1
|
Sha HN, Lu YM, Zhan PP, Chen J, Qiu QF, Xiong JB. Beneficial effects of probiotics on Litopenaeus vannamei growth and immune function via the recruitment of gut Rhodobacteraceae symbionts. Zool Res 2025; 46:388-400. [PMID: 40091533 PMCID: PMC12000132 DOI: 10.24272/j.issn.2095-8137.2024.364] [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: 12/19/2024] [Accepted: 01/14/2025] [Indexed: 03/19/2025] Open
Abstract
Probiotic supplementation enhances the abundance of gut-associated Rhodobacteraceae species, critical symbionts contributing to the health and physiological fitness of Litopenaeus vannamei. Understanding the role of Rhodobacteraceae in shaping the shrimp gut microbiota is essential for optimizing probiotic application. This study investigated whether probiotics benefit shrimp health and fitness via the recruitment of Rhodobacteraceae commensals in the gut. Probiotic supplementation significantly enhanced feed conversion efficiency, digestive enzyme activity, and immune responses, thereby promoting shrimp growth. Additionally, probiotics induced pronounced shifts in gut microbial composition, enriched gut Rhodobacteraceae abundance, and reduced community variability, leading to a more stable gut microbiome. Network analysis revealed that the removal of Rhodobacteraceae nodes disrupted gut microbial connectivity more rapidly than the removal of non-Rhodobacteraceae nodes, indicating a disproportionate role of Rhodobacteraceae in maintaining network stability. Probiotic supplementation facilitated the migration of Rhodobacteraceae taxa from the aquatic environment to the shrimp gut while reinforcing deterministic selection in gut microbiota assembly. Transcriptomic analysis revealed that up-regulation of amino acid metabolism and NF-κB signaling pathways was positively correlated with Rhodobacteraceae abundance. These findings demonstrate that probiotic supplementation enriches key Rhodobacteraceae taxa, stabilizes gut microbial networks, and enhances host digestive and immune functions, ultimately improving shrimp growth performance. This study provides novel perspectives on the ecological and molecular mechanisms underlying the beneficial effects of probiotics on shrimp fitness.
Collapse
Affiliation(s)
- Hao-Nan Sha
- State Key Laboratory for Quality and Safety of Agro-Products, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang 315211, China
- Key Laboratory of Aquacultural Biotechnology, Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Yang-Ming Lu
- State Key Laboratory for Quality and Safety of Agro-Products, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang 315211, China
- Key Laboratory of Aquacultural Biotechnology, Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Ping-Ping Zhan
- Key Laboratory of Aquacultural Biotechnology, Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Jiong Chen
- State Key Laboratory for Quality and Safety of Agro-Products, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang 315211, China
- Key Laboratory of Aquacultural Biotechnology, Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Qiong-Fen Qiu
- Key Laboratory of Aquacultural Biotechnology, Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Jin-Bo Xiong
- State Key Laboratory for Quality and Safety of Agro-Products, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang 315211, China
- Key Laboratory of Aquacultural Biotechnology, Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang 315211, China. E-mail:
| |
Collapse
|
2
|
Meiring S, Aydin Ö, van Baar ACG, van der Vossen EWJ, Rampanelli E, van Grieken NCT, Holleman F, Nieuwdorp M, Bergman JJGHM. From Endoscopic Inspection to Gene-Expression: A Thorough Assessment of the Duodenal Mucosa After Resurfacing-A Prospective Study. Dig Dis Sci 2025; 70:1052-1063. [PMID: 39779586 PMCID: PMC11920325 DOI: 10.1007/s10620-024-08710-4] [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: 02/08/2024] [Accepted: 10/22/2024] [Indexed: 01/11/2025]
Abstract
AIMS Duodenal Mucosal Resurfacing (DMR) is an endoscopic ablation technique aimed at improving glycemia in patients with type 2 diabetes mellitus (T2DM). Although the exact underlying mechanism is still unclear, it is postulated that the DMR-induced improvements are the result of changes in the duodenal mucosa. For this reason, we assessed macroscopic and microscopic changes in the duodenal mucosa induced by DMR + GLP-1RA. METHODS We included 16 patients with T2DM using basal insulin that received a combination treatment of a single DMR and GLP-1RA. Endoscopic evaluation was performed before the DMR procedure and 3 month after, and duodenal biopsies were obtained. Histological evaluation was performed and L and K cell density was calculated. In addition, gene-expression analysis and Western blotting was performed. RESULTS Endoscopic evaluation at 3 month showed duodenal mucosa with a normal appearance. In line, microscopic histological evaluation showed no signs of villous atrophy or inflammation and unchanged L and K cell density. Unbiased transcriptome profiling and western blotting revealed that PDZK1 expression was higher in responders at baseline and after DMR. GATA6 expression was significantly increased in responders after DMR compared to non-responders. CONCLUSION The absence of macroscopic and microscopic changes after 3 month suggest that improvements in glycemic parameters after DMR do not result from significant histological changes in duodenal mucosa. It is more likely that these improvements result from more subtle changes in enteroendocrine signaling. PDZK1 and GATA6 expression might play a role in DMR; this needs to be confirmed in pre-clinical studies.
Collapse
Affiliation(s)
- S Meiring
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centres, Location AMC, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
| | - Ö Aydin
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centres, Location AMC, Amsterdam, The Netherlands
- Department of Bariatric Surgery, Spaarne Gasthuis, Hoofddorp, The Netherlands
| | - A C G van Baar
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centres, Location AMC, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands.
| | - E W J van der Vossen
- Department of Experimental Vascular Medicine, Amsterdam University Medical Centres, Location AMC, Amsterdam, The Netherlands
| | - E Rampanelli
- Department of Experimental Vascular Medicine, Amsterdam University Medical Centres, Location AMC, Amsterdam, The Netherlands
| | - N C T van Grieken
- Department of Pathology, Amsterdam University Medical Centres, Location VUmc, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - F Holleman
- Department of Internal Medicine, Amsterdam University Medical Centres, Location AMC, Amsterdam, The Netherlands
| | - M Nieuwdorp
- Department of Experimental Vascular Medicine, Amsterdam University Medical Centres, Location AMC, Amsterdam, The Netherlands
| | - J J G H M Bergman
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centres, Location AMC, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
| |
Collapse
|
3
|
Zhang Y, Ma R, Suolangduoerji, Ma S, Nuertai A, He K, Liu H, Zhu Y. Annual cycle variations in the gut microbiota of migratory black-necked cranes. Front Microbiol 2025; 16:1533282. [PMID: 39990144 PMCID: PMC11844351 DOI: 10.3389/fmicb.2025.1533282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 01/16/2025] [Indexed: 02/25/2025] Open
Abstract
Introduction Migratory birds exhibit unique annual cycles that complicate their gut microbiota. However, the annual dynamics of gut microbiota in migratory birds remain unclear, hindering our understanding of their environmental adaptation. Methods Here, we collected fecal samples from black-necked cranes (Grus nigricollis) across four seasons at their breeding grounds and used wintering ground data from databases to characterize their gut microbial compositions throughout the year. Results and discussion The results showed that the gut microbiota was clustered by season (Bray-Curtis: R 2 = 0.348, p < 0.001; UniFrac: R 2 = 0.352, p < 0.001). And the summer samples exhibited higher alpha (Simpson and Shannon), beta diversity (Bray-Curtis and UniFrac) and more diverse functions in gut microbiota compared to other seasons. Furthermore, in summer, the gut microbiota exhibited several balanced relative abundances at the family level, whereas Lactobacillaceae family dominated during the other seasons. Thirty-six ASVs were identified by random forest analysis to distinguish samples from distinct seasons. Despite having greater diversity, the summer gut microbiota had a simpler network structure than the other seasons (fewer edges and nodes). The dispersal limitation during random processes also significantly influenced gut microbial community assembly. Overall, the gut microbiota of the black-necked crane undergoes dynamic adjustments to adapt to seasonal environmental changes, which may be associated with the variations in diet across seasons. These results enhance our understanding of the gut microbiota of wild migratory birds and support further research on black-necked cranes.
Collapse
Affiliation(s)
- Yujia Zhang
- College of Animal Science and Veterinary Medicine, Southwest Minzu University, Chengdu, Sichuan, China
| | - Ruifeng Ma
- College of Grassland Resources, Institute of Qinghai-Tibetan Plateau, Sichuan Provincial Forest and Grassland Key Laboratory of Alpine Grassland Conservation and Utilization of Qinghai-Tibetan Plateau, Southwest Minzu University, Chengdu, Sichuan, China
| | - Suolangduoerji
- Sichuan Ruoergai Wetland National Nature Reserve Administration, Ruoergai, Ruoergai, Aba Tibetan and Qiang Autonomous Prefecture, China
| | - Shujuan Ma
- College of Grassland Resources, Institute of Qinghai-Tibetan Plateau, Sichuan Provincial Forest and Grassland Key Laboratory of Alpine Grassland Conservation and Utilization of Qinghai-Tibetan Plateau, Southwest Minzu University, Chengdu, Sichuan, China
- Luxian NO.1 High School, Luzhou, Luzhou, Sichuan, China
| | - Akebota Nuertai
- College of Grassland Resources, Institute of Qinghai-Tibetan Plateau, Sichuan Provincial Forest and Grassland Key Laboratory of Alpine Grassland Conservation and Utilization of Qinghai-Tibetan Plateau, Southwest Minzu University, Chengdu, Sichuan, China
| | - Ke He
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang A & F University, Hangzhou, China
| | - Hongyi Liu
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang A & F University, Hangzhou, China
| | - Ying Zhu
- College of Grassland Resources, Institute of Qinghai-Tibetan Plateau, Sichuan Provincial Forest and Grassland Key Laboratory of Alpine Grassland Conservation and Utilization of Qinghai-Tibetan Plateau, Southwest Minzu University, Chengdu, Sichuan, China
| |
Collapse
|
4
|
Patel RA, Panche AN, Harke SN. Gut microbiome-gut brain axis-depression: interconnection. World J Biol Psychiatry 2025; 26:1-36. [PMID: 39713871 DOI: 10.1080/15622975.2024.2436854] [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: 08/07/2024] [Revised: 11/26/2024] [Accepted: 11/28/2024] [Indexed: 12/24/2024]
Abstract
OBJECTIVES The relationship between the gut microbiome and mental health, particularly depression, has gained significant attention. This review explores the connection between microbial metabolites, dysbiosis, and depression. The gut microbiome, comprising diverse microorganisms, maintains physiological balance and influences health through the gut-brain axis, a communication pathway between the gut and the central nervous system. METHODS Dysbiosis, an imbalance in the gut microbiome, disrupts this axis and worsens depressive symptoms. Factors like diet, antibiotics, and lifestyle can cause this imbalance, leading to changes in microbial composition, metabolism, and immune responses. This imbalance can induce inflammation, disrupt neurotransmitter regulation, and affect hormonal and epigenetic processes, all linked to depression. RESULTS Microbial metabolites, such as short-chain fatty acids and neurotransmitters, are key to gut-brain communication, influencing immune regulation and mood. The altered production of these metabolites is associated with depression. While progress has been made in understanding the gut-brain axis, more research is needed to clarify causative relationships and develop new treatments. The emerging field of psychobiotics and microbiome-targeted therapies shows promise for innovative depression treatments by harnessing the gut microbiome's potential. CONCLUSIONS Epigenetic mechanisms, including DNA methylation and histone modifications, are crucial in how the gut microbiota impacts mental health. Understanding these mechanisms offers new prospects for preventing and treating depression through the gut-brain axis.
Collapse
Affiliation(s)
- Ruhina Afroz Patel
- Institute of Biosciences and Technology, MGM University, Aurangabad, India
| | - Archana N Panche
- Institute of Biosciences and Technology, MGM University, Aurangabad, India
| | - Sanjay N Harke
- Institute of Biosciences and Technology, MGM University, Aurangabad, India
| |
Collapse
|
5
|
Peng C, Ghanbari M, May A, Abeel T. Effects of antibiotic growth promoter and its natural alternative on poultry cecum ecosystem: an integrated analysis of gut microbiota and host expression. Front Microbiol 2024; 15:1492270. [PMID: 39687871 PMCID: PMC11646981 DOI: 10.3389/fmicb.2024.1492270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 11/05/2024] [Indexed: 12/18/2024] Open
Abstract
Background In-feed antibiotic growth promoters (AGPs) have been a cornerstone in the livestock industry due to their role in enhancing growth and feed efficiency. However, concerns over antibiotic resistance have driven a shift away from AGPs toward natural alternatives. Despite the widespread use, the exact mechanisms of AGPs and alternatives are not fully understood. This necessitates holistic studies that investigate microbiota dynamics, host responses, and the interactions between these elements in the context of AGPs and alternative feed additives. Methods In this study, we conducted a multifaceted investigation of how Bacitracin, a common AGP, and a natural alternative impact both cecum microbiota and host expression in chickens. In addition to univariate and static differential abundance and expression analyses, we employed multivariate and time-course analyses to study this problem. To reveal host-microbe interactions, we assessed their overall correspondence and identified treatment-specific pairs of species and host expressed genes that showed significant correlations over time. Results Our analysis revealed that factors such as developmental age substantially impacted the cecum ecosystem more than feed additives. While feed additives significantly altered microbial compositions in the later stages, they did not significantly affect overall host gene expression. The differential expression indicated that with AGP administration, host transmembrane transporters and metallopeptidase activities were upregulated around day 21. Together with the modulated kininogen binding and phenylpyruvate tautomerase activity over time, this likely contributes to the growth-promoting effects of AGPs. The difference in responses between AGP and PFA supplementation suggests that these additives operate through distinct mechanisms. Conclusion We investigated the impact of a common AGP and its natural alternative on poultry cecum ecosystem through an integrated analysis of both the microbiota and host responses. We found that AGP appears to enhance host nutrient utilization and modulate immune responses. The insights we gained are critical for identifying and developing effective AGP alternatives to advance sustainable livestock farming practices.
Collapse
Affiliation(s)
- Chengyao Peng
- Delft Bioinformatics Lab, Delft University of Technology, Delft, Netherlands
| | - Mahdi Ghanbari
- dsm-firmenich, Animal Nutrition and Health R&D Center, Tulln, Austria
| | - Ali May
- dsm-firmenich, Science and Research, Delft, Netherlands
| | - Thomas Abeel
- Delft Bioinformatics Lab, Delft University of Technology, Delft, Netherlands
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| |
Collapse
|
6
|
Pepke ML, Hansen SB, Limborg MT. Unraveling host regulation of gut microbiota through the epigenome-microbiome axis. Trends Microbiol 2024; 32:1229-1240. [PMID: 38839511 DOI: 10.1016/j.tim.2024.05.006] [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: 01/26/2024] [Revised: 05/13/2024] [Accepted: 05/15/2024] [Indexed: 06/07/2024]
Abstract
Recent studies of dynamic interactions between epigenetic modifications of a host organism and the composition or activity of its associated gut microbiota suggest an opportunity for the host to shape its microbiome through epigenetic alterations that lead to changes in gene expression and noncoding RNA activity. We use insights from microbiota-induced epigenetic changes to review the potential of the host to epigenetically regulate its gut microbiome, from which a bidirectional 'epigenome-microbiome axis' emerges. This axis embeds environmentally induced variation, which may influence the adaptive evolution of host-microbe interactions. We furthermore present our perspective on how the epigenome-microbiome axis can be understood and investigated within a holo-omic framework with potential applications in the applied health and food sciences.
Collapse
Affiliation(s)
- Michael L Pepke
- Center for Evolutionary Hologenomics, Globe Institute, Faculty of Health and Medical Sciences, University of Copenhagen, Øster Farimagsgade 5, DK-1353 Copenhagen, Denmark.
| | - Søren B Hansen
- Center for Evolutionary Hologenomics, Globe Institute, Faculty of Health and Medical Sciences, University of Copenhagen, Øster Farimagsgade 5, DK-1353 Copenhagen, Denmark
| | - Morten T Limborg
- Center for Evolutionary Hologenomics, Globe Institute, Faculty of Health and Medical Sciences, University of Copenhagen, Øster Farimagsgade 5, DK-1353 Copenhagen, Denmark.
| |
Collapse
|
7
|
Iurk VB, Ingles M, Correa GS, Silva CR, Staichak G, Pileggi SAV, Christo SW, Domit C, Pileggi M. The potential influence of microplastics on the microbiome and disease susceptibility in sea turtles. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 946:174298. [PMID: 38944299 DOI: 10.1016/j.scitotenv.2024.174298] [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: 10/01/2023] [Revised: 06/10/2024] [Accepted: 06/24/2024] [Indexed: 07/01/2024]
Abstract
Microplastics (MPs) are particles with sizes of ≤5 mm formed when plastic materials break down. These contaminants are often found in marine environments, making it easy for sea turtles to ingest them and for their microbiome to be exposed. MPs can disrupt microbiome balance, leading to dysbiosis and making organisms more susceptible to diseases. Owing to the significance of these processes, it is crucial to dedicate research to studying the metabolic and genetic analysis of the gut microbiome in sea turtles. The objective of this study was to describe the effects of exposure to MPs on the gut microbiome of sea turtles, based on current knowledge. This review also aimed to explore the potential link between MP exposure and disease susceptibility in these animals. We show that the metabolites produced by the gut microbiome, such as short-chain fatty acids (SCFAs), polyamines, and polysaccharide A, can regulate the expression of host genes. Regulation occurs through various mechanisms, including histone acetylation, DNA methylation, and the modulation of cytokine gene expression. These processes are essential for preserving the integrity of the gut mucosa and enhancing the functionality of immune cells. Exposure to MPs disrupts the gut microbiome and alters gene expression, leading to immune system disturbances in sea turtles. This vulnerability makes turtles more susceptible to opportunistic microorganisms such as chelonid alphaherpesvirus 5 (ChAHV5), which is linked to the development of fibropapillomatosis (FP). Additionally, targeted dietary interventions or the use of live microorganisms such as probiotics can help restore microbial biodiversity and recover lost metabolic pathways. The goal of these interventions is to restore the functionality of the immune system in sea turtles undergoing rehabilitation at specialized centers. The gut microbiome plays a crucial role in sea turtle health, sparking discussions and investigations that can potentially lead to promising treatments for these animals.
Collapse
Affiliation(s)
- Vitória Bonfim Iurk
- Laboratório de Ecologia e Conservação, Centro de Estudos do Mar, Universidade Federal do Paraná, PR 832555-000, Brazil; Laboratório de Microbiologia Ambiental, Departamento de Biologia Estrutural, Molecular e Genética, Setor de Ciências Biológicas e da Saúde, Universidade Estadual de Ponta Grossa, PR 84030-000, Brazil
| | - Mariana Ingles
- Laboratório de Ecologia e Conservação, Centro de Estudos do Mar, Universidade Federal do Paraná, PR 832555-000, Brazil
| | - Giovana Sequinel Correa
- Laboratório de Virologia Aplicada, Centro de Ciências Biológicas, Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de Santa Catarina, SC 88040-900, Brazil
| | - Caroline Rosa Silva
- Departamento de Biotecnologia, Genética e Biologia Celular, Universidade Estadual de Maringá, PR 87020-900, Brazil
| | - Gabriel Staichak
- Instituto de Biociências da Universidade Federal de Mato Grosso, Universidade Federal do Mato Grosso, MT 79070-900, Brazil
| | - Sônia Alvim Veiga Pileggi
- Laboratório de Microbiologia Ambiental, Departamento de Biologia Estrutural, Molecular e Genética, Setor de Ciências Biológicas e da Saúde, Universidade Estadual de Ponta Grossa, PR 84030-000, Brazil.
| | - Susete Wambier Christo
- Laboratório de Zoologia, Departamento de Biologia Geral, Setor de Ciências Biológicas e da Saúde, Universidade Estadual de Ponta Grossa, PR 84030-000, Brazil
| | - Camila Domit
- Laboratório de Ecologia e Conservação, Centro de Estudos do Mar, Universidade Federal do Paraná, PR 832555-000, Brazil.
| | - Marcos Pileggi
- Laboratório de Microbiologia Ambiental, Departamento de Biologia Estrutural, Molecular e Genética, Setor de Ciências Biológicas e da Saúde, Universidade Estadual de Ponta Grossa, PR 84030-000, Brazil.
| |
Collapse
|
8
|
Wang ZY, Xie WQ, Xiang ZY, Zhang CY, Xie YG, Quah RYC, Ding GH. Exploring the effects of environmentally relevant concentrations of tris(2-chloroethyl) phosphate on tadpole health: A comprehensive analysis of intestinal microbiota and hepatic transcriptome. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 946:174428. [PMID: 38964390 DOI: 10.1016/j.scitotenv.2024.174428] [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: 03/31/2024] [Revised: 06/10/2024] [Accepted: 06/30/2024] [Indexed: 07/06/2024]
Abstract
Tris(2-chloroethyl) phosphate (TCEP), a chlorinated organophosphate ester, is commonly found in aquatic environments. Due to its various toxic effects, it may pose a risk to the health of aquatic organisms. However, the potential impacts of TCEP exposure on the intestinal microbiota and hepatic function in amphibians have not been reported. This study investigated the impact of long-term exposure to environmentally relevant concentrations of TCEP (0, 3, and 90 μg/L) on the intestinal microbiota and hepatic transcriptome of Polypedates megacephalus tadpoles. The results showed that the body size of the tadpoles decreased significantly with an increase in TCEP concentration. Additionally, TCEP exposure affected the diversity and composition of the intestinal microbiota in tadpoles, leading to significant changes in the relative abundance of certain bacterial groups (the genera Aeromonas decreased and Citrobacter increased) and potentially promoting a more even distribution of microbial species, as indicated by a significant increase in the Simpson index. Moreover, the impact of TCEP on hepatic gene expression profiles in tadpoles was significant, with the majority of differentially expressed genes (DEGs) (709 out of 906 total DEGs in 3 μg/L of TCEP versus control, and 344 out of 387 DEGs in 90 μg/L of TCEP versus control) being significantly down-regulated, which were primarily related to immune response and immune system process. Notably, exposure to TCEP significantly reduced the relative abundance of the genera Aeromonas and Cetobacterium in the tadpole intestine. This reduction was positively correlated with the down-regulated expression of immune-related genes in the liver of corresponding tadpoles. In summary, these findings provide empirical evidence of the potential health risks to tadpoles exposed to TCEP at environmentally relevant concentrations.
Collapse
Affiliation(s)
- Zi-Ying Wang
- Laboratory of Amphibian Diversity Investigation, College of Ecology, Lishui University, Lishui, Zhejiang, China; College of Animal Science and Technology, Zhejiang A & F University, Lin'an, Zhejiang, China
| | - Wen-Qi Xie
- Laboratory of Amphibian Diversity Investigation, College of Ecology, Lishui University, Lishui, Zhejiang, China
| | - Zi-Yong Xiang
- Laboratory of Amphibian Diversity Investigation, College of Ecology, Lishui University, Lishui, Zhejiang, China
| | - Chi-Ying Zhang
- Laboratory of Amphibian Diversity Investigation, College of Ecology, Lishui University, Lishui, Zhejiang, China
| | - Yi-Ge Xie
- Laboratory of Amphibian Diversity Investigation, College of Ecology, Lishui University, Lishui, Zhejiang, China
| | - Roy You Chen Quah
- Department of Biological Sciences, National University of Singapore, Singapore
| | - Guo-Hua Ding
- Laboratory of Amphibian Diversity Investigation, College of Ecology, Lishui University, Lishui, Zhejiang, China.
| |
Collapse
|
9
|
Huang Q, Wen C, Gu S, Jie Y, Li G, Yan Y, Tian C, Wu G, Yang N. Synergy of gut microbiota and host genome in driving heterosis expression of chickens. J Genet Genomics 2024; 51:1121-1134. [PMID: 38950856 DOI: 10.1016/j.jgg.2024.06.011] [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: 04/09/2024] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 07/03/2024]
Abstract
Heterosis has been widely utilized in agricultural production. Despite over a century of extensive research, the underlying mechanisms of heterosis remain elusive. Most hypotheses and research have focused on the genetic basis of heterosis. However, the potential role of gut microbiota in heterosis has been largely ignored. Here, we carefully design a crossbreeding experiment with two distinct broiler breeds and conduct 16S rRNA amplicon and transcriptome sequencing to investigate the synergistic role of gut microbiota and host genes in driving heterosis. We find that the breast muscle weight of hybrids exhibits a high heterosis, 6.28% higher than the mid-parent value. A notable difference is observed in the composition and potential function of cecal microbiota between hybrids and their parents. Over 90% of differentially colonized microbiota and differentially expressed genes exhibit nonadditive patterns. Integrative analyses uncover associations between nonadditive genes and nonadditive microbiota, including a connection between the expression of cellular signaling pathways and metabolism-related genes and the abundance of Odoribacter, Oscillibacter, and Alistipes in hybrids. Moreover, higher abundances of these microbiota are related to better meat yield. In summary, these findings highlight the importance of gut microbiota in heterosis, serving as crucial factors that modulate heterosis expression in chickens.
Collapse
Affiliation(s)
- Qiang Huang
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing 100193, China; National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Chaoliang Wen
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing 100193, China; National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; Sanya Institute of China Agricultural University, Hainan 572025, China.
| | - Shuang Gu
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing 100193, China; National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yuchen Jie
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing 100193, China; National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Guangqi Li
- Beijing Huadu Yukou Poultry Industry Co. Ltd., Beijing 101206, China
| | - Yiyuan Yan
- Beijing Huadu Yukou Poultry Industry Co. Ltd., Beijing 101206, China
| | - Chuanyao Tian
- Beijing Huadu Yukou Poultry Industry Co. Ltd., Beijing 101206, China
| | - Guiqin Wu
- Beijing Huadu Yukou Poultry Industry Co. Ltd., Beijing 101206, China
| | - Ning Yang
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center for Molecular Design Breeding, China Agricultural University, Beijing 100193, China; National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; Sanya Institute of China Agricultural University, Hainan 572025, China.
| |
Collapse
|
10
|
Kim JA, Park YS, Kim JH, Choi CY. Impact of water temperature on oxidative stress and intestinal microbiota in pearl-spot chromis, Chromis notata (Temminck & Schlegel, 1843). Comp Biochem Physiol B Biochem Mol Biol 2024; 275:111029. [PMID: 39243810 DOI: 10.1016/j.cbpb.2024.111029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 09/03/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
Water temperature is an abiotic factor influencing fish metabolism and physiological responses. As poikilothermic creatures, fish are notable sensitivity to fluctuations in water temperature, which also significantly influences intestinal microbial proliferation. This study aimed to investigate the impact of both low (8 °C) and high (28 °C) water temperatures on oxidative stress and the intestinal microbiota of Chromis notata, a species that has recently migrated northward owing to changes in sea water temperature. Laboratory experiments were conducted to assess changes in superoxide dismutase (SOD), catalase (CAT), and lysozyme activities, as well as changes in the abundance and diversity of intestinal microbiota. The activities of antioxidant enzymes, specifically SOD and CAT, in C. notata exposed to low and high temperatures, showed an increase compared to the control group (maintained at 18 °C). Moreover, liver H2O2 levels exhibited a significant increase over time. Conversely, plasma lysozyme activity significantly decreased in groups subjected to low and high water temperatures compared to the control group. Analyzing changes in the intestinal microbiota, we observed an increase in the proportion of Firmicutes but a decrease in Proteobacteria, which are known for their role in immune enhancement, in C. notata exposed to both low and high water temperatures. We propose that alterations in water temperature impact the antioxidant enzyme activity of C. notata, leading to compromised immune responses and disruption of the biological balance of the intestinal microbiota, potentially affecting the host's survival.
Collapse
Affiliation(s)
- Jin A Kim
- Department of Convergence Study on the Ocean Science and Technology, National Korea Maritime and Ocean University, Busan 49112, Republic of Korea
| | - Young-Su Park
- Department of Nursing, Catholic University of Pusan, Busan 46252, Republic of Korea
| | - Jun-Hwan Kim
- Department of Aquatic Life Medicine, Jeju National University, Jeju 63243, Republic of Korea.
| | - Cheol Young Choi
- Department of Convergence Study on the Ocean Science and Technology, National Korea Maritime and Ocean University, Busan 49112, Republic of Korea; Division of Marine BioScience, National Korea Maritime and Ocean University, Busan 49112, Republic of Korea.
| |
Collapse
|
11
|
Ou X, Chen J, Li B, Yang Y, Liu X, Xu Z, Xiang X, Wang Q. Multiomics reveals the ameliorating effect and underlying mechanism of aqueous extracts of polygonatum sibiricum rhizome on obesity and liver fat accumulation in high-fat diet-fed mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155843. [PMID: 38971026 DOI: 10.1016/j.phymed.2024.155843] [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: 09/18/2023] [Revised: 05/21/2024] [Accepted: 06/24/2024] [Indexed: 07/08/2024]
Abstract
BACKGROUND Polygonatum sibiricum polysaccharides protect against obesity and NAFLD. However, the potential effects of PS rhizome aqueous extracts (PSRwe) on adiposity and hepatic lipid accumulation remains unexplored. PURPOSE Elucidating the impact and underlying mechanism of PSRwe on HFD-induced obesity and liver fat depostition. STUDY DESIGN 56 male mice, aged eight weeks, were divided into seven groups: Positive, four doses of PSRwe, Model, and Control. HFD was fed for eight weeks, followed by alternate-day gavage of orlistat and PSRwe for an additional eight-week period. Integrative analysis encompassing multiomics, physiological and histopathological, and biochemical indexes was employed. METHODS Body weight (BW); liver, fat and Lee's indexes; TC, TG, LDL-C, HDL-C, AST, ALT, FFA, leptin, and adiponectin in the liver and blood; TNFα, IL-6, and LPS in the colon, plasma, and liver; H&E, PAS and oil red O staining on adipose and liver samples were examined. OGTT and ITT were conducted The gut microbiome, microbial metabolome, colonic and liver transcriptome, plasma and liver metabolites were investigated. RESULTS PSRwe at the dosage of 7.5 mg/kg demonstrated significant and consistent reduction in BW and hepatic fat deposition than orlistat. PSRwe significantly decreased TC, TG, LDL-C, LEP, FFA levels in blood and liver. PSRwe significantly enhanced the relative abundance of probiotics including Akkermansia muciniphila, Bifidobacterium pseudolongum, Lactobacillus reuteri, and metabolic pathways including glycolysis and fatty acids β-oxidation. The 70 up-regulated microbial metabolites in PSRwe-treated mice mainly involved in nucleotides and amino acids metabolism, while 40 decreased metabolites primarily associated with lipid metabolism. The up-regulated colonic differentially expressed genes (DEGs) participate in JAK-STAT/PI3K-Akt/FoxO signaling pathway, serotonergic/cholinergic/glutamatergic synapses, while the down-regulated DEGs predominantly focused on fat absorption and transport. The up-regulated liver DEGs mainly concentrated on fatty acid oxidation and metabolism. Liver metabolisms revealed 131 differential metabolites, among which carnitine and oxidized lipids significantly increased in PSRwe-treated mice. In plasma, the 58 up-regulated metabolites mainly participate in co-factors/vitamins metabolism while 154 down-regulated ones in fatty acids biosynthesis. Comprehensive multiomics association analysis revealed significant associations between gut microbiota and colonic/liver gene expression, and suggested exogenous and endogenous betaine may be active compound in alleviating HFD-induced symptoms. CONCLUSION PSRwe effectively mitigate HFD-induced obesity and hepatic steatosis by increasing beneficial bacteria, reducing colonic fat digestion/absorption, increasing hepatic lipid metabolism, and elevating betaine levels.
Collapse
Affiliation(s)
- Xiaobin Ou
- Gansu Key Laboratory of Protection and Utlization for Biological Resources and Ecological Restoration in Longdong, Longdong University, Qingyang 745000, Gansu, China; College of Life Sciences and Technology, Longdong University, Qingyang 745000, Gansu, China.
| | - Juanjuan Chen
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou 730030, Gansu, China
| | - Boping Li
- Gansu Key Laboratory of Protection and Utlization for Biological Resources and Ecological Restoration in Longdong, Longdong University, Qingyang 745000, Gansu, China; College of Medicine, Longdong University, Qingyang 745000, Gansu, China
| | - Yan Yang
- Department of Endocrinology and Metabolism, Lanzhou University Second Hospital, Lanzhou 730030, Gansu, China
| | - Xiuli Liu
- Gansu Key Laboratory of Protection and Utlization for Biological Resources and Ecological Restoration in Longdong, Longdong University, Qingyang 745000, Gansu, China; College of Life Sciences and Technology, Longdong University, Qingyang 745000, Gansu, China
| | - Zaoxu Xu
- Gansu Key Laboratory of Protection and Utlization for Biological Resources and Ecological Restoration in Longdong, Longdong University, Qingyang 745000, Gansu, China; College of Life Sciences and Technology, Longdong University, Qingyang 745000, Gansu, China
| | - Xuesong Xiang
- National Institute of Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Qi Wang
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou 730030, Gansu, China.
| |
Collapse
|
12
|
Jin DM, Morton JT, Bonneau R. Meta-analysis of the human gut microbiome uncovers shared and distinct microbial signatures between diseases. mSystems 2024; 9:e0029524. [PMID: 39078158 PMCID: PMC11334437 DOI: 10.1128/msystems.00295-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 05/08/2024] [Indexed: 07/31/2024] Open
Abstract
Microbiome studies have revealed gut microbiota's potential impact on complex diseases. However, many studies often focus on one disease per cohort. We developed a meta-analysis workflow for gut microbiome profiles and analyzed shotgun metagenomic data covering 11 diseases. Using interpretable machine learning and differential abundance analysis, our findings reinforce the generalization of binary classifiers for Crohn's disease (CD) and colorectal cancer (CRC) to hold-out cohorts and highlight the key microbes driving these classifications. We identified high microbial similarity in disease pairs like CD vs ulcerative colitis (UC), CD vs CRC, Parkinson's disease vs type 2 diabetes (T2D), and schizophrenia vs T2D. We also found strong inverse correlations in Alzheimer's disease vs CD and UC. These findings, detected by our pipeline, provide valuable insights into these diseases. IMPORTANCE Assessing disease similarity is an essential initial step preceding a disease-based approach for drug repositioning. Our study provides a modest first step in underscoring the potential of integrating microbiome insights into the disease similarity assessment. Recent microbiome research has predominantly focused on analyzing individual diseases to understand their unique characteristics, which by design excludes comorbidities in individuals. We analyzed shotgun metagenomic data from existing studies and identified previously unknown similarities between diseases. Our research represents a pioneering effort that utilizes both interpretable machine learning and differential abundance analysis to assess microbial similarity between diseases.
Collapse
Affiliation(s)
- Dong-Min Jin
- Center for Genomics and Systems Biology, New York University, New York, New York, USA
| | - James T. Morton
- Center for Computational Biology, Flatiron Institute, Simons Foundation, New York, New York, USA
| | - Richard Bonneau
- Center for Genomics and Systems Biology, New York University, New York, New York, USA
- Genentech, New York, New York, USA
| |
Collapse
|
13
|
Ren J, Li Y, Ni H, Zhang Y, Zhao P, Xiao Q, Hong X, Zhang Z, Yin Y, Li X, Zhang Y, Yang Y. Gut microbiota derived from fecal microbiota transplantation enhances body weight of Mimas squabs. Anim Biosci 2024; 37:1428-1439. [PMID: 38575121 PMCID: PMC11222855 DOI: 10.5713/ab.23.0475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 01/17/2024] [Accepted: 02/08/2024] [Indexed: 04/06/2024] Open
Abstract
OBJECTIVE Compared to Mimas pigeons, Shiqi pigeons exhibit greater tolerance to coarse feeding because of their abundant gut microbiota. Here, to investigate the potential of utilizing intestinal flora derived from Shiqi pigeons, the intestinal flora and body indices of Mimas squabs were evaluated after fecal microbiota transplantation (FMT) from donors. METHODS A total of 90 one-day-old squabs were randomly divided into the control group (CON), the low-concentration group (LC) and the high-concentration group (HC): gavaged with 200 μL of bacterial solution at concentrations of 0, 0.1, and 0.2 g/15 mL, respectively. RESULTS The results suggested that FMT improved the body weight of Mimas squabs in the HC and LC groups (p<0.01), and 0.1 g/15 mL was the optimal dose during FMT. After 16S rRNA sequencing was performed, compared to those in the CON group, the abundance levels of microflora, especially Lactobacillus, Muribaculaceae, and Megasphaera (p<0.05), in the FMT-treated groups were markedly greater. Random forest analysis indicated that the main functions of key microbes involve pathways associated with metabolism, further illustrating their important role in the host body. CONCLUSION FMT has been determined to be a viable method for augmenting the weight and intestinal microbiota of squabs, representing a unique avenue for enhancing the economic feasibility of squab breeding.
Collapse
Affiliation(s)
- Jing Ren
- College of Animal Science, Jilin University, Changchun 130062,
China
| | - Yumei Li
- College of Animal Science, Jilin University, Changchun 130062,
China
| | - Hongyu Ni
- College of Animal Science, Jilin University, Changchun 130062,
China
| | - Yan Zhang
- College of Animal Science and Technology, Jilin Agricultural Science and Technology University, Jilin 132109,
China
| | - Puze Zhao
- College of Animal Science, Jilin University, Changchun 130062,
China
| | - Qingxing Xiao
- College of Animal Science, Jilin University, Changchun 130062,
China
| | - Xiaoqing Hong
- College of Animal Science, Jilin University, Changchun 130062,
China
| | - Ziyi Zhang
- College of Animal Science, Jilin University, Changchun 130062,
China
| | - Yijing Yin
- College of Animal Science, Jilin University, Changchun 130062,
China
| | - Xiaohui Li
- Center of Animal Experiment, College of Basic Medical Sciences, Jilin University, Changchun 130021,
China
| | - Yonghong Zhang
- College of Animal Science, Jilin University, Changchun 130062,
China
| | - Yuwei Yang
- College of Animal Science, Jilin University, Changchun 130062,
China
| |
Collapse
|
14
|
Kim B, Song A, Son A, Shin Y. Gut microbiota and epigenetic choreography: Implications for human health: A review. Medicine (Baltimore) 2024; 103:e39051. [PMID: 39029010 PMCID: PMC11398772 DOI: 10.1097/md.0000000000039051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/21/2024] Open
Abstract
The interwoven relationship between gut microbiota and the epigenetic landscape constitutes a pivotal axis in understanding human health and disease. Governed by a myriad of dietary, genetic, and environmental influences, the gut microbiota orchestrates a sophisticated metabolic interplay, shaping nutrient utilization, immune responses, and defenses against pathogens. Recent strides in genomics and metabolomics have shed light on the intricate connections between these microbial influencers and the host's physiological dynamics, presenting a dynamic panorama across diverse disease spectra. DNA methylation and histone modifications, as key players in epigenetics, intricately align with the dynamic orchestration of the gut microbiota. This seamless collaboration, notably evident in conditions like inflammatory bowel disease and obesity, has captured the attention of researchers, prompting an exploration of its nuanced choreography. Nevertheless, challenges abound. Analyzing data is intricate due to the multifaceted nature of the gut microbiota and the limitations of current analytical methods. This underscores the need for a multidisciplinary approach, where diverse disciplines converge to pave innovative research pathways. The integration of insights from microbiome and epigenome studies assumes paramount importance in unraveling the complexities of this intricate partnership. Deciphering the synchronized interactions within this collaboration offers a deeper understanding of these delicate interplays, potentially heralding revolutionary strides in treatment modalities and strategies for enhancing public health.
Collapse
Affiliation(s)
- Bailee Kim
- Crescenta Valley High School, La Crescenta, CA
| | - Angel Song
- Harvard-Westlake School, Studio City, CA
| | - Andrew Son
- Bellarmine College Preparatory, San Jose, CA
| | - Yonghwan Shin
- Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA
| |
Collapse
|
15
|
Anka IZ, Uren Webster TM, Berbel-Filho WM, Hitchings M, Overland B, Weller S, Garcia de Leaniz C, Consuegra S. Microbiome and epigenetic variation in wild fish with low genetic diversity. Nat Commun 2024; 15:4725. [PMID: 38830879 PMCID: PMC11148108 DOI: 10.1038/s41467-024-49162-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 05/23/2024] [Indexed: 06/05/2024] Open
Abstract
Non-genetic sources of phenotypic variation, such as the epigenome and the microbiome, could be important contributors to adaptive variation for species with low genetic diversity. However, little is known about the complex interaction between these factors and the genetic diversity of the host, particularly in wild populations. Here, we examine the skin microbiome composition of two closely-related mangrove killifish species with different mating systems (self-fertilising and outcrossing) under sympatric and allopatric conditions. This allows us to partition the influence of the genotype and the environment on their microbiome and (previously described) epigenetic profiles. We find the diversity and community composition of the skin microbiome are strongly shaped by the environment and, to a lesser extent, by species-specific influences. Heterozygosity and microbiome alpha diversity, but not epigenetic variation, are associated with the fluctuating asymmetry of traits related to performance (vision) and behaviour (aggression). Our study identifies that a proportion of the epigenetic diversity and microbiome differentiation is unrelated to genetic variation, and we find evidence for an associative relationship between microbiome and epigenetic diversity in these wild populations. This suggests that both mechanisms could potentially contribute to variation in species with low genetic diversity.
Collapse
Affiliation(s)
- Ishrat Z Anka
- Department of Biosciences, Centre for Sustainable Aquatic Research, Swansea University, Swansea, Wales, SA2 8PP, UK
- Department of Aquaculture, Chattogram Veterinary and Animal Sciences University, Chattogram, 4225, Bangladesh
| | - Tamsyn M Uren Webster
- Department of Biosciences, Centre for Sustainable Aquatic Research, Swansea University, Swansea, Wales, SA2 8PP, UK
| | - Waldir M Berbel-Filho
- Department of Biology, University of Oklahoma, Norman, OK, 73019, USA
- Department of Biology, University of West Florida, Pensacola, FL, USA
| | - Matthew Hitchings
- Institute of Life Science, Swansea University, Swansea, Wales, SA2 8PP, UK
| | - Benjamin Overland
- Department of Biosciences, Centre for Sustainable Aquatic Research, Swansea University, Swansea, Wales, SA2 8PP, UK
| | - Sarah Weller
- Department of Biosciences, Centre for Sustainable Aquatic Research, Swansea University, Swansea, Wales, SA2 8PP, UK
| | - Carlos Garcia de Leaniz
- Department of Biosciences, Centre for Sustainable Aquatic Research, Swansea University, Swansea, Wales, SA2 8PP, UK
- Marine Research Centre (CIM-UVIGO), Universidade de Vigo, Vigo, Spain
| | - Sofia Consuegra
- Department of Biosciences, Centre for Sustainable Aquatic Research, Swansea University, Swansea, Wales, SA2 8PP, UK.
- Grupo de Biotecnología Acuática, Departamento de Biotecnología y Acuicultura, Instituto de Investigacións Mariñas, IIM-CSIC, Vigo, Spain.
| |
Collapse
|
16
|
Nelson BN, Friedman JE. Developmental Programming of the Fetal Immune System by Maternal Western-Style Diet: Mechanisms and Implications for Disease Pathways in the Offspring. Int J Mol Sci 2024; 25:5951. [PMID: 38892139 PMCID: PMC11172957 DOI: 10.3390/ijms25115951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Maternal obesity and over/undernutrition can have a long-lasting impact on offspring health during critical periods in the first 1000 days of life. Children born to mothers with obesity have reduced immune responses to stimuli which increase susceptibility to infections. Recently, maternal western-style diets (WSDs), high in fat and simple sugars, have been associated with skewing neonatal immune cell development, and recent evidence suggests that dysregulation of innate immunity in early life has long-term consequences on metabolic diseases and behavioral disorders in later life. Several factors contribute to abnormal innate immune tolerance or trained immunity, including changes in gut microbiota, metabolites, and epigenetic modifications. Critical knowledge gaps remain regarding the mechanisms whereby these factors impact fetal and postnatal immune cell development, especially in precursor stem cells in bone marrow and fetal liver. Components of the maternal microbiota that are transferred from mothers consuming a WSD to their offspring are understudied and identifying cause and effect on neonatal innate and adaptive immune development needs to be refined. Tools including single-cell RNA-sequencing, epigenetic analysis, and spatial location of specific immune cells in liver and bone marrow are critical for understanding immune system programming. Considering the vital role immune function plays in offspring health, it will be important to understand how maternal diets can control developmental programming of innate and adaptive immunity.
Collapse
Affiliation(s)
- Benjamin N. Nelson
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Jacob E. Friedman
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
- Department of Physiology and Biochemistry, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Department of Pediatrics, Section of Diabetes and Endocrinology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
17
|
Zhang J, Liu H, Shen Y, Cheng D, Tang H, Zhang Q, Li C, Liu M, Yao W, Ran R, Hou Q, Zhao X, Wang JS, Sun X, Zhang T, Zhou J. Macrophage AHR-TLR4 cross-talk drives p-STAT3 (Ser727)-mediated mitochondrial oxidative stress and upregulates IDO/ICAM-1 in the steatohepatitis induced by aflatoxin B 1. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 923:171377. [PMID: 38458463 DOI: 10.1016/j.scitotenv.2024.171377] [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: 01/13/2024] [Revised: 02/23/2024] [Accepted: 02/27/2024] [Indexed: 03/10/2024]
Abstract
Aflatoxin B1 (AFB1) is a major mycotoxin contaminant showing in the environment and foods. In this study, the molecular initiating events (MIEs) of AFB1-induced steatohepatitis were explored in mice and human cell model. We observed dose-dependent steatohepatitis in the AFB1-treated mice, including triglyceride accumulation, fibrotic collagen secretion, enrichment of CD11b + and F4/80+ macrophages/Kupffer cells, cell death, lymphocytes clusters and remarkable atrophy areas. The gut barrier and gut-microbiota were also severely damaged after the AFB1 treatment and pre-conditioned colitis in the experimental mice aggravated the steatohepatitis phenotypes. We found that macrophages cells can be pro-inflammatorily activated to M1-like phenotype by AFB1 through an AHR/TLR4/p-STAT3 (Ser727)-mediated mitochondrial oxidative stress. The phenotypes can be rescued by AHR inhibitors in the mice model and human cell model. We further showed that this signaling axis is based on the cross-talk interaction between AHR and TLR4. Gene knock-up experiment found that the signaling is dependent on AFB1 ligand-binding with AHR, but not protein expressions of TLR4. The signaling elevated NLRP3 and two immune metabolic enzymes ICAM-1 and IDO that are associated with macrophage polarization. Results from intervention experiments with natural anti-oxidant and AHR inhibitor CH223191 suggest that the macrophage polarization may rely on AHR and ROS. Our study provides novel and critical references to the food safety and public health regulation of AFB1.
Collapse
Affiliation(s)
- Jing Zhang
- Division of Toxicology, Shandong Center for Disease Control and Prevention, Jinan 250014, China
| | - Hui Liu
- Jinan Municipal Center for Disease Control and Prevention, Jinan, Shandong 250021, China
| | - Yang Shen
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Dong Cheng
- Division of Toxicology, Shandong Center for Disease Control and Prevention, Jinan 250014, China.
| | - Hui Tang
- Division of Toxicology, Shandong Center for Disease Control and Prevention, Jinan 250014, China
| | - Qi Zhang
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Chao Li
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong 250062, China.
| | - Ming Liu
- Jinan Municipal Center for Disease Control and Prevention, Jinan, Shandong 250021, China
| | - Wenhuan Yao
- Division of Toxicology, Shandong Center for Disease Control and Prevention, Jinan 250014, China
| | - Rongrong Ran
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Qingzhen Hou
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Shandong 250012, China.
| | - Xiulan Zhao
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| | - Jia-Sheng Wang
- Interdisciplinary Toxicology Program and Department of Environmental Health Science, College of Public Health, University of Georgia, Athens, GA 30602, USA.
| | - Xiulan Sun
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Foods, School of Food Science Synergetic Innovation Center of Food Safety and Nutrition, Jiangnan University, Wuxi, Jiangsu 214122, China.
| | - Tianliang Zhang
- Division of Toxicology, Shandong Center for Disease Control and Prevention, Jinan 250014, China
| | - Jun Zhou
- Division of Toxicology, Shandong Center for Disease Control and Prevention, Jinan 250014, China; Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| |
Collapse
|
18
|
Lecoeur A, Blanc F, Gourichon D, Bruneau N, Burlot T, Pinard-van der Laan MH, Calenge F. Host genetics drives differences in cecal microbiota composition and immune traits of laying hens raised in the same environment. Poult Sci 2024; 103:103609. [PMID: 38547541 PMCID: PMC11000118 DOI: 10.1016/j.psj.2024.103609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 02/12/2024] [Accepted: 02/28/2024] [Indexed: 04/11/2024] Open
Abstract
Vaccination is one of the most effective strategies for preventing infectious diseases but individual vaccine responses are highly heterogeneous. Host genetics and gut microbiota composition are 2 likely drivers of this heterogeneity. We studied 94 animals belonging to 4 lines of laying hens: a White Leghorn experimental line genetically selected for a high antibody response against the Newcastle Disease Virus (NDV) vaccine (ND3) and its unselected control line (CTR), and 2 commercial lines (White Leghorn [LEG] and Rhode Island Red [RIR]). Animals were reared in the same conditions from hatching to 42 d of age, and animals from different genetic lines were mixed. Animals were vaccinated at 22 d of age and their humoral vaccine response against NDV was assessed by hemagglutination inhibition assay and ELISA from blood samples collected at 15, 19, and 21 d after vaccination. The immune parameters studied were the 3 immunoglobulins subtypes A, M, and Y and the blood cell composition was assessed by flow cytometry. The composition of the cecal microbiota was assessed at the end of the experiment by analyzing amplified 16S rRNA gene sequences to obtain amplicon sequence variants (ASV). The 4 lines showed significantly different levels of NDV vaccine response at the 3 measured points, with, logically, a higher response of the genetically selected ND3 line, and intermediate and low responses for the unselected CTR control line and for the 2 commercial lines, respectively. The ND3 line displayed also a higher proportion of immunoglobulins (IgA, IgM, and IgY). The RIR line showed the most different blood cell composition. The 4 lines showed significantly different microbiota characteristics: composition, abundances at all taxonomic levels, and correlations between genera and vaccine response. The tested genetic lines differ for immune parameters and gut microbiota composition and functions. These phenotypic differences can be attributed to genetic differences between lines. Causal relationships between both types of parameters are discussed and will be investigated in further studies.
Collapse
Affiliation(s)
- Alexandre Lecoeur
- Université Paris-Saclay, INRAE, AgroParisTech, Jouy-en-Josas 78350, France.
| | - Fany Blanc
- Université Paris-Saclay, INRAE, AgroParisTech, Jouy-en-Josas 78350, France
| | | | - Nicolas Bruneau
- Université Paris-Saclay, INRAE, AgroParisTech, Jouy-en-Josas 78350, France
| | | | | | - Fanny Calenge
- Université Paris-Saclay, INRAE, AgroParisTech, Jouy-en-Josas 78350, France
| |
Collapse
|
19
|
Liu Y, Ma L, Riqing D, Qu J, Chen J, Zhandu D, Li B, Jiang M. Microbial Metagenomes and Host Transcriptomes Reveal the Dynamic Changes of Rumen Gene Expression, Microbial Colonization and Co-Regulation of Mineral Element Metabolism in Yaks from Birth to Adulthood. Animals (Basel) 2024; 14:1365. [PMID: 38731369 PMCID: PMC11083404 DOI: 10.3390/ani14091365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 04/26/2024] [Accepted: 04/30/2024] [Indexed: 05/13/2024] Open
Abstract
Yaks are the main pillar of plateau animal husbandry and the material basis of local herdsmen's survival. The level of mineral elements in the body is closely related to the production performance of yaks. In this study, we performed a comprehensive analysis of rumen epithelial morphology, transcriptomics and metagenomics to explore the dynamics of rumen functions, microbial colonization and functional interactions in yaks from birth to adulthood. Bacteria, eukaryotes, archaea and viruses colonized the rumen of yaks from birth to adulthood, with bacteria being the majority. Bacteroidetes and Firmicutes were the dominant phyla in five developmental stages, and the abundance of genus Lactobacillus and Fusobacterium significantly decreased with age. Glycoside hydrolase (GH) genes were the most highly represented in five different developmental stages, followed by glycosyltransferases (GTs) and carbohydrate-binding modules (CBMs), where the proportion of genes coding for CBMs increased with age. Integrating host transcriptome and microbial metagenome revealed 30 gene modules related to age, muscle layer thickness, nipple length and width of yaks. Among these, the MEmagenta and MEturquoise were positively correlated with these phenotypic traits. Twenty-two host genes involved in transcriptional regulation related to metal ion binding (including potassium, sodium, calcium, zinc, iron) were positively correlated with a rumen bacterial cluster 1 composed of Alloprevotella, Paludibacter, Arcobacter, Lactobacillus, Bilophila, etc. Therefore, these studies help us to understand the interaction between rumen host and microorganisms in yaks at different ages, and further provide a reliable theoretical basis for the development of feed and mineral element supplementation for yaks at different ages.
Collapse
Affiliation(s)
- Yili Liu
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation, College of Animal & Veterinary Sciences, Southwest Minzu University, Chengdu 610041, China; (Y.L.); (D.R.); (B.L.)
| | - Liangliang Ma
- College of Grassland Resources, Southwest Minzu University, Chengdu 610041, China;
| | - Daojie Riqing
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation, College of Animal & Veterinary Sciences, Southwest Minzu University, Chengdu 610041, China; (Y.L.); (D.R.); (B.L.)
| | - Jiu Qu
- Agriculture and Rural Affairs Bureau of Naqu City, Naqu 852000, China; (J.Q.); (D.Z.)
| | - Jiyong Chen
- Yushu Prefecture Animal Disease Prevention and Control Center, Yushu 815000, China;
| | - Danzeng Zhandu
- Agriculture and Rural Affairs Bureau of Naqu City, Naqu 852000, China; (J.Q.); (D.Z.)
| | - Biao Li
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation, College of Animal & Veterinary Sciences, Southwest Minzu University, Chengdu 610041, China; (Y.L.); (D.R.); (B.L.)
| | - Mingfeng Jiang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation, College of Animal & Veterinary Sciences, Southwest Minzu University, Chengdu 610041, China; (Y.L.); (D.R.); (B.L.)
| |
Collapse
|
20
|
Kolzhetsov N, Markelova N, Frolova M, Alikina O, Glazunova O, Safonova L, Kalashnikova I, Yudin V, Makarov V, Keskinov A, Yudin S, Troshina D, Rechkina V, Shcherbakova V, Shavkunov K, Ozoline O. Enterotype-Dependent Probiotic-Mediated Changes in the Male Rat Intestinal Microbiome In Vivo and In Vitro. Int J Mol Sci 2024; 25:4558. [PMID: 38674145 PMCID: PMC11049970 DOI: 10.3390/ijms25084558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/08/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024] Open
Abstract
Beneficial properties of lactic acid bacteria have been known long ago, but particular interest in probiotics has arisen in the last two decades due to the understanding of the important role of intestinal microflora in human life. Thus, the ability of probiotics to support healthy homeostasis of gut microbiomes has received particular attention. Here, we evaluated the effect of a probiotic consisting of Bifidobacterium longum and Lacticaseibacillus paracasei on the gut microbiome of male rats, assessed their persistence in the fecal biota, and compared probiotic-mediated changes in vitro and in vivo. As expected, microbiomes of two enterotypes were identified in the feces of 21 animals, and it turned out that even a single dose of the probiotic altered the microbial composition. Upon repeated administration, the E1 biota temporarily acquired properties of the E2 type. Being highly sensitive to the intervention of probiotic bacteria at the phylum and genus levels, the fecal microbiomes retained the identity of their enterotypes when transferred to a medium optimized for gut bacteria. For the E2 biota, even similarities between probiotic-mediated reactions in vitro and in vivo were detected. Therefore, fecal-derived microbial communities are proposed as model consortia to optimize the response of resident bacteria to various agents.
Collapse
Affiliation(s)
- Nikolay Kolzhetsov
- Laboratory of Functional Genomics of Prokaryotes, Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, 142290 Pushchino, Russia; (N.K.); (N.M.); (M.F.); (O.A.); (O.G.); (K.S.)
| | - Natalia Markelova
- Laboratory of Functional Genomics of Prokaryotes, Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, 142290 Pushchino, Russia; (N.K.); (N.M.); (M.F.); (O.A.); (O.G.); (K.S.)
| | - Maria Frolova
- Laboratory of Functional Genomics of Prokaryotes, Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, 142290 Pushchino, Russia; (N.K.); (N.M.); (M.F.); (O.A.); (O.G.); (K.S.)
| | - Olga Alikina
- Laboratory of Functional Genomics of Prokaryotes, Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, 142290 Pushchino, Russia; (N.K.); (N.M.); (M.F.); (O.A.); (O.G.); (K.S.)
| | - Olga Glazunova
- Laboratory of Functional Genomics of Prokaryotes, Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, 142290 Pushchino, Russia; (N.K.); (N.M.); (M.F.); (O.A.); (O.G.); (K.S.)
| | - Lubov Safonova
- Federal State Budgetary Institution “Centre for Strategic Planning and Management of Biomedical Health Risks” of the Federal Medical and Biological Agency, 119121 Moscow, Russia; (L.S.); (I.K.); (V.Y.); (V.M.); (A.K.); (S.Y.)
| | - Irina Kalashnikova
- Federal State Budgetary Institution “Centre for Strategic Planning and Management of Biomedical Health Risks” of the Federal Medical and Biological Agency, 119121 Moscow, Russia; (L.S.); (I.K.); (V.Y.); (V.M.); (A.K.); (S.Y.)
| | - Vladimir Yudin
- Federal State Budgetary Institution “Centre for Strategic Planning and Management of Biomedical Health Risks” of the Federal Medical and Biological Agency, 119121 Moscow, Russia; (L.S.); (I.K.); (V.Y.); (V.M.); (A.K.); (S.Y.)
| | - Valentin Makarov
- Federal State Budgetary Institution “Centre for Strategic Planning and Management of Biomedical Health Risks” of the Federal Medical and Biological Agency, 119121 Moscow, Russia; (L.S.); (I.K.); (V.Y.); (V.M.); (A.K.); (S.Y.)
| | - Anton Keskinov
- Federal State Budgetary Institution “Centre for Strategic Planning and Management of Biomedical Health Risks” of the Federal Medical and Biological Agency, 119121 Moscow, Russia; (L.S.); (I.K.); (V.Y.); (V.M.); (A.K.); (S.Y.)
| | - Sergey Yudin
- Federal State Budgetary Institution “Centre for Strategic Planning and Management of Biomedical Health Risks” of the Federal Medical and Biological Agency, 119121 Moscow, Russia; (L.S.); (I.K.); (V.Y.); (V.M.); (A.K.); (S.Y.)
| | - Daria Troshina
- Faculty of Biotechnology, Lomonosov Moscow State University, 119991 Moscow, Russia;
| | - Viktoria Rechkina
- Laboratory of Anaerobic Microorganisms, Institute of Biochemistry and Physiology of Microorganisms of the Russian Academy of Sciences, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, 142290 Pushchino, Russia; (V.R.); (V.S.)
| | - Viktoria Shcherbakova
- Laboratory of Anaerobic Microorganisms, Institute of Biochemistry and Physiology of Microorganisms of the Russian Academy of Sciences, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, 142290 Pushchino, Russia; (V.R.); (V.S.)
| | - Konstantin Shavkunov
- Laboratory of Functional Genomics of Prokaryotes, Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, 142290 Pushchino, Russia; (N.K.); (N.M.); (M.F.); (O.A.); (O.G.); (K.S.)
| | - Olga Ozoline
- Laboratory of Functional Genomics of Prokaryotes, Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, 142290 Pushchino, Russia; (N.K.); (N.M.); (M.F.); (O.A.); (O.G.); (K.S.)
| |
Collapse
|
21
|
Eltayeb LB. Analyzing bacterial persistence and dormancy: A bibliometric exploration of 21st century scientific literature. Saudi J Biol Sci 2024; 31:103936. [PMID: 38327662 PMCID: PMC10847988 DOI: 10.1016/j.sjbs.2024.103936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/08/2024] [Accepted: 01/19/2024] [Indexed: 02/09/2024] Open
Abstract
In response to growing concerns about the efficacy of antibiotic treatment, there has been a significant increase in research on bacteria that are resistant to antibiotics over the past two centuries. Such investigations might bring a spotlight on the field's evolution and future prospects. The study was aimed at conducting a measurable bibliometric review of the scientific literature on bacterial persistence and dormancy in the 21st Century. A scientific literature published during 21st Century was analyzed to gain insights into and identify research trends and outputs in persistent bacteria. Bibliometrix (R language package) and the VOS viewer were used to conduct a bibliometric investigation to determine the globally indexed persistent bacteria research output. WoS Core Collection databases were searched for persistent bacteria selected as the subject. A total of 1,160 published documents from 495 sources from the preceding two decades were reviewed. Maximum publications of 112 were observed in 2021 with 860 citations; however, 82 publications appeared in 2015 and were able to get the highest number of citations (4,214), only 43 (3.7%) were single-authored, whereas 1,117 (96.3%) publications are the result of collaborative works. Out of the top 10 countries ranked for publications, the USA took the top spot for the most highly productive country with 435 articles. Dormancy' appeared 2,351 times, followed by 'Escherichia coli" (1,744, and 'Growth' 1,184 times) in research publications on bacterial persistence research. The findings from this study will aid in the creation of strategies and guidelines for regulating and avoiding bacterial persistence status.
Collapse
Affiliation(s)
- Lienda Bashier Eltayeb
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University – Al-Kharj, 11942 Riyadh, Saudi Arabia
| |
Collapse
|
22
|
Jin DM, Morton JT, Bonneau R. Meta-analysis of the human gut microbiome uncovers shared and distinct microbial signatures between diseases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.27.582333. [PMID: 38464323 PMCID: PMC10925178 DOI: 10.1101/2024.02.27.582333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Microbiome studies have revealed gut microbiota's potential impact on complex diseases. However, many studies often focus on one disease per cohort. We developed a meta-analysis workflow for gut microbiome profiles and analyzed shotgun metagenomic data covering 11 diseases. Using interpretable machine learning and differential abundance analysis, our findings reinforce the generalization of binary classifiers for Crohn's disease (CD) and colorectal cancer (CRC) to hold-out cohorts and highlight the key microbes driving these classifications. We identified high microbial similarity in disease pairs like CD vs ulcerative colitis (UC), CD vs CRC, Parkinson's disease vs type 2 diabetes (T2D), and schizophrenia vs T2D. We also found strong inverse correlations in Alzheimer's disease vs CD and UC. These findings detected by our pipeline provide valuable insights into these diseases.
Collapse
Affiliation(s)
- Dong-Min Jin
- Center for Genomics and Systems Biology, New York University, New York, NY, USA
| | - James T. Morton
- Center for Computational Biology, Flatiron Institute, Simons Foundation, New York, NY, USA
| | - Richard Bonneau
- Center for Genomics and Systems Biology, New York University, New York, NY, USA
- Genentech, New York, NY, USA
| |
Collapse
|
23
|
Öling S, Struck E, Noreen-Thorsen M, Zwahlen M, von Feilitzen K, Odeberg J, Pontén F, Lindskog C, Uhlén M, Dusart P, Butler LM. A human stomach cell type transcriptome atlas. BMC Biol 2024; 22:36. [PMID: 38355543 PMCID: PMC10865703 DOI: 10.1186/s12915-024-01812-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 01/02/2024] [Indexed: 02/16/2024] Open
Abstract
BACKGROUND The identification of cell type-specific genes and their modification under different conditions is central to our understanding of human health and disease. The stomach, a hollow organ in the upper gastrointestinal tract, provides an acidic environment that contributes to microbial defence and facilitates the activity of secreted digestive enzymes to process food and nutrients into chyme. In contrast to other sections of the gastrointestinal tract, detailed descriptions of cell type gene enrichment profiles in the stomach are absent from the major single-cell sequencing-based atlases. RESULTS Here, we use an integrative correlation analysis method to predict human stomach cell type transcriptome signatures using unfractionated stomach RNAseq data from 359 individuals. We profile parietal, chief, gastric mucous, gastric enteroendocrine, mitotic, endothelial, fibroblast, macrophage, neutrophil, T-cell, and plasma cells, identifying over 1600 cell type-enriched genes. CONCLUSIONS We uncover the cell type expression profile of several non-coding genes strongly associated with the progression of gastric cancer and, using a sex-based subset analysis, uncover a panel of male-only chief cell-enriched genes. This study provides a roadmap to further understand human stomach biology.
Collapse
Affiliation(s)
- S Öling
- Department of Clinical Medicine, Translational Vascular Research, The Arctic University of Norway, 9019, Tromsø, Norway
| | - E Struck
- Department of Clinical Medicine, Translational Vascular Research, The Arctic University of Norway, 9019, Tromsø, Norway
| | - M Noreen-Thorsen
- Department of Clinical Medicine, Translational Vascular Research, The Arctic University of Norway, 9019, Tromsø, Norway
| | - M Zwahlen
- Science for Life Laboratory, Department of Protein Science, Royal Institute of Technology (KTH), 171 21, Stockholm, Sweden
| | - K von Feilitzen
- Science for Life Laboratory, Department of Protein Science, Royal Institute of Technology (KTH), 171 21, Stockholm, Sweden
| | - J Odeberg
- Department of Clinical Medicine, Translational Vascular Research, The Arctic University of Norway, 9019, Tromsø, Norway
- Science for Life Laboratory, Department of Protein Science, Royal Institute of Technology (KTH), 171 21, Stockholm, Sweden
- The University Hospital of North Norway (UNN), 9019, Tromsø, Norway
- Department of Haematology, Coagulation Unit, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - F Pontén
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, 752 37, Uppsala, Sweden
| | - C Lindskog
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, 752 37, Uppsala, Sweden
| | - M Uhlén
- Science for Life Laboratory, Department of Protein Science, Royal Institute of Technology (KTH), 171 21, Stockholm, Sweden
| | - P Dusart
- Science for Life Laboratory, Department of Protein Science, Royal Institute of Technology (KTH), 171 21, Stockholm, Sweden
- Clinical Chemistry and Blood Coagulation Research, Department of Molecular Medicine and Surgery, Karolinska Institute, 171 76, Stockholm, Sweden
- Clinical Chemistry, Karolinska University Laboratory, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - L M Butler
- Department of Clinical Medicine, Translational Vascular Research, The Arctic University of Norway, 9019, Tromsø, Norway.
- Science for Life Laboratory, Department of Protein Science, Royal Institute of Technology (KTH), 171 21, Stockholm, Sweden.
- Clinical Chemistry and Blood Coagulation Research, Department of Molecular Medicine and Surgery, Karolinska Institute, 171 76, Stockholm, Sweden.
- Clinical Chemistry, Karolinska University Laboratory, Karolinska University Hospital, 171 76, Stockholm, Sweden.
| |
Collapse
|
24
|
Ahn JS, Koo BC, Choi YJ, Jung WW, Kim HS, Lee SJ, Hong ST, Chung HJ. Identification of Muscle Strength-Related Gut Microbes through Human Fecal Microbiome Transplantation. Int J Mol Sci 2024; 25:662. [PMID: 38203833 PMCID: PMC10779158 DOI: 10.3390/ijms25010662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 12/28/2023] [Accepted: 12/31/2023] [Indexed: 01/12/2024] Open
Abstract
The gut microbiome is well known for its influence on human physiology and aging. Therefore, we speculate that the gut microbiome may affect muscle strength in the same way as the host's own genes. To demonstrate candidates for gut microbes affecting muscle strength, we remodeled the original gut microbiome of mice into human intestinal microbiome through fecal microbiome transplantation (FMT), using human feces and compared the changes in muscle strength in the same mice before and three months after FMT. After comparing before and after FMT, the mice were divided into three groups based on the observed changes in muscle strength: positive, none, and negative changes in muscle strength. As a result of analyzing the α-diversity, β-diversity, and co-occurrence network of the intestinal microbial community before and after FMT, it was observed that a more diverse intestinal microbial community was established after FMT in all groups. In particular, the group with increased muscle strength had more gut microbiome species and communities than the other groups. Fold-change comparison showed that Eisenbergiella massiliensis and Anaeroplasma abactoclasticum from the gut microbiome had positive contributions to muscle strength, while Ileibacterium valens and Ethanoligenens harbinense had negative effects. This study identifies candidates for the gut microbiome that contribute positively and those that contribute negatively to muscle strength.
Collapse
Affiliation(s)
- Ji-Seon Ahn
- Gwangju Center, Korea Basic Science Institute, Gwangju 61751, Jeolla, Republic of Korea; (J.-S.A.); (Y.-J.C.)
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju 54907, Jeonbuk, Republic of Korea
| | - Bon-Chul Koo
- Division of Bioconvergence Analysis, Korea Basic Science Institute, Ochang 28119, Chungbuk, Republic of Korea;
- Department of Biomedical Laboratory Science, Cheongju University, Cheongju 28503, Chungbuk, Republic of Korea; (W.-W.J.); (H.-S.K.); (S.-J.L.)
| | - Yu-Jin Choi
- Gwangju Center, Korea Basic Science Institute, Gwangju 61751, Jeolla, Republic of Korea; (J.-S.A.); (Y.-J.C.)
| | - Woon-Won Jung
- Department of Biomedical Laboratory Science, Cheongju University, Cheongju 28503, Chungbuk, Republic of Korea; (W.-W.J.); (H.-S.K.); (S.-J.L.)
| | - Hyun-Sook Kim
- Department of Biomedical Laboratory Science, Cheongju University, Cheongju 28503, Chungbuk, Republic of Korea; (W.-W.J.); (H.-S.K.); (S.-J.L.)
| | - Suk-Jun Lee
- Department of Biomedical Laboratory Science, Cheongju University, Cheongju 28503, Chungbuk, Republic of Korea; (W.-W.J.); (H.-S.K.); (S.-J.L.)
| | - Seong-Tshool Hong
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju 54907, Jeonbuk, Republic of Korea
| | - Hea-Jong Chung
- Gwangju Center, Korea Basic Science Institute, Gwangju 61751, Jeolla, Republic of Korea; (J.-S.A.); (Y.-J.C.)
| |
Collapse
|
25
|
König E, Heponiemi P, Kivinen S, Räkköläinen J, Beasley S, Borman T, Collado MC, Hukkinen V, Junnila J, Lahti L, Norring M, Piirainen V, Salminen S, Heinonen M, Valros A. Fewer culturable Lactobacillaceae species identified in faecal samples of pigs performing manipulative behaviour. Sci Rep 2024; 14:132. [PMID: 38168466 PMCID: PMC10762183 DOI: 10.1038/s41598-023-50791-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 12/25/2023] [Indexed: 01/05/2024] Open
Abstract
Manipulative behaviour that consists of touching or close contact with ears or tails of pen mates is common in pigs and can become damaging. Manipulative behaviour was analysed from video recordings of 45-day-old pigs, and 15 manipulator-control pairs (n = 30) were formed. Controls neither received nor performed manipulative behaviour. Rectal faecal samples of manipulators and controls were compared. 16S PCR was used to identify Lactobacillaceae species and 16S amplicon sequencing to determine faecal microbiota composition. Seven culturable Lactobacillaceae species were identified in control pigs and four in manipulator pigs. Manipulators (p = 0.02) and females (p = 0.005) expressed higher Lactobacillus amylovorus, and a significant interaction was seen (sex * status: p = 0.005) with this sex difference being more marked in controls. Females (p = 0.08) and manipulator pigs (p = 0.07) tended to express higher total Lactobacillaceae. A tendency for an interaction was seen in Limosilactobacillus reuteri (sex * status: p = 0.09). Results suggest a link between observed low diversity in Lactobacillaceae and the development of manipulative behaviour.
Collapse
Affiliation(s)
- Emilia König
- Research Centre for Animal Welfare, Department of Production Animal Medicine, University of Helsinki, 00790, Helsinki, Finland.
| | | | - Sanni Kivinen
- Functional Foods Forum, University of Turku, 20520, Turku, Finland
| | | | - Shea Beasley
- Vetcare Ltd., 04600, Mäntsälä, Finland
- Sheaps Oy, 03250, Ojakkala, Finland
| | - Tuomas Borman
- Department of Computing, University of Turku, 20500, Turku, Finland
| | - Maria Carmen Collado
- Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), 46980, Paterna, Valencia, Spain
| | - Vilja Hukkinen
- Research Centre for Animal Welfare, Department of Production Animal Medicine, University of Helsinki, 00790, Helsinki, Finland
| | | | - Leo Lahti
- Department of Computing, University of Turku, 20500, Turku, Finland
| | - Marianna Norring
- Research Centre for Animal Welfare, Department of Production Animal Medicine, University of Helsinki, 00790, Helsinki, Finland
| | - Virpi Piirainen
- Research Centre for Animal Welfare, Department of Production Animal Medicine, University of Helsinki, 00790, Helsinki, Finland
| | - Seppo Salminen
- Functional Foods Forum, University of Turku, 20520, Turku, Finland
| | - Mari Heinonen
- Research Centre for Animal Welfare, Department of Production Animal Medicine, University of Helsinki, 00790, Helsinki, Finland
| | - Anna Valros
- Research Centre for Animal Welfare, Department of Production Animal Medicine, University of Helsinki, 00790, Helsinki, Finland
| |
Collapse
|
26
|
Beaven E, Kumar R, An JM, Mendoza H, Sutradhar SC, Choi W, Narayan M, Lee YK, Nurunnabi M. Potentials of ionic liquids to overcome physical and biological barriers. Adv Drug Deliv Rev 2024; 204:115157. [PMID: 38104896 PMCID: PMC10787599 DOI: 10.1016/j.addr.2023.115157] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 12/04/2023] [Accepted: 12/10/2023] [Indexed: 12/19/2023]
Abstract
Over the last decades, ionic liquids (IL) have shown great potential in non-invasive delivery starting from synthetic small molecules to biological large molecules. ILs are emerging as a particular class of drug delivery systems due to their unique physiochemical properties, simple surface modification, and functionalization. These features of IL help achieve specific design principles that are essential for a non-invasive drug delivery system. In this review, we have discussed IL and their applications in non-invasive drug delivery systems. We evaluated state-of-the-art development and advances of IL aiming to mitigate the biological and physical barriers to improve transdermal and oral delivery, summarized in this review. We also provided an overview of the various factors determining the systemic transportation of IL-based formulation. Additionally, we have emphasized how the ILs facilitate the transportation of therapeutic molecules by overcoming biological barriers.
Collapse
Affiliation(s)
- Elfa Beaven
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, TX 79902, United States; Biomedical Engineering Program, College of Engineering, University of Texas at El Paso, El Paso, TX 79968, United States
| | - Raj Kumar
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, TX 79902, United States; Biomedical Engineering Program, College of Engineering, University of Texas at El Paso, El Paso, TX 79968, United States
| | - Jeong Man An
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 04763, Republic of Korea
| | - Hannia Mendoza
- Department of Chemistry and Biochemistry, College of Science, University of Texas at El Paso, El Paso, TX 79968, United States
| | - Sabuj Chandra Sutradhar
- 4D Convergence Technology Institute, Korea National University of Transportation, Jungpyeong 27909, Republic of Korea
| | - Wonho Choi
- 4D Convergence Technology Institute, Korea National University of Transportation, Jungpyeong 27909, Republic of Korea
| | - Mahesh Narayan
- Department of Chemistry and Biochemistry, College of Science, University of Texas at El Paso, El Paso, TX 79968, United States
| | - Yong-Kyu Lee
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 04763, Republic of Korea; Department of Chemical and Biological Engineering, College of Engineering, Korea National University of Transportation, Chungju 380-702, Republic of Korea; 4D Convergence Technology Institute, Korea National University of Transportation, Jungpyeong 27909, Republic of Korea.
| | - Md Nurunnabi
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, TX 79902, United States; Biomedical Engineering Program, College of Engineering, University of Texas at El Paso, El Paso, TX 79968, United States; Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968, United States.
| |
Collapse
|
27
|
Chetty A, Blekhman R. Multi-omic approaches for host-microbiome data integration. Gut Microbes 2024; 16:2297860. [PMID: 38166610 PMCID: PMC10766395 DOI: 10.1080/19490976.2023.2297860] [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: 07/19/2023] [Accepted: 12/18/2023] [Indexed: 01/05/2024] Open
Abstract
The gut microbiome interacts with the host through complex networks that affect physiology and health outcomes. It is becoming clear that these interactions can be measured across many different omics layers, including the genome, transcriptome, epigenome, metabolome, and proteome, among others. Multi-omic studies of the microbiome can provide insight into the mechanisms underlying host-microbe interactions. As more omics layers are considered, increasingly sophisticated statistical methods are required to integrate them. In this review, we provide an overview of approaches currently used to characterize multi-omic interactions between host and microbiome data. While a large number of studies have generated a deeper understanding of host-microbiome interactions, there is still a need for standardization across approaches. Furthermore, microbiome studies would also benefit from the collection and curation of large, publicly available multi-omics datasets.
Collapse
Affiliation(s)
- Ashwin Chetty
- Committee on Genetics, Genomics and Systems Biology, The University of Chicago, Chicago, IL, USA
| | - Ran Blekhman
- Section of Genetic Medicine, Department of Medicine, The University of Chicago, Chicago, IL, USA
| |
Collapse
|
28
|
Onarman Umu ÖC, Mydland LT, Chen C, de Nanclares MP, Shurson GC, Urriola PE, Sørum H, Øverland M. Integrated multi-omics approach reveals novel associations in the rapeseed diet-microbiota-host axis in pigs. ISME COMMUNICATIONS 2024; 4:ycae061. [PMID: 38800131 PMCID: PMC11128262 DOI: 10.1093/ismeco/ycae061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 04/18/2024] [Accepted: 04/18/2024] [Indexed: 05/29/2024]
Abstract
Diet-mediated host-microbiota interplay is a key factor in optimizing the gut function and overall health of the host. Gaining insight into the biological mechanisms behind this relationship is fundamental to finding sustainable, environment-friendly feed solutions in livestock production systems. Here, we apply a multi-omics integration approach to elucidate sustainable diet-associated host-gut microbiota interactions in pigs and we demonstrate novel and biologically relevant host-microbe associations in the gut, driven by a rapeseed meal-based feed (RSF). Interestingly, RSF-diet promoted the abundance of segmented filamentous bacteria Candidatus Arthromitus that was associated with the maintenance of mucosal immunity in the ileum of pigs. In the colon, RSF diet affected host mRNA splicing functions, which may result in different host gene products, through host-microbiota associations, particularly with the Faecalibacterium population, and through the interaction of dietary components such as sinapic acid with the host cells. Moreover, telomere maintenance and organization functions that may determine the overall health of the host were upregulated and notably associated with Subdoligranulum population in the colon of RSF diet-fed pigs. This integrative multi-omics approach provides more insight into the diet-microbiota-host axis, and a better understanding of mechanisms and opportunities to find new strategies for modulating host health and potentially improving caloric and nutritional efficiency in animal production.
Collapse
Affiliation(s)
- Özgün C Onarman Umu
- Department of Paraclinical Sciences, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås N-1432, Norway
| | - Liv Torunn Mydland
- Department of Animal and Aquacultural Sciences, Faculty of Biosciences, Norwegian University of Life Sciences, Ås N-1432, Norway
| | - Chi Chen
- Department of Food Science and Nutrition, University of Minnesota, St Paul, MN 55108, United States
| | - Marta Pérez de Nanclares
- Department of Animal and Aquacultural Sciences, Faculty of Biosciences, Norwegian University of Life Sciences, Ås N-1432, Norway
| | - Gerald C Shurson
- Department of Animal Science, University of Minnesota, St Paul, MN 55108, United States
| | - Pedro E Urriola
- Department of Animal Science, University of Minnesota, St Paul, MN 55108, United States
| | - Henning Sørum
- Department of Paraclinical Sciences, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås N-1432, Norway
| | - Margareth Øverland
- Department of Animal and Aquacultural Sciences, Faculty of Biosciences, Norwegian University of Life Sciences, Ås N-1432, Norway
| |
Collapse
|
29
|
Joo MK, Lee JW, Woo JH, Kim HJ, Kim DH, Choi JH. Regulation of colonic neuropeptide Y expression by the gut microbiome in patients with ulcerative colitis and its association with anxiety- and depression-like behavior in mice. Gut Microbes 2024; 16:2319844. [PMID: 38404132 PMCID: PMC10900276 DOI: 10.1080/19490976.2024.2319844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 02/13/2024] [Indexed: 02/27/2024] Open
Abstract
Patients with inflammatory bowel disease (IBD), including ulcerative colitis (UC), show an increased incidence of anxiety and depression; however, the association between UC-associated psychiatric disorders and the gut microbiota is unclear. This study aimed to examine whether gut microbiota from patients with UC can alter colonic gene expression, leading to anxiety- and depression-like behavior in mice receiving fecal microbiota transplantation (FMT). RNA sequencing transcriptome analyses revealed a difference in colonic gene expression between mice receiving FMT from patients with UC (UC-FMT mice) and those receiving FMT from healthy controls (HC-FMT mice). Gene ontology analysis revealed the downregulation of neuropeptide signaling pathways, including neuropeptide Y (NPY) expression, in the colons of UC-FMT mice. The protein levels of NPY also decreased in the colon and plasma of UC-FMT mice compared to those in HC-FMT mice. The oral administration of Enterococcus mundtii (EM), a bacterium isolated from the feces of patients with UC, reduced NPY expression in the colons of mice and induced intestinal inflammation, anxiety, and depression-like behavior. Reduced NPY protein levels were also observed in the plasma and hippocampus of EM-treated mice. Intraperitoneal administration of NPY significantly alleviated anxiety- and depressive-like behaviors induced by EM in mice. Capsular polysaccharide in EM was associated with EM-induced NPY downregulation in the colon. Analysis of Gene Expression Omnibus datasets showed markedly reduced NPY expression in the inflamed colons of patients with UC compared with that in the colons of healthy controls. In summary, EM-induced reduction in the colonic expression of NPY may be associated with a decrease in hippocampal NPY and anxiety- and depression-like behavior in mice.
Collapse
Affiliation(s)
- Min-Kyung Joo
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul, Korea
- College of Pharmacy, Kyung Hee University, Seoul, Korea
| | - Jae-Won Lee
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul, Korea
- College of Pharmacy, Kyung Hee University, Seoul, Korea
| | - Jeong-Hwa Woo
- College of Pharmacy, Kyung Hee University, Seoul, Korea
| | - Hyo-Jong Kim
- Department of Internal Medicine, School of Medicine, Kyung Hee University, Seoul, Korea
| | - Dong-Hyun Kim
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul, Korea
- College of Pharmacy, Kyung Hee University, Seoul, Korea
| | - Jung-Hye Choi
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul, Korea
- College of Pharmacy, Kyung Hee University, Seoul, Korea
| |
Collapse
|
30
|
Feng R, Yang W, Feng W, Huang X, Cen M, Peng G, Wu W, Wang Z, Jing Y, Long T, Liu Y, Li Z, Chang G, Huang K. Time-restricted feeding ameliorates non-alcoholic fatty liver disease through modulating hepatic nicotinamide metabolism via gut microbiota remodeling. Gut Microbes 2024; 16:2390164. [PMID: 39154362 PMCID: PMC11332628 DOI: 10.1080/19490976.2024.2390164] [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: 01/22/2024] [Revised: 07/31/2024] [Accepted: 08/05/2024] [Indexed: 08/20/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) has emerged as a global health concern, lacking specific therapeutic strategies. Time-restricted feeding (TRF) regimen demonstrated beneficial effects in NAFLD; however, the underlying mechanisms remain unclear. In this study, we established a NAFLD mouse model through a high-fat diet (HFD) and implemented the 16:8 TRF regimen for a duration of 6 weeks. We demonstrated that TRF remarkably alleviated hepatic steatosis in HFD mice. Of note, aldehyde oxidase 1 (AOX1), a key enzyme in hepatic nicotinamide (NAM) catabolism, exhibited apparent upregulation in response to HFD, leading to abnormal accumulation of N-Methyl-6-pyridone-3-carboxamide (N-Me-6-PY, also known as 2PY) and N-Methyl-4-pyridone-5-carboxamide (N-Me-4-PY, also known as 4PY), whereas it was almost restored by TRF. Both N-Me-6-PY and N-Me-4-PY promoted de novo lipogenesis and fatty acid uptake capacities in hepatocyte, and aggravated hepatic steatosis in mice either fed chow diet or HFD. In contrast, pharmacological inhibition of AOX1 was sufficient to ameliorate the hepatic steatosis and lipid metabolic dysregulation induced by HFD. Moreover, transplantation of fecal microbiota efficiently mimicked the modulatory effect of TRF on NAM metabolism, thus mitigating hepatic steatosis and lipid metabolic disturbance, suggesting a gut microbiota-dependent manner. In conclusion, our study reveals the intricate relationship between host NAM metabolic modification and gut microbiota remodeling during the amelioration of NAFLD by TRF, providing promising insights into the prevention and treatment of NAFLD.
Collapse
Affiliation(s)
- Ruijia Feng
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wenchao Yang
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Weiqi Feng
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiuyi Huang
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Meifeng Cen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Guiyan Peng
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wenrui Wu
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhecun Wang
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yexiang Jing
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ting Long
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yunchong Liu
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zilun Li
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Guangqi Chang
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Kan Huang
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
31
|
Shah A, Lee YY, Suzuki H, Tan-Loh J, Siah KTH, Gwee KA, Fairlie T, Talley NJ, Ghoshal UC, Wang YP, Kim YS, Holtmann G. A pathophysiologic framework for the overlap of disorders of gut-brain interaction and the role of the gut microbiome. Gut Microbes 2024; 16:2413367. [PMID: 39482844 PMCID: PMC11540069 DOI: 10.1080/19490976.2024.2413367] [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: 03/07/2024] [Revised: 07/24/2024] [Accepted: 10/02/2024] [Indexed: 11/03/2024] Open
Abstract
The International Rome Committee defines Disorders of Gut-Brain Interactions (DGBI) based upon distinct combinations of chronic and/or recurrent unexplained gastrointestinal symptoms. Yet patients often experience overlapping DGBI. Patients with DGBI frequently also suffer from extraintestinal symptoms, including fatigue, sleep disturbances, anxiety, and depression. Patients with overlapping DGBI typically experience more severe GI symptoms and increased psychosocial burden. Concerning the pathophysiology, DGBI are associated with disruptions in gut motility, function of the brain and enteric neurons, immune function, and genetic markers, with recent findings revealing gut microbiome alterations linked to these mechanisms of DGBI. Emerging evidence summarized in this review suggests that the microbiome influences various established disease mechanisms of different DGBI groups. Overall, changes in the gastrointestinal microbiome do not seem to be linked to a specific DGBI subgroup but may play a key role in the manifestation of different DGBI and, subsequently, overlap of DGBI. Understanding these shared mechanisms and the role of the gastrointestinal microbiome, particularly for overlapping DGBI, might aid in developing more precise diagnostic criteria and treatment strategies while developing personalized interventions that target specific mechanisms to improve patient outcomes.
Collapse
Affiliation(s)
- Ayesha Shah
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
- Department of Gastroenterology and Hepatology, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Australia
| | - Yeong Yeh Lee
- School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, Malaysia
| | - Hidekazu Suzuki
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Tokai University School of Medicine, Isehara, Japan
| | - Joash Tan-Loh
- Division of Gastroenterology Hepatology, Department of Internal Medicine, Hospital Kuala Lumpur, Kuala Lumpur, Malaysia
| | - Kewin Tien Ho Siah
- Division of Gastroenterology and Hepatology, University Medicine Cluster, National University Hospital, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore City, Singapore
| | - Kok-Ann Gwee
- Division of Gastroenterology and Hepatology, University Medicine Cluster, National University Hospital, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore City, Singapore
| | - Thomas Fairlie
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
- Department of Gastroenterology and Hepatology, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Australia
| | - Nicholas J. Talley
- School of Medicine and Public Health, and Hunter Medical Research Institute, the University of Newcastle, Newcastle, Australia
| | - Uday C Ghoshal
- Institute of Gastrosciences & Liver Transplantation, Apollo Multispeciality Hospitals, Kolkata, India
| | - Yen-Po Wang
- Endoscopy centre for Diagnosis of Treatment, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yong Sung Kim
- Digestive Disease Research Institute, Wonkwang University College of Medicine, Iksan, Korea
- Good Breath Clinic, Gunpo, Korea
| | - Gerald Holtmann
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
- Department of Gastroenterology and Hepatology, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Australia
| |
Collapse
|
32
|
Moon JH, Roh DH, Kwack KH, Lee JH. Bacterial single-cell transcriptomics: Recent technical advances and future applications in dentistry. JAPANESE DENTAL SCIENCE REVIEW 2023; 59:253-262. [PMID: 37674900 PMCID: PMC10477369 DOI: 10.1016/j.jdsr.2023.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 06/17/2023] [Accepted: 08/09/2023] [Indexed: 09/08/2023] Open
Abstract
Metagenomics and metatranscriptomics have enhanced our understanding of the oral microbiome and its impact on oral health. However, these approaches have inherent limitations in exploring individual cells and the heterogeneity within mixed microbial communities, which restricts our current understanding to bulk cells and species-level information. Fortunately, recent technical advances have enabled the application of single-cell RNA sequencing (scRNA-seq) for studying bacteria, shedding light on cell-to-cell diversity and interactions between host-bacterial cells at the single-cell level. Here, we address the technical barriers in capturing RNA from single bacterial cells and highlight pioneering studies from the past decade. We also discuss recent achievements in host-bacterial dual transcriptional profiling at the single-cell level. Bacterial scRNA-seq provides advantages in various research fields, including the investigation of phenotypic heterogeneity within genetically identical bacteria, identification of rare cell types, detection of antibiotic-resistant or persistent cells, analysis of individual gene expression patterns and metabolic activities, and characterization of specific microbe-host interactions. Integrating single-cell techniques with bulk approaches is essential to gain a comprehensive understanding of oral diseases and develop targeted and personalized treatment in dentistry. The reviewed pioneering studies are expected to inspire future research on the oral microbiome at the single-cell level.
Collapse
Affiliation(s)
- Ji-Hoi Moon
- Department of Oral Microbiology, College of Dentistry, Kyung Hee University, Seoul, Republic of Korea
| | - Dae-Hyun Roh
- Department of Oral Physiology, College of Dentistry, Kyung Hee University, Seoul, Republic of Korea
| | - Kyu Hwan Kwack
- Department of Oral Microbiology, College of Dentistry, Kyung Hee University, Seoul, Republic of Korea
| | - Jae-Hyung Lee
- Department of Oral Microbiology, College of Dentistry, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
33
|
Otálora-Otálora BA, López-Rivera JJ, Aristizábal-Guzmán C, Isaza-Ruget MA, Álvarez-Moreno CA. Host Transcriptional Regulatory Genes and Microbiome Networks Crosstalk through Immune Receptors Establishing Normal and Tumor Multiomics Metafirm of the Oral-Gut-Lung Axis. Int J Mol Sci 2023; 24:16638. [PMID: 38068961 PMCID: PMC10706695 DOI: 10.3390/ijms242316638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/13/2023] [Accepted: 11/18/2023] [Indexed: 12/18/2023] Open
Abstract
The microbiome has shown a correlation with the diet and lifestyle of each population in health and disease, the ability to communicate at the cellular level with the host through innate and adaptative immune receptors, and therefore an important role in modulating inflammatory process related to the establishment and progression of cancer. The oral cavity is one of the most important interaction windows between the human body and the environment, allowing the entry of an important number of microorganisms and their passage across the gastrointestinal tract and lungs. In this review, the contribution of the microbiome network to the establishment of systemic diseases like cancer is analyzed through their synergistic interactions and bidirectional crosstalk in the oral-gut-lung axis as well as its communication with the host cells. Moreover, the impact of the characteristic microbiota of each population in the formation of the multiomics molecular metafirm of the oral-gut-lung axis is also analyzed through state-of-the-art sequencing techniques, which allow a global study of the molecular processes involved of the flow of the microbiota environmental signals through cancer-related cells and its relationship with the establishment of the transcription factor network responsible for the control of regulatory processes involved with tumorigenesis.
Collapse
Affiliation(s)
| | - Juan Javier López-Rivera
- Grupo de Investigación INPAC, Specialized Laboratory, Clinica Universitaria Colombia, Clínica Colsanitas S.A., Bogotá 111321, Colombia;
| | - Claudia Aristizábal-Guzmán
- Grupo de Investigación INPAC, Unidad de Investigación, Fundación Universitaria Sanitas, Bogotá 110131, Colombia;
| | - Mario Arturo Isaza-Ruget
- Keralty, Sanitas International Organization, Grupo de Investigación INPAC, Fundación Universitaria Sanitas, Bogotá 110131, Colombia;
| | - Carlos Arturo Álvarez-Moreno
- Infectious Diseases Department, Clinica Universitaria Colombia, Clínica Colsanitas S.A., Bogotá 111321, Colombia;
| |
Collapse
|
34
|
Miteva D, Peshevska-Sekulovska M, Snegarova V, Peruhova M, Vasilev GH, Vasilev GV, Sekulovski M, Lazova S, Gulinac M, Tomov L, Mihova A, Velikova T. Microbiome and Genetic Factors in the Pathogenesis of Liver Diseases. GASTROENTEROLOGY INSIGHTS 2023; 14:575-597. [DOI: 10.3390/gastroent14040041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2024] Open
Abstract
Our genetic background has not changed over the past century, but chronic diseases are on the rise globally. In addition to the genetic component, among the critical factors for many diseases are inhabitants of our intestines (gut microbiota) as a crucial environmental factor. Dysbiosis has been described in liver diseases with different etiologies like non-alcoholic fatty liver disease (NAFLD), alcohol-related liver disease (ALD), viral hepatitis, autoimmune hepatitis (AIH), primary sclerosing cholangitis (PSC), primary biliary cholangitis (PBC), cirrhosis, hepatocellular carcinoma (HCC). On the other hand, new technologies have increased our understanding of liver disease genetics and treatment options. Genome-wide association studies (GWAS) identify unknown genetic risk factors, positional cloning of unknown genes associated with different diseases, gene tests for single nucleotide variations (SNVs), and next-generation sequencing (NGS) of selected genes or the complete genome. NGS also allowed studying the microbiome and its role in various liver diseases has begun. These genes have proven their effect on microbiome composition in host genome–microbiome association studies. We focus on altering the intestinal microbiota, and supplementing some bacterial metabolites could be considered a potential therapeutic strategy. The literature data promote probiotics/synbiotics role in reducing proinflammatory cytokines such as TNF-α and the interleukins (IL-1, IL-6, IL-8), therefore improving transaminase levels, hepatic steatosis, and NAFLD activity score. However, even though microbial therapy appears to be risk-free, evaluating side effects related to probiotics or synbiotics is imperative. In addition, safety profiles for long-term usage should be researched. Thus, this review focuses on the human microbiome and liver diseases, recent GWASs on liver disease, the gut-liver axis, and the associations with the microbiome and microbiome during/after liver disease therapy.
Collapse
Affiliation(s)
- Dimitrina Miteva
- Department of Genetics, Faculty of Biology, Sofia University St. Kliment Ohridski, 8 Dragan Tzankov Str., 1164 Sofia, Bulgaria
- Medical Faculty, Sofia University St. Kliment Ohridski, 1 Kozyak Str., 1407 Sofia, Bulgaria
| | - Monika Peshevska-Sekulovska
- Medical Faculty, Sofia University St. Kliment Ohridski, 1 Kozyak Str., 1407 Sofia, Bulgaria
- Department of Gastroenterology, University Hospital Lozenetz, Kozyak 1 Str., 1407 Sofia, Bulgaria
| | - Violeta Snegarova
- Clinic of Internal Diseases, Naval Hospital—Varna, Military Medical Academy, Medical Faculty, Medical University, Blvd. Hristo Smirnenski 3, 9000 Varna, Bulgaria
| | - Milena Peruhova
- Department of Gastroenterology, Heart and Brain Hospital, Zdrave 1 Str., 8000 Burgas, Bulgaria
| | - Georgi H. Vasilev
- Medical Faculty, Sofia University St. Kliment Ohridski, 1 Kozyak Str., 1407 Sofia, Bulgaria
- Laboratory of Hematopathology and Immunology, National Specialized Hospital for Active Treatment of Hematological Diseases, “Plovdivsko Pole” Str. 6, 1756 Sofia, Bulgaria
| | - Georgi V. Vasilev
- Medical Faculty, Sofia University St. Kliment Ohridski, 1 Kozyak Str., 1407 Sofia, Bulgaria
- Department of Emergency Medicine and Clinic of Neurology, University Hospital “Sv. Georgi”, Blvd. Peshtersko Shose 66, 4000 Plovdiv, Bulgaria
| | - Metodija Sekulovski
- Medical Faculty, Sofia University St. Kliment Ohridski, 1 Kozyak Str., 1407 Sofia, Bulgaria
- Department of Anesthesiology and Intensive Care, University Hospital Lozenetz, 1 Kozyak Str., 1407 Sofia, Bulgaria
| | - Snezhina Lazova
- Medical Faculty, Sofia University St. Kliment Ohridski, 1 Kozyak Str., 1407 Sofia, Bulgaria
- Pediatric Department, University Hospital “N. I. Pirogov”, 21 “General Eduard I. Totleben” Blvd, 1606 Sofia, Bulgaria
- Department of Healthcare, Faculty of Public Health, “Prof. Tsekomir Vodenicharov, MD, DSc”, Medical University of Sofia, Bialo More 8 Str., 1527 Sofia, Bulgaria
| | - Milena Gulinac
- Medical Faculty, Sofia University St. Kliment Ohridski, 1 Kozyak Str., 1407 Sofia, Bulgaria
- Department of General and Clinical Pathology, Medical University of Plovdiv, Bul. Vasil Aprilov 15A, 4000 Plovdiv, Bulgaria
| | - Latchezar Tomov
- Medical Faculty, Sofia University St. Kliment Ohridski, 1 Kozyak Str., 1407 Sofia, Bulgaria
- Department of Informatics, New Bulgarian University, Montevideo 21 Str., 1618 Sofia, Bulgaria
| | - Antoaneta Mihova
- SMDL Ramus, Department of Immunology, Blvd. Kap. Spisarevski 26, 1527 Sofia, Bulgaria
| | - Tsvetelina Velikova
- Medical Faculty, Sofia University St. Kliment Ohridski, 1 Kozyak Str., 1407 Sofia, Bulgaria
| |
Collapse
|
35
|
Zhao J, Duan G, Zhu Y, Zhu D. Gut microbiota and transcriptome response of earthworms (Metaphire guillelmi) to polymyxin B exposure. J Environ Sci (China) 2023; 133:37-47. [PMID: 37451787 DOI: 10.1016/j.jes.2022.07.033] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 07/22/2022] [Accepted: 07/22/2022] [Indexed: 07/18/2023]
Abstract
Polymyxin B (PMB) has received widespread attention for its use as a last-line therapy against multidrug-resistant bacterial infection. However, the consequences of unintended PMB exposure on organisms in the surrounding environment remain inconclusive. Therefore, this study investigated the effects of soil PMB residue on the gut microbiota and transcriptome of earthworms (Metaphire guillelmi). The results indicated that the tested doses of PMB (0.01-100 mg/kg soil) did not significantly affect the richness and Shannon's diversity index of the earthworm gut microbiota, but PMB altered its community structure and taxonomic composition. Moreover, PMB significantly affected Lysobacter, Aeromonas, and Sphingomonas in the soil microbiota, whereas Pseudomonas was significantly impacted the earthworm gut microbiota. Furthermore, active bacteria responded more significantly to PMB than the total microbial community. Bacterial genera such as Acinetobacter and Bacillus were highly correlated with differential expression of some genes, including up-regulated genes associated with folate biosynthesis, sulphur metabolism, and the IL-17 signalling pathway, and downregulated genes involved in vitamin digestion and absorption, salivary secretion, other types of O-glycan biosynthesis, and the NOD-like receptor signalling pathway. These results suggest that adaptation to PMB stress by earthworms involves changes in energy metabolism, their immune and digestive systems, as well as glycan biosynthesis. The study findings help elucidate the relationship between earthworms and their microbiota, while providing a reference for understanding the environmental risks of PMB.
Collapse
Affiliation(s)
- Jun Zhao
- State Key Lab of Urban and Regional Ecology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guilan Duan
- State Key Lab of Urban and Regional Ecology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yongguan Zhu
- State Key Lab of Urban and Regional Ecology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China; Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China
| | - Dong Zhu
- State Key Lab of Urban and Regional Ecology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China; Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China.
| |
Collapse
|
36
|
Tizabi Y, Bennani S, El Kouhen N, Getachew B, Aschner M. Interaction of Heavy Metal Lead with Gut Microbiota: Implications for Autism Spectrum Disorder. Biomolecules 2023; 13:1549. [PMID: 37892231 PMCID: PMC10605213 DOI: 10.3390/biom13101549] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Autism Spectrum Disorder (ASD), a neurodevelopmental disorder characterized by persistent deficits in social interaction and communication, manifests in early childhood and is followed by restricted and stereotyped behaviors, interests, or activities in adolescence and adulthood (DSM-V). Although genetics and environmental factors have been implicated, the exact causes of ASD have yet to be fully characterized. New evidence suggests that dysbiosis or perturbation in gut microbiota (GM) and exposure to lead (Pb) may play important roles in ASD etiology. Pb is a toxic heavy metal that has been linked to a wide range of negative health outcomes, including anemia, encephalopathy, gastroenteric diseases, and, more importantly, cognitive and behavioral problems inherent to ASD. Pb exposure can disrupt GM, which is essential for maintaining overall health. GM, consisting of trillions of microorganisms, has been shown to play a crucial role in the development of various physiological and psychological functions. GM interacts with the brain in a bidirectional manner referred to as the "Gut-Brain Axis (GBA)". In this review, following a general overview of ASD and GM, the interaction of Pb with GM in the context of ASD is emphasized. The potential exploitation of this interaction for therapeutic purposes is also touched upon.
Collapse
Affiliation(s)
- Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington, DC 20059, USA
| | - Samia Bennani
- Faculty of Medicine and Pharmacy of Casablanca, Hassan II University, Casablanca 20100, Morocco
| | - Nacer El Kouhen
- Faculty of Medicine and Pharmacy of Casablanca, Hassan II University, Casablanca 20100, Morocco
| | - Bruk Getachew
- Department of Pharmacology, Howard University College of Medicine, Washington, DC 20059, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA;
| |
Collapse
|
37
|
Kellnerová S, Huber S, Massri M, Fleischer V, Losso K, Sarg B, Kremser L, Talasz H, He X, Varrone E, Brigotti M, Ardissino G, Orth-Höller D, Würzner R. Enzymatic Cleavage of Stx2a in the Gut and Identification of Pancreatic Elastase and Trypsin as Possible Main Cleavers. Microorganisms 2023; 11:2487. [PMID: 37894145 PMCID: PMC10609011 DOI: 10.3390/microorganisms11102487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 09/15/2023] [Accepted: 10/02/2023] [Indexed: 10/29/2023] Open
Abstract
Shiga toxins (Stxs), especially the Stx2a subtype, are the major virulence factors involved in enterohemorrhagic Escherichia coli (EHEC)-associated hemolytic uremic syndrome (eHUS), a life-threatening disease causing acute kidney injury, especially in children. After oral transmission and colonization in the gut, EHEC release Stx. Intracellular cleavage of the Stx A subunit, when followed by reduction, boosts the enzymatic activity that causes damage to targeted cells. This cleavage was assumed to be mostly mediated by furin during Stx intracellular trafficking. To investigate whether this cleavage could occur in the intestine, even prior to entering target cells, Stx2a A subunit structure (intact or cleaved) was characterized after its exposure to specific host factors present in human stool. The molecular weight of Stx2a A subunit/fragments was determined by immunoblotting after electrophoretic separation under reducing conditions. In this study, it was demonstrated that Stx2a is cleaved by certain human stool components. Trypsin and chymotrypsin-like elastase 3B (CELA3B), two serine proteases, were identified as potential candidates that can trigger the extracellular cleavage of Stx2a A subunit directly after its secretion by EHEC in the gut. Whether the observed cleavage indeed translates to natural infections and plays a role in eHUS pathogenesis has yet to be determined. If so, it seems likely that a host's protease profile could affect disease development by changing the toxin's biological features.
Collapse
Affiliation(s)
- Sára Kellnerová
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (S.K.); (S.H.); (M.M.); (V.F.)
| | - Silke Huber
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (S.K.); (S.H.); (M.M.); (V.F.)
| | - Mariam Massri
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (S.K.); (S.H.); (M.M.); (V.F.)
| | - Verena Fleischer
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (S.K.); (S.H.); (M.M.); (V.F.)
| | - Klemens Losso
- Institute of Analytical Chemistry and Radiochemistry, University of Innsbruck, 6020 Innsbruck, Austria;
- Department of Food Technology and Nutrition, MCI|The Entrepreneurial School, 6020 Innsbruck, Austria
| | - Bettina Sarg
- Protein Core Facility, Institute of Medical Biochemistry, Center of Chemistry and Biomedicine, Medical University of Innsbruck, 6020 Innsbruck, Austria; (B.S.); (L.K.); (H.T.)
| | - Leopold Kremser
- Protein Core Facility, Institute of Medical Biochemistry, Center of Chemistry and Biomedicine, Medical University of Innsbruck, 6020 Innsbruck, Austria; (B.S.); (L.K.); (H.T.)
| | - Heribert Talasz
- Protein Core Facility, Institute of Medical Biochemistry, Center of Chemistry and Biomedicine, Medical University of Innsbruck, 6020 Innsbruck, Austria; (B.S.); (L.K.); (H.T.)
| | - Xiaohua He
- Western Regional Research Center, U.S. Department of Agriculture, Agricultural Research Service, Albany, CA 74710, USA;
| | - Elisa Varrone
- Department of Medical and Surgical Sciences, School of Medicine, University of Bologna, 40126 Bologna, Italy; (E.V.); (M.B.)
| | - Maurizio Brigotti
- Department of Medical and Surgical Sciences, School of Medicine, University of Bologna, 40126 Bologna, Italy; (E.V.); (M.B.)
| | - Gianluigi Ardissino
- Center for HUS Prevention, Control and Management at Pediatric Nephrology, Dialysis and Transplant Unit, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | - Dorothea Orth-Höller
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (S.K.); (S.H.); (M.M.); (V.F.)
- MB-LAB–Clinical Microbiology Laboratory, 6020 Innsbruck, Austria
| | - Reinhard Würzner
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (S.K.); (S.H.); (M.M.); (V.F.)
| |
Collapse
|
38
|
Dhatt PS, Chiu S, Moon TS. Microbial thermogenesis is dependent on ATP concentrations and the protein kinases ArcB, GlnL, and YccC. PLoS Biol 2023; 21:e3002180. [PMID: 37862351 PMCID: PMC10619766 DOI: 10.1371/journal.pbio.3002180] [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: 05/14/2023] [Revised: 11/01/2023] [Accepted: 09/14/2023] [Indexed: 10/22/2023] Open
Abstract
Organisms necessarily release heat energy in their pursuit of survival. This process is known as cellular thermogenesis and is implicated in many processes from cancer metabolism to spontaneous farm fires. However, the molecular basis for this fundamental phenomenon is yet to be elucidated. Here, we show that the major players involved in the cellular thermogenesis of Escherichia coli are the protein kinases ArcB, GlnL, and YccC. We also reveal the substrate-level control of adenosine triphosphate (ATP)-driven autophosphorylation that governs cellular thermogenesis. Specifically, through live cell microcalorimetry, we find these regulatory proteins, when knocked out in a model E. coli strain, dysregulate cellular thermogenesis. This dysregulation can be seen in an average 25% or greater increase in heat output by these cells. We also discover that both heat output and intracellular ATP levels are maximal during the late log phase of growth. Additionally, we show that microbial thermogenesis can be engineered through overexpressing glnL. Our results demonstrate a correlation between ATP concentrations in the cell and a cell's ability to generate excess heat. We expect this work to be the foundation for engineering thermogenically tuned organisms for a variety of applications.
Collapse
Affiliation(s)
- Puneet Singh Dhatt
- Department of Energy, Environmental and Chemical Engineering, Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - Stephen Chiu
- Department of Energy, Environmental and Chemical Engineering, Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - Tae Seok Moon
- Department of Energy, Environmental and Chemical Engineering, Washington University in St. Louis, St. Louis, Missouri, United States of America
- Division of Biology and Biomedical Sciences, Washington University in St. Louis, St. Louis, Missouri, United States of America
| |
Collapse
|
39
|
Magnuson JT, Monticelli G, Schlenk D, Bisesi JH, Pampanin DM. Connecting gut microbiome changes with fish health conditions in juvenile Atlantic cod (Gadus morhua) exposed to dispersed crude oil. ENVIRONMENTAL RESEARCH 2023; 234:116516. [PMID: 37399986 DOI: 10.1016/j.envres.2023.116516] [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: 05/04/2023] [Revised: 06/23/2023] [Accepted: 06/25/2023] [Indexed: 07/05/2023]
Abstract
Polycyclic aromatic hydrocarbons found in crude oil can impair fish health following sublethal exposure. However, the dysbiosis of microbial communities within the fish host and influence it has on the toxic response of fish following exposure has been less characterized, particularly in marine species. To better understand the effect of dispersed crude oil (DCO) on juvenile Atlantic cod (Gadus morhua) microbiota composition and potential targets of exposure within the gut, fish were exposed to 0.05 ppm DCO for 1, 3, 7, or 28 days and 16 S metagenomic and metatranscriptomic sequencing on the gut and RNA sequencing on intestinal content were conducted. In addition to assessing species composition, richness, and diversity from microbial gut community analysis and transcriptomic profiling, the functional capacity of the microbiome was determined. Mycoplasma and Aliivibrio were the two most abundant genera after DCO exposure and Photobacterium the most abundant genus in controls, after 28 days. Metagenomic profiles were only significantly different between treatments after a 28-day exposure. The top identified pathways were involved in energy and the biosynthesis of carbohydrates, fatty acids, amino acids, and cellular structure. Biological processes following fish transcriptomic profiling shared common pathways with microbial functional annotations such as energy, translation, amide biosynthetic process, and proteolysis. There were 58 differently expressed genes determined from metatranscriptomic profiling after 7 days of exposure. Predicted pathways that were altered included those involved in translation, signal transduction, and Wnt signaling. EIF2 signaling was consistently dysregulated following exposure to DCO, regardless of exposure duration, with impairments in IL-22 signaling and spermine and spermidine biosynthesis in fish after 28 days. Data were consistent with predictions of a potentially reduced immune response related to gastrointestinal disease. Herein, transcriptomic-level responses helped explain the relevance of differences in gut microbial communities in fish following DCO exposure.
Collapse
Affiliation(s)
- Jason T Magnuson
- Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, Stavanger, Norway.
| | - Giovanna Monticelli
- Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, Stavanger, Norway
| | - Daniel Schlenk
- Department of Environmental Sciences, University of California, Riverside, Riverside, CA, USA
| | - Joseph H Bisesi
- Department of Environmental and Global Health, University of Florida, Gainesville, FL, USA
| | - Daniela M Pampanin
- Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, Stavanger, Norway
| |
Collapse
|
40
|
Weiland-Bräuer N, Koutsouveli V, Langfeldt D, Schmitz RA. First insights into the Aurelia aurita transcriptome response upon manipulation of its microbiome. Front Microbiol 2023; 14:1183627. [PMID: 37637120 PMCID: PMC10448538 DOI: 10.3389/fmicb.2023.1183627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 07/18/2023] [Indexed: 08/29/2023] Open
Abstract
Introduction The associated diverse microbiome contributes to the overall fitness of Aurelia aurita, particularly to asexual reproduction. However, how A. aurita maintains this specific microbiome or reacts to manipulations is unknown. Methods In this report, the response of A. aurita to manipulations of its native microbiome was studied by a transcriptomics approach. Microbiome-manipulated polyps were generated by antibiotic treatment and challenging polyps with a non-native, native, and potentially pathogenic bacterium. Total RNA extraction followed by RNAseq resulted in over 155 million reads used for a de novo assembly. Results The transcriptome analysis showed that the antibiotic-induced change and resulting reduction of the microbiome significantly affected the host transcriptome, e.g., genes involved in processes related to immune response and defense mechanisms were highly upregulated. Similarly, manipulating the microbiome by challenging the polyp with a high load of bacteria (2 × 107 cells/polyp) resulted in induced transcription of apoptosis-, defense-, and immune response genes. A second focus was on host-derived quorum sensing interference as a potential defense strategy. Quorum Quenching (QQ) activities and the respective encoding QQ-ORFs of A. aurita were identified by functional screening a cDNA-based expression library generated in Escherichia coli. Corresponding sequences were identified in the transcriptome assembly. Moreover, gene expression analysis revealed differential expression of QQ genes depending on the treatment, strongly suggesting QQ as an additional defense strategy. Discussion Overall, this study allows first insights into A. aurita's response to manipulating its microbiome, thus paving the way for an in-depth analysis of the basal immune system and additional fundamental defense strategies.
Collapse
Affiliation(s)
| | - Vasiliki Koutsouveli
- GEOMAR Helmholtz Center for Ocean Research Kiel, Düsternbrooker Weg, Kiel, Germany
| | | | - Ruth A. Schmitz
- Institute of General Microbiology, Kiel University, Kiel, Germany
| |
Collapse
|
41
|
Kutluk H, Bastounis EE, Constantinou I. Integration of Extracellular Matrices into Organ-on-Chip Systems. Adv Healthc Mater 2023; 12:e2203256. [PMID: 37018430 PMCID: PMC11468608 DOI: 10.1002/adhm.202203256] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/20/2023] [Indexed: 04/07/2023]
Abstract
The extracellular matrix (ECM) is a complex, dynamic network present within all tissues and organs that not only acts as a mechanical support and anchorage point but can also direct fundamental cell behavior, function, and characteristics. Although the importance of the ECM is well established, the integration of well-controlled ECMs into Organ-on-Chip (OoC) platforms remains challenging and the methods to modulate and assess ECM properties on OoCs remain underdeveloped. In this review, current state-of-the-art design and assessment of in vitro ECM environments is discussed with a focus on their integration into OoCs. Among other things, synthetic and natural hydrogels, as well as polydimethylsiloxane (PDMS) used as substrates, coatings, or cell culture membranes are reviewed in terms of their ability to mimic the native ECM and their accessibility for characterization. The intricate interplay among materials, OoC architecture, and ECM characterization is critically discussed as it significantly complicates the design of ECM-related studies, comparability between works, and reproducibility that can be achieved across research laboratories. Improving the biomimetic nature of OoCs by integrating properly considered ECMs would contribute to their further adoption as replacements for animal models, and precisely tailored ECM properties would promote the use of OoCs in mechanobiology.
Collapse
Affiliation(s)
- Hazal Kutluk
- Institute of Microtechnology (IMT)Technical University of BraunschweigAlte Salzdahlumer Str. 20338124BraunschweigGermany
- Center of Pharmaceutical Engineering (PVZ)Technical University of BraunschweigFranz‐Liszt‐Str. 35a38106BraunschweigGermany
| | - Effie E. Bastounis
- Institute of Microbiology and Infection Medicine (IMIT)Eberhard Karls University of TübingenAuf der Morgenstelle 28, E872076TübingenGermany
- Cluster of Excellence “Controlling Microbes to Fight Infections” EXC 2124Eberhard Karls University of TübingenAuf der Morgenstelle 2872076TübingenGermany
| | - Iordania Constantinou
- Institute of Microtechnology (IMT)Technical University of BraunschweigAlte Salzdahlumer Str. 20338124BraunschweigGermany
- Center of Pharmaceutical Engineering (PVZ)Technical University of BraunschweigFranz‐Liszt‐Str. 35a38106BraunschweigGermany
| |
Collapse
|
42
|
Ahn JS, Choi YJ, Kim HB, Chung HJ, Hong ST. Identification of the Intestinal Microbes Associated with Locomotion. Int J Mol Sci 2023; 24:11392. [PMID: 37511151 PMCID: PMC10380270 DOI: 10.3390/ijms241411392] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/08/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Given the impact of the gut microbiome on human physiology and aging, it is possible that the gut microbiome may affect locomotion in the same way as the host's own genes. There is not yet any direct evidence linking the gut microbiome to locomotion, though there are some potential connections, such as regular physical activity and the immune system. In this study, we demonstrate that the gut microbiome can contribute differently to locomotion. We remodeled the original gut microbiome of mice through fecal microbiota transplantation (FMT) using human feces and compared the changes in locomotion of the same mice before and three months after FMT. We found that FMT affected locomotion in three different ways: positive, none (the same), and negative. Analysis of the phylogenesis, α-diversities, and β-diversities of the gut microbiome in the three groups showed that a more diverse group of intestinal microbes was established after FMT in each of the three groups, indicating that the human gut microbiome is more diverse than that of mice. The FMT-remodeled gut microbiome in each group was also different from each other. Fold change and linear correlation analyses identified Lacrimispora indolis, Pseudoflavonifractor phocaeensis, and Alistipes senegalensis in the gut microbiome as positive contributors to locomotion, while Sphingobacterium cibi, Prevotellamassilia timonensis, Parasutterella excrementihominis, Faecalibaculum rodentium, and Muribaculum intestinale were found to have negative effects. This study not only confirms the presence of gut microbiomes that contribute differently to locomotion, but also explains the mixed results in research on the association between the gut microbiome and locomotion.
Collapse
Affiliation(s)
- Ji-Seon Ahn
- Gwangju Center, Korea Basic Science Institute, Gwangju 61751, Republic of Korea
| | - Yu-Jin Choi
- Gwangju Center, Korea Basic Science Institute, Gwangju 61751, Republic of Korea
| | - Han-Byeol Kim
- Gwangju Center, Korea Basic Science Institute, Gwangju 61751, Republic of Korea
- Department of Biomedical Sciences and Institute for Medical Science, Chonbuk National University Medical School, Jeonju 54907, Republic of Korea
| | - Hea-Jong Chung
- Gwangju Center, Korea Basic Science Institute, Gwangju 61751, Republic of Korea
| | - Seong-Tshool Hong
- Department of Biomedical Sciences and Institute for Medical Science, Chonbuk National University Medical School, Jeonju 54907, Republic of Korea
| |
Collapse
|
43
|
Lou S, Yang M, Li T, Zhao W, Cevasco H, Yang YT, Gerstein M. Constructing a full, multiple-layer interactome for SARS-CoV-2 in the context of lung disease: Linking the virus with human genes and microbes. PLoS Comput Biol 2023; 19:e1011222. [PMID: 37410793 PMCID: PMC10325097 DOI: 10.1371/journal.pcbi.1011222] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 05/28/2023] [Indexed: 07/08/2023] Open
Abstract
The COVID-19 pandemic caused by the SARS-CoV-2 virus has resulted in millions of deaths worldwide. The disease presents with various manifestations that can vary in severity and long-term outcomes. Previous efforts have contributed to the development of effective strategies for treatment and prevention by uncovering the mechanism of viral infection. We now know all the direct protein-protein interactions that occur during the lifecycle of SARS-CoV-2 infection, but it is critical to move beyond these known interactions to a comprehensive understanding of the "full interactome" of SARS-CoV-2 infection, which incorporates human microRNAs (miRNAs), additional human protein-coding genes, and exogenous microbes. Potentially, this will help in developing new drugs to treat COVID-19, differentiating the nuances of long COVID, and identifying histopathological signatures in SARS-CoV-2-infected organs. To construct the full interactome, we developed a statistical modeling approach called MLCrosstalk (multiple-layer crosstalk) based on latent Dirichlet allocation. MLCrosstalk integrates data from multiple sources, including microbes, human protein-coding genes, miRNAs, and human protein-protein interactions. It constructs "topics" that group SARS-CoV-2 with genes and microbes based on similar patterns of co-occurrence across patient samples. We use these topics to infer linkages between SARS-CoV-2 and protein-coding genes, miRNAs, and microbes. We then refine these initial linkages using network propagation to contextualize them within a larger framework of network and pathway structures. Using MLCrosstalk, we identified genes in the IL1-processing and VEGFA-VEGFR2 pathways that are linked to SARS-CoV-2. We also found that Rothia mucilaginosa and Prevotella melaninogenica are positively and negatively correlated with SARS-CoV-2 abundance, a finding corroborated by analysis of single-cell sequencing data.
Collapse
Affiliation(s)
- Shaoke Lou
- Program in Computational Biology & Bioinformatics, Yale University, New Haven, Connecticut, United States of America
- Department of Molecular Biophysics & Biochemistry, Yale University, New Haven, Connecticut, United States of America
| | - Mingjun Yang
- School of Electronic Engineering and Computer Science, Queen Mary University of London, Mile End Road, London, United Kingdom
| | - Tianxiao Li
- Program in Computational Biology & Bioinformatics, Yale University, New Haven, Connecticut, United States of America
- Department of Molecular Biophysics & Biochemistry, Yale University, New Haven, Connecticut, United States of America
| | - Weihao Zhao
- Program in Computational Biology & Bioinformatics, Yale University, New Haven, Connecticut, United States of America
- Department of Molecular Biophysics & Biochemistry, Yale University, New Haven, Connecticut, United States of America
| | - Hannah Cevasco
- Program in Computational Biology & Bioinformatics, Yale University, New Haven, Connecticut, United States of America
| | - Yucheng T. Yang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Shanghai, China
- MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Mark Gerstein
- Program in Computational Biology & Bioinformatics, Yale University, New Haven, Connecticut, United States of America
- Department of Molecular Biophysics & Biochemistry, Yale University, New Haven, Connecticut, United States of America
- Department of Computer Science, Yale University, New Haven, Connecticut, United States of America
- Department of Statistics & Data Science Yale University, New Haven, Connecticut, United States of America
- Department of Biomedical Informatics & Data Science, Yale University, New Haven, Connecticut, United States of America
| |
Collapse
|
44
|
Blejwas K, Beard L, Buchanan J, Lausen CL, Neubaum D, Tobin A, Weller TJ. COULD WHITE-NOSE SYNDROME MANIFEST DIFFERENTLY IN MYOTIS LUCIFUGUS IN WESTERN VERSUS EASTERN REGIONS OF NORTH AMERICA? A REVIEW OF FACTORS. J Wildl Dis 2023; 59:381-397. [PMID: 37270186 DOI: 10.7589/jwd-d-22-00050] [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: 04/27/2022] [Accepted: 02/28/2023] [Indexed: 06/05/2023]
Abstract
White-nose syndrome (WNS) has notably affected the abundance of Myotis lucifugus (little brown myotis) in North America. Thus far, substantial mortality has been restricted to the eastern part of the continent where the cause of WNS, the invasive fungus Pseudogymnoascus destructans, has infected bats since 2006. To date, the state of Washington is the only area in the Western US or Canada (the Rocky Mountains and further west in North America) with confirmed cases of WNS in bats, and there the disease has spread more slowly than it did in Eastern North America. Here, we review differences between M. lucifugus in western and eastern parts of the continent that may affect transmission, spread, and severity of WNS in the West and highlight important gaps in knowledge. We explore the hypothesis that western M. lucifugus may respond differently to WNS on the basis of different hibernation strategies, habitat use, and greater genetic structure. To document the effect of WNS on M. lucifugus in the West most effectively, we recommend focusing on maternity roosts for strategic disease surveillance and monitoring abundance. We further recommend continuing the challenging work of identifying hibernation and swarming sites to better understand the microclimates, microbial communities, and role in disease transmission of these sites, as well as the ecology and hibernation physiology of bats in noncavernous hibernacula.
Collapse
Affiliation(s)
- Karen Blejwas
- Alaska Department of Fish and Game, PO Box 110024, Juneau, Alaska 99811, USA
- Except for the first author, all others are listed in alphabetical order
| | - Laura Beard
- Wyoming Game and Fish Department, 260 Buena Vista, Lander, Wyoming 82520, USA
| | - Joseph Buchanan
- Washington Department of Fish and Wildlife, PO Box 43200, Olympia, Washington 98501, USA
| | - Cori L Lausen
- Wildlife Conservation Society Canada, 202 B Avenue, Kaslo, British Columbia V0G 1M0, Canada
| | - Daniel Neubaum
- Colorado Parks and Wildlife, 711 Independent Ave., Grand Junction, Colorado 81507, USA
| | - Abigail Tobin
- Washington Department of Fish and Wildlife, PO Box 43200, Olympia, Washington 98501, USA
| | - Theodore J Weller
- USDA Forest Service, Pacific Southwest Research Station, 1700 Bayview Drive, Arcata, California 95521, USA
| |
Collapse
|
45
|
Aziz T, Khan AA, Tzora A, Voidarou CC, Skoufos I. Dietary Implications of the Bidirectional Relationship between the Gut Microflora and Inflammatory Diseases with Special Emphasis on Irritable Bowel Disease: Current and Future Perspective. Nutrients 2023; 15:2956. [PMID: 37447285 DOI: 10.3390/nu15132956] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/19/2023] [Accepted: 06/24/2023] [Indexed: 07/15/2023] Open
Abstract
The immune system is vital for safeguarding the human body against infections and inflammatory diseases. The role of diet and meal patterns in modulating immune function is complex, and highlighting this topic is crucial for identifying potential ways to improve immune health. In Europe, the Mediterranean diet and Western diet are the most common dietary patterns, and gaining an understanding of how they affect immune function is essential for public health. There are numerous inflammatory diseases that are observed in younger and older people. Some of the common diseases include polymyalgia rheumatica (PMR), spinal muscular atrophy (SMA), vasculitis, sarcopenia, cirrhosis, cancer, and fibromyalgia, but the main focus in this review article is on irritable bowel disease (IBD). In general, dietary choices can have an immense impact on the microbial flora of the gut in people with inflammatory diseases. The intake of Mediterranean-style foods promotes the growth of healthy bacteria that enhances the function of the immune system. On the other hand, it is mostly seen that the intake of Western-style foods leads to the growth of harmful gut bacteria that contributes to inflammation and disease development by weakening the immune system. Additionally, inflammation in the gut can impact brain function, leading to mood disorders, such as anxiety and depression. Rare inflammatory diseases, such as psoriasis and sarcoidosis, are of main interest in this article. All the above-mentioned common and rare inflammatory diseases have a certain relationship with the microbiota of the gut. The gut microbiome plays a significant role in IBD; fiber and prebiotic interventions may represent promising adjunct therapies for pediatric IBD by targeting the gut microbiome. By advancing a good overall arrangement of microorganisms in the stomach through dietary mediations, working on the side effects and alleviating of diseases might be conceivable. The gut microbiota can be affected differently by various dietary fatty acid types. There is also an involvement of genetics in the progression of IBD, such as transcriptional factors, and one gene of interest is the LCT gene, which encodes for lactase, an enzyme responsible for digesting lactose in the gut.
Collapse
Affiliation(s)
- Tariq Aziz
- Laboratory of Animal Health, Food Hygiene and Quality, Department of Agriculture, University of Ioannina, 47100 Arta, Greece
| | - Ayaz Ali Khan
- Department of Biotechnology, University of Malakand, Chakdara 18800, Pakistan
| | - Athina Tzora
- Laboratory of Animal Health, Food Hygiene and Quality, Department of Agriculture, University of Ioannina, 47100 Arta, Greece
| | - Chrysoula Chrysa Voidarou
- Laboratory of Animal Health, Food Hygiene and Quality, Department of Agriculture, University of Ioannina, 47100 Arta, Greece
| | - Ioannis Skoufos
- Laboratory of Animal Health, Food Hygiene and Quality, Department of Agriculture, University of Ioannina, 47100 Arta, Greece
| |
Collapse
|
46
|
Henry LP, Bergelson J. Evolutionary implications of host genetic control for engineering beneficial microbiomes. CURRENT OPINION IN SYSTEMS BIOLOGY 2023; 34:None. [PMID: 37287906 PMCID: PMC10242548 DOI: 10.1016/j.coisb.2023.100455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Engineering new functions in the microbiome requires understanding how host genetic control and microbe-microbe interactions shape the microbiome. One key genetic mechanism underlying host control is the immune system. The immune system can promote stability in the composition of the microbiome by reshaping the ecological dynamics of its members, but the degree of stability will depend on the interplay between ecological context, immune system development, and higher-order microbe-microbe interactions. The eco-evolutionary interplay affecting composition and stability should inform the strategies used to engineer new functions in the microbiome. We conclude with recent methodological developments that provide an important path forward for both engineering new functionality in the microbiome and broadly understanding how ecological interactions shape evolutionary processes in complex biological systems.
Collapse
|
47
|
Jiang X, Zhang X, Sun Y, Sun Z, Li X, Liu L. Effects of Salmonella Enteritidis infection on TLRs gene expression and microbial diversity in cecum of laying hens. Heliyon 2023; 9:e16414. [PMID: 37265619 PMCID: PMC10230204 DOI: 10.1016/j.heliyon.2023.e16414] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 05/11/2023] [Accepted: 05/16/2023] [Indexed: 06/03/2023] Open
Abstract
Salmonella Enteritidis (SE) is an important foodborne pathogen primarily causing human disease through contaminated food and water. In the current study, to assess the effect of Salmonella Enteritidis infection on the immune system and the microbial diversity of cecum and oviduct in chickens, twelve 24-week-old SE-negative White Leghorn layers were randomly selected and divided into 2 groups. Chickens in the challenge group were orally inoculated with SE, and chickens in the control group received an equal amount of sterilized Phosphate Buffered Saline solution. Serum and tissue samples (cecum, oviduct, ovary, liver, spleen, and pancreas) were collected at 7 days and 14 days post-infection (dpi). Quantitative PCR was used to detect the expression of Toll-like receptors (TLRs) in the cecum, oviduct and ovary. To understand the influence of SE infection on the microbial profile of the cecum and oviduct, microbial community composition of the cecal contents and oviducal contents were analyzed through 16S rRNA sequencing. Results showed that SE infection caused damage to the digestive organs, reproductive organs, and immune organs in laying hens. The expression of TLR1a, TLR1b, TLR2, TLR4, TLR5, TLR7 and TLR15 in the cecum were induced, and the content of IFN-γ, TNF-α, IL-2 and IL-18 in serum increased after SE infection. The composition of the microbial community significantly changed in cecal content, the dominant phylum of Firmicutes increased, and Bacteroidetes decreased significantly. In the oviduct, the microbial diversity became complicated, the dominant bacteria Faecalibacterium was significantly increased, and Bacteroides was significantly decreased. This study investigated the effects of SE infection in laying hens, including host innate immunity, the expression of TLRs, and changes in the composition of microbes in the cecum and reproductive tract. Our results may provide a scientific basis for the Salmonella Enteritidis control in chicken, the maintenance of oviduct function, and the guarantee of clean egg production.
Collapse
Affiliation(s)
- Xintong Jiang
- College of Life Science, Shandong Agricultural University, Tai'an, Shandong, China
| | - Xiao Zhang
- College of Life Science, Shandong Agricultural University, Tai'an, Shandong, China
| | - Yuqing Sun
- College of Life Science, Shandong Agricultural University, Tai'an, Shandong, China
| | - Zhongtao Sun
- College of Life Science, Shandong Agricultural University, Tai'an, Shandong, China
| | - Xianyao Li
- College of Animal Science and Technology, Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Shandong Agricultural University, Tai'an, 271000, Shandong, China
| | - Liying Liu
- College of Life Science, Shandong Agricultural University, Tai'an, Shandong, China
| |
Collapse
|
48
|
Moore BN, Pluznick JL. Commensal microbiota regulate renal gene expression in a sex-specific manner. Am J Physiol Renal Physiol 2023; 324:F511-F520. [PMID: 37053490 PMCID: PMC10202489 DOI: 10.1152/ajprenal.00303.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 03/28/2023] [Accepted: 04/09/2023] [Indexed: 04/15/2023] Open
Abstract
The gut microbiome impacts host gene expression not only in the colon but also at distal sites including the liver, white adipose tissue, and spleen. The gut microbiome also influences the kidney and is associated with renal diseases and pathologies; however, a role for the gut microbiome to modulate renal gene expression has not been examined. To determine if microbes modulate renal gene expression, we used whole organ RNA sequencing to compare gene expression in C57Bl/6 mice that were germ free (lacking gut microbiota) versus conventionalized (gut microbiota reintroduced using an oral gavage of a fecal slurry composed of mixed stool). 16S sequencing showed that male and female mice were similarly conventionalized, although Verrucomicrobia was higher in male mice. We found that renal gene expression was differentially regulated in the presence vs. absence of microbiota and that these changes were largely sex specific. Although microbes also influenced gene expression in the liver and large intestine, most differentially expressed genes (DEGs) in the kidney were not similarly regulated in the liver or large intestine. This demonstrates that the influence of the gut microbiota on gene expression is tissue specific. However, a minority of genes (n = 4 in males and n = 6 in females) were similarly regulated in all three tissues examined, including genes associated with circadian rhythm (period 1 in males and period 2 in females) and metal binding (metallothionein 1 and metallothionein 2 in both males and females). Finally, using a previously published single-cell RNA-sequencing dataset, we assigned a subset of DEGs to specific kidney cell types, revealing clustering of DEGs by cell type and/or sex.NEW & NOTEWORTHY It is unknown whether the microbiome influences host gene expression in the kidney. Here, we utilized an unbiased, bulk RNA-sequencing approach to compare gene expression in the kidneys of male and female mice with or without gut microbiota. This report demonstrates that renal gene expression is modulated by the microbiome in a sex- and tissue-specific manner.
Collapse
Affiliation(s)
- Brittni N Moore
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Jennifer L Pluznick
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| |
Collapse
|
49
|
Nemoto S, Kubota T, Ohno H. Exploring body weight-influencing gut microbiota by elucidating the association with diet and host gene expression. Sci Rep 2023; 13:5593. [PMID: 37019989 PMCID: PMC10076326 DOI: 10.1038/s41598-023-32411-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 03/27/2023] [Indexed: 04/07/2023] Open
Abstract
We aimed to identify gut microbiota that influences body weight by elucidating the association with diets and host genes. Germ-free (GF) mice with and without fecal microbiota transplant (FMT) were fed a normal, high-carbohydrate, or high-fat diet. FMT mice exhibited greater total body weight; adipose tissue and liver weights; blood glucose, insulin, and total cholesterol levels; and oil droplet size than the GF mice, regardless of diet. However, the extent of weight gain and metabolic parameter levels associated with gut microbiota depended on the nutrients ingested. For example, a disaccharide- or polysaccharide-rich diet caused more weight gain than a monosaccharide-rich diet. An unsaturated fatty acid-rich diet had a greater microbial insulin-increasing effect than a saturated fatty acid-rich diet. Perhaps the difference in microbial metabolites produced from substances taken up by the host created metabolic differences. Therefore, we analyzed such dietary influences on gut microbiota, differentially expressed genes between GF and FMT mice, and metabolic factors, including body weight. The results revealed a correlation between increased weight gain, a fat-rich diet, increased Ruminococcaceae abundance, and decreased claudin 22 gene expression. These findings suggest that weight regulation might be possible through the manipulation of the gut microbiota metabolism using the host's diet.
Collapse
Affiliation(s)
- Shino Nemoto
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan.
| | - Tetsuya Kubota
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
- Division of Diabetes and Metabolism, The Institute of Medical Science, Asahi Life Foundation, Tokyo, Japan
- Department of Clinical Nutrition, National Institutes of Biomedical Innovation, Health and Nutrition, Tokyo, Japan
- Division of Cardiovascular Medicine, Toho University Ohashi Medical Center, Tokyo, Japan
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroshi Ohno
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
- Laboratory for Immune Regulation, Graduate School of Medical and Pharmaceutical Sciences, Chiba University, Chiba, Japan
- Immunobiology Laboratory, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan
| |
Collapse
|
50
|
Mohamed AR, Ochsenkühn MA, Kazlak AM, Moustafa A, Amin SA. The coral microbiome: towards an understanding of the molecular mechanisms of coral-microbiota interactions. FEMS Microbiol Rev 2023; 47:fuad005. [PMID: 36882224 PMCID: PMC10045912 DOI: 10.1093/femsre/fuad005] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 02/10/2023] [Accepted: 02/15/2023] [Indexed: 03/09/2023] Open
Abstract
Corals live in a complex, multipartite symbiosis with diverse microbes across kingdoms, some of which are implicated in vital functions, such as those related to resilience against climate change. However, knowledge gaps and technical challenges limit our understanding of the nature and functional significance of complex symbiotic relationships within corals. Here, we provide an overview of the complexity of the coral microbiome focusing on taxonomic diversity and functions of well-studied and cryptic microbes. Mining the coral literature indicate that while corals collectively harbour a third of all marine bacterial phyla, known bacterial symbionts and antagonists of corals represent a minute fraction of this diversity and that these taxa cluster into select genera, suggesting selective evolutionary mechanisms enabled these bacteria to gain a niche within the holobiont. Recent advances in coral microbiome research aimed at leveraging microbiome manipulation to increase coral's fitness to help mitigate heat stress-related mortality are discussed. Then, insights into the potential mechanisms through which microbiota can communicate with and modify host responses are examined by describing known recognition patterns, potential microbially derived coral epigenome effector proteins and coral gene regulation. Finally, the power of omics tools used to study corals are highlighted with emphasis on an integrated host-microbiota multiomics framework to understand the underlying mechanisms during symbiosis and climate change-driven dysbiosis.
Collapse
Affiliation(s)
- Amin R Mohamed
- Biology Program, New York University Abu Dhabi, Abu Dhabi 129188, United Arab Emirates
| | - Michael A Ochsenkühn
- Biology Program, New York University Abu Dhabi, Abu Dhabi 129188, United Arab Emirates
| | - Ahmed M Kazlak
- Systems Genomics Laboratory, American University in Cairo, New Cairo 11835, Egypt
- Biotechnology Graduate Program, American University in Cairo, New Cairo 11835, Egypt
| | - Ahmed Moustafa
- Systems Genomics Laboratory, American University in Cairo, New Cairo 11835, Egypt
- Biotechnology Graduate Program, American University in Cairo, New Cairo 11835, Egypt
- Department of Biology, American University in Cairo, New Cairo 11835, Egypt
| | - Shady A Amin
- Biology Program, New York University Abu Dhabi, Abu Dhabi 129188, United Arab Emirates
- Center for Genomics and Systems Biology (CGSB), New York University Abu Dhabi, Abu Dhabi 129188, United Arab Emirates
| |
Collapse
|