NS, Villarino AV, Furth PA, Hennighausen L. STAT5B leukemic mutations, altering SH2 tyrosine 665, have opposing impacts on immune gene programs. Life Sci Alliance 2025; 8:e202503222. [PMID: 40228864 PMCID: PMC11999048 DOI: 10.26508/lsa.202503222] [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: 01/14/2025] [Revised: 03/31/2025] [Accepted: 03/31/2025] [Indexed: 04/16/2025] Open
Abstract
STAT5B is a vital transcription factor for lymphocytes. Here, the function of two STAT5B mutations from human T-cell leukemias: one substituting tyrosine 665 with phenylalanine (STAT5BY665F) and the other with histidine (STAT5BY665H), was interrogated. In silico modeling predicted divergent energetic effects on homodimerization with a range of pathogenicity. In primary T cells in vitro, STAT5BY665F showed gain-of-function, whereas STAT5BY665H demonstrated loss-of-function. Introducing the mutation into the mouse genome illustrated that the gain-of-function Stat5b Y665F mutation resulted in accumulation of CD8+ effector and memory and CD4+ regulatory T cells, altering CD8+/CD4+ ratios. In contrast, STAT5BY665H "knock-in" mice showed diminished CD8+ effector and memory and CD4+ regulatory T cells. In contrast to WT STAT5B, the STAT5BY665F variant displayed greater STAT5 phosphorylation, DNA binding, and transcriptional activity after cytokine activation, whereas the STAT5BY665H variant resembled a null. The work exemplifies how joining in silico and in vivo studies of single nucleotides deepens our understanding of disease-associated variants, revealing structural determinants of altered function, defining mechanistic roles, and, specifically here, identifying a gain-of-function variant that does not directly induce hematopoietic malignancy.
Collapse
Affiliation(s)
- Hye Kyung Lee
- Laboratory of Genetics and Physiology, National Institute of Diabetes and Digestive and Kidney Diseases, US National Institutes of Health, Bethesda, MD, USA
| | - Jichun Chen
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Rachael L Philips
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sung-Gwon Lee
- Laboratory of Genetics and Physiology, National Institute of Diabetes and Digestive and Kidney Diseases, US National Institutes of Health, Bethesda, MD, USA
| | - Xingmin Feng
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Zhijie Wu
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Chengyu Liu
- Transgenic Core, National Heart, Lung, and Blood Institute, US National Institutes of Health, Bethesda, MD, USA
| | - Aaron B Schultz
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Molly Dalzell
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | | | - Claudia Haferlach
- Munich Leukemia Laboratory (MLL) Max-Lebsche-Platz 31, München, Germany
| | - Foster Birnbaum
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Computational and Systems Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Joel A Sexton
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Amy E Keating
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - John J O'Shea
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Neal S Young
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alejandro V Villarino
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Priscilla A Furth
- Laboratory of Genetics and Physiology, National Institute of Diabetes and Digestive and Kidney Diseases, US National Institutes of Health, Bethesda, MD, USA
| | - Lothar Hennighausen
- Laboratory of Genetics and Physiology, National Institute of Diabetes and Digestive and Kidney Diseases, US National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
33
|
Liu DH, Liu QR, Tojibaev KS, Sukhorukov AP, Wariss HM, Zhao Y, Yang L, Li WJ. Phylogenomics provides new insight into the phylogeny and diversification of Asian Lappula (Boraginaceae). Mol Phylogenet Evol 2025; 208:108361. [PMID: 40287026 DOI: 10.1016/j.ympev.2025.108361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 03/26/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
The application of omics data serves as a powerful tool for investigating the roles of incomplete lineage sorting (ILS) and hybridization in shaping genomic diversity, offering deeper insights into complex evolutionary processes. In this study, we utilized deep genome sequencing data from 76 individuals of Lappula and its closely allied genera, collected from China and Central Asia. By employing the HybPiper and Easy353 pipelines, we recovered 262-279 single-copy nuclear genes (SCNs) and 352-353 Angiosperms353 genes, respectively. We analyzed multiple datasets, including complete chloroplast genomes and a filtered set of 475 SCNs, to conduct phylogenetic analyses using both concatenated and coalescent-based methods. Furthermore, we employed Quartet Sampling (QS), coalescent simulations, MSCquartets, HyDe, and reticulate network analyses to investigate the sources of phylogenetic discordance. Our results confirm that Lappula is polyphyletic, with L. mogoltavica clustering with Pseudolappula sinaica and forming a sister relationship with other taxa included in this study. Additionally, three Lepechiniella taxa nested within distinct clades of Lappula. Significant gene tree discordance was observed at several nodes within Lappula. Coalescent simulations and hybrid detection analyses suggest that both ILS and hybridization contribute to these discrepancies. Flow cytometry (FCM) analyses confirmed the presence of both diploid and tetraploid taxa within Lappula. Phylogenetic network analyses further revealed that Clades IV and VII likely originated through hybridization, with the tetraploids in Clade IV arising from two independent hybridization events. Additionally, the "ghost lineage" identified as sister to Lappula redowskii serves as one of the donors in allopolyploidization. In conclusion, our study provides new insights into the deep phylogenetic relationships of Asian Lappula and its closely allied genera, contributing to a more comprehensive understanding of the evolution and diversification of Lappula.
Collapse
Affiliation(s)
- Dan-Hui Liu
- State Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Urumqi 830011, China; Xinjiang Key Lab of Conservation and Utilization of Plant Gene Resources, Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Urumqi 830011, China; China-Tajikistan Belt and Road Joint Laboratory on Biodiversity Conservation and Sustainable Use, Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Urumqi 830011, China
| | - Quan-Ru Liu
- College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Komiljon Sh Tojibaev
- Institute of Botany, Uzbekistan Academy of Sciences, Tashkent 100125, Uzbekistan
| | - Alexander P Sukhorukov
- Department of Higher Plants, Biological Faculty, Lomonosov Moscow State University, Moscow 119234, Russia
| | - Hafiz Muhammad Wariss
- State Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Urumqi 830011, China; Department of Botany, University of Sargodha, Sargodha, 40100, Pakistan
| | - Yue Zhao
- State Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Urumqi 830011, China
| | - Lei Yang
- State Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Urumqi 830011, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wen-Jun Li
- State Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Urumqi 830011, China; Xinjiang Key Lab of Conservation and Utilization of Plant Gene Resources, Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Urumqi 830011, China; China-Tajikistan Belt and Road Joint Laboratory on Biodiversity Conservation and Sustainable Use, Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Urumqi 830011, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
34
|
Benevides VP, Saraiva MMS, Campos IC, Guerra PR, Silva SR, Miranda VFO, Almeida AM, Christensen H, Olsen JE, Berchieri Junior A. Salmonella Heidelberg isolates from poultry in Brazil and the United States share a large number of resistance and virulence determinants. Microb Pathog 2025; 204:107523. [PMID: 40180234 DOI: 10.1016/j.micpath.2025.107523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/20/2025] [Accepted: 03/31/2025] [Indexed: 04/05/2025]
Abstract
Salmonella enterica subps. enterica serovar Heidelberg (SH) is one of the most common serovars isolated from poultry and associated with severe infections in humans. Commonly considered multidrug-resistant, it represents a risk to public health. We analyzed 317 SH genomes, including 314 from the Enterobase database from Brazil and the United States (US), and added three recently sequenced Brazilian isolates. In genomes from both countries, the main identified resistance genes were: aac(6')-Iaa, fosA7, sul2, tet(A), and blaCMY-2. Mutations in GyrA (S83Y only from US genomes and S83F and D87N from Brazilian genomes) were observed in 17 % and 90.62 % of genomes from US and Brazil, respectively, and ParC mutation (T57S), was identified in all genomes. The plasmid replicons most identified in both countries were ColpVC, IncC, IncI1-I(Gamma), and IncX1. The core and soft-core genes were utilized as the basis for conducting a phylogenetic analysis, showing seven clusters of strains, of which only one was shared between strains from the US and Brazil. Overall, this study highlights the variation in genomic profiles of SH circulating in poultry production in both countries, emphasizing the need for improved surveillance measures to protect human and animal populations from potential outbreaks worldwide.
Collapse
Affiliation(s)
- Valdinete P Benevides
- Postgraduate Program in Agricultural Microbiology, São Paulo State University (UNESP), School of Agricultural and Veterinary Sciences, Jaboticabal, 14884-900, Brazil; São Paulo State University (UNESP), School of Agricultural and Veterinarian Sciences, Jaboticabal, SP, 14884-900, Brazil; Department of Veterinary and Animal Sciences, University of Copenhagen, 1870, Frederiksberg, Denmark.
| | - Mauro M S Saraiva
- São Paulo State University (UNESP), School of Agricultural and Veterinarian Sciences, Jaboticabal, SP, 14884-900, Brazil; Department of Veterinary and Animal Sciences, University of Copenhagen, 1870, Frederiksberg, Denmark.
| | - Isabella C Campos
- São Paulo State University (UNESP), School of Agricultural and Veterinarian Sciences, Jaboticabal, SP, 14884-900, Brazil.
| | - Priscila R Guerra
- Department of Veterinary and Animal Sciences, University of Copenhagen, 1870, Frederiksberg, Denmark
| | - Saura R Silva
- Laboratory of Plant Systematics, Department of Biology, São Paulo State University (UNESP), School of Agricultural and Veterinary Sciences, Jaboticabal, 14884-900, Brazil.
| | - Vitor F O Miranda
- Laboratory of Plant Systematics, Department of Biology, São Paulo State University (UNESP), School of Agricultural and Veterinary Sciences, Jaboticabal, 14884-900, Brazil.
| | - Adriana M Almeida
- São Paulo State University (UNESP), School of Agricultural and Veterinarian Sciences, Jaboticabal, SP, 14884-900, Brazil.
| | - Henrik Christensen
- Department of Veterinary and Animal Sciences, University of Copenhagen, 1870, Frederiksberg, Denmark.
| | - John E Olsen
- Department of Veterinary and Animal Sciences, University of Copenhagen, 1870, Frederiksberg, Denmark.
| | - Angelo Berchieri Junior
- São Paulo State University (UNESP), School of Agricultural and Veterinarian Sciences, Jaboticabal, SP, 14884-900, Brazil.
| |
Collapse
|
35
|
Luo T, Vattathil SM, Lori A, Schneider JA, Bennett DA, Wingo TS, Wingo AP. Brain microRNAs differentially expressed in age-related cerebral pathologies. Neurobiol Aging 2025; 151:42-53. [PMID: 40228357 DOI: 10.1016/j.neurobiolaging.2025.03.014] [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: 03/04/2024] [Revised: 03/28/2025] [Accepted: 03/31/2025] [Indexed: 04/16/2025]
Abstract
Multiple brain pathologies accumulate with age, but their underlying biology remains unclear. We investigated the role of microRNAs (miRNAs) in ten age-related cerebral pathologies. Using miRNA sequencing profiles from the dorsolateral prefrontal cortex of 617 brain donors, we identified miRNAs associated with Alzheimer's disease (AD) pathology, Lewy body pathology, arteriolosclerosis, cerebral amyloid angiopathy, and LATE-NC after adjusting for age, sex, and education. After additionally adjusting for co-existing cerebral pathologies, we found miRNAs specifically associated with AD pathology (n = 75), Lewy body pathology (n = 45), arteriolosclerosis (n = 3), cerebral amyloid angiopathy (n = 1), and LATE-NC (n = 4). While some miRNAs were pathology-specific, 14 miRNAs (including those in the miR-132/212 cluster) were associated with both AD pathology and Lewy body pathology, and one (miR-193a-5p) was associated with both AD pathology and cerebral amyloid angiopathy. Gene set enrichment analysis showed that miRNAs associated with arteriolosclerosis target genes involved in glutathione metabolism, synaptic functions, cellular transport, and innate immune response. These findings highlight the role of miRNAs in age-related cerebral pathologies and provide a foundation for future mechanistic studies.
Collapse
Affiliation(s)
- Tianze Luo
- Department of Neurology, Emory University, Atlanta, GA, USA
| | - Selina M Vattathil
- Department of Neurology, University of California Davis, Sacramento, CA, USA
| | - Adriana Lori
- Department of Psychiatry, Emory University, Atlanta, GA, USA
| | - Julie A Schneider
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Thomas S Wingo
- Department of Neurology, University of California Davis, Sacramento, CA, USA; Alzheimer's Disease Research Center, University of California Davis, Sacramento, CA, USA.
| | - Aliza P Wingo
- Department of Psychiatry, University of California Davis, Sacramento, CA, USA; Veterans Affairs Northern California Health Care System, Sacramento, CA, USA.
| |
Collapse
|
36
|
Zhu Y, Li R, Yan S, Li Y, Xie S. Copper contamination determined the impact of phages on microbially-driven nitrogen cycling in coastal wetland sediments. JOURNAL OF HAZARDOUS MATERIALS 2025; 490:137870. [PMID: 40056518 DOI: 10.1016/j.jhazmat.2025.137870] [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: 01/09/2025] [Revised: 02/24/2025] [Accepted: 03/05/2025] [Indexed: 03/10/2025]
Abstract
Phages have garnered increasing attention due to their potential roles in biogeochemical cycling. However, their impacts on nitrogen cycling have primarily been inferred from the presence of putative auxiliary metabolic genes (AMGs) and the virus-host linkage, despite of very limited direct experimental evidence. In this study, a series of microcosms were established with the inoculation of either native or non-native phages to simulate coastal wetlands with different phage sources and different levels of copper (Cu) contamination. Metagenomics and metatranscriptomics were combined to reveal phages' regulation on microbially-driven nitrogen cycling and to explore how the effects were mediated by Cu stress. Phages significantly impacted denitrification-related genes, with their effects depending on Cu level. Phages inhibited nirK-type denitrification under Cu stress but led to up-regulation of nirS gene in the treatments without Cu addition. Non-native phages also promoted the transcription of genes related to nitrogen assimilation and organic nitrogen transformation. Detection of viral AMGs involved in glutamate synthesis suggested that horizontal gene transfer may be a crucial pathway for phages to facilitate microbial nitrogen uptake. Overall, these findings enhance the understanding of phages' impact on biogeochemical metabolism in coastal wetland, offering novel insights into the links of phages' regulation on microbial nitrogen cycling with Cu stress.
Collapse
Affiliation(s)
- Ying Zhu
- State Key Joint Laboratory of Environmental Simulation and Pollution Control, College of Environmental Sciences and Engineering, Peking University, Beijing 100871, China
| | - Ruili Li
- School of Environment and Energy, Peking University Shenzhen Graduate School, Shenzhen 518055, China; Guangdong Mangrove Engineering Technology Research Center, Peking University Shenzhen Graduate School, Shenzhen 518055, China.
| | - Shuang Yan
- State Key Joint Laboratory of Environmental Simulation and Pollution Control, College of Environmental Sciences and Engineering, Peking University, Beijing 100871, China
| | - Yangyang Li
- State Key Joint Laboratory of Environmental Simulation and Pollution Control, College of Environmental Sciences and Engineering, Peking University, Beijing 100871, China
| | - Shuguang Xie
- State Key Joint Laboratory of Environmental Simulation and Pollution Control, College of Environmental Sciences and Engineering, Peking University, Beijing 100871, China.
| |
Collapse
|
37
|
Stocksdale JT, Leventhal MJ, Lam S, Xu YX, Wang YO, Wang KQ, Thomas R, Faghihmonzavi Z, Raghav Y, Smith C, Wu J, Miramontes R, Sarda K, Johnston H, Shin MG, Huang T, Foster M, Barch M, Amirani N, Paiz C, Easter L, Duderstadt E, Vaibhav V, Sundararaman N, Felsenfeld DP, Vogt TF, Van Eyk J, Finkbeiner S, Kaye JA, Fraenkel E, Thompson LM. Intersecting impact of CAG repeat and huntingtin knockout in stem cell-derived cortical neurons. Neurobiol Dis 2025; 210:106914. [PMID: 40258535 DOI: 10.1016/j.nbd.2025.106914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 04/13/2025] [Accepted: 04/14/2025] [Indexed: 04/23/2025] Open
Abstract
Huntington's Disease (HD) is caused by a CAG repeat expansion in the gene encoding huntingtin (HTT). While normal HTT function appears impacted by the mutation, the specific pathways unique to CAG repeat expansion versus loss of normal function are unclear. To understand the impact of the CAG repeat expansion, we evaluated biological signatures of HTT knockout (HTT KO) versus those that occur from the CAG repeat expansion by applying multi-omics, live cell imaging, survival analysis and a novel feature-based pipeline to study cortical neurons (eCNs) derived from an isogenic human embryonic stem cell series (RUES2). HTT KO and the CAG repeat expansion influence developmental trajectories of eCNs, with opposing effects on growth. Network analyses of differentially expressed genes and proteins associated with enriched epigenetic motifs identified subnetworks common to CAG repeat expansion and HTT KO that include neuronal differentiation, cell cycle regulation, and mechanisms related to transcriptional repression, and may represent gain-of-function mechanisms that cannot be explained by HTT loss of function alone. A combination of dominant and loss-of-function mechanisms are likely involved in the aberrant neurodevelopmental and neurodegenerative features of HD that can help inform therapeutic strategies.
Collapse
Affiliation(s)
| | - Matthew J Leventhal
- MIT PhD Program in Computational and Systems Biology, Cambridge, MA 02139, USA; MIT Department of Biological Engineering, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Stephanie Lam
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Yu-Xin Xu
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Yang Oliver Wang
- Advanced Clinical Biosystems Research Institute, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Keona Q Wang
- Department of Neurobiology and Behavior, UC Irvine, Irvine, CA 92677, USA
| | - Reuben Thomas
- Institute of Data Science and Biotechnology, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Zohreh Faghihmonzavi
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Yogindra Raghav
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Charlene Smith
- Department of Psychiatry and Human Behavior, UC Irvine, Irvine, CA 92697, USA
| | - Jie Wu
- Department of Biological Chemistry, UC Irvine, Irvine, CA 92697, USA
| | - Ricardo Miramontes
- Institute for Memory Impairments and Neurological Disorders, UC Irvine, Irvine, CA 92697, USA
| | - Kanchan Sarda
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Heather Johnston
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Min-Gyoung Shin
- Institute of Data Science and Biotechnology, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Terry Huang
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Mikelle Foster
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Mariya Barch
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Naufa Amirani
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Chris Paiz
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Lindsay Easter
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Erse Duderstadt
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Vineet Vaibhav
- Advanced Clinical Biosystems Research Institute, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Niveda Sundararaman
- Advanced Clinical Biosystems Research Institute, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | | | | | - Jennifer Van Eyk
- Advanced Clinical Biosystems Research Institute, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Steve Finkbeiner
- Institute of Data Science and Biotechnology, Gladstone Institutes, San Francisco, CA 94158, USA; Department of Physiology, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Neurology, University of California San Francisco, San Francisco, CA 94158, USA; Taube/Koret Center for Neurodegenerative Disease Research, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Julia A Kaye
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, CA 94158, USA; Taube/Koret Center for Neurodegenerative Disease Research, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Ernest Fraenkel
- MIT PhD Program in Computational and Systems Biology, Cambridge, MA 02139, USA; MIT Department of Biological Engineering, Cambridge, MA 02139, USA
| | - Leslie M Thompson
- Department of Neurobiology and Behavior, UC Irvine, Irvine, CA 92677, USA; Department of Psychiatry and Human Behavior, UC Irvine, Irvine, CA 92697, USA; Department of Biological Chemistry, UC Irvine, Irvine, CA 92697, USA; Institute for Memory Impairments and Neurological Disorders, UC Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
38
|
Wang L, Gu Y, Shen C. Transcriptome analysis and lncRNA expression profile in brain tissues of neonatal hypoxic-ischemic brain damage rat model. Gene 2025; 952:149363. [PMID: 40064305 DOI: 10.1016/j.gene.2025.149363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 02/20/2025] [Accepted: 02/24/2025] [Indexed: 03/21/2025]
Abstract
BACKGROUND AND OBJECTIVE Neonatal hypoxic-ischemic encephalopathy (HIE) remains a critical challenge in perinatal medicine. This study aimed to elucidate the transcriptomic landscape, focusing on long non-coding RNAs (lncRNAs) expression patterns in the brain tissues of a neonatal rat model of HIE. METHODOLOGY We employed a modified Rice-Vannucci model to induce HIE in postnatal day 4 (P4) rats. The experimental groups were subjected to either 5 or 7 min of hypoxia (0 % O2, 100 % N2), while control animals were exposed to normoxic conditions. RESULTS RNA sequencing revealed a complex transcriptomic landscape in HIE brains, with approximately 80 million differentially expressed lncRNAs compared to controls. ELISA results demonstrated a significant upregulation of pro-inflammatory cytokines (IL-1β, IL-6, TNF-α) and a concomitant decrease in anti-inflammatory IL-10 levels in brain tissue of HIE rats. qRT-PCR analysis revealed aberrant expression of several miRNAs. Biochemical assays indicated a marked reduction in superoxide dismutase (SOD) activity and an increase in malondialdehyde (MDA) content in HIE brain tissues. CONCLUSIONS This study highlights the potential regulatory roles of lncRNAs in HIE brains. The intricate interplay between lncRNAs, miRNAs, and mRNAs and alterations in inflammatory and oxidative stress markers suggests a complex regulatory network governing HIE pathogenesis.
Collapse
Affiliation(s)
- Limin Wang
- Department of Pediatrics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China
| | - Yanni Gu
- Department of Fever Emergency, Shanghai TCM-Integrated Hospital, Shanghai 200082, China
| | - Chaobin Shen
- Department of Pediatrics, Shanghai TCM-Integrated Hospital, Shanghai 200082, China.
| |
Collapse
|
39
|
Zhang RM, Chen XJ, Li YF, Tan HZ, Huang WQ, Li LL, Li XT, Xie J, Chong YX, Lian XL, Huang XH. Removal of antibiotic resistance from wastewater in aquatic ecosystems dominated by submerged macrophytes. JOURNAL OF HAZARDOUS MATERIALS 2025; 489:137706. [PMID: 40010218 DOI: 10.1016/j.jhazmat.2025.137706] [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: 10/18/2024] [Revised: 02/15/2025] [Accepted: 02/20/2025] [Indexed: 02/28/2025]
Abstract
Submerged macrophytes in constructed wetlands (CWs) can effectively improve wastewater quality. However, the effectiveness of different submerged macrophytes in removing antibiotic-resistant genes (ARGs) from wastewater remains unexplored. Additionally, wastewater loading in wetlands can fluctuate due to climate change, potentially affecting ARG removal efficiency. In this study, we systematically constructed microscale wetlands using three submerged plants: Vallisneria natans (VN), Sagittaria pygmaea (SP), and Myriophyllum spicatum (MS). Their effectiveness in ARGs removal was analyzed at hydraulic retention times (HRTs) of 0, 3, 6, and 9 days under high (HWL) and low (LWL) wastewater loading. The results indicated that under LWL conditions, all ecosystems exhibited a higher reduction rate of ARG diversity and relative abundance (RS) compared to HWL conditions. The efficiency of all ecosystems in reducing ARG diversity and abundance followed the order: MS > VN > SP. The sul resistance gene exhibited the highest RS and was degraded most rapidly in all samples. Additionally, sulfadimidine concentrations significantly decreased under LWL conditions, which was significantly correlated with sul reduction. Chemical oxygen demand, total phosphorus, total nitrogen, ammonium nitrogen, and nitrate nitrogen were identified as key factors influencing bacterial and ARG profiles. The increase in rhizobial bacteria and decrease in aerobic denitrifying bacteria likely contributed significantly to ARGs removal. This study offers new insights into ARG removal by submerged macrophytes in CWs, emphasizing the role of wastewater loading and the potential of MS in enhancing ARG degradation. These findings enhance CW design and management to mitigate ARG contamination in wastewater.
Collapse
Affiliation(s)
- Rong-Min Zhang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China; Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xiao-Jie Chen
- Key Laboratory of Animal Antimicrobial Resistance Surveillance, Ministry of Agriculture and Rural Affairs, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beiling 100081, China
| | - Ya-Fei Li
- Institute of Quality Standard and Monitoring Technology for Agro-Products of Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
| | - Hui-Zhen Tan
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China; Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Wen-Qing Huang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China; Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Ling-Lin Li
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China; Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xin-Ting Li
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China; Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Jie Xie
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China; Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yun-Xiao Chong
- Guangdong Provincial Key Laboratory of Agricultural & Rural Pollution Abatement and Environmental Safety, College of Natural Resources and Environment, South China Agricultural University, Guangzhou 510642, China
| | - Xin-Lei Lian
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China; Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xian-Hui Huang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China; Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.
| |
Collapse
|
40
|
Su H, Xu W, Hu X, Xu Y, Wen G, Cao Y. The impact of microplastics on antibiotic resistance genes, metal resistance genes, and bacterial community in aquaculture environment. JOURNAL OF HAZARDOUS MATERIALS 2025; 489:137704. [PMID: 39987738 DOI: 10.1016/j.jhazmat.2025.137704] [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: 11/12/2024] [Revised: 01/14/2025] [Accepted: 02/20/2025] [Indexed: 02/25/2025]
Abstract
Microplastics are emerging contaminants. However, their effects on antibiotic resistance genes (ARGs), metal resistance genes (MRGs), and the structure and abundance of bacterial communities, particularly pathogens, in aquaculture environments remains poorly understood. Therefore, this study investigated the effect of microplastics of different sizes on the abundance and distribution of ARGs, MRGs, and bacterial communities in aquaculture environments. The results revealed that, compared with pond water, large microplastics harbored significantly higher ARG abundances, particularly for multidrug-resistant genes; notably, level-I- and -II-risk ARGs were more prevalent on microplastics, highlighting the potential for horizontal gene transfer. Microplastics also exhibited a propensity to aggregate pathogenic bacteria such as Brucella and Pseudomonas, which could pose direct risks to aquatic product safety and public health. Network and differential network analyses revealed significant correlations between bacterial genera and ARG/MRG abundance, particularly on microplastics. Therefore, our findings suggest that microplastics act as vectors for the spread of ARGs, MRGs, and pathogens in aquaculture, potentially leading to the formation of complexes of these materials that threaten ecosystem health and human well-being. This study provides critical insights into the need for targeted management strategies to mitigate microplastic pollution in aquaculture settings.
Collapse
Affiliation(s)
- Haochang Su
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences /Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangzhou 510300, China; Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture and Rural Affairs /Guangdong Provincial Key Laboratory of Fishery Ecology and Environment, 231 West Xingang Road, Guangzhou 510301, China
| | - Wujie Xu
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences /Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangzhou 510300, China; Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture and Rural Affairs /Guangdong Provincial Key Laboratory of Fishery Ecology and Environment, 231 West Xingang Road, Guangzhou 510301, China
| | - Xiaojuan Hu
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences /Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangzhou 510300, China; Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture and Rural Affairs /Guangdong Provincial Key Laboratory of Fishery Ecology and Environment, 231 West Xingang Road, Guangzhou 510301, China
| | - Yu Xu
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences /Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangzhou 510300, China; Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture and Rural Affairs /Guangdong Provincial Key Laboratory of Fishery Ecology and Environment, 231 West Xingang Road, Guangzhou 510301, China
| | - Guoliang Wen
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences /Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangzhou 510300, China; Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture and Rural Affairs /Guangdong Provincial Key Laboratory of Fishery Ecology and Environment, 231 West Xingang Road, Guangzhou 510301, China
| | - Yucheng Cao
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences /Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangzhou 510300, China; Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture and Rural Affairs /Guangdong Provincial Key Laboratory of Fishery Ecology and Environment, 231 West Xingang Road, Guangzhou 510301, China.
| |
Collapse
|
41
|
Liu H, Chen X, Wang H, Zhuang G, Zhu ZJ, Zhuang M. ZBTB17/MIZ1 promotes peroxisome biogenesis by transcriptional regulation of PEX13. J Cell Biol 2025; 224:e202407198. [PMID: 40243840 PMCID: PMC12005116 DOI: 10.1083/jcb.202407198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 01/16/2025] [Accepted: 02/26/2025] [Indexed: 04/18/2025] Open
Abstract
Peroxisomes are integral metabolic organelles involved in both catabolic and anabolic processes in humans, with defects linked to diseases. The functions of peroxisomes are regulated at transcriptional, translational, and posttranslational levels. In this study, we employed the CRISPR/Cas9-based screening of a ubiquitin ligase library to identify regulators of human peroxisomes. We discovered that ZBTB17 (MIZ1) plays a role in regulating the import of proteins into peroxisomes. Independent of its ubiquitin ligase activity, ZBTB17/MIZ1 operates as a transcription factor to modulate the expression of key importer PEX13, influencing the localization of peroxisomal enzymes. Furthermore, metabolomic profiling reveals that knockdown of ZBTB17 or PEX13 results in similar metabolic alterations, with downregulated purine synthesis. Collectively, we identify ZBTB17 as a key regulator of peroxisomal protein import, thereby affecting peroxisomal function and nucleotide metabolism. Our findings provide insights into the multifaceted regulation of peroxisomes in complex human cells and shed light on the molecular mechanisms underlying ZBTB17's role as a transcriptional regulator.
Collapse
Affiliation(s)
- Hongqin Liu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Xi Chen
- University of Chinese Academy of Sciences, Beijing, China
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Hanlin Wang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, China
| | - Guanglei Zhuang
- State Key Laboratory of Systems Medicine for Cancer, Department of Obstetrics and Gynecology, Shanghai Cancer Institute, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zheng-Jiang Zhu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- Shanghai Key Laboratory of Aging Studies, Shanghai, China
| | - Min Zhuang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| |
Collapse
|
42
|
Hong S, Moon JS, Lee YJ, Kim HY. Whole-genome sequencing-based characterization of Listeria monocytogenes isolated from cattle and pig slaughterhouses. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2025; 130:105737. [PMID: 40068769 DOI: 10.1016/j.meegid.2025.105737] [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: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 03/17/2025]
Abstract
Listeria monocytogenes is a foodborne pathogen that causes human listeriosis and may be transmitted to humans via the food chain, beginning at slaughter and extending through food production and consumption. In this study, we performed whole-genome sequencing (WGS) analysis to determine the genetic characteristics of L. monocytogenes from the carcasses and environments of cattle and pig slaughterhouses in Korea. In total, 50 L. monocytogenes isolates were collected from 46 cattle and 47 pig slaughterhouses nationwide from 2014 to 2022. They were classified into two lineages, 12 sublineages, 12 sequence types, 11 clonal complexes (CCs), and 15 core-genome multilocus sequence types. L. monocytogenes isolates were divided into two lineages: lineage I (serotypes 1/2b and 4b) and lineage II (serotypes 1/2a and 1/2c). The most frequent CCs were CC9 (46.0 %), followed by CC224 (16.0 %) and CC155 (14.0 %). Although all isolates exhibited highly conserved LIPI-1, 20.0 % and 2.0 % contained LIPI-3 or LIPI-4, respectively. Moreover, 96.0 % of the isolates had full-length inlA. Interestingly, 21 of the 23 CC9 isolates contained mutations in inlA resulting from premature stop codon (PMSC). The mdrL and Listeria genomic island-2 (LGI-2) were identified in all L. monocytogenes isolates, whereas LGI-3 was identified in 32.0 % of the isolates. The L. monocytogenes isolates contained various antimicrobial resistance genes, moreover, the plasmid-borne resistance genes tetM and mprF were also identified in 34.0 % and 100 % of the isolates, respectively. Twenty-four isolates (48.0 %) harbored one or two plasmids (pLM33, DOp1, pLGUG1, and pLM5578), and 29 isolates (58.0 %) harbored at least one insertion sequence, composite transposon, and integrative conjugative element. Four isolates showed two CRISPR-Cas types IB and II-A. In addition, phage sequences associated with the spacer constituting the CRISPR array were identified in 26 Listeria phages from 14 L. monocytogenes isolates. The genetic composition of L. monocytogenes was conserved in a collinearity relationship between each of the five L. monocytogenes isolates from the cattle and pig slaughterhouses. These findings suggest that L. monocytogenes isolated from cattle and pig slaughterhouses have the ability to cause human disease and exhibit virulent characteristics.
Collapse
Affiliation(s)
- Serim Hong
- College of Veterinary Medicine & Institute for Veterinary Biomedical Science, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Jin-San Moon
- Avian Disease Research Division, Animal and Plant Quarantine Agency, Gimcheon 39660, Republic of Korea
| | - Young Ju Lee
- College of Veterinary Medicine & Institute for Veterinary Biomedical Science, Kyungpook National University, Daegu 41566, Republic of Korea.
| | - Ha-Young Kim
- Foot-and-Mouth Disease Research Division, Animal and Plant Quarantine Agency, Gimcheon 39660, Republic of Korea.
| |
Collapse
|
43
|
Paudel D, Parrish SB, Peng Z, Parajuli S, Deng Z. A chromosome-scale and haplotype-resolved genome assembly of tetraploid blackberry ( Rubus L. subgenus Rubus Watson). HORTICULTURE RESEARCH 2025; 12:uhaf052. [PMID: 40271456 PMCID: PMC12015472 DOI: 10.1093/hr/uhaf052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 02/13/2025] [Indexed: 04/25/2025]
Abstract
Blackberries (Rubus spp.) are globally consumed and well known for their rich anthocyanin and antioxidant content and distinct flavors. Improving blackberries has been challenging due to genetic complexity of traits and limited genomic resources. The blackberry genome has been particularly challenging to assemble due to its polyploid nature. Here, we present the first chromosome-scale and haplotype-phased assembly for the primocane-fruiting, thornless tetraploid blackberry selection BL1 (Rubus L. subgenus Rubus Watson). The genome assembly was generated using Oxford Nanopore Technology and Hi-C scaffolding, resulting in a 919 Mb genome distributed across 27 pseudochromosomes, with an N50 of 35.73 Mb. This assembly covers >92% of the genome length and contains over 98% of complete BUSCOs. Approximately, 58% of the assembly consists of repetitive sequences, with long terminal repeats being the most abundant class. A total of 87,968 protein-coding genes were predicted, of which, 82% were functionally annotated. Genome mining and RNA-Seq analyses identified possible candidate genes and transcription factors related to thornlessness and the key structural genes and transcription factors for anthocyanin biosynthesis. Activator genes including PAP1 and TTG1 and repressor genes such as ANL2 and MYBPA1 play an important role in the fine tuning of anthocyanin production during blackberry development. Resequencing of seven tetraploid blackberry cultivars/selections with different horticultural characteristics revealed candidate genes that could impact fruiting habit and disease resistance/susceptibility. This tetraploid reference genome should provide a valuable resource for accelerating genetic analysis of blackberries and facilitating the development of new improved cultivars with enhanced horticultural and nutritional traits.
Collapse
Affiliation(s)
- Dev Paudel
- Gulf Coast Research and Education Center, Department of Environmental Horticulture, University of Florida, IFAS, 14625 County Road 672, Wimauma, FL 33598, USA
| | - S Brooks Parrish
- Gulf Coast Research and Education Center, Department of Environmental Horticulture, University of Florida, IFAS, 14625 County Road 672, Wimauma, FL 33598, USA
| | - Ze Peng
- Gulf Coast Research and Education Center, Department of Environmental Horticulture, University of Florida, IFAS, 14625 County Road 672, Wimauma, FL 33598, USA
| | - Saroj Parajuli
- Gulf Coast Research and Education Center, Department of Environmental Horticulture, University of Florida, IFAS, 14625 County Road 672, Wimauma, FL 33598, USA
| | - Zhanao Deng
- Gulf Coast Research and Education Center, Department of Environmental Horticulture, University of Florida, IFAS, 14625 County Road 672, Wimauma, FL 33598, USA
| |
Collapse
|
44
|
Ishihara H, Yamashita R, Ishiyama R, Ikeda T, Fukuda H, Yoshida K, Hirai T, Iizuka J, Kondo T, Nagashima Y, Takagi T. Genome-wide transcriptome and DNA methylome profiling of acquired cystic disease-associated renal cell carcinoma. Pathology 2025; 57:495-501. [PMID: 39984417 DOI: 10.1016/j.pathol.2024.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 09/11/2024] [Accepted: 11/01/2024] [Indexed: 02/23/2025]
Abstract
Acquired cystic disease (ACD)-associated renal cell carcinoma (RCC) develops uniquely and frequently in patients receiving long-term dialysis for end-stage renal disease (ESRD). In our previous study, the molecular alteration profiles of ACD-associated RCC were partially similar to those of papillary RCC (PRCC). However, the specific profiles of molecular alterations in ACD-associated RCC and their pathogenic mechanisms remain largely unknown. Therefore, we compared genome-wide transcription and DNA methylation profiles of 12 ACD-associated RCC and 26 PRCC samples, which comprised eight ESRD-induced and 18 sporadic (arising in non-dialysis kidney) PRCC samples. RNA-seq and Infinium Methylation EPIC were used to identify the unique genetic and epigenetic profiles in ACD-associated RCC. ACD-associated RCC harboured a unique expression profile from that of PRCC. Its profile was characterised by the upregulation of pathways related to amino acid metabolism. In addition, ACD-associated RCC exhibited a unique DNA methylation profile that was characterised by the hypomethylation of pathways related to amino acid metabolism. This reflected a significant difference between the expression profiles of ACD-associated RCC and PRCC. The present genome-wide transcriptome and DNA methylome profiling revealed that aberrant activation of amino acid metabolism-related pathways, potentially induced by DNA hypomethylation, may be involved in the pathogenesis of ACD-associated RCC.
Collapse
Affiliation(s)
- Hiroki Ishihara
- Department of Urology, Tokyo Women's Medical University, Shinjuku-ku, Tokyo, Japan.
| | - Riu Yamashita
- Division of Translational Informatics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Chiba, Japan
| | - Ryo Ishiyama
- Department of Urology, Tokyo Women's Medical University, Shinjuku-ku, Tokyo, Japan
| | - Takashi Ikeda
- Department of Urology, Tokyo Women's Medical University, Shinjuku-ku, Tokyo, Japan
| | - Hironori Fukuda
- Department of Urology, Tokyo Women's Medical University, Shinjuku-ku, Tokyo, Japan
| | - Kazuhiko Yoshida
- Department of Urology, Tokyo Women's Medical University, Shinjuku-ku, Tokyo, Japan
| | - Toshihito Hirai
- Department of Urology, Tokyo Women's Medical University, Shinjuku-ku, Tokyo, Japan
| | - Junpei Iizuka
- Department of Urology, Tokyo Women's Medical University, Shinjuku-ku, Tokyo, Japan
| | - Tsunenori Kondo
- Department of Urology, Tokyo Women's Medical University Adachi Medical Center, Tokyo, Japan
| | - Yoji Nagashima
- Department of Surgical Pathology, Tokyo Women's Medical University, Tokyo, Japan
| | - Toshio Takagi
- Department of Urology, Tokyo Women's Medical University, Shinjuku-ku, Tokyo, Japan
| |
Collapse
|
45
|
Xu J, Shen Z, Hao T, Su H, Chen M, Pan X, Yi Z. Exploring the evolution of anaerobes within ciliate class Prostomatea by transcriptomics. Mol Phylogenet Evol 2025; 207:108345. [PMID: 40174813 DOI: 10.1016/j.ympev.2025.108345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 03/18/2025] [Accepted: 03/30/2025] [Indexed: 04/04/2025]
Abstract
Mitochondrion-related organelles (MROs) enable anaerobic eukaryotes to thrive in anoxic environments, and the independent ciliate lineages of anaerobes serve as excellent candidates for investigating the convergent evolutionary transition from mitochondria to MROs. Previous studies have demonstrated that the adaptations of ciliates to anaerobic conditions may be lineage-specific. However, our understanding of the diverse metabolic peculiarities of MROs is limited to a few ciliate lineages. In this study, we sequenced the transcriptomes of four anaerobic species from two genera (Apolagynus and Holophrya), which are classified within the predominantly aerobic class Prostomatea, and predicted their mitochondrial metabolisms. The ecological niches of prostomatean anaerobes were mapped onto newly constructed phylogenomic trees and small subunit (SSU) rDNA trees. Results showed that paraphyletic class Prostomatea containing six clades (Clade Ⅰ-Ⅵ) has a close relationship with Oligohymenophorea and Plagiopylea. Notably, all prostomatean species within Clade Ⅱ are anaerobic, while anaerobes are only sporadically present in other clades. The MROs of anaerobic prostomatean species display at least two distinct phenotypes. Holophrya ovum in Clade Ⅰ produces ATP by oxidative phosphorylation under aerobic conditions and via substrate-level phosphorylation via acetate: succinate CoA transferase (ASCT) and succinyl CoA synthetase (SCS) as well as adenylate kinase (AK) under anaerobic conditions. In contrast, three species of Apolagynus in Clade Ⅱ possess reduced electron transport chain (ETC), and are capable of ATP generation via substrate-level phosphorylation mediated by ASCT/SCS and propionyl-CoA. Additionally, these three Apolagynus species possess [FeFe] hydrogenase probably producing H2. A comparison of the ETC pathways among various anaerobic ciliates further showed that the MROs of these organisms have originated from repeated convergent evolution. Our findings shed lights on evolutionary history of anaerobes within the ciliate class Prostomatea.
Collapse
Affiliation(s)
- Jiahui Xu
- Key Laboratory of Biodiversity of Aquatic Organisms, Harbin Normal University, Harbin 150025, China; Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Zhuo Shen
- Earth, Ocean and Atmospheric Sciences Thrust, Function Hub, The Hong Kong University of Science and Technology (Guangzhou), Guangzhou 511458, China; School of Marine Sciences, Sun Yat-sen University, Zhuhai 519082, China
| | - Tingting Hao
- Key Laboratory of Biodiversity of Aquatic Organisms, Harbin Normal University, Harbin 150025, China; Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Hua Su
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Miaoying Chen
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Xuming Pan
- Key Laboratory of Biodiversity of Aquatic Organisms, Harbin Normal University, Harbin 150025, China.
| | - Zhenzhen Yi
- Key Laboratory of Biodiversity of Aquatic Organisms, Harbin Normal University, Harbin 150025, China; Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, School of Life Science, South China Normal University, Guangzhou 510631, China.
| |
Collapse
|
46
|
Zhang H, Ruan Y, Kuzyakov Y, Sun H, Huang Q, Guo S, Shen Q, Ling N. Viruses Facilitate Energy Acquisition Potential by Their Bacterial Hosts in Rhizosphere of Grafted Plants. PLANT, CELL & ENVIRONMENT 2025; 48:4599-4610. [PMID: 40038896 DOI: 10.1111/pce.15458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 02/03/2025] [Accepted: 02/21/2025] [Indexed: 03/06/2025]
Abstract
Viruses alter the ecological and evolutionary trajectories of bacterial host communities. Plant grafting is a technique that integrates two species or varietiies and have consequences on the rhizosphere functioning. The grafting effects on the taxonomic and functional assembly of viruses and their bacterial host in the plant rhizosphere remain largely elusive. Using shotgun metagenome sequencing, we recover a total of 1441 viral operational taxonomic units from the rhizosphere of grafted and ungrafted plants after 8-year continuous monoculture. In the grafted and ungrafted rhizosphere, the Myoviridae, Zobellviridae and Kyanoviridae emerged as the predominant viral families, collectively representing around 40% of the viral community in each respective environment. Grafting enriched the members in viral family Kyanoviridae, Tectiviridae, Peduoviridae and Suoliviridae, and auxiliary metabolic genes related to pyruvate metabolism and energy acquisition (e.g., gloB, DNMT1 and dcyD). The virus-bacterial interactions increased the rapid growth potential of bacteria, which explains the strong increase in abundance of specific bacterial hosts (i.e., Chitinophagaceae, Cyclobacteriaceae and Spirosomaceae) in the grafted-plant rhizosphere. Overall, these results deepen our understanding of microbial community assembly and ecological services from the perspective of virus-host interactions.
Collapse
Affiliation(s)
- He Zhang
- Key Lab of Organic-Based Fertilizers of China and Jiangsu Provincial Key Lab for Solid Organic Waste Utilization, Nanjing Agricultural University, Nanjing, China
| | - Yang Ruan
- Key Lab of Organic-Based Fertilizers of China and Jiangsu Provincial Key Lab for Solid Organic Waste Utilization, Nanjing Agricultural University, Nanjing, China
- State Key Laboratory of Herbage Improvement and Grassland Agro-Ecosystems, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, China
| | - Yakov Kuzyakov
- Department of Soil Science of Temperate Ecosystems, Department of Agricultural Soil Science, University of Goettingen, Göttingen, Germany
- Peoples Friendship University of Russia (RUDN University), Moscow, Russia
| | - Hong Sun
- Key Lab of Organic-Based Fertilizers of China and Jiangsu Provincial Key Lab for Solid Organic Waste Utilization, Nanjing Agricultural University, Nanjing, China
| | - Qiwei Huang
- Key Lab of Organic-Based Fertilizers of China and Jiangsu Provincial Key Lab for Solid Organic Waste Utilization, Nanjing Agricultural University, Nanjing, China
| | - Shiwei Guo
- Key Lab of Organic-Based Fertilizers of China and Jiangsu Provincial Key Lab for Solid Organic Waste Utilization, Nanjing Agricultural University, Nanjing, China
| | - Qirong Shen
- Key Lab of Organic-Based Fertilizers of China and Jiangsu Provincial Key Lab for Solid Organic Waste Utilization, Nanjing Agricultural University, Nanjing, China
| | - Ning Ling
- Key Lab of Organic-Based Fertilizers of China and Jiangsu Provincial Key Lab for Solid Organic Waste Utilization, Nanjing Agricultural University, Nanjing, China
- State Key Laboratory of Herbage Improvement and Grassland Agro-Ecosystems, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, China
| |
Collapse
|
47
|
Boggs TE, Gross JB. Elevated Blood Hemoglobin in Different Cavefish Populations Evolves Through Diverse Hemoglobin Gene Expression Patterns. JOURNAL OF EXPERIMENTAL ZOOLOGY. PART B, MOLECULAR AND DEVELOPMENTAL EVOLUTION 2025; 344:175-181. [PMID: 39930703 PMCID: PMC12046277 DOI: 10.1002/jez.b.23289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/09/2024] [Accepted: 01/29/2025] [Indexed: 05/03/2025]
Abstract
Cave-dwelling animals thrive in isolated caves despite the pressures of darkness, starvation, and reduced oxygen. Prior work revealed that Astyanax cave-dwelling morphs derived from different cave localities express significantly higher levels of blood hemoglobin compared to surface-dwelling fish. Interestingly, this elevation is maintained in different populations of cavefish, despite captive rearing in normal oxygen conditions. We capitalized on the consistent response of elevated hemoglobin in captive cavefish, which were derived from geographically distinct regions, to determine if this elevation is underpinned by expression of the same Hb genes. Blood hemoglobin proteins are encoded by a large family of hemoglobin (Hb) gene family members, which demonstrate coordinated expression patterns, subject to various organismal (e.g., period of life history) and environmental influences (e.g., oxygen availability). Surprisingly, we found that geographically distinct populations showed mostly divergent patterns of Hb gene expression. Cavefish from two cave localities, Pachón and Tinaja, have a more recent shared origin, and show more similar Hb expression patterns as adults. However, during embryonic phases, Pachón and Tinaja show significant variability in timing of peak expression of Hb family members. In sum, the transcriptomic underpinnings of Hb gene expression represents a complex composite of shared and divergent expression patterns across three captive cavefish populations. We conclude that these differential patterns are likely influenced by life history, and the unique cave conditions in which these animals evolved.
Collapse
Affiliation(s)
- Tyler E. Boggs
- Department of Biological SciencesUniversity of CincinnatiCincinnatiOhioUSA
| | - Joshua B. Gross
- Department of Biological SciencesUniversity of CincinnatiCincinnatiOhioUSA
| |
Collapse
|
48
|
Wang N, Li Y, Li X, Li H, Bian C, Chen X, Jafari H, Chen N, Lei C. Genome-wide analysis of genetic diversity and selection signatures in Fuzhou cattle. Anim Genet 2025; 56:e70015. [PMID: 40324879 DOI: 10.1111/age.70015] [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: 01/13/2025] [Revised: 04/17/2025] [Accepted: 04/21/2025] [Indexed: 05/07/2025]
Abstract
The Fuzhou cattle breed, native to northeast China, is widely recognized for its adaptability, disease resistance, and docility. Despite being known for these qualities, its population has declined recently, and there is a significant lack of genomic studies on this species. We sequenced 21 samples from a primary breeding farm to determine the genetic structure, diversity, and selection signature to address this. Additionally, we combined 100 published genomic datasets from diverse geographical regions to characterize the genomic variation of Fuzhou cattle. There were 53 752 978 bi-allelic SNPs retained for downstream analysis. In population structure analysis, Fuzhou cattle show a predominantly East Asian taurine ancestry, with strong genetic affinities to Hanwoo and Yanbian cattle. Despite high nucleotide diversity within the Bos taurine lineage, genetic diversity analysis also revealed significant levels of inbreeding in Fuzhou cattle populations, indicating the need for conservation. Utilizing various methods such as θπ, iHS, FST, π-ratio, and XP-EHH, we identified genes associated with traits like growth, meat quality, energy metabolism, and immunity. Several genes related to cold adaptation were identified, including PLIN5, PLB1, and CPT2. These findings provide a basis for conservation strategies to safeguard the genetic resources of Fuzhou cattle.
Collapse
Affiliation(s)
- Nan Wang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Yushan Li
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Xinyi Li
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Hao Li
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Chenqi Bian
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Xinyu Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Halima Jafari
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Ningbo Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Chuzhao Lei
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| |
Collapse
|
49
|
Moon JH, Woo J, Park JY, Noh MH, Kim D, Jung GY. Biosensor-guided evolution boosts itaconic acid production, unveiling unique insights into the stringent response. BIORESOURCE TECHNOLOGY 2025; 426:132326. [PMID: 40024573 DOI: 10.1016/j.biortech.2025.132326] [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: 07/26/2024] [Revised: 01/20/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
Acetate is a cost-effective and sustainable carbon source that, despite its potential, remains underutilized. This study employed biosensor-assisted adaptive laboratory evolution (ALE) to enhance itaconic acid production and acetate metabolism in Escherichia coli. The evolved E. coli W strains exhibited 65% increase in itaconic acid production and 71% increase in growth rate, and 45% increase in itaconic acid yield. A common 31-kb genomic deletion was identified in the evolved strains, with two genes, ecw_m2276 and ecw_m2277, driving the observed phenotypic changes. The evolved strains exhibited an intensified stringent response, which enhanced the acetate-utilizing pathway and resulted in over a 5,000% increase in the expression of the glyoxylate shunt, thereby boosting microbial growth. Overexpression of relA further replicated these enhanced phenotypes. Our findings highlight not only significant physiological improvements but also present a novel strategy for enhancing microbial growth and bioproduction from acetate, offering valuable insights for industrial biotechnology applications.
Collapse
Affiliation(s)
- Jo Hyun Moon
- Department of Chemical Engineering, Pohang University of Science and Technology, 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk 37673, Republic of Korea
| | - Jihoon Woo
- School of Energy and Chemical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Joon Young Park
- School of Energy and Chemical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Myung Hyun Noh
- Center for Bio-based Chemistry, Korea Research Institute of Chemical Technology (KRICT), 406-30, Jongga-ro, Jung-gu, Ulsan 44429, Republic of Korea.
| | - Donghyuk Kim
- School of Energy and Chemical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea.
| | - Gyoo Yeol Jung
- Department of Chemical Engineering, Pohang University of Science and Technology, 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk 37673, Republic of Korea; School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk 37673, Republic of Korea.
| |
Collapse
|
50
|
Rusu EC, Clavero-Mestres H, Sánchez-Álvarez M, Veciana-Molins M, Bertran L, Monfort-Lanzas P, Aguilar C, Camaron J, Auguet T. Uncovering hepatic transcriptomic and circulating proteomic signatures in MASH: A meta-analysis and machine learning-based biomarker discovery. Comput Biol Med 2025; 191:110170. [PMID: 40220593 DOI: 10.1016/j.compbiomed.2025.110170] [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: 10/20/2024] [Revised: 04/05/2025] [Accepted: 04/05/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Metabolic-associated steatohepatitis (MASH), the progressive form of metabolic-associated steatotic liver disease (MASLD), poses significant risks for liver fibrosis and cardiovascular complications. Despite extensive research, reliable biomarkers for MASH diagnosis and progression remain elusive. This study aimed to identify hepatic transcriptomic and circulating proteomic signatures specific to MASH, and to develop a machine learning-based biomarker discovery model. METHODS A systematic review of RNA-Seq and proteomic datasets was conducted, retrieving 7 hepatic transcriptomics and 3 circulating proteomics studies, encompassing 483 liver samples and 169 serum/plasma samples, respectively. Differential gene and protein expression analyses were performed, and pathways were enriched using gene set enrichment analysis. A machine learning (ML) model was developed to identify MASH-specific biomarkers, utilizing biologically significant protein ratios. KEY FINDINGS Hepatic transcriptomic analysis identified 5017 differentially expressed genes (DEGs), with significant enrichment of extracellular matrix (ECM) pathways. Serum proteomics revealed six differentially expressed proteins (DEPs), including complement-related proteins. Integration of transcriptomic and proteomic data highlighted the complement cascade as a key pathway in MASH, with discordant regulation between the liver and circulation. The ML-based biomarker discovery model, utilizing protein ratios, achieved an F1 scores of 0.83 and 0.64 in the training sets and 0.67 in an external validation set. CONCLUSION Our findings indicate ECM deregulation and complement system involvement in MASH progression. The novel ML model incorporating protein ratios offers a potential tool for MASH diagnosis. However, further refinement and validation across larger and more diverse cohorts is needed to generalize these results.
Collapse
Affiliation(s)
- Elena Cristina Rusu
- GEMMAIR research Unit (AGAUR) - Applied Medicine (URV). Department of Medicine and Surgery. University Rovira I Virgili (URV), Health Research Institute Pere Virgili (IISPV), 43007, Tarragona, Spain; Institute for Integrative Systems Biology (I2SysBio), University of Valencia and the Spanish National Research Council (CSIC), 46980, Valencia, Spain.
| | - Helena Clavero-Mestres
- GEMMAIR research Unit (AGAUR) - Applied Medicine (URV). Department of Medicine and Surgery. University Rovira I Virgili (URV), Health Research Institute Pere Virgili (IISPV), 43007, Tarragona, Spain.
| | - Mario Sánchez-Álvarez
- GEMMAIR research Unit (AGAUR) - Applied Medicine (URV). Department of Medicine and Surgery. University Rovira I Virgili (URV), Health Research Institute Pere Virgili (IISPV), 43007, Tarragona, Spain.
| | - Marina Veciana-Molins
- GEMMAIR research Unit (AGAUR) - Applied Medicine (URV). Department of Medicine and Surgery. University Rovira I Virgili (URV), Health Research Institute Pere Virgili (IISPV), 43007, Tarragona, Spain.
| | - Laia Bertran
- GEMMAIR research Unit (AGAUR) - Applied Medicine (URV). Department of Medicine and Surgery. University Rovira I Virgili (URV), Health Research Institute Pere Virgili (IISPV), 43007, Tarragona, Spain.
| | - Pablo Monfort-Lanzas
- Institute of Medical Biochemistry, Biocenter, Medical University of Innsbruck, 6020, Innsbruck, Austria; Institute of Bioinformatics, Biocenter, Medical University of Innsbruck, 6020, Innsbruck, Austria.
| | - Carmen Aguilar
- GEMMAIR research Unit (AGAUR) - Applied Medicine (URV). Department of Medicine and Surgery. University Rovira I Virgili (URV), Health Research Institute Pere Virgili (IISPV), 43007, Tarragona, Spain.
| | - Javier Camaron
- Internal Medicine Unit, Joan XXIII University Hospital of Tarragona, 43007, Tarragona, Spain.
| | - Teresa Auguet
- GEMMAIR research Unit (AGAUR) - Applied Medicine (URV). Department of Medicine and Surgery. University Rovira I Virgili (URV), Health Research Institute Pere Virgili (IISPV), 43007, Tarragona, Spain; Internal Medicine Unit, Joan XXIII University Hospital of Tarragona, 43007, Tarragona, Spain.
| |
Collapse
|